T-18, a stemonamide synthetic intermediate inhibits Pim kinase activity and induces cell apoptosis, acting as a potent anticancer drug

  • Authors:
    • Zhen Wang
    • Xing-Min Li
    • Kun Shang
    • Peng Zhang
    • Chao-Fu Wang
    • Yu-Hu Xin
    • Lu Zhou
    • Ying-Yi Li
  • View Affiliations

  • Published online on: January 11, 2013     https://doi.org/10.3892/or.2013.2233
  • Pages: 1245-1251
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pim-3 kinase has been shown to be aberrantly expressed in premalignant and malignant lesions of endoderm-derived organs such as the liver, pancreas, colon and stomach. Pim-3 kinase inactivates the Bad protein, a proapoptotic molecule, and improves the expression of Bcl-xL, an antiapoptotic molecule, to promote cell proliferation. Thus, blocking Pim-3 kinase activity may be a new strategy for the treatment of pancreatic cancer. In this study, we screened low molecular compounds and observed that the stemonamide synthetic intermediate, T-18, potently inhibited Pim kinase activity. Moreover, T-18 inhibited the proliferation of human pancreatic, as well as that of hepatocellular and colon cancer cells in vitro. It also induced the apoptosis of human pancreatic carcinoma cells in vitro by decreasing the levels of phospho-Ser112-Bad; the levels of Pim-3 kinase and total Bad protein were not altered. Furthermore, T-18 inhibited the growth of human pancreatic cancer cells in nude mice without apparent adverse effects when the tumor was palpable. These observations indicate that stemonamide synthetic intermediates may be novel drugs for the treatment of gastrointestinal cancers, particularly pancreatic cancer.

Introduction

Pancreatic cancer is one of the common malignant tumors in Western countries with the highest incidence rate, and is also one of the cancers with the highest mortality rate (1). The characteristics of pancreatic cancer include high-grade malignancy, specific biological behavior, non-specific symptoms and tendency for early metastasis, insensitivity to chemotherapy and radiotherapy and a low rate of surgical resection (2). Compared with other cancers, the survival rate is so low that the incidence rate is basically equal to the mortality rate (3). Thus, molecular targeted therapy may be the most effective treatment for pancreatic cancer.

The proto-oncogene Pim family exhibits serine/threonine kinase activity. In humans, the Pim family includes 3 members: Pim-1, Pim-2 and Pim-3 (4). Crystal structures of Pim family kinases have revealed that the ATP-binding sites of Pim family kinases share high structural homology (57). The overexpression of Pim-1 and Pim-2 has been found in a variety of human hematopoietic malignancies (811), such as leukemia and lymphoma and some solid tumors, such as prostate cancer (1214). Pim-3 has mainly been found in solid tumors, particularly endoderm-derived organs such as the liver, stomach, pancreas and colon (1517) and Pim kinases have been demonstrated to inhibit apoptosis by phosphorylating the pro-apoptotic molecule, Bad at Ser112. Thus, Pim-3 may be an effective target for the treatment of cancer of endoderm-derived organs, particularly the pancreas (1821). Several Pim kinase inhibitors have been reported (22,23), but only a few of them are effective against all Pim family kinases (2427). This prompted us to develop a specific inhibitor for each Pim kinase.

In this study, we investigated whether T-18, a stemonamide synthetic intermediate, may be used as a Pim-3 kinase inhibitor. T-18 reduced the levels of phosphorylated Bad at Ser112, resulting in the decreased growth of pancreatic carcinoma cells in vitro and the induction of apoptosis. Furthermore, T-18 also inhibited the growth of human pancreatic cancer cells in nude mice even when the forming tumor was observed. These results indicate that T-18 may be a lead compound that can be modified to exert a more potent inhibitory effect on Pim-3 kinase, and may thus be effectively used in the treatment of cancers with an aberrant Pim-3 overexpression.

