Synergistic effect of all‑trans retinoic acid in combination with protein kinase C 412 in FMS-like tyrosine kinase 3-mutated acute myeloid leukemia cells

  • Authors:
    • Hoang Thanh Chi
    • Bui Thi Kim Ly
    • Hoang Anh Vu
    • Yuko Sato
    • Phu Chi Dung
    • Phan Thi Xinh
  • View Affiliations

  • Published online on: January 15, 2015     https://doi.org/10.3892/mmr.2015.3203
  • Pages: 3969-3975
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute myeloid leukemia (AML) is a heterogeneous disease. Numerous molecular abnormalities have been identified in AML and, amongst these, FMS‑like tyrosine kinase 3 (FLT3) mutations are one of the most common somatic alterations detected. In the present study, an in vitro investigation was performed to evaluate the effects of all‑trans retinoic acid (ATRA) and PKC412, alone and in combination, in FLT3‑mutated AML cell lines. Trypan blue exclusion test, as well as morphological, western blot and isobologram analyses were conducted. The results indicated that the combined ATRA and PKC412 treatment exhibited additive or synergistic effects in FLT3‑mutated AML cell lines. These results provided in vitro evidence for the future clinical trials evaluating the effects of a combination treatment using PKC412 and ATRA on AML patients with FLT3‑mutations.

Introduction

Acute myeloid leukemia (AML) is a common hematological malignancy, which is characterized by the arrest of myeloid cells at various differentiation stages, resulting in their accumulation as they are unable to differentiate into mature, functional blood cells (1,2). Numerous genetic abnormalities have been identified in AML. Among these, FMS-like tyrosine kinase 3 (FLT3) abnormality is common and a previous study has demonstrated that FLT3 was abnormally activated in 70–90% of patients with AML, including overexpression of wild-type FLT3 and FLT3 mutations (3).

AML patients with FLT3 abnormalities present unfavorable prognoses (4), including a high risk of relapse and lower long-term survival rates compared with patients with wild-type FLT3 (5,6). The use of cytarabine- and anthracycline-based intensive chemotherapy, in combination with advanced supportive care and the introduction of allogeneic stem cell transplantation has been demonstrated to initially improve the outcome of patient responses (7,8). However, these responses were found to be transient, lasting for weeks to months, followed by progressive disease development and subsequent drug-resistance (9). The median survival of FLT3-mutated patients with AML has been reported to be ≤5 months, following the first disease relapse (10,11). To date, multiple small molecule FLT3-tyrosine kinase inhibitors (FLT3-TKIs) have been developed, and their effect in AML patients as single agents or in combination with chemotherapy have been evaluated (12). Protein kinase C (PKC)412 (N-benzoyl-staurosporine) is a FLT3-TKI that was originally developed as a PKC and vascular growth factor receptor inhibitor. PKC412 has been used in phase I clinical trials for the treatment of solid tumors, which demonstrated a tolerable dose (13,14). Subsequently, PKC412 was found to specifically and potently inhibit the growth of leukemic cell lines expressing FLT3-internal tandem duplication (ITD)-induced-myeloproliferative-like syndrome in the nanomolar range (15). The results of phase II and IIB trials, where patients with FLT3 mutations were treated with PKC412, indicated that PKC412 was generally well-tolerated. These results suggested a potentially effective strategy comprising a combination of PKC412 and other agents, including chemotherapy, in AML patients with FLT3-mutations (16,17).

Individuals with a subtype of AML known as acute promyelocytic leukaemia (APL) are frequently treated with all-trans retinoic acid (ATRA). ATRA is a derivative of vitamin A (retinoids), also known as tretinoin (Vesanoid®). ATRA induces leukemia cell maturation and differentiation, and is therefore able to rapidly reduce leukemia symptoms by preventing myeloid cell accumulation (18). In addition, ATRA has been demonstrated to induce cell growth arrest, differentiation and cell death of various types of cancer cells in vitro (19,20). However, the clinical applications of ATRA are limited by side-effects, which include acute retinoid resistance, hypertriglyceridemia, mucocutaneous dryness, headaches, disease relapse following a brief remission period and drug resistance (2123). In addition, its clinical applications are further limited due to its low plasma concentrations. Therefore, in order to circumvent these limitations, combinations of ATRA and other anticancer drugs have been investigated (24,25). Previous studies have indicated that ATRA was able to enhance the cytotoxic effect of chemotherapeutic drugs (26,27). Furthermore, certain preclinical trials have demonstrated the efficacy of using a combination of retinoids and cytotoxic drugs (2729).

