32 results on '"Sanborn JZ"'
Search Results
2. Abstract P2-09-04: Identification of a neoantigen targeted by tumor-infiltrating lymphocytes in a patient with Her2+ breast cancer
- Author
-
Reimann, H, primary, Nguyen, A, additional, Hübner, H, additional, Erber, R, additional, Bausenwein, J, additional, Van der Meijden, ED, additional, Lux, MP, additional, Jud, S, additional, Griffioen, M, additional, Rauh, C, additional, Sanborn, JZ, additional, Benz, SC, additional, Rabizadeh, S, additional, Beckmann, MW, additional, Mackensen, A, additional, Rübner, M, additional, Fasching, PA, additional, and Kremer, AN, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P2-04-26: Identifying patient-specific neoepitopes for cell-based and vaccine immunotherapy across breast cancer classifications reveals rarely shared recurrent neoepitopes
- Author
-
Nguyen, A, primary, Sanborn, JZ, additional, Vaske, CJ, additional, Rabizadeh, S, additional, Niazi, K, additional, Soon-Shiong, P, additional, and Benz, SC, additional
- Published
- 2017
- Full Text
- View/download PDF
4. Abstract P6-04-14: Integrating whole genome sequencing data with RNAseq, pathway analysis, and quantitative proteomics to determine prognosis after standard adjuvant treatment with trastuzumab and chemotherapy in primary breast cancer patients
- Author
-
Benz, SC, primary, Rabizadeh, S, additional, Cecchi, F, additional, Beckman, MW, additional, Brucker, SY, additional, Hartmann, A, additional, Golovato, J, additional, Hembrough, T, additional, Janni, W, additional, Rack, B, additional, Sanborn, JZ, additional, Schneeweiss, A, additional, Vaske, CJ, additional, Soon-Shiong, P, additional, and Fasching, PA, additional
- Published
- 2016
- Full Text
- View/download PDF
5. Integrated genomic analyses of ovarian carcinoma
- Author
-
Bell, D, Berchuck, A, Birrer, M, Chien, J, Cramer, DW, Dao, F, Dhir, R, DiSaia, P, Gabra, H, Glenn, P, Godwin, AK, Triche, T, Berman, BP, Van den Berg, DJ, Buckley, J, Baylin, SB, Zhang, J, Spellman, PT, Purdom, E, Iacocca, M, Shelton, T, Voet, D, Neuvial, P, Bengtsson, H, Jakkula, LR, Durinck, S, Han, J, Dorton, S, Marr, H, Zhang, H, Choi, YG, Wang, V, Wilkinson, J, Nguyen, H, Wang, NJ, Imielinski, M, Ngai, J, Conboy, JG, Parvin, B, Feiler, HS, Speed, TP, Gray, JW, Wu, CJ, Bloom, T, Levine, DA, Li, L, Socci, ND, Liang, Y, Taylor, BS, Kalloger, S, Schultz, N, Borsu, L, Lash, AE, Brennan, C, Ardlie, K, Viale, A, Shukla, S, Grimm, D, Sander, C, Ladanyi, M, Hoadley, KA, Meng, S, Du, Y, Karlan, BY, Shi, Y, Fennell, T, Cibulskis, K, Lawrence, MS, Meyerson, M, Hatfield, M, Mills, GB, Sivachenko, A, Jing, R, Park, RW, Liu, Y, Park, PJ, Ramos, AH, Noble, M, Chin, L, Carter, H, Kim, D, Morris, S, Winckler, W, Karchin, R, Morrison, C, Baldwin, J, Korkola, JE, Yena, P, Heiser, LM, Getz, G, Cho, RJ, Hu, Z, Gabriel, S, Mutch, D, Cerami, E, Rhodes, P, Olshen, A, Verhaak, RGW, Lander, ES, Reva, B, Antipin, Y, Shen, R, Olvera, N, Mankoo, P, Sheridan, R, Ciriello, G, Sherman, M, Chang, WK, Bernanke, JA, Hayes, DN, Carter, SL, Haussler, D, Orsulic, S, Benz, CC, Paulauskis, J, Stuart, JM, Zhang, N, Benz, SC, Sanborn, JZ, Vaske, CJ, Mermel, CH, Zhu, J, Szeto, C, Scott, GK, Yau, C, Rabeno, B, Ding, L, Park, K, Balu, S, Perou, CM, Saksena, G, Wilkerson, MD, Millis, S, Kahn, A, Turman, YJ, Fulton, RS, Onofrio, RC, Greene, JM, Sfeir, R, Jensen, MA, Chen, J, Whitmore, J, Alonso, S, Jordan, J, Chu, A, Rader, JS, Koboldt, DC, Zang, D, Gross, J, Barker, A, Compton, C, Eley, G, Ferguson, M, Fielding, P, Gerhard, DS, Myles, R, McLellan, MD, Schaefer, C, Helms, EB, Shaw, KRM, Sikic, BI, Vaught, J, Vockley, JB, Good, PJ, Guyer, MS, Ozenberger, B, Wylie, T, Peterson, J, Thomson, E, Smith-McCune, K, Sood, AK, Bowtell, D, Hubbard, D, Penny, R, Testa, JR, Chang, K, Walker, J, Dinh, HH, Drummond, JA, Fowler, G, Zhou, X, Gunaratne, P, Hawes, AC, Kovar, CL, Lewis, LR, Gupta, S, Morgan, MB, O'Laughlin, M, Newsham, IF, Santibanez, J, Reid, JG, Trevino, LR, Wu, J, Wu, Y-Q, Wang, M, Muzny, DM, Wheeler, DA, Gibbs, RA, Crenshaw, A, Sivachenko, AY, Topal, MD, Sougnez, C, Dooling, DJ, Fulton, L, Akbani, R, Abbott, R, Dees, ND, Zhang, Q, Kandoth, C, Wendl, M, Schierding, W, Shen, D, Harris, CC, Baggerly, KA, Schmidt, H, Wilson, RK, Kalicki, J, Delehaunty, KD, Fronick, CC, Demeter, R, Cook, L, Wallis, JW, Lin, L, Magrini, VJ, Yung, WK, Hodges, JS, Protopopov, A, Eldred, JM, Smith, SM, Pohl, CS, Vandin, F, Raphael, BJ, Weinstock, GM, Mardis, R, Kim, TM, Hartmann, L, Perna, I, Xiao, Y, Ren, G, Sathiamoorthy, N, Petrelli, N, Lee, E, Kucherlapati, R, Absher, DM, Huang, M, Waite, L, Sherlock, G, Brooks, JD, Li, JZ, Weinstein, JN, Xu, J, Myers, RM, Laird, PW, Cope, L, Herman, JG, Shen, H, Huntsman, DG, Weisenberger, DJ, Noushmehr, H, Pan, F, Massachusetts Institute of Technology. Department of Biology, Lander, Eric S., and Meyerson, Matthew L.
