Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 2/2008

01-07-2008 | Original Article

Antiproliferative effects of rapamycin as a single agent and in combination with carboplatin and paclitaxel in head and neck cancer cell lines

Authors: Nasredine Aissat, Christophe Le Tourneau, Aïda Ghoul, Maria Serova, Ivan Bieche, François Lokiec, Eric Raymond, Sandrine Faivre

Published in: Cancer Chemotherapy and Pharmacology | Issue 2/2008

Login to get access

Abstract

Purpose

Recent data suggested that combining targeted therapies with chemotherapy may counteract drug resistance. Activation of the PI3K/AKT/mTOR pathway downstream to kinase receptors, such as EGFR, was found in 57–81% of head and neck squamous cell carcinoma (HNSCC), and was eventually associated with a loss of PTEN function. mTOR was shown to modulate cell proliferation, apoptosis, invasion, and angiogenesis. This study aimed to evaluate molecular and cellular effects of rapamycin in a panel of cell lines either as single agent or in combination with cytotoxics commonly used in HNSCC.

Methods

Antiproliferative effects of rapamycin, carboplatin, and paclitaxel were evaluated in a panel of three HNSCC cell lines (SCC61, SQ20B and HEP2). Cells were exposed to rapamycin for 48 h, to carboplatin for 48 h, or to paclitaxel for 24 h. Antiproliferative effects of simultaneous and sequential rapamycin-based combinations were studied using MTT assay and median effect plot analysis. Cell cycle effects were analysed using flow cytometry.

Results

Rapamycin induced concentration dependent antiproliferative effects in HNSCC cell lines with IC50 of 5 ± 1, 12 ± 2 and 20 ± 2 μM in SCC61, SQ20B, and HEP2 cells, respectively. Higher antiproliferative effects were observed in SCC61 cells overexpressing NOXA and cyclin D1 than in HEP2 that overexpressed MDR1 and BCL2. In our panel, antiproliferative effects of rapamycin were associated with G0/G1 cell cycle accumulation and apoptosis induction, at concentrations ranging 3–30 μM. Combinations of rapamycin with paclitaxel and carboplatin displayed synergistic and additive effects. Synergistic effects were observed with paclitaxel in SQ20B and HEP2 cells and with carboplatin in SQ20B cells, when cells were exposed to cytotoxics prior to rapamycin.

