36 results on '"Ziyan Y. Pessetto"'
Search Results
2. Supplementary Tables 1 - 6, Figures 1 - 4 from Drug Repurposing for Gastrointestinal Stromal Tumor
- Author
-
Andrew K. Godwin, Melinda A. Broward, Geetika Sethi, Scott J. Weir, and Ziyan Y. Pessetto
- Abstract
PDF file - 1279K, Table S1: List of FDA-approved drug library; Table S2: Screen parameters; Table S3: Curve classification criteria; Table S4: Screening of FDA-approved drugs on Hs 919.T. cell line hits summary; Table S5: Drug structures of candidate hits; Table S6: Known pharmacological data of auranofin (Prometheus Laboratories Inc. (San Diego, CA, USA); Figure S1: Drug screen assay development; Figure S2: TrxR1, TrxR2, TrxR3 transcript expression analysis from a GIST microarray dataset GS31802; Figure S3: In vitro effects of auranofin treatment on TrxR/Trx activity; Figure S4: GIST-T1, GIST T1-10R, and GIST 882 cells were treated with or without auranofin at the concentration indicated in the figure.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Drug Repurposing Identifies a Synergistic Combination Therapy with Imatinib Mesylate for Gastrointestinal Stromal Tumor
- Author
-
Andrew K. Godwin, Scott J. Weir, Margaret von Mehren, Jeff J. Hirst, Yan Ma, and Ziyan Y. Pessetto
- Abstract
Gastrointestinal stromal tumor (GIST) is a rare and therefore often neglected disease. Introduction of the kinase inhibitor imatinib mesylate radically improved the clinical response of patients with GIST; however, its effects are often short-lived, with GISTs demonstrating a median time-to-progression of approximately two years. Although many investigational drugs, approved first for other cancers, have been subsequently evaluated for the management of GIST, few have greatly affected the overall survival of patients with advanced disease. We employed a novel, focused, drug-repurposing effort for GIST, including imatinib mesylate–resistant GIST, evaluating a large library of FDA-approved drugs regardless of current indication. As a result of the drug-repurposing screen, we identified eight FDA-approved drugs, including fludarabine phosphate (F-AMP), that showed synergy with and/or overcame resistance to imatinib mesylate. F-AMP induces DNA damage, Annexin V, and caspase-3/7 activities as the cytotoxic effects on GIST cells, including imatinib mesylate–resistant GIST cells. F-AMP and imatinib mesylate combination treatment showed greater inhibition of GIST cell proliferation when compared with imatinib mesylate and F-AMP alone. Successful in vivo experiments confirmed the combination of imatinib mesylate with F-AMP enhanced the antitumor effects compared with imatinib mesylate alone. Our results identified F-AMP as a promising, repurposed drug therapy for the treatment of GISTs, with potential to be administered in combination with imatinib mesylate or for treatment of imatinib mesylate–refractory tumors. Mol Cancer Ther; 13(10); 2276–87. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Drug Repurposing for Gastrointestinal Stromal Tumor
- Author
-
Andrew K. Godwin, Melinda A. Broward, Geetika Sethi, Scott J. Weir, and Ziyan Y. Pessetto
- Abstract
Despite significant treatment advances over the past decade, metastatic gastrointestinal stromal tumor (GIST) remains largely incurable. Rare diseases, such as GIST, individually affect small groups of patients but collectively are estimated to affect 25 to 30 million people in the United States alone. Given the costs associated with the discovery, development, and registration of new drugs, orphan diseases such as GIST are often not pursued by mainstream pharmaceutical companies. As a result, “drug repurposing” or “repositioning,” has emerged as an alternative to the traditional drug development process. In this study, we screened 796 U.S. Food and Drug Administration (FDA)-approved drugs and found that two of these compounds, auranofin (Ridaura) and fludarabine phosphate, effectively and selectively inhibited the proliferation of GISTs, including imatinib-resistant cells. One of the most notable drug hits, auranofin, an oral, gold-containing agent approved by the FDA in 1985 for the treatment of rheumatoid arthritis, was found to inhibit thioredoxin reductase activity and induce reactive oxygen species (ROS) production, leading to dramatic inhibition of GIST cell growth and viability. Importantly, the anticancer activity associated with auranofin was independent of imatinib-resistant status, but was closely related to the endogenous and inducible levels of ROS. Coupled with the fact that auranofin has an established safety profile in patients, these findings suggest for the first time that auranofin may have clinical benefit for patients with GIST, particularly in those suffering from imatinib-resistant and recurrent forms of this disease. Mol Cancer Ther; 12(7); 1299–309. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figures 1 through 15 and Supplementary Table 1 from Drug Repurposing Identifies a Synergistic Combination Therapy with Imatinib Mesylate for Gastrointestinal Stromal Tumor
- Author
-
Andrew K. Godwin, Scott J. Weir, Margaret von Mehren, Jeff J. Hirst, Yan Ma, and Ziyan Y. Pessetto
- Abstract
Figure S1. Analysis of combination index (CI) value of IM combined with FDA-approved drugs in GIST-T1 cells. Figure S2. Analysis of CI value of IM combined with FDA-approved drugs in GIST 882 cells. Figure S3. Analysis of CI value of IM combined with FDA-approved drugs in GIST T1-10R cells. Figure S4. Drug combination assays using the checkerboard experimental design. Figure S5. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with idarubicin on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S6. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with digoxin on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S7. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with bortezomib on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S8. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with auranofin on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S9. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with ouabain on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S10. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with gentian violet on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S11. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with carbimazole on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S12. Representative dose-effect, median-effect and normalized isobologram (Chou-Chou Plot) plots for IM in combination with F-AMP on A) GIST-T1, B) GIST 882, and C) GIST T1-10R cells. Figure S13. Apoptosis analysis of GIST cells treated with vehicle, F-AMP, IM, or the drug combination. Figure S14. Mice body weights of each treatment group in the in vivo study. Figure S15. Representative H and E staining photographs of livers, kidneys and spleens from each treatment group in the in vivo study. Table S1. Optimized drug doses for the apoptosis assays.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figures from Licofelone Enhances the Efficacy of Paclitaxel in Ovarian Cancer by Reversing Drug Resistance and Tumor Stem-like Properties
- Author
-
Andrew K. Godwin, Katherine F. Roby, Adam J. Krieg, Devin C. Koestler, Stefan Graw, Thuc Ly, Ziyan Y. Pessetto, Stephen Hyter, Harsh B. Pathak, and Jeff Hirst
- Abstract
This document contains supplemental figures 1-17. Figure legends corresponding to each SF are on the subsequent page. Supplemental Figure 1. MCTS formation across a panel of ovarian cancer cell lines. Supplemental Figure 2. The number of proliferating cells is universally reduced in ovarian cancer cells grown as spheroids. Supplemental Figure 3. HIO cell lines form small spheroids which lack stem-like properties. Supplemental Figure 4. JMJD1A protein expression is enhanced in ovarian cancer cells when grown in 3D. Supplemental Figure 5. Targeted therapies used in ovarian cancer clinical trials induce a variable pattern of expression of stem-like associated genes. Supplemental Figure 6. CellTiter-Glo measured cell viability and drug response in MCTS. Supplemental Figure 7. 3D drug screening in non-tumorigenic cell lines did not identify 3D Hits. Supplemental Figure 8. Secondary screening validated 3D hits across multiple cell lines. Supplemental Figure 9. Dosage response validation of a 2D hit and 2D and 3D hit. Supplemental Figure 10. Licofelone showed enhanced activity in paclitaxel resistant 2D cells. Supplemental Figure 11. Licofelone had more activity against ovarian cancer cell-derived MCTS than the COX-2 specific inhibitor celecoxib. Supplemental Figure 12. Combination index values show strong synergy between with paclitaxel following licofelone pretreatment. Supplemental Figure 13. Pretreatment of ovarian cancer cell-derived MCTS with paclitaxel synergistically enhances licofelone activity. Supplemental Figure 14. Combination of licofelone and paclitaxel leads to increases in apoptosis in ovarian cancer cell-derived MCTS. Supplemental Figure 15. Licofelone can also sensitize ovarian cancer cells grown as spheroids to carboplatin. Supplemental Figure 16. Licofelone treatment decreases mitochondrial metabolism in OVCAR8 spheroids. Supplemental Figure 17. Changes in Cell Proliferation in Tumor Cells Isolated from Recurrent PDX Ascites Fluid.