- Subjects
endocrine system diseases ,Serous carcinoma ,Messenger ,Gene Dosage ,Cancer Genome Atlas Research Network ,GYNECOLOGIC-ONCOLOGY-GROUP ,GRADE SEROUS CARCINOMA ,0302 clinical medicine ,Ovarian carcinoma ,Aged ,Carcinoma ,DNA Methylation ,Female ,Gene Expression Profiling ,Gene Expression Regulation, Neoplastic ,Humans ,MicroRNAs ,Middle Aged ,Mutation ,Ovarian Neoplasms ,RNA, Messenger ,Genomics ,Multidisciplinary ,Genetics ,HYBRID SELECTION ,0303 health sciences ,female genital diseases and pregnancy complications ,3. Good health ,Multidisciplinary Sciences ,Serous fluid ,BRCA MUTATION CARRIERS ,030220 oncology & carcinogenesis ,DNA methylation ,PARP inhibitor ,Science & Technology - Other Topics ,General Science & Technology ,Biology ,Article ,03 medical and health sciences ,CLEAR-CELL CARCINOMA ,MD Multidisciplinary ,microRNA ,HIGH-THROUGHPUT ANNOTATION ,medicine ,DRIVER MUTATIONS ,Gene ,030304 developmental biology ,Neoplastic ,Science & Technology ,MUTANT-CELLS ,SOMATIC MUTATIONS ,medicine.disease ,CANCER STATISTICS ,Gene Expression Regulation ,Cancer research ,RNA ,Ovarian cancer - Abstract
A catalogue of molecular aberrations that cause ovarian cancer is critical for developing and deploying therapies that will improve patients’ lives. The Cancer Genome Atlas project has analysed messenger RNA expression, microRNA expression, promoter methylation and DNA copy number in 489 high-grade serous ovarian adenocarcinomas and the DNA sequences of exons from coding genes in 316 of these tumours. Here we report that high-grade serous ovarian cancer is characterized by TP53 mutations in almost all tumours (96%); low prevalence but statistically recurrent somatic mutations in nine further genes including NF1, BRCA1, BRCA2, RB1 and CDK12; 113 significant focal DNA copy number aberrations; and promoter methylation events involving 168 genes. Analyses delineated four ovarian cancer transcriptional subtypes, three microRNA subtypes, four promoter methylation subtypes and a transcriptional signature associated with survival duration, and shed new light on the impact that tumours with BRCA1/2 (BRCA1 or BRCA2) and CCNE1 aberrations have on survival. Pathway analyses suggested that homologous recombination is defective in about half of the tumours analysed, and that NOTCH and FOXM1 signalling are involved in serous ovarian cancer pathophysiology., National Institutes of Health (U.S.) (Grant U54HG003067), National Institutes of Health (U.S.) (Grant U54HG003273), National Institutes of Health (U.S.) (Grant U54HG003079), National Institutes of Health (U.S.) (Grant U24CA126543), National Institutes of Health (U.S.) (Grant U24CA126544), National Institutes of Health (U.S.) (Grant U24CA126546), National Institutes of Health (U.S.) (Grant U24CA126551), National Institutes of Health (U.S.) (Grant U24CA126554), National Institutes of Health (U.S.) (Grant U24CA126561), National Institutes of Health (U.S.) (Grant U24CA126563), National Institutes of Health (U.S.) (Grant U24CA143882), National Institutes of Health (U.S.) (Grant U24CA143731), National Institutes of Health (U.S.) (Grant U24CA143835), National Institutes of Health (U.S.) (Grant U24CA143845), National Institutes of Health (U.S.) (Grant U24CA143858), National Institutes of Health (U.S.) (Grant U24CA144025), National Institutes of Health (U.S.) (Grant U24CA143866), National Institutes of Health (U.S.) (Grant U24CA143867), National Institutes of Health (U.S.) (Grant U24CA143848), National Institutes of Health (U.S.) (Grant U24CA143843), National Institutes of Health (U.S.) (Grant R21CA135877)
- Published
- 2010
6. Abstract P2-06-05: Single-cell RNA sequencing of paclitaxol-treated breast cancer cell lines to find individual cell response
- Author
-
Vaske, CJ, primary, Lee, W, additional, Benz, SC, additional, Sanborn, JZ, additional, Emerson, BM, additional, Pourmand, N, additional, and Lopez, Diaz F, additional
- Published
- 2012
- Full Text
- View/download PDF
7. P3-06-07: Integrated Genomic and Pathway Analysis Reveals Key Pathways across Breast Subtypes.
- Author
-
Benz, S, primary, Sanborn, JZ, additional, and Vaske, C, additional
- Published
- 2011
- Full Text
- View/download PDF
8. PD03-04: SuperPathway Analyses of Luminal and Basaloid Breast Cancers from the Cancer Genome Atlas (TCGA) Program.
- Author
-
Yau, C, primary, Benz, S, additional, Sanborn, JZ, additional, Stuart, J, additional, Haussler, D, additional, and Benz, C, additional
- Published
- 2011
- Full Text
- View/download PDF
9. UCSC cancer genomics browser.
- Author
-
Zhu, J, primary, Sanborn, JZ, additional, Wang, T, additional, Hsu, F, additional, Benz, S, additional, Szeto, C, additional, Esserman, L, additional, and Haussler, D, additional
- Published
- 2009
- Full Text
- View/download PDF
10. Efficient tumor clearance and diversified immunity through neoepitope vaccines and combinatorial immunotherapy
- Author
-
Lee, KL, primary, Benz, SC, additional, Hicks, KC, additional, Nguyen, A, additional, Gameiro, SR, additional, Palena, C, additional, Sanborn, JZ, additional, Su, Z, additional, Ordentlich, P, additional, Rohlin, L, additional, Lee, JH, additional, Rabizadeh, S, additional, Soon-Shiong, P, additional, Niazi, K, additional, Schlom, J, additional, and Hamilton, DH, additional
- Full Text
- View/download PDF
11. Identification and validation of expressed HLA-binding breast cancer neoepitopes for potential use in individualized cancer therapy.
- Author
-
Reimann H, Nguyen A, Sanborn JZ, Vaske CJ, Benz SC, Niazi K, Rabizadeh S, Spilman P, Mackensen A, Ruebner M, Hein A, Beckmann MW, van der Meijden ED, Bausenwein J, Kretschmann S, Griffioen M, Schlom J, Gulley JL, Lee KL, Hamilton DH, Soon-Shiong P, Fasching PA, and Kremer AN
- Subjects
- Female, Humans, Antigens, Neoplasm genetics, Breast Neoplasms genetics, Histocompatibility Antigens Class I immunology, Immunotherapy methods
- Abstract
Background: Therapeutic regimens designed to augment the immunological response of a patient with breast cancer (BC) to tumor tissue are critically informed by tumor mutational burden and the antigenicity of expressed neoepitopes. Herein we describe a neoepitope and cognate neoepitope-reactive T-cell identification and validation program that supports the development of next-generation immunotherapies., Methods: Using GPS Cancer, NantOmics research, and The Cancer Genome Atlas databases, we developed a novel bioinformatic-based approach which assesses mutational load, neoepitope expression, human leukocyte antigen (HLA)-binding prediction, and in vitro confirmation of T-cell recognition to preferentially identify targetable neoepitopes. This program was validated by application to a BC cell line and confirmed using tumor biopsies from two patients with BC enrolled in the Tumor-Infiltrating Lymphocytes and Genomics (TILGen) study., Results: The antigenicity and HLA-A2 restriction of the BC cell line predicted neoepitopes were determined by reactivity of T cells from HLA-A2-expressing healthy donors. For the TILGen subjects, tumor-infiltrating lymphocytes (TILs) recognized the predicted neoepitopes both as peptides and on retroviral expression in HLA-matched Epstein-Barr virus-lymphoblastoid cell line and BC cell line MCF-7 cells; PCR clonotyping revealed the presence of T cells in the periphery with T-cell receptors for the predicted neoepitopes. These high-avidity immune responses were polyclonal, mutation-specific and restricted to either HLA class I or II. Interestingly, we observed the persistence and expansion of polyclonal T-cell responses following neoadjuvant chemotherapy., Conclusions: We demonstrate our neoepitope prediction program allows for the successful identification of neoepitopes targeted by TILs in patients with BC, providing a means to identify tumor-specific immunogenic targets for individualized treatment, including vaccines or adoptively transferred cellular therapies., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
12. Reconstructing tumor history in breast cancer: signatures of mutational processes and response to neoadjuvant chemotherapy ⋆ .