Conclusion

Our results show that rapamycin displays antiproliferative effects and induces apoptosis in HNSCC cell lines, cellular effects being more potent in cells that do not express BCL2 and MDR1. Additive and synergistic effects were observed when rapamycin was combined with carboplatin and paclitaxel.
Literature
1.
go back to reference Grandis JR, Tweardy DJ (1993) Elevated levels of transforming growth factor alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer. Cancer Res 53:3579–3584PubMed Grandis JR, Tweardy DJ (1993) Elevated levels of transforming growth factor alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer. Cancer Res 53:3579–3584PubMed
2.
go back to reference Grandis JR, Melhem MF, Gooding WE et al (1998) Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst 90:824–832CrossRef Grandis JR, Melhem MF, Gooding WE et al (1998) Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst 90:824–832CrossRef
3.
go back to reference Vivanco I, Sawyers CL (2002) The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer 2:489–501PubMedCrossRef Vivanco I, Sawyers CL (2002) The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer 2:489–501PubMedCrossRef
4.
go back to reference Brown EJ, Albers MW, Shin TB et al (1994) A mammalian protein targeted by G1-arrestingreceptor complex. Nature 369:756–768PubMedCrossRef Brown EJ, Albers MW, Shin TB et al (1994) A mammalian protein targeted by G1-arrestingreceptor complex. Nature 369:756–768PubMedCrossRef
5.
go back to reference Bjornsti MA, Houghton PJ (2004) The TOR pathway: a target for cancer therapy. Nat Rev Cancer 4:335–348PubMedCrossRef Bjornsti MA, Houghton PJ (2004) The TOR pathway: a target for cancer therapy. Nat Rev Cancer 4:335–348PubMedCrossRef
6.
go back to reference Vignot S, Faivre S, Aguirre D et al (2005) mTOR-targeted therapy of cancer with rapamycin derivatives. Ann Oncol 16:525–537PubMedCrossRef Vignot S, Faivre S, Aguirre D et al (2005) mTOR-targeted therapy of cancer with rapamycin derivatives. Ann Oncol 16:525–537PubMedCrossRef
7.
go back to reference Bjornsti MA, Houghton PJ (2004) Lost in translation: dysregulation of cap-dependent translation and cancer. Cancer Cell 5:519–523PubMedCrossRef Bjornsti MA, Houghton PJ (2004) Lost in translation: dysregulation of cap-dependent translation and cancer. Cancer Cell 5:519–523PubMedCrossRef
8.
go back to reference Gera JF, Mellinghoff IK, Shi Y et al (2004) AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem 4:2737–2746 Gera JF, Mellinghoff IK, Shi Y et al (2004) AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem 4:2737–2746
9.
go back to reference Faivre S, Kroemer G, Raymond E (2006) Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov 5(8):671–688PubMedCrossRef Faivre S, Kroemer G, Raymond E (2006) Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov 5(8):671–688PubMedCrossRef
10.
go back to reference Grewe M, Gansauge F, Schmid RM et al (1999) Regulation of cell growth and cyclin D1 expression by the constitutively active FRAP-p70s6K pathway in human pancreatic cancer cells. Cancer Res 59:3581–3587PubMed Grewe M, Gansauge F, Schmid RM et al (1999) Regulation of cell growth and cyclin D1 expression by the constitutively active FRAP-p70s6K pathway in human pancreatic cancer cells. Cancer Res 59:3581–3587PubMed
11.
go back to reference Dong J, Peng J, Zhang H et al (2005) Role of glycogen synthase kinase 3beta in rapamycin-mediated cell cycle regulation and chemosensitivity. Cancer Res 65(5):1961–1972PubMedCrossRef Dong J, Peng J, Zhang H et al (2005) Role of glycogen synthase kinase 3beta in rapamycin-mediated cell cycle regulation and chemosensitivity. Cancer Res 65(5):1961–1972PubMedCrossRef
12.
go back to reference Oki E, Baba H, Tokunaga E et al (2005) Akt phosphorylation associates with LOH of PTEN and leads to chemoresistance for gastric cancer. Int J Cancer 117(3):376–380PubMedCrossRef Oki E, Baba H, Tokunaga E et al (2005) Akt phosphorylation associates with LOH of PTEN and leads to chemoresistance for gastric cancer. Int J Cancer 117(3):376–380PubMedCrossRef
13.
go back to reference Geoerger B, Kerr K, Tang CB et al (2001) Antitumor activity of the Rapamycin analog CCI-779 in human primitive neuroectodermal Tumor/Medulloblastoma models as single agent and in combination Chemotherapy. Cancer Res 61:1527–1532PubMed Geoerger B, Kerr K, Tang CB et al (2001) Antitumor activity of the Rapamycin analog CCI-779 in human primitive neuroectodermal Tumor/Medulloblastoma models as single agent and in combination Chemotherapy. Cancer Res 61:1527–1532PubMed
