Galeterone and its analogs inhibit Mnk-eIF4E axis, synergize with gemcitabine, impede pancreatic cancer cell migration, invasion and proliferation and inhibit tumor growth in mice

Andrew K Kwegyir-Afful, Francis N Murigi, Puranik Purushottamachar, Vidya P Ramamurthy, Marlena S Martin, Vincent C O Njar, Andrew K Kwegyir-Afful, Francis N Murigi, Puranik Purushottamachar, Vidya P Ramamurthy, Marlena S Martin, Vincent C O Njar

Abstract

Survival rate for pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) is poor, with about 80% of patients presenting with the metastatic disease. Gemcitabine, the standard chemotherapeutic agent for locally advanced and metastatic PDAC has limited efficacy, attributed to innate/acquired resistance and activation of pro-survival pathways. The Mnk1/2-eIF4E and NF-κB signaling pathways are implicated in PDAC disease progression/metastasis and also associated with gemcitabine-induced resistance in PDAC. Galeterone (gal), a multi-target, agent in phase III clinical development for prostate cancer has also shown effects on the aforementioned pathways. We show for the first time, that gal/analogs (VNPT55, VNPP414 and VNPP433-3β) profoundly inhibited cell viability of gemcitabine-naive/resistance PDAC cell lines and strongly synergized with gemcitabine in gemcitabine-resistant PDAC cells. In addition, to inducing G1 cell cycle arrest, gal/analogs induced caspase 3-mediated cell-death of PDAC cells. Gal/analogs caused profound downregulation of Mnk1/2, peIF4E and NF-κB (p-p65), metastatic inducing factors (N-cadherin, MMP-1/-2/-9, Slug, Snail and CXCR4) and putative stem cell factors, (β-Catenin, Nanog, BMI-1 and Oct-4). Gal/analog also depleted EZH2 and upregulated E-Cadherin. These effects resulted in significant inhibition of PDAC cell migration, invasion and proliferation. Importantly, we also observed strong MiaPaca-2 tumor xenograft growth inhibition (61% to 92%). Collectively, these promising findings strongly support further development of gal/analogs as novel therapeutics for PDAC.

Keywords: galeterone (gal) and its analogs; pancreatic cancer resistance.

Conflict of interest statement

CONFLICTS OF INTEREST Vincent C. O. Njar is the lead inventor of galeterone (gal, VN/124-1 or TOK-001), the new analog, patents and technologies thereof owned by the University of Maryland, Baltimore, and licensed to Tokai Pharmaceuticals, Inc. Puranik Purushottamachar and Andrew K. Kwegyir-Afful are co-inventors of VNPT55 and related compounds. Patent applications to protect these novel compounds and their potential utility against pancreatic cancer have been filed. The other authors declare no potential conflicts of interest.

Figures

Figure 1. Anti-proliferative activities of ARDAs and…
Figure 1. Anti-proliferative activities of ARDAs and gemcitabine in PDAC cells
A. Structures of Androgen Receptor Degrading Agents (ARDA). B. Gal and analogs decrease cell viability of MiaPaCa-2 cells utilizing MTT cell viability assays. C. MTT cell viability assays in drug-naive and drug-resistant (MiaPaCa-2, MiaPaCa-GR and MiaPaCa-GTR) show a decrease in gemcitabine activity in drug-resistant cells D. Gal and analogs potentiate effects of gemcitabine and strikingly decrease GI50values in gemcitabine-resistant PDAC cells (**p < 0.001). PDAC cells were first treated with gal and analogs, then with gemcitabine. E. Gal and gemcitabine synergistically inhibit cell viability of gemcitabine-resistant PDAC cells. Cell viability assays were conducted for gal and gemcitabine individually and GI50 values calculated. Both compounds were combined at a constant ratio and utilized in cell viability assays. Fractional effects of single agents and in combination were calculated and analyzed by the calcusyn software to compute the combination indices (CI) at ED50, ED75 and ED90. CI value at ED90 reveal strong synergy between the two compounds in MiaPaCa-GR cells.