Materials and methods

Cell culture and antibodies

The human pancreatic cancer cell lines, PCI35 (28), PCI55 (28), PCI66 (28), PANC-1 (29) and MIA PaCa-2 (30), and the human colon cancer cell line, SW480 (31), were cultured in RPMI-1640 medium, while another human pancreatic cancer cell line, L3.6pl (32), and the human colon cancer cell line, SW48 (31), were maintained in minimum essential medium. The human colon cancer cell lines, HCT29 (33) and HCT116 (34), were cultured in McCoy’s 5A medium and the human hepatocarcinoma cell lines, HepG2, Hep3B (35) and HuH7 (36), were maintained in Dulbecco’s modified Eagle’s medium. All media were supplemented with 10% FBS, 50 U/ml of penicillin and 50 U/ml of streptomycin. The following antibodies were used: mouse anti-human Bad (Santa Cruz Biotechnology, Santa Cruz, CA, USA), rabbit anti-phosphorylated (p-) Ser112-Bad (Cell Signaling Technology, Beverly, MA, USA) and anti-β-actin antibodies (Sigma-Aldrich). The rabbit anti-human Pim-3 antibodies were prepared as previously described (16).

Preparation of stemonamide synthetic intermediates

The stemonamide synthetic intermediates were synthesized as previously described (37). T-10 and T-18 were dissolved in DMSO (Sigma-Aldrich) at the concentration of 100 mM as stocking solution and the solutions were stable within 1 week. T-18 was dissolved in DMSO at the concentration of 100 mg/ml and the solution was diluted with olive oil to reduce the final DMSO concentration to 4% v/v for the animal experiment.

In vitro Pim kinase assay and Akt kinase assay

Based on the ability of Pim kinases to phosphorylate Bad at Ser112, in vitro kinase assay for Pim-1, Pim-2 and Pim-3 was established and conducted as previously described (38,39). The IC50 values were calculated using logistic regression.

Akt kinase activities were determined based on their capacity to phosphorylate Bad at Ser136 as described previously (38,39). Either recombinant Akt-1 (10 ng) (Cell Signaling Technology) or Akt-2 (10 ng) (Assay Designs, Ann Arbor, MI, USA) was used as the source of the enzymes. Akt-1/2 kinase inhibitor (Sigma-Aldrich) was used as the positive control inhibitor.

Cell viability assay

Cells were inoculated into 96-multiwell plate at 3,000 cells/100 μl per well and incubated at 37°C for 18 h. The chemicals were then added to each well at the indicated concentrations and the cells were incubated for the indicated periods of time. After removing the supernatants, the medium containing CCK-8 (Dojin Chemicals, Kumamoto, Japan) was added to each well followed by a 2-h incubation. Subsequently, the optical density was measured at 450 nm. The number of viable cells at day 0 was regarded as the control to determine the ratios of viable cell numbers. Logistic regression was used to calculate the IC50 values.

Cell apoptosis assay

The cell apoptosis assay was operated according to the instruction manual of human Annexin V-FITC apoptosis kit (Bender MedSystems GmbH, Vienna, Austria). The cells were harvested after exposure to the chemicals and 20,000 stained cells were acquired for the analysis on the FACSCalibur system (BD Biosciences).

Western blot analysis

The L3.6pl or MIA PaCa-2 cells (2×106 cells) were seeded in 100-mm culture plates. After an 18-h incubation, the chemicals were added to the plate at the final concentration of 10 μM. Subsequently, 60 μl CellLytic™ Cell Lysis Reagent (Sigma-Aldrich) with Complete Proteinase Inhibitor Cocktail (Roche Diagnostics AG, Rotkreutz, Switzerland) was added followed by ultrasound sonication after treatment with the chemicals. Following centrifugation, the supernatants were collected for western blot analysis using the antibodies mentioned above.

Animal experiments

L3.6pl cells were suspended in HBSS at the concentration of 5×106 cells/ml and 100 μl cell suspensions were inoculated subcutaneously into the back of BALB/c nu/nu mice (SLC, Shizuoka, Japan). Twelve days after tumor injection, the chemical (20 mg/kg) or olive oil vehicle was injected intraperitoneally once a day for 5 days as one course. The course was repeated 3 times with an interval of 2 days. Tumor sizes were measured every 3–4 days with a caliper and tumor volumes were determined using the following the formula: tumor volume (mm3) = (the longest diameter) (mm) × (the shortest diameter)2 (mm)/2. On day 32 after the tumor cell injection, blood was taken simultaneously at tumor removal. Serum levels of alanine aminotransferase (ALT) were measured using the Fuji Drichem 5500V analyzer (Fujifilm Medical Systems, Tokyo, Japan).

