41 results on '"Katherine E. Varley"'
Search Results
2. Data from Expression of the MHC Class II Pathway in Triple-Negative Breast Cancer Tumor Cells Is Associated with a Good Prognosis and Infiltrating Lymphocytes
- Author
-
Katherine E. Varley, Albert F. LoBuglio, Richard M. Myers, Donald J. Buchsbaum, Christos Vaklavas, Todd C. Burwell, Erinn Downs-Kelly, Natalie D. Merz, Katherine L. Updike, William E. Grizzle, Dongquan Chen, Yufeng Li, and Andres Forero
- Abstract
Triple-negative breast cancer (TNBC) is a subtype with heterogeneous patient outcomes. Approximately 40% of patients experience rapid relapse, while the remaining patients have long-term disease-free survival. To determine if there are molecular differences between primary tumors that predict prognosis, we performed RNA-seq on 47 macrodissected tumors from newly diagnosed patients with TNBC (n = 47; 22 relapse, 25 no relapse; follow-up median, 8 years; range, 2–11 years). We discovered that expression of the MHC class II (MHC II) antigen presentation pathway in tumor tissue was the most significant pathway associated with progression-free survival (HR, 0.36; log-rank P = 0.0098). The association between MHC II pathway expression and good prognosis was confirmed in a public gene expression database of 199 TNBC cases (HR, 0.28; log-rank P = 4.5 × 10–8). Further analysis of immunohistochemistry, laser-capture microdissected tumors, and TNBC cell lines demonstrated that tumor cells, in addition to immune cells, aberrantly express the MHC II pathway. MHC II pathway expression was also associated with B-cell and T-cell infiltration in the tumor. Together, these data support the model that aberrant expression of the MHC II pathway in TNBC tumor cells may trigger an antitumor immune response that reduces the rate of relapse and enhances progression-free survival. Cancer Immunol Res; 4(5); 390–9. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
3. A human breast cancer-derived xenograft and organoid platform for drug discovery and precision oncology
- Author
-
Katrin P. Guillen, Maihi Fujita, Andrew J. Butterfield, Sandra D. Scherer, Matthew H. Bailey, Zhengtao Chu, Yoko S. DeRose, Ling Zhao, Emilio Cortes-Sanchez, Chieh-Hsiang Yang, Jennifer Toner, Guoying Wang, Yi Qiao, Xiaomeng Huang, Jeffery A. Greenland, Jeffery M. Vahrenkamp, David H. Lum, Rachel E. Factor, Edward W. Nelson, Cindy B. Matsen, Jane M. Poretta, Regina Rosenthal, Anna C. Beck, Saundra S. Buys, Christos Vaklavas, John H. Ward, Randy L. Jensen, Kevin B. Jones, Zheqi Li, Steffi Oesterreich, Lacey E. Dobrolecki, Satya S. Pathi, Xing Yi Woo, Kristofer C. Berrett, Mark E. Wadsworth, Jeffrey H. Chuang, Michael T. Lewis, Gabor T. Marth, Jason Gertz, Katherine E. Varley, Bryan E. Welm, and Alana L. Welm
- Subjects
Organoids ,Cancer Research ,Oncology ,Drug Discovery ,Heterografts ,Humans ,Triple Negative Breast Neoplasms ,Precision Medicine ,Xenograft Model Antitumor Assays ,United States - Abstract
Models that recapitulate the complexity of human tumors are urgently needed to develop more effective cancer therapies. We report a bank of human patient-derived xenografts (PDXs) and matched organoid cultures from tumors that represent the greatest unmet need: endocrine-resistant, treatment-refractory and metastatic breast cancers. We leverage matched PDXs and PDX-derived organoids (PDxO) for drug screening that is feasible and cost-effective with in vivo validation. Moreover, we demonstrate the feasibility of using these models for precision oncology in real time with clinical care in a case of triple-negative breast cancer (TNBC) with early metastatic recurrence. Our results uncovered a Food and Drug Administration (FDA)-approved drug with high efficacy against the models. Treatment with this therapy resulted in a complete response for the individual and a progression-free survival (PFS) period more than three times longer than their previous therapies. This work provides valuable methods and resources for functional precision medicine and drug development for human breast cancer.
- Published
- 2022
- Full Text
- View/download PDF
4. Data from TBCRC 019: A Phase II Trial of Nanoparticle Albumin-Bound Paclitaxel with or without the Anti-Death Receptor 5 Monoclonal Antibody Tigatuzumab in Patients with Triple-Negative Breast Cancer
- Author
-
Antonio C. Wolff, Richard M. Myers, William Grizzle, Paul Haluska, Jennifer F. De Los Santos, Helen Krontiras, William J. Irvin, Julie R. Nangia, Catherine H. Van Poznak, Sujata Patil, Tiffany A. Traina, Anna M. Storniolo, Rita Nanda, Hope S. Rugo, Minetta C. Liu, Nancy U. Lin, Christos Vaklavas, Yufeng Li, Vandana G. Abramson, Katherine E. Varley, and Andres Forero-Torres
- Abstract
Purpose: Tigatuzumab (TIG), an agonistic anti-DR5 antibody, triggers apoptosis in DR5+ human tumor cells without crosslinking. TIG has strong in vitro/in vivo activity against basal-like breast cancer cells enhanced by chemotherapy agents. This study evaluates activity of TIG and chemotherapy in patients with metastatic triple-negative breast cancer (TNBC).Experimental Design: Randomized 2:1 phase II trial of albumin-bound paclitaxel (nab-PAC) ± TIG in patients with TNBC stratified by prior chemotherapy. Patients received nab-PAC weekly × 3 ± TIG every other week, every 28 days. Primary objective was within-arm objective response rate (ORR). Secondary objectives were safety, progression-free survival (PFS), clinical benefit, and TIG immunogenicity. Metastatic research biopsies were required.Results: Among 64 patients (60 treated; TIG/nab-PAC n = 39 and nab-PAC n = 21), there were 3 complete remissions (CR), 8 partial remissions (PR; 1 almost CR), 11 stable diseases (SD), and 17 progressive diseases (PD) in the TIG/nab-PAC arm (ORR, 28%), and no CRs, 8 PRs, 4 SDs, and 9 PDs in the nab-PAC arm (ORR, 38%). There was a numerical increase in CRs and several patients had prolonged PFS (1,025+, 781, 672, 460, 334) in the TIG/nab-PAC arm. Grade 3 toxicities were 28% and 29%, respectively, with no grade 4–5. Exploratory analysis suggests an association of ROCK1 gene pathway activation with efficacy in the TIG/nab-PAC arm.Conclusions: ORR and PFS were similar in both. Preclinical activity of TIG in basal-like breast cancer and prolonged PFS in few patients in the combination arm support further investigation of anti-DR5 agents. ROCK pathway activation merits further evaluation. Clin Cancer Res; 21(12); 2722–9. ©2015 AACR.See related article by Paoletti et al., p. 2771
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Data from A Multigene Assay Determines Risk of Recurrence in Patients with Triple-Negative Breast Cancer
- Author
-
Katherine E. Varley, Philip S. Bernard, Kenneth M. Boucher, N. Lynn Henry, Rachel E. Factor, Katherine L. Updike, and Rachel L. Stewart
- Abstract
Supplemental Figures 1-7 include a graphical outline of the study, graphs depicting probe performance, correlations between MHCII and TIL scores, ROC statistics, and additional Kaplan Meier curves.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figure 1 from TBCRC 019: A Phase II Trial of Nanoparticle Albumin-Bound Paclitaxel with or without the Anti-Death Receptor 5 Monoclonal Antibody Tigatuzumab in Patients with Triple-Negative Breast Cancer
- Author
-
Antonio C. Wolff, Richard M. Myers, William Grizzle, Paul Haluska, Jennifer F. De Los Santos, Helen Krontiras, William J. Irvin, Julie R. Nangia, Catherine H. Van Poznak, Sujata Patil, Tiffany A. Traina, Anna M. Storniolo, Rita Nanda, Hope S. Rugo, Minetta C. Liu, Nancy U. Lin, Christos Vaklavas, Yufeng Li, Vandana G. Abramson, Katherine E. Varley, and Andres Forero-Torres
- Abstract
1. Supplemental Data: Differential of Toxicity and Eligibility Critique.; 2. Supplemental Figure 1: Gene Expression in Patients With Response to Therapy.
- Published
- 2023
- Full Text
- View/download PDF
7. Data from A Multigene Assay Determines Risk of Recurrence in Patients with Triple-Negative Breast Cancer
- Author
-
Katherine E. Varley, Philip S. Bernard, Kenneth M. Boucher, N. Lynn Henry, Rachel E. Factor, Katherine L. Updike, and Rachel L. Stewart
- Abstract
Approximately 40% of patients with stage I–III triple-negative breast cancer (TNBC) recur after standard treatment, whereas the remaining 60% experience long-term disease-free survival (DFS). There are currently no clinical tests to assess the risk of recurrence in TNBC patients. We previously determined that TNBC patients with MHC class II (MHCII) pathway expression in their tumors experienced significantly longer DFS. To translate this discovery into a clinical test, we developed an MHCII Immune Activation assay, which measures expression of 36 genes using NanoString technology. Preanalytical testing confirmed that the assay is accurate and reproducible in formalin-fixed paraffin-embedded (FFPE) tumor specimens. The assay measurements were concordant with RNA-seq, MHCII protein expression, and tumor-infiltrating lymphocyte counts. In a training set of 44 primary TNBC tumors, the MHCII Immune Activation Score was significantly associated with longer DFS (HR = 0.17; P = 0.015). In an independent validation cohort of 56 primary FFPE TNBC tumors, the Immune Activation Score was significantly associated with longer DFS (HR = 0.19; P = 0.011) independent of clinical stage. An Immune Activation Score threshold for identifying patients with very low risk of relapse in the training set provided 100% specificity in the validation cohort. The assay format enables adoption as a standardized clinical prognostic test for identifying TNBC patients with a low risk of recurrence. Correlative data support future studies to determine if the assay can identify patients in whom chemotherapy can be safely deescalated and patients likely to respond to immunotherapy.Significance:The MHCII Immune Activation assay identifies TNBC patients with a low risk of recurrence, addressing a critical need for prognostic biomarker tests that enable precision medicine for TNBC patients.