- Published
- 2023
- Full Text
- View/download PDF
7. Table S1 from Licofelone Enhances the Efficacy of Paclitaxel in Ovarian Cancer by Reversing Drug Resistance and Tumor Stem-like Properties
- Author
-
Andrew K. Godwin, Katherine F. Roby, Adam J. Krieg, Devin C. Koestler, Stefan Graw, Thuc Ly, Ziyan Y. Pessetto, Stephen Hyter, Harsh B. Pathak, and Jeff Hirst
- Abstract
Expression of ABC transporter genes in 2D and in 3D cultures.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Licofelone Enhances the Efficacy of Paclitaxel in Ovarian Cancer by Reversing Drug Resistance and Tumor Stem-like Properties
- Author
-
Andrew K. Godwin, Katherine F. Roby, Adam J. Krieg, Devin C. Koestler, Stefan Graw, Thuc Ly, Ziyan Y. Pessetto, Stephen Hyter, Harsh B. Pathak, and Jeff Hirst
- Abstract
Drug development for first-line treatment of epithelial ovarian cancer (EOC) has been stagnant for almost three decades. Traditional cell culture methods for primary drug screening do not always accurately reflect clinical disease. To overcome this barrier, we grew a panel of EOC cell lines in three-dimensional (3D) cell cultures to form multicellular tumor spheroids (MCTS). We characterized these MCTS for molecular and cellular features of EOC and performed a comparative screen with cells grown using two-dimensional (2D) cell culture to identify previously unappreciated anticancer drugs. MCTS exhibited greater resistance to chemotherapeutic agents, showed signs of senescence and hypoxia, and expressed a number of stem cell–associated transcripts including ALDH1A and CD133, also known as PROM1. Using a library of clinically repurposed drugs, we identified candidates with preferential activity in MCTS over 2D cultured cells. One of the lead compounds, the dual COX/LOX inhibitor licofelone, reversed the stem-like properties of ovarian MCTS. Licofelone also synergized with paclitaxel in ovarian MCTS models and in a patient-derived tumor xenograft model. Importantly, the combination of licofelone with paclitaxel prolonged the median survival of mice (>141 days) relative to paclitaxel (115 days), licofelone (37 days), or vehicle (30 days). Increased efficacy was confirmed by Mantel–Haenszel HR compared with vehicle (HR = 0.037) and paclitaxel (HR = 0.017). These results identify for the first time an unappreciated, anti-inflammatory drug that can reverse chemotherapeutic resistance in ovarian cancer, highlighting the need to clinically evaluate licofelone in combination with first-line chemotherapy in primary and chemotherapy-refractory EOC.Significance: This study highlights the use of an in vitro spheroid 3D drug screening model to identify new therapeutic approaches to reverse chemotherapy resistance in ovarian cancer. Cancer Res; 78(15); 4370–85. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Table S3 from Licofelone Enhances the Efficacy of Paclitaxel in Ovarian Cancer by Reversing Drug Resistance and Tumor Stem-like Properties
- Author
-
Andrew K. Godwin, Katherine F. Roby, Adam J. Krieg, Devin C. Koestler, Stefan Graw, Thuc Ly, Ziyan Y. Pessetto, Stephen Hyter, Harsh B. Pathak, and Jeff Hirst
- Abstract
Supplemental Table 3. Mante-Haenszel hazard ratio shows licofelone combination increases the efficacy of paclitaxel.
- Published
- 2023
- Full Text
- View/download PDF
10. Prospective longitudinal study of kinetics of humoral response to one, two, or three doses of SARS-CoV-2 vaccine in hematopoietic cell transplant recipients
- Author
-
Qamar J, Khan, Cory R, Bivona, Ben, Liu, Maggie, Nelson, Grace A, Martin, Muhammad Umair, Mushtaq, Priyanka, Sharma, Natalie R, Streeter, Marc, Hoffmann, Gary C, Doolittle, Cuncong, Zhong, Laura, Mitchell, Kevin H, Li, Ziyan Y, Pessetto, Arnab, Ghosh, Harsh B, Pathak, Jun, Zhang, Andrew K, Godwin, and Joseph P, McGuirk
- Subjects
Kinetics ,COVID-19 Vaccines ,SARS-CoV-2 ,Hematopoietic Stem Cell Transplantation ,COVID-19 ,Humans ,Longitudinal Studies ,Prospective Studies ,Antibodies, Viral ,Transplant Recipients - Published
- 2021
11. Nano Pom-Poms Prepared Exosomes for Highly Specific Cancer Biomarker Detection
- Author
-
Sirisha Thippabhotla, Mei He, Zachary Greenberg, Liang Xu, Nan He, Cuncong Zhong, Andrew K. Godwin, Ziyan Y. Pessetto, and Yong Zeng
- Subjects
Chemistry ,medicine ,Cancer research ,Biomarker (medicine) ,Cancer ,medicine.disease ,Microvesicles - Abstract
Extracellular vesicles (EVs), particularly nano-sized small EV exosomes, are emerging biomarker sources. However, due to heterogeneous populations secreted from diverse cell types, mapping exosome multi-omic molecular information specifically to their pathogenesis origin for cancer biomarker identification is still extraordinarily challenging. Herein, we introduced a novel 3D-structured nanographene immunomagnetic particles (NanoPoms) with unique flower pom-poms morphology and photo-click chemistry for specific marker-defined capture and release of intact exosome. This specific exosome isolation approach leads to the expanded identification of targetable cancer biomarkers with enhanced specificity and sensitivity, as demonstrated by multi-omic exosome analysis of bladder cancer patient tissue fluids using the next generation sequencing of somatic DNA mutations, miRNAs, and the global proteome. The NanoPoms prepared exosomes also exhibit distinctive in vivo biodistribution patterns, highlighting the highly viable and integral quality. The developed method is simple and straightforward, which is applicable to nearly all types of biological fluids and amenable for enrichment, scale up, and high-throughput exosome isolation.
- Published
- 2021
- Full Text
- View/download PDF
12. Design, Optimization, and Multisite Evaluation of a Targeted Next-Generation Sequencing Assay System for Chimeric RNAs from Gene Fusions and Exon-Skipping Events in Non–Small Cell Lung Cancer
- Author
-
Richard A. Blidner, Gary J. Latham, Sarah Schmitt, Ziyan Y. Pessetto, Dan Su, Robyn D. Cardwell, Andrew K. Godwin, Stephen Hyter, Léon C van Kempen, Shobha Gokul, Maria Aguirre, Patrick Hurban, and Brian C. Haynes
- Subjects
0301 basic medicine ,Lung Neoplasms ,Computational biology ,Biology ,Article ,DNA sequencing ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Chimeric RNA ,Cell Line, Tumor ,Carcinoma, Non-Small-Cell Lung ,Humans ,Gene ,Sequence Analysis, RNA ,Alternative splicing ,Computational Biology ,High-Throughput Nucleotide Sequencing ,RNA ,Exons ,Exon skipping ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,RNA splicing ,Nucleic acid ,Molecular Medicine ,Gene Fusion - Abstract
Lung cancer accounts for approximately 14% of all newly diagnosed cancers and is the leading cause of cancer-related deaths. Chimeric RNA resulting from gene fusions (RNA fusions) and other RNA splicing errors are driver events and clinically addressable targets for non-small cell lung cancer (NSCLC). The reliable assessment of these RNA markers by next-generation sequencing requires integrated reagents, protocols, and interpretive software that can harmonize procedures and ensure consistent results across laboratories. We describe the development and verification of a system for targeted RNA sequencing for the analysis of challenging, low-input solid tumor biopsies that includes reagents for nucleic acid quantification and library preparation, run controls, and companion bioinformatics software. Assay development reconciled sequence discrepancies in public databases, created predictive formalin-fixed, paraffin-embedded RNA qualification metrics, and eliminated read misidentification attributable to index hopping events on the next-generation sequencing flow cell. The optimized and standardized system was analytically verified internally and in a multiphase study conducted at five independent laboratories. The results show accurate, reproducible, and sensitive detection of RNA fusions, alternative splicing events, and other expression markers of NSCLC. This comprehensive approach, combining sample quantification, quality control, library preparation, and interpretive bioinformatics software, may accelerate the routine implementation of targeted RNA sequencing of formalin-fixed, paraffin-embedded samples relevant to NSCLC.
- Published
- 2019
- Full Text
- View/download PDF
13. Nano Pom-poms Prepared Highly Specific Extracellular Vesicles Expand the Detectable Cancer Biomarkers
- Author
-
Ziyan Y. Pessetto, Yong Zeng, Nan He, Cuncong Zhong, Mei He, Andrew K. Godwin, Liang Xu, Zachary Greenberg, and Sirisha Thippabhotla
- Subjects
Cell type ,Somatic cell ,Chemistry ,microRNA ,Proteome ,medicine ,Cancer ,Cancer biomarkers ,medicine.disease ,Microvesicles ,Biomarker (cell) ,Cell biology - Abstract
Extracellular vesicles (EVs), particularly exosomes, are emerging biomarker sources. However, due to heterogeneous populations secreted from diverse cell types, mapping EV multi-omic molecular information specifically to their pathogenesis origin for cancer biomarker identification is still extraordinary challenging. Herein, we introduced a novel 3D-structured nanographene immunomagnetic particles (NanoPoms) with unique flower pom-poms morphology and photo-click chemistry for specific marker-defined capture and release of intact small EVs. This specific EV isolation approach leads to the expanded identification of targetable cancer biomarkers with enhanced specificity and sensitivity, as demonstrated by multi-omic EV analysis of bladder cancer patient tissue fluids using the next generation sequencing of somatic DNA mutations, miRNAs, and the global proteome. The NanoPoms prepared sEVs also exhibit distinctive in vivo biodistribution patterns, highlighting the highly viable and integral quality. The developed method is simple and straightforward, and is applicable to nearly all types of biological fluids and amenable for scale up and high-throughput EV isolation.