Statistical analysis

Data are expressed as the means ± SD unless otherwise indicated. One-way ANOVA and the Tukey-Kramer test were employed to analyze the differences between groups and a value of P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of T-18 on Pim kinase activity

We previously reported that some synthetic intermediates of stemonamide (T-2) can inhibit the activity of Pim-3 kinase in vitro(38). However, the IC50 of T-2 was at a micromolar concentration, which prompted us to find additional chemicals which can exert a more profound inhibitory effect at low doses. T-18 was synthesized based on T-2. We found that T-18, one of the stemonamide synthetic intermediates (Fig. 1A), inhibited Pim kinase activity (Fig. 1B). Compared with T-10 (38), T-18 inhibited Pim kinase activity to a lower extent. Moreover, T-18 inhibited Pim-1 and Pim-2 kinase activity to a similar extent as Pim-3 (Table I). We then examined the effects of T-18 on the human Akt/protein kinase B (PKB) protein kinase, another serine/threonine kinase which can phosphorylate a similar set of proteins, such as Bad (40,41). Akt-1/2, a specific inhibitor of Akt-1 and Akt-2, was used in our study as the positive control for a comparison against T-18 (Table I). Akt-1/2 efficiently inhibited Akt-1 and Akt-2 activity, while T-18 did not exert any effect on the Akt protein kinase. Thus, T-18 can potently and selectively inhibit Pim-3 kinase activity, but not that of Akt.

Table I

Inhibitory activity of T-18 on Pim and Akt kinases.

Table I

Inhibitory activity of T-18 on Pim and Akt kinases.

IC50 (μmol/l)

Chemical namePim-3Pim-1Pim-2Akt-1Akt-2
T-10>2,000>2,000>2,000>2,000>2,000
T-185711827>2,000>2,000
Akt-1/2NDNDND31.672.5

[i] Inhibitory effects were investigated up to T-18 concentrations of 320 μmol/l and IC50 values were calculated. ND, not determined.

Effects of T-18 on cell proliferation in vitro

Two human pancreatic cancer cell lines, MIA PaCa-2 and L3.6pl, were recruited to examine the effects of T-18 on cell proliferation in vitro (Fig. 2). The results showed that T-18 effectively inhibited cell proliferation in a dose-dependent manner. However, T-10 had little effect on the cell proliferation of these cancer cell lines, and had little influence on Pim-3 kinase activity. T-18 was added to the medium of the other human pancreatic cancer cell lines (PCI66, PCI35, PCI55 and PANC-1), hepatocellular carcinoma cell lines (HuH7, HepG2 and Hep3B) and colon cancer cell lines (SW480, SW48, HT29 and HCT116) to determine the effect of T-18 on the proliferation of these cells (Table II). Almost all the cancer cell lines were sensitive to T-18. These observations suggest that T-18 inhibits Pim-3 kinase activity and has an effect on the proliferation of these cells.

Table II

Effects of T-18 on the proliferation of human cancer cells in vitro.

Table II

Effects of T-18 on the proliferation of human cancer cells in vitro.

Cell typeCell line nameProliferation IC50 (μmol/l) T-18
PancreasL3.6pl3.46
MIA PaCa-22.09
PCI662.24
PCI352.25
PCI553.31
PANC-12.96
LiverHuH75.25
HepG221.4
Hep3B-
ColonSW4801.78
SW482.75
HT294.32
HCT1161.79

[i] Data represent the means ± SD calculated from 3 independent experiments.

Effects of T-18 on cell apoptosis

In our previous studies, we demonstrated that a Pim-3 shRNA-mediated reduction in Pim-3 protein expression decreased the levels of phospho-Ser112-Bad and increased the number of apoptotic cells in human pancreatic and colon cancer cell lines (16,17). In this study, we investigated whether T-18 can affect the phosphorylation state of Bad in the human pancreatic cancer cell lines, MIA PaCa-2 and L3.6pl. Consistent with our previous results, the levels of phospho-Ser112-Bad decreased in the cell lines following treatment with T-18. However, the levels of Pim-3 kinase protein or total Bad protein were not altered with the treatment time (Fig. 3). Moreover, early apoptotic and late apoptotic cells were enhanced in both cell lines (Fig. 4) following treatment with T-18 for 24 and 48 h. These observations indicate that T-18 prevents the phosphorylation of Bad, eventually leading to apoptosis in human pancreatic cancer cells.