14.
go back to reference Gupta A, Dai Y, Vethanayagam RR et al (2006) Cyclosporin A, tacrolimus and sirolimus are potent inhibitors of the human breast cancer resistance protein (ABCG2) and reverse resistance to mitoxantrone and topotecan. Cancer Chemother Pharmacol 58(3):374–383PubMedCrossRef Gupta A, Dai Y, Vethanayagam RR et al (2006) Cyclosporin A, tacrolimus and sirolimus are potent inhibitors of the human breast cancer resistance protein (ABCG2) and reverse resistance to mitoxantrone and topotecan. Cancer Chemother Pharmacol 58(3):374–383PubMedCrossRef
15.
go back to reference Nagasawa I, Keng P, Maki C et al (1988) Absence of a radiation-induced first-cycle G1-S arrest in p53+ human tumor cells synchronized by mitotic selection. Cancer Res 58:2036–2041 Nagasawa I, Keng P, Maki C et al (1988) Absence of a radiation-induced first-cycle G1-S arrest in p53+ human tumor cells synchronized by mitotic selection. Cancer Res 58:2036–2041
16.
go back to reference Maggiorella L, Frascogna V, Poullain MG et al (2001) The Olivacine S16020 enhances the Antitumor effect of ionizing radiation without increasing radio-induced Mucositis. Clin Cancer Res 7:2091–2095PubMed Maggiorella L, Frascogna V, Poullain MG et al (2001) The Olivacine S16020 enhances the Antitumor effect of ionizing radiation without increasing radio-induced Mucositis. Clin Cancer Res 7:2091–2095PubMed
17.
go back to reference Brachman DG, Beckett M, Graves D et al (1993) p53 mutation does not correlate with radiosensitivity in 24 head and neck cancer cell lines. Cancer Res 53:3667–3669PubMed Brachman DG, Beckett M, Graves D et al (1993) p53 mutation does not correlate with radiosensitivity in 24 head and neck cancer cell lines. Cancer Res 53:3667–3669PubMed
18.
go back to reference Bièche I, Parfait B, Tozlu S et al (2001) Quantitation of androgen receptor gene expression in sporadic breast tumors by real-time RT-PCR: evidence that MYC is an AR-regulated gene. Carcinogenesis 22:1521–1526PubMedCrossRef Bièche I, Parfait B, Tozlu S et al (2001) Quantitation of androgen receptor gene expression in sporadic breast tumors by real-time RT-PCR: evidence that MYC is an AR-regulated gene. Carcinogenesis 22:1521–1526PubMedCrossRef
19.
go back to reference Chou TC, Talalay P (1984) Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 22:27–55PubMedCrossRef Chou TC, Talalay P (1984) Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 22:27–55PubMedCrossRef
20.
go back to reference Janus A, Robak T, Smolewski P (2005) The mammalian target of rapamycin (mTOR) kinase pathway. It’s role in tumourgenesis and targeted antitumour therapy. Cell Mol Biol Lett 10:479–498PubMed Janus A, Robak T, Smolewski P (2005) The mammalian target of rapamycin (mTOR) kinase pathway. It’s role in tumourgenesis and targeted antitumour therapy. Cell Mol Biol Lett 10:479–498PubMed
21.
go back to reference Wiederrecht GJ, Sabers CJ, Brunn GJ et al (1995) Mechanism of action of rapamycin: new insights into the regulation of G1-phase progression in eukaryotic cells. Prog Cell Cycle Res 1:53–71PubMed Wiederrecht GJ, Sabers CJ, Brunn GJ et al (1995) Mechanism of action of rapamycin: new insights into the regulation of G1-phase progression in eukaryotic cells. Prog Cell Cycle Res 1:53–71PubMed
22.
go back to reference Neshat MS, Mellinghoff IK, Tran C et al (2001) Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci USA 98:10314–10319PubMedCrossRef Neshat MS, Mellinghoff IK, Tran C et al (2001) Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci USA 98:10314–10319PubMedCrossRef
23.
go back to reference Mondesire W, Jian W, Zhang H et al (2004) Targeting mammalian target of rapamycin synergistically enhances chemotherapy-induced cytotoxicity in breast cancer cells. Clin Cancer Res 10:7031–7042PubMedCrossRef Mondesire W, Jian W, Zhang H et al (2004) Targeting mammalian target of rapamycin synergistically enhances chemotherapy-induced cytotoxicity in breast cancer cells. Clin Cancer Res 10:7031–7042PubMedCrossRef
24.
