68 results on '"James S. Hardwick"'
Search Results
2. Data from Downregulation of MHC-I Expression Is Prevalent but Reversible in Merkel Cell Carcinoma
- Author
-
Paul Nghiem, Jürgen C. Becker, Michele A. Cleary, Mary L. Disis, James S. Hardwick, Shigetoshi Sano, Hideki Nakajima, Shailender Bhatia, Margaret Madeleine, David M. Koelle, Aaron Seo, William T. Simonson, Kotaro Nagase, Renee Thibodeau, Shinichi Koba, David Schrama, Olga K. Afanasiev, Jayasri G. Iyer, Christoph Willmes, Andrew Tegeder, and Kelly G. Paulson
- Abstract
Merkel cell carcinoma (MCC) is an aggressive, polyomavirus-associated skin cancer. Robust cellular immune responses are associated with excellent outcomes in patients with MCC, but these responses are typically absent. We determined the prevalence and reversibility of major histocompatibility complex class I (MHC-I) downregulation in MCC, a potentially reversible immune-evasion mechanism. Cell-surface MHC-I expression was assessed on five MCC cell lines using flow cytometry as well as immunohistochemistry on tissue microarrays representing 114 patients. Three additional patients were included who had received intralesional IFN treatment and had evaluable specimens before and after treatment. mRNA expression analysis of antigen presentation pathway genes from 35 MCC tumors was used to examine the mechanisms of downregulation. Of note, 84% of MCCs (total n = 114) showed reduced MHC-I expression as compared with surrounding tissues, and 51% had poor or undetectable MHC-I expression. Expression of MHC-I was lower in polyomavirus-positive MCCs than in polyomavirus-negative MCCs (P < 0.01). The MHC-I downregulation mechanism was multifactorial and did not depend solely on HLA gene expression. Treatment of MCC cell lines with ionizing radiation, etoposide, or IFN resulted in MHC-I upregulation, with IFNs strongly upregulating MHC-I expression in vitro, and in 3 of 3 patients treated with intralesional IFNs. MCC tumors may be amenable to immunotherapy, but downregulation of MHC-I is frequently present in these tumors, particularly those that are positive for polyomavirus. This downregulation is reversible with any of several clinically available treatments that may thus promote the effectiveness of immune-stimulating therapies for MCC. Cancer Immunol Res; 2(11); 1071–9. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Identification of biomarkers for tumor endothelial cell proliferation through gene expression profiling
- Author
-
Laura Sepp-Lorenzino, John R. Lamb, Michael E. Severino, Nancy E. Kohl, Edward J. Weinstein, Robert L. Phillips, Robert Wasserman, Hongyue Dai, Susan L. Hill, Elizabeth J. Koury, Rosemary McFall, Bin Shi, Chunsheng Zhang, Yi Yang, and James S. Hardwick
- Abstract
Extensive efforts are under way to identify antiangiogenic therapies for the treatment of human cancers. Many proposed therapeutics target vascular endothelial growth factor (VEGF) or the kinase insert domain receptor (KDR/VEGF receptor-2/FLK-1), the mitogenic VEGF receptor tyrosine kinase expressed by endothelial cells. Inhibition of KDR catalytic activity blocks tumor neoangiogenesis, reduces vascular permeability, and, in animal models, inhibits tumor growth and metastasis. Using a gene expression profiling strategy in rat tumor models, we identified a set of six genes that are selectively overexpressed in tumor endothelial cells relative to tumor cells and whose pattern of expression correlates with the rate of tumor endothelial cell proliferation. In addition to being potential targets for antiangiogenesis tumor therapy, the expression patterns of these genes or their protein products may aid the development of pharmacodynamic assays for small molecule inhibitors of the KDR kinase in human tumors.
- Published
- 2023
- Full Text
- View/download PDF
4. Data from A Phase I Pharmacokinetic and Pharmacodynamic Study of Dalotuzumab (MK-0646), an Anti-Insulin-like Growth Factor-1 Receptor Monoclonal Antibody, in Patients with Advanced Solid Tumors
- Author
-
José Baselga, Ronald B. Langdon, Susana Roselló, Emiliano Calvo, Karl Hsu, William D. Hanley, Robert A. Beckman, James S. Hardwick, Jason Clark, Holly Brown, Serena Di Cosimo, Jordi Rodon, Cristina Gamez, Jorge Guijarro, Amparo Domingo, Edith Rodríguez-Braun, Jordi Andreu, Ludmila Prudkin, Andrés Cervantes, Josep Tabernero, and Francesco Atzori
- Abstract
Purpose: Insulin-like growth factor-1 receptor (IGF-1R) mediates cellular processes in cancer and has been proposed as a therapeutic target. Dalotuzumab (MK-0646) is a humanized IgG1 monoclonal antibody that binds to IGF-1R preventing receptor activation. This study was designed to evaluate the safety and tolerability of dalotuzumab, determine the pharmacokinetic (PK) and pharmacodynamic (PD) profiles, and identify a recommended phase II dose.Experimental Design: Patients with tumors expressing IGF-1R protein were allocated to dose-escalating cohorts of three or more patients each and received intravenous dalotuzumab weekly, every 2 or 3 weeks. Plasma was collected for PK analysis. Paired baseline and on-treatment skin and tumor biopsy samples were collected for PD analyses.Results: Eighty patients with chemotherapy-refractory solid tumors were enrolled. One dose-limiting toxicity was noted, but a maximum-tolerated dose was not identified. Grade 1 to 3 hyperglycemia, responsive to metformin, occurred in 15 (19%) patients. At dose levels or more than 5 mg/kg, dalotuzumab mean terminal half-life was 95 hours or more, mean Cmin was more than 25 μg/mL, clearance was constant, and serum exposures were approximately dose proportional. Decreases in tumor IGF-1R, downstream receptor signaling, and Ki67 expression were observed. 18F-Fluorodeoxy-glucose positron emission tomography metabolic responses occurred in three patients. One patient with Ewing's sarcoma showed a mixed radiologic response. The recommended phase II doses were 10, 20, and 30 mg/kg for the weekly, every other week, and every third week schedules, respectively.Conclusions: Dalotuzumab was generally well-tolerated, exhibited dose-proportional PK, inhibited IGF-1R pathway signaling and cell proliferation in treated tumors, and showed clinical activity. The low clearance rate and long terminal half-life support more extended dosing intervals. Clin Cancer Res; 17(19); 6304–12. ©2011 AACR.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 1 from A Phase I Pharmacokinetic and Pharmacodynamic Study of Dalotuzumab (MK-0646), an Anti-Insulin-like Growth Factor-1 Receptor Monoclonal Antibody, in Patients with Advanced Solid Tumors
- Author
-
José Baselga, Ronald B. Langdon, Susana Roselló, Emiliano Calvo, Karl Hsu, William D. Hanley, Robert A. Beckman, James S. Hardwick, Jason Clark, Holly Brown, Serena Di Cosimo, Jordi Rodon, Cristina Gamez, Jorge Guijarro, Amparo Domingo, Edith Rodríguez-Braun, Jordi Andreu, Ludmila Prudkin, Andrés Cervantes, Josep Tabernero, and Francesco Atzori
- Abstract
PDF file - 85K
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 3 from A Phase I Pharmacokinetic and Pharmacodynamic Study of Dalotuzumab (MK-0646), an Anti-Insulin-like Growth Factor-1 Receptor Monoclonal Antibody, in Patients with Advanced Solid Tumors
- Author
-
José Baselga, Ronald B. Langdon, Susana Roselló, Emiliano Calvo, Karl Hsu, William D. Hanley, Robert A. Beckman, James S. Hardwick, Jason Clark, Holly Brown, Serena Di Cosimo, Jordi Rodon, Cristina Gamez, Jorge Guijarro, Amparo Domingo, Edith Rodríguez-Braun, Jordi Andreu, Ludmila Prudkin, Andrés Cervantes, Josep Tabernero, and Francesco Atzori
- Abstract
PDF file - 301K
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S1 from Identification of biomarkers for tumor endothelial cell proliferation through gene expression profiling
- Author
-
Laura Sepp-Lorenzino, John R. Lamb, Michael E. Severino, Nancy E. Kohl, Edward J. Weinstein, Robert L. Phillips, Robert Wasserman, Hongyue Dai, Susan L. Hill, Elizabeth J. Koury, Rosemary McFall, Bin Shi, Chunsheng Zhang, Yi Yang, and James S. Hardwick
- Abstract
Supplementary Table S1 from Identification of biomarkers for tumor endothelial cell proliferation through gene expression profiling
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figures 1-5, Tables 1-2 from Inhibition of NOTCH Signaling by Gamma Secretase Inhibitor Engages the RB Pathway and Elicits Cell Cycle Exit in T-Cell Acute Lymphoblastic Leukemia Cells
- Author
-
Christopher G. Winter, Peter R. Strack, A. Thomas Look, Giulio F. Draetta, Pamela M. Carroll, Lex H.T. Van der Ploeg, Victoria M. Richon, Nancy E. Kohl, Jing Yuan, Edyta Tyminski, Theresa Zhang, Xudong Dai, James S. Hardwick, Cole D. Liberator, Jennifer O'Neil, and Sudhir S. Rao
- Abstract
Supplementary Figures 1-5, Tables 1-2 from Inhibition of NOTCH Signaling by Gamma Secretase Inhibitor Engages the RB Pathway and Elicits Cell Cycle Exit in T-Cell Acute Lymphoblastic Leukemia Cells
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Inhibition of NOTCH Signaling by Gamma Secretase Inhibitor Engages the RB Pathway and Elicits Cell Cycle Exit in T-Cell Acute Lymphoblastic Leukemia Cells
- Author
-
Christopher G. Winter, Peter R. Strack, A. Thomas Look, Giulio F. Draetta, Pamela M. Carroll, Lex H.T. Van der Ploeg, Victoria M. Richon, Nancy E. Kohl, Jing Yuan, Edyta Tyminski, Theresa Zhang, Xudong Dai, James S. Hardwick, Cole D. Liberator, Jennifer O'Neil, and Sudhir S. Rao
- Abstract
NOTCH signaling is deregulated in the majority of T-cell acute lymphoblastic leukemias (T-ALL) as a result of activating mutations in NOTCH1. Gamma secretase inhibitors (GSI) block proteolytic activation of NOTCH receptors and may provide a targeted therapy for T-ALL. We have investigated the mechanisms of GSI sensitivity across a panel of T-ALL cell lines, yielding an approach for patient stratification based on pathway activity and also providing a rational combination strategy for enhanced response to GSI. Whereas the NOTCH1 mutation status does not serve as a predictor of GSI sensitivity, a gene expression signature of NOTCH pathway activity does correlate with response, and may be useful in the selection of patients more likely to respond to GSI. Furthermore, inhibition of the NOTCH pathway activity signature correlates with the induction of the cyclin-dependent kinase inhibitors CDKN2D (p19INK4d) and CDKN1B (p27Kip1), leading to derepression of RB and subsequent exit from the cell cycle. Consistent with this evidence of cell cycle exit, short-term exposure of GSI resulted in sustained molecular and phenotypic effects after withdrawal of the compound. Combination treatment with GSI and a small molecule inhibitor of CDK4 produced synergistic growth inhibition, providing evidence that GSI engagement of the CDK4/RB pathway is an important mechanism of GSI action and supports further investigation of this combination for improved efficacy in treating T-ALL. [Cancer Res 2009;69(7):3060–8]
- Published
- 2023
- Full Text
- View/download PDF
10. Preclinical Evaluation of 89Zr-Df-IAB22M2C PET as an Imaging Biomarker for the Development of the GUCY2C-CD3 Bispecific PF-07062119 as a T Cell Engaging Therapy
- Author
-
Eve H. Pickering, Keith Kobylarz, Jianqing Chen, Jonathan Golas, Ohad Ilovich, Lindsay King, Sripad Ram, Kevin P. Maresca, Timothy J. McCarthy, Laigao Chen, James S. Hardwick, Amira Hussein, Jatin Narula, Edward Rosfjord, Anand Giddabasappa, Edmund J. Keliher, Ian D. Wilson, Adam Root, David Schaer, Divya Mathur, Paul A. Rejto, and Kevin Staton
- Subjects
Cancer Research ,Adoptive cell transfer ,T cell ,T-Lymphocytes ,89Zr-IAB22M2C PET imaging ,Receptors, Enterotoxin ,Standardized uptake value ,Mice, SCID ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,medicine ,Cytotoxic T cell ,Animals ,Radiology, Nuclear Medicine and imaging ,030304 developmental biology ,0303 health sciences ,business.industry ,GUCY2C bispecific antibody ,Immuno-oncology ,Imaging agent ,medicine.anatomical_structure ,Oncology ,CD8 T cell ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,Cancer research ,Zirconium ,business ,Preclinical imaging ,Ex vivo ,CD8 ,Biomarkers ,Research Article - Abstract
Purpose A sensitive and specific imaging biomarker to monitor immune activation and quantify pharmacodynamic responses would be useful for development of immunomodulating anti-cancer agents. PF-07062119 is a T cell engaging bispecific antibody that binds to CD3 and guanylyl cyclase C, a protein that is over-expressed by colorectal cancers. Here, we used 89Zr-Df-IAB22M2C (89Zr-Df-Crefmirlimab), a human CD8-specific minibody to monitor CD8+ T cell infiltration into tumors by positron emission tomography. We investigated the ability of 89Zr-Df-IAB22M2C to track anti-tumor activity induced by PF-07062119 in a human CRC adoptive transfer mouse model (with injected activated/expanded human T cells), as well as the correlation of tumor radiotracer uptake with CD8+ immunohistochemical staining. Procedures NOD SCID gamma mice bearing human CRC LS1034 tumors were treated with four different doses of PF-07062119, or a non-targeted CD3 BsAb control, and imaged with 89Zr-Df-IAB22M2C PET at days 4 and 9. Following PET/CT imaging, mice were euthanized and dissected for ex vivo distribution analysis of 89Zr-Df-IAB22M2C in tissues on days 4 and 9, with additional data collected on day 6 (supplementary). Data were analyzed and reported as standard uptake value and %ID/g for in vivo imaging and ex vivo tissue distribution. In addition, tumor tissues were evaluated by immunohistochemistry for CD8+ T cells. Results The results demonstrated substantial mean uptake of 89Zr-Df-IAB22M2C (%ID/g) in PF-07062119-treated tumors, with significant increases in comparison to non-targeted BsAb-treated controls, as well as PF-07062119 dose-dependent responses over time of treatment. A moderate correlation was observed between tumor tissue radioactivity uptake and CD8+ cell density, demonstrating the value of the imaging agent for non-invasive assessment of intra-tumoral CD8+ T cells and the mechanism of action for PF-07062119. Conclusion Immune-imaging technologies for quantitative cellular measures would be a valuable biomarker in immunotherapeutic clinical development. We demonstrated a qualification of 89Zr-IAB22M2C PET to evaluate PD responses (mice) to a novel immunotherapeutic.