Effects of T-18 on tumor growth in vivo

Finally, we investigated the effects of T-18 on tumor growth in vivo. We subcutaneously injected L3.6pl pancreatic cancer cells into nude mice as described above and T-18 treatment commenced on day 12 after the tumor injection, when the tumor was palpable (Fig. 5A). The tumors of the mice in the vehicle-treated group grew more rapidly than those of the mice in the T-18-treated group (Fig. 5B). During the course of the experiment, all mice tolerated T-18 well, as evidenced by the fact that no body weight loss was observed (Fig. 5C) and the changes in serum ALT levels (Fig. 5D). These results suggest that T-18 suppresses tumor growth in vivo without any significant side-effects.

Discussion

Pancreatic cancer is a malignant tumor of the pancreas, and poses a threat to human health. The majority of patients are diagnosed at an advanced stage, thus surgical treatment is not valid and the only treatment options are radiotherapy or chemotherapy, with the survival time being less than a year (42,43). Thus, molecular targeted therapy is considered to be a very promising treatment. These targeted molecules are not expressed in normal tissues but are aberrantly expressed in tumor tissues and play a key role in regulating tumor cell proliferation. The Pim-3 kinase is aberrantly expressed in premalignant and malignant lesions of endoderm-derived organs, such as the liver, pancreas, colon and stomach (16,17,44,45) but not in normal tissues. The Pim-3 kinase inactivates the Bad protein by phosphorylating Bad at Ser112 to promote cell proliferation (25), providing the characteristics of a targeted molecule. Thus, blocking Pim-3 kinase activity may be a new strategy for the treatment of pancreatic cancer.

The crystal structure of Pim-1 and Pim-2 reveals that there is a special hinge region that can connect the 2 lobes of the protein kinase domain, which presents a unique way for ATP to bind with Pim kinases instead of other protein kinases (5,46,47). Thus, it may be possible to develop an inhibitor that selectively targets Pim kinases, not other seine/threonine kinases (48). Existing small molecule inhibitors against Pim kinases that have been reported include flavonol quercetargetin (7), imidazole[1,2-b]pyridazines (26,49), bezylindene-thiazolidine-2,4-dione (5052), 3,5-disubstituted indole derivatives (53), pyrazolo[3,4-g]quinoxaline derivatives (54) and N-10 substituted pyrrolo[2,3-a]carbazole derivatives (5557). These molecules have been certified to inhibit the proliferation of human cancer cells in vitro and/or in vivo. However, most of the compounds are multi-Pim kinase inhibitors and Pim-3 has received the least attention among the Pim family kinases. Thus, a novel and safe Pim-3 targeting inhibitor is required. We have previously demonstrated that other stemonamide synthetic intermediates (T-2, T-5) can inhibit Pim kinases activity and suppress cancer cell proliferation in vitro, as well as tumor growth in vivo(38). However, the IC50 of T-2 was at a micromolar concentration. In this study, we investigated another low molecular compound (T-18) that shares homoplastic structure with T-2 and may exert a more effective anticancer effect.

To recognize a Pim-3 inhibitor, a series of low molecule chemicals were screened and a stemonamide synthetic intermediate, T-18, was observed, which was similar to T-2 which was investigated in our previous study (38). In the current study, T-18 inhibited the activity of Pim-3, Pim-1 and Pim-2 but not that of Akt-1 or Akt-2 kinase, showing the specific inhibitory effect on Pim family kinases and not on Akt kinases. Moreover, T-18 suppressed the proliferation of human pancreatic carcinoma cells in vitro, as well as the in vivo tumor growth of human pancreatic cancer cells injected into nude mice, without any severe adverse effects. Furthermore, T-18 prevented Bad phosphorylation at Ser112, while increasing the number of apoptotic pancreatic cancer cells; however, the levels of Pim-3 protein and total Bad protein were not altered. Since Pim-1 and Pim-2 protein can phosphorylate Bad at Ser112, which were not found in human pancreatic cancer cells (unpublished data), Pim-3 kinase was regarded as the target of T-18. T-18 inhibited Pim-3 activity, inactivating Bad and finally inducing apoptosis.

As we have previously demonstrated, T-2 (38), similar to T-18, can inhibit Pim-3 activity and reduce the growth of pancreatic cancer cells in vitro and in vivo. T-2 also inhibited in vivo tumor growth effectively. However, as shown in this study, T-18 inhibited the proliferation of human cancer cells in vitro with a lower IC50 value. Perhaps T-18 is an ideal compound that can be modified to exert a more potent inhibitory effect without any severe side-effects.

