Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 2/2013

01-02-2013 | Original Article

In vitro cytotoxicity, pharmacokinetics, tissue distribution, and metabolism of small-molecule protein kinase D inhibitors, kb-NB142-70 and kb-NB165-09, in mice bearing human cancer xenografts

Authors: Jianxia Guo, Dana M. Clausen, Jan H. Beumer, Robert A. Parise, Merrill J. Egorin, Karla Bravo-Altamirano, Peter Wipf, Elizabeth R. Sharlow, Qiming Jane Wang, Julie L. Eiseman

Published in: Cancer Chemotherapy and Pharmacology | Issue 2/2013

Login to get access

Abstract

Purpose

Protein kinase D (PKD) mediates diverse biological responses including cell growth and survival. Therefore, PKD inhibitors may have therapeutic potential. We evaluated the in vitro cytotoxicity of two PKD inhibitors, kb-NB142-70 and its methoxy analogue, kb-NB165-09, and examined their in vivo efficacy and pharmacokinetics.

Methods

The in vitro cytotoxicities of kb-NB142-70 and kb-NB165-09 were evaluated by MTT assay against PC-3, androgen-independent prostate cancer cells, and CFPAC-1 and PANC-1, pancreatic cancer cells. Efficacy studies were conducted in mice bearing either PC-3 or CPFAC-1 xenografts. Tumor-bearing mice were euthanized between 5 and 1,440 min after iv dosing, and plasma and tissue concentrations were measured by HPLC–UV. Metabolites were characterized by LC–MS/MS.

Results

kb-NB142-70 and kb-NB165-09 inhibited cellular growth in the low–mid μM range. The compounds were inactive when administered to tumor-bearing mice. In mice treated with kb-NB142-70, the plasma C max was 36.9 nmol/mL, and the PC-3 tumor C max was 11.8 nmol/g. In mice dosed with kb-NB165-09, the plasma C max was 61.9 nmol/mL, while the PANC-1 tumor C max was 8.0 nmol/g. The plasma half-lives of kb-NB142-70 and kb-NB165-09 were 6 and 14 min, respectively. Both compounds underwent oxidation and glucuronidation.