- Published
- 2023
- Full Text
- View/download PDF
8. Loss of G0/G1 switch gene 2 (G0S2) promotes disease progression and drug resistance in chronic myeloid leukaemia (CML) by disrupting glycerophospholipid metabolism
- Author
-
Mayra A. Gonzalez, Idaly M. Olivas, Alfonso E. Bencomo‐Alvarez, Andres J. Rubio, Christian Barreto‐Vargas, Jose L. Lopez, Sara K. Dang, Jonathan P. Solecki, Emily McCall, Gonzalo Astudillo, Vanessa V. Velazquez, Katherine Schenkel, Kelaiah Reffell, Mariah Perkins, Nhu Nguyen, Jehu N. Apaflo, Efren Alvidrez, James E. Young, Joshua J. Lara, Dongqing Yan, Anna Senina, Jonathan Ahmann, Katherine E. Varley, Clinton C. Mason, Christopher A. Eide, Brian J. Druker, Md Nurunnabi, Osvaldo Padilla, Sudip Bajpeyi, and Anna M. Eiring
- Subjects
Mice ,Drug Resistance, Neoplasm ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Disease Progression ,Fusion Proteins, bcr-abl ,Animals ,Humans ,Molecular Medicine ,Medicine (miscellaneous) ,Cell Cycle Proteins ,Glycerophospholipids ,Protein Kinase Inhibitors ,Genes, Switch - Abstract
Tyrosine kinase inhibitors (TKIs) targeting BCR::ABL1 have turned chronic myeloid leukaemia (CML) from a fatal disease into a manageable condition for most patients. Despite improved survival, targeting drug-resistant leukaemia stem cells (LSCs) remains a challenge for curative CML therapy. Aberrant lipid metabolism can have a large impact on membrane dynamics, cell survival and therapeutic responses in cancer. While ceramide and sphingolipid levels were previously correlated with TKI response in CML, the role of lipid metabolism in TKI resistance is not well understood. We have identified downregulation of a critical regulator of lipid metabolism, G0/G1 switch gene 2 (G0S2), in multiple scenarios of TKI resistance, including (1) BCR::ABL1 kinase-independent TKI resistance, (2) progression of CML from the chronic to the blast phase of the disease, and (3) in CML versus normal myeloid progenitors. Accordingly, CML patients with low G0S2 expression levels had a worse overall survival. G0S2 downregulation in CML was not a result of promoter hypermethylation or BCR::ABL1 kinase activity, but was rather due to transcriptional repression by MYC. Using CML cell lines, patient samples and G0s2 knockout (G0s2
- Published
- 2022
- Full Text
- View/download PDF
9. TXNIP loss expands Myc-dependent transcriptional programs by increasing Myc genomic binding
- Author
-
Tian-Yeh Lim, Blake R. Wilde, Mallory L. Thomas, Kristin E. Murphy, Jeffery M. Vahrenkamp, Megan E. Conway, Katherine E. Varley, Jason Gertz, and Donald E. Ayer
- Subjects
General Immunology and Microbiology ,General Neuroscience ,General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular Biology - Abstract
The c-Myc protooncogene places a demand on glucose uptake to drive glucose-dependent biosynthetic pathways. To meet this demand, c-Myc protein (Myc henceforth) drives the expression of glucose transporters, glycolytic enzymes, and represses the expression of thioredoxin interacting protein (TXNIP), which is a potent negative regulator of glucose uptake. A Mychigh/TXNIPlow gene signature is clinically significant as it correlates with poor clinical prognosis in triple-negative breast cancer (TNBC) but not in other subtypes of breast cancer, suggesting a functional relationship between Myc and TXNIP. To better understand how TXNIP contributes to the aggressive behavior of TNBC, we generated TXNIP null MDA-MB-231 (231:TKO) cells for our study. We show that TXNIP loss drives a transcriptional program that resembles those driven by Myc and increases global Myc genome occupancy. TXNIP loss allows Myc to invade the promoters and enhancers of target genes that are potentially relevant to cell transformation. Together, these findings suggest that TXNIP is a broad repressor of Myc genomic binding. The increase in Myc genomic binding in the 231:TKO cells expands the Myc-dependent transcriptome we identified in parental MDA-MB-231 cells. This expansion of Myc-dependent transcription following TXNIP loss occurs without an apparent increase in Myc’s intrinsic capacity to activate transcription and without increasing Myc levels. Together, our findings suggest that TXNIP loss mimics Myc overexpression, connecting Myc genomic binding and transcriptional programs to the nutrient and progrowth signals that control TXNIP expression.
- Published
- 2022
- Full Text
- View/download PDF
10. Tet2 coordinates with Foxo1 and Runx1 to balance T follicular helper cell and T helper 1 cell differentiation
- Author
-
Andrew Baessler, Camille L. Novis, Zuolian Shen, Jelena Perovanovic, Mark Wadsworth, Kendall A. Thiede, Linda M. Sircy, Malia Harrison-Chau, Nguyen X. Nguyen, Katherine E. Varley, Dean Tantin, and J. Scott Hale
- Subjects
Multidisciplinary ,T Follicular Helper Cells ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Germinal Center ,Lymphocyte Activation - Abstract
In response to various types of infection, naïve CD4 + T cells differentiate into diverse helper T cell subsets; however, the epigenetic programs that regulate differentiation in response to viral infection remain poorly understood. Demethylation of CpG dinucleotides by Tet methylcytosine dioxygenases is a key component of epigenetic programing that promotes specific gene expression, cellular differentiation, and function. We report that following viral infection, Tet2-deficient CD4 + T cells preferentially differentiate into highly functional germinal center T follicular helper (T FH ) cells that provide enhanced help for B cells. Using genome-wide DNA methylation and transcription factor binding analyses, we find that Tet2 coordinates with multiple transcription factors, including Foxo1 and Runx1, to mediate the demethylation and expression of target genes, including genes encoding repressors of T FH differentiation. Our findings establish Tet2 as an important regulator of T FH cell differentiation and reveal pathways that could be targeted to enhance immune responses against infectious disease.
- Published
- 2022
11. The lingering mysteries of metastatic recurrence in breast cancer
- Author
-
Katherine E. Varley, Alana L. Welm, and Alessandra I. Riggio
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Breast Neoplasms ,Review Article ,Disease ,Systemic therapy ,Metastasis ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Neoplasm Recurrence ,Internal medicine ,Animals ,Humans ,Medicine ,Neoplasm Invasiveness ,Neoplasm Metastasis ,030304 developmental biology ,0303 health sciences ,business.industry ,Extramural ,Cancer ,medicine.disease ,Key factors ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business - Abstract
Despite being the hallmark of cancer that is responsible for the highest number of deaths, very little is known about the biology of metastasis. Metastatic disease typically manifests after a protracted period of undetectable disease following surgery or systemic therapy, owing to relapse or recurrence. In the case of breast cancer, metastatic relapse can occur months to decades after initial diagnosis and treatment. In this review, we provide an overview of the known key factors that influence metastatic recurrence, with the goal of highlighting the critical unanswered questions that still need to be addressed to make a difference in the mortality of breast cancer patients.
- Published
- 2020
- Full Text
- View/download PDF
12. STAT3 and GR Cooperate to Drive Gene Expression and Growth of Basal-Like Triple-Negative Breast Cancer
- Author
-
James Turkson, Peibin Yue, Katherine E. Varley, Joy M. McDaniel, Stephanie L. Parker, Donald J. Buchsbaum, Bryan E. Welm, Katrin P. Guillen, Richard M. Myers, Patsy G. Oliver, Megan E. Conway, and James M. Graham
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Chromatin Immunoprecipitation ,Cancer Research ,Triple Negative Breast Neoplasms ,Kaplan-Meier Estimate ,Regulatory Sequences, Nucleic Acid ,Dexamethasone ,Disease-Free Survival ,Article ,03 medical and health sciences ,Receptors, Glucocorticoid ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,Gene expression ,medicine ,Humans ,STAT3 ,Transcription factor ,Triple-negative breast cancer ,Regulation of gene expression ,Binding Sites ,biology ,DNA Methylation ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,biology.protein ,Female ,Estrogen receptor alpha - Abstract
Breast cancers are divided into subtypes with different prognoses and treatment responses based on global differences in gene expression. Luminal breast cancer gene expression and proliferation are driven by estrogen receptor alpha, and targeting this transcription factor is the most effective therapy for this subtype. By contrast, it remains unclear which transcription factors drive the gene expression signature that defines basal-like triple-negative breast cancer, and there are no targeted therapies approved to treat this aggressive subtype. In this study, we utilized integrated genomic analysis of DNA methylation, chromatin accessibility, transcription factor binding, and gene expression in large collections of breast cancer cell lines and patient tumors to identify transcription factors responsible for the basal-like gene expression program. Glucocorticoid receptor (GR) and STAT3 bind to the same genomic regulatory regions, which were specifically open and unmethylated in basal-like breast cancer. These transcription factors cooperated to regulate expression of hundreds of genes in the basal-like gene expression signature, which were associated with poor prognosis. Combination treatment with small-molecule inhibitors of both transcription factors resulted in synergistic decreases in cell growth in cell lines and patient-derived organoid models. This study demonstrates that GR and STAT3 cooperate to regulate the basal-like breast cancer gene expression program and provides the basis for improved therapy for basal-like triple-negative breast cancer through rational combination of STAT3 and GR inhibitors. Significance: This study demonstrates that GR and STAT3 cooperate to activate the canonical gene expression signature of basal-like triple-negative breast cancer and that combination treatment with STAT3 and GR inhibitors could provide synergistic therapeutic efficacy.
- Published
- 2020
- Full Text
- View/download PDF
13. Abstract P1-10-09: The MHCII immune activation assay is prognostic for disease free survival in basal-like TNBC breast cancer patients in the GEICAM/9906 clinical trial
- Author
-
Katherine L. Updike, Álvaro Rodríguez-Lescure, Jesús Herranz, Miguel Martín, Nuria Martin, Lourdes Calvo, Philip S. Bernard, and Katherine E. Varley
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Disease free survival ,business.industry ,medicine.disease ,Clinical trial ,Basal (phylogenetics) ,Breast cancer ,Internal medicine ,medicine ,business ,Immune activation - Abstract
Background: Approximately 40% of patients with stage I-III triple-negative breast cancer (TNBC) recur after standard treatment, while the remaining 60% experience long-term disease-free survival (DFS). There are currently no clinical tests to assess the risk of recurrence in TNBC patients. We previously developed and validated an MHCII Immune Activation assay, which measures a gene expression signature that includes the major histocompatibility complex class II (MHCII) pathway genes and markers of activated tumor infiltrating lymphocytes. The MHCII Immune Activation assay was recently shown to accurately assess the risk of recurrence in Basal-like TNBC patients using FFPE tissues from an institutional cohort. A limitation of the previous study is that the patients’ treatment regimens in the institutional cohort were heterogeneous. The goal of this study was to determine if the MHCII Immune Activation assay can accurately assess the DFS in Basal-like TNBC patients who received uniform chemotherapy regimens in the context of a randomized clinical trial. Methods: The MHCII Immune Activation assay was applied to RNA isolated from 60 Basal-like TNBC FFPE specimens from the GEICAM/9906 clinical trial, a multicenter randomized phase III study evaluating adjuvant chemotherapy in node-positive operable BC patients. Thirty nine (65%) of the patients were treated with fluorouracil, epirubicin, and cyclophosphamide (FEC), and 21 (35%) patients received FEC followed by paclitaxel (FEC-P) (median follow-up 9.7 years). The MHCII Immune Activation Score was calculated from the assay using methods described previously, and patients were categorized into a High Risk of Recurrence (High ROR) group using pre-specified thresholds. The association between the MHCII Immune Activation Score and DFS was assessed using Kaplan-Meier analysis and cox proportional hazards models. Results: Across both arms, 32/60 (53.3%) patients were classified into the High ROR group based on their Low MHCII Immune Activation Scores. Patients in the High ROR group had significantly shorter DFS (Hazard Ratio (HR)=2.9 (CI95%:1.27-6.66), p-value=0.0081). Cox proportional hazard regression of the log10 transformed MHCII Immune Activation Score confirmed that MHCII Immune Activation Score is significantly associated with DFS (Wald Test p=0.0127). In the FEC arm, there were 22/39 (56.41%) patients classified into the High ROR group (Low MHCII Immune Activation Score). Patients in the High ROR group in the FEC arm showed a trend toward shorter DFS (HR=1.99 (CI95%:0.74-5.31)), but it was not significant (p-value=0.1595). In the FEC-P arm, there were 10/21 (47.62%) patients classified into the High ROR group (Low MHCII Immune Activation Score). Patients in the High ROR group in the FEC-P arm had significantly shorter DFS (HR=6.28 (CI95%:1.28-30.73), p-value=0.0111). Larger cohorts will be needed to confirm if the association between DFS and MHCII Immune Activation Score is stronger when patients are treated with chemotherapy regimens that include paclitaxel. Conclusions: The MHCII Immune Activation assay can be applied to RNA from FFPE tumor specimens to assess risk of recurrence in TNBC patients. The MHCII Immune Activation Score is associated with risk of recurrence in TNBC patients treated with uniform adjuvant chemotherapy regimens. Citation Format: Miguel Martín, Katherine L Updike, Alvaro Rodríguez-Lescure, Lourdes Calvo, Jesús Herranz, Nuria Martín, Philip S Bernard, Katherine E Varley. The MHCII immune activation assay is prognostic for disease free survival in basal-like TNBC breast cancer patients in the GEICAM/9906 clinical trial [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P1-10-09.