Figure 2. Gal/analogs and gemcitabine cause cell…
Figure 2. Gal/analogs and gemcitabine cause cell cycle arrest and induce apoptosis in PDAC cells
A. Gal and VNPP433-3B, induce G1 cell cycle arrest in MiaPaCa-2 cells. B. Gal and analogs cause accumulation of cells in the G1 phase of cell cycle in metastatic PDAC cells (S2-VP10). C. Gal and analogs deplete cell cycle regulators (cyclin B1 and cyclin D1) and upregulate p21 in MiaPaCa-2, S2-VP10 and MiaPaCa-GTR cells. D. Cell viability assays indicate that concentrations utilized (5 and 10 μM), over a 24 h time point did not compromise cell viability significantly. E. Gal/analogs and gemcitabine induce apoptosis in S2-013 cells in vitro, using the acridine orange/ethidium bromide assay to determine loss of cell membrane integrity. Cells were treated at 2.5 μM for 72 h. Arrows next to L indicate live cells; arrows pointing to A indicate apoptotic cells; and arrows pointing N indicate necrotic cells F. Gal/VNPP433-3β and gemcitabine (5 and 10 μM) were compared in their ability to induce apoptosis, analyzed by flow cytometry. Cells treated with compounds for 24 h were stained with annexin v and propidium iodide (PI). Early and late apoptotic cells were analyzed by FACS. All compounds show significant apoptotic induction (*p < 0.05, **p < 0.001). G. Gal and analogs enhanced depletion of Bcl-2 and increased expression of cleaved PARP, caspase 3 and Bax in gemcitabine-naive/resistant PDAC cells.
Figure 3. Gal and analogs deplete Mnk1/2…
Figure 3. Gal and analogs deplete Mnk1/2 protein expression and downregulate eIF4E phosphorylation
A and B. ASPC1 and S2-013 cells treated with gal and VNPP433-3β show a dose dependent decrease in K-Ras, Mnk1 and peIF4E. C and D. CaPan-1 and MiaPaCa-GTR cells treated with gal and VNPP414 at 10 μM also show decrease in Ras, Mnk2 and peIF4E protein expression. E. S2-013 cells treated with gal/VNPP433-3β/gemcitabine was analyzed for phosphorylation levels of eIF4G, eIF4E and 4E-BP. Protein expression analysis reveal an enhanced expression of Mnk1 by gemcitabine treatment.
Figure 4. Gal and analogs inhibit metastatic…
Figure 4. Gal and analogs inhibit metastatic inducing pathways
A. MiaPaCa-2 and S2-013 cells treated with gal and VNPP433-3β at increasing concentration show a dose-dependent decrease in p-p65. B. MiaPaCa-GR and MiaPaCa-GTR cells exposed to gal and VNPTP433-3β show profound depletion of EZH2 protein expression MiaPaCa-GTR (left panel) and MiaPaCa-GR (right panel). C. S2-013 cells, left panel, exposed to gal (5 – 10 μM) show downregulation of N-cadherin, and an upregulation of E-cadherin (bottom graph). Densitometric analysis suggests a profound increase in E-Cad: N-Cad ratio, suggesting loss of mesenchymal characteristics. D. S2-VP10 cells exposed to 10 μM of gal/analogs exhibit a downregulation of N-cadherin and an upregulation of E-cadherin resulting in an increase in E-cad N-cad ratio (bottom chart). E. Gal and VNPP433-3β deplete MMP-2/-9 in MiaPaCa-2 and MiaPaCa-GTR cells after 24 h treatment period. F. S2-013 cells transfected with Mnk1 siRNA show downregulation of N-cadherin, MMP-2/-9 and peIF4E.
Figure 5. Downregulation of EMT markers
Figure 5. Downregulation of EMT markers
after A, top panel) Media from S2-013 cells, treated with indicated compounds was separated on a zymogram gel to analyze the proteolytic activity of MMP-1/-9. (A, bottom graph) densitometric analysis of zymogram gel in top panel, reveals a significant decrease in collagenase activity of MMP-1/-9 (**p < 0.001), zymogram assays were repeated at least three times and presented as mean ± S.E.M. B. Densitometry of proteolytically digested bands on zymogram gels analyzed from gal, CGP, gemcitabine and combinations’ treated MiaPaCa-2 cultures, shows significant upregulation of MMP-9 collagenase activity in the presence of gemcitabine. Gal and CGP both decrease gemcitabine-induced MMP-9 activity (*p < 0.05, **p < 0.001). C and D. Gemcitabine-naïve, Gemcitabine-resistant and Gemcitabine/erlotinib resistant, PDAC cells, were treated with gal and VNPP433-3β for 24 h at 10 μM. Protein expression analysis show downregulation of Snail, Slug, Cox-2, CDC42 and CXCR4 in all four cell lines.