In the present study, an in vitro investigation was performed to evaluate the effect of the combination of ATRA and PKC412 in FLT3-mutated cell lines. The results of the present study may establish whether a clinical trial on patients with FLT3 mutations should be conducted.

Materials and methods

Cell lines and culture conditions

Four human leukemia cell lines were used in this study and were obtained as described previously (30). Briefly, the two sister cell lines, MOLM13 and MOLM14, were obtained from a patient with acute monocytic leukemia (M5a) presenting the t(9;11) mutation (31), MV4-11 cell line was acquired from an AML patient carrying the t(4;11) mutation (32) and KOCL-48 cell line was obtained from an infant leukemic patient carrying the t(4;11) mutation (33).

Two mutations within FLT3 exon 14 were detected in MOLM13 and MOLM14 cells, including an ITD (21 bp) corresponding to codons Phe594-Asp600 and a novel missense nucleotide substitution at codon 599 (Tyr599Phe) (34,35). These mutations were located on the same allele (35). In MV4-11 cells, an ITD (30 bp) within FLT3 exon 14, corresponding to codons Tyr591-Asp600, and a Tyr591His mutation were detected (34,35). In the KOCL-48 cell line, only the FLT3-Asp835Glu mutation was detected (34).

All the cell lines were cultured in RPMI-1640 medium (Sigma-Aldrich Japan K.K., Tokyo, Japan) supplemented with 10% heat-inactivated fetal bovine serum (FBS; JRH Biosciences, Lenexa, KS, USA), 100 IU/ml penicillin and 0.1 mg/ml streptomycin (Nakalai Tesque, Kyoto, Japan) in a humidified atmosphere containing 5% CO2 at 37°C.

Reagents

PKC412 was provided by Novartis Pharma AG (Basel, Switzerland) and ATRA was purchased from Wako Pure Chemical Industries, Ltd. (Osaka, Japan). All the drugs were dissolved in dimethylsulfoxide (DMSO; Merck-Millipore, Darmstadt, Germany). Control cells were cultured with the same DMSO concentration as that used for the highest drug dose (1:1000 dilution). To avoid cytotoxicity, the concentration of DMSO was maintained at <0.1%.

Cell proliferation assays

Cell proliferation was determined using the trypan blue dye exclusion test as described previously (30). Briefly, the cells were seeded in six-well plates (1×105 cells/ml) in the presence of various ATRA or PKC412 concentrations for 72 h. Following treatment, 10 μl cell suspension was mixed with 10 μl 0.4% trypan blue (Nacalai Tesque, Tokyo, Japan), and live cells were counted manually using a hemacytometer (Erma, Tokyo, Japan). The results were expressed as the percentage of the values obtained when the cells were grown in the absence of reagents.

Morphological assessment for the detection of apoptotic cells

In order to detect fragmented nuclei and condensed chromatin, the cells (1×105 cells/ml) were treated with 1 μM ATRA. Following incubation for 8 h, the cells were harvested and fixed onto slides using a Cytospin 2 (Shandon Southern Products Ltd., Cheshire, UK). Subsequently, the cells were stained with a Wright-Giemsa solution (Merck Co., Ltd, Tokyo, Japan) and the cell morphology was observed under an inverted microscope (IX70; Olympus Corp. Tokyo, Japan).