- Published
- 2021
- Full Text
- View/download PDF
14. WJMSC‐derived small extracellular vesicle enhance T cell suppression through PD‐L1
- Author
-
Neil Dunavin, Meizhang Li, Rupal Soder, Haitham Abdelhakim, Siddhartha Ganguly, Joseph P. McGuirk, Andrew K. Godwin, Buddhadeb Dawn, Ziyan Y. Pessetto, Harsh B. Pathak, Mitchell W. Braun, Camille V. Trinidad, Sunil Abhyankar, and Clayton Deighan
- Subjects
0301 basic medicine ,viruses ,T-Lymphocytes ,Graft vs Host Disease ,Exosomes ,Lymphocyte Activation ,B7-H1 Antigen ,Gene Knockout Techniques ,0302 clinical medicine ,Pregnancy ,Wharton Jelly ,Research Articles ,Cells, Cultured ,biology ,Chemistry ,virus diseases ,Extracellular vesicle ,acute graft‐versus‐host disease ,respiratory system ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cytokines ,Female ,Research Article ,Adult ,Histology ,Adolescent ,T cell ,Receptors, Antigen, T-Cell ,Wharton's Jelly‐derived mesenchymal stem cells ,small extracellular vesicles ,03 medical and health sciences ,Extracellular Vesicles ,Young Adult ,Immune system ,PD-L1 ,medicine ,Humans ,QH573-671 ,T-cell receptor ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Immune checkpoint ,Microvesicles ,030104 developmental biology ,PD‐L1 ,Cancer research ,biology.protein ,T cell receptor ,Cytology - Abstract
Both mesenchymal stem cells (MSCs) and their corresponding small extracellular vesicles (sEVs, commonly referred to as exosomes) share similar immunomodulatory properties that are potentially beneficial for the treatment of acute graft versus host disease (aGvHD). We report that clinical grade Wharton's Jelly‐derived MSCs (WJMSCs) secrete sEVs enriched in programmed death‐ligand 1 (PD‐L1), an essential ligand for an inhibitory immune checkpoint. A rapid increase in circulating sEV‐associated PD‐L1 was observed in patients with aGvHD and was directly associated with the infusion time of clinical grade WJMSCs. In addition, in vitro inhibitory antibody mediated blocking of sEV‐associated PD‐L1 restored T cell activation (TCA), suggesting a functional inhibitory role of sEVs‐PD‐L1. PD‐L1‐deficient sEVs isolated from WJMSCs following CRISPR‐Cas9 gene editing fail to inhibit TCA. Furthermore, we found that PD‐L1 is essential for WJMSC‐derived sEVs to modulate T cell receptors (TCRs). Our study reveals an important mechanism by which therapeutic WJMSCs modulate TCR‐mediated TCA through sEVs or sEV‐carried immune checkpoints. In addition, our clinical data suggest that sEV‐associated PD‐L1 may be not only useful in predicting the outcomes from WJMSC clinical administration, but also in developing cell‐independent therapy for aGvHD patients.
- Published
- 2021
15. Cancer Therapy and Immunogenicity of COVID Vaccine – CANINE Study
- Author
-
Qamar J. Khan, Cory R. Bivona, Grace A. Martin, Jun Zhang, Ben Liu, Jianghua He, Kevin H. Li, Maggie Nelson, Stephen Williamson, Gary C. Doolittle, Weijing Sun, Dinesh Pal Mudaranthakam, Natalie R. Streeter, Joseph McGuirk, Raed Al-Rajabi, Marc Hoffmann, Anup Kasi, Rahul A. Parikh, Cocong Zhong, Laura Mitchell, Ziyan Y. Pessetto, Harsh Pathak, Ghosh Arnab, Stephanie LaFaver, Priyanka Sharma, and Andrew K. Godwin
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2021
- Full Text
- View/download PDF
16. Adherent cell depletion promotes the expansion of renal cell carcinoma infiltrating T cells with optimal characteristics for adoptive transfer
- Author
-
Andrew K. Godwin, Haitham Abdelhakim, Ziyan Y. Pessetto, Harsh B. Pathak, Devin C. Koestler, Mitchell W. Braun, Neil Dunavin, Meizhang Li, and Stephen Hyter
- Subjects
lymphocytes ,Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,T lymphocytes ,therapies ,chemical and pharmacologic phenomena ,investigational ,Immunotherapy, Adoptive ,Flow cytometry ,Lymphocytes, Tumor-Infiltrating ,TIGIT ,Aldesleukin ,medicine ,Immunology and Allergy ,Humans ,IL-2 receptor ,Carcinoma, Renal Cell ,RC254-282 ,tumor infiltrating ,Pharmacology ,medicine.diagnostic_test ,Immune Cell Therapies and Immune Cell Engineering ,Chemistry ,FOXP3 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,hemic and immune systems ,Immunotherapy ,Kidney Neoplasms ,Oncology ,Cancer research ,Molecular Medicine ,immunotherapy ,CD8 - Abstract
BackgroundTumor-infiltrating lymphocyte (TIL) therapy is a personalized cancer treatment which involves generating ex vivo cultures of tumor-reactive T cells from surgically resected tumors and administering the expanded TILs as a therapeutic infusion. Phase 1 of many TIL production protocols use aldesleukin (IL-2) alone to establish TIL cultures (termed “PreREP” (Pre-Rapid Expansion Protocol)); however, this fails to consistently produce TIL cultures from renal cell carcinoma (RCC) in a timely manner. Adding mitogenic stimulation via anti-CD3/anti-CD28 beads along with IL-2 to the fresh tumor digest (FTD) during TIL generation (termed “FTD+ beads”) increases successful TIL culture rates; however, T cells produced by this method may be suboptimal for adoptive transfer. We hypothesize that adherent cell depletion (ACD) before TIL expansion will produce a superior TIL product by removing the immunosuppressive signals originating from adherent tumor and stromal cells. Here we investigate if “panning,” a technique for ACD prior to TIL expansion, will impact the phenotype, functionality and/or clonality of ex vivo expanded RCC TILs.MethodsTumor specimens from 55 patients who underwent radical or partial nephrectomy at the University of Kansas Medical Center (KUMC) were used to develop the panning method and an additional 19 specimens were used to validate the protocol. Next-generation sequencing, immunohistochemistry/immunocytochemistry and flow cytometry were used during method development. The phenotype, functionality and clonality of autologous TILs generated in parallel by panning, PreREP, and FTD+ beads were assessed by flow cytometry, in vitro co-culture assays, and TCRB CDR3 sequencing.ResultsTIL cultures were successfully generated using the panning protocol from 15/16 clear cell, 0/1 chromophobe, and 0/2 papillary RCC samples. Significantly fewer regulatory (CD4+/CD25+/FOXP3+) (p=0.049, p=0.005), tissue-resident memory (CD8+/CD103+) (p=0.027, p=0.009), PD-1+/TIM-3+ double-positive (p=0.009, p=0.011) and TIGIT+ T cells (p=0.049, p=0.026) are generated by panning relative to PreREP and FTD+ beads respectively. Critically, a subset of TILs generated by panning were able to degranulate and/or produce interferon gamma in response to autologous tumor cells and the average tumor-reactive TIL yield was greatest when using the panning protocol.ConclusionsRemoving immunosuppressive adherent cells within an RCC digest prior to TIL expansion allow for the rapid production of tumor-reactive T cells with optimal characteristics for adoptive transfer.
- Published
- 2020
17. Role of miR-139 as a surrogate marker for tumor aggression in breast cancer
- Author
-
Dan Gallagher, Hongyan Dai, Fang Fan, Andrew K. Godwin, Ziyan Y. Pessetto, Ossama Tawfik, and Sarah Schmitt
- Subjects
0301 basic medicine ,CA15-3 ,Oncology ,medicine.medical_specialty ,Proliferation index ,Biopsy ,Down-Regulation ,CA 15-3 ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Risk Factors ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Neoplasm Invasiveness ,Cell Proliferation ,Retrospective Studies ,Reverse Transcriptase Polymerase Chain Reaction ,Carcinoma, Ductal, Breast ,Middle Aged ,medicine.disease ,Survival Analysis ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Phenotype ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Disease Progression ,Biomarker (medicine) ,Female ,Neoplasm Grading ,Carcinogenesis - Abstract
MicroRNAs are non-protein coding molecules that play a key role in oncogenesis, tumor progression, and metastasis in many types of malignancies including breast cancer. In the current study, we studied the expression of microRNA-139-5p (miR-139) in invasive ductal carcinoma (IDC) of the breast and correlated its expression with tumor grade, molecular subtype, hormonal status, human epidermal growth factor receptor 2 status, proliferation index, tumor size, lymph node status, patient's age, and overall survival in 74 IDC cases. In addition, we compared and correlated miR-139 expression in 18 paired serum and tissue samples from patients with IDC to assess its value as a serum marker. Our data showed that miR-139 was down-regulated in all tumor tissue samples compared with control. More pronounced down-regulation was seen in tumors that were higher grade, estrogen receptor negative, progesterone receptor negative, more proliferative, or larger in size (P.05). Although not statistically significant, lower miR-139 level was frequently associated with human epidermal growth factor receptor 2 overexpression. In addition, significantly lower miR-139 tissue level was seen in patients who were deceased (P = .027), although older age (50 years) and positive local nodal disease did not adversely affect miR-139 expression. In contrast, serum miR-139 profile of the patients appeared similar to that of normal control. In conclusion, our study demonstrated that down-regulation of miR-139 was associated with aggressive tumor behavior and disease progression in breast cancer. miR-139 may serve as a risk assessment biomarker in tailoring treatment options.