Figure 6. Gal and analogs possess anti-migratory…
Figure 6. Gal and analogs possess anti-migratory and anti-invasive properties
A. Scratch wound healing assays reveal that gal/analogs, CGP and gemcitabine at 5 μM, inhibit Panc-1(top panel) and S2-013 (bottom panel) PDAC cell migration. B. Distance migrated by Panc-1 and S2-013 cells after treatment were quantified. Wounds were measured before and after the 12 h time point. Distance migrated were quantified by measuring the difference at time 0 and 12 h and normalized to control. (Distance migrated = Distance at time 0 hour - distance at 12 h/ Distance migrated by control) (*p < 0.05, **p < 0.001). C. Gal/analogs, CGP, gemcitabine at 5 μM, inhibit gemcitabine-naive (CaPan-1), MiaPaCa-GR and MiaPaCa-GTR PDAC cell invasion. D. Quantification of invaded PDAC cells was done by counting cells in quadrants. Only invading cells at the bottom of inserts were counted. Quantified invaded cells shows a significant inhibition of PDAC cell invasion (**p < 0.001).
Figure 7. Gal and analogs inhibit colony…
Figure 7. Gal and analogs inhibit colony formation of PDAC cell
A. Gal/VNPP433-3β and gemcitabine significantly inhibit colony formation of ASPC1 and B. S2-013 cells. C. Gal/VNPP414/VNPP433-3β/CGP and gemcitabine decreased colony formation of S2-VP10 (left panel) and MiaPaCa-2 (right panel) cells. Both VNPP414 and CGP potentiated the effects of Gemcitabine and severely reduced colonies (C, right panel) in MiaPaCa-2 cells. D. Gal and VNPP433-3β significantly inhibited colony forming units (CFU) of gemcitabine-erlotinib-resistant PDAC cells and potentiated the effects of gemcitabine in further decreasing the number of colonies formed, contrary to gemcitabine alone which did not show strong inhibition of colonies. (D, right graph) Colony formation assays were repeated at least three times and colonies counted in four quadrants of the wells. Results are represented as averages with S.E.M. (*p < 0.05, **p < 0.001). Note: The numbers in parenthesis are concentrations in μM.
Figure 8. Gal and analogs downregulate stem…
Figure 8. Gal and analogs downregulate stem cell factors and inhibit sphere formation in S2-013 PDAC cells
A. 200 cells resuspended in media to break up clumps and ensure single cell suspension were seeded in ultra-low adherent 24-well plates. After sphere formation, cells were treated with indicated compounds 2X in 14days and spheroids analyzed by light microscopy and images taken. B and C. Gal and VNPP433-3β deplete protein expression of putative stem cell factors in gemcitabine-naïve and gemcitabine-resistant PDAC cells. β-Catenin, Oct-4, Nanog, BMI-1 and cell proliferation factor c-Myc were downregulated after 24 h treatment period.
Figure 9. In vivo activity of gal…
Figure 9. In vivo activity of gal and analogs
A. Effect of gal, VNPT55, VNPP414 and VNPP433-3β were evaluated in MiaPaCa-2 PDAC xenograft-bearing mice. Mice (n = 5) were administered with gal [0.26 mmol/kg (100.9 mg/kg)/twice daily], VNPT55 [0.26 mmol/kg (125.4 mg/kg)/twice daily], VNPP414 [0.068 mmol/kg (32.6 mg/kg)/twice daily], and VNPP433-3β [0.068 mmol/kg (30 mg/kg)/once daily], by intraperitoneal injection, 5 days per week for 32 days. Tumors were measured twice a week. Gal and analogs significantly inhibited tumor growth (** p< 0.001; %T/C: ratio of tumor volume in treated mice vs. control). Effective criteria for %T/C value according to NCI standard is ≤42% [50, 51]. TGI: Tumor Growth Inhibition index = [1-(mean volume of treated tumors)/(mean volume of control tumors)] × 100%). B. Mean body weights of mice were taken twice a week for the duration of the study. Mean body weights showed no significant toxicities to mice. C. H & E stain of liver, lung and kidney in both compound treated animals and vehicle treated animal show no gross organ abnormalities. D. Western blot analyses show a marked depletion of Mnk1/2, EZH2 and an upregulation of proapoptotic protein Bax by all four compounds in vivo.