- Author
-
Denkert C, Untch M, Benz S, Schneeweiss A, Weber KE, Schmatloch S, Jackisch C, Sinn HP, Golovato J, Karn T, Marmé F, Link T, Budczies J, Nekljudova V, Schmitt WD, Stickeler E, Müller V, Jank P, Parulkar R, Heinmöller E, Sanborn JZ, Schem C, Sinn BV, Soon-Shiong P, van Mackelenbergh M, Fasching PA, Rabizadeh S, and Loibl S
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Humans, Mutation, Prognosis, Prospective Studies, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Neoadjuvant Therapy
- Abstract
Background: Different endogenous and exogenous mutational processes act over the evolutionary history of a malignant tumor, driven by abnormal DNA editing, mutagens or age-related DNA alterations, among others, to generate the specific mutational landscape of each individual tumor. The signatures of these mutational processes can be identified in large genomic datasets. We investigated the hypothesis that genomic patterns of mutational signatures are associated with the clinical behavior of breast cancer, in particular chemotherapy response and survival, with a particular focus on therapy-resistant disease., Patients and Methods: Whole exome sequencing was carried out in 405 pretherapeutic samples from the prospective neoadjuvant multicenter GeparSepto study. We analyzed 11 mutational signatures including biological processes such as APOBEC-mutagenesis, homologous recombination deficiency (HRD), mismatch repair deficiency and also age-related or tobacco-induced alterations., Results: Different subgroups of breast carcinomas were defined mainly by differences in HRD-related and APOBEC-related mutational signatures and significant differences between hormone-receptor (HR)-negative and HR-positive tumors as well as correlations with age, Ki-67 and immunological parameters were observed. We could identify mutational processes that were linked to increased pathological complete response rates to neoadjuvant chemotherapy with high significance. In univariate analyses for HR-positive tumors signatures, S3 (HRD, P < 0.001) and S13 (APOBEC, P = 0.001) as well as exonic mutation rate (P = 0.002) were significantly correlated with increased pathological complete response rates. The signatures S3 (HRD, P = 0.006) and S4 (tobacco, P = 0.011) were prognostic for reduced disease-free survival of patients with chemotherapy-resistant tumors., Conclusion: The results of this investigation suggest that the clinical behavior of a tumor, in particular, response to neoadjuvant chemotherapy and disease-free survival of therapy-resistant tumors, could be predicted by the composition of mutational signatures as an indicator of the individual genomic history of a tumor. After additional validations, mutational signatures might be used to identify tumors with an increased response rate to neoadjuvant chemotherapy and to define therapy-resistant subgroups for future therapeutic interventions., Competing Interests: Disclosure CD reports personal fees from Novartis, personal fees and travel support from Roche, personal fees from MSD Oncology, from Daiichi Sankyo, grants from Myriad Genetics and GBG, other from Sividon Diagnostics/Myriad, outside the submitted work. In addition, CD has a patent EP18209672 pending, a patent EP20150702464 pending and a patent Software (VMscope digital pathology) pending. MU reports personal fees and non-financial support from Abbvie, personal fees and non-financial support from Amgen GmbH, personal fees and non-financial support from AstraZeneca, personal fees from Bristol Myers Squibb (BMS), personal fees and non-financial support from Celgene GmbH, personal fees and non-financial support from Daiji Sankyo, personal fees and non-financial support from Eisai GmbH, personal fees from Lilly Deutschland, personal fees and non-financial support from Lilly Int., personal fees and non-financial support from MSD Merck, personal fees and non-financial support from Mundipharma, personal fees and non-financial support from Myriad Genetics, personal fees and non-financial support from Odonate, personal fees and non-financial support from Pfizer GmbH, personal fees from PUMA Biotechnology, personal fees and non-financial support from Roche Pharma AG, personal fees and non-financial support from Sanofi Aventis Deutschland GmbH, personal fees and non-financial support from TEVA Pharmaceuticals Ind Ltd, personal fees and non-financial support from Novartis, personal fees from Pierre Fabre, personal fees and non-financial support from Clovis Oncology, outside the submitted work. SB reports other from NantOmics, LLC, during the conduct of the study; other from NantOmics, LLC, outside the submitted work; and Employee of NantOmics, LLC with equity interest. AS reports grants from Celgene, grants from Roche, grants from AbbVie, grants from Molecular Partner, personal fees from Roche, personal fees from AstraZeneca, personal fees from Celgene, personal fees from Roche, personal fees from Celgene, personal fees from Pfizer, personal fees from AstraZeneca, personal fees from Novartis, personal fees from MSD, personal fees from Tesaro, personal fees from Lilly, other from Roche, outside the submitted work. CJ reports personal fees from Roche, personal fees from Celgene, personal fees from Amgen, during the conduct of the study. TL reports non-financial support from Pharma Mar, non-financial support from Daiichi Sankyo, personal fees and non-financial support from MSD, personal fees from Amgen, personal fees and non-financial support from Pfizer, personal fees from Novartis, personal fees from Teva, personal fees from Tesaro, personal fees and non-financial support from Roche, personal fees and non-financial support from Clovis, non-financial support from Celgene, outside the submitted work. WDS reports grants from German Breast Group, during the conduct of the study; personal fees from AstraZeneca, outside the submitted work. VM reports grants and personal fees from Roche, during the conduct of the study; personal fees from Amgen, Astra Zeneca, Daiichi-Sankyo, Eisai, Pfizer, MSD, Novartis, Roche, Teva, Seattle Genetics and consultancy honoraria from Genomic Health, Hexal, Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi-Sankyo, Eisai, Lilly, Tesaro, Nektar, personal fees from Genomic Health, Hexal, Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi-Sankyo, Eisai, Lilly, Tesaro and Nektar, other from I Novartis, Roche, Seattle Genetics, Genentech, outside the submitted work. PSS is the CEO of NantOmics and reports non-financial support from NantOmics, outside the submitted work. MvM reports honoraria from Roche, Amgen, Genomic Health, Astra Zeneca, as well as travel support from Lilly and Novartis. PAF reports grants from Novartis, grants from BioNTech, personal fees from Novartis, personal fees from Roche, personal fees from Pfizer, personal fees from Celgene, personal fees from Daiichi-Sankyo, personal fees from AstraZeneca, personal fees from Merck Sharp & Dohme, personal fees from Eisai, personal fees from Puma, grants from Cepheid, personal fees from Lilly, personal fees from Seattle Genetics, during the conduct of the study. SR is the Chief Scientific Officer for NantOmics and reports non-financial support from NantOmics, outside the submitted work. SL reports grants and other from Celgene, grants and other from Roche, during the conduct of the study; grants and other from Abbvie, grants and other from Amgen, grants and other from AstraZeneca, grants and other from Novartis, grants and other from Pfizer, other from Seattle Genetics, other from PriME/Medscape, personal fees from Chugai, grants from Teva, grants from Vifor, grants and other from Daiichi-Sankyo, other from Lilly, other from Samsung, other from Eirgenix, other from BMS, other from Puma, other from MSD, grants from Immunomedics, outside the submitted work. In addition, SL has a patent EP14153692.0 pending. All other authors have declared no conflicts of interest., (Copyright © 2021 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Large scale, robust, and accurate whole transcriptome profiling from clinical formalin-fixed paraffin-embedded samples.
- Author
-
Newton Y, Sedgewick AJ, Cisneros L, Golovato J, Johnson M, Szeto CW, Rabizadeh S, Sanborn JZ, Benz SC, and Vaske C
- Subjects
- Computational Biology, Databases, Factual, Humans, Paraffin Embedding, Reproducibility of Results, Sequence Analysis, RNA, Tissue Fixation methods, Gene Expression Profiling methods, High-Throughput Nucleotide Sequencing methods
- Abstract
Transcriptome profiling can provide information of great value in clinical decision-making, yet RNA from readily available formalin-fixed paraffin-embedded (FFPE) tissue is often too degraded for quality sequencing. To assess the clinical utility of FFPE-derived RNA, we performed ribo-deplete RNA extractions on > 3200 FFPE slide samples; 25 of these had direct FFPE vs. fresh frozen (FF) replicates, 57 were sequenced in 2 different labs, 87 underwent multiple library analyses, and 16 had direct microdissected vs. macrodissected replicates. Poly-A versus ribo-depletion RNA extraction methods were compared using transcriptomes of TCGA cohort and 3116 FFPE samples. Compared to FF, FFPE transcripts coding for nuclear/cytoplasmic proteins involved in DNA packaging, replication, and protein synthesis were detected at lower rates and zinc finger family transcripts were of poorer quality. The greatest difference in extraction methods was in histone transcripts which typically lack poly-A tails. Encouragingly, the overall sequencing success rate was 81%. Exome coverage was highly concordant in direct FFPE and FF replicates, with 98% agreement in coding exon coverage and a median correlation of whole transcriptome profiles of 0.95. We provide strong rationale for clinical use of FFPE-derived RNA based on the robustness, reproducibility, and consistency of whole transcriptome profiling.