- Published
- 2017
- Full Text
- View/download PDF
18. Abstract P13: Prospective voluntary SARS-CoV-2 virus and anti-COVID-19 antibody tests in asymptomatic medical and research staff who work in direct contact with cancer patients: A single center study
- Author
-
Elizabeth Marie Wulff-Burchfield, Natalie Streeter, B.J. Broome, Roy A. Jensen, Tara L. Lin, Terry Tsue, Weijing Sun, Anup Kasi, Joseph McGuirk, Kathan Mehta, Ziyan Y. Pessetto, Andrew K. Godwin, Adam Pessetto, and Joaquina Baranda
- Subjects
Cancer Research ,medicine.medical_specialty ,biology ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Cancer ,Single Center ,medicine.disease ,Asymptomatic ,Virus ,Oncology ,Internal medicine ,biology.protein ,Medicine ,Antibody ,medicine.symptom ,business - Abstract
Background The SARS–CoV-2 pandemic has assaulted all aspects of daily life. Medical professionals in oncology face additional challenges with balancing prompt cancer diagnosis and urgent treatment against potential COVID-19 exposure risk in these high-risk patients. We designed this prospective freewill study to offer testing for SAR2-CoV-2 viral RNA and/or anti-COVID-19, respectively in asymptomatic medical and research staff who work in direct contact with cancer patients. The overall goal was to evaluate the prevalence of infection in this group of asymptomatic healthcare providers to reduce exposure of cancer patients to asymptomatic staff. Methods Asymptomatic medical and research staff who work in direct contact with cancer patients were asked to voluntarily be tested for either SARS–CoV-2 viral RNA or antibodies or both. Either NP swabs and/or blood samples (EDTA tube) were collected. Tests are performed at Sinochips Kansas LLC, Sinochips Diagnostics (CLIA number:17D2176068, CAP number: 8709463). The PCR test is performed with FDA authorized 2019-Novel Coronavirus (2019-nCoV) Real-Time RT-PCR Diagnostic Panel EUA. The Elecsys® Anti-SARS-CoV-2 (Roche Diagnostics) immunoassay was used to qualitative detection of antibodies to SARS-CoV-2 in human plasma. Results From 06/18/2020 to 12/18/2020, 861 participated in the study. 1095 tests were completed for SAR2-CoV-2 virus infection, and 918 were completed for antibody. Amount participants, 530 had both virus and antibody tested. 235 were tested more than once for viral infection and 166 were tested more than once for the antibody. Median age of participants was 39 years (IQR 32-51 years). Among these 84.7% were females, 84.4% white, 6.7% African American, 4.8% Asian and 84.7% non-Hispanic. The cumulative incidence of a positive test for the virus was 2.2% (16/712), and for the antibody test was 3.8% (26/679). 5 had both viral and antibody tests positive, with an average time of 4.1 weeks from viral testing positivity to detectable antibody among 3 cases and 2 cases with both viral infection and antibody detected at same time. There were 3 cases virus was detected more than once after turning positive. 2 remained positive at 16 and 22 days after initial test and one turned negative at 36 days as of last follow up. There were 7 cases where the antibody was tested more than once after turning positive and all 7 remained positive as of last follow up (range 7-103 days). Conclusion Prospective voluntary testing in asymptomatic medical and research staff who work in direct contact with cancer patients was feasible and resulted in identification of asymptomatic carriers who then placed in quarantine, thereby limiting exposure to cancer patients. Medical and research staff who work with cancer patients are general very cautious and the frequency of infections were significantly lower than general society. In addition, it seems that 1) virus and antibody may co-exist in the same person after exposure, and 2) the antibody may last for a relatively long time. Citation Format: Weijing Sun, Andrew K. Godwin, Kathan Mehta, Natalie Streeter, Elizabeth Wulff-Burchfield, Anup Kasi, Tara L. Lin, Joaquina Baranda, Joseph McGuirk, Ziyan Pessetto, Adam Pessetto, B.J. Broome, Terry Tsue, Roy Jensen. Prospective voluntary SARS-CoV-2 virus and anti-COVID-19 antibody tests in asymptomatic medical and research staff who work in direct contact with cancer patients: A single center study [abstract]. In: Proceedings of the AACR Virtual Meeting: COVID-19 and Cancer; 2021 Feb 3-5. Philadelphia (PA): AACR; Clin Cancer Res 2021;27(6_Suppl):Abstract nr P13.
- Published
- 2021
- Full Text
- View/download PDF
19. Overall survival based on MSI and BRAF mutation status for stage II/III colorectal cancer
- Author
-
Sophiya Karki, Rashna Madan, Anup Kasi, Weijing Sun, Sarah Schmitt, Ziyan Y. Pessetto, and Andrew K. Godwin
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,business.industry ,Colorectal cancer ,Stage ii ,medicine.disease ,digestive system diseases ,BRAF V600E ,Internal medicine ,Mutation (genetic algorithm) ,medicine ,Overall survival ,skin and connective tissue diseases ,business ,neoplasms - Abstract
132 Background: Colorectal cancer (CRC) is the second leading cause of cancer-associated deaths in the United States. Some of the poor prognostic factors for metastatic CRC (mCRC) include BRAF V600E mutation and microsatellite instability (MSI) that result from mutation or loss of mismatch-repair genes. While the prognostic value of MSI-high CRC for early-stage patients treated with resection and adjuvant chemotherapy is favorable, the prognostic value of BRAF mutation is still unclear. Furthermore, the impact of BRAF mutation with concurrent microsatellite instability on overall survival has not been well investigated. Methods: Here, we collected BRAF mutation status and MSI status of stage II/III CRC patients (n=106) treated at the University of Kansas Cancer Center between September 2009 and July 2020 and compared overall survival between 4 subtypes:MSI-H/BRAF mutant (n=16), MSS/BRAF mutant (n=4), MSI-H/BRAF WT (n=17) and MSS/BRAF WT (n=69), further stratifying patients by age at diagnosis and tumor location. Molecular data were obtained from molecular oncology laboratory as PCR or IHC-based or acquired from outside records. Subgroup analyses were done for stage II and stage III cancers. Results: Table shows the patient characteristics. From our preliminary analysis, MSI-H CRC was found to be primarily a right-sided tumor (MSI-H/BRAF mutant: 94% and MSI-H/BRAF WT 76%). On the contrary, MSS CRC had a more heterogenous localization, spanning left colon, right colon and rectum. In our patient cohort, median survival was not reached for stage II patients whereas for stage III patients, BRAF mutation was associated with poor median survival irrespective of MSI status (MSS/BRAF mutant: 27 months and MSI-H/BRAF mutant 29 months). Median overall survival was found to be 87 months, not reached, 27 months and 29 months for MSS/BRAF WT, MSI-H/BRAF WT, MSS/BRAF mutant and MSI-H/BRAF mutant, respectively. Although associated with poor survival, MSI-H/BRAF mutant displayed later age at diagnosis (mean age 73) compared to MSS/BRAF mutant (mean age 60, p-value
- Published
- 2021
- Full Text
- View/download PDF
20. Acute myeloid leukemia or myelodysplastic syndrome with chromosome 17 abnormalities and long-term outcomes with or without hematopoietic stem cell transplantation
- Author
-
Siddhartha Ganguly, Joseph P. McGuirk, Leyla Shune, Barry S. Skikne, Shivani Golem, Clint Divine, Tara L. Lin, Ghulam Rehman Mohyuddin, Andrew K. Godwin, Ajoy Dias, Sunil Abhyankar, Brian McClune, Ziyan Y. Pessetto, Anurag K. Singh, and Alec Britt
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Gene mutation ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,Humans ,Medicine ,Aged ,Retrospective Studies ,Aged, 80 and over ,Chromosome Aberrations ,business.industry ,Hematopoietic Stem Cell Transplantation ,Induction chemotherapy ,Myeloid leukemia ,Retrospective cohort study ,Hematology ,Middle Aged ,Prognosis ,Chromosome 17 (human) ,Leukemia, Myeloid, Acute ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Concomitant ,Female ,Tumor Suppressor Protein p53 ,business ,Chromosomes, Human, Pair 17 ,030215 immunology - Abstract
Introduction Chromosome 17 abnormalities, especially disorders of the 17p region and including TP53 gene mutations, result in very low rates of cure for patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) treated with conventional chemotherapy or allogeneic hematopoietic cell transplant (allo-HCT). Our retrospective study analyzed outcomes in patients with chromosome 17 (ch17) abnormalities who received conventional chemotherapy followed by allo-HCT versus those who did not receive a transplant. We analyzed whether poor outcomes extend to patients with all types of ch17 abnormalities and the impact of concomitant TP53 gene mutations assessed by next-generation sequencing (NGS) on prognosis. Methods We retrospectively analyzed diagnostic and outcome data on 98 patients treated at our institution from 2012 to 2018 with AML or MDS who possessed ch17 abnormalities by cytogenetic analysis. The presence of TP53 mutations was analyzed by NGS. Primary endpoint of our study was overall survival (OS). Results 61 patients with AML and 37 with MDS were included. Complete remission (CR) with first line treatment was similar between induction chemotherapy or hypomethylating agents (HMA), 22.9 % versus 21.6 % (p = 0.33). Median OS for all patients (with or without transplant) was 10 months. Patients with abnormal ch17 in conjunction with any TP53 mutation(s) exhibited worse OS compared to patients without a TP53 mutation (10 versus 23 months, p = 0.02). 30 patients (19 AML, 11 MDS) underwent HCT, with a median OS of 11 months. For AML patients who underwent allo-HCT, 18 were in CR (13 with cytogenetic remission) and 1 had persistent disease at transplant. In the MDS cohort, 3 patients were in CR (2 with cytogenetic remission) and 8 had stable disease. Post allo-HCT survival of AML and MDS cohorts did not differ (p = 0.6), although cytogenetic CR at time of HCT trended towards improved OS (17 versus 8 months; p = 0.6). Conclusions AML/MDS patients with ch17 abnormalities have poor outcomes with or without HCT. Our results show that patients with ch17 abnormalities and TP53 mutations have a significantly poorer survival compared to patients who have ch17 abnormalities but no TP53 mutations. Drugs targeting abnormalities of the p53 pathway, improvement in depth of response prior to HCT, and novel maintenance strategies are needed for improved outcomes in these patients.