- Published
- 2021
11. Combining CDK4/6 inhibition with taxanes enhances anti-tumor efficacy by sustained impairment of pRB-E2F pathways in squamous cell lung cancer
- Author
-
James S. Hardwick, Ping Wei, Joan Cao, Hui Wang, Todd VanArsdale, Scott L. Weinrich, Paul A. Rejto, Shibing Deng, Goldie Y. L. Lui, Zhou Zhu, and Timothy Nichols
- Subjects
Bridged-Ring Compounds ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Cell cycle checkpoint ,Pyridines ,medicine.medical_treatment ,Gene Expression ,Mice, Nude ,Angiogenesis Inhibitors ,Palbociclib ,Biology ,Retinoblastoma Protein ,Piperazines ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Human Umbilical Vein Endothelial Cells ,Genetics ,medicine ,Animals ,Humans ,E2F ,Molecular Biology ,Cell Proliferation ,Mice, Inbred BALB C ,Taxane ,Neovascularization, Pathologic ,Retinoblastoma ,Cyclin-Dependent Kinase 4 ,Cancer ,Cyclin-Dependent Kinase 6 ,Immunotherapy ,medicine.disease ,E2F Transcription Factors ,G2 Phase Cell Cycle Checkpoints ,030104 developmental biology ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Cancer research ,Female ,Taxoids ,Signal Transduction - Abstract
The CDK4/6 inhibitor palbociclib reduces tumor growth by decreasing retinoblastoma (RB) protein phosphorylation and inducing cell cycle arrest at the G1/S phase transition. Palbociclib in combination with anti-hormonal therapy brings significant benefit to breast cancer patients. In this study, novel combination approaches and underlying molecular/cellular mechanisms for palbociclib were explored in squamous cell lung cancer (SqCLC), the second most common subtype of non-small cell lung cancer. While approximate 20% lung patients benefit from immunotherapy, most SqCLC patients who receive platinum-doublet chemotherapy as first-line treatment, which often includes a taxane, are still in need of more effective combination therapies. Our results demonstrated enhanced cytotoxicity and anti-tumor effect with palbociclib plus taxanes at clinically achievable doses in multiple SqCLC models with diverse cancer genetic backgrounds. Comprehensive gene expression analysis revealed a sustained disruption of pRB-E2F signaling by combination that was accompanied with enhanced regulation of pleiotropic biological effects. These included several novel mechanisms such as abrogation of G2/M and mitotic spindle assembly checkpoints, as well as impaired induction of hypoxia-inducible factor 1 alpha (HIF-1α). The decrease in HIF-1α modulated a couple key angiogenic and anti-angiogenic factors, resulting in an enhanced anti-angiogenic effect. This preclinical work suggests a new therapeutic opportunity for palbociclib in lung and other cancers currently treated with taxane based chemotherapy as standard of care.
- Published
- 2019
- Full Text
- View/download PDF
12. Abstract P2-07-01: Integrative analyses of immunophenotypes and multi-omics profiles in breast cancers
- Author
-
Sripad Ram, Y.S. Choi, Sy Cho, James S. Hardwick, Timothy Nichols, Pamela Vizcarra, Ying Ding, J. Yu, Keith A. Ching, Young-Ae Park, Zhengyan Kan, Eric L. Powell, and S-H Lee
- Subjects
Therapy naive ,Cancer Research ,Breast cancer ,Oncology ,business.industry ,Immune microenvironment ,Cancer research ,medicine ,Multi omics ,medicine.disease ,business - Abstract
The advent of immuno-oncology (IO) therapies has made it an imperative to characterize intratumoral immune microenvironment in addition to oncogenic alterations through molecular profiling of the tumor. To elucidate the baseline profiles of tumor infiltrating leukocytes (TILs) in breast cancer (BC) in the context of molecular subtypes and oncogenic alterations, we performed whole-exome sequencing (WES) and RNA-Seq of an Asian BC cohort (SMC) consisting of 178 treatment naïve primary tumors. A subset of 120 tumors was further analyzed by H&E and IHC using a panel of 8 TIL markers (CD45, CD4, CD8, CD163, PD1, PD-L1, IDO1 and FOXP3). Using expression signatures representing distinct immune cell types, we classified an expression compendium of 2,781 tumor samples, including SMC and multiple cancers from TCGA, into three immune subtypes with high, medium and low levels of TILs. Basal and HER2 subtypes show higher levels of TILs than Luminal subtypes, consistent with observed clinical responses to checkpoint blockade in clinical trials. Moreover, Asian BCs were significantly enriched in TIL-high subtype (35.3%) compared to the primarily Caucasian TCGA BC cohort (20.2%) while 50.6% of the highly immunogenic Lung adenocarcinoma was TIL-high. We then applied machine learning methods to detect and quantify TILs from H&E images of 120 SMC and 349 TCGA BC tumors. The expression signature analysis results were concordant with independently derived histology based TIL data. Taken together, our findings suggest that IO therapies may be more effective in HR negative BC subtypes and Asian BCs. Leukocyte exclusion (LE), an immunophenotype where TILs concentrate at the tumor periphery, has been linked to worse prognosis and resistance to IO therapies. Visual assessment of whole tumor IHC images identified LE patterns in 25% of SMC cases. We observed differential distribution of LE by molecular subtype and evidence for selective exclusion of immune cell subsets. Covariate analyses with clinical and molecular data while controlling for subtype as a confounder identified significant associations with tumor proliferation index, percent tumor purity and TP53 mutations. LE is also significantly associated with expression signatures of chemokine signaling, macrophages, angiogenesis and hypoxia, indicating that marked distinctions exist in both tumor intrinsic and microenvironment characteristics between TIL excluded and TIL infiltrated tumors. To validate these findings, we independently identified LE for 200 cases of TCGA BCs based on patterns of TILs extracted from H&E images and saw significant concordance of covariate relationships identified between TCGA and SMC. Our study provided a rare comprehensive resource for studying tumor associated immunity in breast cancers by generating the integrated multi-omics and IO profiles for a large cohort of primary tumors. Comparative analyses revealed that TIL activities are highly variable across different intrinsic subtypes and geographic origins of BC, with potential implications for IO therapeutic application. Correlative analyses of immunophenotypes with molecular data further yielded insights into LE's role in immune escape and identified hallmark signatures for LE indicative of causal molecular mechanisms. Citation Format: Kan Z, Powell E, Ram S, Ching K, Ding Y, Vizcarra P, Nichols T, Hardwick J, Lee S-H, Cho SY, Choi Y-L, Yu J-H, Park YH. Integrative analyses of immunophenotypes and multi-omics profiles in breast cancers [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P2-07-01.
- Published
- 2018
- Full Text
- View/download PDF
13. Patterns of Metastatic Spread and Mechanisms of Resistance to Crizotinib in ROS1-Positive Non–Small-Cell Lung Cancer
- Author
-
James S. Hardwick, Alice T. Shaw, Anna F. Farago, Aaron N. Hata, Lorin A. Ferris, Diane Tseng, Katherine Schultz, Beow Y. Yeap, Luc Friboulet, Justin F. Gainor, Satoshi Yoda, Donghui Huang, Leila Dardaei, Zofia Piotrowska, Ibiayi Dagogo-Jack, A. John Iafrate, Jessica J. Lin, Harper Hubbeling, and Mari Mino-Kenudson
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,Crizotinib ,business.industry ,Disease ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,Cohort ,medicine ,ROS1 ,Cancer research ,Anaplastic lymphoma kinase ,Lung cancer ,business ,Tyrosine kinase ,medicine.drug - Abstract
Purpose The ROS1 tyrosine kinase is activated through ROS1 gene rearrangements in 1% to 2% of non–small-cell lung cancers (NSCLCs), which confer sensitivity to treatment with the anaplastic lymphoma kinase (ALK)/ROS1/mesenchymal-epithelial transition factor inhibitor crizotinib. Currently, insights into patterns of metastatic spread and mechanisms of crizotinib resistance among patients with ROS1-positive disease are limited. Patients and Methods We reviewed clinical and radiographic imaging data of patients with ROS1- and ALK-positive NSCLC to compare patterns of metastatic spread at initial metastatic diagnosis. To determine molecular mechanisms of crizotinib resistance, we analyzed repeat biopsy specimens from a cohort of patients with ROS1-positive disease who progressed on crizotinib. Results We identified 39 and 196 patients with advanced ROS1- and ALK-positive NSCLC, respectively. Patients with ROS1-positive disease had significantly lower rates of extrathoracic metastases ( ROS1, 59.0%; ALK, 83.2%; P = .002), including lower rates of brain metastases ( ROS1, 19.4%; ALK, 39.1%; P = .033), at initial metastatic diagnosis. Despite similar overall survival between patients with ALK- and ROS1-positive NSCLC treated with crizotinib (median, 3.0 v 2.5 years, respectively; P = .786), patients with ROS1-positive NSCLC also had a significantly lower cumulative incidence of brain metastases (34% v 73% at 5 years; P < .001). In addition, we identified 16 patients who underwent a total of 17 repeat biopsies after progression on crizotinib. ROS1 resistance mutations were identified in 53% of specimens, including nine (64%) of 14 non–brain metastasis specimens. ROS1 mutations included G2032R (41%), D2033N (6%), and S1986F (6%). Conclusion Compared with ALK rearrangements, ROS1 rearrangements are associated with lower rates of extrathoracic metastases, including fewer brain metastases, at initial metastatic diagnosis. ROS1 resistance mutations, particularly G2032R, appear to be the predominant mechanism of resistance to crizotinib, which underscores the need to develop novel ROS1 inhibitors with activity against these resistant mutants.
- Published
- 2017
- Full Text
- View/download PDF
14. TNER: a novel background error suppression method for mutation detection in circulating tumor DNA
- Author
-
Kai Wang, Shibing Deng, Paul A. Rejto, James S. Hardwick, Maruja E. Lira, Jadwiga Bienkowska, Tao Xie, Jennifer Kinong, Crystal Valdez, and Donghui Huang
- Subjects
0301 basic medicine ,Computer science ,DNA Mutational Analysis ,Normal Distribution ,Early detection ,Computational biology ,Single-nucleotide variant ,Biology ,lcsh:Computer applications to medicine. Medical informatics ,Biochemistry ,DNA sequencing ,Circulating Tumor DNA ,Background noise ,03 medical and health sciences ,Structural Biology ,Neoplasms ,Variant calling ,Error suppression ,Humans ,Mutation detection ,Computer Simulation ,Molecular Biology ,lcsh:QH301-705.5 ,Genetics ,Applied Mathematics ,Methodology Article ,Disease progression ,Healthy subjects ,High-Throughput Nucleotide Sequencing ,ctDNA ,Computer Science Applications ,030104 developmental biology ,lcsh:Biology (General) ,ROC Curve ,Circulating tumor DNA ,Mutation ,Next-generation sequencing ,lcsh:R858-859.7 ,DNA microarray ,Software - Abstract
Background Ultra-deep next-generation sequencing of circulating tumor DNA (ctDNA) holds great promise as a tool for the early detection of cancer and for monitoring disease progression and therapeutic responses. However, the low abundance of ctDNA in the bloodstream coupled with technical errors introduced during library construction and sequencing complicates mutation detection. Results To achieve high accuracy of variant calling via better distinguishing low-frequency ctDNA mutations from background errors, we introduce TNER (Tri-Nucleotide Error Reducer), a novel background error suppression method that provides a robust estimation of background noise to reduce sequencing errors. The results on both simulated data and real data from healthy subjects demonstrate that the proposed algorithm consistently outperforms a current, state-of-the-art, position-specific error polishing model, particularly when the sample size of healthy subjects is small. Conclusions TNER significantly enhances the specificity of downstream ctDNA mutation detection without sacrificing sensitivity. The tool is publicly available at https://github.com/ctDNA/TNER. Electronic supplementary material The online version of this article (10.1186/s12859-018-2428-3) contains supplementary material, which is available to authorized users.
- Published
- 2018
15. An accessible pharmacodynamic transcriptional biomarker for notch target engagement
- Author
-
Donald A. Bergstrom, Luiz M Camargo, Andrey Loboda, Bo Wei, Jeremy Hing, Sanjiv J. Shah, Eva M. Finney, Samuel C. Blackman, Radha Railkar, Amy Harman, Peter Strack, Tim Demuth, Gary A. Herman, Jared Lunceford, Keith Q. Tanis, James S. Hardwick, Joel A. Klappenbach, James Watters, Robert Iannone, and Alexei A. Podtelezhnikov
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Transcription, Genetic ,Pharmacology ,Biomarkers, Pharmacological ,Transcriptome ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Double-Blind Method ,Transcription (biology) ,Benzene Derivatives ,medicine ,Animals ,Humans ,Protease Inhibitors ,Pharmacology (medical) ,Molecular Targeted Therapy ,RNA, Messenger ,Sulfones ,Receptor ,Oligonucleotide Array Sequence Analysis ,Cross-Over Studies ,Dose-Response Relationship, Drug ,Receptors, Notch ,biology ,Gene Expression Profiling ,RNA ,Hair follicle ,Macaca mulatta ,Healthy Volunteers ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Baltimore ,Models, Animal ,biology.protein ,Biomarker (medicine) ,Amyloid Precursor Protein Secretases ,Drug Monitoring ,Propionates ,Hair Follicle ,Amyloid precursor protein secretase - Abstract
γ-Secretase mediates amyloid production in Alzheimer's disease (AD) and oncogenic activity of Notch. γ-Secretase inhibitors (GSIs) are thus of interest for AD and oncology. A peripheral biomarker of Notch activity would aid determination of the therapeutic window and dosing regimen for GSIs, given toxicities associated with chronic Notch inhibition. This study examined the effects of GSI MK-0752 on blood and hair follicle transcriptomes in healthy volunteers. The effects of a structurally diverse GSI on rhesus blood and hair follicles were also compared. Significant dose-related effects of MK-0752 on transcription were observed in hair follicles, but not blood. The GSI biomarker identified in follicles exhibited 100% accuracy in a clinical test cohort, and was regulated in rhesus by a structurally diverse GSI. This study identified a translatable, accessible pharmacodynamic biomarker of GSI target engagement and provides proof of concept of hair follicle RNA as a translatable biomarker source.
