Kouros Owzar

Overview:

cancer pharmacogenomics
drug induced neuropathy, neutropenia and hypertension
statistical genetics
statistical methods for high-dimensional data
copulas
survival analysis
statistical computing

Positions:

Professor of Biostatistics & Bioinformatics

Biostatistics & Bioinformatics
School of Medicine

Director, DCI Bioinformatics

Biostatistics & Bioinformatics
School of Medicine

Member of the Duke Cancer Institute

Duke Cancer Institute
School of Medicine

Education:

Ph.D. 2002

University of North Carolina - Chapel Hill

Grants:

Preoperative Breast Radiotherapy: A Tool to Provide Individualized and Biologically-Based Radiation Therapy

Administered By
Radiation Oncology
Awarded By
Gateway for Cancer Research
Role
Collaborator
Start Date
End Date

mTOR Therapy in Prostate Cancer: Signatures of Response and Biology of Resistance

Administered By
Institutes and Centers
Awarded By
National Institutes of Health
Role
Statistician
Start Date
End Date

Health Disparity in African Americans: A Meta-analysis of Six Phase III Trials in Metastatic Castration-Resistant Prostate Cancer Men treated with Docetaxel

Administered By
Biostatistics & Bioinformatics
Awarded By
Department of Defense
Role
Collaborator
Start Date
End Date

Profiling the Adenosine Axis in Metastatic Colorectal Cancer

Administered By
Medicine, Medical Oncology
Awarded By
MedImmune, Inc.
Role
Statistician
Start Date
End Date

Computational Resources and Dissemination Core

Administered By
Biostatistics & Bioinformatics
Awarded By
University of North Carolina - Chapel Hill
Role
Principal Investigator
Start Date
End Date

Publications:

Clustering Deviation Index (CDI): a robust and accurate internal measure for evaluating scRNA-seq data clustering.

Most single-cell RNA sequencing (scRNA-seq) analyses begin with cell clustering; thus, the clustering accuracy considerably impacts the validity of downstream analyses. In contrast with the abundance of clustering methods, the tools to assess the clustering accuracy are limited. We propose a new Clustering Deviation Index (CDI) that measures the deviation of any clustering label set from the observed single-cell data. We conduct in silico and experimental scRNA-seq studies to show that CDI can select the optimal clustering label set. As a result, CDI also informs the optimal tuning parameters for any given clustering method and the correct number of cluster components.
Authors
Fang, J; Chan, C; Owzar, K; Wang, L; Qin, D; Li, Q-J; Xie, J
MLA Citation
Fang, Jiyuan, et al. “Clustering Deviation Index (CDI): a robust and accurate internal measure for evaluating scRNA-seq data clustering.Genome Biol, vol. 23, no. 1, Dec. 2022, p. 269. Pubmed, doi:10.1186/s13059-022-02825-5.
URI
https://scholars.duke.edu/individual/pub1560494
PMID
36575517
Source
pubmed
Published In
Genome Biology
Volume
23
Published Date
Start Page
269
DOI
10.1186/s13059-022-02825-5

Molecular classification and biomarkers of clinical outcome in breast ductal carcinoma in situ: Analysis of TBCRC 038 and RAHBT cohorts.