Figure 10. Downregulation of oncogenic biomarkers in…
Figure 10. Downregulation of oncogenic biomarkers in vivo
A. Immunohistochemical staining show significant depletion of Mnk1/2, peIF4E, BMI-1, SLUG, vimentin and PCNA from paraffinized sections of tumor tissue. B. Quantified IHC stains, show a significant downregulation of biomarkers in vivo (**p < 0.001).

References

    1. Siegel R, Desantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–117.
    1. Magee CJ, Ghaneh P, Neoptolemos JP. Surgical and medical therapy for pancreatic carcinoma. Best Pract Res Clin Gastroenterol. 2002;16:435–455.
    1. Ottenhof NA, de Wilde RF, Maitra A, Hruban RH, Offerhaus GJ. Molecular characteristics of pancreatic ductal adenocarcinoma. Patholog Res Int. 2011;2011:620601.
    1. di Magliano MP, Logsdon CD. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology. 2013;144:1220–1229.
    1. Mendoza MC, Er EE, Blenis J. The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends Biochem Sci. 2011;36:320–328.
    1. Bitterman PB, Polunovsky VA. Attacking a nexus of the oncogenic circuitry by reversing aberrant eIF4F-mediated translation. Mol Cancer Ther. 2012;11:1051–1061.
    1. Bitterman PB, Polunovsky VA. Translational control of cell fate: from integration of environmental signals to breaching anticancer defense. Cell Cycle. 2012;11:1097–1107.
    1. Hay N. Mnk earmarks eIF4E for cancer therapy. Proc Natl Acad Sci U S A. 2010;107:13975–13976.
    1. Topisirovic I, Ruiz-Gutierrez M, Borden KL. Phosphorylation of the eukaryotic translation initiation factor eIF4E contributes to its transformation and mRNA transport activities. Cancer Res. 2004;64:8639–8642.
    1. Wendel HG, Silva RL, Malina A, Mills JR, Zhu H, Ueda T, Watanabe-Fukunaga R, Fukunaga R, Teruya-Feldstein J, Pelletier J, Lowe SW. Dissecting eIF4E action in tumorigenesis. Genes Dev. 2007;21:3232–3237.
    1. Konicek BW, Dumstorf CA, Graff JR. Targeting the eIF4F translation initiation complex for cancer therapy. Cell Cycle. 2008;7:2466–2471.
    1. Silvera D, Formenti SC, Schneider RJ. Translational control in cancer. Nat Rev Cancer. 2010;10:254–266.
    1. Bhat M, Robichaud N, Hulea L, Sonenberg N, Pelletier J, Topisirovic I. Targeting the translation machinery in cancer. Nat Rev Drug Discov. 2015;14:261–278.
    1. Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, Lolkema MP, Buchholz M, Olive KP, Gress TM, Tuveson DA. Stromal biology and therapy in pancreatic cancer. Gut. 2011;60:861–868.
    1. Vaccaro V, Sperduti I, Milella M. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;365:768–769. author reply 769.
    1. Adesso L, Calabretta S, Barbagallo F, Capurso G, Pilozzi E, Geremia R, Delle Fave G, Sette C. Gemcitabine triggers a pro-survival response in pancreatic cancer cells through activation of the MNK2/eIF4E pathway. Oncogene. 2013;32:2848–2857.
    1. Arlt A, Gehrz A, Muerkoster S, Vorndamm J, Kruse ML, Folsch UR, Schafer H. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene. 2003;22:3243–3251.
    1. Prabhu L, Mundade R, Korc M, Loehrer PJ, Lu T. Critical role of NF-kappaB in pancreatic cancer. Oncotarget. 2014;5:10969–10975. doi: 10.18632/oncotarget.2624.
