Tony Navas
Astellas Pharma
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Tony Navas.
PLOS ONE | 2018
Tony Navas; Thomas D. Pfister; Simona Colantonio; Amina Aziz; Lynda Dieckman; Richard G. Saul; Jan A. Kaczmarczyk; Suzanne Borgel; Sergio Y. Alcoser; Melinda G. Hollingshead; Young Ho Lee; Donald P. Bottaro; Tara Hiltke; Gordon Whiteley; Naoko Takebe; Robert J. Kinders; Ralph E. Parchment; Joseph E. Tomaszewski; James H. Doroshow
The presence of cancer stem cells (CSCs) and the induction of epithelial-to-mesenchymal transition (EMT) in tumors are associated with tumor aggressiveness, metastasis, drug resistance, and poor prognosis, necessitating the development of reagents for unambiguous detection of CSC- and EMT-associated proteins in tumor specimens. To this end, we generated novel antibodies to EMT- and CSC-associated proteins, including Goosecoid, Sox9, Slug, Snail, and CD133. Importantly, unlike several widely used antibodies to CD133, the anti-CD133 antibodies we generated recognize epitopes distal to known glycosylation sites, enabling analyses that are not confounded by differences in CD133 glycosylation. For all target proteins, we selected antibodies that yielded the expected target protein molecular weights by Western analysis and the correct subcellular localization patterns by immunofluorescence microscopy assay (IFA); binding selectivity was verified by immunoprecipitation−mass spectrometry and by immunohistochemistry and IFA peptide blocking experiments. Finally, we applied these reagents to assess modulation of the respective markers of EMT and CSCs in xenograft tumor models by IFA. We observed that the constitutive presence of human hepatocyte growth factor (hHGF) in the tumor microenvironment of H596 non-small cell lung cancer tumors implanted in homozygous hHGF knock-in transgenic mice induced a more mesenchymal-like tumor state (relative to the epithelial-like state when implanted in control SCID mice), as evidenced by the elevated expression of EMT-associated transcription factors detected by our novel antibodies. Similarly, our new anti-CD133 antibody enabled detection and quantitation of drug-induced reductions in CD133-positive tumor cells following treatment of SUM149PT triple-negative breast cancer xenograft models with the CSC/focal adhesion kinase (FAK) inhibitor VS-6063. Thus, our novel antibodies to CSC- and EMT-associated factors exhibit sufficient sensitivity and selectivity for immunofluorescence microscopy studies of these processes in preclinical xenograft tumor specimens and the potential for application with clinical samples.
Cancer Research | 2017
Robert S. Meehan; Alice P. Chen; Geraldine O’Sullivan Coyne; Shivaani Kummar; Jiuping Ji; Rasa Vilimas; Lamin Juwara; Robert J. Kinders; Katherine V. Ferry-Galow; Deborah Wilsker; Yiping Zhang; Angie B. Dull; Tony Navas; Lihua Wang; Ralph E. Parchment; James H. Doroshow
Inhibition of poly (ADP-ribose) polymerase (PARP) sensitizes tumor cells to DNA damage that would normally be repaired through the base excision repair pathway. PARP inhibitors are active clinically against BRCA-deficient ovarian cancers. The PARP inhibitor talazoparib produces cytotoxicity in human cancer cell lines and animal models of tumors that harbor mutations that compromise DNA repair pathways. In this study, single agent talazoparib (1000 µg/day) was administered to patients with deleterious BRCA1 or BRCA2 mutations and advanced solid tumors in 28 day cycles. The primary objective of the trial was to examine pharmacodynamic (PD) effects of talazoparib; the secondary objective was to determine response rate in patients whose tumors carry BRCA mutations. Mandatory paired tumor biopsies were obtained pre-treatment and 3-6 hrs post-treatment on cycle 1 day 8. Optional biopsies were collected at the time of progression. One core from each time point was analyzed for PARP inhibition by a validated ELISA assay while the other core was used for IFA analysis of γH2AX. A total of 9 patients (pts) were enrolled and treated before this trial was closed due to lack of drug availability: [prostate (3), ovarian (2), breast (2), uterine sarcoma (1), pancreatic (1)]. Median age was 63 (range: 33-73 yrs); male-to-female ratio was 4:5; and the median number of prior treatments was 6 (range: 1-12). All 9 pts were evaluable for PD endpoints. One pt progressed during the first cycle of treatment; 8 pts were evaluable for clinical response. Mean time on study for evaluable pts was 8 cycles (range: 2-18); 5 of 8 (62%) pts experienced a documented partial responses [ovarian (2), prostate (2), breast (1)] lasting between 4 and 12 cycles (median: 6 cycles). Two pts had stable disease for 4 to 6 cycles, and one progressed after 2 cycles. The agent was well tolerated; the most frequent adverse events were hematologic including grade (gr) 4 anemia (1) and thrombocytopenia (1), and gr 3 anemia (2), neutropenia (1), lymphopenia (1). Decreases in PAR levels (>75%) were observed in all cycle 1 day 8 biopsy pairs, documenting a primary PD effect. Increased γH2AX expression was observed for 4/6 pts in post-dose biopsies; pre-treatment γH2AX levels, measured as %Nuclear Associated Protein (NAP), increased from a mean ± SD of 1.33 ± 1.08 to a post-treatment %NAP mean of 5.60 ± 0.78; (p=0.018), supporting a role for drug-enhanced DNA double strand breaks in the mechanism of action of talazoparib for BRCA mutant tumors. In summary, talazoparib demonstrated significant clinical activity as a single agent in patients with BRCA-deficient tumors and produced substantial reductions in tumor PAR levels in matched pre and post-treatment tumor biopsies. Citation Format: Robert S. Meehan, Alice P. Chen, Geraldine O’Sullivan Coyne, Shivaani Kummar, Jiuping Ji, Rasa Vilimas, Lamin Juwara, Robert J. Kinders, Katherine Ferry-Galow, Deborah Wilsker, Yiping Zhang, Angie B. Dull, Tony Navas, Lihua Wang, Ralph E. Parchment, James H. Doroshow. Pilot trial of talazoparib (BMN 673), an oral PARP inhibitor, in patients with advanced solid tumors carrying deleterious BRCA mutations [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 4678. doi:10.1158/1538-7445.AM2017-4678
Cancer Research | 2017
Tony Navas; Melinda G. Hollingshead; Suzanne Borgel; John Carter; Angelena Millione; Brad A. Gouker; Donna O. Butcher; Susan Holbeck; Apurva K. Srivastava; Robert J. Kinders; Donald P. Bottaro; Shivaani Kumar; Alice Chen; James H. Doroshow; Ralph E. Parchment
Epithelial-mesenchymal transition (EMT) is a critical process implicated in metastasis. Despite extensive research, the clinical significance of EMT remains unclear, and a quantitative evaluation of this process in human tumors has yet to be demonstrated. We previously reported a validated EMT immunofluorescence assay (EMT-IFA), that utilizes β-catenin as a tumor segmentation marker to delineate tumor tissue from surrounding stroma in FFPE tumor biopsies. The assay accurately quantifies individual expression and co-localization of E-cadherin (E) and vimentin (V) in tumor cells (Navas et al. NCI-EORTC 2015). We used this assay to measure changes in both the total number and the corresponding EMT phenotypes of tumor cells in xenograft tissues following treatment with various anticancer agents currently in clinical trials at the NCI. Daily treatment for 14 days with the multikinase inhibitor pazopanib caused significant tumor regression and delayed regrowth in the epithelial (E+) gastric cancer xenograft model MKN45, and the tumor cells remaining after treatment were significantly shifted toward a mesenchymal (V+) phenotype. In contrast, pazopanib (QDx15) had less anti-tumor efficacy in the mesenchymal gastric cancer xenograft model SNU5. In another instance, daily treatment of the MDA-MB-468 breast cancer xenograft model with the BCR-Abl kinase inhibitor nilotinib for 19 days did not demonstrate any significant anti-tumor efficacy or change in the predominance of the E+ phenotype, whereas a cycle of treatment with the anti-tubulin agent paclitaxel (Q3Dx4) led to MDA-MB-468 tumor regression and delayed tumor regrowth after completion of the treatment cycle. Furthermore, the combination of nilotinib with paclitaxel not only significantly diminished the