- Published
- 2016
- Full Text
- View/download PDF
16. Abstract A2-33: Molecular profiling of patient-derived xenograft models across cancers
- Author
-
Xie Tao, Hui Wang, David Shields, Keith A. Ching, Stephanie T. Shi, Maruja E. Lira, Amy Jackson-Fisher, Xianxian Zheng, Pamela Vizcarra, Zhengyan Kan, Mark Ozeck, Gabriel Troche, Julio Fernandez, Konstantinos Tsaparikos, Xiaorong Li, Edward Rosfjord, Shibing Deng, Eric Upeslacis, Paul A. Rejto, James S. Hardwick, Judy Lucas, Ying Ding, Patrick Lappin, and John Chionis
- Subjects
Genetics ,Cancer Research ,business.industry ,Drug discovery ,In silico ,Computational biology ,medicine.disease ,Primary tumor ,Clinical biomarker ,Oncology ,Cancer genome ,Medicine ,Copy-number variation ,business ,Gene ,Tumor xenograft - Abstract
Patient-Derived Xenograft (PDX) provides important preclinical model for pharmacological testing of oncology drug candidates. Molecular profiling of PDX tumors contributes to many areas of drug discovery from target discovery to development of clinical biomarker hypotheses and clinical trial design. We established a work flow to perform genomic and histopathology analyses of large numbers of PDX tumor models being made available for Pfizer internal research. To date we have generated whole-genome sequencing (WGS), whole-exome sequencing (WES) and whole transcriptome sequencing (RNA-Seq) data on PDX models spanning six cancer types including colon, pancreatic and breast cancers. Bioinformatics pipelines were developed to quantify gene expression and detect genetic alterations including mutation, copy number variations and gene fusions. A controlled evaluation study demonstrated that in silico classification of NGS reads into human/mouse origins is more effective than laboratory-based methods for removing mouse tissue contamination. Comparative analyses of molecular profiles from PDX and primary tumors of the same cancer origins suggest that important patterns of gene expression are retained by PDX models. An integrative genomic classifier was developed using the random forest algorithm, trained on primary tumor data, and shown to identify PDX cancer subtypes with high accuracy. Citation Format: Zhengyan Kan, Edward Rosfjord, James Hardwick, Ying Ding, Xianxian Zheng, Julio Fernandez, Stephanie Shi, Mark Ozeck, Hui Wang, Gabriel Troche, Eric Upeslacis, Amy Jackson-Fisher, Keith Ching, Shibing Deng, Xie Tao, John Chionis, Maruja Lira, Xiaorong Li, Konstantinos Tsaparikos, Patrick Lappin, Pamela Vizcarra, David Shields, Judy Lucas, Paul Rejto. Molecular profiling of patient-derived xenograft models across cancers. [abstract]. In: Proceedings of the AACR Special Conference on Translation of the Cancer Genome; Feb 7-9, 2015; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(22 Suppl 1):Abstract nr A2-33.
- Published
- 2015
- Full Text
- View/download PDF
17. Phase I Trial of Vorinostat Combined With Bortezomib for the Treatment of Relapsing and/or Refractory Multiple Myeloma
- Author
-
Mohamad A. Hussein, Gary J. Schiller, Donna M. Weber, Ronald Sobecks, Sundar Jagannath, James S. Hardwick, Thorsten Graef, and Lisa Lupinacci
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,Hydroxamic Acids ,Bortezomib ,Cohort Studies ,Refractory ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Secondary Prevention ,medicine ,Humans ,Adverse effect ,Vorinostat ,Aged ,business.industry ,Drug Synergism ,Hematology ,Middle Aged ,Boronic Acids ,Surgery ,Clinical trial ,Pyrazines ,Proteasome inhibitor ,Female ,Multiple Myeloma ,business ,Cohort study ,medicine.drug - Abstract
Preclinical studies have shown that targeted combination therapy consisting of vorinostat and bortezomib has antitumor activity in multiple myeloma (MM). We examined this drug combination in advanced relapsing and/or refractory MM patients (n = 34). Although the maximum tolerated dose was not reached, the study found this combination regimen generally well tolerated and clinically active in relapsed and/or refractory MM patients.Development of targeted therapies for MM has improved response rates and increased patient survival, but ultimately the disease becomes refractory and progresses. Vorinostat combined with bortezomib has demonstrated synergistic antiproliferative and proapoptotic activity in preclinical models of MM. The objectives of this study were to determine the maximum tolerated dose for vorinostat with bortezomib in patients with advanced MM and to evaluate the clinical benefit of this new drug combination.Patients ≥ 18 years old with relapsed and/or refractory MM were enrolled into escalating dose cohorts of vorinostat and bortezomib combination therapy. Thirty-four patients were enrolled and were evaluable for safety and efficacy analyses.All patients reported adverse events, 89% of which were mild to moderate in severity. Thirteen patients experienced 29 serious adverse events, 12 (41%) of which were considered drug-related. The maximum tolerated dose was not reached. Partial responses were observed in 9 (27%) patients. Minimal responses were observed in 2 additional patients (6%), and another 20 patients (59%) experienced disease stabilization.Vorinostat with bortezomib is generally well-tolerated and has clinical activity in patients with relapsed and/or refractory MM. Response rates were similar in patients previously exposed to bortezomib and patients who were naive to bortezomib therapy.
- Published
- 2012
- Full Text
- View/download PDF
18. Vorinostat combined with bexarotene for treatment of cutaneous T-cell lymphoma:in vitroand phase I clinical evidence supporting augmentation of retinoic acid receptor/retinoid X receptor activation by histone deacetylase inhibition
- Author
-
James S. Hardwick, Jose Garcia-Vargas, Stanley R. Frankel, Matthias Steinhoff, Chalid Assaf, Wolfram Sterry, Kenneth B. Hymes, Syed Rizvi, Janet D. Ahern, Karl Heath, Katrin Baumann, Marc Beyer, Sean Whittaker, Reinhard Dummer, Mirjam T. Epping, René Bernards, Helmut Kerl, University of Zurich, and Dummer, R
- Subjects
Adult ,Male ,Cancer Research ,Tetrahydronaphthalenes ,Transcription, Genetic ,Cell Survival ,Receptors, Retinoic Acid ,medicine.drug_class ,T cell ,2720 Hematology ,610 Medicine & health ,In Vitro Techniques ,Retinoid X receptor ,Pharmacology ,Hydroxamic Acids ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,1306 Cancer Research ,Vorinostat ,Aged ,Bexarotene ,business.industry ,Histone deacetylase inhibitor ,Cutaneous T-cell lymphoma ,10177 Dermatology Clinic ,Hematology ,Middle Aged ,medicine.disease ,Lymphoma, T-Cell, Cutaneous ,Histone Deacetylase Inhibitors ,Retinoic acid receptor ,Retinoid X Receptors ,medicine.anatomical_structure ,Oncology ,Cancer research ,2730 Oncology ,Female ,Histone deacetylase ,business ,medicine.drug - Abstract
The retinoid X receptor (RXR)-agonist bexarotene and the histone deacetylase inhibitor (HDACI) vorinostat are each established monotherapies for cutaneous T-cell lymphomas (CTCLs). We investigated the combination of HDACI and retinoic acid receptor (RAR)/RXR agonists in vitro and in a phase I, multicenter, open-label, two-part dose-escalation study. The combination of bexarotene with a HDACI in vitro leads to cooperative activation of gene transcription and reduction of cell viability in human tumor cell lines. The primary clinical objective was to determine the maximum tolerated dose (MTD) of bexarotene plus vorinostat in 23 patients with CTCLs. The MTD for part I was established at vorinostat 200 mg/day plus bexarotene 300 mg/m(2)/day. The MTD for part II was not reached. Four patients had an objective response and seven patients experienced pruritus relief. We conclude that concomitant administration of vorinostat and bexarotene is feasible only if lower doses of each drug are administered relative to the product label monotherapy doses.
- Published
- 2012
- Full Text
- View/download PDF
19. Abstract PD2-02: Longitudinal ctDNA sequencing using an expanded genomic panel in the PALOMA3 trial of palbociclib plus fulvestrant
- Author
-
James S. Hardwick, Stephen Huang, S. Loibl, Ben O'Leary, Paul A. Rejto, Fabrice Andre, Maruja E. Lira, N. Turner, Tao Xie, Jennifer Kinong, Xin-Yun Huang, AM DeMichele, C Huang Bartlett, Massimo Cristofanilli, Yuan Liu, and Shibing Deng
- Subjects
Cancer Research ,biology ,Fulvestrant ,Cancer ,Palbociclib ,medicine.disease ,Molecular biology ,Breast cancer ,Oncology ,biology.protein ,medicine ,Digital polymerase chain reaction ,CDKN1B ,Cyclin-dependent kinase 6 ,Gene ,medicine.drug - Abstract
Background: CDK4/6 inhibition combined with endocrine therapy (ET) is now the standard of care for advanced hormone receptor (HR) positive breast cancer patients (pts). Previous ctDNA analysis of paired pre-treatment (pre-T) and end of treatment (EoT) plasma samples from the PALOMA-3 trial of the CDK4/6 inhibitor P with/without F demonstrated that acquired PIK3CA, ESR1, RB1 and rare growth factor receptor mutations (mut) likely contribute to resistance (Turner et al, ASCO 2018, Abstract 1001). We now report results from an extended ctDNA analysis of paired PALOMA-3 plasma samples using hybridization capture-based, next-generation sequencing (NGS) assay covering 87 breast cancer, HR signaling, and cell cycle-related genes. Methods: Pre-/post-menopausal pts whose disease progressed after prior ET (N=521) were randomized 2:1 to receive F 500 mg + P (125 mg/d, 3 wk on/1 wk off) or placebo. Cell-free DNA extracted from paired plasma samples (n=194) was analyzed with a custom 87-gene NGS assay, to identify single nucleotide variants and copy number amplification (CNA), with molecular barcoding, high NGS coverage (10,000-20,000 reads per nucleotide position), and background error correction. Differences in ctDNA mut frequencies detected in P+F compared to F alone were assessed and their association to clinical outcome estimated. Results: Blinded assay qualification and droplet digital PCR validation suggested a mut detection frequency cutoff at 0.3%. A high coefficient of correlation with previously presented data was observed for ESR1 and PIK3CA variants (r=0.94 and 0.97, respectively), with acquisition of PIK3CA and ESR1 mutations at EoT in P+F and F groups. Gene level mut analysis of EoT plasma revealed no significant difference between P+F vs F alone (Table), although there was an increase in RB1 mutations in P+F consistent with previous data. Other acquired muts at EoT (SMAD4, NOTCH2, and CDKN1B) were observed at low frequencies. Muts in CDK4 and CDK6 were rarely observed (< 1% of pts), regardless of treatment arm. Detected Variants, Frequency, n (%) P+FF GenePre-TEoTPre-TEoTPre-T pvalEoT pvalPIK3CA47(37)51(40)19(28)22(33)0.2660.352ESR136(28)45(35)19(28)24(36)11TP5330(24)33(26)23(34)25(37)0.1280.137RB12(2)9(7)2(3)2(3)NA0.336PTEN5(4)7(6)3(4)3(4)11AKT17(6)7(6)2(3)2(3)0.7210.721 Conclusions: Our results corroborate the previously reported results demonstrating that genomic profiles of treatment resistant cancer are similar between P + F and F therapy. Expanded analysis of cell-cycle genes identified no new recurrently mutated genes, and confirmed that RB1 mutations are selected at low frequency after P+F treatment. Alterations in cell cycle genes may not be a common mechanism of resistance to CDK4/6 inhibitors in HR+ HER2- advanced breast cancer. Sponsor: Pfizer Citation Format: O'Leary B, Lira ME, Huang S, Deng S, Xie T, Kinong JK, Hardwick J, Rejto P, Liu Y, Huang X, Huang Bartlett C, André F, Loibl S, DeMichele A, Cristofanilli M, Turner NC. Longitudinal ctDNA sequencing using an expanded genomic panel in the PALOMA3 trial of palbociclib plus fulvestrant [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr PD2-02.
- Published
- 2019
- Full Text
- View/download PDF
20. Abstract LB-215: Analysis of mutations associated with response to glasdegib in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS)
- Author
-
Paul A. Rejto, James S. Hardwick, Jadwiga Bienkowska, Kai Wang, Thomas O’Brien, Mark Ozeck, Donghui Huang, Keith A. Ching, Geoffrey Chan, Jingjin Gao, and Paul D. Lira
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,NPM1 ,Mutation ,business.industry ,Cancer ,Myeloid leukemia ,Context (language use) ,medicine.disease_cause ,medicine.disease ,Oncology ,medicine ,Cytarabine ,Cancer research ,KRAS ,business ,medicine.drug - Abstract
Background: The Hedgehog (Hh) signaling pathway is key to development, differentiation, and stem cell maintenance. In cancer, dysregulation of Hh signaling is associated with solid tumors and hematological malignancies. Hh signaling in AML and MDS is thought to promote the renewal and maintenance of leukemic stem cells, which may lead to chemotherapy resistance and recurrence. Glasdegib is a potent and selective oral inhibitor of Smoothened, a transmembrane protein that stimulates Hh signaling. Here we report the results of an exploratory analysis to profile mutations in 118 bone marrow samples and 77 peripheral blood samples of patients with previously untreated AML or high-risk MDS treated with chemotherapy with or without glasdegib in phase I/II clinical trials. Materials and Methods: We used targeted resequencing to survey 109 genes, and a polymerase chain reaction assay to detect FLT3 internal tandem duplications. Resequencing and response data were obtained from two arms, 1) an intensive group (n=61) treated with a combination of cytarabine/daunorubicin plus glasdegib, and 2) a non-intensive group (n=73) treated with low-dose cytarabine (LDAC) plus glasdegib (n=44) or LDAC alone (n=19). We assessed significantly mutated genes by Fisher's Exact Test at p ≤ 0.05. Results: In the combined dataset we found that mutations in NF1 (P=0.006), KRAS (P=0.028), or a combination of KRAS, NRAS, and NF1 (P=0.013) were associated with a negative response. Mutations in NPM1 (P=0.006), MPL (P=0.041), RB1 (P=0.041), or SF3B1 (P=0.041) were associated with a positive response. When split by arm, in the intensive group mutations in TP53 (P=0.001), NF1 (P=0.035), or CREBBP (P=0.041) were associated with negative response, whereas mutation in NPM1 was associated with positive response (P=0.046). In the non-intensive group, mutations in RSPH10B2 (P=0.036) or a combination of KRAS, NRAS, and NF1 (P=0.031) were associated with negative response, whereas mutation in CREBBP (P=0.007) was associated with positive response. We discuss the implications of these mutations in the context of Smoothened inhibition and the leukemic stem cell hypothesis. Conclusions: Mutational profiling of patients with AML or high-risk MDS treated with glasdegib highlights alterations in genes associated with response and the potential role of Hh signaling in these malignancies. Citation Format: Keith A. Ching, Donghui Huang, Kai Wang, Mark Ozeck, Paul Lira, Jingjin Gao, Jadwiga Bienkowska, Paul Rejto, James Hardwick, Thomas O'Brien, Geoffrey Chan. Analysis of mutations associated with response to glasdegib in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) [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-215.
- Published
- 2018
- Full Text
- View/download PDF
21. Immunohistochemical Detection of Antitumor, Antimetastasis, and Antiangiogenesis Effects of a Vascular Endothelial Growth Factor Receptor 2 Kinase Inhibitor in an Orthotopic Breast Cancer Metastasis Model
- Author
-
Laura Sepp-Lorenzino, Brett Connolly, Nancy E. Kohl, Rosemary C. McFall, Bin Shi, James S. Hardwick, Susan Hill, Kenneth A. Thomas, and Xianzhi Mao
- Subjects
Pathology ,medicine.medical_specialty ,Histology ,Angiogenesis ,business.industry ,Cell ,Kinase insert domain receptor ,medicine.disease ,Primary tumor ,Metastasis ,Medical Laboratory Technology ,Breast cancer ,medicine.anatomical_structure ,Cancer cell ,medicine ,Immunohistochemistry ,Anatomy ,business - Abstract
Antiangiogenic agents such as vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors may prove most efficacious in the setting of early disease and in the prevention of dissemination and growth of micrometastases. This hypothesis was tested in a metastatic orthotopic rat model of breast cancer with the use of a novel orally bioavailable VEGFR2 kinase inhibitor, MK-0888. Mat B III rat mammary cancer cells were implanted into the mammary fat pads of syngeneic female F344 rats. Primary tumor growth was very aggressive, with micrometastases detected 8 days after cell implantation in ipsilateral axillary and inguinal lymph nodes. Lung metastases were detected 15 days after cell implantation by histological analysis. MK-0888 suppressed primary and metastatic tumor growth and reduced the incidence of metastasis in a dose- and schedule-dependent manner. Inhibitions of primary and metastatic tumor growth, as well as intratumoral antiangiogenesis effects, were detected in situ by immunohistochemi...