Another serine/threonine kinase, namely Akt, can phosphorylate similar sets of substrates, such as Bad, as Pim kinases, thus initiating the proliferation of cancer cells (41). Akt is aberrantly activated in various types of tumors and Akt inhibitors have been extensively investigated (40). Although the Akt inhibitor, GSK690693, has exhibited potent antitumor activity in pre-clinical animal experiments (41), the genetic disruption of each Akt kinase gene results in severe phenotypic changes, such as neonatal mortality, severe growth retardation and reduced brain size (5860) and Akt-2 inhibition induces severe hyperglycemia (41). This is a serious impediment for the clinical use of Akt inhibitors in anticancer treatment. The Pim-3 kinase is aberrantly expressed in malignant lesions but not in endoderm-derived normal tissues, such as the liver, pancreas, colon and stomach (16,17,44,45) and promotes tumor development. Moreover, Pim-3 gene deficiency does not induce apparent phenotypic changes, suggesting that Pim-3 may be physiologically dispensable (61). Pim kinases are not localized downstream of the insulin receptor signaling pathway but the Akt kinase is. The inhibition of Pim kinases has few effects on normal metabolism. Thus, Pim kinases may be more effective targets compared to Akt in the molecular targeted therapy of various types of cancer, particularly pancreatic cancer, exerting potent effects and few adverse effects. In this study, we showed that T-18 inhibited Pim kinases activity but failed to suppress Akt-1 and Akt-2 activity, suggesting that T-18 may be a safe drug for use in molecular targeted therapy. However, it may be difficult to discovery a specific inhibitor against either Pim-1 or Pim-3 kinase due to their extraordinarily similar peptide substrate identity (46). It would be of interest to determine whether a specific inhibitor against anyone of the Pim family kinases will prove to be advantageous over existing Pim kinase inhibitors.

Acknowledgements

The authors gratefully acknowledge grant support from The National Science Foundation of China (NSFC) (30973476), the Shanghai Pujiang Program (KW201028464), Fudan University ‘985 Project’ Phase III Cancer Research Projects II (985III-YFX0102), ‘Start Up’ Project of the Shanghai Cancer Center (YJRC0901), and Shanghai Committee of Science and Technology (12DZ2260100). The authors thank Professor Hiroyuki Ishibashi of School of Pharmaceutical Sciences, Kanazawa University, for providing T-18 and T-10.

References

1 

Aune D, Vieira AR, Chan DS, et al: Height and pancreatic cancer risk: a systematic review and meta-analysis of cohort studies. Cancer Causes Control. 23:1213–1222. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Li D, Xie K, Wolff R and Abbruzzese JL: Pancreatic cancer. Lancet. 363:1049–1057. 2004. View Article : Google Scholar

3 

Hochster HS, Haller DG, de Gramont A, et al: Consensus report of the international society of gastrointestinal oncology on therapeutic progress in advanced pancreatic cancer. Cancer. 107:676–685. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Mikkers H, Allen J, Knipscheer P, et al: High-throughput retroviral tagging to identify components of specific signaling pathways in cancer. Nat Genet. 32:153–159. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Qian KC, Wang L, Hickey ER, et al: Structural basis of constitutive activity and a unique nucleotide binding mode of human Pim-1 kinase. J Biol Chem. 280:6130–6137. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Kumar A, Mandiyan V, Suzuki Y, et al: Crystal structures of proto-oncogene kinase Pim1: a target of aberrant somatic hypermutations in diffuse large cell lymphoma. J Mol Biol. 348:183–193. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Holder S, Zemskova M, Zhang C, et al: Characterization of a potent and selective small-molecule inhibitor of the PIM1 kinase. Mol Cancer Ther. 6:163–172. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Alvarado Y, Giles FJ and Swords RT: The PIM kinases in hematological cancers. Expert Rev Hematol. 5:81–96. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Kim KT, Baird K, Ahn JY, et al: Pim-1 is up-regulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival. Blood. 105:1759–1767. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Pasqualucci L, Neumeister P, Goossens T, et al: Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature. 412:341–346. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Gaidano G, Pasqualucci L, Capello D, et al: Aberrant somatic hypermutation in multiple subtypes of AIDS-associated non-Hodgkin lymphoma. Blood. 102:1833–1841. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Dhanasekaran SM, Barrette TR, Ghosh D, et al: Delineation of prognostic biomarkers in prostate cancer. Nature. 412:822–826. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Valdman A, Fang X, Pang ST, Ekman P and Egevad L: Pim-1 expression in prostatic intraepithelial neoplasia and human prostate cancer. Prostate. 60:367–371. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Xu Y, Zhang T, Tang H, et al: Overexpression of PIM-1 is a potential biomarker in prostate carcinoma. J Surg Oncol. 92:326–330. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Liu LM, Zhang JX, Wang XP, Guo HX, Deng H and Luo J: Pim-3 protects against hepatic failure in D-galactosamine (D-GalN)-sensitized rats. Eur J Clin Invest. 40:127–138. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Li YY, Popivanova BK, Nagai Y, Ishikura H, Fujii C and Mukaida N: Pim-3, a proto-oncogene with serine/threonine kinase activity, is aberrantly expressed in human pancreatic cancer and phosphorylates Bad to block Bad-mediated apoptosis in human pancreatic cancer cell lines. Cancer Res. 66:6741–6747. 2006. View Article : Google Scholar