Conclusions

kb-NB142-70 and kb-NB165-09 were rapidly metabolized, and concentrations in tumor were lower than those required for in vitro cytotoxicity. Replacement of the phenolic hydroxyl group with a methoxy group increased the plasma half-life of kb-NB165-09 2.3-fold over that of kb-NB142-70. Rapid metabolism in mice suggests that next-generation compounds will require further structural modifications to increase potency and/or metabolic stability.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rennecke J, Rehberger PA, Fürstenberger G, Johannes FJ, Stöhr M, Marks F, Richter KH (1999) Protein-kinase-Cmu expression correlates with enhanced keratinocyte proliferation in normal and neoplastic mouse epidermis and in cell culture. Int J Cancer 80:98–103PubMedCrossRef Rennecke J, Rehberger PA, Fürstenberger G, Johannes FJ, Stöhr M, Marks F, Richter KH (1999) Protein-kinase-Cmu expression correlates with enhanced keratinocyte proliferation in normal and neoplastic mouse epidermis and in cell culture. Int J Cancer 80:98–103PubMedCrossRef
2.
go back to reference Zhukova E, Sinnett-Smith J, Rozengurt E (2001) Protein kinase D potentiates DNA synthesis and cell proliferation induced by bombesin, vasopressin, or phorbol esters in swiss 3T3 cells. J Biol Chem 276:40298–40305PubMed Zhukova E, Sinnett-Smith J, Rozengurt E (2001) Protein kinase D potentiates DNA synthesis and cell proliferation induced by bombesin, vasopressin, or phorbol esters in swiss 3T3 cells. J Biol Chem 276:40298–40305PubMed
3.
go back to reference Johannes FJ, Horn J, Link G, Haas E, Siemienski K, Wajant H, Pfizenmaier K (1998) Protein kinase Cmu downregulation of tumor-necrosis-factor-induced apoptosis correlates with enhanced expression of nuclear-factor-kappaB-dependent protective genes. Eur J Biochem 257:47–54PubMedCrossRef Johannes FJ, Horn J, Link G, Haas E, Siemienski K, Wajant H, Pfizenmaier K (1998) Protein kinase Cmu downregulation of tumor-necrosis-factor-induced apoptosis correlates with enhanced expression of nuclear-factor-kappaB-dependent protective genes. Eur J Biochem 257:47–54PubMedCrossRef
4.
go back to reference Endo K, Oki E, Biedermann V, Kojima H, Yoshida K, Johannes FJ, Kufe D, Datta R (2000) Proteolytic cleavage and activation of protein kinase C [micro] by caspase-3 in the apoptotic response of cells to 1-beta-d-arabinofuranosylcytosine and other genotoxic agents. J Biol Chem 275:18476–18481PubMedCrossRef Endo K, Oki E, Biedermann V, Kojima H, Yoshida K, Johannes FJ, Kufe D, Datta R (2000) Proteolytic cleavage and activation of protein kinase C [micro] by caspase-3 in the apoptotic response of cells to 1-beta-d-arabinofuranosylcytosine and other genotoxic agents. J Biol Chem 275:18476–18481PubMedCrossRef
5.
go back to reference Brändlin I, Hübner S, Eiseler T, Martinez-Moya M, Horschinek A, Hausser A, Link G, Rupp S, Storz P, Pfizenmaier K, Johannes FJ (2002) Protein kinase C (PKC)eta-mediated PKC mu activation modulates ERK and JNK signal pathways. J Biol Chem 277:6490–6496PubMedCrossRef Brändlin I, Hübner S, Eiseler T, Martinez-Moya M, Horschinek A, Hausser A, Link G, Rupp S, Storz P, Pfizenmaier K, Johannes FJ (2002) Protein kinase C (PKC)eta-mediated PKC mu activation modulates ERK and JNK signal pathways. J Biol Chem 277:6490–6496PubMedCrossRef
6.
go back to reference Rey O, Young SH, Cantrell D, Rozengurt E (2001) Rapid protein kinase D translocation in response to G protein-coupled receptor activation. J Biol Chem 276:32616–32626PubMedCrossRef Rey O, Young SH, Cantrell D, Rozengurt E (2001) Rapid protein kinase D translocation in response to G protein-coupled receptor activation. J Biol Chem 276:32616–32626PubMedCrossRef