Western blot analysis

Cells were plated onto 10-cm dishes at a density of 1×105 cells/ml in the presence of 1 μM ATRA. Following incubation for the indicated time periods, the cells were collected and washed twice with PBS. Next, the cells were dissolved in protein lysis buffer (Wako Pure Chemical Industries, Ltd), consisting of 5 mM EDTA, 50 mM NaF, 10 mM Na2H2P2O7, 0.01% Triton X-100, 5 mM HEPES, 150 mM NaCl, 1 mM Na3VO4, 1 mM phenylmethylsulfonyl fluoride and 75 μg/ml aprotinin, on ice for 30 min with a brief vortex of four times every 10 min. The samples were centrifuged at 16,000 × g at 4°C for 10 min and the total cell lysates were collected for western blot analysis. Subsequently, the protein samples were subjected to 12% polyacrylamide gel electrophoresis and transferred to Hypond-P membranes (GE Healthcare Life Sciences, Little Chalfont, UK) using electroblotting. Following washing with PBS, the membranes were incubated with various antibodies and antibody-binding was detected using enhanced chemiluminescence system (Amersham Pharmacia Biotech, Tokyo, Japan). Rabbit polyclonal anti-actin (A2066; 1:500 in 5% skimmed milk) was obtained from Sigma-Aldrich, rabbit polyclonal anti-caspase-3 (#9662; 1:1,000 in 5% BSA) and mouse anti-caspase-9 (#9508 1:1,000 in 5% BSA) antibodies were purchased from Cell Signaling Technology (Tokyo, Japan) and rabbit-anti-poly-adenosine diphosphate ribose polymerase (PARP) antibody (#9542; 1:1,000 in 5% BSA) was purchased from Wako Pure Chemical Industries, Ltd. Secondary antibodies horseradish peroxidase (HRP)-conjugated anti-rabbit immunoglobulin (Ig)G (sc-2317) and anti-mouse IgG-HRP (sc-2031) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA)

Isobologram analysis

The isobologram method described by Steel and Peckham (36) was used to evaluate the dose-response interactions between ATRA and PKC412 in the MOLM13, MOLM14, MV4-11 and KOCL-48 cells at the IC50 level. IC50 was defined as the concentration of the reagent that produced 50% cell-growth inhibition. The isobologram method used in this study was selected since it can be used to investigate anticancer agents with unclear cytotoxic mechanisms and various dose-response curves (36).

Statistical analysis

Data obtained from the isobolograms were analyzed as described previously (37). The drug combinations were considered to have a synergistic effect when the majority of the observed data points were found in the area of supra-additivity (mean value of observed data less than predicted minimum values). By contrast, an antagonistic effect was considered between the drug combinations when the majority of data points were located in the areas of subadditivity and protection (mean value of observed data more than predicted maximum value). Statistical analyses were performed to analyze the significance of the detected synergism (or antagonism). The Wilcoxon signed-rank test was used to compare the observed data with the predicted minimum or maximum values for additive effects, which were closest to the observed data. Values are expressed as the mean ± standard deviation of three independent experiments. P<0.05 was considered to indicate a statistically significant difference. P≥0.05 was considered to indicate additive to synergistic (or additive to antagonistic) effect. Additional data were analyzed using Student’s t-test. Statistical analyses were performed using StatView 4.01 software program (Abacus Concepts, Berkeley, CA, USA).

Results

ATRA and PKC412 inhibit proliferation of FLT3-mutated AML cells

In order to evaluate the effect of ATRA and PKC412 on cell growth, the cell lines (MOLM13, MOLM14, MV4-11 and KOCL-48) were incubated with DMSO alone (0 μM reagents), with various concentrations of ATRA (0.001–10 μM) or with various concentrations of PKC412 (5–80 nM) for 72 h. Cell proliferation was evaluated using the trypan blue exclusion test. The results indicated that ATRA and PKC412 inhibited the cellular proliferation of MOLM13, MOLM14, MV4-11 and KOCL-48 cells in a dose-dependent manner (Fig. 1).