- Published
- 2010
- Full Text
- View/download PDF
22. Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers
- Author
-
Chunsheng Zhang, Maria O'Donovan, Frances R. Balkwill, Marco Novelli, Vicki Save, Amel Saadi, Madhumita Das, Pierre Lao-Sirieix, Nicholas J. Clemons, James S. Hardwick, Rebecca C. Fitzgerald, Elaine C. Walker, and Nicholas B. Shannon
- Subjects
Stromal cell ,Esophageal Neoplasms ,Adenocarcinoma ,Biology ,Digestive System Neoplasms ,Barrett Esophagus ,Stroma ,Metaplasia ,Endopeptidases ,Gene expression ,medicine ,Humans ,Neoplasm Invasiveness ,Receptors, Cytokine ,Inflammation ,Multidisciplinary ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Serine Endopeptidases ,Membrane Proteins ,Nuclear Proteins ,Cancer ,Oncogenes ,Biological Sciences ,Prognosis ,BCL6 ,medicine.disease ,DNA-Binding Proteins ,medicine.anatomical_structure ,Gelatinases ,Dysplasia ,Immunology ,Proto-Oncogene Proteins c-bcl-6 ,Trans-Activators ,Cancer research ,Cytokines ,Stromal Cells ,medicine.symptom ,Pancreas ,Precancerous Conditions - Abstract
The stromal compartment is increasingly recognized to play a role in cancer. However, its role in the transition from preinvasive to invasive disease is unknown. Most gastrointestinal tumors have clearly defined premalignant stages, and Barrett’s esophagus (BE) is an ideal research model. Supervised clustering of gene expression profiles from microdissected stroma identified a gene signature that could distinguish between BE metaplasia, dysplasia, and esophageal adenocarcinoma (EAC). EAC patients overexpressing any of the five genes ( TMEPAI , JMY , TSP1 , FAPα , and BCL6 ) identified from this stromal signature had a significantly poorer outcome. Gene ontology analysis identified a strong inflammatory component in BE disease progression, and key pathways included cytokine–cytokine receptor interactions and TGF-β. Increased protein levels of inflammatory-related genes significantly up-regulated in EAC compared with preinvasive stages were confirmed in the stroma of independent samples, and in vitro assays confirmed functional relevance of these genes. Gene set enrichment analysis of external datasets demonstrated that the stromal signature was also relevant in the preinvasive to invasive transition of the stomach, colon, and pancreas. These data implicate inflammatory pathways in the genesis of gastrointestinal tract cancers, which can affect prognosis.
- Published
- 2010
- Full Text
- View/download PDF
23. Predicting Response to Histone Deacetylase Inhibitors Using High-Throughput Genomics
- Author
-
Fan Wang, R. Stephanie Huang, Michael Chisamore, Michael L. Maitland, Bonnie LaCroix, Sanja Karovic, Paul Geeleher, Divya Lenkala, Jacqueline Wang, Andrey Loboda, James S. Hardwick, and Michael Nebozhyn
- Subjects
Cancer Research ,medicine.drug_class ,Antineoplastic Agents ,Biology ,Bioinformatics ,Hydroxamic Acids ,Autoantigens ,Chromatin remodeling ,Article ,Predictive Value of Tests ,Cell Line, Tumor ,medicine ,Humans ,Vorinostat ,Regulation of gene expression ,Gene Expression Profiling ,Histone deacetylase inhibitor ,Thrombocytopenia ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Histone Deacetylase Inhibitors ,Oncology ,Histone deacetylase complex ,Histone deacetylase ,CHD4 ,medicine.drug ,Mi-2 Nucleosome Remodeling and Deacetylase Complex - Abstract
Background Many disparate biomarkers have been proposed as predictors of response to histone deacetylase inhibitors (HDI); however, all have failed when applied clinically. Rather than this being entirely an issue of reproducibility, response to the HDI vorinostat may be determined by the additive effect of multiple molecular factors, many of which have previously been demonstrated. Methods We conducted a large-scale gene expression analysis using the Cancer Genome Project for discovery and generated another large independent cancer cell line dataset across different cancers for validation. We compared different approaches in terms of how accurately vorinostat response can be predicted on an independent out-of-batch set of samples and applied the polygenic marker prediction principles in a clinical trial. Results Using machine learning, the small effects that aggregate, resulting in sensitivity or resistance, can be recovered from gene expression data in a large panel of cancer cell lines.This approach can predict vorinostat response accurately, whereas single gene or pathway markers cannot. Our analyses recapitulated and contextualized many previous findings and suggest an important role for processes such as chromatin remodeling, autophagy, and apoptosis. As a proof of concept, we also discovered a novel causative role for CHD4, a helicase involved in the histone deacetylase complex that is associated with poor clinical outcome. As a clinical validation, we demonstrated that a common dose-limiting toxicity of vorinostat, thrombocytopenia, can be predicted (r = 0.55, P = .004) several days before it is detected clinically. Conclusion Our work suggests a paradigm shift from single-gene/pathway evaluation to simultaneously evaluating multiple independent high-throughput gene expression datasets, which can be easily extended to other investigational compounds where similar issues are hampering clinical adoption.
- Published
- 2015
24. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes
- Author
-
Jiangang Liu, Patrick Tan, Ronghua Chen, Jae Moon Bae, Christoph Reinhard, Shawn Liu, Razvan Cristescu, Min Gew Choi, Swee Seong Wong, Sin-Ho Jung, Jason C. Ting, James S. Hardwick, Seung Tae Kim, Sung Kim, Jian Wang, Kun Yu, Jake Fu, Joon-Ho Lee, Jason Gang Jin, Mark Ayers, Xiang S. Ye, Amit Aggarwal, Michael Nebozhyn, Andrey Loboda, Lara Gong, Dai Hongyue, Kyoung-Mee Kim, Insuk Sohn, Yong Gang Yue, Se Hoon Park, Jeeyun Lee, Won Ki Kang, In-Gu Do, and Tae Sung Sohn
- Subjects
Male ,Epithelial-Mesenchymal Transition ,Gene Dosage ,Translational research ,Disease ,Bioinformatics ,Gene dosage ,General Biochemistry, Genetics and Molecular Biology ,Cohort Studies ,Translational Research, Biomedical ,Recurrence ,Stomach Neoplasms ,Medicine ,Humans ,Epithelial–mesenchymal transition ,Aged ,Oligonucleotide Array Sequence Analysis ,Proportional Hazards Models ,Principal Component Analysis ,Helicobacter pylori ,Proportional hazards model ,business.industry ,Gene Expression Profiling ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,Prognosis ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,Tissue Array Analysis ,Mutation ,Disease Progression ,Female ,DNA microarray ,Tumor Suppressor Protein p53 ,business ,Microsatellite Repeats - Abstract
Gastric cancer, a leading cause of cancer-related deaths, is a heterogeneous disease. We aim to establish clinically relevant molecular subtypes that would encompass this heterogeneity and provide useful clinical information. We use gene expression data to describe four molecular subtypes linked to distinct patterns of molecular alterations, disease progression and prognosis. The mesenchymal-like type includes diffuse-subtype tumors with the worst prognosis, the tendency to occur at an earlier age and the highest recurrence frequency (63%) of the four subtypes. Microsatellite-unstable tumors are hyper-mutated intestinal-subtype tumors occurring in the antrum; these have the best overall prognosis and the lowest frequency of recurrence (22%) of the four subtypes. The tumor protein 53 (TP53)-active and TP53-inactive types include patients with intermediate prognosis and recurrence rates (with respect to the other two subtypes), with the TP53-active group showing better prognosis. We describe key molecular alterations in each of the four subtypes using targeted sequencing and genome-wide copy number microarrays. We validate these subtypes in independent cohorts in order to provide a consistent and unified framework for further clinical and preclinical translational research.
- Published
- 2015
25. Abstract 2749: Liquid biopsy testing allows highly-sensitive detection of plasma cfDNA mutations in 87 breast cancer-related genes
- Author
-
Kai Wang, Paul A. Rejto, Maruja E. Lira, Shibing Deng, James S. Hardwick, Nathan V. Lee, Jadwiga Bienkowska, Zhou Zhu, Jingjin Gao, Tao Xie, Jennifer Kinong, and Stephen Huang
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Estrogen receptor ,Single-nucleotide polymorphism ,medicine.disease ,genomic DNA ,Breast cancer ,Oncology ,Cancer research ,medicine ,Liquid biopsy ,International HapMap Project ,business ,Allele frequency ,Gene - Abstract
Liquid biopsies have the potential to revolutionize the way physicians select personalized anti-cancer therapies, monitor patient responses to treatment, and characterize acquired resistance to cancer drugs. New tests that use a simple peripheral blood draw offer snapshots of a patient‘s total tumor DNA mutation profile and are attractive because of their minimally-invasive modality and because they integrate information from both primary and metastatic disease. Currently, most plasma cell-free DNA (cfDNA) mutation detection tests used in clinical research detect known hotspot mutations in a limited number of genes. Technologies that interrogate multi-gene panels in cfDNA are advancing, but commercially-available options suitable for clinical use are limited, come at a high cost, and are not customizable. We designed and developed a customized, next generation sequencing-based, liquid biopsy assay capable of detecting somatic mutations in 87 breast cancer genes involved in cell cycle and estrogen receptor signaling. Targeted regions (147 Kb) were enriched using hybrid capture resulting in an average capture specificity and uniformity of 65.93% and 96.38%, respectively. When tested on cfDNA from healthy donors (n=14), we demonstrated a level of specificity >99.99%. Analytical sensitivity of 0.1% was established on HapMap and reference mutant cell line DNA. Using a pool of HapMap genomic DNA (n=10), 96% (48/50) of SNPs with expected allele frequency of 0.1% were detected. In reference mutant cell line DNA with 1% or 0.1% mutant allele frequencies, we were able to reliably detect all mutations present at 1% and mutations at 0.1% in 50% of the cases. Assay validation on plasma cfDNA with matching tumor from ER+, HER2- breast cancer patients will be presented. In conclusion, we developed a highly sensitive and specific liquid biopsy assay to interrogate 87 breast cancer-related genes. The high level of specificity and sensitivity makes the test ideal not only for detecting known cancer gene hotspot mutations but also for detection of novel gene mutations that may arise during treatment as a result of acquired drug resistance. Citation Format: Maruja E. Lira, Tao Xie, Shibing Deng, Jennifer Kinong, Jingjin Gao, Zhou Zhu, Nathan Lee, Paul Rejto, Jadwiga Bienkowska, James Hardwick, Kai Wang, Stephen Huang. Liquid biopsy testing allows highly-sensitive detection of plasma cfDNA mutations in 87 breast cancer-related genes [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 2749. doi:10.1158/1538-7445.AM2017-2749
- Published
- 2017
- Full Text
- View/download PDF
26. Endoglin as an Important Regulator of Colorectal Cancer Invasion and Metastasis
- Author
-
Lukas J. A. C. Hawinkels, Cornelis F. M. Sier, Charles P. Theuer, Mark J A Schoonderwoerd, Madelon Paauwe, Peter ten Dijke, James S. Hardwick, Hein W. Verspaget, and Gabi W. van Pelt
- Subjects
Oncology ,medicine.medical_specialty ,Hepatology ,Colorectal cancer ,business.industry ,Internal medicine ,Gastroenterology ,Regulator ,medicine ,Endoglin ,medicine.disease ,business ,Metastasis - Published
- 2017
- Full Text
- View/download PDF
27. Genomic landscape and genetic heterogeneity in gastric adenocarcinoma revealed by whole-genome sequencing
- Author
-
Christoph Reinhard, Xiaoqiao Liu, Jason C. Ting, Hongyue Dai, James S. Hardwick, Kun Yu, Jeeyun Lee, Xiang S. Ye, Joon Oh Park, Michael Nebozhyn, Jason Gang Jin, Shawn Liu, Razvan Cristescu, Yong Gang Yue, In Gu Do, Sung Kim, Jian Wang, Kyoung-Mee Kim, So Young Kang, Andrey Loboda, Lara Gong, Swee Seong Wong, Jake Fu, Amit Aggarwal, Chen Ronghua, and Tae Sung Sohn
- Subjects
Adult ,Male ,Somatic cell ,Sequence analysis ,General Physics and Astronomy ,Genomics ,Adenocarcinoma ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Genetic Heterogeneity ,Young Adult ,Stomach Neoplasms ,medicine ,Humans ,Gene ,Aged ,Genetics ,Whole genome sequencing ,Multidisciplinary ,Genetic heterogeneity ,Cancer ,Sequence Analysis, DNA ,General Chemistry ,Middle Aged ,medicine.disease ,CpG site ,Female - Abstract
Gastric cancer (GC) is the second most common cause of cancer-related deaths. It is known to be a heterogeneous disease with several molecular and histological subtypes. Here we perform whole-genome sequencing of 49 GCs with diffuse (N=31) and intestinal (N=18) histological subtypes and identify three mutational signatures, impacting TpT, CpG and TpCp[A/T] nucleotides. The diffuse-type GCs show significantly lower clonality and smaller numbers of somatic and structural variants compared with intestinal subtype. We further divide the diffuse subtype into one with infrequent genetic changes/low clonality and another with relatively higher clonality and mutations impacting TpT dinucleotide. Notably, we discover frequent and exclusive mutations in Ephrins and SLIT/ROBO signalling pathway genes. Overall, this study delivers new insights into the mutational heterogeneity underlying distinct histologic subtypes of GC that could have important implications for future research in the diagnosis and treatment of GC.
- Published
- 2014
- Full Text
- View/download PDF
28. Rapamycin-modulated transcription defines the subset of nutrient-sensitive signaling pathways directly controlled by the Tor proteins
- Author
-
Finny G. Kuruvilla, Alykhan F. Shamji, Stuart L. Schreiber, James S. Hardwick, and Jeffrey K. Tong
- Subjects
Saccharomyces cerevisiae Proteins ,Transcription, Genetic ,Nitrogen ,Prions ,Citric Acid Cycle ,Saccharomyces cerevisiae ,TORC1 signaling ,Repressor ,Cell Cycle Proteins ,Fungal Proteins ,Phosphatidylinositol 3-Kinases ,Cell Cycle Protein ,Oligonucleotide Array Sequence Analysis ,Sirolimus ,Glutathione Peroxidase ,Fungal protein ,Multidisciplinary ,biology ,Activator (genetics) ,Gene Expression Profiling ,Nucleic Acid Hybridization ,Biological Sciences ,biology.organism_classification ,Culture Media ,Repressor Proteins ,Citric acid cycle ,Phosphotransferases (Alcohol Group Acceptor) ,Glucose ,Biochemistry ,Signal transduction ,Glycolysis ,Signal Transduction - Abstract
The immunosuppressant rapamycin inhibits Tor1p and Tor2p (target of rapamycin proteins), ultimately resulting in cellular responses characteristic of nutrient deprivation through a mechanism involving translational arrest. We measured the immediate transcriptional response of yeast grown in rich media and treated with rapamycin to investigate the direct effects of Tor proteins on nutrient-sensitive signaling pathways. The results suggest that Tor proteins directly modulate the glucose activation and nitrogen discrimination pathways and the pathways that respond to the diauxic shift (including glycolysis and the citric acid cycle). Tor proteins do not directly modulate the general amino acid control, nitrogen starvation, or sporulation (in diploid cells) pathways. Poor nitrogen quality activates the nitrogen discrimination pathway, which is controlled by the complex of the transcriptional repressor Ure2p and activator Gln3p. Inhibiting Tor proteins with rapamycin increases the electrophoretic mobility of Ure2p. The work presented here illustrates the coordinated use of genome-based and biochemical approaches to delineate a cellular pathway modulated by the protein target of a small molecule.