    1. Baylot V, Andrieu C, Katsogiannou M, Taieb D, Garcia S, Giusiano S, Acunzo J, Iovanna J, Gleave M, Garrido C, Rocchi P. OGX-427 inhibits tumor progression and enhances gemcitabine chemotherapy in pancreatic cancer. Cell Death Dis. 2011;2:e221.
    1. Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, Gayral M, Cordelier P, Delisle MB, Bousquet-Dubouch MP, Tomasini R, Schmid H, Mathonnet M, et al. Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Mol Med. 2015;7:735–753.
    1. Kumar K, Chow CR, Ebine K, Arslan AD, Kwok B, Bentrem DJ, Eckerdt FD, Platanias LC, Munshi HG. Differential Regulation of ZEB1 and EMT by MAPK-Interacting Protein Kinases (MNK) and eIF4E in Pancreatic Cancer. Mol Cancer Res. 2016;14:216–227.
    1. Njar VCO, Brodie AM. Discovery and development of Galeterone (TOK-001 or VN/124-1) for the treatment of all stages of prostate cancer. J Med Chem. 2015;58:2077–2087.
    1. Bruno RD, Gover TD, Burger AM, Brodie AM, Njar VCO. 17alpha-Hydroxylase/17,20 lyase inhibitor VN/124-1 inhibits growth of androgen-independent prostate cancer cells via induction of the endoplasmic reticulum stress response. Mol Cancer Ther. 2008;7:2828–2836.
    1. Kwegyir-Afful AK, Bruno RD, Purushottamachar P, Murigi FN, Njar VCO. Galeterone and VNPT55 disrupt Mnk-eIF4E to inhibit prostate cancer cell migration and invasion. FEBS J. 2016;283:3898–3918.
    1. Montgomery B, Eisenberger MA, Rettig MB, Chu F, Pili R, Stephenson JJ, Vogelzang NJ, Koletsky AJ, Nordquist LT, Edenfield WJ, Mamlouk K, Ferrante KJ, Taplin ME. Androgen Receptor Modulation Optimized for Response (ARMOR) Phase I and II Studies: Galeterone for the Treatment of Castration-Resistant Prostate Cancer. Clin Cancer Res. 2016;22:1356–1363.
    1. Purushottamachar P, Godbole AM, Gediya LK, Martin MS, Vasaitis TS, Kwegyir-Afful AK, Ramalingam S, Ates-Alagoz Z, Njar VCO. Systematic structure modifications of multitarget prostate cancer drug candidate galeterone to produce novel androgen receptor down-regulating agents as an approach to treatment of advanced prostate cancer. J Med Chem. 2013;56:4880–4898.
    1. Kwegyir-Afful AK, Ramalingam S, Purushottamachar P, Ramamurthy VP, Njar VCO. Galeterone and VNPT55 induce proteasomal degradation of AR/AR-V7, induce significant apoptosis via cytochrome c release and suppress growth of castration resistant prostate cancer xenografts in vivo. Oncotarget. 2015;6:27440–27460. doi: 10.18632/oncotarget.4578.
    1. Greenway BA. Androgen receptor-blocking agents: potential role in pancreatic cancer. Drugs Aging. 2000;17:161–163.
    1. Konduri S, Schwarz MA, Cafasso D, Schwarz RE. Androgen receptor blockade in experimental combination therapy of pancreatic cancer. J Surg Res. 2007;142:378–386.
    1. Kwegyir-Afful AK, Purushottamachar P, Martin MS, Njar VCO. Galeterone and its novel analogs induce profound anti-cancer activities in human pancreatic cancer cell lines: Implications for pancreatic cancer therapy. Cancer Reserach. 2015;75:1764–1764.
    1. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984;22:27–55.
    1. Sparreboom A, de Jonge MJ, Verweij J. The use of oral cytotoxic and cytostatic drugs in cancer treatment. Eur J Cancer. 2002;38:18–22.
    1. Rixe O, Fojo T. Is cell death a critical end point for anticancer therapies or is cytostasis sufficient? Clinical cancer research. 2007;13:7280–7287.
    1. Millar AW, Lynch KP. Rethinking clinical trials for cytostatic drugs. Nature reviews Cancer. 2003;3:540–545.
    1. Wang W, Riedel K, Lynch P, Chien CY, Montelione GT, Krug RM. RNA binding by the novel helical domain of the influenza virus NS1 protein requires its dimer structure and a small number of specific basic amino acids. RNA. 1999;5:195–205.