ATRA induces apoptosis in FLT3-mutated AML cells

Previous studies demonstrated that ATRA was able to induce the differentiation of immature leukemic blasts into terminally differentiated granulocytic cells (38,39), or the apoptosis of specific tumor cells (340,41). To elucidate the mechanism underlying the ATRA-induced suppression of cellular proliferation in MOLM13, MOLM14, MV4-11 and KOCL-48 cells, the cell morphology and expression levels of apoptotic markers in these cells following treatment with 1 μM ATRA were evaluated. After 8 h of treatment with 1 μM ATRA, higher apoptosis was observed in the cell membranes of MOLM13 cells compared with the non-treated cells (0-h treatment; Fig. 2A and B). Furthermore, bands of cleaved caspase-9 were detected after 8 h of incubation with 1 μM ATRA. Cleaved caspase-9 induced the activation of caspase 3 (indicated by the presence of the cleaved caspase-3 band), which subsequently inactived an enzymes involved in DNA repair, known as PARP (Fig. 2C). The caspase 3-mediated proteolytic cleavage of PARP is a key event in apoptosis. In addition, apoptotic bodies were also observed after 24 h of treatment with 1 μM ATRA in MOLM13 cells (Fig. 2B), which indicated that ATRA induced apoptosis in cell lines exhibiting FLT3 mutations. Analogous results were observed following evaluation of apoptosis in the remainder cell lines (data not shown).

Synergistic effect of combined ATRA and PKC412 treatment

ATRA was demonstrated to suppress cellular proliferation by inducing apoptosis, whereas PKC412 is known to be a specific FLT3-inhibitor that suppresses cell proliferation by inhibiting FLT3 expression in FLT3-mutated cell lines (15). The present study aimed to elucidate whether a combination of ATRA and PKC412 was able to enhance the effect of these drugs on the suppression of cell proliferation in FLT3-mutated cell lines. The results revealed that combined treatment with ATRA and PKC412 had a synergistic effect on MOLM13 and MOLM14 cells and an additive effect on KOCL-48 and MV4-11 cells (Fig. 3). In the MOLM13 and MOLM14 cells, the combined data points were detected in the areas of supra-additivity and additivity. The mean values obtained were lower compared with the predicted minimum values (Table I), and the differences were found to be significant (P<0.01), indicating synergistic effects. However, in the KOCL-48 and MV4-11 cells, the combined data points were detected within the envelope of additivity, indicating an additive effect. In addition, the mean values detected for the KOCL-48 and MV4-11 cells were lower compared with the predicted maximum valies and higher than the predicted minimum values (Table I), confirming the presence of additive effects.

Table I

Mean values of observed data and predicted minimum and maximum values of the combination of all-trans retinoic acid and protein kinase C 412.

Table I

Mean values of observed data and predicted minimum and maximum values of the combination of all-trans retinoic acid and protein kinase C 412.

Predicted values for an additive effect

Cell linenObserved dataMinimumMaximumEffect
MOLM1340.1500.7710.895Synergistic (<0.01)
MOLM1440.2310.2650.584Synergistic (<0.01)
MV4-1150.4420.2450.653Additive
KOCL-4860.4320.3350.632Additive

Discussion

Despite the positive response to certain therapeutic strategies, the decreased ability of cancer cells to undergo apoptosis by malignant evolution represents a major challenge in the development of effective therapeutic approaches (19). Currently, novel selective strategies aiming to manipulate cancer cells, but not healthy cells, towards apoptosis are under development as potential therapies (42). Therefore, apoptosis-inducing agents that do not induce cytotoxicity in normal cells represent a potential anticancer treatment. The results of the present study demonstrated that ATRA inhibited cell proliferation and induced apoptosis in FLT3-mutated cell lines, indicating that ATRA may be a potentially useful drug for the treatment of AML patients with FLT3 mutations.