go back to reference Harada H, Andersen J, Mann M et al (2001) p70S6 kinase signals cell survival as well as growth, inactivating the pro-apoptotic molecule BAD. Porc Natl Acad Sci USA 98:9666–9670CrossRef Harada H, Andersen J, Mann M et al (2001) p70S6 kinase signals cell survival as well as growth, inactivating the pro-apoptotic molecule BAD. Porc Natl Acad Sci USA 98:9666–9670CrossRef
25.
go back to reference Le X, Hittelman W, Liu J et al (2003) Paclitaxel induces inactivation of p70S6 kinase and phosphorylation of The421 and Ser424 via multiple signaling pathways in mitosis. Oncogene 22:484–497PubMedCrossRef Le X, Hittelman W, Liu J et al (2003) Paclitaxel induces inactivation of p70S6 kinase and phosphorylation of The421 and Ser424 via multiple signaling pathways in mitosis. Oncogene 22:484–497PubMedCrossRef
26.
go back to reference Wan X, Helman L (2002) Effect of insulin-like growth factor II on protecting myoblast cells against cisplatin-induced apoptosis through p70S6 kinase pathway. Neoplasia 4:400–408PubMedCrossRef Wan X, Helman L (2002) Effect of insulin-like growth factor II on protecting myoblast cells against cisplatin-induced apoptosis through p70S6 kinase pathway. Neoplasia 4:400–408PubMedCrossRef
27.
go back to reference Yamamoto K, Ichijo H, Korsmeyer S (1999) BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-Terminal protein kinase pathway normally activated at G2/M. Mol Cell Biol 19:8469–8478PubMed Yamamoto K, Ichijo H, Korsmeyer S (1999) BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-Terminal protein kinase pathway normally activated at G2/M. Mol Cell Biol 19:8469–8478PubMed
28.
go back to reference Floros K, Thomadaki H, Katsaros N et al (2004) mRNA expression analysis of a variety of apoptosis-related genes, including the novel gene of the BCL2-family, BCL2L12, in HL-60 leukemia cells after treatment with carboplatin and doxorubicin. J Biol Chem 385:1099–1103CrossRef Floros K, Thomadaki H, Katsaros N et al (2004) mRNA expression analysis of a variety of apoptosis-related genes, including the novel gene of the BCL2-family, BCL2L12, in HL-60 leukemia cells after treatment with carboplatin and doxorubicin. J Biol Chem 385:1099–1103CrossRef
29.
go back to reference Raymond E, Alexandre J, Faivre S et al (2002) Dosage adjustment and pharmacokinetic profile of irinotecan in cancer patients with hepatic dysfunction. J Clin Oncol 20:4303–4012PubMedCrossRef Raymond E, Alexandre J, Faivre S et al (2002) Dosage adjustment and pharmacokinetic profile of irinotecan in cancer patients with hepatic dysfunction. J Clin Oncol 20:4303–4012PubMedCrossRef
30.
go back to reference Ratain MJ, Napoli KL, Knightley Moshier K et al (2007) A phase 1b study of oral rapamycin (sirolimus) in patients with advanced malignancies. J Clin Oncol 25:3510 Ratain MJ, Napoli KL, Knightley Moshier K et al (2007) A phase 1b study of oral rapamycin (sirolimus) in patients with advanced malignancies. J Clin Oncol 25:3510
31.
go back to reference Aguirre D, Boya P, Bellet D et al (2004) Bcl-2 and CCND1/CDK4 expression levels predict the cellular effects of mTOR inhibitors in human ovarian carcinoma. Apoptosis 6:797–805CrossRef Aguirre D, Boya P, Bellet D et al (2004) Bcl-2 and CCND1/CDK4 expression levels predict the cellular effects of mTOR inhibitors in human ovarian carcinoma. Apoptosis 6:797–805CrossRef
32.
go back to reference Noh W, Mondesire W, Peng J et al (2004) Determinants of Rapamycin sensitivity in breast cancer cells. Clin Cancer Res 10:1013–1023PubMedCrossRef Noh W, Mondesire W, Peng J et al (2004) Determinants of Rapamycin sensitivity in breast cancer cells. Clin Cancer Res 10:1013–1023PubMedCrossRef
33.
go back to reference Wu C, Wangpaichitr M, Feun L et al (2005) Overcoming cispaltin resistance by mTOR inhibitor in lung cancer. Molecular Cancer 4:1–10CrossRef Wu C, Wangpaichitr M, Feun L et al (2005) Overcoming cispaltin resistance by mTOR inhibitor in lung cancer. Molecular Cancer 4:1–10CrossRef
Metadata
Title
Antiproliferative effects of rapamycin as a single agent and in combination with carboplatin and paclitaxel in head and neck cancer cell lines
Authors
Nasredine Aissat
Christophe Le Tourneau
Aïda Ghoul
Maria Serova
Ivan Bieche
François Lokiec
Eric Raymond
Sandrine Faivre
Publication date
01-07-2008
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 2/2008
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-007-0609-2

Other articles of this Issue 2/2008

Cancer Chemotherapy and Pharmacology 2/2008 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