- Published
- 2020
- Full Text
- View/download PDF
14. A breast cancer patient-derived xenograft and organoid platform for drug discovery and precision oncology
- Author
-
Ward Jh, Randy L. Jensen, Ling Zhao, Greenland Ja, Maihi Fujita, Rosenthal R, Saundra S. Buys, Anna C. Beck, Guoying Wang, Wadsworth Me, Emilio Cortes-Sanchez, Sandra D. Scherer, Chieh-Hsiang Yang, Katrin P. Guillen, Jeffrey H. Chuang, Cindy B. Matsen, Zheqi Li, Kristopher Berrett, Jeffery M. Vahrenkamp, David H. Lum, Alana L. Welm, Michael T. Lewis, Toner J, Jason Gertz, Chu Z, Poretta Jm, Yoko S. DeRose, Lacey E. Dobrolecki, Gabor T. Marth, Katherine E. Varley, Rachel E. Factor, Andrew Butterfield, Bryan E. Welm, Xiaomeng Huang, Edward W. Nelson, Matthew H. Bailey, Pathi Ss, Xing Yi Woo, Yi Qiao, Christos Vaklavas, Steffi Oesterreich, and Kevin B. Jones
- Subjects
Drug ,Oncology ,medicine.medical_specialty ,Drug discovery ,business.industry ,media_common.quotation_subject ,Cancer ,Precision medicine ,medicine.disease ,Breast cancer ,Drug development ,In vivo ,Internal medicine ,Medicine ,business ,Triple-negative breast cancer ,media_common - Abstract
Model systems that recapitulate the complexity of human tumors and the reality of variable treatment responses are urgently needed to better understand cancer biology and to develop more effective cancer therapies. Here we report development and characterization of a large bank of patient-derived xenografts (PDX) and matched organoid cultures from tumors that represent some of the greatest unmet needs in breast cancer research and treatment. These include endocrine-resistant, treatment-refractory, and metastatic breast cancers and, in some cases, multiple tumor collections from the same patients. The models can be grown long-term with high fidelity to the original tumors. We show that development of matched PDX and PDX-derived organoid (PDxO) models facilitates high-throughput drug screening that is feasible and cost-effective, while also allowing in vivo validation of results. Our data reveal consistency between drug screening results in organoids and drug responses in breast cancer PDX. Moreover, we demonstrate the feasibility of using these patient-derived models for precision oncology in real time with patient care, using a case of a triple negative breast cancer with early metastatic recurrence as an example. Our results uncovered an FDA-approved drug with high efficacy against the models. Treatment with the PDxO-directed therapy resulted in a complete response for the patient and a progression-free survival period more than three times longer than her previous therapies. This work provides valuable new methods and resources for functional precision medicine and drug development for human breast cancer.Graphical Abstract
- Published
- 2021
- Full Text
- View/download PDF
15. Tet2-mediated programing balances T follicular helper cell and T helper 1 cell differentiation
- Author
-
J. Scott Hale, Andrew Baessler, Camille L Novis, Zuolian Shen, Jelena Perovanovic, Mark Wadsworth, Linda M Sircy, Malia Harrison-Chau, Katherine E Varley, and Dean R Tantin
- Subjects
Immunology ,Immunology and Allergy - Abstract
The differentiation of CD4+ T cell subsets in response to infection has been studied extensively, however the epigenetic programs that regulate these processes remain poorly understood. Active demethylation by tet methylcytosine dioxygenases of CpG dinucleotides within DNA is a key component of epigenetic programing that promotes lineage specific gene expression and contributes to cellular differentiation and function. Here we report that Tet2 acts to restrict the differentiation of T follicular helper (Tfh) cells in CD4+ T cells responding to viral infection. Using an adoptive transfer model of virus-specific CD4+ cells we found that Tet2-deficient CD4+ T cells skew away from the Th1 lineage and instead preferentially differentiate into highly functional germinal center (GC) Tfh cells that provide enhanced help for B cell responses. We found that the impact of Tet2-mediated programing on CD4+ T cell differentiation is cell intrinsic and the shift in lineage differentiation occurs as early as 2 days post infection. Using genome-wide expression, DNA methylation and transcription factor binding analyses, we found that Tet2 coordinates with multiple transcription factors to mediate the demethylation and expression of their target genes following activation. Our findings establish Tet2 as an important regulator of Tfh cell differentiation and reveal pathways that could be targeted to enhance GC responses against infectious disease. Supported by grants from NIH (R01 AI137238 to J.S.H., T32 AI055434 to L.M.S., and T32 AI138945 to A.B.
- Published
- 2022
- Full Text
- View/download PDF
16. CaBagE: a Cas9-based Background Elimination strategy for targeted, long-read DNA sequencing
- Author
-
Thomas A Sasani, Amelia D. Wallace, Aaron R. Quinlan, Jordan Swanier, Brooke L. Gates, Brent S. Pedersen, Katherine E. Varley, and Jeff Greenland
- Subjects
Tandem repeat ,Cas9 ,Minion ,Microsatellite ,Human genome ,Computational biology ,Nanopore sequencing ,Biology ,Genome ,DNA sequencing - Abstract
A substantial fraction of the human genome is difficult to interrogate with short-read DNA sequencing technologies due to paralogy, complex haplotype structures, or tandem repeats. Long-read sequencing technologies, such as Oxford Nanopore’s MinION, enable direct measurement of complex loci without introducing many of the biases inherent to short-read methods, though they suffer from relatively lower throughput. This limitation has motivated recent efforts to develop amplification-free strategies to target and enrich loci of interest for subsequent sequencing with long reads. Here, we present CaBagE, a novel method for target enrichment that is efficient and useful for sequencing large, structurally complex targets. The CaBagE method leverages the stable binding of Cas9 to its DNA target to protect desired fragments from digestion with exonuclease. Enriched DNA fragments are then sequenced with Oxford Nanopore’s MinION long-read sequencing technology. Enrichment with CaBagE resulted in up to 416X coverage of target loci when tested on five genomic targets ranging from 4-20kb in length using healthy donor DNA. Four cancer gene targets were enriched in a single reaction and multiplexed on a single MinION flow cell. We further demonstrate the utility of CaBagE in two ALS patients with C9orf72 short tandem repeat expansions to produce genotype estimates commensurate with genotypes derived from repeat-primed PCR for each individual. With CaBagE there is a physical enrichment of on-target DNA in a given sample prior to sequencing. This feature allows adaptability across sequencing platforms and potential use as an enrichment strategy for applications beyond sequencing. CaBagE is a rapid enrichment method that can illuminate regions of the ‘hidden genome’ underlying human disease.
- Published
- 2020
- Full Text
- View/download PDF
17. Spatially-resolved quantification of proteins in triple negative breast cancers reveals differences in the immune microenvironment associated with prognosis
- Author
-
Rachel E. Factor, Rachel L. Stewart, Anna P. Matynia, and Katherine E. Varley
- Subjects
Stromal cell ,Immune microenvironment ,lcsh:Medicine ,Triple Negative Breast Neoplasms ,Biology ,Article ,Inducible T-Cell Co-Stimulator Protein ,Prognostic markers ,Breast cancer ,Immune system ,Recurrence ,Cell Line, Tumor ,Tumor Microenvironment ,Humans ,Lymphocytes ,lcsh:Science ,Triple negative ,Triple-negative breast cancer ,MHC class II ,Multidisciplinary ,Tumor-infiltrating lymphocytes ,Spatially resolved ,lcsh:R ,HLA-DR Antigens ,Translational research ,Prognosis ,Neoplasm Proteins ,CD4 Antigens ,Cancer research ,biology.protein ,lcsh:Q ,Stromal Cells - Abstract
Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype. Recent studies have shown that MHC class II (MHCII) expression and tumor infiltrating lymphocytes are important prognostic factors in patients with TNBC, although the relative importance of lymphocyte subsets and associated protein expression is incompletely understood. NanoString Digital Spatial Profiling (DSP) allows for spatially resolved, highly multiplexed quantification of proteins in clinical samples. In this study, we sought to determine if DSP could be used to characterize expression of MHCII and other immune related proteins in tumor epithelial versus stromal compartments of patient-derived TNBCs (N = 10) using a panel of 39 markers. We confirmed that a subset of TNBCs have elevated expression of HLA-DR in tumor epithelial cells; HLA-DR expression was also significantly higher in the tumors of patients with long-term disease-free survival when compared to patients that relapsed. HLA-DR expression in the epithelial compartment was correlated with high expression of CD4 and ICOS in the stromal compartment of the same tumors. We also identified candidate protein biomarkers with significant differential expression between patients that relapsed versus those that did not. In conclusion, DSP is a powerful method that allows for quantification of proteins in the immune microenvironment of TNBCs.