total MDA-MB-468 tumor cell number compared to single-agent arms, but also effectively transformed the EMT phenotype of the tumor, with only a subset of cancer cells surviving by the last day of treatment, which were mostly CD44+CD133+ mesenchymal cells and potential cancer stem cells. The changes in EMT phenotype brought on by effective drug treatments occurred via preferential killing of cells with E+ phenotype, suggesting that V+ and V+E+ tumor cells may be more resistant to therapy. The EMT-IFA provides a much-needed analysis tool suitable for clinical investigation of the proposed role of EMT in tumor progression, metastasis, and acquired drug resistance, and is able to detect changes in EMT signature which may serve as early indicators of treatment efficacy and tumor resistance. Funded by NCI Contract No. HHSN261200800001E. Citation Format: Tony Navas, Melinda G. Hollingshead, Suzanne Borgel, John P. Carter, Angelena Millione, Brad A. Gouker, Donna Butcher, Susan Holbeck, Apurva K. Srivastava, Robert J. Kinders, Donald P. Bottaro, Shivaani Kumar, Alice Chen, James H. Doroshow, Ralph E. Parchment. A clinically validated multiplex immunofluorescence assay for the quantitative assessment of changes in EMT phenotypes in FFPE tumor tissues in response to cancer therapeutics [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 845. doi:10.1158/1538-7445.AM2017-845
Cancer Research | 2016
David M. Evans; Rene Delosh; Julie Laudeman; Chad Ogle; Russell Reinhart; Michael Selby; Silvers Thomas; Robert J. Kinders; Tony Navas; Scott M. Lawrence; Anne Monks; Annamaria Rapisarda; Ralph E. Parchment; James H. Doroshow; Beverly A. Teicher
Three-dimensional (3D) cultures have been proposed as higher fidelity models of in vivo tumors than 2D cultures. To determine whether this idea extends to drug pharmacodynamics, we examined whether 3D cultures of the human melanoma line A375 (grown as tumor spheroids) could replicate the timing and magnitude of the nuclear γH2AX response to the topoisomerase 1 inhibitor, topotecan observed in A375 tumor xenografts (Kinders et al, Clin Can Res 2010). The appearance of γH2AX-positive nuclear foci has been used as a biomarker for drug- and radiation-induced double-strand breaks in DNA; we reported previously that treatment of nu/nu mice harboring A375 xenografts with a single-dose of topotecan induced nuclear γH2AX foci in a dose- and time-dependent manner, which peaked 4 hours after drug administration. A375 spheroids were generated by seeding cells into ULA U-bottom plates; 4 hours of exposure to 0.1 μM topotecan elicited a γH2AX signal in these 3D cultures while cell viability and intracellular ATP levels remained unchanged, indicating a DNA damage repair response at the maximally tolerated topotecan concentration for human hematopoietic cells (Erickson-Miller et al, Can Chemo Pharmacol 2009). Extending drug exposure to 24 hours caused substantial loss of viable cells (calcein AM+) and 50% decline in ATP levels but no further increase in γH2AX. In contrast, the HT29 tumor line was refractory to topotecan in vivo, and exposing 3D cultures of HT-29 spheroids to 0.1 μM topotecan for 24 hours elicited a strong nuclear γH2AX biomarker response in only a small fraction of cells at the surface of the spheroids. Longer exposure durations or supra-pharmacological concentrations (1 μM) of topotecan were required to achieve a strong nuclear γH2AX response in HT-29 spheroids. These results support the hypothesis that the 3D pharmacodynamics (PD) of drug response is similar to PD drug response in vivo for the camptothecin class of topoisomerase 1 inhibitors. Funded by NCI Contract No. HHSN261200800001E. Citation Format: David M. Evans, Rene Delosh, Julie Laudeman, Chad Ogle, Russell Reinhart, Michael Selby, Silvers Thomas, Robert Kinders, Tony Navas, Scott Lawrence, Anne Monks, Annamaria Rapisarda, Ralph E. Parchment, James H. Doroshow, Beverly Teicher. Similar 3D pharmacodynamic (3D-PD) responses of human tumor spheroids and xenografts to topoisomerase 1 inhibitor-induced DNA damage. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 606.