ATRA has been demonstrated to inhibit vascular endothelial growth factor, which is crucial for the process of angiogenesis (29), and represents a major development in the treatment of APL with differentiation therapy. However, the duration of remission induced and maintained by ATRA therapy alone has been found to be short-lived, and ATRA alone failed to induce a second remission in the majority of patients following relapse (43,44). To circumvent these limitations, improving the effectiveness of ATRA on first drug application is required. Notably, AML results from by a combination of at least two pathophysiological problems. Therefore, the application of a therapy targeting only one pathophysiological pathway may not be sufficient to induce a major response, unless using a therapeutic combination. Furthermore, anticancer drugs may induce severe cytotoxic side-effects, which limit the doses that can be administered during treatment, thereby limiting the potential effectiveness of these therapeutic approaches (45,46). The use of differential combinations of anticancer drugs may circumvent these limitations by improving the effectiveness of cancer chemotherapy (4547). The majority of anticancer drugs have distinct mechanisms of action and are associated with specific cytotoxic side-effects. In addition, an upper concentration limit exists for each drug in order to achieve effective inhibition of tumor-cell proliferation, whilst minimizing the extent of damage to healthy cells. Therefore, an ideal anticancer drug combination should maximize the therapeutic efficacy and minimize the associated cytotoxic side-effects (4850).

Based on the aforementioned principals, the present study aimed to evaluate the effect of a combination of ATRA and PKC412 on FLT3-mutated AML cell lines. Preliminary preclinical data obtained in the present study demonstrated that the combination of ATRA and PKC412 had a synergistic/additive cytotoxic effect on FLT3-mutated cell lines as compared to each agent alone. For instance, the IC50 of ATRA alone in MOLM13 cells was found to be 0.01 μM; however, upon combined treatment with PKC412 (3.5 nM), the IC50 concentration of ATRA was significantly reduced to 0.001 μM (P<0.01). Similarly, the IC50 of PKC412 alone in MOLM13 cells was found to be 20 nM; however, upon combined treatment with ATRA (0.001 μM), the IC50 concentration of PKC412 was reduced to 3.5 nM (P<0.01).

A previous study indicated that the side-effects associated with PKC412 treatment were associated with the dosage administered (14). Therefore, in the treatment combination, ATRA and PKC412 may maximize the therapeutic efficacy and minimize cytotoxic side-effects. In conclusion, the results of the present study provided experimental evidence for the effect of a combined ATRA and PKC412 therapeutic strategy in the prevention and treatment of AML patients with FLT3-mutations.

References

1 

Breitenbuecher F, Schnittger S, Grundler R, Markova B, Carius B, Brecht A, Duyster J, Haferlach T, Huber C and Fischer T: Identification of a novel type of ITD mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor. Blood. 113:4074–4077. 2009. View Article : Google Scholar

2 

O’Donnell MR, Appelbaum FR, Coutre SE, Damon LE, Erba HP, Foran J, et al: Acute myeloid leukemia. J Natl Compr Canc Netw. 6:962–993. 2008.

3 

Foran JM: New prognostic markers in acute myeloid leukemia: perspective from the clinic. Hematology Am Soc Hematol Educ Program. 2010:47–55. 2010. View Article : Google Scholar

4 

Scholl C, Gilliland DG and Frohling S: Deregulation of signaling pathways in acute myeloid leukemia. Semin Oncol. 35:336–345. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Kuchenbauer F, Kern W, Schoch C, Kohlmann A, Hiddemann W, Haferlach T, et al: Detailed analysis of FLT3 expression levels in acute myeloid leukemia. Haematologica. 90:1617–1625. 2005.PubMed/NCBI

6 

Kang HJ, Lee JW, Kho SH, Kim MJ, Seo YJ, Kim H, et al: High transcript level of FLT3 associated with high risk of relapse in pediatric acute myeloid leukemia. J Korean Med Sci. 25:841–845. 2005. View Article : Google Scholar

7 

Burnett AK: Acute myeloid leukemia: treatment of adults under 60 years. Rev Clin Exp Hematol. 6:26–45. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Löwenberg B, Downing JR and Burnett A: Acute myeloid leukemia. N Engl J Med. 341:1051–1062. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Chu SH and Small D: Mechanisms of resistance to FLT3 inhibitors. Drug Resist Updat. 12:8–16. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Ravandi F, Kantarjian H, Faderl S, Garcia-Manero G, O’Brien S, Koller C, Pierce S, Brandt M, Kennedy D, Cortes J and Beran M: Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk Res. 34:752–756. 2010. View Article : Google Scholar