Ductal carcinoma in situ (DCIS) is the most common precursor of invasive breast cancer (IBC), with variable propensity for progression. We perform multiscale, integrated molecular profiling of DCIS with clinical outcomes by analyzing 774 DCIS samples from 542 patients with 7.3 years median follow-up from the Translational Breast Cancer Research Consortium 038 study and the Resource of Archival Breast Tissue cohorts. We identify 812 genes associated with ipsilateral recurrence within 5 years from treatment and develop a classifier that predicts DCIS or IBC recurrence in both cohorts. Pathways associated with recurrence include proliferation, immune response, and metabolism. Distinct stromal expression patterns and immune cell compositions are identified. Our multiscale approach employed in situ methods to generate a spatially resolved atlas of breast precancers, where complementary modalities can be directly compared and correlated with conventional pathology findings, disease states, and clinical outcome.
Authors
Strand, SH; Rivero-Gutiérrez, B; Houlahan, KE; Seoane, JA; King, LM; Risom, T; Simpson, LA; Vennam, S; Khan, A; Cisneros, L; Hardman, T; Harmon, B; Couch, F; Gallagher, K; Kilgore, M; Wei, S; DeMichele, A; King, T; McAuliffe, PF; Nangia, J; Lee, J; Tseng, J; Storniolo, AM; Thompson, AM; Gupta, GP; Burns, R; Veis, DJ; DeSchryver, K; Zhu, C; Matusiak, M; Wang, J; Zhu, SX; Tappenden, J; Ding, DY; Zhang, D; Luo, J; Jiang, S; Varma, S; Anderson, L; Straub, C; Srivastava, S; Curtis, C; Tibshirani, R; Angelo, RM; Hall, A; Owzar, K; Polyak, K; Maley, C; Marks, JR; Colditz, GA; Hwang, ES; West, RB
MLA Citation
Strand, Siri H., et al. “Molecular classification and biomarkers of clinical outcome in breast ductal carcinoma in situ: Analysis of TBCRC 038 and RAHBT cohorts.Cancer Cell, vol. 40, no. 12, Dec. 2022, pp. 1521-1536.e7. Pubmed, doi:10.1016/j.ccell.2022.10.021.
URI
https://scholars.duke.edu/individual/pub1556629
PMID
36400020
Source
pubmed
Published In
Cancer Cell
Volume
40
Published Date
Start Page
1521
End Page
1536.e7
DOI
10.1016/j.ccell.2022.10.021

Leveraging patient derived models of FGFR2 fusion positive intrahepatic cholangiocarcinoma to identify synergistic therapies.

Intrahepatic cholangiocarcinoma (ICC) remains a deadly malignancy lacking systemic therapies for advanced disease. Recent advancements include selective FGFR1-3 inhibitors for the 15% of ICC patients harboring fusions, although survival is limited by poor response and resistance. Herein we report generation of a patient-derived FGFR2 fusion-positive ICC model system consisting of a cell line, organoid, and xenograft, which have undergone complete histologic, genomic, and phenotypic characterization, including testing standard-of-care systemic therapies. Using these FGFR2 fusion-positive ICC models, we conducted an unbiased high-throughput small molecule screen to prioritize combination strategies with FGFR inhibition, from which HDAC inhibition together with pemigatinib was validated in vitro and in vivo as a synergistic therapy for ICC. Additionally, we demonstrate broad utility of the FGFR/HDAC combination for other FGFR fusion-positive solid tumors. These data are directly translatable and justify early phase trials to establish dosing, safety, and therapeutic efficacy of this synergistic combination.
Authors
Lidsky, ME; Wang, Z; Lu, M; Liu, A; Hsu, SD; McCall, SJ; Sheng, Z; Granek, JA; Owzar, K; Anderson, KS; Wood, KC
MLA Citation
Lidsky, Michael E., et al. “Leveraging patient derived models of FGFR2 fusion positive intrahepatic cholangiocarcinoma to identify synergistic therapies.Npj Precis Oncol, vol. 6, no. 1, Oct. 2022, p. 75. Pubmed, doi:10.1038/s41698-022-00320-5.
URI
https://scholars.duke.edu/individual/pub1554318
PMID
36274097
Source
pubmed
Published In
Npj Precis Oncol
Volume
6
Published Date
Start Page
75
DOI
10.1038/s41698-022-00320-5

Correction: Bevacizumab-induced hypertension and proteinuria: a genome-wide study of more than 1000 patients.