7.
go back to reference Paolucci L, Rozengurt E (1999) Protein kinase D in small cell lung cancer cells: rapid activation through protein kinase C. Cancer Res 59:572–577PubMed Paolucci L, Rozengurt E (1999) Protein kinase D in small cell lung cancer cells: rapid activation through protein kinase C. Cancer Res 59:572–577PubMed
8.
go back to reference Sidorenko SP, Law CL, Klaus SJ, Chandran KA, Takata M, Kurosaki T, Clark EA (1996) Protein kinase C mu (PKC mu) associates with the B cell antigen receptor complex and regulates lymphocyte signaling. Immunity 5:353–363PubMedCrossRef Sidorenko SP, Law CL, Klaus SJ, Chandran KA, Takata M, Kurosaki T, Clark EA (1996) Protein kinase C mu (PKC mu) associates with the B cell antigen receptor complex and regulates lymphocyte signaling. Immunity 5:353–363PubMedCrossRef
9.
go back to reference Rozengurt E, Sinnett-Smith J, Van Lint J, Valverde AM (1995) Protein kinase D: a novel target for diacylglycerol and phorbol esters. Mutat Res 326:545–551 Rozengurt E, Sinnett-Smith J, Van Lint J, Valverde AM (1995) Protein kinase D: a novel target for diacylglycerol and phorbol esters. Mutat Res 326:545–551
10.
go back to reference Haworth RS, Sinnett-Smith J, Rozengurt E, Avkiran M (1999) Protein kinase D inhibits plasma membrane Na+/H+ exchanger activity. Am J Physiol 277:C1202–C1209PubMed Haworth RS, Sinnett-Smith J, Rozengurt E, Avkiran M (1999) Protein kinase D inhibits plasma membrane Na+/H+ exchanger activity. Am J Physiol 277:C1202–C1209PubMed
11.
go back to reference Rykx A, Kimpe LD, Mikhalap S, Vantus T, Seufferlein T, Vandenheede JR, Lint JV (2003) Protein kinase D: a family affair. FEBS Lett 546:81–86PubMedCrossRef Rykx A, Kimpe LD, Mikhalap S, Vantus T, Seufferlein T, Vandenheede JR, Lint JV (2003) Protein kinase D: a family affair. FEBS Lett 546:81–86PubMedCrossRef
12.
go back to reference Eiseler T, Döppler H, Yan IK, Goodison S, Storz P (2009) Protein kinase D1 regulates matrix metalloproteinase expression and inhibits breast cancer cell invasion. Breast Cancer Res 11(1):R13PubMedCrossRef Eiseler T, Döppler H, Yan IK, Goodison S, Storz P (2009) Protein kinase D1 regulates matrix metalloproteinase expression and inhibits breast cancer cell invasion. Breast Cancer Res 11(1):R13PubMedCrossRef
13.
go back to reference Papazyan R, Rozengurt E, Rey O (2006) The C-terminal tail of protein kinase D2 and protein kinase D3 regulates their intracellular distribution. Biochem Biophys Res Commun 342(3):685–689PubMedCrossRef Papazyan R, Rozengurt E, Rey O (2006) The C-terminal tail of protein kinase D2 and protein kinase D3 regulates their intracellular distribution. Biochem Biophys Res Commun 342(3):685–689PubMedCrossRef
14.
go back to reference Lu G, Chen J, Espinoza LA, Garfield S, Toshiyuki S, Akiko H, Huppler A, Wang QJ (2007) Protein kinase D 3 is localized in vesicular structures and interacts with vesicle-associated membrane protein 2. Cell Signal 19(4):867–879PubMedCrossRef Lu G, Chen J, Espinoza LA, Garfield S, Toshiyuki S, Akiko H, Huppler A, Wang QJ (2007) Protein kinase D 3 is localized in vesicular structures and interacts with vesicle-associated membrane protein 2. Cell Signal 19(4):867–879PubMedCrossRef
15.
go back to reference Marklund U, Lightfoot K, Cantrell D (2003) Intracellular location and cell context-dependent function of protein kinase D. Immunity 19(4):491–501PubMedCrossRef Marklund U, Lightfoot K, Cantrell D (2003) Intracellular location and cell context-dependent function of protein kinase D. Immunity 19(4):491–501PubMedCrossRef
16.