- Published
- 1999
- Full Text
- View/download PDF
29. Protein phosphatase 2A interacts with the 70-kDa S6 kinase and is activated by inhibition of FKBP12–rapamycinassociated protein
- Author
-
Stuart L. Schreiber, Randall T. Peterson, Bimal N. Desai, and James S. Hardwick
- Subjects
Recombinant Fusion Proteins ,Phosphatase ,macromolecular substances ,Biology ,Transfection ,Models, Biological ,environment and public health ,Tacrolimus Binding Proteins ,Dephosphorylation ,Jurkat Cells ,chemistry.chemical_compound ,Eukaryotic initiation factor ,Phosphoprotein Phosphatases ,Humans ,Protein Phosphatase 2 ,Enzyme Inhibitors ,Immunophilins ,Phosphorylation ,Oxazoles ,Glutathione Transferase ,Sirolimus ,Multidisciplinary ,Kinase ,Ribosomal Protein S6 Kinases ,TOR Serine-Threonine Kinases ,Protein phosphatase 2 ,Biological Sciences ,Cell biology ,Kinetics ,Phosphotransferases (Alcohol Group Acceptor) ,enzymes and coenzymes (carbohydrates) ,chemistry ,Biochemistry ,Marine Toxins ,Carrier Proteins ,Calyculin - Abstract
The FKBP12–rapamycin-associated protein (FRAP; also called RAFT1/mTOR) regulates translation initiation and entry into the cell cycle. Depriving cells of amino acids or treating them with the small molecule rapamycin inhibits FRAP and results in rapid dephosphorylation and inactivation of the translational regulators 4E-BP1(eukaryotic initiation factor 4E-binding protein 1) and p70 s6k (the 70-kDa S6 kinase). Data published recently have led to the view that FRAP acts as a traditional mitogen-activated kinase, directly phosphorylating 4E-BP1 and p70 s6k in response to mitogenic stimuli. We present evidence that FRAP controls 4E-BP1 and p70 s6k phosphorylation indirectly by restraining a phosphatase. A calyculin A-sensitive phosphatase is required for the rapamycin- or amino acid deprivation-induced dephosphorylation of p70 s6k , and treatment of Jurkat I cells with rapamycin increases the activity of the protein phosphatase 2A (PP2A) toward 4E-BP1. PP2A is shown to associate with p70 s6k but not with a mutated p70 s6k that is resistant to rapamycin- and amino acid deprivation-mediated dephosphorylation. FRAP also is shown to phosphorylate PP2A in vitro , consistent with a model in which phosphorylation of PP2A by FRAP prevents the dephosphorylation of 4E-BP1 and p70 s6k , whereas amino acid deprivation or rapamycin treatment inhibits FRAP’s ability to restrain the phosphatase.
- Published
- 1999
- Full Text
- View/download PDF
30. Downregulation of MHC-I expression is prevalent but reversible in Merkel cell carcinoma
- Author
-
Kelly G. Paulson, David Schrama, Hideki Nakajima, Christoph Willmes, Margaret M. Madeleine, Jayasri G. Iyer, Jürgen C. Becker, James S. Hardwick, Mary L. Disis, Shailender Bhatia, Kotaro Nagase, William T. Simonson, Paul Nghiem, Shinichi Koba, David M. Koelle, Aaron Seo, Olga K. Afanasiev, Michele A. Cleary, Renee Thibodeau, Andrew Tegeder, and Shigetoshi Sano
- Subjects
Cancer Research ,Skin Neoplasms ,medicine.medical_treatment ,Immunology ,Medizin ,Down-Regulation ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,Human leukocyte antigen ,Major histocompatibility complex ,Article ,Downregulation and upregulation ,Cell Line, Tumor ,MHC class I ,medicine ,Carcinoma ,Humans ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Merkel cell carcinoma ,Histocompatibility Antigens Class I ,food and beverages ,Interferon-beta ,Immunotherapy ,Flow Cytometry ,medicine.disease ,Immunohistochemistry ,Carcinoma, Merkel Cell ,Tissue Array Analysis ,biology.protein ,Tumor Escape ,Skin cancer - Abstract
Merkel cell carcinoma (MCC) is an aggressive, polyomavirus-associated skin cancer. Robust cellular immune responses are associated with excellent outcomes in patients with MCC, but these responses are typically absent. We determined the prevalence and reversibility of major histocompatibility complex class I (MHC-I) downregulation in MCC, a potentially reversible immune-evasion mechanism. Cell-surface MHC-I expression was assessed on five MCC cell lines using flow cytometry as well as immunohistochemistry on tissue microarrays representing 114 patients. Three additional patients were included who had received intralesional IFN treatment and had evaluable specimens before and after treatment. mRNA expression analysis of antigen presentation pathway genes from 35 MCC tumors was used to examine the mechanisms of downregulation. Of note, 84% of MCCs (total n = 114) showed reduced MHC-I expression as compared with surrounding tissues, and 51% had poor or undetectable MHC-I expression. Expression of MHC-I was lower in polyomavirus-positive MCCs than in polyomavirus-negative MCCs (P < 0.01). The MHC-I downregulation mechanism was multifactorial and did not depend solely on HLA gene expression. Treatment of MCC cell lines with ionizing radiation, etoposide, or IFN resulted in MHC-I upregulation, with IFNs strongly upregulating MHC-I expression in vitro, and in 3 of 3 patients treated with intralesional IFNs. MCC tumors may be amenable to immunotherapy, but downregulation of MHC-I is frequently present in these tumors, particularly those that are positive for polyomavirus. This downregulation is reversible with any of several clinically available treatments that may thus promote the effectiveness of immune-stimulating therapies for MCC. Cancer Immunol Res; 2(11); 1071–9. ©2014 AACR.
- Published
- 2014
31. The Activated Form of the Lck Tyrosine Protein Kinase in Cells Exposed to Hydrogen Peroxide Is Phosphorylated at Both Tyr-394 and Tyr-505
- Author
-
Bartholomew M. Sefton and James S. Hardwick
- Subjects
inorganic chemicals ,T-Lymphocytes ,Blotting, Western ,Protein tyrosine phosphatase ,SH2 domain ,Biochemistry ,Catalysis ,Receptor tyrosine kinase ,SH3 domain ,Jurkat Cells ,Animals ,Humans ,Protein phosphorylation ,Src family kinase ,Phosphorylation ,Molecular Biology ,Tyrosine-protein kinase CSK ,biology ,Chemistry ,Hydrogen Peroxide ,Cell Biology ,Fibroblasts ,Rats ,Enzyme Activation ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,biology.protein ,Tyrosine ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Members of the Src family of non-receptor tyrosine protein kinases are known to be inhibited by the intramolecular association between a phosphorylated carboxyl-terminal tyrosine residue and the SH2 domain. We have previously shown that exposure of cells to H2O2 strongly activates Lck, a lymphocyte-specific Src family kinase, by inducing phosphorylation on Tyr-394, an absolutely conserved residue within the activation loop of the catalytic domain. Here we show that Lck that has been activated by H2O2 is simultaneously phosphorylated at both the carboxyl-terminal tyrosine (Tyr-505) and Tyr-394. Thus, dephosphorylation of Tyr-505 is not a prerequisite for either phosphorylation of Lck at Tyr-394 or catalytic activation of the kinase. These results indicate that activation of Lck by phosphorylation of Tyr-394 is dominant over any inhibition induced by phosphorylation of Tyr-505. We propose that these results may be extended to all Src family members.
- Published
- 1997
- Full Text
- View/download PDF
32. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma
- Author
-
Ming Qiu, Ping Wei, Zhiyu Peng, Melinda D. Willard, Ke Hao, Yingrui Li, Heather Estrella, Paul A. Rejto, Jia Ju, Hongyue Dai, Shengpei Chen, Jiangang Liu, Sheung Tat Fan, John M. Luk, James S. Hardwick, Rui Ye, Qinghui Zhang, Mariam Ehsani, Bo Wang, Isabella H. Wulur, Guan Wang, Xiaomei Fan, Jiangchun Xu, Jun Wang, Thomas D. Barber, Andrey Loboda, Lin Li, Zhao Lin, Wei Zhou, Jie Yang, Zhuolin Gong, Wing-Kin Sung, Hancheng Zheng, Ronghua Chen, Zhengyan Kan, Amit Aggarwal, Chunsheng Zhang, Julio Fernandez, Christoph Reinhard, Kang Yi, Mao Mao, Nikki P. Lee, Kai Wang, Ronnie T.P. Poon, Robert Hauptschein, Shibing Deng, Jian Wang, Huan Gao, Xiao Liu, and Shuyu Li
- Subjects
Male ,Hepatitis B virus ,Carcinoma, Hepatocellular ,Virus Integration ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Genetics ,medicine ,Carcinoma ,Humans ,Amino Acid Sequence ,Wnt Signaling Pathway ,Genetics (clinical) ,beta Catenin ,Whole genome sequencing ,Mutation ,Oncogene ,Janus kinase 1 ,Genome, Human ,Research ,Liver Neoplasms ,Wnt signaling pathway ,Janus Kinase 1 ,Sequence Analysis, DNA ,medicine.disease ,digestive system diseases ,STAT Transcription Factors ,Hepatocellular carcinoma ,DNA, Viral ,Cancer research ,Female ,Tumor Suppressor Protein p53 - Abstract
Hepatocellular carcinoma (HCC) is one of the most deadly cancers worldwide and has no effective treatment, yet the molecular basis of hepatocarcinogenesis remains largely unknown. Here we report findings from a whole-genome sequencing (WGS) study of 88 matched HCC tumor/normal pairs, 81 of which are Hepatitis B virus (HBV) positive, seeking to identify genetically altered genes and pathways implicated in HBV-associated HCC. We find beta-catenin to be the most frequently mutated oncogene (15.9%) and TP53 the most frequently mutated tumor suppressor (35.2%). The Wnt/beta-catenin and JAK/STAT pathways, altered in 62.5% and 45.5% of cases, respectively, are likely to act as two major oncogenic drivers in HCC. This study also identifies several prevalent and potentially actionable mutations, including activating mutations of Janus kinase 1 (JAK1), in 9.1% of patients and provides a path toward therapeutic intervention of the disease.
- Published
- 2013
- Full Text
- View/download PDF
33. Stimulation of Phosphorylation of Tyr394 by Hydrogen Peroxide Reactivates Biologically Inactive, Non-membrane-bound Forms of Lck
- Author
-
James S. Hardwick, Lara K. Yurchak, Kathryn Pierno, Bartholomew M. Sefton, and Kurt E. Amrein
- Subjects
inorganic chemicals ,Molecular Sequence Data ,Protein tyrosine phosphatase ,Biology ,Biochemistry ,Catalysis ,Cell Line ,Substrate Specificity ,Phosphorylation cascade ,Enzyme Reactivators ,Palmitoylation ,Animals ,Protein phosphorylation ,Amino Acid Sequence ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Base Sequence ,Cell Membrane ,Autophosphorylation ,Hydrogen Peroxide ,Cell Biology ,Rats ,src-Family Kinases ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,Tyrosine ,Fatty acylation - Abstract
Lck, a lymphocyte-specific tyrosine protein kinase, is bound to cellular membranes as the result of myristoylation and palmitoylation of its amino terminus. Its activity is inhibited by phosphorylation of tyrosine 394. The Tyr-505 --> Phe mutant of Lck (F505Lck) exhibits elevated biological activity and constitutive phosphorylation of Tyr-394 in vivo. Mutations at sites of fatty acylation that prevent F505Lck from associating with cellular membranes abolish the biological activity as a protein kinase in vivo and in vitro, and eliminate the phosphorylation of Tyr-394. Here, we show that exposure of cells expressing cytoplasmic or nuclear forms of F505Lck to H2O2, a general inhibitor of tyrosine protein phosphatases, restores the catalytic activity of these mutant proteins through stimulation of phosphorylation of Tyr-394. H2O2 treatment induced the phosphorylation of Tyr-394 therefore need not occur by autophosphorylation. Thus, there appear to be two mechanisms through which the phosphorylation of Lck at Tyr-394 can occur. One is restricted to the plasma membrane and does not require the presence of oxidants. The other is operational in the nucleus as well as the cytosol and is responsive to oxidants.
- Published
- 1996
- Full Text
- View/download PDF
34. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma
- Author
-
Angela M. Liu, Yingrui Li, Ronghua Chen, Zhao Lin, Hongyue Dai, Chunsheng Zhang, KL Chan, Guan Wang, Yujie Hu, Wen-Chi Chou, Nikki P. Lee, Wing-Kin Sung, Wah Heng Lee, Zhengyan Kan, Mao Mao, Amit Aggarwal, Jun Wang, Ronnie T.P. Poon, Thomas D. Barber, Sheung Tat Fan, Wei Zhou, James S. Hardwick, Zhuolin Gong, Jiangchun Xu, Qinghui Zhang, Hancheng Zheng, Chandana Tennakoon, John M. Luk, Xiao Liu, Ke Hao, Kwong F. Wong, Christoph Reinhard, Shuyu Li, Carolyn A. Buser, Pramila N. Ariyaratne, and Fabianus Hendriyan Mulawadi
- Subjects
Male ,Hepatitis B virus ,Carcinoma, Hepatocellular ,DNA Copy Number Variations ,Virus Integration ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Genome ,symbols.namesake ,Chromosomal Instability ,Cyclin E ,Genetics ,medicine ,Humans ,Gene ,Telomerase ,Sanger sequencing ,Oncogene Proteins ,Massive parallel sequencing ,Base Sequence ,Liver Neoplasms ,virus diseases ,Histone-Lysine N-Methyltransferase ,HCCS ,Middle Aged ,medicine.disease ,Virology ,digestive system diseases ,DNA-Binding Proteins ,Survival Rate ,Hepatocellular carcinoma ,DNA, Viral ,symbols ,RNA, Viral ,Female ,Liver cancer - Abstract
To survey hepatitis B virus (HBV) integration in liver cancer genomes, we conducted massively parallel sequencing of 81 HBV-positive and 7 HBV-negative hepatocellular carcinomas (HCCs) and adjacent normal tissues. We found that HBV integration is observed more frequently in the tumors (86.4%) than in adjacent liver tissues (30.7%). Copy-number variations (CNVs) were significantly increased at HBV breakpoint locations where chromosomal instability was likely induced. Approximately 40% of HBV breakpoints within the HBV genome were located within a 1,800-bp region where the viral enhancer, X gene and core gene are located. We also identified recurrent HBV integration events (in ≥ 4 HCCs) that were validated by RNA sequencing (RNA-seq) and Sanger sequencing at the known and putative cancer-related TERT, MLL4 and CCNE1 genes, which showed upregulated gene expression in tumor versus normal tissue. We also report evidence that suggests that the number of HBV integrations is associated with patient survival.