- Published
- 2020
- Full Text
- View/download PDF
21. Licofelone enhances the efficacy of paclitaxel in ovarian cancer by reversing drug resistance and tumor stem-like properties
- Author
-
Andrew K. Godwin, Adam J. Krieg, Stefan Graw, Thuc Ly, Jeff Hirst, Devin C. Koestler, Ziyan Y. Pessetto, Harsh B. Pathak, Katherine F. Roby, and Stephen Hyter
- Subjects
0301 basic medicine ,Cancer Research ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Cancer ,Drug resistance ,medicine.disease ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Paclitaxel ,chemistry ,Drug development ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,Ovarian cancer ,Licofelone - Abstract
Drug development for first-line treatment of epithelial ovarian cancer (EOC) has been stagnant for almost three decades. Traditional cell culture methods for primary drug screening do not always accurately reflect clinical disease. To overcome this barrier, we grew a panel of EOC cell lines in three-dimensional (3D) cell cultures to form multicellular tumor spheroids (MCTS). We characterized these MCTS for molecular and cellular features of EOC and performed a comparative screen with cells grown using two-dimensional (2D) cell culture to identify previously unappreciated anticancer drugs. MCTS exhibited greater resistance to chemotherapeutic agents, showed signs of senescence and hypoxia, and expressed a number of stem cell–associated transcripts including ALDH1A and CD133, also known as PROM1. Using a library of clinically repurposed drugs, we identified candidates with preferential activity in MCTS over 2D cultured cells. One of the lead compounds, the dual COX/LOX inhibitor licofelone, reversed the stem-like properties of ovarian MCTS. Licofelone also synergized with paclitaxel in ovarian MCTS models and in a patient-derived tumor xenograft model. Importantly, the combination of licofelone with paclitaxel prolonged the median survival of mice (>141 days) relative to paclitaxel (115 days), licofelone (37 days), or vehicle (30 days). Increased efficacy was confirmed by Mantel–Haenszel HR compared with vehicle (HR = 0.037) and paclitaxel (HR = 0.017). These results identify for the first time an unappreciated, anti-inflammatory drug that can reverse chemotherapeutic resistance in ovarian cancer, highlighting the need to clinically evaluate licofelone in combination with first-line chemotherapy in primary and chemotherapy-refractory EOC. Significance: This study highlights the use of an in vitro spheroid 3D drug screening model to identify new therapeutic approaches to reverse chemotherapy resistance in ovarian cancer. Cancer Res; 78(15); 4370–85. ©2018 AACR.
- Published
- 2018
22. Beyond standard therapy: drugs under investigation for the treatment of gastrointestinal stromal tumor
- Author
-
Hani J. Alturkmani, Andrew K. Godwin, and Ziyan Y. Pessetto
- Subjects
Oncology ,medicine.medical_specialty ,Gastrointestinal Stromal Tumors ,medicine.drug_class ,medicine.medical_treatment ,Antineoplastic Agents ,PDGFRA ,Pharmacology ,Malignancy ,Article ,Tyrosine-kinase inhibitor ,Targeted therapy ,Internal medicine ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,Stromal tumor ,neoplasms ,Gastrointestinal Neoplasms ,GiST ,business.industry ,Drugs, Investigational ,General Medicine ,medicine.disease ,digestive system diseases ,Survival Rate ,Clinical trial ,Imatinib mesylate ,Drug Design ,Disease Progression ,business - Abstract
Gastrointestinal stromal tumor (GIST) is the most common nonepithelial malignancy of the GI tract. With the discovery of KIT and later platelet-derived growth factor α (PDGFRA) gain-of-function mutations as factors in the pathogenesis of the disease, GIST was the quintessential model for targeted therapy. Despite the successful clinical use of imatinib mesylate, a selective receptor tyrosine kinase (RTK) inhibitor that targets KIT, PDGFRA and BCR-ABL, we still do not have treatment for the long-term control of advanced GIST.This review summarizes the drugs that are under investigation or have been assessed in trials for GIST treatment. The article focuses on their mechanisms of actions, the preclinical evidence of efficacy, and the clinical trials concerning safety and efficacy in humans.It is known that KIT and PDGFRA mutations in GIST patients influence the response to treatment. This observation should be taken into consideration when investigating new drugs. RECIST was developed to help uniformly report efficacy trials in oncology. Despite the usefulness of this system, many questions are being addressed about its validity in evaluating the true efficacy of drugs knowing that new targeted therapies do not affect the tumor size as much as they halt progression and prolong survival.
- Published
- 2015
- Full Text
- View/download PDF
23. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study
- Author
-
Stephen K. Williamson, Kishore Polireddy, Jun Yu, Ziyan Y. Pessetto, Gregory A. Reed, Mark Levine, Jeanne Drisko, Ruochen Dong, Qi Chen, Ping Chen, Andrew K. Godwin, Pierre-Christian Violet, and Fang Fan
- Subjects
0301 basic medicine ,Programmed cell death ,Mice, Nude ,lcsh:Medicine ,Apoptosis ,Ascorbic Acid ,Deoxycytidine ,Article ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Animals ,Humans ,Medicine ,Cytotoxic T cell ,Infusions, Parenteral ,Tissue Distribution ,Prospective Studies ,Neoplasm Metastasis ,lcsh:Science ,Cell Proliferation ,Multidisciplinary ,business.industry ,lcsh:R ,Cancer ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Gemcitabine ,3. Good health ,Pancreatic Neoplasms ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Toxicity ,cardiovascular system ,Cancer research ,Female ,lcsh:Q ,business ,Follow-Up Studies ,medicine.drug - Abstract
Pancreatic cancer is among the most lethal cancers with poorly tolerated treatments. There is increasing interest in using high-dose intravenous ascorbate (IVC) in treating this disease partially because of its low toxicity. IVC bypasses bioavailability barriers of oral ingestion, provides pharmacological concentrations in tissues, and exhibits selective cytotoxic effects in cancer cells through peroxide formation. Here, we further revealed its anti-pancreatic cancer mechanisms and conducted a phase I/IIa study to investigate pharmacokinetic interaction between IVC and gemcitabine. Pharmacological ascorbate induced cell death in pancreatic cancer cells with diverse mutational backgrounds. Pharmacological ascorbate depleted cellular NAD+ preferentially in cancer cells versus normal cells, leading to depletion of ATP and robustly increased α-tubulin acetylation in cancer cells. While ATP depletion led to cell death, over-acetylated tubulin led to inhibition of motility and mitosis. Collagen was increased, and cancer cell epithelial-mesenchymal transition (EMT) was inhibited, accompanied with inhibition in metastasis. IVC was safe in patients and showed the possibility to prolong patient survival. There was no interference to gemcitabine pharmacokinetics by IVC administration. Taken together, these data revealed a multi-targeting mechanism of pharmacological ascorbate’s anti-cancer action, with minimal toxicity, and provided guidance to design larger definitive trials testing efficacy of IVC in treating advanced pancreatic cancer.
- Published
- 2017
- Full Text
- View/download PDF
24. Developing a genetic signature to predict drug response in ovarian cancer
- Author
-
Rama Raghavan, Jeff Hirst, Devin C. Koestler, Andrew K. Godwin, Stephen Hyter, Dong Pei, Ziyan Y. Pessetto, and Harsh B. Pathak
- Subjects
0301 basic medicine ,Drug ,Auranofin ,Bevacizumab ,media_common.quotation_subject ,In silico ,gene signature ,03 medical and health sciences ,Ovarian tumor ,0302 clinical medicine ,medicine ,ovarian ,media_common ,business.industry ,AUY922 ,auranofin ,Gene signature ,TCGA ,medicine.disease ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Pharmacogenomics ,Cancer research ,Ovarian cancer ,business ,medicine.drug ,Research Paper - Abstract
There is a lack of personalized treatment options for women with recurrent platinum-resistant ovarian cancer. Outside of bevacizumab and a group of poly ADP-ribose polymerase inhibitors, few options are available to women that relapse. We propose that efficacious drug combinations can be determined via molecular characterization of ovarian tumors along with pre-established pharmacogenomic profiles of repurposed compounds. To that end, we selectively performed multiple two-drug combination treatments in ovarian cancer cell lines that included reactive oxygen species inducers and HSP90 inhibitors. This allowed us to select cell lines that exhibit disparate phenotypes of proliferative inhibition to a specific drug combination of auranofin and AUY922. We profiled altered mechanistic responses from these agents in both reactive oxygen species and HSP90 pathways, as well as investigated PRKCI and lncRNA expression in ovarian cancer cell line models. Generation of dual multi-gene panels implicated in resistance or sensitivity to this drug combination was produced using RNA sequencing data and the validity of the resistant signature was examined using high-density RT-qPCR. Finally, data mining for the prevalence of these signatures in a large-scale clinical study alluded to the prevalence of resistant genes in ovarian tumor biology. Our results demonstrate that high-throughput viability screens paired with reliable in silico data can promote the discovery of effective, personalized therapeutic options for a currently untreatable disease.