- Published
- 2020
- Full Text
- View/download PDF
14. A risk-associated Active transcriptome phenotype expressed by histologically normal human breast tissue and linked to a pro-tumorigenic adipocyte population.
- Author
-
Kang T, Yau C, Wong CK, Sanborn JZ, Newton Y, Vaske C, Benz SC, Krings G, Camarda R, Henry JE, Stuart J, Powell M, and Benz CC
- Subjects
- Adipocytes metabolism, Adult, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms metabolism, Carcinogenesis metabolism, Carcinogenesis pathology, Cross-Sectional Studies, Female, Humans, Middle Aged, Obesity genetics, Obesity metabolism, Obesity pathology, Phenotype, Prognosis, Transcriptome, Adipocytes pathology, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: Previous studies have identified and validated a risk-associated Active transcriptome phenotype commonly expressed in the cancer-adjacent and histologically normal epithelium, stroma, and adipose containing peritumor microenvironment of clinically established invasive breast cancers, conferring a 2.5- to 3-fold later risk of dying from recurrent breast cancer. Expression of this Active transcriptome phenotype has not yet been evaluated in normal breast tissue samples unassociated with any benign or malignant lesions; however, it has been associated with increased peritumor adipocyte composition., Methods: Detailed histologic and transcriptomic (RNAseq) analyses were performed on normal breast biopsy samples from 151 healthy, parous, non-obese (mean BMI = 29.60 ± 7.92) women, ages 27-66 who donated core breast biopsy samples to the Komen Tissue Bank, and whose average breast cancer risk estimate (Gail score) at the time of biopsy (1.27 ± 1.34) would not qualify them for endocrine prevention therapy., Results: Full genome RNA sequencing (RNAseq) identified 52% (78/151) of these normal breast samples as expressing the Active breast phenotype. While Active signature genes were found to be most variably expressed in mammary adipocytes, donors with the Active phenotype had no difference in BMI but significantly higher Gail scores (1.46 vs. 1.18; p = 0.007). Active breast samples possessed 1.6-fold more (~ 80%) adipocyte nuclei, larger cross-sectional adipocyte areas (p < 0.01), and 0.5-fold fewer stromal and epithelial cell nuclei (p < 1e-6). Infrequent low-level expression of cancer gene hotspot mutations was detected but not enriched in the Active breast samples. Active samples were enriched in gene sets associated with adipogenesis and fat metabolism (FDR q ≤ 10%), higher signature scores for cAMP-dependent lipolysis known to drive breast cancer progression, white adipose tissue browning (Wilcoxon p < 0.01), and genes associated with adipocyte activation (leptin, adiponectin) and remodeling (CAV1, BNIP3), adipokine growth factors (IGF-1, FGF2), and pro-inflammatory fat signaling (IKBKG, CCL13)., Conclusions: The risk-associated Active transcriptome phenotype first identified in cancer-adjacent breast tissues also occurs commonly in healthy women without breast disease who do not qualify for breast cancer chemoprevention, and independently of breast expressed cancer-associated mutations. The risk-associated Active phenotype appears driven by a pro-tumorigenic adipocyte microenvironment that can predate breast cancer development.
- Published
- 2020
- Full Text
- View/download PDF
15. Transcriptomic silencing as a potential mechanism of treatment resistance.
- Author
-
Adashek JJ, Kato S, Parulkar R, Szeto CW, Sanborn JZ, Vaske CJ, Benz SC, Reddy SK, and Kurzrock R
- Subjects
- Female, High-Throughput Nucleotide Sequencing, Humans, Male, Middle Aged, Gene Silencing, Germ-Line Mutation, Neoplasms genetics, Neoplasms metabolism, Neoplasms therapy, Polymorphism, Single Nucleotide, Transcriptome
- Abstract
Next-generation sequencing (NGS) has not revealed all the mechanisms underlying resistance to genomically matched drugs. Here, we performed in 1417 tumors whole-exome tumor (somatic)/normal (germline) NGS and whole-transcriptome sequencing, the latter focusing on a clinically oriented 50-gene panel in order to examine transcriptomic silencing of putative driver alterations. In this large-scale study, approximately 13% of the somatic single nucleotide variants (SNVs) were unexpectedly not expressed as RNA; 23% of patients had ≥1 nonexpressed SNV. SNV-bearing genes consistently transcribed were TP53, PIK3CA, and KRAS; those with lower transcription rates were ALK, CSF1R, ERBB4, FLT3, GNAS, HNF1A, KDR, PDGFRA, RET, and SMO. We also determined the frequency of tumor mutations being germline, rather than somatic, in these and an additional 462 tumors with tumor/normal exomes; 33.8% of germline SNVs within the gene panel were rare (not found after filtering through variant information domains) and at risk of being falsely reported as somatic. Both the frequency of silenced variant transcription and the risk of falsely identifying germline mutations as somatic/tumor related are important phenomena. Therefore, transcriptomics is a critical adjunct to genomics when interrogating patient tumors for actionable alterations, because, without expression of the target aberrations, there will likely be therapeutic resistance.
- Published
- 2020
- Full Text
- View/download PDF
16. Prediction of Benefit from Checkpoint Inhibitors in Mismatch Repair Deficient Metastatic Colorectal Cancer: Role of Tumor Infiltrating Lymphocytes.
- Author
-
Loupakis F, Depetris I, Biason P, Intini R, Prete AA, Leone F, Lombardi P, Filippi R, Spallanzani A, Cascinu S, Bonetti LR, Maddalena G, Valeri N, Sottoriva A, Zapata L, Salmaso R, Munari G, Rugge M, Dei Tos AP, Golovato J, Sanborn JZ, Nguyen A, Schirripa M, Zagonel V, Lonardi S, and Fassan M
- Subjects
- Biomarkers, Tumor genetics, DNA Mismatch Repair genetics, Humans, Microsatellite Instability, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Lymphocytes, Tumor-Infiltrating
- Abstract
Background: Immunotherapy with immune checkpoint inhibitors (ICIs) is highly effective in microsatellite instability-high (MSI-H) metastatic colorectal cancer (mCRC); however, specific predictive biomarkers are lacking., Patients and Methods: Data and samples from 85 patients with MSI-H mCRC treated with ICIs were gathered. Tumor infiltrating lymphocytes (TILs) and tumor mutational burden (TMB) were analyzed in an exploratory cohort of "super" responders and "clearly" refractory patients; TILs were then evaluated in the whole cohort of patients. Primary objectives were the correlation between the number of TILs and TMB and their role as biomarkers of ICI efficacy. Main endpoints included response rate (RR), progression-free survival (PFS), and overall survival (OS)., Results: In the exploratory cohort, an increasing number of TILs correlated to higher TMB (Pearson's test, p = .0429). In the whole cohort, median number of TILs was 3.6 in responders compared with 1.8 in nonresponders (Mann-Whitney test, p = .0448). RR was 70.6% in patients with high number of TILs (TILs-H) compared with 42.9% in patients with low number of TILs (odds ratio = 3.20, p = .0291). Survival outcomes differed significantly in favor of TILs-H (PFS: hazard ratio [HR] = 0.42, p = .0278; OS: HR = 0.41, p = .0463)., Conclusion: A significant correlation between higher TMB and increased number of TILs was shown. A significantly higher activity and better PFS and OS with ICI in MSI-H mCRC were reported in cases with high number of TILs, thus supporting further studies of TIL count as predictive biomarker of ICI efficacy., Implications for Practice: Microsatellite instability is the result of mismatch repair protein deficiency, caused by germline mutations or somatic modifications in mismatch repair genes. In metastatic colorectal cancer (mCRC), immunotherapy (with immune checkpoint inhibitors [ICIs]) demonstrated remarkable clinical benefit in microsatellite instability-high (MSI-H) patients. ICI primary resistance has been observed in approximately 25% of patients with MSI-H mCRC, underlining the need for predictive biomarkers. In this study, tumor mutational burden (TMB) and tumor infiltrating lymphocyte (TIL) analyses were performed in an exploratory cohort of patients with MSI-H mCRC treated with ICIs, demonstrating a significant correlation between higher TMB and increased number of TILs. Results also demonstrated a significant correlation between high number of TILs and clinical responses and survival benefit in a large data set of patients with MSI-H mCRC treated with ICI. TMB and TILs could represent predictive biomarkers of ICI efficacy in MSI-H mCRC and should be incorporated in future trials testing checkpoint inhibitors in colorectal cancer., (© AlphaMed Press 2020.)