go back to reference Hassan S, Biswas MH, Zhang C, Du C, Balaji KC (2009) Heat shock protein 27 mediates repression of androgen receptor function by protein kinase D1 in prostate cancer cells. Oncogene 28(49):4386–4396PubMedCrossRef Hassan S, Biswas MH, Zhang C, Du C, Balaji KC (2009) Heat shock protein 27 mediates repression of androgen receptor function by protein kinase D1 in prostate cancer cells. Oncogene 28(49):4386–4396PubMedCrossRef
17.
go back to reference Matthews SA, Navarro MN, Sinclair LV, Emslie E, Feijoo-Carnero C, Cantrell DA (2010) Unique functions for protein kinase D1 and protein kinase D2 in mammalian cells. Biochem J 432(1):153–163PubMedCrossRef Matthews SA, Navarro MN, Sinclair LV, Emslie E, Feijoo-Carnero C, Cantrell DA (2010) Unique functions for protein kinase D1 and protein kinase D2 in mammalian cells. Biochem J 432(1):153–163PubMedCrossRef
18.
go back to reference American Cancer Society: Cancer Facts and Figures 2009. Atlanta, Ga: American Cancer Society, 2009. Also available online. Last accessed May 7, 2009 American Cancer Society: Cancer Facts and Figures 2009. Atlanta, Ga: American Cancer Society, 2009. Also available online. Last accessed May 7, 2009
19.
go back to reference Amadesi S, Grant AD, Cottrell GS, Vaksman N, Poole DP, Rozengurt E, Bunnett NW (2009) Protein kinase D isoforms are expressed in rat and mouse primary sensory neurons and are activated by agonists of protease-activated receptor 2. J Comp Neurol 516(2):141–156PubMedCrossRef Amadesi S, Grant AD, Cottrell GS, Vaksman N, Poole DP, Rozengurt E, Bunnett NW (2009) Protein kinase D isoforms are expressed in rat and mouse primary sensory neurons and are activated by agonists of protease-activated receptor 2. J Comp Neurol 516(2):141–156PubMedCrossRef
20.
go back to reference Chen LA, Li J, Silva SR, Jackson LN, Zhou Y, Watanabe H, Ives KL, Hellmich MR, Evers BM (2009) PKD3 is the predominant protein kinase D isoform in mouse exocrine pancreas and promotes hormone-induced amylase secretion. J Biol Chem 284(4):2459–2471PubMedCrossRef Chen LA, Li J, Silva SR, Jackson LN, Zhou Y, Watanabe H, Ives KL, Hellmich MR, Evers BM (2009) PKD3 is the predominant protein kinase D isoform in mouse exocrine pancreas and promotes hormone-induced amylase secretion. J Biol Chem 284(4):2459–2471PubMedCrossRef
21.
go back to reference Rey O, Yuan J, Rozengurt E (2003) Intracellular redistribution of protein kinase D2 in response to G-protein-coupled receptor agonists. Biochem Biophys Res Commun 302:817–824PubMedCrossRef Rey O, Yuan J, Rozengurt E (2003) Intracellular redistribution of protein kinase D2 in response to G-protein-coupled receptor agonists. Biochem Biophys Res Commun 302:817–824PubMedCrossRef
22.
go back to reference Rey O, Yuan J, Young SH, Rozengurt E (2003) Protein kinase C nu/protein kinase D3 nuclear localization, catalytic activation, and intracellular redistribution in response to G protein-coupled receptor agonists. J Biol Chem 278:23773–23785PubMedCrossRef Rey O, Yuan J, Young SH, Rozengurt E (2003) Protein kinase C nu/protein kinase D3 nuclear localization, catalytic activation, and intracellular redistribution in response to G protein-coupled receptor agonists. J Biol Chem 278:23773–23785PubMedCrossRef
23.
go back to reference Yuan J, Rozengurt E (2008) PKD, PKD2, and p38 MAPK mediate Hsp27 serine-82 phosphorylation induced by neurotensin in pancreatic cancer PANC-1 cells. J Cell Biochem 103(2):648–662PubMedCrossRef Yuan J, Rozengurt E (2008) PKD, PKD2, and p38 MAPK mediate Hsp27 serine-82 phosphorylation induced by neurotensin in pancreatic cancer PANC-1 cells. J Cell Biochem 103(2):648–662PubMedCrossRef