- Published
- 2020
- Full Text
- View/download PDF
18. MS4A3 Promotes Differentiation in Chronic Myeloid Leukemia By Enhancing Common β Chain Cytokine Receptor Endocytosis
- Author
-
Jason Gertz, Derek L. Stirewalt, Amber D. Bowler, Sooryanarayana Varambally, Bret Helton, Jeffrey W. Tyner, Michael W. Deininger, Shannon K. McWeeney, Dongqing Yan, Jae Yeon Hwang, Phillip M. Clair, Matthew S. Zabriskie, Katherine E. Varley, Hannah M. Redwine, Kristofer C. Berrett, Anna M. Eiring, Anna V. Senina, Anthony D. Pomicter, Jamshid S. Khorashad, Siddharth M. Iyer, Helong Zhao, Anupriya Agarwal, Brian J. Druker, Anca Franzini, Jeffrey M Vahrenkamp, Vivian G. Oehler, Jonathan M. Ahmann, and Jerald P. Radich
- Subjects
Myeloid Neoplasia ,Myeloid ,Stem Cells ,medicine.medical_treatment ,Cellular differentiation ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Leukemia, Myeloid, Acute ,Cytokine ,medicine.anatomical_structure ,Downregulation and upregulation ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Imatinib Mesylate ,medicine ,Humans ,Progenitor cell ,Cytokine receptor ,Tyrosine kinase ,health care economics and organizations - Abstract
Background Chronic phase chronic myeloid leukemia (CP-CML) is characterized by overproduction of differentiating myeloid cells, while blast phase CML (BP-CML) cells exhibit a differentiation block. Tyrosine kinase inhibitors (TKIs) are effective in CP-CML, but resistance is common in BP-CML and can occur without explanatory BCR-ABL1 kinase mutations (BCR-ABL1-independent resistance). Similarly, CML stem/progenitor cells (LSPCs) are insensitive to TKIs, and residual leukemia persists in the majority of CML patients on TKI therapy. We previously reported overlap between the transcriptomes of CD34 + cells from BP-CML and TKI-naïve CP-CML patients with primary TKI resistance, pointing to commonalities between LSPC quiescence, BCR-ABL1-independent TKI resistance, and BP-CML. Results To identify common mechanisms, we performed a meta-analysis of published CML transcriptomes. We identified a small set of genes with consistently low expression in LSPC quiescence, BCR-ABL1-independent TKI resistance, and BP-CML, including Membrane Spanning 4-Domains A3 (MS4A3), a signaling protein previously reported to inhibit hematopoietic cell cycle progression. Low MS4A3 in CD34 + cells from TKI-naïve CP-CML patients was associated with shorter survival on subsequent TKI therapy, suggesting that MS4A3 governs TKI response. To understand the function of MS4A3, we lentivirally introduced MS4A3 shRNA or an MS4A3 expression vector into CML CD34 + LSPCs. MS4A3 knockdown increased clonogenicity and imatinib resistance, while ectopic MS4A3 expression had opposite effects. MS4A3 KD also increased LSPC persistence ex vivo in LTC-IC assays, and in vivo in NSG mice xenografts, while modulating MS4A3 expression had no effect on normal CD34 + cells. We next generated Ms4a3+/+│-/-; Scl-tTA+; TRE-BCR-ABL1+ compound transgenic mice. Upon BCR-ABL1 induction, Ms4a3-/-; Scl-tTA+; TRE-BCR-ABL1+ mice developed leukocytosis comparable to Ms4a3+/+ controls. However, BM of Ms4a3-/-; Scl-tTA+; TRE-BCR-ABL1+ mice showed increased short-term HSCs and multipotent progenitor cells, and reduced granulocyte-macrophage progenitors. When Lin - BM cells from leukemic mice were transplanted into irradiated recipients, Ms4a3-/-; Scl-tTA+; TRE-BCR-ABL1+ cells showed increased engraftment and myeloid leukocytosis, validating our observations in human cells. To determine how MS4A3 is downregulated in CML, we expressed BCR-ABL1in 32D-cl3 cells. p210 BCR-ABL1 drastically reduced Ms4a3 expression, while kinase-inactive p210 BCR-ABL1-K271R had no effect. Moreover, we found that suppression of C/EBPε by MECOM reduces MS4A3, consistent with previous reports of MECOM as a driver of TKI resistance and progression to BP. Treatment of CML CD34 + cells with a library of epigenetic pathway inhibitors revealed that MS4A3 is suppressed by both DNA methylation and PRC2/EZH2-mediated H3K27 trimethylation, which was confirmed by patch-PCR and ChIPseq. These data indicate that multi-levelled mechanisms cooperate in the suppression of MS4A3 in CML. To determine how MS4A3 regulates clonogenicity and TKI response, we expressed MS4A3-EGFP fusion protein in LAMA-84 CML cells. We found that MS4A3 resides on the plasma membrane and in endosomes. Surface protein biotin labelling and tandem mass spectrometry ± MS4A3 KD showed that MS4A3 controls endocytosis of membrane proteins, including common β chain (βc) cytokine receptors. Specifically, MS4A3 promotes endocytosis of βc cytokine receptors upon GM-CSF/IL-3 stimulation of primary LSPCs and enhances downstream signaling and differentiation, suggesting that restoring MS4A3 expression has therapeutic efficacy. To test this, we manufactured a prototype MS4A3 protein-loaded liposomal nanoparticle (NP) using coating with the CD34 CD62L for targeted delivery to CD34 + cells. Compared to MS4A3-free NPs, MS4A3 NPs increased CD34 +CD38 + and CD34 -CD38 + at the expense of CD34 +CD38 - cells, reduced clonogenicity, and increased sensitivity to TKIs, mimicking ectopic MS4A3 expression. Conclusion MS4A3 governs response to differentiating myeloid cytokines, providing a unifying mechanism for the differentiation block characteristic of primitive LSPCs and BP-CML cells. We posit that LSPCs downregulate MS4A3 to evade βc cytokine-induced differentiation to maintain a primitive, TKI-insensitive state. MS4A3 re-expression or delivery of ectopic MS4A3 may eliminate LSPCs. Figure 1 Figure 1. Disclosures Druker: Aptose Therapeutics: Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Cepheid: Consultancy, Membership on an entity's Board of Directors or advisory committees; EnLiven: Consultancy, Research Funding; Blueprint Medicines: Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Aileron: Membership on an entity's Board of Directors or advisory committees; Amgen: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Research Funding; ALLCRON: Consultancy, Membership on an entity's Board of Directors or advisory committees; GRAIL: Current equity holder in publicly-traded company; Iterion Therapeutics: Membership on an entity's Board of Directors or advisory committees; Merck & Co: Patents & Royalties; Nemucore Medical Innovations, Inc.: Consultancy; Novartis Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; Pfizer: Research Funding; Recludix Pharma, Inc.: Consultancy; The RUNX1 Research Program: Membership on an entity's Board of Directors or advisory committees; Third Coast Therapeutics: Membership on an entity's Board of Directors or advisory committees; VB Therapeutics: Membership on an entity's Board of Directors or advisory committees; Vincerx Pharma: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Vivid Biosciences: Membership on an entity's Board of Directors or advisory committees. Tyner: Agios: Research Funding; Astrazeneca: Research Funding; Array: Research Funding; Genentech: Research Funding; Janssen: Research Funding; Takeda: Research Funding; Gilead: Research Funding; Incyte: Research Funding; Petra: Research Funding; Seattle Genetics: Research Funding; Constellation: Research Funding; Schrodinger: Research Funding. Oehler: BMS: Consultancy; OncLive: Honoraria; Pfizer: Research Funding; Takeda: Consultancy; Blueprint Medicines: Consultancy. Radich: BMS: Membership on an entity's Board of Directors or advisory committees; Genentech: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees. Deininger: Sangamo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Part of a Study Management Committee, Research Funding; Incyte: Consultancy, Honoraria, Research Funding; Fusion Pharma, Medscape, DisperSol: Consultancy; Novartis: Consultancy, Research Funding; SPARC, DisperSol, Leukemia & Lymphoma Society: Research Funding; Blueprint Medicines Corporation: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Part of a Study Management Committee, Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
19. A novel AGGF1-PDGFRb fusion in pediatric T-cell acute lymphoblastic leukemia
- Author
-
Michael W. Deininger, Orlando Antelope, Katherine E. Varley, Thomas O'Hare, Rodney R. Miles, Anupam Verma, Reha M. Toydemir, Matthew S. Zabriskie, Thai Hoa Tran, Christopher A. Eide, Brian J. Druker, James M. Graham, Jae Yeon Hwang, Anthony D. Pomicter, Elizabeth A. Raetz, Lauren Draper, and Jeffrey W. Tyner
- Subjects
Male ,0301 basic medicine ,Fatal outcome ,Oncogene Proteins, Fusion ,Oncogene Proteins ,Lymphoblastic Leukemia ,T cell ,PDGFRB ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Receptor, Platelet-Derived Growth Factor beta ,03 medical and health sciences ,Fatal Outcome ,0302 clinical medicine ,Text mining ,Humans ,Medicine ,Angiogenic Proteins ,Online Only Articles ,Receptor ,Protein Kinase Inhibitors ,Gene Rearrangement ,business.industry ,Hematology ,Gene rearrangement ,030104 developmental biology ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,Cancer research ,business - Published
- 2017
- Full Text
- View/download PDF
20. A Multigene Assay Determines Risk of Recurrence in Patients with Triple-Negative Breast Cancer
- Author
-
Rachel E. Factor, Katherine L. Updike, Philip S. Bernard, Kenneth M. Boucher, Rachel L. Stewart, N. Lynn Henry, and Katherine E. Varley
- Subjects
0301 basic medicine ,Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Lymphocyte ,Triple Negative Breast Neoplasms ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Lymphocytes, Tumor-Infiltrating ,Risk Factors ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Stage (cooking) ,Triple-negative breast cancer ,Aged ,Chemotherapy ,business.industry ,Standard treatment ,Histocompatibility Antigens Class II ,Immunotherapy ,Middle Aged ,Precision medicine ,medicine.disease ,Prognosis ,Survival Rate ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
Approximately 40% of patients with stage I–III triple-negative breast cancer (TNBC) recur after standard treatment, whereas the remaining 60% experience long-term disease-free survival (DFS). There are currently no clinical tests to assess the risk of recurrence in TNBC patients. We previously determined that TNBC patients with MHC class II (MHCII) pathway expression in their tumors experienced significantly longer DFS. To translate this discovery into a clinical test, we developed an MHCII Immune Activation assay, which measures expression of 36 genes using NanoString technology. Preanalytical testing confirmed that the assay is accurate and reproducible in formalin-fixed paraffin-embedded (FFPE) tumor specimens. The assay measurements were concordant with RNA-seq, MHCII protein expression, and tumor-infiltrating lymphocyte counts. In a training set of 44 primary TNBC tumors, the MHCII Immune Activation Score was significantly associated with longer DFS (HR = 0.17; P = 0.015). In an independent validation cohort of 56 primary FFPE TNBC tumors, the Immune Activation Score was significantly associated with longer DFS (HR = 0.19; P = 0.011) independent of clinical stage. An Immune Activation Score threshold for identifying patients with very low risk of relapse in the training set provided 100% specificity in the validation cohort. The assay format enables adoption as a standardized clinical prognostic test for identifying TNBC patients with a low risk of recurrence. Correlative data support future studies to determine if the assay can identify patients in whom chemotherapy can be safely deescalated and patients likely to respond to immunotherapy. Significance: The MHCII Immune Activation assay identifies TNBC patients with a low risk of recurrence, addressing a critical need for prognostic biomarker tests that enable precision medicine for TNBC patients.
- Published
- 2018
21. Single-Cell Transcriptomes Distinguish Stem Cell State Changes and Lineage Specification Programs in Early Mammary Gland Development
- Author
-
Christopher Dravis, Chi-Yeh Chung, Cheng Fan, Rajshekhar R. Giraddi, Mark Novotny, Benjamin T. Spike, Elnaz Mirzaei Mehrabad, Berhane M. Hagos, Ozlen Balcioglu, Roger S. Lasken, Katherine E. Varley, Richard E. Heinz, Christy L. Trejo, Luo Wei Rodewald, Charles M. Perou, Jae Y. Hwang, and Geoffrey M. Wahl
- Subjects
0301 basic medicine ,Mammary gland ,Cell ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,Mammary Glands, Animal ,Gene expression ,medicine ,Animals ,Humans ,lcsh:QH301-705.5 ,Stem Cells ,Cell Differentiation ,Gene signature ,Embryonic stem cell ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,Female ,Stem cell ,Carcinogenesis - Abstract
SUMMARY The mammary gland consists of cells with gene expression patterns reflecting their cellular origins, function, and spatiotemporal context. However, knowledge of developmental kinetics and mechanisms of lineage specification is lacking. We address this significant knowledge gap by generating a single-cell transcriptome atlas encompassing embryonic, postnatal, and adult mouse mammary development. From these data, we map the chronology of transcriptionally and epigenetically distinct cell states and distinguish fetal mammary stem cells (fMaSCs) from their precursors and progeny. fMaSCs show balanced co-expression of factors associated with discrete adult lineages and a metabolic gene signature that subsides during maturation but reemerges in some human breast cancers and metastases. These data provide a useful resource for illuminating mammary cell heterogeneity, the kinetics of differentiation, and developmental correlates of tumorigenesis., Graphical Abstract, In Brief Single-cell RNA sequencing of developing mouse mammary epithelia reveals the timing of lineage specification. Giraddi et al. find that fetal mammary stem cells co-express factors that define distinct lineages in their progeny and bear functionally relevant metabolic program signatures that change with differentiation and are resurrected in human breast cancers and metastases.