11 

Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S, Erba H, Stuart RK, Baccarani M, Cripe LD, et al: Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood. 117:3294–3301. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Kindler T, Lipka DB and Fischer T: FLT3 as a therapeutic target in AML: still challenging after all these years. Blood. 116:5089–5102. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Monnerat C, Henriksson R, Le Chevalier T, Novello S, Berthaud P, Faivre S, et al: Phase I study of PKC412 (N-benzoyl-staurosporine), a novel oral protein kinase C inhibitor, combined with gemcitabine and cisplatin in patients with non-small-cell lung cancer. Ann Oncol. 15:316–323. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Propper DJ, McDonald AC, Man A, Thavasu P, Balkwill F, Braybrooke JP, et al: Phase I and pharmacokinetic study of PKC412, an inhibitor of protein kinase C. J Clin Oncol. 19:1485–1492. 2001.PubMed/NCBI

15 

Weisberg E, Boulton C, Kelly LM, Manley P, Fabbro D, Meyer T, Gilliland DG and Griffin JD: Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell. 1:433–443. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Stone RM, DeAngelo DJ, Klimek V, Galinsky I, Estey E, Nimer SD, Grandin W, Lebwohl D, Wang Y, Cohen P, et al: Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood. 105:54–60. 2005. View Article : Google Scholar

17 

Fischer T, Stone RM, Deangelo DJ, Galinsky I, Estey E, Lanza C, et al: Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol. 28:4339–4345. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Lotan R: Suppression of squamous cell carcinoma growth and differentiation by retinoids. Cancer Res. 54(Suppl 7): 1987s–1990s. 1994.PubMed/NCBI

19 

Caliaro MJ, Marmouget C, Guichard S, Mazars P, Valette A, Moisand A, Bugat R and Jozan S: Response of four human ovarian carcinoma cell lines to all-trans retinoic acid: relationship with induction of differentiation and retinoic acid receptor expression. Int J Cancer. 56:743–748. 1994. View Article : Google Scholar : PubMed/NCBI

20 

Amos B and Lotan R: Retinoid-sensitive cells and cell lines. Methods Enzymol. 190:217–225. 1990. View Article : Google Scholar : PubMed/NCBI

21 

Frankel SR, Eardley A, Lauwers G, Weiss M and Warrell RP Jr: The “retinoic acid syndrome” in acute promyelocytic leukemia. Ann Intern Med. 117:292–296. 1992. View Article : Google Scholar : PubMed/NCBI

22 

Muindi J, Frankel SR, Miller WH Jr, Jakubowski A, Scheinberg DA, Young CW, et al: Continuous treatment with all-trans retinoic acid causes a progressive reduction in plasma drug concentrations: implications for relapse and retinoid “resistance” in patients with acute promyelocytic leukemia. Blood. 79:299–303. 1992.PubMed/NCBI

23 

Conley BA, Egorin MJ, Sridhara R, Finley R, Hemady R, Wu S, et al: Phase I clinical trial of all-trans-retinoic acid with correlation of its pharmacokinetics and pharmacodynamics. Cancer Chemother Pharmacol. 39:291–299. 1997. View Article : Google Scholar : PubMed/NCBI

24 

Karmakar S, Banik NL and Ray SK: Combination of all-trans retinoic acid and paclitaxel-induced differentiation and apoptosis in human glioblastoma U87MG xenografts in nude mice. Cancer. 112:596–607. 2008. View Article : Google Scholar