24.
go back to reference Seufferlein T (2002) Novel protein kinases in pancreatic cell growth and cancer. Int J Gastrointest Cancer 31(1–3):15–21PubMedCrossRef Seufferlein T (2002) Novel protein kinases in pancreatic cell growth and cancer. Int J Gastrointest Cancer 31(1–3):15–21PubMedCrossRef
25.
go back to reference Guha S, Rey O, Rozengurt E (2002) Neurotension induces protein kinase C-dependent protein kinase D activation and DNA synthesis in human pancreatic carcinoma cell line PANC-1. Cancer Res 62:1632–1640PubMed Guha S, Rey O, Rozengurt E (2002) Neurotension induces protein kinase C-dependent protein kinase D activation and DNA synthesis in human pancreatic carcinoma cell line PANC-1. Cancer Res 62:1632–1640PubMed
26.
go back to reference Du C, Jaggi M, Zhang C, Balaji KC (2009) Protein kinase D1-mediated phosphorylation and subcellular localization of beta-catenin. Cancer Res 69(3):1117–1124PubMedCrossRef Du C, Jaggi M, Zhang C, Balaji KC (2009) Protein kinase D1-mediated phosphorylation and subcellular localization of beta-catenin. Cancer Res 69(3):1117–1124PubMedCrossRef
27.
go back to reference Mak P, Jaggi M, Syed V, Chauhan SC, Hassan S, Biswas H, Balaji KC (2008) Protein kinase D1 (PKD1) influences androgen receptor (AR) function in prostate cancer cells. Biochem Biophys Res Commun 373(4):618–623PubMedCrossRef Mak P, Jaggi M, Syed V, Chauhan SC, Hassan S, Biswas H, Balaji KC (2008) Protein kinase D1 (PKD1) influences androgen receptor (AR) function in prostate cancer cells. Biochem Biophys Res Commun 373(4):618–623PubMedCrossRef
28.
go back to reference Chen J, Deng F, Singh SV, Wang QJ (2008) Protein kinase D3 (PKD3) contributes to prostate cancer cell growth and survival through a PKCepsilon/PKD3 pathway downstream of Akt and ERK 1/2. Cancer Res 68(10):3844–3853PubMedCrossRef Chen J, Deng F, Singh SV, Wang QJ (2008) Protein kinase D3 (PKD3) contributes to prostate cancer cell growth and survival through a PKCepsilon/PKD3 pathway downstream of Akt and ERK 1/2. Cancer Res 68(10):3844–3853PubMedCrossRef
29.
go back to reference Sturany S, Lint JV, Gilchrist A, Vandenheede JR, Adler G, Seufferlein T (2002) Mechanism of activation of protein kinase D2(PKD2) by the CCKB/gastrin receptor. J Biol Chem 277:29431–29436PubMedCrossRef Sturany S, Lint JV, Gilchrist A, Vandenheede JR, Adler G, Seufferlein T (2002) Mechanism of activation of protein kinase D2(PKD2) by the CCKB/gastrin receptor. J Biol Chem 277:29431–29436PubMedCrossRef
30.
go back to reference Jaggi M, Rao PS, Smith DJ, Wheelock MJ, Johnson KR, Hemstreet GP, Balaji KC (2005) E-cadherin phosphorylation by protein kinase D1/protein kinase C{mu} is associated with altered cellular aggregation and motility in prostate cancer. Cancer Res 65:483–492PubMed Jaggi M, Rao PS, Smith DJ, Wheelock MJ, Johnson KR, Hemstreet GP, Balaji KC (2005) E-cadherin phosphorylation by protein kinase D1/protein kinase C{mu} is associated with altered cellular aggregation and motility in prostate cancer. Cancer Res 65:483–492PubMed
31.
go back to reference Harikumar KB, Kunnumakkara AB, Ochi N, Tong Z, Deorukhkar A, Sung B, Kelland L, Jamieson S, Sutherland R, Raynham T, Charles M, Bagherzadeh A, Foxton C, Boakes A, Farooq M, Maru D, Diagaradjane P, Matsuo Y, Sinnett-Smith J, Gelovani J, Krishnan S, Aggarwal BB, Rozengurt E, Ireson CR, Guha S (2010) A novel small-molecule inhibitor of protein kinase D blocks pancreatic cancer growth in vitro and in vivo. Mol Cancer Ther 9:1136–1145PubMedCrossRef Harikumar KB, Kunnumakkara AB, Ochi N, Tong Z, Deorukhkar A, Sung B, Kelland L, Jamieson S, Sutherland R, Raynham T, Charles M, Bagherzadeh A, Foxton C, Boakes A, Farooq M, Maru D, Diagaradjane P, Matsuo Y, Sinnett-Smith J, Gelovani J, Krishnan S, Aggarwal BB, Rozengurt E, Ireson CR, Guha S (2010) A novel small-molecule inhibitor of protein kinase D blocks pancreatic cancer growth in vitro and in vivo. Mol Cancer Ther 9:1136–1145PubMedCrossRef