- Published
- 2020
- Full Text
- View/download PDF
17. Multiregion exome sequencing of ovarian immature teratomas reveals 2N near-diploid genomes, paucity of somatic mutations, and extensive allelic imbalances shared across mature, immature, and disseminated components.
- Author
-
Heskett MB, Sanborn JZ, Boniface C, Goode B, Chapman J, Garg K, Rabban JT, Zaloudek C, Benz SC, Spellman PT, Solomon DA, and Cho RJ
- Subjects
- Adolescent, Adult, Alleles, Child, Diploidy, Female, Humans, Ovarian Neoplasms pathology, Ovarian Neoplasms surgery, Teratoma pathology, Teratoma surgery, Exome Sequencing, Young Adult, Allelic Imbalance, Mutation, Ovarian Neoplasms genetics, Teratoma genetics
- Abstract
Immature teratoma is a subtype of malignant germ cell tumor of the ovary that occurs most commonly in the first three decades of life, frequently with bilateral ovarian disease. Despite being the second most common malignant germ cell tumor of the ovary, little is known about its genetic underpinnings. Here we performed multiregion whole-exome sequencing to interrogate the genetic zygosity, clonal relationship, DNA copy number, and mutational status of 52 pathologically distinct tumor components from ten females with ovarian immature teratomas, with bilateral tumors present in five cases and peritoneal dissemination in seven cases. We found that ovarian immature teratomas are genetically characterized by 2N near-diploid genomes with extensive loss of heterozygosity and an absence of genes harboring recurrent somatic mutations or known oncogenic variants. All components within a single ovarian tumor (immature teratoma, mature teratoma with different histologic patterns of differentiation, and yolk sac tumor) were found to harbor an identical pattern of loss of heterozygosity across the genome, indicating a shared clonal origin. In contrast, the four analyzed bilateral teratomas showed distinct patterns of zygosity changes in the right versus left sided tumors, indicating independent clonal origins. All disseminated teratoma components within the peritoneum (including gliomatosis peritonei) shared a clonal pattern of loss of heterozygosity with either the right or left primary ovarian tumor. The observed genomic loss of heterozygosity patterns indicate that diverse meiotic errors contribute to the formation of ovarian immature teratomas, with 11 out of the 15 genetically distinct clones determined to result from nondisjunction errors during meiosis I or II. Overall, these findings suggest that copy-neutral loss of heterozygosity resulting from meiotic abnormalities may be sufficient to generate ovarian immature teratomas from germ cells.
- Published
- 2020
- Full Text
- View/download PDF
18. Efficient Tumor Clearance and Diversified Immunity through Neoepitope Vaccines and Combinatorial Immunotherapy.
- Author
-
Lee KL, Benz SC, Hicks KC, Nguyen A, Gameiro SR, Palena C, Sanborn JZ, Su Z, Ordentlich P, Rohlin L, Lee JH, Rabizadeh S, Soon-Shiong P, Niazi K, Schlom J, and Hamilton DH
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, Cell Line, Tumor, Combined Modality Therapy, Disease Models, Animal, Female, Gene Expression Profiling, Humans, Immunomodulation, Immunotherapy methods, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Mice, Neoplasms genetics, Neoplasms pathology, Neoplasms therapy, Treatment Outcome, Tumor Burden, Vaccination, Antigens, Neoplasm immunology, Cancer Vaccines immunology, Epitopes immunology, Neoplasms immunology
- Abstract
Progressive tumor growth is associated with deficits in the immunity generated against tumor antigens. Vaccines targeting tumor neoepitopes have the potential to address qualitative defects; however, additional mechanisms of immune failure may underlie tumor progression. In such cases, patients would benefit from additional immune-oncology agents targeting potential mechanisms of immune failure. This study explores the identification of neoepitopes in the MC38 colon carcinoma model by comparison of tumor to normal DNA and tumor RNA sequencing technology, as well as neoepitope delivery by both peptide- and adenovirus-based vaccination strategies. To improve antitumor efficacies, we combined the vaccine with a group of rationally selected immune-oncology agents. We utilized an IL15 superagonist to enhance the development of antigen-specific immunity initiated by the neoepitope vaccine, PD-L1 blockade to reduce tumor immunosuppression, and a tumor-targeted IL12 molecule to facilitate T-cell function within the tumor microenvironment. Analysis of tumor-infiltrating leukocytes demonstrated this multifaceted treatment regimen was required to promote the influx of CD8
+ T cells and enhance the expression of transcripts relating to T-cell activation/effector function. Tumor-targeted IL12 resulted in a marked increase in clonality of T-cell repertoire infiltrating the tumor, which when sculpted with the addition of either a peptide or adenoviral neoepitope vaccine promoted efficient tumor clearance. In addition, the neoepitope vaccine induced the spread of immunity to neoepitopes expressed by the tumor but not contained within the vaccine. These results demonstrate the importance of combining neoepitope-targeting vaccines with a multifaceted treatment regimen to generate effective antitumor immunity., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
19. Cell of origin and mutation pattern define three clinically distinct classes of sebaceous carcinoma.
- Author
-
North JP, Golovato J, Vaske CJ, Sanborn JZ, Nguyen A, Wu W, Goode B, Stevers M, McMullen K, Perez White BE, Collisson EA, Bloomer M, Solomon DA, Benz SC, and Cho RJ
- Subjects
- Carcinoma, Squamous Cell classification, Carcinoma, Squamous Cell diagnosis, Carcinoma, Squamous Cell pathology, DNA Mutational Analysis, Diagnosis, Differential, Exome, Eye Neoplasms classification, Eye Neoplasms diagnosis, Eye Neoplasms pathology, Humans, Keratinocytes metabolism, Keratinocytes pathology, Keratinocytes radiation effects, Microsatellite Repeats, Sebaceous Gland Neoplasms classification, Sebaceous Gland Neoplasms diagnosis, Sebaceous Gland Neoplasms pathology, Skin Neoplasms classification, Skin Neoplasms diagnosis, Skin Neoplasms etiology, Terminology as Topic, Transcriptome, Ultraviolet Rays adverse effects, Exome Sequencing, Carcinoma, Squamous Cell genetics, Eye Neoplasms genetics, Microsatellite Instability, Mutation, Sebaceous Gland Neoplasms genetics, Skin Neoplasms genetics
- Abstract
Sebaceous carcinomas (SeC) are cutaneous malignancies that, in rare cases, metastasize and prove fatal. Here we report whole-exome sequencing on 32 SeC, revealing distinct mutational classes that explain both cancer ontogeny and clinical course. A UV-damage signature predominates in 10/32 samples, while nine show microsatellite instability (MSI) profiles. UV-damage SeC exhibited poorly differentiated, infiltrative histopathology compared to MSI signature SeC (p = 0.003), features previously associated with dissemination. Moreover, UV-damage SeC transcriptomes and anatomic distribution closely resemble those of cutaneous squamous cell carcinomas (SCC), implicating sun-exposed keratinocytes as a cell of origin. Like SCC, this UV-damage subclass harbors a high somatic mutation burden with >50 mutations per Mb, predicting immunotherapeutic response. In contrast, ocular SeC acquires far fewer mutations without a dominant signature, but show frequent truncations in the ZNF750 epidermal differentiation regulator. Our data exemplify how different mutational processes convergently drive histopathologically related but clinically distinct cancers.
- Published
- 2018
- Full Text
- View/download PDF
20. Comprehensive genomic transcriptomic tumor-normal gene panel analysis for enhanced precision in patients with lung cancer.