25 

Ortiz MA, Bayon Y, Lopez-Hernandez FJ and Piedrafita FJ: Retinoids in combination therapies for the treatment of cancer: mechanisms and perspectives. Drug Resist Updat. 5:162–175. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Arrieta O, González-De la Rosa CH, Aréchaga-Ocampo E, Villanueva-Rodríguez G, Cerón-Lizárraga TL, Martínez-Barrera L, Vázquez-Manríquez ME, et al: Randomized phase II trial of All-trans-retinoic acid with chemotherapy based on paclitaxel and cisplatin as first-line treatment in patients with advanced non-small-cell lung cancer. J Clin Oncol. 28:3463–3471. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Boorjian SA, Milowsky MI, Kaplan J, Albert M, Cobham MV, Coll DM, Mongan NP, Shelton G, Petrylak D, Gudas LJ and Nanus DM: Phase 1/2 clinical trial of interferon alpha2b and weekly liposome-encapsulated all-trans retinoic acid in patients with advanced renal cell carcinoma. J Immunother. 30:655–662. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Bryan M, Pulte ED, Toomey KC, Pliner L, Pavlick AC, Saunders T and Wieder R: A pilot phase II trial of all-trans retinoic acid (Vesanoid) and paclitaxel (Taxol) in patients with recurrent or metastatic breast cancer. Invest New Drugs. 29:1482–1487. 2011. View Article : Google Scholar

29 

Kini AR, Peterson LA, Tallman MS and Lingen MW: Angiogenesis in acute promyelocytic leukemia: induction by vascular endothelial growth factor and inhibition by all-trans retinoic acid. Blood. 97:3919–3924. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Ly BT, Chi HT, Yamagishi M, et al: Inhibition of FLT3 expression by green tea catechins in FLT3 mutated-AML cells. PLoS One. 8:e663782013. View Article : Google Scholar : PubMed/NCBI

31 

Matsuo Y, MacLeod RA, Uphoff CC, Drexler HG, Nishizaki C, Katayama Y, Kimura G, Fujii N, Omoto E, Harada M and Orita K: Two acute monocytic leukemia (AML-M5a) cell lines (MOLM-13 and MOLM-14) with interclonal phenotypic heterogeneity showing MLL-AF9 fusion resulting from an occult chromosome insertion, ins (11;9) (q23;p22p23). Leukemia. 11:1469–1477. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Lange B, Valtieri M, Santoli D, Caracciolo D, Mavilio F, Gemperlein I, et al: Growth factor requirements of childhood acute leukemia: establishment of GM-CSF-dependent cell lines. Blood. 70:192–199. 1987.PubMed/NCBI

33 

Iida S, Saito M, Okazaki T, Seto M, Yamamoto K, Akao Y, et al: Phenotypic and genotypic characterization of 14 leukemia and lymphoma cell lines with 11q23 translocations. Leuk Res. 16:1155–1163. 1992. View Article : Google Scholar : PubMed/NCBI

34 

Taketani T, Taki T, Sugita K, Furuichi Y, Ishii E, Hanada R, Tsuchida M, Ida K and Hayashi Y: FLT3 mutations in the activation loop of tyrosine kinase domain are frequently found in infant ALL with MLL rearrangements and pediatric ALL with hyperdiploidy. Blood. 103:1085–1088. 2004. View Article : Google Scholar

35 

Furukawa Y, Vu HA, Akutsu M, Odgerel T, Izumi T, Tsunoda S, et al: Divergent cytotoxic effects of PKC412 in combination with conventional antileukemic agents in FLT3 mutation-positive versus -negative leukemia cell lines. Leukemia. 21:1005–1014. 2007.PubMed/NCBI

36 

Steel GG and Peckham MJ: Exploitable mechanisms in combined radiotherapy-chemotherapy: the concept of additivity. Int J Radiat Oncol Biol Phys. 5:85–91. 1979. View Article : Google Scholar : PubMed/NCBI

37 

Kano Y, Akutsu M, Tsunoda S, Mori K, Suzuki K and Adachi KI: In vitro schedule-dependent interaction between paclitaxel and SN-38 (the active metabolite of irinotecan) in human carcinoma cell lines. Cancer Chemother Pharmacol. 42:91–98. 1998. View Article : Google Scholar : PubMed/NCBI

38 

Breitman TR, Collins SJ and Keene BR: Terminal differentiation of human promyelocytic leukemic cells in primary culture in response to retinoic acid. Blood. 57:1000–10004. 1981.PubMed/NCBI