- Published
- 2017
25. The prognosis of NF1 mutations in newly diagnosed AML: A single-center retrospective study
- Author
-
Haitham Abdelhakim, Nicole Balmaceda, Joseph McGuirk, Eyad Z. Gharaibeh, Andrew K. Godwin, Mohammad Telfah, Ziyan Y. Pessetto, and Tara L. Lin
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Myeloid leukemia ,Retrospective cohort study ,Newly diagnosed ,business ,Single Center ,neoplasms - Abstract
e18525 Background: Several recurrent genetic mutations have been described in acute myeloid leukemia (AML), which have both prognostic and therapeutic implications. Recurrent mutations in the Neurofibromin 1 ( NF1) gene are reported in 1-5 % of AML patients; however, there are limited data regarding its prognostic implications. Here, we report the outcomes for patients with newly diagnosed AML with a somatic NF1 mutation at our center. Methods: A retrospective chart review included patients with newly diagnosed AML at KUMC from 01/2016 through 09/2018. All patients had targeted next-generation sequencing (NGS) at diagnosis. Baseline characteristics were compared between patients with NF1 mutations and those who were wild-type using Fisher’s exact test for categorical variables, and the Wilcoxon rank-sum test for continuous variables. The primary outcome was overall survival (OS), which was measured from the time of diagnosis to the time of death from any cause. A stepwise Cox proportional-hazard model was used to adjust for potential confounders. Results: Data on 110 patients were included. Out of the 110 patients, 15 (13.6%) had a delectable NF1 mutation, while 95 (86.4%) patients were NF1 wild-type. The baseline characteristics of the two groups are displayed below. Median OS for patients with an NF1 mutation was 7.3 months, while it was 18.4 months for patients with NF1 wild-type, Log-rank test p-value 0.02. After adjusting for potential confounders, including age, ELN risk category, induction regimen, presence of other mutations such as TP53, the hazard of death remained significantly higher for patients with NF1 mutations, HR 2.4, CI (1.05-5.6), p-value 0.04. Conclusions: In this single-center retrospective study, the presence of a NF1 mutation was associated with worse overall survival in patients with newly diagnosis AML. [Table: see text]
- Published
- 2019
- Full Text
- View/download PDF
26. Niraparib in metastatic pancreatic cancer after previous chemotherapy (NIRA-PANC): A phase 2 trial
- Author
-
Andrew K. Godwin, Anup Kasi, Carolyn Foster, Stephen K. Williamson, Ziyan Y. Pessetto, Timothy M. Schmitt, Raed Al-Rajabi, Anwaar Saeed, Malgorzata A. Witek, Joaquina Baranda, Sean C. Kumer, Weijing Sun, Steven A. Soper, and Prabhakar Chalise
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Metastatic pancreatic cancer ,medicine ,Adenocarcinoma ,business ,030215 immunology - Abstract
TPS4168 Background: Attempts to improve therapy for patients with pancreatic adenocarcinoma with traditional chemotherapy have largely failed to meaningfully improve survival. Therefore, there is a critical need for identification of specific molecular changes that define prognosis and potentially guide therapy decisions. Defective DNA damage response pathways in pancreatic cancer represent a targeted opportunity for treatment. PARP inhibitors exert activity in tumor cells that may not be effectively able to repair initially single-stranded and cumulatively double-stranded DNA breaks and can have a heightened susceptibility in tumor cells over normal tissue. This concept is referred to as synthetic lethality. Niraparib is an orally available, potent, highly selective PARP-1 and -2 inhibitor. We are studying the efficacy of Niraparib in pancreatic cancer patients that harbor DNA repair defects. Methods: This study is funded by a research grant from TESARO. Pre-screening of patients to find biomarker positive patients is funded by KU Cancer Center. This is a phase II open label single arm trial in metastatic pancreatic cancer patients with germline or somatic mutations, either already known, or tested after consent to pre-screening tumor tissue analysis in BRCA1/2, PALB2, ATM, NBN, ATR, BRIP1, IDH1/2, RAD51, RAD51B/C/D, RAD54L, CDK12, BARD1, FAM175A, BAP1, CHEK1/2, GEN1, MRE11A, XRCC2, SHFM1, FANCD2, FANCA, FANCC, FANCG, RPA1, ARID1A. Patients are being treated with Niraparib 300mg or 200mg by mouth daily for 28 days (1 cycle = 28 days) (200mg dose is for participants whose baseline weight is < 77 kg [169.756 lbs] or baseline platelet count is < 150,000 µL). The primary objective is to assess antitumor efficacy of niraparib using Objective Response Rate per RECIST 1.1. Secondary objectives include PFS, OS, DCR, DOR, and safety. Eligible patients received > 1 line of therapy, no prior PARP inhibitor(s), have measurable disease, and ECOG PS 0-1. Accrual target enrollment of 18 patients over a period of 24 months with a study duration of 30 months. Correlative studies include assessment of pharmacokinetics, circulating tumor cells and storing samples for future research. The trial is currently enrolling. Clinical trial information: NCT03553004.
- Published
- 2019
- Full Text
- View/download PDF
27. Drug Repurposing for Gastrointestinal Stromal Tumor
- Author
-
Scott Weir, Melinda Broward, Andrew K. Godwin, Geetika Sethi, and Ziyan Y. Pessetto
- Subjects
Drug ,Cancer Research ,Auranofin ,Gastrointestinal Stromal Tumors ,media_common.quotation_subject ,Cell Culture Techniques ,Apoptosis ,Disease ,Pharmacology ,Article ,Cell Line, Tumor ,medicine ,Humans ,Stromal tumor ,Cell Proliferation ,media_common ,GiST ,business.industry ,Drug Repositioning ,medicine.disease ,Drug repositioning ,Oncology ,Drug development ,Rheumatoid arthritis ,business ,Vidarabine ,medicine.drug - Abstract
Despite significant treatment advances over the past decade, metastatic gastrointestinal stromal tumor (GIST) remains largely incurable. Rare diseases, such as GIST, individually affect small groups of patients but collectively are estimated to affect 25 to 30 million people in the United States alone. Given the costs associated with the discovery, development, and registration of new drugs, orphan diseases such as GIST are often not pursued by mainstream pharmaceutical companies. As a result, “drug repurposing” or “repositioning,” has emerged as an alternative to the traditional drug development process. In this study, we screened 796 U.S. Food and Drug Administration (FDA)-approved drugs and found that two of these compounds, auranofin (Ridaura) and fludarabine phosphate, effectively and selectively inhibited the proliferation of GISTs, including imatinib-resistant cells. One of the most notable drug hits, auranofin, an oral, gold-containing agent approved by the FDA in 1985 for the treatment of rheumatoid arthritis, was found to inhibit thioredoxin reductase activity and induce reactive oxygen species (ROS) production, leading to dramatic inhibition of GIST cell growth and viability. Importantly, the anticancer activity associated with auranofin was independent of imatinib-resistant status, but was closely related to the endogenous and inducible levels of ROS. Coupled with the fact that auranofin has an established safety profile in patients, these findings suggest for the first time that auranofin may have clinical benefit for patients with GIST, particularly in those suffering from imatinib-resistant and recurrent forms of this disease. Mol Cancer Ther; 12(7); 1299–309. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
28. In silico and in vitro drug screening identifies new therapeutic approaches for Ewing sarcoma
- Author
-
Hani J. Alturkmani, Bin Chen, Atul J. Butte, Ziyan Y. Pessetto, Scott Weir, Howard G. Rosenthal, Michael Baltezor, Andrew K. Godwin, Yan Ma, Stephen Hyter, Colleen A. Flynn, and Kathleen A. Neville
- Subjects
0301 basic medicine ,Gerontology ,Oncology ,Oncogene Proteins, Fusion ,Ganetespib ,Drug Screening Assays ,Hsp90 inhibitor ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,media_common ,Cancer ,Oncogene Proteins ,Pediatric ,Tumor ,drug repurposing ,auranofin ,Sarcoma ,3. Good health ,Gene Expression Regulation, Neoplastic ,Drug repositioning ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Development of treatments and therapeutic interventions ,medicine.drug ,Research Paper ,Biotechnology ,Drug ,medicine.medical_specialty ,Pediatric Research Initiative ,Auranofin ,Cell Survival ,In silico ,media_common.quotation_subject ,ganetespib ,Oncology and Carcinogenesis ,Sarcoma, Ewing ,In Vitro Techniques ,high-throughput screening ,Cell Line ,03 medical and health sciences ,Rare Diseases ,Pharmacokinetics ,In vivo ,Internal medicine ,Cell Line, Tumor ,Ewing ,medicine ,Humans ,Computer Simulation ,Fusion ,Cell Proliferation ,Neoplastic ,business.industry ,Proto-Oncogene Protein c-fli-1 ,Antitumor ,Triazoles ,030104 developmental biology ,Orphan Drug ,Gene Expression Regulation ,Drug Screening Assays, Antitumor ,RNA-Binding Protein EWS ,business ,Ewing sarcoma ,Transcription Factors - Abstract
The long-term overall survival of Ewing sarcoma (EWS) patients remains poor; less than 30% of patients with metastatic or recurrent disease survive despite aggressive combinations of chemotherapy, radiation and surgery. To identify new therapeutic options, we employed a multi-pronged approach using in silico predictions of drug activity via an integrated bioinformatics approach in parallel with an in vitro screen of FDA-approved drugs. Twenty-seven drugs and forty-six drugs were identified, respectively, to have anti-proliferative effects for EWS, including several classes of drugs in both screening approaches. Among these drugs, 30 were extensively validated as mono-therapeutic agents and 9 in 14 various combinations in vitro. Two drugs, auranofin, a thioredoxin reductase inhibitor, and ganetespib, an HSP90 inhibitor, were predicted to have anti-cancer activities in silico and were confirmed active across a panel of genetically diverse EWS cells. When given in combination, the survival rate in vivo was superior compared to auranofin or ganetespib alone. Importantly, extensive formulations, dose tolerance, and pharmacokinetics studies demonstrated that auranofin requires alternative delivery routes to achieve therapeutically effective levels of the gold compound. These combined screening approaches provide a rapid means to identify new treatment options for patients with a rare and often-fatal disease.