17 

Popivanova BK, Li YY, Zheng H, et al: Proto-oncogene, Pim-3 with serine/threonine kinase activity, is aberrantly expressed in human colon cancer cells and can prevent Bad-mediated apoptosis. Cancer Sci. 98:321–328. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Lilly M, Sandholm J, Cooper JJ, Koskinen PJ and Kraft A: The PIM-1 serine kinase prolongs survival and inhibits apoptosis-related mitochondrial dysfunction in part through a bcl-2-dependent pathway. Oncogene. 18:4022–4031. 1999. View Article : Google Scholar

19 

Fox CJ, Hammerman PS, Cinalli RM, Master SR, Chodosh LA and Thompson CB: The serine/threonine kinase Pim-2 is a transcriptionally regulated apoptotic inhibitor. Genes Dev. 17:1841–1854. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Yan B, Zemskova M, Holder S, et al: The PIM-2 kinase phosphorylates BAD on serine 112 and reverses BAD-induced cell death. J Biol Chem. 278:45358–45367. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Macdonald A, Campbell DG, Toth R, McLauchlan H, Hastie CJ and Arthur JS: Pim kinases phosphorylate multiple sites on Bad and promote 14-3-3 binding and dissociation from Bcl-XL. BMC Cell Biol. 7:12006. View Article : Google Scholar : PubMed/NCBI

22 

Brault L, Gasser C, Bracher F, Huber K, Knapp S and Schwaller J: PIM serine/threonine kinases in the pathogenesis and therapy of hematologic malignancies and solid cancers. Haematologica. 95:1004–1015. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Anizon F, Shtil AA, Danilenko VN and Moreau P: Fighting tumor cell survival: advances in the design and evaluation of Pim inhibitors. Curr Med Chem. 17:4114–4133. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Akue-Gedu R, Rossignol E, Azzaro S, et al: Synthesis, kinase inhibitory potencies, and in vitro antiproliferative evaluation of new Pim kinase inhibitors. J Med Chem. 52:6369–6381. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Tao ZF, Hasvold LA, Leverson JD, et al: Discovery of 3H-benzo[4,5]thieno[3,2-d]pyrimidin-4-ones as potent, highly selective, and orally bioavailable inhibitors of the human protooncogene proviral insertion site in moloney murine leukemia virus (PIM) kinases. J Med Chem. 52:6621–6636. 2009.

26 

Chen LS, Redkar S, Bearss D, Wierda WG and Gandhi V: Pim kinase inhibitor, SGI-1776, induces apoptosis in chronic lymphocytic leukemia cells. Blood. 114:4150–4157. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Mumenthaler SM, Ng PY, Hodge A, et al: Pharmacologic inhibition of Pim kinases alters prostate cancer cell growth and resensitizes chemoresistant cells to taxanes. Mol Cancer Ther. 8:2882–2893. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Yano T, Ishikura H, Kato H, Ogawa Y, Kondo S and Yoshiki T: Vaccination effect of interleukin-6-producing pancreatic cancer cells in nude mice: a model of tumor prevention and treatment in immune-compromised patients. Jpn J Cancer Res. 92:83–87. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Lieber M, Mazzetta J, Nelson-Rees W, Kaplan M and Todaro G: Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrine pancreas. Int J Cancer. 15:741–747. 1975. View Article : Google Scholar : PubMed/NCBI

