Skip to main content
Top
Published in: Archives of Gynecology and Obstetrics 2/2013

01-08-2013 | Gynecologic Oncology

Valproic acid suppresses cervical cancer tumor progression possibly via activating Notch1 signaling and enhances receptor-targeted cancer chemotherapeutic via activating somatostatin receptor type II

Authors: Cheguo Tsai, Juliana S. Leslie, Laura G. Franko-Tobin, Monica C. Prasnal, Tong Yang, L. Vienna Mackey, Joseph A. Fuselier, David H. Coy, Mingqiu Liu, Changyuan Yu, Lichun Sun

Published in: Archives of Gynecology and Obstetrics | Issue 2/2013

Login to get access

Abstract

Purpose

We investigated the effects of the anti-epilepsy drug valproic acid (VPA) alone and in combination in treating cervical cancer.

Methods

VPA was investigated for its effects on cervical cancer Hela cell proliferation and tumor growth via in vitro and in vivo assays.

Results

VPA induce cell growth suppression and cell cycle arrest, with an increase of Notch1 that acts as a tumor suppressor and the change of other tumor-associated genes such as p21, p63 and PCNA. VPA was also found to induce cell morphological change, with an increase of certain cell transformation markers such as snail1, snail2 and N-cadherin. Moreover, VPA could significantly up-regulate somatostatin receptor type II (SSTR2). Our in vivo study further demonstrated that VPA via inducing SSTR2 up-regulation extremely enhanced the anti-tumor ability of the SSTR2-preferential cytotoxic COL-SST conjugate in xenografts.