- Published
- 2017
29. Clinical and biomarker results from phase I/II study of PI3K inhibitor BYL 719 (alpelisib) plus nab-paclitaxel in HER2-negative metastatic breast cancer
- Author
-
Stephen K. Williamson, Stephanie LaFaver, Jecinta Scott, Marc Hoffmann, Qamar J. Khan, Bruce F. Kimler, Anne O'Dea, Jilliann A De Jong, Vandana G. Abramson, Julia Urban, Andrew K. Godwin, Sharon Lewis, Jaimie Heldstab, Greg Reed, Priyanka Sharma, Ziyan Y. Pessetto, Harsh B. Pathak, Manana Elia, Yen Y. Wang, and Karissa Finke
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,HER2 negative ,medicine.disease ,Metastatic breast cancer ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Phase i ii ,Oncology ,030220 oncology & carcinogenesis ,Concomitant ,Cancer research ,Medicine ,Biomarker (medicine) ,business ,PI3K/AKT/mTOR pathway ,Nab-paclitaxel - Abstract
1018Background: Activation of Phosphatidylinositol-3-kinase (PI3K) pathway may confer resistance to taxanes and in preclinical models concomitant inhibition of the PI3K pathway enhances efficacy of...
- Published
- 2018
- Full Text
- View/download PDF
30. ALK alteration association with prognosis in metastatic colorectal cancer: A single institution retrospective cohort study
- Author
-
Nikhil Gupta, Anusha Chidharla, Sarah Schmitt, Ravi Kumar Paluri, Anup Kasi, Andrew K. Godwin, Anwaar Saeed, and Ziyan Y. Pessetto
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Anaplastic Lymphoma ,business.industry ,Colorectal cancer ,Incidence (epidemiology) ,Retrospective cohort study ,medicine.disease ,hemic and lymphatic diseases ,Internal medicine ,Medicine ,Single institution ,business - Abstract
676 Background: The Anaplastic Lymphoma Kinas (ALK) gene is altered in various malignancies which can be targeted with several inhibitors. However, the incidence of mutations in ALK are not well defined in colorectal cancers (CRC). We aim to study the prevalence of ALK mutations in metastatic CRC (mCRC) patients and correlate with clinical outcomes. Methods: We retrospectively evaluated 54 mCRC patients treated with chemotherapy at Kansas University Cancer Center between June 2010 and September 2017 who underwent our inhouse targeted 17 gene next-generation sequencing test using an illumina MiSeq technology. The panel included AKT1, ALK, BRAF, EGFR, FLT3, HRAS, IDH1, IDH2, JAK2, KIT, KRAS, NPM1, NRAS, PDGFRA, PIK3CA, PTEN, and TP53 genes. We compared baseline characteristics, tumor location (left vs right) and clinical outcomes between patients with ALK mutations versus ALK wild type. Results: We retrospectively evaluated 54 mCRC patients treated with chemotherapy at Kansas University Cancer Center between June 2010 and September 2017 who underwent our inhouse targeted 17 gene next-generation sequencing test using an illumina MiSeq technology. We compared baseline characteristics, tumor location (left vs right) and clinical outcomes between patients with ALK mutations versus ALK wild type. Results: We found 10 pts (18.5%) with ALK mutations and compared with 44 patients that were ALK wild type. Demographic and baseline characteristics (ALK mutation/wild type) were as follows: Median age 61 vs 61 years, Gender (male): 80% vs 50%, Left sided tumor 70% vs 60%. Of 41 evaluable patients (ALK mutation vs wild type), Objective response rate (stable + partial response) was 50% vs 91% [p = 0.03], 6-month PFS 33% vs 77%, Median PFS 12 months vs 17.7 months [p = 0.2], Median OS was 14.5 months vs not reached [HR 28.98, p = 0.025]. Conclusions: Patients harboring ALK mutations had a statistically significant poorer objective response rate and OS compared to ALK wild type patients. Findings of our study highlight the importance of assessing ALK gene alteration as a prognostic biomarker for mCRC. Therefore ALK mutation appears to be poor prognostic factor and needs to be validated in larger cohorts.
- Published
- 2018
- Full Text
- View/download PDF
31. Abstract 1764: Accurate and reproducible detection of fusions and exon skipping events in NSCLC-derived samples using a comprehensive, targeted RNA-Seq system across multiple laboratories
- Author
-
T Halsey, Ziyan Y. Pessetto, Victor J. Weigman, Gary J. Latham, Dan Su, Andrew K. Godwin, Patrick Hurban, Richard A. Blidner, Shobha Gokul, Maria Aguirre, Brian C. Haynes, Stephen Hyter, Léon C van Kempen, and Maria Curtis
- Subjects
Genetics ,Cancer Research ,Oncology ,RNA-Seq ,Biology ,Exon skipping - Abstract
Introduction: Reliable assessment of cancer-associated RNA markers in lung cancer produced by gene-fusions or exon skipping events by next-generation sequencing requires integrated reagents, protocols, and interpretive software that can harmonize procedures and ensure consistent results across laboratories. We evaluated a comprehensive system for targeted RNA-Seq that includes reagents for nucleic acid quantification, library prep, run controls, and companion bioinformatics software. The reproducibility of this system was evaluated in a multi-phase study design at 5 independent laboratories. Methods: Total nucleic acid (TNA) was isolated from formalin-fixed, paraffin-embedded (FFPE) residual non-small cell lung cancer (NSCLC) tumor biopsies and cell-lines (RT-112, H596, HCC78). These TNA isolates were used to prepare a set of 30 test samples, including a dilution series to assess assay sensitivity. A non-template control and kit positive control were also included. The sample set was evaluated using the QuantideX® NGS RNA Lung Cancer Kit RUO (Asuragen) and sequenced on the MiSeq® system (Illumina) at Asuragen and 4 independent laboratories. Analyses were conducted using QuantideX® NGS Reporter RUO (Asuragen), a software suite that includes a FASTQ processing pipeline and incorporates pre-analytical QC information into the fusion-caller algorithm and reporting tool. Results: Laboratories were trained on the assay workflow and companion bioinformatics software in less than two days followed by independent library preparation and sequencing workflows. A total of 266 sample libraries were evaluated from inputs down to Conclusions: The accuracy of this novel targeted NGS assay for RNA fusions and splice variants in NSCLC was demonstrated in a multi-site laboratory evaluation using clinically-relevant specimens and low inputs of TNA. The ability of the panel to detect both common and rare gene fusion transcripts and exon skipping events within an integrated wet- and dry-bench workflow provides a foundation for the reliable detection of oncogenic RNA fusions and aberrant splicing events that can respond to current and emerging targeted therapies. This study highlighted the ease of implementation and consistent performance that can be achieved in different laboratories when the process from sample-to-report is highly integrated. Citation Format: Gary J. Latham, Richard Blidner, Brian C. Haynes, Shobha Gokul, Maria L. Aguirre, Stephen Hyter, Ziyan Y. Pessetto, Maria Curtis, Dan Su, Tom Halsey, Victor Weigman, Patrick Hurban, Andrew K. Godwin, Leon C. van Kempen. Accurate and reproducible detection of fusions and exon skipping events in NSCLC-derived samples using a comprehensive, targeted RNA-Seq system across multiple laboratories [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 1764. doi:10.1158/1538-7445.AM2017-1764
- Published
- 2017
- Full Text
- View/download PDF
32. Drug repurposing identifies a synergistic combination therapy with imatinib mesylate for gastrointestinal stromal tumor
- Author
-
Ziyan Y. Pessetto, Andrew K. Godwin, Scott Weir, Yan Ma, Margaret von Mehren, and Jeff Hirst
- Subjects
Cancer Research ,Gastrointestinal Stromal Tumors ,Mice, Nude ,Apoptosis ,Pharmacology ,Piperazines ,Article ,Mice ,Pharmacotherapy ,In vivo ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Animals ,Humans ,Stromal tumor ,neoplasms ,Cell Proliferation ,GiST ,business.industry ,Drug Repositioning ,Cancer ,Imatinib ,Drug Synergism ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,High-Throughput Screening Assays ,Drug repositioning ,Imatinib mesylate ,Pyrimidines ,Oncology ,Benzamides ,Cancer research ,Imatinib Mesylate ,Female ,business ,Vidarabine Phosphate ,medicine.drug - Abstract
Gastrointestinal stromal tumor (GIST) is a rare and therefore often neglected disease. Introduction of the kinase inhibitor imatinib mesylate radically improved the clinical response of patients with GIST; however, its effects are often short-lived, with GISTs demonstrating a median time-to-progression of approximately two years. Although many investigational drugs, approved first for other cancers, have been subsequently evaluated for the management of GIST, few have greatly affected the overall survival of patients with advanced disease. We employed a novel, focused, drug-repurposing effort for GIST, including imatinib mesylate–resistant GIST, evaluating a large library of FDA-approved drugs regardless of current indication. As a result of the drug-repurposing screen, we identified eight FDA-approved drugs, including fludarabine phosphate (F-AMP), that showed synergy with and/or overcame resistance to imatinib mesylate. F-AMP induces DNA damage, Annexin V, and caspase-3/7 activities as the cytotoxic effects on GIST cells, including imatinib mesylate–resistant GIST cells. F-AMP and imatinib mesylate combination treatment showed greater inhibition of GIST cell proliferation when compared with imatinib mesylate and F-AMP alone. Successful in vivo experiments confirmed the combination of imatinib mesylate with F-AMP enhanced the antitumor effects compared with imatinib mesylate alone. Our results identified F-AMP as a promising, repurposed drug therapy for the treatment of GISTs, with potential to be administered in combination with imatinib mesylate or for treatment of imatinib mesylate–refractory tumors. Mol Cancer Ther; 13(10); 2276–87. ©2014 AACR.