- Published
- 2017
22. PO-487 Prognosis of triple negative breast cancer is associated with MHC II genes
- Author
-
G.B. Bond, Yufeng Li, Andres Forero, Albert F. LoBuglio, A. Zhao, and Katherine E. Varley
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,CD74 ,business.industry ,medicine.medical_treatment ,Odds ratio ,medicine.disease ,Targeted therapy ,Breast cancer ,Internal medicine ,Progesterone receptor ,medicine ,CIITA ,Progression-free survival ,business ,Triple-negative breast cancer - Abstract
Introduction Triple negative breast cancer (TNBC) is clinically to denote women with invasive breast cancer whose tumours lack expression of oestrogen receptor (ER-), progesterone receptor (PR-), or overexpression of HER2/Neu. TNB tumours behave aggressively and are not candidates for ER or HER2/Neu targeted therapy. In previous published study of 47 TNBC patients with RNA-seq data, we discovered that twenty-four genes related MCH pathway exhibited significantly higher expression in tumour tissue from patients who did not relapse, including eleven genes representation of the MHC II pathway. In this study, we further investigated 24 genes in MCH pathway in prognosis of TNBC. This work will lead in future guidance in treatment of TNB based on patient’s’ gene profile and also provides means to assess prognosis in TNBC. Material and methods Forty-seven snap frozen primary TNBC tumour specimens were analysed using RNA-seq. Descriptive analysis and logistic regression model were used to identify genes that can predict TNB good prognosis (no relapsed). Odds ratio and 95% two-sided confidence intervals were presented. The optimal cut point of expression level was determined from the receiver operating characteristic analysis and sensitivity and specificity were estimated. This study has approval of Institutional Review Board. Results and discussions MCH II genes CD74 and CIITA had significantly association with TNBC progression free survival. When TNBC patients had high CIITA, they were 10 times likely to have no relapsed than patients with low expression (odds ratio OR=10.8, 95% CI 2.8–41.9, p=0.006). The sensitivity and specificity is 75% and 78.3% respectively; TNB who had higher expression of CD74 had 3 times more likely to have no relapsed (OR=3.1, 95% CI 0.95–10.28, p=0.0609) with sensitivity and specificity of 62.5% and 65.2% respectively. We used multivariable approach with all 24-gene panel and identified three genes, CIITA, CTSH and KRT14 together can predict no relapsed with 87.5% sensitivity 78.3%. The accuracy is 83% if TNBC had high expression of CIITA, CTSH and KRT14. With a total of 47 patients, we predicted 17 out 22 relapsed, and 21 out 25 no relapsed correctly. The results was validated using the public microarray data from 199 patients with TNBC confirmed. Conclusion Higher expression of genes in MCII pathway, e.g. CIITA, CD74, not only associated with TNBC progression free survival, they also have high accuracy to predict TNBC patient’s no relapse regardless of age, race, and tumour grade and tumour stage.
- Published
- 2018
- Full Text
- View/download PDF
23. Abstract P1-15-02: Long term follow-up of the neo-adjuvant pilot trial evaluating activity of letrozole in combination with bevacizumab in post-menopausal women with newly diagnosed estrogen and/or progesterone receptor positive primary breast cancer
- Author
-
Helen Krontiras, Andres Forero, Carla I. Falkson, Richard M. Myers, Lisle Nabell, K Bowen, Katherine E. Varley, Valerie Caterinicchia, John T. Carpenter, J DeLos Santos, Albert F. LoBuglio, Janis P. O'Malley, C. Jones, Mansoor N. Saleh, and Y. Li
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Letrozole ,medicine.medical_treatment ,Cancer ,medicine.disease ,Inflammatory breast cancer ,Surgery ,Regimen ,Breast cancer ,Internal medicine ,medicine ,Clinical endpoint ,business ,Neoadjuvant therapy ,medicine.drug - Abstract
Introduction: Vascular endothelial growth factor overexpression has been associated with resistance to anti-estrogen therapy (Cancer Res 2008; 68: 6232); our preclinical data showed that anti-VEGF therapy reverse resistance to estrogen therapy. We postulated that anti-VEGF therapy would enhance anti-estrogen therapy and thus designed a pilot study to assess the feasibility and efficacy of neoadjuvant letrozole and bevacizumab in post-menopausal women with stage II/III, ER/PR positive breast cancer. Patients and Methods: Eligible patients were treated with a neo-adjuvant regimen of letrozole, 2.5 mg/day (PO) and bevacizumab 15 mg/kg every 3 weeks (IV) for a total of 24 weeks prior to surgical treatment of their breast cancer. Patients were followed for toxicity at three week intervals and for tumor assessment at 6 week intervals. Research tumor biopsies were taken before and 6 weeks after initiation of therapy. The primary endpoint was pathological complete remission (pCR). Patients with inflammatory breast cancer were excluded. Results: Twenty six patients were enrolled and 25 were treated (one patient had a TIA the day before initiation of therapy). The regimen was well tolerated with 2 patients taken off-study due to uncontrolled hypertension. Objective clinical response occurred in 68% of the patients (17/25), 16% with CR and 52% with partial response (PR). Sixteen percent of the patients (4/25) had clinical stable disease (SD) and 2 patients progressed (PD) while on therapy. Three patients had pCR and 1 patient had microscopic residual tumor cells in the LNs but not in the breast (pCR 16%). Thirty two percent of the patients attained stage 0 or 1 status. None of the pCR patients received adjuvant chemotherapy and none have relapsed after a median follow-up of 6.1 years (range, 5.8+ to 7.5+). Eight of the 13 patients with PR did not receive chemotherapy and only one relapsed with a median follow-up of 6.2 years (range, 3.7 to 7.7+). At a median follow-up of 6.4 years, 88% of the patients have not relapsed and 12% relapsed (1 PD [basal-like], 1 PR [Luminal B], 1 SD [HER2] relapsed at 1.7, 4, and 6.8 years respectively). Of the 17 patients with CR and PR, only 1 has relapsed (6%). Next Generation Sequencing Analysis and evaluation of markers of proliferation/apoptosis are underway. Conclusion: Combination neoadjuvant therapy with letrozole and bevacizumab was well tolerated and resulted in an impressive pCR of 16%. At a median of 6.4 years, the relapse free survival is 88% for all comers and 94% for responding patients (Luminal A and B). Full correlation of clinical and genomic/biomarker analysis will be presented at the time of the meeting. This encouraging data has led The Breast Cancer Translational Research Consortium to complete a randomized phase II trial (TBCRC002) of letrozole ± bevacizumab in this patient population. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P1-15-02.
- Published
- 2013
- Full Text
- View/download PDF
24. Dynamic DNA methylation across diverse human cell lines and tissues
- Author
-
Richard M. Myers, Brian A. Williams, Barbara J. Wold, Stephanie L. Parker, Florencia Pauli-Behn, Jason Gertz, John A. Stamatoyannopoulos, Timothy E. Reddy, Gregory E. Crawford, Devin Absher, Katherine E. Varley, Kevin M. Bowling, and Marie K. Cross
- Subjects
Resource ,Genetics ,Gene Expression Profiling ,EZH2 ,Sequence Analysis, DNA ,Methylation ,DNA Methylation ,Biology ,Chromatin ,Epigenetics of physical exercise ,Gene Expression Regulation ,Cell Line, Tumor ,Histone methylation ,DNA methylation ,Humans ,Sulfites ,Illumina Methylation Assay ,CpG Islands ,Promoter Regions, Genetic ,Sequence Alignment ,RNA-Directed DNA Methylation ,Genetics (clinical) ,Oligonucleotide Array Sequence Analysis ,Epigenomics - Abstract
As studies of DNA methylation increase in scope, it has become evident that methylation has a complex relationship with gene expression, plays an important role in defining cell types, and is disrupted in many diseases. We describe large-scale single-base resolution DNA methylation profiling on a diverse collection of 82 human cell lines and tissues using reduced representation bisulfite sequencing (RRBS). Analysis integrating RNA-seq and ChIP-seq data illuminates the functional role of this dynamic mark. Loci that are hypermethylated across cancer types are enriched for sites bound by NANOG in embryonic stem cells, which supports and expands the model of a stem/progenitor cell signature in cancer. CpGs that are hypomethylated across cancer types are concentrated in megabase-scale domains that occur near the telomeres and centromeres of chromosomes, are depleted of genes, and are enriched for cancer-specific EZH2 binding and H3K27me3 (repressive chromatin). In noncancer samples, there are cell-type specific methylation signatures preserved in primary cell lines and tissues as well as methylation differences induced by cell culture. The relationship between methylation and expression is context-dependent, and we find that CpG-rich enhancers bound by EP300 in the bodies of expressed genes are unmethylated despite the dense gene-body methylation surrounding them. Non-CpG cytosine methylation occurs in human somatic tissue, is particularly prevalent in brain tissue, and is reproducible across many individuals. This study provides an atlas of DNA methylation across diverse and well-characterized samples and enables new discoveries about DNA methylation and its role in gene regulation and disease.
- Published
- 2013
- Full Text
- View/download PDF
25. CTCF/cohesin-mediated DNA looping is required for protocadherin α promoter choice
- Author
-
Ya Guo, Jason Gertz, Tom Maniatis, Haiyang Wu, Kevin Monahan, Qiang Wu, Wei Li, Richard M. Myers, and Katherine E. Varley
- Subjects
CCCTC-Binding Factor ,Transcription, Genetic ,Cohesin complex ,Chromosomal Proteins, Non-Histone ,Protocadherin ,Cell Cycle Proteins ,Biology ,Models, Biological ,Cell Line ,Epigenesis, Genetic ,Humans ,Protein Isoforms ,Promoter Regions, Genetic ,Enhancer ,Neurons ,Genetics ,Stochastic Processes ,Multidisciplinary ,Models, Genetic ,Cohesin ,Nuclear Proteins ,Promoter ,DNA ,Biological Sciences ,Cadherins ,Phosphoproteins ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Gene Expression Regulation ,CTCF ,DNA methylation ,CpG Islands - Abstract
The closely linked human protocadherin ( Pcdh ) α, β, and γ gene clusters encode 53 distinct protein isoforms, which are expressed in a combinatorial manner to generate enormous diversity on the surface of individual neurons. This diversity is a consequence of stochastic promoter choice and alternative pre-mRNA processing. Here, we show that Pcdhα promoter choice is achieved by DNA looping between two downstream transcriptional enhancers and individual promoters driving the expression of alternate Pcdhα isoforms. In addition, we show that this DNA looping requires specific binding of the CTCF/cohesin complex to two symmetrically aligned binding sites in both the transcriptionally active promoters and in one of the enhancers. These findings have important implications regarding enhancer/promoter interactions in the generation of complex Pcdh cell surface codes for the establishment of neuronal identity and self-avoidance in individual neurons.
- Published
- 2012
- Full Text
- View/download PDF
26. Genistein and bisphenol A exposure cause estrogen receptor 1 to bind thousands of sites in a cell type-specific manner
- Author
-
Jason Gertz, Timothy E. Reddy, Katherine E. Varley, Richard M. Myers, and Michael J. Garabedian
- Subjects
medicine.medical_specialty ,Transcription, Genetic ,medicine.drug_class ,Estrogen receptor ,Genistein ,Phytoestrogens ,Biology ,chemistry.chemical_compound ,Phenols ,Cell Line, Tumor ,Internal medicine ,Gene expression ,Genetics ,medicine ,Humans ,Estrogens, Non-Steroidal ,Benzhydryl Compounds ,Gene ,Genetics (clinical) ,Estrogen receptor beta ,Regulation of gene expression ,Binding Sites ,Estradiol ,Genome, Human ,Sequence Analysis, RNA ,Research ,Estrogen Receptor alpha ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Endocrinology ,chemistry ,Genetic Loci ,Organ Specificity ,Estrogen ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists - Abstract
Endogenous estrogens that are synthesized in the body impact gene regulation by activating estrogen receptors in diverse cell types. Exogenous compounds that have estrogenic properties can also be found circulating in the blood in both children and adults. The genome-wide impact of these environmental estrogens on gene regulation is unclear. To obtain an integrated view of gene regulation in response to environmental and endogenous estrogens on a genome-wide scale, we performed ChIP-seq to identify estrogen receptor 1 (ESR1; previously estrogen receptor α) binding sites, and RNA-seq in endometrial cancer cells exposed to bisphenol A (BPA; found in plastics), genistein (GEN; found in soybean), or 17β-estradiol (E2; an endogenous estrogen). GEN and BPA treatment induces thousands of ESR1 binding sites and >50 gene expression changes, representing a subset of E2-induced gene regulation changes. Genes affected by E2 were highly enriched for ribosome-associated proteins; however, GEN and BPA failed to regulate most ribosome-associated proteins and instead enriched for transporters of carboxylic acids. Treatment-dependent changes in gene expression were associated with treatment-dependent ESR1 binding sites, with the exception that many genes up-regulated by E2 harbored a BPA-induced ESR1 binding site but failed to show any expression change after BPA treatment. GEN and BPA exhibited a similar relationship to E2 in the breast cancer line T-47D, where cell type specificity played a much larger role than treatment specificity. Overall, both environmental estrogens clearly regulate gene expression through ESR1 on a genome-wide scale, although with lower potency resulting in less ESR1 binding sites and less gene expression changes compared to the endogenous estrogen, E2.