Conclusions

VPA could not only suppress tumor progression but also provide a novel promising therapeutic choice in combination with a receptor-targeted cytotoxic conjugate via activating the specific receptor.
Literature
1.
go back to reference Maillard I, Pear WS (2003) Notch and cancer: best to avoid the ups and downs. Cancer Cell 3:203–205PubMedCrossRef Maillard I, Pear WS (2003) Notch and cancer: best to avoid the ups and downs. Cancer Cell 3:203–205PubMedCrossRef
3.
4.
go back to reference Talora C, Cialfi S, Segatto O (2005) Constitutively active Notch1 induces growth arrest of HPV-positive cervical cancer cells via separate signaling pathways. Exp Cell Res 305:343–354PubMedCrossRef Talora C, Cialfi S, Segatto O (2005) Constitutively active Notch1 induces growth arrest of HPV-positive cervical cancer cells via separate signaling pathways. Exp Cell Res 305:343–354PubMedCrossRef
5.
go back to reference Franko-Tobin LG, Mackey LV, Huang W (2011) Notch1-mediated tumor suppression in cervical cancer with the involvement of sst signaling and its application in enhanced SSTR-targeted therapeutics. Oncologist 17:220–232CrossRef Franko-Tobin LG, Mackey LV, Huang W (2011) Notch1-mediated tumor suppression in cervical cancer with the involvement of sst signaling and its application in enhanced SSTR-targeted therapeutics. Oncologist 17:220–232CrossRef
6.
go back to reference Kostrouchová M, Kostrouch Z, Kostrouchová M (2007) Valproic acid, a molecular lead to multiple regulatory pathways. Folia Biol (Praha) 53:37–49 Kostrouchová M, Kostrouch Z, Kostrouchová M (2007) Valproic acid, a molecular lead to multiple regulatory pathways. Folia Biol (Praha) 53:37–49
7.
go back to reference Platta CS, Greenblatt DY, Kunnimalaiyaan M (2008) Valproic acid induces Notch1 signaling in small cell lung cancer cells. J Surg Res 148:31–37PubMedCrossRef Platta CS, Greenblatt DY, Kunnimalaiyaan M (2008) Valproic acid induces Notch1 signaling in small cell lung cancer cells. J Surg Res 148:31–37PubMedCrossRef
8.
go back to reference Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C (2008) Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors. Cancer Treat Rev 34:206–222PubMedCrossRef Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C (2008) Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors. Cancer Treat Rev 34:206–222PubMedCrossRef
9.
go back to reference Shabbeer S, Kortenhorst MS, Kachhap S, Galloway N, Rodriguez R, Carducci MA (2007) Multiple molecular pathways explain the anti-proliferative effect of valproic acid on prostate cancer cells in vitro and in vivo. Prostate 67:1099–1110PubMedCrossRef Shabbeer S, Kortenhorst MS, Kachhap S, Galloway N, Rodriguez R, Carducci MA (2007) Multiple molecular pathways explain the anti-proliferative effect of valproic acid on prostate cancer cells in vitro and in vivo. Prostate 67:1099–1110PubMedCrossRef
10.
go back to reference Adler JT, Hottinger DG, Kunnimalaiyaan M (2010) Inhibition of growth in medullary thyroid cancer cells with histone deacetylase inhibitors and lithium chloride. J Surg Res 159:640–644PubMedCrossRef Adler JT, Hottinger DG, Kunnimalaiyaan M (2010) Inhibition of growth in medullary thyroid cancer cells with histone deacetylase inhibitors and lithium chloride. J Surg Res 159:640–644PubMedCrossRef
11.
go back to reference Sami S, Höti N, Xu HM, Shen Z, Huang X (2008) Valproic acid inhibits the growth of cervical cancer both in vitro and in vivo. J Biochem 144:357–362PubMedCrossRef Sami S, Höti N, Xu HM, Shen Z, Huang X (2008) Valproic acid inhibits the growth of cervical cancer both in vitro and in vivo. J Biochem 144:357–362PubMedCrossRef
12.
go back to reference Sun L, Coy DH (2011) Somatostatin receptor-targeted anti-cancer therapy. Cur Drug Deliv 8:2–10CrossRef Sun L, Coy DH (2011) Somatostatin receptor-targeted anti-cancer therapy. Cur Drug Deliv 8:2–10CrossRef
13.
go back to reference Sun L, Luo J, Mackey VL (2007) Effects of camptothecin on tumor cell proliferation and angiogenesis when coupled to a bombesin analog used as a targeted delivery vector. Anticancer Drugs 18:341–348PubMedCrossRef Sun L, Luo J, Mackey VL (2007) Effects of camptothecin on tumor cell proliferation and angiogenesis when coupled to a bombesin analog used as a targeted delivery vector. Anticancer Drugs 18:341–348PubMedCrossRef
14.
go back to reference Eastham AM, Spencer H, Soncin F (2007) Epithelial-mesenchymal transition events during human embryonic stem cell differentiation. Cancer Res 67:11254–11262PubMedCrossRef Eastham AM, Spencer H, Soncin F (2007) Epithelial-mesenchymal transition events during human embryonic stem cell differentiation. Cancer Res 67:11254–11262PubMedCrossRef
15.
go back to reference Kato Y, Yashiro M, Noda S (2010) Establishment and characterization of a new hypoxia-resistant cancer cell line, OCUM-12/Hypo, derived from a scirrhous gastric carcinoma. Br J Cancer 102:898–907PubMedCrossRef Kato Y, Yashiro M, Noda S (2010) Establishment and characterization of a new hypoxia-resistant cancer cell line, OCUM-12/Hypo, derived from a scirrhous gastric carcinoma. Br J Cancer 102:898–907PubMedCrossRef
16.
go back to reference Forino M, Torregrossa R, Ceol M (2006) TGFb1 induces epithelial–mesenchymal transition, but not myofibroblast transdifferentiation of human kidney tubular epithelial cells in primary culture. Int J Exp Pathol 87:197–208PubMedCrossRef Forino M, Torregrossa R, Ceol M (2006) TGFb1 induces epithelial–mesenchymal transition, but not myofibroblast transdifferentiation of human kidney tubular epithelial cells in primary culture. Int J Exp Pathol 87:197–208PubMedCrossRef
17.
go back to reference Lossi L, Gambino G, Ferrini F (2009) Posttranslational regulation of BCL2 levels in cerebellar granule cells: a mechanism of neuronal survival. Dev Neurobiol 69:855–870PubMedCrossRef Lossi L, Gambino G, Ferrini F (2009) Posttranslational regulation of BCL2 levels in cerebellar granule cells: a mechanism of neuronal survival. Dev Neurobiol 69:855–870PubMedCrossRef
18.
go back to reference Higashikawa K, Yoneda S, Tobiume K (2007) Snail-induced down-regulation of DeltaNp63alpha acquires invasive phenotype of human squamous cell carcinoma. Cancer Res 67:9207–9213PubMedCrossRef Higashikawa K, Yoneda S, Tobiume K (2007) Snail-induced down-regulation of DeltaNp63alpha acquires invasive phenotype of human squamous cell carcinoma. Cancer Res 67:9207–9213PubMedCrossRef
19.
go back to reference Gagnon V, Mathieu I, Sexton É (2004) AKT involvement in cisplatin chemoresistance of human uterine cancer cells. Gynecol Oncol 94:785–795PubMedCrossRef Gagnon V, Mathieu I, Sexton É (2004) AKT involvement in cisplatin chemoresistance of human uterine cancer cells. Gynecol Oncol 94:785–795PubMedCrossRef
20.
go back to reference Georgeseu MM (2010) PTEN Tumor Suppressor Network in PI3K-Akt Pathway Control. Genes Cancer 1:1170–1177CrossRef Georgeseu MM (2010) PTEN Tumor Suppressor Network in PI3K-Akt Pathway Control. Genes Cancer 1:1170–1177CrossRef
21.
go back to reference Thiery JP (2002) Epithelial-mesenchymal transitions in tumor progression. Nat Rev Cancer 2:442–454PubMedCrossRef Thiery JP (2002) Epithelial-mesenchymal transitions in tumor progression. Nat Rev Cancer 2:442–454PubMedCrossRef
22.
go back to reference Reubi JC (2003) Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocr Rev 24:389–427PubMedCrossRef Reubi JC (2003) Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocr Rev 24:389–427PubMedCrossRef
Metadata
Title
Valproic acid suppresses cervical cancer tumor progression possibly via activating Notch1 signaling and enhances receptor-targeted cancer chemotherapeutic via activating somatostatin receptor type II
Authors
Cheguo Tsai
Juliana S. Leslie
Laura G. Franko-Tobin
Monica C. Prasnal
Tong Yang
L. Vienna Mackey
Joseph A. Fuselier
David H. Coy
Mingqiu Liu
Changyuan Yu
Lichun Sun
Publication date
01-08-2013
Publisher
Springer Berlin Heidelberg
Published in
Archives of Gynecology and Obstetrics / Issue 2/2013
Print ISSN: 0932-0067
Electronic ISSN: 1432-0711
DOI
https://doi.org/10.1007/s00404-013-2762-7

Other articles of this Issue 2/2013

Archives of Gynecology and Obstetrics 2/2013 Go to the issue