Authors
Quintanilha, JCF; Wang, J; Sibley, AB; Jiang, C; Etheridge, AS; Shen, F; Jiang, G; Mulkey, F; Patel, JN; Hertz, DL; Dees, EC; McLeod, HL; Bertagnolli, M; Rugo, H; Kindler, HL; Kelly, WK; Ratain, MJ; Kroetz, DL; Owzar, K; Schneider, BP; Lin, D; Innocenti, F
MLA Citation
Quintanilha, Julia C. F., et al. “Correction: Bevacizumab-induced hypertension and proteinuria: a genome-wide study of more than 1000 patients.Br J Cancer, vol. 126, no. 1, Jan. 2022, p. 162. Pubmed, doi:10.1038/s41416-021-01617-1.
URI
https://scholars.duke.edu/individual/pub1502770
PMID
34853435
Source
pubmed
Published In
Br J Cancer
Volume
126
Published Date
Start Page
162
DOI
10.1038/s41416-021-01617-1

Plasma Protein Biomarkers in Advanced or Metastatic Colorectal Cancer Patients Receiving Chemotherapy With Bevacizumab or Cetuximab: Results from CALGB 80405 (Alliance).

PURPOSE: CALGB 80405 compared the combination of first-line chemotherapy with cetuximab or bevacizumab in the treatment of advanced or metastatic colorectal cancer (mCRC). Although similar clinical outcomes were observed in the cetuximab-chemotherapy group and the bevacizumab-chemotherapy group, biomarkers could identify patients deriving more benefit from either biologic agent. PATIENTS AND METHODS: In this exploratory analysis, the Angiome, a panel of 24 soluble protein biomarkers were measured in baseline plasma samples in CALGB 80405. Prognostic biomarkers were determined using univariate Cox proportional hazards models. Predictive biomarkers were identified using multivariable Cox regression models including interaction between biomarker level and treatment. RESULTS: In the total population, high plasma levels of Ang-2, CD73, HGF, ICAM-1, IL6, OPN, TIMP-1, TSP-2, VCAM-1, and VEGF-R3 were identified as prognostic of worse progression-free survival (PFS) and overall survival (OS). PlGF was identified as predictive of lack of PFS benefit from bevacizumab [bevacizumab HR, 1.51; 95% confidence interval (CI), 1.10-2.06; cetuximab HR, 0.94; 95% CI, 0.71-1.25; Pinteraction = 0.0298] in the combined FOLFIRI/FOLFOX regimens. High levels of VEGF-D were predictive of lack of PFS benefit from bevacizumab in patients receiving FOLFOX regimen only (FOLFOX/bevacizumab HR, 1.70; 95% CI, 1.19-2.42; FOLFOX/cetuximab HR, 0.92; 95% CI, 0.68-1.24; Pinteraction = 0.0097). CONCLUSIONS: In this exploratory, hypothesis-generating analysis, the Angiome identified multiple prognostic biomarkers and two potential predictive biomarkers for patients with mCRC enrolled in CALGB 80405. PlGF and VEGF-D predicted lack of benefit from bevacizumab in a chemo-dependent manner. See related commentaries by Mishkin and Kohn, p. 2722 and George and Bertagnolli, p. 2725.
Authors
Nixon, AB; Sibley, AB; Liu, Y; Hatch, AJ; Jiang, C; Mulkey, F; Starr, MD; Brady, JC; Niedzwiecki, D; Venook, AP; Baez-Diaz, L; Lenz, H-J; O'Neil, BH; Innocenti, F; Meyerhardt, JA; O'Reilly, EM; Owzar, K; Hurwitz, HI
MLA Citation
Nixon, Andrew B., et al. “Plasma Protein Biomarkers in Advanced or Metastatic Colorectal Cancer Patients Receiving Chemotherapy With Bevacizumab or Cetuximab: Results from CALGB 80405 (Alliance).Clin Cancer Res, vol. 28, no. 13, July 2022, pp. 2779–88. Pubmed, doi:10.1158/1078-0432.CCR-21-2389.
URI
https://scholars.duke.edu/individual/pub1504729
PMID
34965954
Source
pubmed
Published In
Clinical Cancer Research
Volume
28
Published Date
Start Page
2779
End Page
2788
DOI
10.1158/1078-0432.CCR-21-2389