30 

Yunis AA, Arimura GK and Russin DJ: Human pancreatic carcinoma (MIA PaCa-2) in continuous culture: sensitivity to asparaginase. Int J Cancer. 19:128–135. 1977. View Article : Google Scholar : PubMed/NCBI

31 

Leibovitz A, Stinson JC, McCombs WB III, McCoy CE, Mazur KC and Mabry ND: Classification of human colorectal adenocarcinoma cell lines. Cancer Res. 36:4562–4569. 1976.PubMed/NCBI

32 

Bruns CJ, Harbison MT, Kuniyasu H, Eue I and Fidler IJ: In vivo selection and characterization of metastatic variants from human pancreatic adenocarcinoma by using orthotopic implantation in nude mice. Neoplasia. 1:50–62. 1999. View Article : Google Scholar

33 

Marshall CJ, Franks LM and Carbonell AW: Markers of neoplastic transformation in epithelial cell lines derived from human carcinomas. J Natl Cancer Inst. 58:1743–1751. 1977.PubMed/NCBI

34 

Brattain MG, Brattain DE, Fine WD, et al: Initiation and characterization of cultures of human colonic carcinoma with different biological characteristics utilizing feeder layers of confluent fibroblasts. Oncodev Biol Med. 2:355–366. 1981.

35 

Knowles BB, Howe CC and Aden DP: Human hepatocellular carcinoma cell lines secrete the major plasma proteins and hepatitis B surface antigen. Science. 209:497–499. 1980. View Article : Google Scholar : PubMed/NCBI

36 

Nakabayashi H, Taketa K, Miyano K, Yamane T and Sato J: Growth of human hepatoma cells lines with differentiated functions in chemically defined medium. Cancer Res. 42:3858–3863. 1982.PubMed/NCBI

37 

Taniguchi T, Tanabe G, Muraoka O and Ishibashi H: Total synthesis of (+/-)-stemonamide and (+/-)-isostemonamide using a radical cascade. Org Lett. 10:197–199. 2008.

38 

Li YY, Wang YY, Taniguchi T, et al: Identification of stemonamide synthetic intermediates as a novel potent anticancer drug with an apoptosis-inducing ability. Int J Cancer. 127:474–484. 2010.PubMed/NCBI

39 

Wang YY, Taniguchi T, Baba T, Li YY, Ishibashi H and Mukaida N: Identification of a phenanthrene derivative as a potent anticancer drug with Pim kinase inhibitory activity. Cancer Sci. 103:107–115. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Amaravadi R and Thompson CB: The survival kinases Akt and Pim as potential pharmacological targets. J Clin Invest. 115:2618–2624. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Rhodes N, Heerding DA, Duckett DR, et al: Characterization of an Akt kinase inhibitor with potent pharmacodynamic and antitumor activity. Cancer Res. 68:2366–2374. 2008. View Article : Google Scholar : PubMed/NCBI

42 

André T, Balosso J, Louvet C, et al: Adenocarcinoma of the pancreas. General characteristics. Presse Med. 27:533–536. 1998.(In French).

43 

Jian J, Hu ZF and Huang Y: Effect of ginsenoside Rg3 on Pim-3 and Bad proteins in human pancreatic cancer cell line PANC-1. Ai Zheng. 28:461–465. 2009.(In Chinese).

44 

Fujii C, Nakamoto Y, Lu P, et al: Aberrant expression of serine/threonine kinase Pim-3 in hepatocellular carcinoma development and its role in the proliferation of human hepatoma cell lines. Int J Cancer. 114:209–218. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Zheng HC, Tsuneyama K, Takahashi H, et al: Aberrant Pim-3 expression is involved in gastric adenoma-adenocarcinoma sequence and cancer progression. J Cancer Res Clin Oncol. 134:481–488. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Bullock AN, Debreczeni J, Amos AL, Knapp S and Turk BE: Structure and substrate specificity of the Pim-1 kinase. J Biol Chem. 280:41675–41682. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Bullock AN, Russo S, Amos A, et al: Crystal structure of the PIM2 kinase in complex with an organoruthenium inhibitor. PLoS One. 4:e71122009. View Article : Google Scholar : PubMed/NCBI

48 

Swords R, Kelly K, Carew J, et al: The Pim kinases: new targets for drug development. Curr Drug Targets. 12:2059–2066. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Pogacic V, Bullock AN, Fedorov O, et al: Structural analysis identifies imidazo[1,2-b]pyridazines as PIM kinase inhibitors with in vitro antileukemic activity. Cancer Res. 67:6916–6924. 2007.PubMed/NCBI