Molecular Cancer Therapeutics | 2015
Tony Navas; Robert J. Kinders; Scott M. Lawrence; Katherine V. Ferry-Galow; Thomas D. Pfister; Apurva K. Srivastava; Sergio Y. Alcoser; Melinda G. Hollingshead; Lindsay M. Dutko; Brad A. Gouker; Donna O. Butcher; Hala Makhlouf; Rodrigo Chuaqui; Donald P. Bottaro; Shivaani Kummar; Alice Chen; James H. Doroshow; Ralph E. Parchment
Epithelial-mesenchymal transition (EMT) is a dynamic process whereby epithelial cells acquire mesenchymal properties. Despite the clinical significance of the acquired mesenchymal properties for metastasis and drug resistance, histopathological evidence of transitional cells in patient samples is lacking and EMT remains an unproven clinical hypothesis. We previously developed and validated a multiplex immunofluorescence assay (IFA) that quantifies the levels of EMT biomarkers (E-Cadherin, Vimentin, β-catenin) in snap-frozen, formalin-fixed, paraffin-embedded (FFPE) tumor tissue (Navas et al, AACR 2013). Building upon that method, we now report a precise, quantitative and unbiased IFA method of tissue analysis (EMT-IFA) using Definiens® software to quantify co-expression of the epithelial marker E-cadherin (E) and mesenchymal marker Vimentin (V) at the cellular level in FFPE clinical biopsies. FFPE human tumor xenografts and cell lines serve as calibrators and reference materials for establishing initial image acquisition parameters for segmented tumor regions of interest. Flanking HE 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr LB-B18.
Cancer Research | 2011
Tony Navas; Trisha Haubrich; Jenny Llamas; Hector Aviña; Zhang Chunying; Myra Perez; Linnette Capo; Monica Leavitt; Rou-Yu Chen; Alla Verlinsky; Zili An; James Secrest; Nandini Ganguly; Daulet Satpayev; Karen Jane Meyrick Morrison; Art Raitano; Pia M. Challita-Eid; Xiao-Chi Jia; Kendall Morrison; Alan Hartford; David R. Stover
Prostate stem cell antigen (PSCA) is a cysteine-rich cell surface glycoprotein expressed in about 50% of pancreatic and prostate cancers. AGS-PSCA is a hybridoma-derived fully human IgG1κ monoclonal antibody (MAb) targeting PSCA previously reported to have anti-tumor efficacy in prostate and pancreatic tumor models. AGS-1C4D4 is a CHO-derived antibody generated from the same gene as AGS-PSCA, with similar specificity and binding affinity to PSCA (Kd = 2.0 × 10-10M). Since hybridoma and CHO-derived MAbs displayed disparate glycosylation patterns by mass spectroscopy, we further evaluated their MAb effector functions. AGS-1C4D4 was found to have more potent ADCC activity in vitro on PSCA-expressing pancreatic cell lines, HPAC and Panc0203, using human PBMCs from multiple normal donors. However, both MAbs were similarly effective in mediating CDC on PSCA-expressing recombinant B300.19 cells. Deglycosylation greatly reduced the relative ADCC and CDC activities of both MAbs compared to their intact versions. Additional studies to elucidate the role of the effector functions of AGS-1C4D4 are currently being conducted in vivo using intact and deglycosylated MAbs and will be presented. While neither intact antibody had any direct cytotoxic activity on HPAC cells in vitro, both MAbs significantly inhibited tumor formation, local invasion and metastases to distant sites in orthotopic HPAC xenograft tumor models in vivo. In addition, AGS-1C4D4 significantly inhibited the growth and metastasis of established orthotopic HPAC tumors in combination with Gemcitabine. AGS-1C4D4 is currently being evaluated in a Phase 2 clinical study for pancreatic cancer in combination with Gemcitabine. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4566. doi:10.1158/1538-7445.AM2011-4566
Archive | 2017
Apurva K. Srivastava; Thomas D. Pfister; Tony Navas; Ralph E. Parchment; James E. Doroshow
Journal of Clinical Oncology | 2018
Geraldine Helen O'Sullivan Coyne; Kevin C. Conlon; Naoko Takebe; Howard Streicher; Mary Flanagan Quinn; Ashley Bruns; Tony Navas; Ralph E. Parchment; Larry Rubinstein; Elad Sharon; James H. Doroshow; Thomas A. Waldmann; Alice P. Chen
Journal of Clinical Oncology | 2018
Ralph E. Parchment; Tony Navas; Kristin Fino; Andy Fung; Facundo Cutuli; Lihua Wang; Robert J. Kinders; James H. Doroshow
Annals of Translational Medicine | 2017
Apurva K. Srivastava; Tony Navas; William G. Herrick; Melinda G. Hollingshead; Donald P. Bottaro; James H. Doroshow; Ralph E. Parchment