- Author
-
Rabizadeh S, Garner C, Sanborn JZ, Benz SC, Reddy S, and Soon-Shiong P
- Abstract
A CMS approved test for lung cancer is based on tumor-only analysis of a targeted 35 gene panel, specifically excluding the use of the patient's normal germline tissue. However, this tumor-only approach increases the risk of mistakenly identifying germline single nucleotide polymorphisms (SNPs) as somatically-derived cancer driver mutations (false positives). 621 patients with 30 different cancer types, including lung cancer, were studied to compare the precision of tumor somatic variant calling in 35 genes using tumor-only DNA sequencing versus tumor-normal DNA plus RNA sequencing. When sequencing of lung cancer was performed using tumor genomes alone without normal germline controls, 94% of variants identified were SNPs and thus false positives. Filtering for common SNPs still resulted in as high as 48% false positive variant calling. With tumor-only sequencing, 29% of lung cancer patients had a false positive variant call in at least one of twelve genes with directly targetable drugs. RNA analysis showed 18% of true somatic variants were not expressed. Thus, sequencing and analysis of both normal germline and tumor genomes is necessary for accurate identification of molecular targets. Treatment decisions based on tumor-only analysis may result in the administration of ineffective therapies while also increasing the risk of negative drug-related side effects., Competing Interests: CONFLICTS OF INTEREST Authors are affiliated with NantOmics, LLC or NantHealth, Inc as listed above.
- Published
- 2018
- Full Text
- View/download PDF
21. Multiple Hereditary Infundibulocystic Basal Cell Carcinoma Syndrome Associated With a Germline SUFU Mutation.
- Author
-
Schulman JM, Oh DH, Sanborn JZ, Pincus L, McCalmont TH, and Cho RJ
- Subjects
- DNA Mutational Analysis, Exome genetics, Humans, Middle Aged, Skin pathology, Skin Neoplasms pathology, Syndrome, Carcinoma, Basal Cell genetics, Germ-Line Mutation genetics, Hamartoma Syndrome, Multiple genetics, Repressor Proteins genetics, Skin Neoplasms genetics
- Abstract
Importance: Multiple hereditary infundibulocystic basal cell carcinoma syndrome (MHIBCC) is a rare genodermatosis in which numerous indolent, well-differentiated basal cell carcinomas develop primarily on the face and genitals, without other features characteristic of basal cell nevus syndrome. The cause is unknown. The purpose of the study was to identify a genetic basis for the syndrome and a mechanism by which the associated tumors develop., Observations: Whole-exome sequencing of 5 tumors and a normal buccal mucosal sample from a patient with MHIBCC was performed. A conserved splice-site mutation in 1 copy of the suppressor of fused gene (SUFU) was identified in all tumor and normal tissue samples. Additional distinct deletions of the trans SUFU allele were identified in all tumor samples, none of which were present in the normal sample., Conclusions and Relevance: A germline SUFU mutation was present in a patient with MHIBCC, and additional acquired SUFU mutations underlie the development of infundibulocystic basal cell carcinomas. The downstream location of the SUFU gene within the sonic hedgehog pathway may explain why its loss is associated with relatively well-differentiated tumors and suggests that MHIBCC will not respond to therapeutic strategies, such as smoothened inhibitors, that target upstream components of this pathway.
- Published
- 2016
- Full Text
- View/download PDF
22. Exome sequencing of desmoplastic melanoma identifies recurrent NFKBIE promoter mutations and diverse activating mutations in the MAPK pathway.
- Author
-
Shain AH, Garrido M, Botton T, Talevich E, Yeh I, Sanborn JZ, Chung J, Wang NJ, Kakavand H, Mann GJ, Thompson JF, Wiesner T, Roy R, Olshen AB, Gagnon A, Gray JW, Huh N, Hur JS, Busam KJ, Scolyer RA, Cho RJ, Murali R, and Bastian BC
- Subjects
- Humans, Melanoma enzymology, Melanoma pathology, Exome, I-kappa B Proteins genetics, MAP Kinase Signaling System, Melanoma genetics, Mutation, Promoter Regions, Genetic, Proto-Oncogene Proteins genetics
- Abstract
Desmoplastic melanoma is an uncommon variant of melanoma with sarcomatous histology, distinct clinical behavior and unknown pathogenesis. We performed low-coverage genome and high-coverage exome sequencing of 20 desmoplastic melanomas, followed by targeted sequencing of 293 genes in a validation cohort of 42 cases. A high mutation burden (median of 62 mutations/Mb) ranked desmoplastic melanoma among the most highly mutated cancers. Mutation patterns strongly implicate ultraviolet radiation as the dominant mutagen, indicating a superficially located cell of origin. Newly identified alterations included recurrent promoter mutations of NFKBIE, encoding NF-κB inhibitor ɛ (IκBɛ), in 14.5% of samples. Common oncogenic mutations in melanomas, in particular in BRAF (encoding p.Val600Glu) and NRAS (encoding p.Gln61Lys or p.Gln61Arg), were absent. Instead, other genetic alterations known to activate the MAPK and PI3K signaling cascades were identified in 73% of samples, affecting NF1, CBL, ERBB2, MAP2K1, MAP3K1, BRAF, EGFR, PTPN11, MET, RAC1, SOS2, NRAS and PIK3CA, some of which are candidates for targeted therapies.
- Published
- 2015
- Full Text
- View/download PDF
23. Phylogenetic analyses of melanoma reveal complex patterns of metastatic dissemination.
- Author
-
Sanborn JZ, Chung J, Purdom E, Wang NJ, Kakavand H, Wilmott JS, Butler T, Thompson JF, Mann GJ, Haydu LE, Saw RP, Busam KJ, Lo RS, Collisson EA, Hur JS, Spellman PT, Cleaver JE, Gray JW, Huh N, Murali R, Scolyer RA, Bastian BC, and Cho RJ
- Subjects
- Humans, Melanoma genetics, Neoplasm Metastasis, Melanoma pathology, Phylogeny
- Abstract
Melanoma is difficult to treat once it becomes metastatic. However, the precise ancestral relationship between primary tumors and their metastases is not well understood. We performed whole-exome sequencing of primary melanomas and multiple matched metastases from eight patients to elucidate their phylogenetic relationships. In six of eight patients, we found that genetically distinct cell populations in the primary tumor metastasized in parallel to different anatomic sites, rather than sequentially from one site to the next. In five of these six patients, the metastasizing cells had themselves arisen from a common parental subpopulation in the primary, indicating that the ability to establish metastases is a late-evolving trait. Interestingly, we discovered that individual metastases were sometimes founded by multiple cell populations of the primary that were genetically distinct. Such establishment of metastases by multiple tumor subpopulations could help explain why identical resistance variants are identified in different sites after initial response to systemic therapy. One primary tumor harbored two subclones with different oncogenic mutations in CTNNB1, which were both propagated to the same metastasis, raising the possibility that activation of wingless-type mouse mammary tumor virus integration site (WNT) signaling may be involved, as has been suggested by experimental models.
- Published
- 2015
- Full Text
- View/download PDF
24. Glioblastoma adaptation traced through decline of an IDH1 clonal driver and macro-evolution of a double-minute chromosome.