- Published
- 2012
35. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors
- Author
-
E. Rodríguez-Braun, Karl Hsu, James S. Hardwick, Jordi Andreu, Jordi Rodon, Ludmila Prudkin, José Baselga, Josep Tabernero, Francesco Atzori, Amparo Soler Domingo, Jorge Guijarro, Robert A. Beckman, Jason Clark, Susana Roselló, Emiliano Calvo, William D. Hanley, Andrés Cervantes, Ronald B. Langdon, Cristina Gamez, Serena Di Cosimo, and Holly Brown
- Subjects
Adult ,Male ,Cancer Research ,Maximum Tolerated Dose ,medicine.medical_treatment ,Antineoplastic Agents ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Drug Administration Schedule ,Receptor, IGF Type 1 ,Insulin-like growth factor ,Pharmacokinetics ,Neoplasms ,medicine ,Humans ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,Dalotuzumab ,business.industry ,Cancer ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Oncology ,Tolerability ,Pharmacodynamics ,Monoclonal ,Toxicity ,Female ,business - Abstract
Purpose: Insulin-like growth factor-1 receptor (IGF-1R) mediates cellular processes in cancer and has been proposed as a therapeutic target. Dalotuzumab (MK-0646) is a humanized IgG1 monoclonal antibody that binds to IGF-1R preventing receptor activation. This study was designed to evaluate the safety and tolerability of dalotuzumab, determine the pharmacokinetic (PK) and pharmacodynamic (PD) profiles, and identify a recommended phase II dose. Experimental Design: Patients with tumors expressing IGF-1R protein were allocated to dose-escalating cohorts of three or more patients each and received intravenous dalotuzumab weekly, every 2 or 3 weeks. Plasma was collected for PK analysis. Paired baseline and on-treatment skin and tumor biopsy samples were collected for PD analyses. Results: Eighty patients with chemotherapy-refractory solid tumors were enrolled. One dose-limiting toxicity was noted, but a maximum-tolerated dose was not identified. Grade 1 to 3 hyperglycemia, responsive to metformin, occurred in 15 (19%) patients. At dose levels or more than 5 mg/kg, dalotuzumab mean terminal half-life was 95 hours or more, mean Cmin was more than 25 μg/mL, clearance was constant, and serum exposures were approximately dose proportional. Decreases in tumor IGF-1R, downstream receptor signaling, and Ki67 expression were observed. 18F-Fluorodeoxy-glucose positron emission tomography metabolic responses occurred in three patients. One patient with Ewing's sarcoma showed a mixed radiologic response. The recommended phase II doses were 10, 20, and 30 mg/kg for the weekly, every other week, and every third week schedules, respectively. Conclusions: Dalotuzumab was generally well-tolerated, exhibited dose-proportional PK, inhibited IGF-1R pathway signaling and cell proliferation in treated tumors, and showed clinical activity. The low clearance rate and long terminal half-life support more extended dosing intervals. Clin Cancer Res; 17(19); 6304–12. ©2011 AACR.
- Published
- 2011
36. Single-dose fosaprepitant for the prevention of chemotherapy-induced nausea and vomiting associated with cisplatin therapy: randomized, double-blind study protocol--EASE
- Author
-
Fausto Roila, Anish Maru, Judith A. Boice, Suzanne DeVandry, Alexandra D. Carides, Elizabeth Beckford, Steven M. Grunberg, José Dinis, Arlene Taylor, James S. Hardwick, Jørn Herrstedt, and Daniel Chua
- Subjects
Adult ,Male ,Cancer Research ,medicine.drug_class ,Vomiting ,Morpholines ,Antineoplastic Agents ,Rolapitant ,Fosaprepitant ,Dexamethasone ,Drug Administration Schedule ,Ondansetron ,chemistry.chemical_compound ,Double-Blind Method ,medicine ,Antiemetic ,Netupitant ,Humans ,Aprepitant ,Aged ,Aged, 80 and over ,business.industry ,Nausea ,Middle Aged ,Regimen ,Treatment Outcome ,Oncology ,chemistry ,Anesthesia ,Antiemetics ,Female ,Cisplatin ,business ,Chemotherapy-induced nausea and vomiting ,medicine.drug - Abstract
Purpose Addition of aprepitant, a neurokinin-1 receptor antagonist (NK1RA), to an ondansetron and dexamethasone regimen improves prevention of chemotherapy-induced nausea/vomiting (CINV), particularly during the delayed phase (DP; 25 to 120 hours). Therefore, recommended antiemetic regimens include multiple-day NK1RA administration. Preliminary data suggested that single-dose aprepitant before chemotherapy could provide CINV protection throughout the overall risk phase (OP; 0 to 120 hours). This study compared a 3-day oral aprepitant schedule to a regimen containing a single dose of the intravenous NK1RA fosaprepitant. Patients and Methods A randomized, double-blind, active-control design was used to test whether fosaprepitant is noninferior to aprepitant. Patients receiving cisplatin ≥ 70 mg/m2 for the first time received ondansetron and dexamethasone with a standard aprepitant regimen (125 mg on day 1, 80 mg on day 2, 80 mg on day 3) or a single-dose fosaprepitant regimen (150 mg on day 1). The primary end point was complete response (CR; no vomiting, no rescue medication) during OP. Secondary end points were CR during DP and no vomiting during OP. Accrual of 1,113 evaluable patients per treatment arm was planned to confirm noninferiority with expected CR of 67.7% and noninferiority margin of minus 7 percentage points. Results A total of 2,322 patients were randomly assigned, and 2,247 were evaluable for efficacy. Antiemetic protection with aprepitant and fosaprepitant was equivalent within predefined bounds for noninferiority. Both regimens were well tolerated, although more frequent infusion site pain/erythema/thrombophlebitis was seen with fosaprepitant relative to aprepitant (2.7% v 0.3%, respectively). Conclusion Given with ondansetron and dexamethasone, single-dose intravenous fosaprepitant (150 mg) was noninferior to standard 3-day oral aprepitant in preventing CINV during OP and DP.
- Published
- 2011
- Full Text
- View/download PDF
37. A 4-gene signature predicts survival of patients with resected adenocarcinoma of the esophagus, junction, and gastric cardia
- Author
-
Christopher J, Peters, Jonathan R E, Rees, Richard H, Hardwick, James S, Hardwick, Sarah L, Vowler, Chin-Ann J, Ong, Chunsheng, Zhang, Vicki, Save, Maria, O'Donovan, Doris, Rassl, Derek, Alderson, Carlos, Caldas, Rebecca C, Fitzgerald, and N, Maynard
- Subjects
Oncology ,Adult ,Genetic Markers ,Male ,Pathology ,medicine.medical_specialty ,Esophageal Neoplasms ,Kaplan-Meier Estimate ,Biology ,TNM staging system ,Adenocarcinoma ,Esophagus ,Predictive Value of Tests ,Stomach Neoplasms ,Internal medicine ,medicine ,Humans ,Aged ,Aged, 80 and over ,Tissue microarray ,Hepatology ,Gene Expression Profiling ,Incidence ,ADCY9 ,Gastroenterology ,Reproducibility of Results ,Cardia ,Deoxycytidine kinase ,Gene signature ,Middle Aged ,medicine.disease ,Prognosis ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Female ,Esophagogastric Junction - Abstract
Background & Aims The incidence of esophageal and junctional adenocarcinoma has increased 6-fold in the past 30 years and 5-year survival remains approximately 20%. Current staging is limited in its ability to predict survival which has ramifications for treatment choices. The aim of this study was to generate and validate a molecular prognostic signature for esophageal adenocarcinoma. Methods Gene expression profiling was performed and the resulting 42,000 gene signatures correlated with clinical and pathologic features for 75 snap-frozen esophageal and junctional resection specimens. External validation of selected targets was performed on 371 independent cases using immunohistochemistry to maximize clinical applicability. Results A total of 119 genes were associated significantly with survival and 270 genes with the number of involved lymph nodes. Filtering of these lists resulted in a shortlist of 10 genes taken forward to validation. Four genes proved to be prognostic at the protein level (deoxycytidine kinase [DCK], 3′-phosphoadenosine 5′-phosphosulfate synthase 2 [PAPSS2], sirtuin 2 [SIRT2], and tripartite motif-containing 44 [TRIM44]) and were combined to create a molecular prognostic signature. This 4-gene signature was highly predictive of survival in the independent external validation cohort (0/4 genes dysregulated 5-year survival, 58%; 95% confidence interval [CI], 36%–80%; 1–2/4 genes dysregulated 5-year survival, 26%; 95% CI, 20%–32%; and 3–4/4 genes dysregulated 5-year survival, 14%; 95% CI, 4%–24% ( P = .001). Furthermore, this 4-gene signature was independently prognostic in a multivariable model together with the existing clinical TNM staging system ( P = .013). Conclusions This study has generated a clinically applicable prognostic gene signature that independently predicts survival in an external validation cohort and may inform management decisions.
- Published
- 2010
38. Phase I and pharmacokinetic study of vorinostat (suberoylanilide hydroxamic acid) in Japanese patients with solid tumors
- Author
-
Yutaka Fujiwara, Tetsuya Otsuki, James S. Hardwick, Shinichi Kanazu, Takashi Iwasa, Yasuhide Yamada, Noboru Yamamoto, Kazuhiko Yamada, and Tomohide Tamura
- Subjects
Adult ,Male ,Cancer Research ,Maximum Tolerated Dose ,medicine.drug_class ,Initial dose ,Anorexia ,Pharmacology ,Hydroxamic Acids ,Pharmacokinetics ,Suberoylanilide Hydroxamic Acid ,Neoplasms ,Medicine ,Humans ,Tissue Distribution ,Dosing ,Enzyme Inhibitors ,Vorinostat ,Aged ,business.industry ,Histone deacetylase inhibitor ,General Medicine ,Middle Aged ,Prognosis ,Histone Deacetylase Inhibitors ,Survival Rate ,Oncology ,Maximum tolerated dose ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
Vorinostat (suberoylanilide hydroxamic acid), a potent, oral histone deacetylase inhibitor, has demonstrated clinical activity in non-Japanese patients with various hematological and solid tumors. We sought to determine the maximum tolerated dose and a recommended phase II dose for 18 Japanese patients with solid tumors (median age, 58 years; range, 25-72 years) who failed standard therapy. Patients received vorinostat for 14 days followed by a 7-day rest. The initial dose was 100 mg twice daily escalating by 100 mg twice daily. Once-daily dosing was tested at 400 and 500 mg. A maximum tolerated dose could not be identified. Dose-limiting toxicities (thrombocytopenia, anorexia, and fatigue) were observed in two of six patients receiving 200 mg twice daily and in one of six patients receiving 500 mg once daily. In the 100-500 mg dose range, vorinostat area under the concentration-time curve increased in proportion to dose with a pharmacokinetic profile similar to that established in non-Japanese patients. Vorinostat doses of 200 mg twice daily or 500 mg once daily for 14 days followed by a 7-day rest were well tolerated and are candidate doses for phase II trials, although a maximum tolerated dose for vorinostat was not reached.
- Published
- 2009
39. Aprepitant for the prevention of chemotherapy-induced nausea and vomiting associated with a broad range of moderately emetogenic chemotherapies and tumor types: a randomized, double-blind study
- Author
-
Arlene Taylor, Judith A. Boice, James S. Hardwick, Hans-Joachim Schmoll, Timothy Webb, Bernardo Leon Rapoport, Karin Jordan, Carole Brown, and Alexandra D. Carides
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Nausea ,Vomiting ,Morpholines ,Antineoplastic Agents ,Rolapitant ,Fosaprepitant ,Dexamethasone ,chemistry.chemical_compound ,Double-Blind Method ,Internal medicine ,Neoplasms ,Netupitant ,Medicine ,Humans ,Prospective Studies ,Aprepitant ,Aged ,business.industry ,Middle Aged ,Ondansetron ,Regimen ,Treatment Outcome ,chemistry ,Anesthesia ,Antiemetics ,Drug Therapy, Combination ,Female ,medicine.symptom ,business ,medicine.drug ,Chemotherapy-induced nausea and vomiting - Abstract
Aprepitant was shown previously to be effective for prevention of chemotherapy-induced nausea and vomiting (CINV) with moderately emetogenic chemotherapy (MEC) in breast cancer patients receiving an anthracycline and cyclophosphamide (AC)-based regimen. This study assessed aprepitant in patients receiving a broad range of MEC regimens with a variety of tumor types.This phase III, randomized, gender-stratified, double-blind trial enrolled patients with confirmed malignancies, naïve to MEC or highly emetogenic chemotherapy, who were scheduled to receive a single dose of at least one MEC agent. Patients received an aprepitant triple-therapy regimen (aprepitant, ondansetron, and dexamethasone) or a control regimen (ondansetron and dexamethasone) administered orally. Primary and key secondary efficacy endpoints were proportions of patients with no vomiting and complete response (no vomiting and no rescue medication), respectively, during the 120 h post-chemotherapy.Of 848 randomized patients, 77% were female, and 52% received non-AC-based antineoplastic regimens. Significantly, more patients in the aprepitant group achieved no vomiting and complete response, regardless of whether they received AC or non-AC regimens, in the 120 h after chemotherapy. Overall, the incidences of adverse events were generally similar in the aprepitant (62.8%) and control groups (67.2%).The aprepitant regimen provided superior efficacy in the treatment of CINV in a broad range of patients receiving MEC (non-AC or AC) in both no vomiting and complete response endpoints. Aprepitant was generally well tolerated. These results show the benefit of including aprepitant as part of the standard antiemetic regimen for cancer patients receiving MEC.