- Published
- 2012
- Full Text
- View/download PDF
27. Transposase mediated construction of RNA-seq libraries
- Author
-
Scott Kuersten, Igor Y. Goryshin, Nicholas S. Davis, Jason Gertz, Richard M. Myers, Ramesh Vaidyanathan, Katherine E. Varley, and Bradley Baas
- Subjects
Transposable element ,DNA, Complementary ,Library ,genetic processes ,Transposases ,Method ,RNA-Seq ,Biology ,Deoxyribonuclease (Pyrimidine Dimer) ,Cell Line, Tumor ,Complementary DNA ,Genetics ,Humans ,natural sciences ,Genomic library ,RNA, Messenger ,Cloning, Molecular ,Uracil-DNA Glycosidase ,Genetics (clinical) ,Illumina dye sequencing ,Transposase ,Gene Library ,cDNA library ,Sequence Analysis, DNA ,DNA Transposable Elements - Abstract
RNA-seq has been widely adopted as a gene-expression measurement tool due to the detail, resolution, and sensitivity of transcript characterization that the technique provides. Here we present two transposon-based methods that efficiently construct high-quality RNA-seq libraries. We first describe a method that creates RNA-seq libraries for Illumina sequencing from double-stranded cDNA with only two enzymatic reactions. We generated high-quality RNA-seq libraries from as little as 10 pg of mRNA (∼1 ng of total RNA) with this approach. We also present a strand-specific RNA-seq library construction protocol that combines transposon-based library construction with uracil DNA glycosylase and endonuclease VIII to specifically degrade the second strand constructed during cDNA synthesis. The directional RNA-seq libraries maintain the same quality as the nondirectional libraries, while showing a high degree of strand specificity, such that 99.5% of reads map to the expected genomic strand. Each transposon-based library construction method performed well when compared with standard RNA-seq library construction methods with regard to complexity of the libraries, correlation between biological replicates, and the percentage of reads that align to the genome as well as exons. Our results show that high-quality RNA-seq libraries can be constructed efficiently and in an automatable fashion using transposition technology.
- Published
- 2011
- Full Text
- View/download PDF
28. Abstract LB-038: Predicting breast cancer therapy response using a patient-derived xenograft organoid screening platform
- Author
-
Sandra D. Scherer, Maihi Fujita, Alana L. Welm, Jason Gertz, Satya S. Pathi, James M. Graham, Katherine E. Varley, Yi Qiao, Yoko S. DeRose, Bryan E. Welm, Katrin P. Guillen, and Gabor T. Marth
- Subjects
Cancer Research ,Cancer ,Biology ,medicine.disease ,Breast cancer ,Therapy response ,Oncology ,Stroma ,Conditioned medium ,Cancer research ,medicine ,Organoid ,Doubling time ,Tumor xenograft - Abstract
Patient-derived xenografts (PDX) are valuable, clinically-relevant models of cancer. Their close genomic, phenotypic, and temporal association with patient tumors makes them well-suited for pre-clinical and co-clinical studies that assess the potential of new therapeutics. However, PDX models are not amenable to large-scale drug sensitivity studies due to their high cost and low throughput capacity. To address this, we established and characterized long-term PDX organoid (PDxO) cultures from breast cancer (BC) PDXs and evaluated their utility in therapeutic studies. To establish PDxO culture conditions, we extensively tested medium supplements, including growth factors, kinase inhibitors, conditioned medium, and antioxidants, along with extracellular matrix composition in 3D gels. Using optimized culture conditions for each BC subtype, we established PDxOs from 16 PDX lines in the HCI series, with a PDX to PDxO success rate of 85%. Around 20% of PDxOs contained aggressive mouse stroma, which had to be eliminated by FACS for long-term culture success. PDxOs were maintained for over 1 year, during which we tracked viability, doubling time, organoid size, genomics, epithelial character, and tumorigenicity. Doubling times stabilized after 60 days of initial culture, with a mean of 6.4±1.7 days for triple negative (TN) lines and 7.2±1 days for ER+ lines. Mean organoid size remained stable, with ER+ PDxOs significantly smaller than TN PDxOs, at 91 and 262 cells/organoid respectively (p=0.0012). PDxOs histologically resembled their PDX counterparts when stained for H&E, Vimentin, and CAM 5.2. RNA-Seq and copy-number variation analyses showed that PDxOs clustered with their PDXs and patient tumors. To assess if culturing affected tumorigenicity, we re-implanted PDxOs into mice following early and late passaging. 5/5 early passage and 5/6 late passage PDxOs engrafted and formed tumors. Resulting tumors showed similar gene expression profiles as their original PDXs by RNA-Seq. These data suggest that PDxOs generally recapitulate the molecular and genomic features of their originating PDXs and patient tumors. Having established PDxO cultures, we evaluated their utility as a therapeutic screening platform. We developed a screen to differentiate compounds with cytotoxic and cytostatic effects. NCI CTEP and clinically-approved BC therapies (n=40) were screened in quadruplicate 8-point dose response curves on all 16 PDxOs across three biological replicates. Drug response profiles were stable across biological replicates, spanning up to 1 year in culture. PDxOs exhibited diverse responses to therapies targeting cIAP1, PI3K, and CHK1/2. Initial work to evaluate concordance between PDxO and in vivo PDX responses returned trending linear correlations between PDxO dose response data and change in PDX growth rate following treatment (R2 = 0.52-0.77; p = 0.045-0.12, across 3 PDXs and 9 compounds). Ongoing work aims to confirm concordance between PDX and PDxO models. Our work demonstrates that PDxO models are cost-efficient, easy to maintain, and grow indefinitely - making them renewable and accessible cancer models. PDxOs are a powerful parallel resource to PDX models, especially useful for efficient determination of PDX drug response. We are currently expanding our PDxO bank to include 100 models which will be deposited with the NCI as part of the PDXNet effort. Citation Format: Katrin P. Guillen, Sandra D. Scherer, Yi Qiao, Satya S. Pathi, James M. Graham, Maihi Fujita, Yoko S. DeRose, Jason Gertz, Gabor T. Marth, Katherine E. Varley, Alana L. Welm, Bryan E. Welm. Predicting breast cancer therapy response using a patient-derived xenograft organoid screening platform [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 LB-038.
- Published
- 2018
- Full Text
- View/download PDF
29. Nested Patch PCR enables highly multiplexed mutation discovery in candidate genes
- Author
-
Katherine E. Varley and Robi D. Mitra
- Subjects
Candidate gene ,Genes, APC ,DNA Mutational Analysis ,Nonsense mutation ,Pilot Projects ,Single-nucleotide polymorphism ,Biology ,Polymerase Chain Reaction ,Polymorphism, Single Nucleotide ,Genome ,law.invention ,law ,Multiplex polymerase chain reaction ,Methods ,Genetics ,Humans ,Multiplex ,Genetics (clinical) ,Polymerase chain reaction ,DNA Primers ,Base Sequence ,Reproducibility of Results ,DNA, Neoplasm ,Exons ,genomic DNA ,Codon, Nonsense ,Colonic Neoplasms ,Mutation - Abstract
Medical resequencing of candidate genes in individual patient samples is becoming increasingly important in the clinic and in clinical research. Medical resequencing requires the amplification and sequencing of many candidate genes in many patient samples. Here we introduce Nested Patch PCR, a novel method for highly multiplexed PCR that is very specific, can sensitively detect SNPs and mutations, and is easy to implement. This is the first method that couples multiplex PCR with sample-specific DNA barcodes and next-generation sequencing to enable highly multiplex mutation discovery in candidate genes for multiple samples in parallel. In our pilot study, we amplified exons from colon cancer and matched normal human genomic DNA. From each sample, we successfully amplified 96% (90 of 94) targeted exons from across the genome, totaling 21.6 kbp of sequence. Ninety percent of all sequencing reads were from targeted exons, demonstrating that Nested Patch PCR is highly specific. We found that the abundance of reads per exon was reproducible across samples. We reliably detected germline SNPs and discovered a colon tumor specific nonsense mutation in APC, a gene causally implicated in colorectal cancer. With Nested Patch PCR, candidate gene mutation discovery across multiple individual patient samples can now utilize the power of second-generation sequencing.