- Author
-
Favero F, McGranahan N, Salm M, Birkbak NJ, Sanborn JZ, Benz SC, Becq J, Peden JF, Kingsbury Z, Grocok RJ, Humphray S, Bentley D, Spencer-Dene B, Gutteridge A, Brada M, Roger S, Dietrich PY, Forshew T, Gerlinger M, Rowan A, Stamp G, Eklund AC, Szallasi Z, and Swanton C
- Subjects
- Adult, Antineoplastic Agents, Alkylating therapeutic use, Brain Neoplasms enzymology, Brain Neoplasms pathology, Brain Neoplasms therapy, Chemotherapy, Adjuvant, Cyclin-Dependent Kinase 4 genetics, Dacarbazine analogs & derivatives, Dacarbazine therapeutic use, Disease Progression, Fatal Outcome, Female, Genetic Association Studies, Genetic Predisposition to Disease, Glioblastoma enzymology, Glioblastoma pathology, Glioblastoma therapy, Humans, Imatinib Mesylate therapeutic use, Neoplasm Grading, Neoplasm Recurrence, Local, Neurosurgical Procedures, Phenotype, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-kit genetics, Receptor, Platelet-Derived Growth Factor alpha genetics, Temozolomide, Time Factors, Treatment Outcome, Brain Neoplasms genetics, Chromosomes, Human, Glioblastoma genetics, Isocitrate Dehydrogenase genetics, Mutation
- Abstract
Background: Glioblastoma (GBM) is the most common malignant brain cancer occurring in adults, and is associated with dismal outcome and few therapeutic options. GBM has been shown to predominantly disrupt three core pathways through somatic aberrations, rendering it ideal for precision medicine approaches., Methods: We describe a 35-year-old female patient with recurrent GBM following surgical removal of the primary tumour, adjuvant treatment with temozolomide and a 3-year disease-free period. Rapid whole-genome sequencing (WGS) of three separate tumour regions at recurrence was carried out and interpreted relative to WGS of two regions of the primary tumour., Results: We found extensive mutational and copy-number heterogeneity within the primary tumour. We identified a TP53 mutation and two focal amplifications involving PDGFRA, KIT and CDK4, on chromosomes 4 and 12. A clonal IDH1 R132H mutation in the primary, a known GBM driver event, was detectable at only very low frequency in the recurrent tumour. After sub-clonal diversification, evidence was found for a whole-genome doubling event and a translocation between the amplified regions of PDGFRA, KIT and CDK4, encoded within a double-minute chromosome also incorporating miR26a-2. The WGS analysis uncovered progressive evolution of the double-minute chromosome converging on the KIT/PDGFRA/PI3K/mTOR axis, superseding the IDH1 mutation in dominance in a mutually exclusive manner at recurrence, consequently the patient was treated with imatinib. Despite rapid sequencing and cancer genome-guided therapy against amplified oncogenes, the disease progressed, and the patient died shortly after., Conclusion: This case sheds light on the dynamic evolution of a GBM tumour, defining the origins of the lethal sub-clone, the macro-evolutionary genomic events dominating the disease at recurrence and the loss of a clonal driver. Even in the era of rapid WGS analysis, cases such as this illustrate the significant hurdles for precision medicine success., (© The Author 2015. Published by Oxford University Press on behalf of the European Society for Medical Oncology.)
- Published
- 2015
- Full Text
- View/download PDF
25. Transcription restores DNA repair to heterochromatin, determining regional mutation rates in cancer genomes.
- Author
-
Zheng CL, Wang NJ, Chung J, Moslehi H, Sanborn JZ, Hur JS, Collisson EA, Vemula SS, Naujokas A, Chiotti KE, Cheng JB, Fassihi H, Blumberg AJ, Bailey CV, Fudem GM, Mihm FG, Cunningham BB, Neuhaus IM, Liao W, Oh DH, Cleaver JE, LeBoit PE, Costello JF, Lehmann AR, Gray JW, Spellman PT, Arron ST, Huh N, Purdom E, and Cho RJ
- Subjects
- DNA Packaging genetics, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Gene Expression Regulation, Neoplastic, Germ Cells metabolism, Humans, Proto-Oncogene Proteins genetics, Carcinoma, Squamous Cell genetics, DNA Repair genetics, Genome, Human genetics, Heterochromatin genetics, Mutation Rate, Skin Neoplasms genetics, Transcription, Genetic
- Abstract
Somatic mutations in cancer are more frequent in heterochromatic and late-replicating regions of the genome. We report that regional disparities in mutation density are virtually abolished within transcriptionally silent genomic regions of cutaneous squamous cell carcinomas (cSCCs) arising in an XPC(-/-) background. XPC(-/-) cells lack global genome nucleotide excision repair (GG-NER), thus establishing differential access of DNA repair machinery within chromatin-rich regions of the genome as the primary cause for the regional disparity. Strikingly, we find that increasing levels of transcription reduce mutation prevalence on both strands of gene bodies embedded within H3K9me3-dense regions, and only to those levels observed in H3K9me3-sparse regions, also in an XPC-dependent manner. Therefore, transcription appears to reduce mutation prevalence specifically by relieving the constraints imposed by chromatin structure on DNA repair. We model this relationship among transcription, chromatin state, and DNA repair, revealing a new, personalized determinant of cancer risk., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
26. The somatic genomic landscape of glioblastoma.
- Author
-
Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, Beroukhim R, Bernard B, Wu CJ, Genovese G, Shmulevich I, Barnholtz-Sloan J, Zou L, Vegesna R, Shukla SA, Ciriello G, Yung WK, Zhang W, Sougnez C, Mikkelsen T, Aldape K, Bigner DD, Van Meir EG, Prados M, Sloan A, Black KL, Eschbacher J, Finocchiaro G, Friedman W, Andrews DW, Guha A, Iacocca M, O'Neill BP, Foltz G, Myers J, Weisenberger DJ, Penny R, Kucherlapati R, Perou CM, Hayes DN, Gibbs R, Marra M, Mills GB, Lander E, Spellman P, Wilson R, Sander C, Weinstein J, Meyerson M, Gabriel S, Laird PW, Haussler D, Getz G, and Chin L
- Subjects
- Brain Neoplasms metabolism, Female, Gene Expression Profiling, Gene Regulatory Networks, Glioblastoma metabolism, Humans, Male, Mutation, Proteome analysis, Signal Transduction, Brain Neoplasms genetics, Glioblastoma genetics
- Abstract
We describe the landscape of somatic genomic alterations based on multidimensional and comprehensive characterization of more than 500 glioblastoma tumors (GBMs). We identify several novel mutated genes as well as complex rearrangements of signature receptors, including EGFR and PDGFRA. TERT promoter mutations are shown to correlate with elevated mRNA expression, supporting a role in telomerase reactivation. Correlative analyses confirm that the survival advantage of the proneural subtype is conferred by the G-CIMP phenotype, and MGMT DNA methylation may be a predictive biomarker for treatment response only in classical subtype GBM. Integrative analysis of genomic and proteomic profiles challenges the notion of therapeutic inhibition of a pathway as an alternative to inhibition of the target itself. These data will facilitate the discovery of therapeutic and diagnostic target candidates, the validation of research and clinical observations and the generation of unanticipated hypotheses that can advance our molecular understanding of this lethal cancer., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
27. Double minute chromosomes in glioblastoma multiforme are revealed by precise reconstruction of oncogenic amplicons.
- Author
-
Sanborn JZ, Salama SR, Grifford M, Brennan CW, Mikkelsen T, Jhanwar S, Katzman S, Chin L, and Haussler D
- Subjects
- Brain Neoplasms, Case-Control Studies, Chromosome Mapping, Cohort Studies, Gene Dosage, Humans, In Situ Hybridization, Fluorescence, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Sequence Analysis, DNA, Biomarkers, Tumor genetics, Brain metabolism, Chromosome Aberrations, Chromosomes, Human genetics, Genome, Human, Glioblastoma genetics
- Abstract
DNA sequencing offers a powerful tool in oncology based on the precise definition of structural rearrangements and copy number in tumor genomes. Here, we describe the development of methods to compute copy number and detect structural variants to locally reconstruct highly rearranged regions of the tumor genome with high precision from standard, short-read, paired-end sequencing datasets. We find that circular assemblies are the most parsimonious explanation for a set of highly amplified tumor regions in a subset of glioblastoma multiforme samples sequenced by The Cancer Genome Atlas (TCGA) consortium, revealing evidence for double minute chromosomes in these tumors. Further, we find that some samples harbor multiple circular amplicons and, in some cases, further rearrangements occurred after the initial amplicon-generating event. Fluorescence in situ hybridization analysis offered an initial confirmation of the presence of double minute chromosomes. Gene content in these assemblies helps identify likely driver oncogenes for these amplicons. RNA-seq data available for one double minute chromosome offered additional support for our local tumor genome assemblies, and identified the birth of a novel exon made possible through rearranged sequences present in the double minute chromosomes. Our method was also useful for analysis of a larger set of glioblastoma multiforme tumors for which exome sequencing data are available, finding evidence for oncogenic double minute chromosomes in more than 20% of clinical specimens examined, a frequency consistent with previous estimates.