- Published
- 2009
40. Inhibition of NOTCH signaling by gamma secretase inhibitor engages the RB pathway and elicits cell cycle exit in T-cell acute lymphoblastic leukemia cells
- Author
-
Sudhir Rao, Jennifer O'Neil, Lex H.T. Van der Ploeg, Nancy E. Kohl, Peter Strack, Cole Liberator, Pamela Carroll, Theresa Zhang, Victoria M. Richon, Jing Yuan, Xudong Dai, A. Thomas Look, James S. Hardwick, Edyta Tyminski, Christopher Winter, and Giulio Draetta
- Subjects
Cancer Research ,Cell signaling ,Tumor suppressor gene ,Transcription, Genetic ,Notch signaling pathway ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Transfection ,Retinoblastoma Protein ,S Phase ,Cell Line, Tumor ,Thiadiazoles ,Humans ,Protease Inhibitors ,CDKN2D ,Cyclin-Dependent Kinase Inhibitor p19 ,Phosphorylation ,Receptor, Notch1 ,Gamma secretase ,Gene Expression Profiling ,G1 Phase ,Intracellular Signaling Peptides and Proteins ,Cyclin-Dependent Kinase 4 ,Cell cycle ,Cyclic S-Oxides ,Oncology ,Cancer research ,CDKN1B ,Signal transduction ,Amyloid Precursor Protein Secretases ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction - Abstract
NOTCH signaling is deregulated in the majority of T-cell acute lymphoblastic leukemias (T-ALL) as a result of activating mutations in NOTCH1. Gamma secretase inhibitors (GSI) block proteolytic activation of NOTCH receptors and may provide a targeted therapy for T-ALL. We have investigated the mechanisms of GSI sensitivity across a panel of T-ALL cell lines, yielding an approach for patient stratification based on pathway activity and also providing a rational combination strategy for enhanced response to GSI. Whereas the NOTCH1 mutation status does not serve as a predictor of GSI sensitivity, a gene expression signature of NOTCH pathway activity does correlate with response, and may be useful in the selection of patients more likely to respond to GSI. Furthermore, inhibition of the NOTCH pathway activity signature correlates with the induction of the cyclin-dependent kinase inhibitors CDKN2D (p19INK4d) and CDKN1B (p27Kip1), leading to derepression of RB and subsequent exit from the cell cycle. Consistent with this evidence of cell cycle exit, short-term exposure of GSI resulted in sustained molecular and phenotypic effects after withdrawal of the compound. Combination treatment with GSI and a small molecule inhibitor of CDK4 produced synergistic growth inhibition, providing evidence that GSI engagement of the CDK4/RB pathway is an important mechanism of GSI action and supports further investigation of this combination for improved efficacy in treating T-ALL. [Cancer Res 2009;69(7):3060–8]
- Published
- 2009
41. Phase II Trial of Vorinostat in Recurrent Glioblastoma Multiforme: A North Central Cancer Treatment Group Study
- Author
-
Patrick J. Flynn, Andrey Loboda, Valeria R. Fantin, James A. Zwiebel, Matthew M. Ames, Caterina Giannini, Bernd W. Scheithauer, Joel M. Reid, John F. Reilly, James S. Hardwick, Victoria M. Richon, Michael Nebozhyn, Kurt A. Jaeckle, Evanthia Galanis, Matthew J. Maurer, Dennis F. Moore, and Jan C. Buckner
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_treatment ,Hydroxamic Acids ,Gastroenterology ,Histones ,Immunoenzyme Techniques ,Risk Factors ,Tissue Distribution ,Vorinostat ,Molecular Structure ,Brain Neoplasms ,Histone deacetylase inhibitor ,Area under the curve ,Acetylation ,Middle Aged ,Prognosis ,Survival Rate ,Treatment Outcome ,Oncology ,Toxicity ,Female ,Cyclin-Dependent Kinase Inhibitor p27 ,medicine.drug ,Adult ,Cyclin-Dependent Kinase Inhibitor p21 ,medicine.medical_specialty ,medicine.drug_class ,Antineoplastic Agents ,Internal medicine ,Original Reports ,medicine ,Humans ,Survival rate ,Aged ,Neoplasm Staging ,Chemotherapy ,business.industry ,Gene Expression Profiling ,medicine.disease ,Histone Deacetylase Inhibitors ,Drug Resistance, Neoplasm ,Hypernatremia ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,Progressive disease ,Follow-Up Studies - Abstract
Purpose Vorinostat, a histone deacetylase inhibitor, represents a rational therapeutic target in glioblastoma multiforme (GBM). Patients and Methods Patients with recurrent GBM who had received one or fewer chemotherapy regimens for progressive disease were eligible. Vorinostat was administered at a dose of 200 mg orally twice a day for 14 days, followed by a 7-day rest period. Results A total of 66 patients were treated. Grade 3 or worse nonhematologic toxicity occurred in 26% of patients and consisted mainly of fatigue (17%), dehydration (6%), and hypernatremia (5%); grade 3 or worse hematologic toxicity occurred in 26% of patients and consisted mainly of thrombocytopenia (22%). Pharmacokinetic analysis showed lower vorinostat maximum concentration and area under the curve (0 to 24 hours) values in patients treated with enzyme-inducing anticonvulsants, although this did not reach statistical significance. The trial met the prospectively defined primary efficacy end point, with nine of the first 52 patients being progression-free at 6 months. Median overall survival from study entry was 5.7 months (range, 0.7 to 28+ months). Immunohistochemical analysis performed in paired baseline and post-vorinostat treatment samples in a separate surgical subgroup of five patients with recurrent GBM showed post treatment increase in acetylation of histones H2B and H4 (four of five patients) and of histone H3 (three of five patients). Microarray RNA analysis in the same samples showed changes in genes regulated by vorinostat, such as upregulation of E-cadherin (P = .02). Conclusion Vorinostat monotherapy is well tolerated in patients with recurrent GBM and has modest single-agent activity. Histone acetylation analysis and RNA expression profiling indicate that vorinostat in this dose and schedule affects target pathways in GBM. Additional testing of vorinostat in combination regimens is warranted.
- Published
- 2009
42. Development of vorinostat: current applications and future perspectives for cancer therapy
- Author
-
James S. Hardwick, Jose Garcia-Vargas, and Victoria M. Richon
- Subjects
Cancer Research ,Radiation-Sensitizing Agents ,medicine.drug_class ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,Biology ,Hydroxamic Acids ,Histone Deacetylases ,Histones ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Vorinostat ,DNA Modification Methylases ,Histone deacetylase 5 ,Clinical Trials, Phase I as Topic ,HDAC11 ,Histone deacetylase 2 ,HDAC10 ,Histone deacetylase inhibitor ,Acetylation ,Histone Deacetylase Inhibitors ,Histone ,Oncology ,Cancer research ,biology.protein ,Histone deacetylase ,medicine.drug - Abstract
Vorinostat is a potent histone deacetylase inhibitor that blocks the catalytic site of these enzymes. A large number of cellular proteins are modified post-translationally by acetylation, leading to altered structure and/or function. Many of these proteins, such as core nucleosomal histones and transcription factors, function in key cellular processes and signal transduction pathways that regulate cell growth, migration, and differentiation. At concentrations that are non-toxic to normal cells, vorinostat dramatically alters cellular acetylation patterns and causes growth arrest and death and in a wide range of transformed cells, both in vitro and in animal tumor models. Vorinostat has shown promising clinical activity against hematologic and solid tumors at doses that have been well tolerated by patients. Recent non-clinical experiments that explored the effects of vorinostat in combination with other chemotherapeutic agents have begun to illuminate potential mechanisms of action for this histone deacetylase inhibitor and are providing guidance for new avenues of clinical investigation.
- Published
- 2008
43. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes
- Author
-
Valeria R. Fantin, H. Yang, Carlos E. Bueso-Ramos, Victoria M. Richon, J. Paul Secrist, Alessandra Ferrajoli, William G. Wierda, Gary L. Rosner, James S. Hardwick, Justin L. Ricker, Hagop M. Kantarjian, Guillermo Garcia-Manero, Jorge E. Cortes, Gail Morris, Sophia Randolph, Stanley R. Frankel, Charles Koller, Andrey Loboda, Cong Chen, Stefan Faderl, and John F. Reilly
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,Drug-Related Side Effects and Adverse Reactions ,medicine.drug_class ,Chronic lymphocytic leukemia ,medicine.medical_treatment ,Immunology ,Hydroxamic Acids ,Biochemistry ,Histone Deacetylases ,Histones ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Enzyme Inhibitors ,Vorinostat ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Chemotherapy ,Hematology ,Leukemia ,Clinical Trials, Phase I as Topic ,Dose-Response Relationship, Drug ,business.industry ,Myelodysplastic syndromes ,Gene Expression Profiling ,Histone deacetylase inhibitor ,Myeloid leukemia ,Acetylation ,Cell Biology ,Drug Tolerance ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Histone Deacetylase Inhibitors ,Myelodysplastic Syndromes ,Female ,business ,medicine.drug - Abstract
Vorinostat (suberoylanilide hydroxamic acid, SAHA) is a histone deacetylase inhibitor active clinically in cutaneous T-cell lymphoma and preclinically in leukemia. A phase 1 study was conducted to evaluate the safety and activity of oral vorinostat 100 to 300 mg twice or thrice daily for 14 days followed by 1-week rest. Patients with relapsed or refractory leukemias or myelodysplastic syndromes (MDS) and untreated patients who were not candidates for chemotherapy were eligible. Of 41 patients, 31 had acute myeloid leukemia (AML), 4 chronic lymphocytic leukemia, 3 MDS, 2 acute lymphoblastic leukemia, and 1 chronic myelocytic leukemia. The maximum tolerated dose (MTD) was 200 mg twice daily or 250 mg thrice daily. Dose-limiting toxicities were fatigue, nausea, vomiting, and diarrhea. Common drug-related adverse experiences were diarrhea, nausea, fatigue, and anorexia and were mild/moderate in severity. Grade 3/4 drug–related adverse experiences included fatigue (27%), thrombocytopenia (12%), and diarrhea (10%). There were no drug-related deaths; 7 patients had hematologic improvement response, including 2 complete responses and 2 complete responses with incomplete blood count recovery (all with AML treated at/below MTD). Increased histone acetylation was observed at all doses. Antioxidant gene expression may confer vorinostat resistance. Further evaluation of vorinostat in AML/MDS is warranted.
- Published
- 2007
44. Abstract A42: Palbociclib potentiates nab-paclitaxel efficacy in pancreatic ductal adenocarcinoma
- Author
-
Todd VanArsdale, Beatriz Salvador, James S. Hardwick, Camino Menéndez, Pedro P. López-Casas, Xianxian Zheng, Tao Xie, Paul A. Rejto, Peter Olson, David J. Shields, Jing Yuan, John Chionis, and Manuel Hidalgo
- Subjects
Cancer Research ,Pancreatic ductal adenocarcinoma ,endocrine system diseases ,Colorectal cancer ,business.industry ,Cancer ,Palbociclib ,medicine.disease ,medicine.disease_cause ,Gemcitabine ,Clinical trial ,Oncology ,Pancreatic cancer ,Immunology ,medicine ,Cancer research ,KRAS ,business ,medicine.drug - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, with a 5 year survival rate of less than 5%. Deaths caused by pancreatic cancer are projected to exceed the number from colorectal carcinoma by 2020, making PDAC the second leading cause of cancer-related death in the United States, behind only NSCLC. At the molecular level, PDAC is enriched for a number of genetic events central to CDK4/6:CyclinD1 control of cell cycle progression - 90% of tumors harbor oncogenic KRAS mutations, which are synthetic lethal with CDK4/6 inhibition, while the majority of PDAC cases also feature loss of p16INK4A, the endogenous inhibitor of CDK4/6. Rb loss is uncommon in PDAC and phosphorylation of Rb, the canonical CDK4/6 substrate, is detectable at high frequencies, suggesting that aberrant CDK4/6 signaling may be central to loss of cell cycle control in pancreatic cancer. To determine the significance of CDK4/6 activity in pancreatic cancer, patient-derived xenograft models of early and late stage disease were used to evaluate the selective CDK4/6 inhibitor, palbociclib. Combinatorial efficacy of palbociclib with the standard of care agents, Gemcitabine and nab-paclitaxel (Gem/nab-Pac) was also assessed. The majority of models display greater than 50% tumor growth inhibition following treatment with single agent palbociclib, which is comparable to the response to Gem/nab-Pac in those models. Addition of palbociclib to Gem/nab-Pac confers further benefit in most models by increasing the degree of tumor response on therapy and/or maintaining tumor response after drug removal. To dissect the driver activities in the triplet combination, palbociclib was assessed in combination with either Gem or nab-Pac in a series of tumor growth inhibition/delay studies. Palbociclib and nab-Pac are the dominant agents in the combination - palbociclib/nab-Pac doublet activity surpasses the anti-tumor effects of Gem/nab-Pac in the majority of models, while palbociclib/nab-Pac is equivalent or superior to the triple combination in all models. In contrast, the addition of palbociclib to Gem yields little signs of positive combinatorial activity. Based on these data, the palbociclib/nab-Pac combination will be evaluated in an upcoming clinical trial for PDAC patients. Ongoing pre-clinical studies are focused on the mechanistic and molecular basis for the robust activity of the palbociclib/nab-Pac combination in pancreatic cancer. Citation Format: Manuel Hidalgo, Camino Menendez, Jing Yuan, Beatriz Salvador, Tao Xie, John Chionis, Pedro Lopez-Casas, Xianxian Zheng, James Hardwick, Paul Rejto, Peter Olson, Todd VanArsdale, David J. Shields. Palbociclib potentiates nab-paclitaxel efficacy in pancreatic ductal adenocarcinoma. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr A42.
- Published
- 2015
- Full Text
- View/download PDF
45. Abstract A43: RB pathway disruption by the CDK4/6 inhibitor palbociclib enhances responses to chemotherapy in squamous cell lung cancer
- Author
-
Todd VanArsdale, Paul A. Rejto, James S. Hardwick, Goldie Y. L. Lui, Joan Cao, Hui Wang, Konstantinos Tsaparikos, Ping Wei, Kim Arndt, and David Shields
- Subjects
Cisplatin ,Cancer Research ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Cancer ,Cell cycle ,Palbociclib ,Pharmacology ,medicine.disease ,Oncology ,Docetaxel ,medicine ,Cancer research ,Adenocarcinoma ,Lung cancer ,business ,medicine.drug - Abstract
Lung cancer remains one of the leading causes of cancer-related mortality. Squamous cell lung cancer (SqCLC) is the second most common subtype of non-small cell lung cancer (NSCLC). Despite recent development of effective targeted therapeutic agents for lung adenocarcinoma, patients with SqCLC often receive conventional cytotoxic chemotherapy as this cancer subtype lacks genomic alterations that can be targeted by personalized medicine. Hence, novel approaches that enhance the efficacy of chemotherapy will benefit treatment outcomes in this patient population. CDK inhibitors comprise a class of drugs that targets the dysregulated cell cycle in malignant cells. Treatment of tumor cells with the CDK4/6 inhibitor palbociclib inhibits tumor growth by decreasing retinoblastoma (RB) protein phosphorylation and inducing cell cycle arrest at the G1/S phase transition. Based on promising clinical trial data, palbociclib in combination with letrozole was granted accelerated approval by the US FDA for the treatment of postmenopausal women with ER-positive, HER2-negative advanced breast cancer. In this preclinical study, we explored the effect of palbociclib on several chemotherapies (taxanes, platins, and antimetabolites) in preclinical models of SqCLC. Because the activity of chemotherapy generally requires cell cycle progression, careful combination/sequencing of this class of drugs with CDK inhibitors may be important to achieve synergy as well as avoid potential antagonism. To obtain optimal activity of palbociclib and chemotherapy combinations, we investigated several combination/sequencing regimens (concurrent, chemotherapy followed by palbociclib or the reverse sequence) in several SqCLC cell lines. We did not encounter antagonism of chemotherapy-mediated cytotoxicity by palbociclib in any of the tested regimens. Rather, we observed robust combinatorial anti-cancer cell activity in all settings. Combination of palbociclib with chemotherapy was associated with reduction of RB phosphorylation and FOXM1 protein levels, and the induction of p21. Our studies demonstrated that, while palbociclib partially antagonized chemotherapy-induced apoptosis, it significantly synergized with chemotherapy to induce cell cycle arrest as well as a senescence-like phenotype. Cells pretreated with palbociclib plus cisplatin or palbociclib plus docetaxel displayed less cell growth upon drug removal compared to those treated with monotherapies. Finally, palbociclib treatment that followed docetaxel, nab-paclitaxel or cisplatin treatment significantly enhanced the antitumor activity of the chemotherapies in several cell line-derived or patient-derived xenograft models. Our results suggest that treatment with optimal palbociclib and chemotherapy combination/sequencing could lead to better clinical outcomes for SqCLC patients. Citation Format: Ping Wei, Joan Cao, Goldie Lui, Hui Wang, Konstantinos Tsaparikos, David Shields, Kim Arndt, Paul Rejto, Todd VanArsdale, James Hardwick. RB pathway disruption by the CDK4/6 inhibitor palbociclib enhances responses to chemotherapy in squamous cell lung cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr A43.
