6 results on '"Ryan W. Feathers"'
Search Results
2. Integration of Comprehensive Genomic Analysis and Functional Screening of Affected Molecular Pathways to Inform Cancer Therapy
- Author
-
Stephen J. Murphy, Minetta C. Liu, Faye R. Harris, Sarah H. Johnson, George Vasmatzis, Farhad Kosari, Ryan W. Feathers, Mitesh J. Borad, James B. Smadbeck, Sowjanya Reganti, Janet L. Schaefer Klein, Lin Yang, E. Aubrey Thompson, John Cheville, and Panos Z. Anastasiadis
- Subjects
Afatinib ,DNA Mutational Analysis ,Breast Neoplasms ,Article ,CDH1 ,Metastasis ,Breast cancer ,Germline mutation ,medicine ,Humans ,Neoplasm Metastasis ,Precision Medicine ,Loss function ,medicine.diagnostic_test ,biology ,business.industry ,General Medicine ,Genomics ,Middle Aged ,medicine.disease ,Precision medicine ,Carcinoma, Lobular ,biology.protein ,Cancer research ,Female ,business ,Algorithms ,medicine.drug ,Fluorescence in situ hybridization ,Genes, Neoplasm - Abstract
Objective To select optimal therapies based on the detection of actionable genomic alterations in tumor samples is a major challenge in precision medicine. Methods We describe an effective process (opened December 1, 2017) that combines comprehensive genomic and transcriptomic tumor profiling, custom algorithms and visualization software for data integration, and preclinical 3-dimensiona ex vivo models for drug screening to assess response to therapeutic agents targeting specific genomic alterations. The process was applied to a patient with widely metastatic, weakly hormone receptor positive, HER2 nonamplified, infiltrating lobular breast cancer refractory to standard therapy. Results Clinical testing of liver metastasis identified BRIP1, NF1, CDH1, RB1, and TP53 mutations pointing to potential therapies including PARP, MEK/RAF, and CDK inhibitors. The comprehensive genomic analysis identified 395 mutations and several structural rearrangements that resulted in loss of function of 36 genes. Meta-analysis revealed biallelic inactivation of TP53, CDH1, FOXA1, and NIN, whereas only one allele of NF1 and BRIP1 was mutated. A novel ERBB2 somatic mutation of undetermined significance (P702L), high expression of both mutated and wild-type ERBB2 transcripts, high expression of ERBB3, and a LITAF-BCAR4 fusion resulting in BCAR4 overexpression pointed toward ERBB-related therapies. Ex vivo analysis validated the ERBB-related therapies and invalidated therapies targeting mutations in BRIP1 and NF1. Systemic patient therapy with afatinib, a HER1/HER2/HER4 small molecule inhibitor, resulted in a near complete radiographic response by 3 months. Conclusion Unlike clinical testing, the combination of tumor profiling, data integration, and functional validation accurately assessed driver alterations and predicted effective treatment.