    1. Shin YJ, Kim JH. The role of EZH2 in the regulation of the activity of matrix metalloproteinases in prostate cancer cells. PloS one. 2012;7:e30393.
    1. Gress TM, Muller-Pillasch F, Lerch MM, Friess H, Buchler M, Adler G. Expression and in-situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. International journal of cancer Journal international du cancer. 1995;62:407–413.
    1. Maatta M, Soini Y, Liakka A, Autio-Harmainen H. Differential expression of matrix metalloproteinase (MMP)-2, MMP-9, and membrane type 1-MMP in hepatocellular and pancreatic adenocarcinoma: implications for tumor progression and clinical prognosis. Clinical cancer research. 2000;6:2726–2734.
    1. Harvey SR, Hurd TC, Markus G, Martinick MI, Penetrante RM, Tan D, Venkataraman P, DeSouza N, Sait SN, Driscoll DL, Gibbs JF. Evaluation of urinary plasminogen activator, its receptor, matrix metalloproteinase-9, and von Willebrand factor in pancreatic cancer. Clinical cancer research. 2003;9:4935–4943.
    1. Soifer HS, Souleimanian N, Wu S, Voskresenskiy AM, Collak FK, Cinar B, Stein CA. Direct regulation of androgen receptor activity by potent CYP17 inhibitors in prostate cancer cells. J Biol Chem. 2012;287:3777–3787.
    1. Shekhar MP, Pauley R, Heppner G. Host microenvironment in breast cancer development: extracellular matrix-stromal cell contribution to neoplastic phenotype of epithelial cells in the breast. Breast cancer research. 2003;5:130–135.
    1. Poser I, Dominguez D, de Herreros AG, Varnai A, Buettner R, Bosserhoff AK. Loss of E-cadherin expression in melanoma cells involves up-regulation of the transcriptional repressor Snail. J Biol Chem. 2001;276:24661–24666.
    1. Nakajima S, Doi R, Toyoda E, Tsuji S, Wada M, Koizumi M, Tulachan SS, Ito D, Kami K, Mori T, Kawaguchi Y, Fujimoto K, Hosotani R, Imamura M. N-cadherin expression and epithelial-mesenchymal transition in pancreatic carcinoma. Clinical cancer research. 2004;10:4125–4133.
    1. Sahai E, Marshall CJ. RHO-GTPases and cancer. Nat Rev Cancer. 2002;2:133–142.
    1. Tang Y, Olufemi L, Wang MT, Nie D. Role of Rho GTPases in breast cancer. Front Biosci. 2008;13:759–776.
    1. Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiD lenalidomide. Anticancer Res. 2011;31:3747–3756.
    1. Sarkar FH, Li Y, Wang Z, Kong D. Pancreatic cancer stem cells and EMT in drug resistance and metastasis. Minerva Chir. 2009;64:489–500.
    1. Bao B, Wang Z, Ali S, Ahmad A, Azmi AS, Sarkar SH, Banerjee S, Kong D, Li Y, Thakur S, Sarkar FH. Metformin inhibits cell proliferation, migration and invasion by attenuating CSC function mediated by deregulating miRNAs in pancreatic cancer cells. Cancer Prev Res (Phila) 2012;5:355–364.
    1. Yu Y, Kanwar SS, Patel BB, Nautiyal J, Sarkar FH, Majumdar AP. Elimination of Colon Cancer Stem-Like Cells by the Combination of Curcumin and FOLFOX. Transl Oncol. 2009;2:321–328.
    1. Bissery MC, Chabot GG. [History and new development of screening and evaluation methods of anticancer drugs used in vivo and in vitro]. [Article in French] Bull Cancer. 1991;78:587–602.
    1. Hollingshead MG. Antitumor efficacy testing in rodents. J Natl Cancer Inst. 2008;100:1500–1510.
    1. Venkannagari S, Fiskus W, Peth K, Atadja P, Hidalgo M, Maitra A, Bhalla KN. Superior efficacy of co-treatment with dual PI3K/mTOR inhibitor NVP-BEZ235 and pan-histone deacetylase inhibitor against human pancreatic cancer. Oncotarget. 2012;3:1416–1427. doi: 10.18632/oncotarget.724.