- Published
- 2015
- Full Text
- View/download PDF
46. Abstract 1469: Patient derived xenograft (PDX) models: improving predictability of experimental cancer therapies
- Author
-
Zhengyan Kan, James S. Hardwick, Hans-Peter Gerber, Judy Lucas, Fred Immermann, Danielle Leahy, Jonathon Golas, Justin Lucas, Paul A. Rejto, Edward Rosfjord, Erik Upeslacis, Jeremy S. Myers, Puja Sapra, Eric L. Powell, Xin Han, Andrea T. Hooper, and Bingwen Lu
- Subjects
N of 1 trial ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,business.industry ,Direct transfer ,Test agent ,medicine.disease ,Preclinical data ,Internal medicine ,Medicine ,Clinical efficacy ,business ,Human cancer ,Tumor xenograft ,Progressive disease - Abstract
Clinical development of cancer therapies is associated with attrition rates as high as 80-95%. This high attrition suggests that standard preclinical pharmacology models do not accurately reflect clinical responses. The development of more predictive preclinical models requires several considerations; the relevance of the in vivo model, the administration of test agent, and the interpretation of efficacy data. PDX are cancer models developed from the direct transfer of patient tumor tissue into immunocompromised mice. A collection of PDX models, by retaining the genetic and histologic characteristics of the patients from which they were derived, represents the complexity and heterogeneity of human cancer. To minimize the clinical attrition rates of oncology compounds, we are developing hundreds of PDX models in seven major cancer indications. The collection is being molecularly profiled by RNAseq, WES, and proteomics. Profiling has identified models with robust expression of target proteins or mutant oncogenes that are likely to respond in preclinical efficacy tests. Conversely, the PDX models may provide an understanding of resistance, for example evaluating models with good target expression that fail to respond to therapy. Patient and tumor information, if known, has been collected for each PDX model including age, sex, cancer stage and grade, diagnosis, primary or metastatic site, and prior treatments. In addition to the improvements provided by the PDX models, a preclinical paradigm shift away from treatment with maximally tolerated dose towards clinically relevant dose (CRD), taking into consideration such aspects as exposure, formulation, route and schedule, is critical when attempting to predict clinical outcome from preclinical data. Also essential is the incorporation of clinically meaningful endpoints (regression) when assessing preclinical activity. We have initiated studies on cohorts of non small cell lung and breast PDX models to predict the likely clinical efficacy of candidate compounds for clinical development and to determine the CRD for standard of care (SOC) regimens required to define the most promising Phase II/III combination therapies. Anti-tumor activities were characterized using RECIST criteria of progressive disease (PD), stable disease (SD), partial response (PR), and complete response (CR). Target expression was evaluated by RNA, proteomics and immunohistochemistry. Preliminary results demonstrate a spectrum of responses against experimental therapeutics, including Phase I ADCs and are defining the CRD required for combination treatments with SOC. Identification of the most critical parameters of PDX models predicting clinical outcome will help in validating the utility of ‘n of 1′ studies with the PDX collection, inform patient enrollment strategies, guide combination therapies, and provide insight for identifying new tumor indications. Citation Format: Edward Rosfjord, Xin Han, Danielle Leahy, Erik Upeslacis, Justin Lucas, Jonathon Golas, Andrea Hooper, Fred Immermann, Bingwen Lu, Jeremy Myers, Zhengyan Kan, James Hardwick, Eric Powell, Puja Sapra, Paul Rejto, Hans-Peter Gerber, Judy Lucas. Patient derived xenograft (PDX) models: improving predictability of experimental cancer therapies. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1469. doi:10.1158/1538-7445.AM2015-1469
- Published
- 2015
- Full Text
- View/download PDF
47. Abstract 764: Rational combination of PF-06463922 (next-generation ALK inhibitor) with PI3K pathway inhibitors overcomes ALKi resistance in EML4-ALK+ NSCLC models
- Author
-
Tod Smeal, Konstantinos Tsaparikos, Nathan V. Lee, Timothy Sargis, Ming Qiu, Ping Wei, Conglin Fan, Goldie Y. L. Lui, Hui Wang, Paul A. Rejto, Justine L. Lam, James S. Hardwick, Valeria Fantin, Maruja E. Lira, and Joan Cao
- Subjects
Cancer Research ,Crizotinib ,medicine.drug_class ,business.industry ,Phases of clinical research ,Pharmacology ,Tyrosine-kinase inhibitor ,ALK inhibitor ,Oncology ,ROS1 ,medicine ,Cancer research ,Progression-free survival ,business ,Protein kinase B ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Crizotinib (PF-02341066) is a small molecule tyrosine kinase inhibitor of ALK, ROS1 and c-MET that is approved in over 70 countries for the treatment of ALK fusion positive non-small cell lung cancer (ALK+ NSCLC). Crizotinib achieved robust objective response rates of approximately 60% in ALK+ NSCLC and significantly improved progression free survival compared to chemotherapy. The emergence of secondary mutations within the ALK kinase domain or the activation of compensatory signaling pathways in crizotinib and other ALKi refractory tumors prompted searches for next generation of ALKi active against resistance mutations as single agents or in combination with other treatments. Such effort led to our recent discovery of PF-06463922, an ALK/ROS1 inhibitor with greatly improved ALK potency, brain penetration, and broad spectrum activity against all known clinical ALKi-resistant mutations. PF-06463922 is being tested in a Phase I clinical trial in both ALK+ and ROS1 fusion positive NSCLC in treatment naive or ALKi relapsed patients. In our current preclinical study, we explored rational combination strategies to further improve the efficacy of PF-06463922 in ALKi resistant cells or tumors. Our results show that compared to PF-06463922 alone, the combination of this compound with PI3K pathway inhibitors, such as PF-05212384 (PI3K/mTOR), GDC0941 (pan-PI3K) or GDC0032 (beta-sparing) leads to more robust anti-proliferative activity in vitro and greater duration of efficacy in vivo in the ALKi resistant models. These PI3K pathway inhibitors also partially overcome EGF or HGF ligand-induced resistance to PF-06463922. Interestingly, in addition to AKT signaling, both compounds inhibit ERK signaling as well, which may be essential for their enhancement of PF-06463922 cell activity or tumor efficacy in combination settings. Studies are ongoing to identify optimal partners for PF-06463922 combination using isoform selective PI3Ki, AKTi and mTORi. We are also exploring the breadth of efficacy of this combination in overcoming resistance to crizotinib, PF-06463922 or other ALKi. The findings provide important evidence that will help define the clinical development path for PF-06463922. This research effort may ultimately lead to more effective approaches to treat ALKi refractory patients in the clinic. Citation Format: Ping Wei, Ming Qiu, Nathan Lee, Joan Cao, Hui Wang, Konstantinos Tsaparikos, Conglin Fan, Timothy Sargis, Justine Lam, Maruja E. Lira, Goldie Lui, James Hardwick, Valeria Fantin, Paul Rejto, Tod Smeal. Rational combination of PF-06463922 (next-generation ALK inhibitor) with PI3K pathway inhibitors overcomes ALKi resistance in EML4-ALK+ NSCLC models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 764. doi:10.1158/1538-7445.AM2015-764
- Published
- 2015
- Full Text
- View/download PDF
48. Abstract 940: Identification of Palbociclib response signature across indications
- Author
-
Xianxian Zheng, David Shields, Zhou Zhu, Keith A. Ching, Paul A. Rejto, Todd VanArsdale, James S. Hardwick, and Mark Ozeck
- Subjects
Cancer Research ,Retinoblastoma ,Melanoma ,Estrogen receptor ,Cancer ,Cell cycle ,Biology ,Palbociclib ,medicine.disease ,Head and neck squamous-cell carcinoma ,Oncology ,medicine ,Cancer research ,E2F - Abstract
Cellular proliferation is dependent on an orderly movement through the various phases of cell cycle. Progression through the G1 phase in particular requires phosphorylation of the retinoblastoma (Rb) protein which in turn releases E2F transcription factors resulting in transcriptional activation of response gene necessary for progression into S-phase. The cyclin D-CDK4/6 signaling pathway represent a critical regulatory pathway controlling transition from G1 into S-phase and greater than 90% of human tumors have mutations in this pathway. Palbociclib is a potent, selective, and orally bioavailable inhibitor of Cdk4/6. In human tumor xenograft models, Palbociclib has significant antitumor activities. The clinical activities of Palbociclib have also be demonstrated in the phase II PALOMA-1 trial for dramatic efficacy of postmenopausal patients with locally advanced or newly diagnosed estrogen receptor (ER)-positive, HER2-negative metastatic breast cancer in combination with letrozole. To better understand the molecular mechanisms of Palbociclib response, we have identified a common set of gene signatures for ER+ BC as well as melanoma models. The physiological role of these genes regulated by Palbociclib is associated with DNA replication and repair, cell cycle, signal transduction, and Mitosis. Many of these genes have been previously identified as E2F signatures. In this study, we are aiming to expand this analysis to include two additional indications including Head and Neck squamous cell carcinoma (HNSCC) as well as squamous cell lung carcinoma (Sq lung) using Illumina RNASeq technology platform. Results from these studies will be presented and core Palbo-response signature will be discussed. In addition, we will also present the pharmacological effects of Letrozole on the gene expression changes for these core signatures in ER+ BC models. Citation Format: Xianxian Zheng, Mark Ozeck, Zhou Zhu, Keith Ching, David Shields, James Hardwick, Paul Rejto, Todd VanArsdale. Identification of Palbociclib response signature across indications. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 940. doi:10.1158/1538-7445.AM2015-940
- Published
- 2015
- Full Text
- View/download PDF
49. Abstract 3518: Growth inhibition of SCLC cell lines by treatment with LSD1 inhibitors is associated with modulation of neuroendocrine pathways
- Author
-
Shikhar Sharma, Tao Xie, Thomas A Paul, Martin James Wythes, Michael J. Greig, Jill Hallin, James S. Hardwick, Dominique Verhelle, and Timothy Nichols
- Subjects
Cancer Research ,animal structures ,Cell cycle checkpoint ,Cell ,Cancer ,Biology ,medicine.disease ,Neuroendocrine differentiation ,medicine.anatomical_structure ,Oncology ,Cell culture ,medicine ,Cancer research ,Neural cell adhesion molecule ,Epigenetics ,Cell adhesion - Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy with a high propensity for early metastasis. Response rates to first-line chemotherapy are typically high, but short lived. We describe an epigenetic-based mechanism for targeting SCLC using inhibitors for the LSD1 histone lysine demethylase. Mechanistically reversible and irreversible LSD1 inhibitors (GSK690 and OG-86) demonstrate induction of either cell cycle arrest or apoptosis in 54% of SCLC cell lines (12/22) that becomes apparent upon continuous treatment for 7-to-10 days. Maximal rates of growth inhibition in sensitive cell lines vary from 50-to-95% and plateaus between 18-to-21 days of LSD1 inhibitor treatment. Thus, heterogeneous sensitivity to LSD1 inhibitors exists between cell line models as well as within subpopulations of cells in the same cell line. To understand the mechanisms underlying LSD1 inhibitor activity in SCLC we have performed RNA-seq and ChIP-seq experiments coupled with bioinformatic analysis of expression signatures in sensitive and resistant models. Our data indicate that although LSD1 is over-expressed in SCLC cell lines and patient samples relative to non-small cell lung cancers, high LSD1 expression does not predict sensitivity to LSD1 inhibitors. Pathway analysis demonstrates that LSD1 inhibition modulates the expression of genes involved in cell adhesion and axon guidance including members of the Ephrin and Semaphorins families. At LSD1 target genes, we demonstrate site-specific H3K4me2 histone methylation changes overlapping LSD1 binding sites, however no global changes in H3K4me2 were observed. Interestingly we find morphological and cell adhesion changes in sensitive cell lines that coincide with expression changes in markers of neuroendocrine differentiation of SCLC such as GRP, NCAM, and NEUROD1. Based on these data, we propose a model that LSD1 inhibition modulates the neuroendocrine differentiation program of SCLC cells promoting tumor inhibition in sensitive SCLC models. Citation Format: Thomas A. Paul, Shikhar Sharma, Jill Hallin, Tao Xie, Timothy Nichols, Mike Greig, James Hardwick, Martin Wythes, Dominique Verhelle. Growth inhibition of SCLC cell lines by treatment with LSD1 inhibitors is associated with modulation of neuroendocrine pathways. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3518. doi:10.1158/1538-7445.AM2015-3518
- Published
- 2015
- Full Text
- View/download PDF
50. Identification of biomarkers for tumor endothelial cell proliferation through gene expression profiling
- Author
-
James S. Hardwick, Nancy E. Kohl, John R. Lamb, Hongyue Dai, Elizabeth J. Koury, Laura Sepp-Lorenzino, Rosemary C. McFall, Chunsheng Zhang, Edward Weinstein, Robert Wasserman, Bin Shi, Michael E. Severino, Yi Yang, Robert L. Phillips, and Susan L. Hill
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Time Factors ,Tumor M2-PK ,Vascular permeability ,Angiogenesis Inhibitors ,Biology ,Vascular endothelial growth inhibitor ,chemistry.chemical_compound ,Cell Line, Tumor ,Biomarkers, Tumor ,Animals ,Humans ,Neoplasm Metastasis ,Cell Proliferation ,DNA Primers ,Oligonucleotide Array Sequence Analysis ,Neovascularization, Pathologic ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Microcirculation ,Endothelial Cells ,Kinase insert domain receptor ,Molecular biology ,Immunohistochemistry ,Vascular Endothelial Growth Factor Receptor-2 ,Rats, Inbred F344 ,Rats ,Gene expression profiling ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Oncology ,chemistry ,Microscopy, Fluorescence ,Cancer research ,Endothelium, Vascular ,Tyrosine kinase ,Neoplasm Transplantation - Abstract
Extensive efforts are under way to identify antiangiogenic therapies for the treatment of human cancers. Many proposed therapeutics target vascular endothelial growth factor (VEGF) or the kinase insert domain receptor (KDR/VEGF receptor-2/FLK-1), the mitogenic VEGF receptor tyrosine kinase expressed by endothelial cells. Inhibition of KDR catalytic activity blocks tumor neoangiogenesis, reduces vascular permeability, and, in animal models, inhibits tumor growth and metastasis. Using a gene expression profiling strategy in rat tumor models, we identified a set of six genes that are selectively overexpressed in tumor endothelial cells relative to tumor cells and whose pattern of expression correlates with the rate of tumor endothelial cell proliferation. In addition to being potential targets for antiangiogenesis tumor therapy, the expression patterns of these genes or their protein products may aid the development of pharmacodynamic assays for small molecule inhibitors of the KDR kinase in human tumors.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.