50 

Dakin LA, Block MH, Chen H, et al: Discovery of novel benzylidene-1,3-thiazolidine-2,4-diones as potent and selective inhibitors of the PIM-1, PIM-2, and PIM-3 protein kinases. Bioorg Med Chem Lett. 22:4599–4604. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Xia Z, Knaak C, Ma J, et al: Synthesis and evaluation of novel inhibitors of Pim-1 and Pim-2 protein kinases. J Med Chem. 52:74–86. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Lin YW, Beharry ZM, Hill EG, et al: A small molecule inhibitor of Pim protein kinases blocks the growth of precursor T-cell lymphoblastic leukemia/lymphoma. Blood. 115:824–833. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Nishiguchi GA, Atallah G, Bellamacina C, et al: Discovery of novel 3,5-disubstituted indole derivatives as potent inhibitors of Pim-1, Pim-2, and Pim-3 protein kinases. Bioorg Med Chem Lett. 21:6366–6369. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Gavara L, Saugues E, Alves G, Debiton E, Anizon F and Moreau P: Synthesis and biological activities of pyrazolo[3,4-g]quinoxaline derivatives. Eur J Med Chem. 45:5520–5526. 2010.

55 

Akue-Gedu R, Letribot B, Saugues E, Debiton E, Anizon F and Moreau P: Kinase inhibitory potencies and in vitro antiproliferative activities of N-10 substituted pyrrolo[2,3-a]carbazole derivatives. Bioorg Med Chem Lett. 22:3807–3809. 2012.PubMed/NCBI

56 

Akue-Gedu R, Nauton L, Thery V, et al: Synthesis, Pim kinase inhibitory potencies and in vitro antiproliferative activities of diversely substituted pyrrolo[2,3-a]carbazoles. Bioorg Med Chem. 18:6865–6873. 2010.PubMed/NCBI

57 

Letribot B, Akue-Gedu R, Santio NM, et al: Use of copper(I) catalyzed azide alkyne cycloaddition (CuAAC) for the preparation of conjugated pyrrolo[2,3-a]carbazole Pim kinase inhibitors. Eur J Med Chem. 50:304–310. 2012.PubMed/NCBI

58 

Chen WS, Xu PZ, Gottlob K, et al: Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene. Genes Dev. 15:2203–2208. 2001. View Article : Google Scholar : PubMed/NCBI

59 

Easton RM, Cho H, Roovers K, et al: Role for Akt3/protein kinase Bgamma in attainment of normal brain size. Mol Cell Biol. 25:1869–1878. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Woulfe D, Jiang H, Morgans A, Monks R, Birnbaum M and Brass LF: Defects in secretion, aggregation, and thrombus formation in platelets from mice lacking Akt2. J Clin Invest. 113:441–450. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Mukaida N, Wang YY and Li YY: Roles of Pim-3, a novel survival kinase, in tumorigenesis. Cancer Sci. 102:1437–1442. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 29 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Z, Li X, Shang K, Zhang P, Wang C, Xin Y, Zhou L and Li Y: T-18, a stemonamide synthetic intermediate inhibits Pim kinase activity and induces cell apoptosis, acting as a potent anticancer drug. Oncol Rep 29: 1245-1251, 2013
APA
Wang, Z., Li, X., Shang, K., Zhang, P., Wang, C., Xin, Y. ... Li, Y. (2013). T-18, a stemonamide synthetic intermediate inhibits Pim kinase activity and induces cell apoptosis, acting as a potent anticancer drug. Oncology Reports, 29, 1245-1251. https://doi.org/10.3892/or.2013.2233
MLA
Wang, Z., Li, X., Shang, K., Zhang, P., Wang, C., Xin, Y., Zhou, L., Li, Y."T-18, a stemonamide synthetic intermediate inhibits Pim kinase activity and induces cell apoptosis, acting as a potent anticancer drug". Oncology Reports 29.3 (2013): 1245-1251.
Chicago
Wang, Z., Li, X., Shang, K., Zhang, P., Wang, C., Xin, Y., Zhou, L., Li, Y."T-18, a stemonamide synthetic intermediate inhibits Pim kinase activity and induces cell apoptosis, acting as a potent anticancer drug". Oncology Reports 29, no. 3 (2013): 1245-1251. https://doi.org/10.3892/or.2013.2233