- Published
- 2013
- Full Text
- View/download PDF
28. The UCSC Interaction Browser: multidimensional data views in pathway context.
- Author
-
Wong CK, Vaske CJ, Ng S, Sanborn JZ, Benz SC, Haussler D, and Stuart JM
- Subjects
- Colorectal Neoplasms genetics, Computer Graphics, DNA Copy Number Variations, DNA Methylation, Gene Expression, Genomics, Humans, Internet, Mutation, Protein Interaction Mapping, Gene Regulatory Networks, Software
- Abstract
High-throughput data sets such as genome-wide protein-protein interactions, protein-DNA interactions and gene expression data have been published for several model systems, especially for human cancer samples. The University of California, Santa Cruz (UCSC) Interaction Browser (http://sysbio.soe.ucsc.edu/nets) is an online tool for biologists to view high-throughput data sets simultaneously for the analysis of functional relationships between biological entities. Users can access several public interaction networks and functional genomics data sets through the portal as well as upload their own networks and data sets for analysis. Users can navigate through correlative relationships for focused sets of genes belonging to biological pathways using a standard web browser. Using a new visual modality called the CircleMap, multiple 'omics' data sets can be viewed simultaneously within the context of curated, predicted, directed and undirected regulatory interactions. The Interaction Browser provides an integrative viewing of biological networks based on the consensus of many observations about genes and their products, which may provide new insights about normal and disease processes not obvious from any isolated data set.
- Published
- 2013
- Full Text
- View/download PDF
29. The UCSC Cancer Genomics Browser: update 2011.
- Author
-
Sanborn JZ, Benz SC, Craft B, Szeto C, Kober KM, Meyer L, Vaske CJ, Goldman M, Smith KE, Kuhn RM, Karolchik D, Kent WJ, Stuart JM, Haussler D, and Zhu J
- Subjects
- DNA Copy Number Variations, Gene Expression, Genome, Human, Humans, Internet, Neoplasms metabolism, Neoplasms pathology, Software, Databases, Genetic, Genomics, Neoplasms genetics
- Abstract
The UCSC Cancer Genomics Browser (https://genome-cancer.ucsc.edu) comprises a suite of web-based tools to integrate, visualize and analyze cancer genomics and clinical data. The browser displays whole-genome views of genome-wide experimental measurements for multiple samples alongside their associated clinical information. Multiple data sets can be viewed simultaneously as coordinated 'heatmap tracks' to compare across studies or different data modalities. Users can order, filter, aggregate, classify and display data interactively based on any given feature set including clinical features, annotated biological pathways and user-contributed collections of genes. Integrated standard statistical tools provide dynamic quantitative analysis within all available data sets. The browser hosts a growing body of publicly available cancer genomics data from a variety of cancer types, including data generated from the Cancer Genome Atlas project. Multiple consortiums use the browser on confidential prepublication data enabled by private installations. Many new features have been added, including the hgMicroscope tumor image viewer, hgSignature for real-time genomic signature evaluation on any browser track, and 'PARADIGM' pathway tracks to display integrative pathway activities. The browser is integrated with the UCSC Genome Browser; thus inheriting and integrating the Genome Browser's rich set of human biology and genetics data that enhances the interpretability of the cancer genomics data.
- Published
- 2011
- Full Text
- View/download PDF
30. Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM.
- Author
-
Vaske CJ, Benz SC, Sanborn JZ, Earl D, Szeto C, Zhu J, Haussler D, and Stuart JM
- Subjects
- Breast Neoplasms genetics, DNA Copy Number Variations, Female, Gene Expression Profiling methods, Glioblastoma genetics, Humans, Genomics methods, Neoplasms genetics, Software
- Abstract
Motivation: High-throughput data is providing a comprehensive view of the molecular changes in cancer tissues. New technologies allow for the simultaneous genome-wide assay of the state of genome copy number variation, gene expression, DNA methylation and epigenetics of tumor samples and cancer cell lines. Analyses of current data sets find that genetic alterations between patients can differ but often involve common pathways. It is therefore critical to identify relevant pathways involved in cancer progression and detect how they are altered in different patients., Results: We present a novel method for inferring patient-specific genetic activities incorporating curated pathway interactions among genes. A gene is modeled by a factor graph as a set of interconnected variables encoding the expression and known activity of a gene and its products, allowing the incorporation of many types of omic data as evidence. The method predicts the degree to which a pathway's activities (e.g. internal gene states, interactions or high-level 'outputs') are altered in the patient using probabilistic inference. Compared with a competing pathway activity inference approach called SPIA, our method identifies altered activities in cancer-related pathways with fewer false-positives in both a glioblastoma multiform (GBM) and a breast cancer dataset. PARADIGM identified consistent pathway-level activities for subsets of the GBM patients that are overlooked when genes are considered in isolation. Further, grouping GBM patients based on their significant pathway perturbations divides them into clinically-relevant subgroups having significantly different survival outcomes. These findings suggest that therapeutics might be chosen that target genes at critical points in the commonly perturbed pathway(s) of a group of patients., Availability: Source code available at http://sbenz.github.com/Paradigm,., Supplementary Information: Supplementary data are available at Bioinformatics online.
- Published
- 2010
- Full Text
- View/download PDF
31. The UCSC Cancer Genomics Browser.
- Author
-
Zhu J, Sanborn JZ, Benz S, Szeto C, Hsu F, Kuhn RM, Karolchik D, Archie J, Lenburg ME, Esserman LJ, Kent WJ, Haussler D, and Wang T
- Subjects
- Humans, Biomarkers, Tumor genetics, Chromosome Mapping methods, DNA, Neoplasm genetics, Gene Expression Profiling methods, Neoplasm Proteins genetics, Neoplasms genetics, Software, User-Computer Interface
- Published
- 2009
- Full Text
- View/download PDF
32. Comparative genomics search for losses of long-established genes on the human lineage.
- Author
-
Zhu J, Sanborn JZ, Diekhans M, Lowe CB, Pringle TH, and Haussler D
- Subjects
- Animals, Dogs, Genetic Variation genetics, Genomics methods, Humans, Mice, Biological Evolution, Chromosome Mapping methods, DNA Mutational Analysis methods, Evolution, Molecular, Gene Deletion, Genome, Human genetics, Pseudogenes genetics
- Abstract
Taking advantage of the complete genome sequences of several mammals, we developed a novel method to detect losses of well-established genes in the human genome through syntenic mapping of gene structures between the human, mouse, and dog genomes. Unlike most previous genomic methods for pseudogene identification, this analysis is able to differentiate losses of well-established genes from pseudogenes formed shortly after segmental duplication or generated via retrotransposition. Therefore, it enables us to find genes that were inactivated long after their birth, which were likely to have evolved nonredundant biological functions before being inactivated. The method was used to look for gene losses along the human lineage during the approximately 75 million years (My) since the common ancestor of primates and rodents (the euarchontoglire crown group). We identified 26 losses of well-established genes in the human genome that were all lost at least 50 My after their birth. Many of them were previously characterized pseudogenes in the human genome, such as GULO and UOX. Our methodology is highly effective at identifying losses of single-copy genes of ancient origin, allowing us to find a few well-known pseudogenes in the human genome missed by previous high-throughput genome-wide studies. In addition to confirming previously known gene losses, we identified 16 previously uncharacterized human pseudogenes that are definitive losses of long-established genes. Among them is ACYL3, an ancient enzyme present in archaea, bacteria, and eukaryotes, but lost approximately 6 to 8 Mya in the ancestor of humans and chimps. Although losses of well-established genes do not equate to adaptive gene losses, they are a useful proxy to use when searching for such genetic changes. This is especially true for adaptive losses that occurred more than 250,000 years ago, since any genetic evidence of the selective sweep indicative of such an event has been erased.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.