- Published
- 2008
- Full Text
- View/download PDF
30. Abstract 1756: Genomic reclassification of endometrial carcinoma predicts drug response and facilitates refinement of current management based on PDX-guided efficacy outcomes
- Author
-
Margit M. Janát-Amsbury, Katherine E. Varley, Elke A. Jarboe, Matthew Peterson, David K. Gaffney, Chieh-Hsiang Yang, Jason Gertz, and Theresa L. Werner
- Subjects
Cancer Research ,Colorectal cancer ,business.industry ,Endometrial cancer ,Microsatellite instability ,Disease ,medicine.disease ,Bioinformatics ,Clinical trial ,Breast cancer ,Oncology ,Carcinoma ,medicine ,Copy-number variation ,business - Abstract
Endometrial cancer (EC) is the most common gynecological malignancy among American women. Steadily increasing numbers of newly diagnosed cases and deaths emphasize the necessity of improving current management strategies. Rather than the commonly known two types of EC based on histological classification, the Cancer Genome Atlas (TCGA) Research Network recently reported a reclassification that categorizes EC into four subtypes based on genomic characterization. Molecular similarities between these subtypes and other cancers, including colorectal, ovarian, and breast cancers, were identified. But to date, no prospective validation has delivered an improved understanding on how to translate these findings to the clinic in the form of treatments adapting genomic guidance. Objectives: To evaluate drug response according to TCGA classification using a panel of twenty-seven established endometrial cancer patient-derived xenografts (EC-PDXs). Methods: Twenty-seven EC-PDXs representing various stages of disease and histological subtypes were orthotopically transplanted and propagated over multiple generations. Upon thorough characterization including histology, metastatic status, and molecular features, EC-PDXs were further categorized into the four TCGA molecular subtypes on the basis of somatic copy number variations (CNV), microsatellite instability (MSI), and POLE mutations. A PDX clinical trial (PCT) was performed to re-evaluate current standard treatments in comparison to therapeutic agents adopted from treatment regimens for other cancer types based on their molecular similarities. Results: EC-PDXs established from patients with recurrent disease exhibited minimal activity to first-line chemotherapeutic drugs, as expected, but remained sensitive to other second-line drugs. None of the tested drugs exhibited sufficient activity in POLE EC-PDXs. MSI EC-PDXs were found to be less sensitive to platinum-based treatment and resistant to 5-FU as reported for MSI colorectal cancer. CNV-high EC-PDXs were more sensitive to drugs adopted from current ovarian and breast cancer treatment regimens. Conclusions: This is the first study that demonstrates the utility of TCGA classification for treatment guidance based on a PCT. Patients with recurrent and persistent disease are in desperate need of new therapeutic options. Our findings open up the possibility to utilize drugs which are currently used in the treatment of other cancers for the treatment of EC based on molecular similarities. In addition, this is the first study that successfully demonstrated the use of molecular classification to enable the prediction of drug response, and thus may facilitate the refinement of current EC management in a more precise and personalized way. Citation Format: Chieh-Hsiang Yang, David K. Gaffney, Theresa L. Werner, Elke A. Jarboe, Jason Gertz, Katherine E. Varley, Matthew Peterson, Margit M. Janat-Amsbury. Genomic reclassification of endometrial carcinoma predicts drug response and facilitates refinement of current management based on PDX-guided efficacy outcomes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1756. doi:10.1158/1538-7445.AM2017-1756
- Published
- 2017
- Full Text
- View/download PDF
31. Abstract 648: Integrated genomic characterization of endometrial cancer tumor grafts: a step toward genomic-guided treatment
- Author
-
Jason Gertz, Elke A. Jarboe, C. Matthew Peterson, Chieh-Hsiang Yang, Katherine E. Varley, and Margit M. Janát-Amsbury
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,business.industry ,Endometrial cancer ,Microsatellite instability ,Histology ,Disease ,medicine.disease ,Metastasis ,Clinical trial ,In vivo ,Internal medicine ,Medicine ,Histopathology ,business - Abstract
Endometrial cancer (EC) is the fourth most frequently diagnosed gynecological cancer among American Women. Steady increased numbers of newly diagnosed cases and deaths necessitates an added attention. Despite survival rates between 60-80% for localized, early stage disease, only marginal advances have been made in the treatment of distant (16%) and recurrent (15% of early stage) EC over the past two decades. New treatment strategies, guided by more predictive preclinical disease models incorporating molecular characteristics are crucial. Recent findings from The Cancer Genome Atlas (TCGA) Research Network suggested that the current histo-pathologically-guided classification of EC may not suffice to direct treatment recommendations, and introduced the potential adequacy of genomic profiling for reclassifying EC. However, these findings are hampered by the lack of appropriate in vivo models, which adequately facilitate obtaining the necessary preclinical evidence before translating findings to genome-guided clinical trials. Objectives: To establish patient derived orthotopic endometrial tumor grafts recapitulating histology, metastasis, and molecular characteristics of the original tumor specimen, and integrating TCGA genomic characterization to facilitate the development of genomic-guided treatment. Methods: Patient derived EC tissues were orthotopically transplanted to murine uteri and propagated over multiple generations. Generated tumor grafts were characterized for metastases, histopathology, and molecular profiles to ensure grafts resemblance of the original tumor samples. Tumor grafts were then further categorized into the four reported TCGA clusters on the basis of mutation frequency, somatic copy number alterations, microsatellite instability, and POLE mutation. Results: Twenty EC tumor grafts were successfully generated. Histological features, including tumor grade and hormone receptor status, as well as genomic profiles were maintained for up to 7 generations. Xenografts were capable to form metastasis closely parallel to clinical relevant sites. Results from a survival study demonstrated that the established tumor grafts reflected clinical stage-, and grade-based disease progression, and recapitulate TCGA cluster-based survival findings. Conclusions: This study reports the first successful generation of orthotopic EC patient-derived tumor grafts, which retain crucial histo-pathological characteristics, the capacity to form distant metastasis following known clinical patterns, as well as recapitulating molecular features of the original human tumor specimen. Additionally, our model can indeed be used as a tool to investigate the need for reclassifying EC into four clusters rather than two simplified disease subtypes and feasibility in directing genomic-guided treatment based on TCGA recommendations. Citation Format: Chieh-Hsiang Yang, Elke A. Jarboe, Jason Gertz, Katherine E. Varley, C. Matthew Peterson, Margit M. Janát-Amsbury. Integrated genomic characterization of endometrial cancer tumor grafts: a step toward genomic-guided treatment. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 648.
- Published
- 2016
- Full Text
- View/download PDF
32. Distinct properties of cell-type-specific and shared transcription factor binding sites
- Author
-
Richard M. Myers, Jason Gertz, Alexias Safi, Timothy E. Reddy, Katherine E. Varley, Gregory E. Crawford, E. Christopher Partridge, Daniel Savic, Gregory M. Cooper, and Preti Jain
- Subjects
Biology ,Response Elements ,Transfection ,Models, Biological ,Article ,Cell Line ,Evolution, Molecular ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Receptors, Glucocorticoid ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,Promoter Regions, Genetic ,Transcription factor ,Molecular Biology ,Conserved Sequence ,030304 developmental biology ,Regulation of gene expression ,Genetics ,0303 health sciences ,Binding Sites ,Estradiol ,Pioneer factor ,Estrogen Receptor alpha ,Estrogens ,Cell Biology ,DNA Methylation ,Chromatin ,Cell biology ,DNA binding site ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,DNA methylation ,Thermodynamics ,RNA Interference ,Estrogen receptor alpha ,Transcription Factors - Abstract
Most human transcription factors bind a small subset of potential genomic sites and often use different subsets in different cell types. To identify mechanisms that govern cell-type-specific transcription factor binding, we used an integrative approach to study estrogen receptor α (ER). We found that ER exhibits two distinct modes of binding. Shared sites, bound in multiple cell types, are characterized by high-affinity estrogen response elements (EREs), inaccessible chromatin, and a lack of DNA methylation, while cell-specific sites are characterized by a lack of EREs, co-occurrence with other transcription factors, and cell-type-specific chromatin accessibility and DNA methylation. These observations enabled accurate quantitative models of ER binding that suggest tethering of ER to one-third of cell-specific sites. The distinct properties of cell-specific binding were also observed with glucocorticoid receptor and for ER in primary mouse tissues, representing an elegant genomic encoding scheme for generating cell-type-specific gene regulation.
- Published
- 2012
33. Global Genomic Analysis of Prostate, Breast and Pancreatic Cancer
- Author
-
Richard M. Myers, Katherine E. Varley, and Marie K. Cross
- Subjects
Oncology ,CA15-3 ,PCA3 ,medicine.medical_specialty ,Cancer ,Biology ,medicine.disease ,Prostate cancer ,Breast cancer ,Internal medicine ,Pancreatic cancer ,DNA methylation ,medicine ,Survival rate - Abstract
Prostate cancer and breast cancer are the most prevalent cancers in men and women, respectively, and pancreatic cancer, while more rare than prostate and breast cancer, is an extremely lethal cancer, with a 5-year survival rate of less than 5%. With the recent advances in next generation sequencing technology, there is an opportunity to identify genomic aberrations that will provide knowledge about the biology driving these cancers and serve as novel diagnostic and prognostic biomarkers for these diseases. We performed genome-wide measurements of mRNA, microRNA, and DNA methylation from tumor and patient-matched non-tumor tissues. We have identified DNA methylation and gene expression signatures associated tumor formation, disease recurrence, and treatment sensitivity. These candidate biomarkers may be useful in predicting clinical outcomes for patients, and may suggest pathways that can be targeted with novel treatment regimens.
- Published
- 2012
- Full Text
- View/download PDF
34. Widespread plasticity in CTCF occupancy linked to DNA methylation
- Author
-
Hao Wang, Molly Weaver, Kristen Lee, Rajinder Kaul, Richard M. Myers, Florencia Pauli, Richard Sandstrom, Hongzhu Qu, Robert E. Thurman, Jason Gertz, John A. Stamatoyannopoulos, Theresa K. Canfield, Matthew T. Maurano, and Katherine E. Varley
- Subjects
Regulation of gene expression ,Genetics ,CCCTC-Binding Factor ,Chromatin Immunoprecipitation ,Binding Sites ,Research ,Bisulfite sequencing ,High-Throughput Nucleotide Sequencing ,Methylation ,Biology ,DNA Methylation ,Chromatin ,Cell Line ,Repressor Proteins ,CpG site ,Gene Expression Regulation ,CTCF ,DNA methylation ,Cluster Analysis ,Humans ,CpG Islands ,Chromatin immunoprecipitation ,Genetics (clinical) - Abstract
CTCF is a ubiquitously expressed regulator of fundamental genomic processes including transcription, intra- and interchromosomal interactions, and chromatin structure. Because of its critical role in genome function, CTCF binding patterns have long been assumed to be largely invariant across different cellular environments. Here we analyze genome-wide occupancy patterns of CTCF by ChIP-seq in 19 diverse human cell types, including normal primary cells and immortal lines. We observed highly reproducible yet surprisingly plastic genomic binding landscapes, indicative of strong cell-selective regulation of CTCF occupancy. Comparison with massively parallel bisulfite sequencing data indicates that 41% of variable CTCF binding is linked to differential DNA methylation, concentrated at two critical positions within the CTCF recognition sequence. Unexpectedly, CTCF binding patterns were markedly different in normal versus immortal cells, with the latter showing widespread disruption of CTCF binding associated with increased methylation. Strikingly, this disruption is accompanied by up-regulation of CTCF expression, with the result that both normal and immortal cells maintain the same average number of CTCF occupancy sites genome-wide. These results reveal a tight linkage between DNA methylation and the global occupancy patterns of a major sequence-specific regulatory factor.
- Published
- 2012
35. Analysis of DNA methylation in a three-generation family reveals widespread genetic influence on epigenetic regulation
- Author
-
Stephanie L. Parker, Huntington F. Willard, Katerina S. Kucera, Richard M. Myers, Kevin M. Bowling, Timothy E. Reddy, Katherine E. Varley, Jason Gertz, and Florencia Pauli
- Subjects
Male ,Cancer Research ,Heredity ,lcsh:QH426-470 ,Genomic imprinting ,Chromosomes, Human, Pair 21 ,Molecular Sequence Data ,Gene Expression ,Biology ,Polymorphism, Single Nucleotide ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Epigenetics of physical exercise ,Genetics ,Humans ,Epigenetics ,Gene Silencing ,Molecular Biology ,RNA-Directed DNA Methylation ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,Alleles ,030304 developmental biology ,0303 health sciences ,Chromosomes, Human, X ,Base Sequence ,Methylation ,Sequence Analysis, DNA ,Genomics ,DNA Methylation ,Pedigree ,Functional Genomics ,lcsh:Genetics ,Differentially methylated regions ,Reduced representation bisulfite sequencing ,DNA methylation ,CpG Islands ,Female ,DNA modification ,X chromosome inactivation ,030217 neurology & neurosurgery ,Chromosomes, Human, Pair 8 ,Research Article - Abstract
The methylation of cytosines in CpG dinucleotides is essential for cellular differentiation and the progression of many cancers, and it plays an important role in gametic imprinting. To assess variation and inheritance of genome-wide patterns of DNA methylation simultaneously in humans, we applied reduced representation bisulfite sequencing (RRBS) to somatic DNA from six members of a three-generation family. We observed that 8.1% of heterozygous SNPs are associated with differential methylation in cis, which provides a robust signature for Mendelian transmission and relatedness. The vast majority of differential methylation between homologous chromosomes (>92%) occurs on a particular haplotype as opposed to being associated with the gender of the parent of origin, indicating that genotype affects DNA methylation of far more loci than does gametic imprinting. We found that 75% of genotype-dependent differential methylation events in the family are also seen in unrelated individuals and that overall genotype can explain 80% of the variation in DNA methylation. These events are under-represented in CpG islands, enriched in intergenic regions, and located in regions of low evolutionary conservation. Even though they are generally not in functionally constrained regions, 22% (twice as many as expected by chance) of genes harboring genotype-dependent DNA methylation exhibited allele-specific gene expression as measured by RNA-seq of a lymphoblastoid cell line, indicating that some of these events are associated with gene expression differences. Overall, our results demonstrate that the influence of genotype on patterns of DNA methylation is widespread in the genome and greatly exceeds the influence of imprinting on genome-wide methylation patterns., Author Summary DNA methylation is a dynamic epigenetic mark that is essential for mammalian organismal development. DNA methylation levels can be influenced by environment, a chromosome's parental origin, and genome sequence. In this study, we evaluated the impact that DNA sequence has on DNA methylation by analyzing methylation levels in a three-generation family as well as unrelated individuals. By following DNA methylation patterns through the family along with nearby SNPs, we found that allelic differences between chromosomes play a much larger role in determining DNA methylation than the parental origin of the chromosome, indicating that DNA sequence has a larger impact on DNA methylation than gametic imprinting. We also found that allelic differences in DNA methylation found in the family can also be observed in unrelated individuals. In fact, the majority of variation in DNA methylation can be explained by genotype. Our results emphasize the importance of genome sequence in setting patterns of DNA methylation and indicate that genotype will need to be taken into account when assessing DNA methylation in the context of disease.