39 

Warrell RP Jr, Frankel SR, Miller WH Jr, Scheinberg DA, Itri LM, Hittelman WN, Vyas R, And-reeff M, Tafuri A, Jakubowski A, et al: Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid). N Engl J Med. 324:1385–1393. 1991. View Article : Google Scholar : PubMed/NCBI

40 

Lotan R: Retinoids as modulators of tumor cells invasion and metastasis. Semin Cancer Biol. 2:197–208. 1991.PubMed/NCBI

41 

Hofmann SL: Retinoids--”differentiation agents” for cancer treatment and prevention. Am J Med Sci. 304:202–213. 1992. View Article : Google Scholar : PubMed/NCBI

42 

Ashkenazi A, Holland P and Eckhardt SG: Ligand-based targeting of apoptosis in cancer: the potential of recombinant human apoptosis ligand 2/Tumor necrosis factor-related apoptosis-inducing ligand (rhApo2L/TRAIL). J Clin Oncol. 26:3621–3630. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Warrell RP Jr, de Thé H, Wang ZY and Degos L: Acute promyelocytic leukemia. N Engl J Med. 329:177–189. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Degos L, Dombret H, Chomienne C, Daniel MT, Micléa JM, Chastang C, Castaigne S and Fenaux P: All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia. Blood. 85:2643–2653. 1995.PubMed/NCBI

45 

Das A, Banik NL and Ray SK: Retinoids induced astrocytic differentiation with down regulation of telomerase activity and enhanced sensitivity to taxol for apoptosis in human glioblastoma T98G and U87MG cells. J Neurooncol. 87:9–22. 2008. View Article : Google Scholar

46 

Nagai S, Takenaka K, Sonobe M, Wada H and Tanaka F: Schedule-dependent synergistic effect of pemetrexed combined with gemcitabine against malignant pleural mesothelioma and non-small cell lung cancer cell lines. Chemotherapy. 54:166–175. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Ridwelski K, Gebauer T, Fahlke J, Kröning H, Kettner E, Meyer F, et al: Combination chemotherapy with docetaxel and cisplatin for locally advanced and metastatic gastric cancer. Ann Oncol. 12:47–51. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Sarkar K and Yang H: Encapsulation and extended release of anti-cancer anastrozole by stealth nanoparticles. Drug Deliv. 15:343–346. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Sofou S: Radionuclide carriers for targeting of cancer. Int J Nanomedicine. 3:181–199. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Torchilin V: Antibody-modified liposomes for cancer chemotherapy. Expert Opin Drug Deliv. 5:1003–1025. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 11 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chi HT, Ly BT, Vu HA, Sato Y, Dung PC and Xinh PT: Synergistic effect of all‑trans retinoic acid in combination with protein kinase C 412 in FMS-like tyrosine kinase 3-mutated acute myeloid leukemia cells. Mol Med Rep 11: 3969-3975, 2015
APA
Chi, H.T., Ly, B.T., Vu, H.A., Sato, Y., Dung, P.C., & Xinh, P.T. (2015). Synergistic effect of all‑trans retinoic acid in combination with protein kinase C 412 in FMS-like tyrosine kinase 3-mutated acute myeloid leukemia cells. Molecular Medicine Reports, 11, 3969-3975. https://doi.org/10.3892/mmr.2015.3203
MLA
Chi, H. T., Ly, B. T., Vu, H. A., Sato, Y., Dung, P. C., Xinh, P. T."Synergistic effect of all‑trans retinoic acid in combination with protein kinase C 412 in FMS-like tyrosine kinase 3-mutated acute myeloid leukemia cells". Molecular Medicine Reports 11.5 (2015): 3969-3975.
Chicago
Chi, H. T., Ly, B. T., Vu, H. A., Sato, Y., Dung, P. C., Xinh, P. T."Synergistic effect of all‑trans retinoic acid in combination with protein kinase C 412 in FMS-like tyrosine kinase 3-mutated acute myeloid leukemia cells". Molecular Medicine Reports 11, no. 5 (2015): 3969-3975. https://doi.org/10.3892/mmr.2015.3203