- Published
- 2019
3. Three-dimensional microscale hanging drop arrays with geometric control for drug screening and live tissue imaging
- Author
-
Ariana Mostafa, Alvaro G. Hernandez, C. L. Wright, Jenny Drnevich, Anurup Ganguli, Jacob Berger, Vahid Faramarzi, Andrew M. Smith, Rashid Bashir, Phuong Le, George Vasmatzis, Olaoluwa O. Adeniba, Panos Z. Anastasiadis, Yongdeok Kim, Wan-Hsin Lin, Ryan W. Feathers, C. Saavedra, Karla P. Ramos-Cruz, Farhad Kosari, and G. J. Pagan Diaz
- Subjects
Materials science ,Cell Culture Techniques ,Drug Evaluation, Preclinical ,Geometric shape ,GeneralLiterature_MISCELLANEOUS ,law.invention ,Mice ,03 medical and health sciences ,Engineering ,0302 clinical medicine ,Tissue engineering ,Hardware_GENERAL ,Confocal microscopy ,law ,Spheroids, Cellular ,Microscopy ,Animals ,Humans ,Health and Medicine ,Hardware_REGISTER-TRANSFER-LEVELIMPLEMENTATION ,Research Articles ,Microscale chemistry ,Cancer ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Tissue Engineering ,business.industry ,Drug discovery ,Spheroid ,SciAdv r-articles ,High-Throughput Screening Assays ,030220 oncology & carcinogenesis ,Personalized medicine ,business ,Research Article ,Biomedical engineering - Abstract
Tiny silicon microchip–based high-throughput hanging drop arrays allow drug screening and geometric control., Existing three-dimensional (3D) culture techniques are limited by trade-offs between throughput, capacity for high-resolution imaging in living state, and geometric control. Here, we introduce a modular microscale hanging drop culture where simple design elements allow high replicates for drug screening, direct on-chip real-time or high-resolution confocal microscopy, and geometric control in 3D. Thousands of spheroids can be formed on our microchip in a single step and without any selective pressure from specific matrices. Microchip cultures from human LN229 glioblastoma and patient-derived mouse xenograft cells retained genomic alterations of originating tumors based on mate pair sequencing. We measured response to drugs over time with real-time microscopy on-chip. Last, by engineering droplets to form predetermined geometric shapes, we were able to manipulate the geometry of cultured cell masses. These outcomes can enable broad applications in advancing personalized medicine for cancer and drug discovery, tissue engineering, and stem cell research.
- Published
- 2021
- Full Text
- View/download PDF
4. PLEKHA7, an apical adherens junction protein, suppresses inflammatory breast cancer in the context of high E-cadherin and p120-catenin expression
- Author
-
Aziza Nassar, Idris Tolgay Ocal, Sotiris Sotiriou, Paul A. Decker, Sejal Subhash Shah, Antonis Kourtidis, Panos Z. Anastasiadis, Mary T. Haddad, Ryan W. Feathers, and Lindy J. Pence
- Subjects
Delta Catenin ,Mice, SCID ,adherens junction ,Polyethylene Glycols ,lcsh:Chemistry ,Mice ,PLEKHA7 ,lcsh:QH301-705.5 ,Spectroscopy ,Antibiotics, Antineoplastic ,Catenins ,Adherens Junctions ,General Medicine ,Cadherins ,Tumor Burden ,Computer Science Applications ,Gene Expression Regulation, Neoplastic ,Lymphatic Metastasis ,Immunohistochemistry ,Female ,Inflammatory Breast Neoplasms ,Signal Transduction ,Context (language use) ,Inflammatory breast cancer ,Article ,Catalysis ,Inorganic Chemistry ,Adherens junction ,Breast cancer ,Antigens, CD ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,Cell Proliferation ,cadherin–catenin complexes ,Cadherin ,business.industry ,Organic Chemistry ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,lcsh:Biology (General) ,lcsh:QD1-999 ,Doxorubicin ,Cancer research ,Caco-2 Cells ,Carrier Proteins ,business - Abstract
Background: Inflammatory breast cancer is a highly aggressive form of breast cancer that robustly forms clusters of tumor emboli in dermal lymphatics and readily metastasizes. Inflammatory breast cancers express high levels of E-cadherin, the major protein of adherens junctions, which may enhance the ability of tumor cells to form such clusters and contribute to metastasis. Seemingly contradictory, E-cadherin has both tumor-suppressing and tumor-promoting roles in cancer; previous studies suggest that this depends on the balance between apical and basolateral cadherin-catenin complexes. Methods: In the present study, we use immunohistochemistry of inflammatory breast cancer patient samples and biochemical analysis of cell lines to determine the expression of PLEKHA7, an apical adherens junction protein. We use viral transduction to ectopically express PLEKHA7 in