32.
go back to reference Stewart JR, Christman KL, O’Brian CA (2000) Effects of resveratrol on the autophosphorylation of phorbol ester-responsive protein kinases: inhibition of protein kinase D but not protein kinase C isozyme autophosphorylation. Biochem Pharmacol 60(9):1355–1359PubMedCrossRef Stewart JR, Christman KL, O’Brian CA (2000) Effects of resveratrol on the autophosphorylation of phorbol ester-responsive protein kinases: inhibition of protein kinase D but not protein kinase C isozyme autophosphorylation. Biochem Pharmacol 60(9):1355–1359PubMedCrossRef
33.
go back to reference Sharlow ER, Giridhar KV, LaValle CR, Chen J, Leimgruber S, Barrett R, Bravo-Altamirano K, Wipf P, Lazo JS, Wang QJ (2008) Potent and selective disruption of protein kinase D functionality by a benzoxoloazepinolone. J Biol Chem 283(48):33516–33526PubMedCrossRef Sharlow ER, Giridhar KV, LaValle CR, Chen J, Leimgruber S, Barrett R, Bravo-Altamirano K, Wipf P, Lazo JS, Wang QJ (2008) Potent and selective disruption of protein kinase D functionality by a benzoxoloazepinolone. J Biol Chem 283(48):33516–33526PubMedCrossRef
34.
go back to reference LaValle CR, Bravo-Altamirano K, Giridhar KV, Chen J, Sharlow E, Lazo JS, Wipf P, Wang QJ (2010) Novel protein kinase D inhibitors cause potent arrest in prostate cancer cell growth and motility. BMC Chem Biol 10:5PubMedCrossRef LaValle CR, Bravo-Altamirano K, Giridhar KV, Chen J, Sharlow E, Lazo JS, Wipf P, Wang QJ (2010) Novel protein kinase D inhibitors cause potent arrest in prostate cancer cell growth and motility. BMC Chem Biol 10:5PubMedCrossRef
35.
go back to reference Bravo-Altamirano K, George KM, Frantz MC, Lavalle CR, Tandon M, Leimgruber S, Sharlow ER, Lazo JS, Wang QJ, Wipf P (2011) Synthesis and structure-activity relationships of benzothienothiazepinone inhibitors of protein kinase D. ACS Med Chem Lett 2(2):154–159PubMedCrossRef Bravo-Altamirano K, George KM, Frantz MC, Lavalle CR, Tandon M, Leimgruber S, Sharlow ER, Lazo JS, Wang QJ, Wipf P (2011) Synthesis and structure-activity relationships of benzothienothiazepinone inhibitors of protein kinase D. ACS Med Chem Lett 2(2):154–159PubMedCrossRef
36.
go back to reference George KM, Frantz M-CL, Bravo-Altamirano K, Lavalle CR, Tandon M, Leimgruber S, Sharlow ER, Lazo JS, Wang QJ, Wipf P (2011) Design, synthesis, and biological evaluation of PKD inhibitors. Pharmaceutics 3(2):186–228PubMedCrossRef George KM, Frantz M-CL, Bravo-Altamirano K, Lavalle CR, Tandon M, Leimgruber S, Sharlow ER, Lazo JS, Wang QJ, Wipf P (2011) Design, synthesis, and biological evaluation of PKD inhibitors. Pharmaceutics 3(2):186–228PubMedCrossRef
37.
go back to reference D’Argenio DZ, Schumitzky A. ADAPT, Biomedical Simulation Resource, USC. bmsr.usc.edu/Software/ADAPT/ADAPT.html D’Argenio DZ, Schumitzky A. ADAPT, Biomedical Simulation Resource, USC. bmsr.usc.edu/Software/ADAPT/ADAPT.html
38.
go back to reference Yeh KC, Kwan KC (1978) A comparison of numerical integrating algorithms by trapezoidal, Lagrange, and spline approximation. J Pharmacokinet Biopharm 6:79–98PubMedCrossRef Yeh KC, Kwan KC (1978) A comparison of numerical integrating algorithms by trapezoidal, Lagrange, and spline approximation. J Pharmacokinet Biopharm 6:79–98PubMedCrossRef
39.