- Published
- 2011
36. Intra-tumor heterogeneity of MLH1 promoter methylation revealed by deep single molecule bisulfite sequencing
- Author
-
Robi D. Mitra, David G. Mutch, Paul J. Goodfellow, Katherine E. Varley, and Tina Bocker Edmonston
- Subjects
Bisulfite sequencing ,Biology ,Gene Regulation, Chromatin and Epigenetics ,medicine.disease_cause ,Polymerase Chain Reaction ,Epigenesis, Genetic ,03 medical and health sciences ,Endometrium ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Sulfites ,Epigenetics ,Allele ,Promoter Regions, Genetic ,Alleles ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,0303 health sciences ,Genetic heterogeneity ,Genetic Variation ,Nuclear Proteins ,Methylation ,Sequence Analysis, DNA ,DNA Methylation ,Molecular biology ,3. Good health ,Endometrial Neoplasms ,030220 oncology & carcinogenesis ,DNA methylation ,Illumina Methylation Assay ,Female ,Carcinogenesis ,MutL Protein Homolog 1 - Abstract
A single tumor may contain cells with different somatic mutations. By characterizing this genetic heterogeneity within tumors, advances have been made in the prognosis, treatment and understanding of tumorigenesis. In contrast, the extent of epigenetic intra-tumor heterogeneity and how it influences tumor biology is under-explored. We have characterized epigenetic heterogeneity within individual tumors using next-generation sequencing. We used deep single molecule bisulfite sequencing and sample-specific DNA barcodes to determine the spectrum of MLH1 promoter methylation across an average of 1000 molecules in each of 33 individual samples in parallel, including endometrial cancer, matched blood and normal endometrium. This first glimpse, deep into each tumor, revealed unexpectedly heterogeneous patterns of methylation at the MLH1 promoter within a subset of endometrial tumors. This high-resolution analysis allowed us to measure the clonality of methylation in individual tumors and gain insight into the accumulation of aberrant promoter methylation on both alleles during tumorigenesis.
- Published
- 2009
37. Quantification of rare allelic variants from pooled genomic DNA
- Author
-
F. Sessions Cole, Scott W. Doniger, Jacqueline A. Bonds, Justin C. Fay, Daniel J. Wegner, Katherine E. Varley, Olivia L Knowles, Aaron Hamvas, Sarah W Robison, Robi D. Mitra, Francesco Vallania, and Todd E. Druley
- Subjects
DNA nanoball sequencing ,Sequence analysis ,Molecular Sequence Data ,Biology ,Biochemistry ,Polymorphism, Single Nucleotide ,Sensitivity and Specificity ,Deep sequencing ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Genetic variation ,Molecular Biology ,Allele frequency ,Gene ,Exome sequencing ,030304 developmental biology ,Genetics ,0303 health sciences ,Base Sequence ,Chromosome Mapping ,Genetic Variation ,Reproducibility of Results ,Cell Biology ,DNA ,Sequence Analysis, DNA ,genomic DNA ,Sequence Alignment ,030217 neurology & neurosurgery ,Algorithms ,Software ,Biotechnology - Abstract
Rare germline variants are difficult to identify using traditional sequencing due to relatively high cost and low throughput. Using second-generation sequencing, we report a targeted, cost-effective method to quantify rare SNPs from pooled genomic DNA. We pooled DNA from 1,111 individuals and targeted four genes. Our novel base-calling algorithm, SNPSeeker, derived from Large Deviation theory, can detect SNPs present at frequencies below the raw error rate of the sequencing platform
- Published
- 2008
38. MHC II antigen presentation pathway expression in triple-negative breast cancer
- Author
-
Andres Forero-Torres, Donald J. Buchsbaum, Albert F. LoBuglio, Dongquan Chen, Todd C. Burwell, Yufeng Li, Richard M. Myers, William E. Grizzle, Katherine E. Varley, and Erinn Downs-Kelly
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Gene signature ,Log-rank test ,Internal medicine ,Gene expression ,Immunology ,medicine ,biology.protein ,Immunohistochemistry ,Progression-free survival ,Antibody ,business ,Gene ,Triple-negative breast cancer - Abstract
1066 Background: The purpose of this study was to identify gene expression differences between triple negative breast cancer (TNBC) tumors from patients who did or did not have disease relapse. Methods: 47 snap frozen macro-dissected primary TNBC tumors from treatment-naive patients, with an adequate follow up (greater than 24 months), were analyzed using RNA-seq to identify gene expression differences between patients with disease relapse and patients who did not relapse. In addition, archived de-identified primary TNBC tumors underwent standard immunohistochemical analysis with anti-CD74 and anti-HLA-DPB1 antibodies. Results: Database included 22 patients with disease relapse and 25 who did not relapse. 24 genes had significantly higher expression in tumor tissue from patients who did not relapse and 11 of these genes were integral members of the MHC II antigen presentation pathway. The 24 gene signature was significantly associated with progression free survival (PFS) (HR = 0.24; log rank p = 0.00016)....
- Published
- 2015
- Full Text
- View/download PDF
39. Abstract LB-412: TargetRich™ cancer gene panels: targeted next generation sequencing in cancer samples
- Author
-
Irina Vasenkova, Tatiana Shvetsova, Richard M. Myers, D. Troy Moore, Randall C. Bachmeyer, Katherine E. Varley, and Katherine J. Spayd
- Subjects
Genetics ,Cancer genome sequencing ,Cancer Research ,Cancer ,Ion semiconductor sequencing ,Biology ,medicine.disease ,DNA sequencing ,genomic DNA ,Exon ,Oncology ,medicine ,Cancer gene ,Gene - Abstract
Crucial to the adoption of next-generation sequencing within clinical cancer research is the ability to deeply sequence hundreds to thousands of targeted genomic regions across a large number of patient samples. Several targeting (PCR) and capture (hybridization) methods have been developed for next-generation sequencing, however these methods have limited utility with cancer patient specimens due to drawbacks such as large input DNA quantity requirements, wasted off-target sequencing, high cost of reagents, low sample throughput, specialized equipment, and inflexible content design. We developed TargetRich cancer gene panels employing Nested PatchPCR, which enables the targeted sequencing of hundreds of genomic regions in large numbers of patient samples to fully and efficiently utilize next-generation sequencers. Nested PatchPCR required only 250 ng genomic DNA, and was compatible with the fragmented DNA obtained from formalin fixed paraffin embedded samples (FFPE). No special equipment was needed, only a standard PCR machine. Patient samples were processed in parallel in a single 96-well plate. For each patient sample, we simultaneously amplified hundreds of exons from genes that are frequently mutated in cancer. We utilized standard library construction and barcoding methods to sequence targeted genes from 96 patient samples on a single Illumina GAIIx flowcell. The TargetRich cancer gene panels were highly specific; 95% of sequencing reads aligned to the targeted regions. The high percentage of on-target sequence data and focused content, allowed us to achieve high coverage across the loci (average of 400X read depth) for the sensitive detection of cancer mutations. We have created two cancer panels: CR (157 regions covering exons from 10 genes) and CS (760 regions covering exons from 62 genes) for use on the Illumina MiSeq, GAIIx & HiSeq platforms. Additionally, a CR panel for the Ion Torrent PGM has been developed with platform-specific primers added during the Nested PatchPCR reaction, allowing direct sequencing of products without time-consuming library construction. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-412. doi:1538-7445.AM2012-LB-412
- Published
- 2012
- Full Text
- View/download PDF
40. Nested Patch PCR for Highly Multiplexed Amplification of Genomic Loci
- Author
-
Katherine E. Varley and Robi D. Mitra
- Subjects
Mutation ,Models, Genetic ,Extramural ,Genomics ,Single-nucleotide polymorphism ,DNA ,Computational biology ,Biology ,medicine.disease_cause ,Polymerase Chain Reaction ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,genomic DNA ,Genetic Techniques ,law ,Polymorphism (computer science) ,Dna barcodes ,medicine ,Humans ,Polymerase chain reaction ,DNA Primers - Abstract
INTRODUCTIONNested Patch polymerase chain reaction (PCR) amplifies a large number (greater than 90) of targeted loci from genomic DNA simultaneously in the same reaction. These amplified loci can then be sequenced on a second-generation sequencing machine to detect single nucleotide polymorphisms (SNPs) and mutations. The reaction is highly specific: 90% of sequencing reads match targeted loci. Nested Patch PCR can be performed on many samples in parallel, and by using sample-specific DNA barcodes, these can be pooled and sequenced in a single reaction. Thus, the Nested Patch PCR protocol that is described here provides an easy workflow to identify SNPs and mutations across many targeted loci for many samples in parallel.
- Published
- 2009
- Full Text
- View/download PDF
41. Evidence for Active Maintenance of Inverted Repeat Structures Identified by a Comparative Genomic Approach
- Author
-
Guoyan Zhao, Gary D. Stormo, Katherine E. Varley, and Kuan Y. Chang
- Subjects
Genetic Speciation ,Inverted Repeat Sequences ,Inverted repeat ,lcsh:Medicine ,Sequence alignment ,Computational Biology/Comparative Sequence Analysis ,Biology ,Genome ,Genomic Instability ,Evolution, Molecular ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,Gene Duplication ,Sequence Homology, Nucleic Acid ,Animals ,Direct repeat ,Gene conversion ,Insertion sequence ,Caenorhabditis elegans ,lcsh:Science ,Phylogeny ,030304 developmental biology ,Genetics ,Comparative genomics ,Comparative Genomic Hybridization ,0303 health sciences ,Multidisciplinary ,lcsh:R ,DNA, Helminth ,Evolutionary biology ,Caenorhabditis ,DNA, Intergenic ,lcsh:Q ,Genetics and Genomics/Comparative Genomics ,Sequence Alignment ,030217 neurology & neurosurgery ,Research Article - Abstract
Inverted repeats have been found to occur in both prokaryotic and eukaryotic genomes. Usually they are short and some have important functions in various biological processes. However, long inverted repeats are rare and can cause genome instability. Analyses of C. elegans genome identified long, nearly-perfect inverted repeat sequences involving both divergently and convergently oriented homologous gene pairs and complete intergenic sequences. Comparisons with the orthologous regions from the genomes of C. briggsae and C. remanei show that the inverted repeat structures are often far more conserved than the sequences. This observation implies that there is an active mechanism for maintaining the inverted repeat nature of the sequences.
- 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.