the SUM149 inflammatory breast cancer cell line. The effect of PLEKHA7 on the aggressiveness of inflammatory breast cancer in 2D, 3D and in-vivo were examined. Results: We determined that PLEKHA7 was deregulated in inflammatory breast cancer, demonstrating improper localization or lost expression in a strong majority of patient samples and very low expression in cell line models. We found that re-expressing PLEKHA7 is sufficient to suppress proliferation, anchorage independent growth, spheroid viability, and tumor growth in-vivo. We also observed a negative-selection pressure within the xenograft tumors to lose PLEKHA7 function or expression.Conclusions: The data indicate that PLEKHA7 is frequently deregulated and acts as a suppressor of inflammatory breast cancer. They also suggest that the resulting imbalance between apical and basolateral cadherin-catenin complexes contributes to growth, survival and emboli-forming capacities of inflammatory breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
5. Combination targeted treatment may enhance antitumor activity in ERBB3 amplified high-grade serous endometrial cancer cells resistant to single agent targeted therapy
- Author
-
Andrea Mariani, Janet L. Schaefer Klein, Jamie Lynch, Aaron S. Mansfield, S. John Weroha, Leila A. Jones, Michael T. Barrett, James B. Smadbeck, Alyssa Larish, Panos Z. Anastasiadis, George Vasmatzis, Mitesh J. Borad, Stephen J. Murphy, Faye R. Harris, Dorsay Sadeghian, Wan-Hsin Lin, Maureen A. Lemens, Angela Emanuel, and Ryan W. Feathers
- Subjects
Cobimetinib ,business.industry ,medicine.medical_treatment ,Cmax ,Obstetrics and Gynecology ,medicine.disease ,Primary tumor ,Targeted therapy ,chemistry.chemical_compound ,Serous fluid ,Oncology ,chemistry ,ErbB ,Cancer research ,Medicine ,Viability assay ,business ,PI3K/AKT/mTOR pathway - Abstract
Objectives: Up-regulation of ERBB pathways represent targetable alterations in many high grade endometrial cancers (EC); ERBB3 amplifications in particular may contribute to ineffective pathway targeting due to persistent co-activation of other ERBB binding partners, leading to tumor growth and survival. The efficacy of downstream dual-inhibition of MEK+AKT or MEK+PI3K in an ERBB3 amplified EC primary tumor was studied using a microcancer 3D ex-vivo tumor cell viability assay. Methods: Tumor was prospectively collected from a patient with stage II, FIGO grade 3, serous EC and genomic characterization was performed using multiple sequencing methods: whole exome, RNA, and MatePair analysis. Somatic mutations, structural variance, with transcriptomic profiling was used to identify potential driver pathways for subsequent pharmacologic inhibition. Primary tumor cells were grown in a three-dimensional environment and the formed microcancers were subjected to drug treatment. Cell viability was determined by the CellTiter-Glow Luminescent Assay. Data transformation and dose-response curves were generated using GraphPad PRISM using four parameter logistic regression. CompuSyn software with the Chou-Talalay method was used to analyze drug interactions and synergy. The Fractional response-Combination index (Fa-CI) plot was generated with Microsoft Excel. Capivasertib, GDC-0084, and cobimetinib were used to inhibit AKT, PI3K/mTOR, and MEK, respectively. Inhibitory effect was defined as percent reduction in ATP at clinically-defined predicted plasma maximum concentration (Cmax) values. Results: Genomic sequencing revealed overexpression of a mutated ERBB3 (c.889G>T; p.Asp297Tyr) transcript within a 6N focal amplification. Additional pertinent alterations included a MYC mid-level gain, and TP53 double hit (loss and exonic splicing silencer (ESS)). Inhibition of cell viability was moderate by single agents: capivasertib, cobimetanib, or GDC-0084, as shown by inhibitory effect values of 33.9%, 35.9% and 49.9%, respectively. Two combinations with cobimetinib demonstrated increasing inhibitory effect values of 76.2% for cobimetanib/capivasertib at the Cmax of capivasertib and 79.7% for cobimetanib/GDC-0084 at the Cmax of cobimetanib. Synergy was evidenced by a combination index 0.8 (Figure 1). Download : Download high-res image (185KB) Download : Download full-size image Conclusions: Combined inhibition of MEK and PI3K-AKT-mTOR pathways synergistically suppress viability in a patient-derived high-grade serous EC harboring a ERBB3 amplification. Further ex-vivo testing should be performed in endometrial tumors with other ERBB3 aberrations to allow generalization of results.