    1. Skrypek N, Duchene B, Hebbar M, Leteurtre E, van Seuningen I, Jonckheere N. The MUC4 mucin mediates gemcitabine resistance of human pancreatic cancer cells via the Concentrative Nucleoside Transporter family. Oncogene. 2013;32:1714–1723.
    1. Fujii S, Fukamachi K, Tsuda H, Ito K, Ito Y, Ochiai A. RAS oncogenic signal upregulates EZH2 in pancreatic cancer. Biochemical and biophysical research communications. 2012;417:1074–1079.
    1. Li X, Ma Q, Xu Q, Duan W, Lei J, Wu E. Targeting the cancer-stroma interaction: a potential approach for pancreatic cancer treatment. Current pharmaceutical design. 2012;18:2404–2415.
    1. Stamenkovic I. Matrix metalloproteinases in tumor invasion and metastasis. Seminars in cancer biology. 2000;10:415–433.
    1. Okami J, Yamamoto H, Fujiwara Y, Tsujie M, Kondo M, Noura S, Oshima S, Nagano H, Dono K, Umeshita K, Ishikawa O, Sakon M, Matsuura N, et al. Overexpression of cyclooxygenase-2 in carcinoma of the pancreas. Clin Cancer Res. 1999;5:2018–2024.
    1. Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ., 3rd Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res. 1999;59:987–990.
    1. Yip-Schneider MT, Barnard DS, Billings SD, Cheng L, Heilman DK, Lin A, Marshall SJ, Crowell PL, Marshall MS, Sweeney CJ. Cyclooxygenase-2 expression in human pancreatic adenocarcinomas. Carcinogenesis. 2000;21:139–146.
    1. Bu X, Zhao C, Dai X. Involvement of COX-2/PGE(2) Pathway in the Upregulation of MMP-9 Expression in Pancreatic Cancer. Gastroenterol Res Pract. 2011;2011:214269.
    1. Endo H, Watanabe T, Sugioka Y, Niioka M, Inagaki Y, Okazaki I. Activation of two distinct MAPK pathways governs constitutive expression of matrix metalloproteinase-1 in human pancreatic cancer cell lines. International journal of oncology. 2009;35:1237–1245.
    1. Hotz B, Arndt M, Dullat S, Bhargava S, Buhr HJ, Hotz HG. Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clinical cancer research. 2007;13:4769–4776.
    1. Yin T, Wang C, Liu T, Zhao G, Zha Y, Yang M. Expression of snail in pancreatic cancer promotes metastasis and chemoresistance. J Surg Res. 2007;141:196–203.
    1. Boutet A, De Frutos CA, Maxwell PH, Mayol MJ, Romero J, Nieto MA. Snail activation disrupts tissue homeostasis and induces fibrosis in the adult kidney. EMBO J. 2006;25:5603–5613.
    1. Wang Z, Ma Q, Li P, Sha H, Li X, Xu J. Aberrant expression of CXCR4 and beta-catenin in pancreatic cancer. Anticancer Res. 2013;33:4103–4110.
    1. Schleger C, Verbeke C, Hildenbrand R, Zentgraf H, Bleyl U. c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Modern pathology. 2002;15:462–469.
    1. Kumar K, Raza SS, Knab LM, Chow CR, Kwok B, Bentrem DJ, Popovic R, Ebine K, Licht JD, Munshi HG. GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Scientific reports. 2015;5:9489.
    1. Purushottamachar P, Kwegyir-Afful AK, Martin MS, Ramamurthy VP, Ramalingam S, Njar VCO. Identification of Novel Steroidal Androgen Receptor Degrading Agents Inspired by Galeterone 3beta-Imidazole Carbamate. ACS Med Chem Lett. 2016;7:708–713.
    1. Purushottamachar P, Murigi FM, Njar VCO. Improved Procedures for Gram-Scale Synthesis of Galeterone 3β-Imidazole and Galeterone 3β-Pyridine Methoxylate, Potent Androgen Receptor/Mnk Degrading Agents. Organic Process Research & Development. 2016;20:1654–1661.

Source: PubMed

3
Se inscrever