- Published
- 2014
33. Epimorphin-Induced MET Sensitizes Ovarian Cancer Cells to Platinum
- Author
-
Kok-Hooi Yew, Ziyan Y. Pessetto, Jennifer Crow, Jeff Hirst, and Andrew K. Godwin
- Subjects
Multidisciplinary ,business.industry ,Science ,lcsh:R ,lcsh:Medicine ,Correction ,Bioinformatics ,Cancer research ,Ovarian cancer cells ,Correct name ,Medicine ,lcsh:Q ,business ,lcsh:Science - Abstract
The name of the fourth author is incorrect. The correct name is: Ziyan Pessetto. The correct citation is: Yew K-H, Crow J, Hirst J, Pessetto Z, Godwin AK (2013) Epimorphin-Induced MET Sensitizes Ovarian Cancer Cells to Platinum. PLoS ONE 8(9): e72637. doi:10.1371/journal.pone.0072637.
- Published
- 2013
34. Casitas B-Lineage Lymphoma RING Domain Inhibitors Protect Mice against High-Fat Diet-Induced Obesity and Insulin Resistance
- Author
-
Min Wu, Xingliang Xie, Qing Su, Wang Zan, Ling Zhong, Xiurong Gao, Zhihe Zang, Yan Zhao, Lin Sun, Yiyi Sun, and Ziyan Y. Pessetto
- Subjects
Blood Glucose ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Gene Expression ,lcsh:Medicine ,Type 2 diabetes ,Diet, High-Fat ,Protein Structure, Secondary ,Receptor tyrosine kinase ,Energy homeostasis ,Diabetes Mellitus, Experimental ,Mice ,Insulin resistance ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Hypoglycemic Agents ,Insulin ,Obesity ,Proto-Oncogene Proteins c-cbl ,lcsh:Science ,Multidisciplinary ,biology ,lcsh:R ,Signal transducing adaptor protein ,medicine.disease ,Recombinant Proteins ,Protein Structure, Tertiary ,3. Good health ,Ubiquitin ligase ,Mice, Inbred C57BL ,Endocrinology ,biology.protein ,Cancer research ,lcsh:Q ,Anti-Obesity Agents ,Insulin Resistance ,Energy Metabolism ,Peptides ,Injections, Intraperitoneal ,Research Article - Abstract
The casitas b-lineage lymphoma (c-Cbl) is an important adaptor protein with an intrinsic E3 ubiquitin ligase activity that interacts with E2 proteins such as UbCH7. c-Cbl plays a vital role in regulating receptor tyrosine kinase signaling. c-Cbl involves in whole-body energy homeostasis, which makes it a potential target for the treatment of type 2 diabetes and obesity. In the present study, we have designed two parental peptides and 55 modified peptides based on the structure of UbCH7 loop L1 and L2. Thirteen of the modified peptides showed increased inhibitory activity in a fluorescence polarization-based assay. In the in vivo proof of study principle, mice treated with peptides 10, 34, 49 and 51 were protected against high-fat diet-induced obesity and insulin resistant. These inhibitors may potentially lead to new therapeutic alternatives for obesity and type 2 diabetes.
- Published
- 2015
- Full Text
- View/download PDF
35. Development of a Fluorescence Polarization Based High-Throughput Assay to Identify Casitas B-Lineage Lymphoma RING Domain Regulators
- Author
-
Yiyi Sun, Yan Zhao, Wang Zan, Xiurong Gao, Min Wu, Qing Su, Ziyan Y. Pessetto, Xingliang Xie, Lin Sun, Zhihe Zang, and Ling Zhong
- Subjects
Multidisciplinary ,Lymphoma ,biology ,lcsh:R ,B-cell receptor ,Proto-Oncogene Proteins c-cbl ,lcsh:Medicine ,Fluorescence Polarization ,Plasma protein binding ,Ubiquitin-conjugating enzyme ,Small molecule ,Ubiquitin ligase ,Biochemistry ,Ubiquitin-Conjugating Enzymes ,biology.protein ,lcsh:Q ,Small molecule binding ,Signal transduction ,lcsh:Science ,Research Article ,Protein Binding - Abstract
The E3 ubiquitin protein ligase Casitas B-lineage Lymphoma (Cbl) proteins and their binding partners play an important role in regulating signal transduction pathways. It is important to utilize regulators to study the protein-protein interactions (PPIs) between these proteins. However, finding specific small-molecule regulators of PPIs remains a significant challenge due to the fact that the interfaces involved in PPIs are not well suited for effective small molecule binding. We report the development of a competitive, homogeneous, high-throughput fluorescence polarization (FP) assay to identify small molecule regulators of Cbl (RING) domain. The FP assay was used to measure binding affinities and inhibition constants of UbCH7 peptides and small molecule regulators of Cbl (RING) domains, respectively. In order to rule out promiscuous, aggregation-based inhibition, two assay conditions were developed and compared side by side. Under optimized conditions, we screened a 10,000 natural compound library in detergent-free and detergent-present (0.01% Triton X-100) systems. The results indicate that the detergent-present system is more suitable for high-throughput screens. Three potential compounds, methylprotodioscin, leonuride and catalpol, have been identified that bind to Cbl (RING) domain and interfere with the Cbl (RING)-UbCH7 protein-protein interaction.
- Published
- 2013
- Full Text
- View/download PDF
36. Irinotecan (Iri) and buparlisib (B) in previously treated patients (pts) with metastatic colorectal cancer (mCRC)
- Author
-
Stephen K. Williamson, Raymond P. Perez, Rashna Madan, Ziyan Y. Pessetto, Maxine Stoltz, Andrew K. Godwin, Greg Reed, Chris Mackay, and Joaquina Baranda
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,Buparlisib ,Cancer ,Pharmacology ,medicine.disease ,Clinical correlation ,Irinotecan ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Toxicity ,Cohort ,medicine ,business ,Previously treated ,medicine.drug - Abstract
655 Background: The PIK3CA pathway has oncogenic role in mCRC. Buparlisib is an oral pan-class PIK3CA inhibitor. It is currently being studied in combination trials for various malignancies. The primary objective of this phase I trial was to identify the maximum tolerated dose (MTD) for Iri plus B in pts with previously treated mCRC, with or without previous Iri therapy. We also performed PK analysis of each drug alone and in combination, determined clinical response to the combination, and evaluated achieval FFPE samples for somatic mutations in a panel of cancer associated genes, including PIK3CA for clinical correlation. Methods: A 3+3 dose titration method was used. Iri was administered intravenously every 14 days (one cycle) and B orally daily. Pts received the first dose of Iri on Cycle1 Day1. B was started 24 hours after the first dose of Iri. Safety and toxicity assessments were performed every cycle. Results: Twenty patients were enrolled: 4 in cohort 0 (Iri 120 mg/m2 + B 50 mg/d), 11 in cohort 1 (Iri 150 mg/m2 + B 50 mg/d), 5 in cohort 2 (Iri 150 mg/m2 + B 80 mg/d). The most common Grades 3/4 adverse events were neutropenia and abdominal pain. The MTD was Iri 150 mg/m2 + B 50 mg/d. The DLTs include male genital mucositis, grade 3 diarrhea, and asymptomatic grade 3 hyponatremia. In cohort 1, one pt experienced grade 2 delirium and a pt in cohort 2 had grade 3 psychosis. Of the 4 pts who received 4 cycles of therapy 2 had stable and 2 had progressive disease. No objective responses were seen. There is no significant change in the PK of Iri between Cycles 1 and 2. B at 50 mg had no consistent effect on the disposition of Iri given at 120 mg/m2 and 150 mg/m2. The Cmax and AUC for B show clear dose proportionality. Using targeted re-sequencing, a 48-gene panel that included AKT1, BRAF, KRAS, and PTEN was performed. The complete analysis of the correlative study will be presented in the meeting. Conclusions: This first human trial established that Buparlisib (50 mg qd) and Iri (150 mg/m2 q14d) are tolerable in combination. However, it is too early to determine the activity of this combination in the treatment of mCRC. Clinical trial information: NCT01304602.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.