- Published
- 2021
- Full Text
- View/download PDF
6. A phase 1 and randomized, placebo-controlled phase 2 trial of bevacizumab plus dasatinib in patients with recurrent glioblastoma: Alliance/North Central Cancer Treatment Group N0872
- Author
-
Caterina Giannini, Xiomara W. Carrero, Jesse G. Dixon, Erin Twohy, S. Keith Anderson, Daniel M. Anderson, Evanthia Galanis, David Schiff, Ryan W. Feathers, Timothy J. Kaufmann, David Tran, Panos Z. Anastasiadis, Jan C. Buckner, Suriya A. Jeyapalan, Galanis E., Anderson S.K., Twohy E.L., Carrero X.W., Dixon J.G., Tran D.D., Jeyapalan S.A., Anderson D.M., Kaufmann T.J., Feathers R.W., Giannini C., Buckner J.C., Anastasiadis P.Z., and Schiff D.
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Kaplan-Meier Estimate ,bevacizumab ,Placebo ,Drug Administration Schedule ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Glioma ,Internal medicine ,Lymphopenia ,Antineoplastic Combined Chemotherapy Protocols ,Outcome Assessment, Health Care ,Clinical endpoint ,Medicine ,Humans ,dasatinib ,030212 general & internal medicine ,Adverse effect ,Fatigue ,Aged ,business.industry ,Brain Neoplasms ,Cancer ,phase 2 trial ,Common Terminology Criteria for Adverse Events ,Middle Aged ,medicine.disease ,Dasatinib ,030220 oncology & carcinogenesis ,Src family kinase inhibitors ,Female ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,medicine.drug ,recurrent glioblastoma - Abstract
Background: Src signaling is markedly upregulated in patients with invasive glioblastoma (GBM) after the administration of bevacizumab. The Src family kinase inhibitor dasatinib has been found to effectively block bevacizumab-induced glioma invasion in preclinical models, which led to the hypothesis that combining bevacizumab with dasatinib could increase bevacizumab efficacy in patients with recurrent GBM. Methods: After the completion of the phase 1 component, the phase 2 trial (ClinicalTrials.gov identifier NCT00892177) randomized patients with recurrent GBM 2:1 to receive 100mg of oral dasatinib twice daily (arm A) or placebo (arm B) on days 1 to 14 of each 14-day cycle combined with 10mg/kg of intravenous bevacizumab on day 1 of each 14-day cycle. The primary endpoint was 6-month progression-free survival (PFS6). Results: In the 121 evaluable patients, the PFS6 rate was numerically, but not statistically, higher in arm A versus arm B (28.9% [95% CI, 19.5%-40.0%] vs 18.4% [95% CI, 7.7%-34.4%]; P=.22). Similarly, there was no significant difference in the median overall survival noted between the treatment arms (7.3months and 7.7months, respectively; P=.93). The objective response rate was 15.7% in arm A and 26.3% in arm B (P=.52), but with a significantly longer duration in patients treated on arm A (16.3months vs 2months). The incidence of grade ≥3 toxicity was comparable between treatment arms, with hematologic toxicities occurring more frequently in arm A versus arm B (15.7% vs 7.9%) (adverse events were assessed as per the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). Correlative tissue analysis demonstrated an association between pSRC/LYN signaling in patient tumors and outcome. Conclusions: Despite upregulation of Src signaling in patients with GBM, the combination of bevacizumab with dasatinib did not appear to significantly improve the outcomes of patients with recurrent GBM compared with bevacizumab alone.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.