go back to reference Rocci ML, Jusko WJ (1983) LAGRAN program for area and moments in pharmacokinetic analysis. Comput Programs Biomed 16:203–216PubMedCrossRef Rocci ML, Jusko WJ (1983) LAGRAN program for area and moments in pharmacokinetic analysis. Comput Programs Biomed 16:203–216PubMedCrossRef
40.
go back to reference Brown RP, Delp MD, Lindstedt SL, Rhomberg LR, Beliles RP (1997) Physiological parameter values for physiologically based pharmacokinetic models. Toxicol Ind Health 13:407–484PubMed Brown RP, Delp MD, Lindstedt SL, Rhomberg LR, Beliles RP (1997) Physiological parameter values for physiologically based pharmacokinetic models. Toxicol Ind Health 13:407–484PubMed
41.
go back to reference LaValle CR, George KM, Sharlow ER, Lazo JS, Wipf P, Wang QJ (2010) Protein kinase D as a potential new target for cancer therapy. Biochim Biophys Acta 1806:183–192PubMed LaValle CR, George KM, Sharlow ER, Lazo JS, Wipf P, Wang QJ (2010) Protein kinase D as a potential new target for cancer therapy. Biochim Biophys Acta 1806:183–192PubMed
42.
go back to reference Ochi N, Tanasanvimon S, Matsuo Y, Tong Z, Sung B, Aggarwal BB, Sinnett-Smith J, Rozengurt E, Guha S (2011) Protein kinase D1 promotes anchorage-independent growth, invasion, and angiogenesis by human pancreatic cancer cells. J Cell Physiol 226:1074–1081PubMedCrossRef Ochi N, Tanasanvimon S, Matsuo Y, Tong Z, Sung B, Aggarwal BB, Sinnett-Smith J, Rozengurt E, Guha S (2011) Protein kinase D1 promotes anchorage-independent growth, invasion, and angiogenesis by human pancreatic cancer cells. J Cell Physiol 226:1074–1081PubMedCrossRef
43.
go back to reference Ristich VL, Bowman PH, Dodd ME, Bollag WB (2006) Protein kinase D distribution in normal human epidermis, basal cell carcinoma and psoriasis. Br J Dermatol 154:586–593PubMedCrossRef Ristich VL, Bowman PH, Dodd ME, Bollag WB (2006) Protein kinase D distribution in normal human epidermis, basal cell carcinoma and psoriasis. Br J Dermatol 154:586–593PubMedCrossRef
44.
go back to reference Kisfalvi K, Hurd C, Guha S, Rozengurt E (2010) Induced overexpression of protein kinase D1 stimulates mitogenic signaling in human pancreatic carcinoma PANC-1 cells. J Cell Physiol 223:309–316PubMed Kisfalvi K, Hurd C, Guha S, Rozengurt E (2010) Induced overexpression of protein kinase D1 stimulates mitogenic signaling in human pancreatic carcinoma PANC-1 cells. J Cell Physiol 223:309–316PubMed
45.
go back to reference Chen J, Giridhar KV, Zhang L, Xu S, Wang QJ (2011) A protein kinase C/protein kinase D pathway protects LNCaP prostate cancer cells from phorbol ester-induced apoptosis by promoting ERK1/2 and NF-{kappa}B activities. Carcinogenesis 32:1198–1206PubMedCrossRef Chen J, Giridhar KV, Zhang L, Xu S, Wang QJ (2011) A protein kinase C/protein kinase D pathway protects LNCaP prostate cancer cells from phorbol ester-induced apoptosis by promoting ERK1/2 and NF-{kappa}B activities. Carcinogenesis 32:1198–1206PubMedCrossRef
Metadata
Title
In vitro cytotoxicity, pharmacokinetics, tissue distribution, and metabolism of small-molecule protein kinase D inhibitors, kb-NB142-70 and kb-NB165-09, in mice bearing human cancer xenografts
Authors
Jianxia Guo
Dana M. Clausen
Jan H. Beumer
Robert A. Parise
Merrill J. Egorin
Karla Bravo-Altamirano
Peter Wipf
Elizabeth R. Sharlow
Qiming Jane Wang
Julie L. Eiseman
Publication date
01-02-2013
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 2/2013
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-012-2010-z

Other articles of this Issue 2/2013

Cancer Chemotherapy and Pharmacology 2/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine