Molecular and therapeutic potential and toxicity of valproic acid

Sébastien Chateauvieux, Franck Morceau, Mario Dicato, Marc Diederich, Sébastien Chateauvieux, Franck Morceau, Mario Dicato, Marc Diederich

Abstract

Valproic acid (VPA), a branched short-chain fatty acid, is widely used as an antiepileptic drug and a mood stabilizer. Antiepileptic properties have been attributed to inhibition of Gamma Amino Butyrate (GABA) transaminobutyrate and of ion channels. VPA was recently classified among the Histone Deacetylase Inhibitors, acting directly at the level of gene transcription by inhibiting histone deacetylation and making transcription sites more accessible. VPA is a widely used drug, particularly for children suffering from epilepsy. Due to the increasing number of clinical trials involving VPA, and interesting results obtained, this molecule will be implicated in an increasing number of therapies. However side effects of VPA are substantially described in the literature whereas they are poorly discussed in articles focusing on its therapeutic use. This paper aims to give an overview of the different clinical-trials involving VPA and its side effects encountered during treatment as well as its molecular properties.

Figures

Figure 1
Figure 1
(a) Structure of Valproic acid, (b) structure of valeric acid, (c) valeriana officinalis (valeriana officinalis in an early stage of flowering, [Belgium] Photo by Maarten. © 2007 Erowid.org: http://www.erowid.org/herbs/show_image.php?i=valerian/valeriana_officinalis_flower__i2005e1334_disp.jpg).
Figure 2
Figure 2
Pharmacological activity of VPA described in the literature. Schematic representation of direct and indirect targets of VPA. Principal direct targets known for VPA are ionic channels and ABAT (in green). Epigenetic action of VPA (in violet) as HDACi activity: VPA targets the transcriptomic system and principally directly inhibits HDAC class I (subcategories 1, 2, and 3), and less strongly class II/a (subcategories 4, 5, and 7), but induces HDAC 9 and 11, and indirectly inhibits the function of SMC and DNMT. Probably due to its epigenetic properties, or interactions not yet established, VPA alters, directly or indirectly, expression of many molecules involved in molecular pathways such as apoptosis, inflammation, differentiation, and proliferation (in red).

References

    1. Burton B. On the propyl derivatives and decomposition products of ethylacetoacetate. American Chemical Journal. 1882;3:385–395.
    1. Meunier H, Carraz G, Neunier Y, Eymard P, Aimard M. Pharmacodynamic properties of N-dipropylacetic acid. Thérapie. 1963;18:435–438.
    1. Lebreton S, Carraz G, Behriel H, Meunier H. Pharmacodynamic properties of 2,2-dipropylacetic Acid. III. Therapie. 1984;19:457–467.
    1. Lebreton S, Carraz G, Meunier H, Behriel H. Pharmacodynamic properties of 2,2-dipropylacetic Acid. 2d report on its anti-epileptic properties. Therapie. 1964;19:451–456.
    1. Mesdjian E, Ciesielski L, Valli M. Sodium valproate: kinetic profile and effects on GABA levels in various brain areas of the rat. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 1982;6(3):223–233.
    1. Zeise ML, Kasparow S, Zieglgansberger W. Valproate suppresses N-methyl-D-aspartate-evoked transient depolarizations in the rat neocortex in vitro. Brain Research. 1991;544(2):345–348.
    1. Gean P-W, Huang C-C, Hung C-R, Tsai J-J. Valproic acid suppresses the synaptic response mediated by the NMDA receptors in rat amygdalar slices. Brain Research Bulletin. 1994;33(3):333–336.
    1. VanDongen AMJ, VanErp MG, Voskuyl RA. Valproate reduces excitability by blockage of sodium and potassium conductance. Epilepsia. 1986;27(3):177–182.
    1. McElroy SL, Keck PE, Jr., Pope HG, Jr., Hudson JI. Valproate in psychiatric disorders: literature review and clinical guidelines. Journal of Clinical Psychiatry. 1989;50(supplement):23–29.
    1. Calabrese JR, Delucchi GA. Phenomenology of rapid cycling manic depression and its treatment with valproate. Journal of Clinical Psychiatry. 1989;(supplement):30–34.
    1. Erenberg G, Rothner AD, Henry CE, Cruse RP. Valproic acid in the treatment of intractable absence seizures in children. A single-blind clinical and quantitative EEG study. American Journal of Diseases of Children. 1982;136(6):526–529.
    1. Coppola G, Auricchio G, Federico R, Carotenuto M, Pascotto A. Lamotrigine versus valproic acid as first-line monotherapy in newly diagnosed typical absence seizures: an open-label, randomized, parallel-group study. Epilepsia. 2004;45(9):1049–1053.
    1. Dean JC, Penry JK. Valproate monotherapy in 30 patients with partial seizures. Epilepsia. 1988;29(2):140–144.
    1. Calleja S, Salas-Puig J, Ribacoba R, Lahoz CH. Evolution of juvenile myoclonic epilepsy treated from the outset with sodium valproate. Seizure. 2001;10(6):424–427.
    1. Friis ML. Valproate in the treatment of epilepsy in people with intellectual disability. Journal of Intellectual Disability Research. 1998;42(supplement 1):32–35.
    1. Rosato RR, Almenara JA, Grant S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1. Cancer Research. 2003;63(13):3637–3645.
    1. Martirosyan A, Leonard S, Shi X, Griffith B, Gannett P, Strobl J. Actions of a histone deacetylase inhibitor NSC3852 (5-nitroso-8-quinolinol) link reactive oxygen species to cell differentiation and apoptosis in MCF-7 human mammary tumor cells. Journal of Pharmacology and Experimental Therapeutics. 2006;317(2):546–552.
    1. Savickiene J, Borutinskaite V-V, Treigyte G, Magnusson K-E, Navakauskiene R. The novel histone deacetylase inhibitor BML-210 exerts growth inhibitory, proapoptotic and differentiation stimulating effects on the human leukemia cell lines. European Journal of Pharmacology. 2006;549(1–3):9–18.
    1. Chen Y, Pan R-L, Zhang X-L, et al. Induction of hepatic differentiation of mouse bone marrow stromal stem cells by the histone deacetylase inhibitor VPA. Journal of Cellular and Molecular Medicine. 2009;13(8 B):2582–2592.
    1. Bradbury CA, Khanim FL, Hayden R, et al. Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia. 2005;19(10):1751–1759.
    1. Bhattacharya SK, Ramchandani S, Cervoni N, Szyf M. A mammalian protein with specific demethylase activity for mCpG DNA. Nature. 1999;397(6720):579–583.
    1. Ramchandani S, Bhattacharya SK, Cervoni N, Szyf M. DNA methylation is a reversible biological signal. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(11):6107–6112.
    1. Cervoni N, Szyf M. Demethylase activity is directed by histone acetylation. Journal of Biological Chemistry. 2001;276(44):40778–40787.
    1. Cervoni N, Detich N, Seo S-B, Chakravarti D, Szyf M. The oncoprotein set/TAF-1β, an inhibitor of histone acetyltransferase, inhibits active demethylation of DNA, integrating DNA methylation and transcriptional silencing. Journal of Biological Chemistry. 2002;277(28):25026–25031.
    1. Marchion DC, Bicaku E, Daud AI, Sullivan DM, Munster PN. Valproic acid alters chromatin structure by regulation of chromatin modulation proteins. Cancer Research. 2005;65(9):3815–3822.
    1. Nightingale KP, Gendreizig S, White DA, Bradbury C, Hollfelder F, Turner BM. Cross-talk between histone modifications in response to histone deacetylase inhibitors: MLL4 links histone H3 acetylation and histone H3K4 methylation. Journal of Biological Chemistry. 2007;282(7):4408–4416.
    1. Harikrishnan KN, Karagiannis TC, Chow MZ, El-Osta A. Effect of valproic acid on radiation-induced DNA damage in euchromatic and heterochromatic compartments. Cell Cycle. 2008;7(4):468–476.
    1. Lunke S, El-Osta A. The emerging role of epigenetic modifications and chromatin remodeling in spinal muscular atrophy. Journal of Neurochemistry. 2009;109(6):1557–1569.
    1. Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nature Reviews Cancer. 2006;6(1):38–51.
    1. Pastorelli R, Ferrari G, Gozzini A. CML blasts modify the acetylation pattern of non histone proteins after short chain fatty acid histone deacetylase inhibitor treatment. The American Society of Hematology. 2005;106(11):p. 2884.
    1. May T, Rambeck B. Serum concentrations of valproic acid: influence of dose and comedication. Therapeutic Drug Monitoring. 1985;7(4):387–390.
    1. Hamilton RA, Garnett WR, Kline BJ, Pellock JM. Effects of food on valproic acid absorption. American Journal of Hospital Pharmacy. 1981;38(10):1490–1493.
    1. Lefebvre S, Burlet P, Liu Q, et al. Correlation between severity and SMN protein level in spinal muscular atrophy. Nature Genetics. 1997;16(3):265–269.
    1. Pan T, Li X, Xie W, Jankovic J, Le W. Valproic acid-mediated Hsp70 induction and anti-apoptotic neuroprotection in SH-SY5Y cells. FEBS Letters. 2005;579(30):6716–6720.
    1. Chen P-S, Peng G-S, Li G, et al. Valproate protects dopaminergic neurons in midbrain neuron/glia cultures by stimulating the release of neurotrophic factors from astrocytes. Molecular Psychiatry. 2006;11(12):1116–1125.
    1. Chen PS, Wang C-C, Bortner CD, et al. Valproic acid and other histone deacetylase inhibitors induce microglial apoptosis and attenuate lipopolysaccharide-induced dopaminergic neurotoxicity. Neuroscience. 2007;149(1):203–212.
    1. Monti B, Polazzi E, Batti L, Crochemore C, Virgili M, Contestabile A. Alpha-synuclein protects cerebellar granule neurons against 6-hydroxydopamine-induced death. Journal of Neurochemistry. 2007;103(2):518–530.
    1. Monti B, Gatta V, Piretti F, Raffaelli SS, Virgili M, Contestabile A. Valproic acid is neuroprotective in the rotenone rat model of Parkinson’s disease: involvement of α-synuclein. Neurotoxicity Research. 2009;17(2):130–140.
    1. Pearce I, Heathfield KWG, Pearce JMS. Valproate sodium in Huntington chorea. Archives of Neurology. 1977;34(5):308–309.
    1. Zádori D, Geisz A, Vámos E, Vécsei L, Klivényi P. Valproate ameliorates the survival and the motor performance in a transgenic mouse model of Huntington’s disease. Pharmacology Biochemistry and Behavior. 2009;94(1):148–153.
    1. Armeanu S, Bitzer M, Lauer UM, et al. Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Research. 2005;65(14):6321–6329.
    1. Kristensen Ø, Lølandsmo T, Isaksen Å, Vederhus J-K, Clausen T. Treatment of polydrug-using opiate dependents during withdrawal: towards a standardisation of treatment. BMC Psychiatry. 2006;6, article no. 54
    1. Lehrman G, Hogue IB, Palmer S, et al. Depletion of latent HIV-1 infection in vivo: a proof-of-concept study. Lancet. 2005;366(9485):549–555.
    1. Sagot-Lerolle N, Lamine A, Chaix M-L, et al. Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS. 2008;22(10):1125–1129.
    1. Gurpur PB, Liu J, Burkin DJ, Kaufman SJ. Valproic acid activates the PI3K/Akt/mTOR pathway in muscle and ameliorates pathology in a mouse model of Duchenne muscular dystrophy. American Journal of Pathology. 2009;174(3):999–1008.
    1. Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. Journal of the National Cancer Institute. 2000;92(15):1210–1216.
    1. Melnick A, Licht JD. Histone deacetylases as therapeutic targets in hematologic malignancies. Current Opinion in Hematology. 2002;9(4):322–332.
    1. Vigushin DM, Coombes RC. Histone deacetylase inhibitors in cancer treatment. Anti-Cancer Drugs. 2002;13(1):1–13.
    1. Cheng Y-C, Lin H, Huang M-J, Chow J-M, Lin S, Liu HE. Downregulation of c-Myc is critical for valproic acid-induced growth arrest and myeloid differentiation of acute myeloid leukemia. Leukemia Research. 2007;31(10):1403–1411.
    1. Schuchmann M, Schulze-Bergkamen H, Fleischer B, et al. Histone deacetylase inhibition by valproic acid down-regulates c-FLIP/CASH and sensitizes hepatoma cells towards CD95- and TRAIL receptor-mediated apoptosis and chemotherapy. Oncology Reports. 2006;15(1):227–230.
    1. Ziauddin MF, Yeow W-S, Maxhimer JB, et al. Valproic acid, an antiepileptic drug with histone deacetylase inhibitory activity, potentiates the cytotoxic effect of Apo2L/TRAIL on cultured thoracic cancer cells through mitochondria-dependent caspase activation. Neoplasia. 2006;8(6):446–457.
    1. Lagneaux L, Gillet N, Stamatopoulos B, et al. Valproic acid induces apoptosis in chronic lymphocytic leukemia cells through activation of the death receptor pathway and potentiates TRAIL response. Experimental Hematology. 2007;35(10):1527–1537.
    1. Iacomino G, Medici MC, Russo GL. Valproic acid sensitizes K562 erythroleukemia cells to TRAIL/Apo2L-induced apoptosis. Anticancer Research. 2008;28(2A):855–864.
    1. Phillips A, Bullock T, Plant N. Sodium valproate induces apoptosis in the rat hepatoma cell line, FaO. Toxicology. 2003;192(2-3):219–227.
    1. Angelucci A, Valentini A, Millimaggi D, et al. Valproic acid induces apoptosis in prostate carcinoma cell lines by activation of multiple death pathways. Anti-Cancer Drugs. 2006;17(10):1141–1150.
    1. Chen G, Huang L-D, Jiang Y-M, Manji HK. The mood-stabilizing agent valproate inhibits the activity of glycogen synthase kinase-3. Journal of Neurochemistry. 1999;72(3):1327–1330.
    1. Marinova Z, Ren M, Wendland JR, et al. Valproic acid induces functional heat-shock protein 70 via Class I histone deacetylase inhibition in cortical neurons: a potential role of Sp1 acetylation. Journal of Neurochemistry. 2009;111(4):976–987.
    1. Greenblatt DY, Cayo MA, Adler JT, et al. Valproic acid activates Notch1 signaling and induces apoptosis in medullary thyroid cancer cells. Annals of Surgery. 2008;247(6):1036–1040.
    1. Lampen A, Siehler S, Ellerbeck U, Göttlicher M, Nau H. New molecular bioassays for the estimation of the teratogenic potency of valproic acid derivatives in vitro: activation of the peroxisomal proliferator-activated receptor (PPARδ) Toxicology and Applied Pharmacology. 1999;160(3):238–249.
    1. Werling U, Siehler S, Litfin M, Nau H, Göttlicher M. Induction of differentiation in F9 cells and activation of peroxisome proliferator-activated receptor δ by valproic acid and its teratogenic derivatives. Molecular Pharmacology. 2001;59(5):1269–1276.
    1. Lan MJ, Yuan P, Chen G, Manji HK. Neuronal peroxisome proliferator-activated receptor γ signaling: regulation by mood-stabilizer valproate. Journal of Molecular Neuroscience. 2008;35(2):225–234.
    1. Togi S, Kamitani S, Kawakami S, et al. HDAC3 influences phosphorylation of STAT3 at serine 727 by interacting with PP2A. Biochemical and Biophysical Research Communications. 2009;379(2):616–620.
    1. Snyder M, Huang X-Y, Zhang JJ. Identification of novel direct Stat3 target genes for control of growth and differentiation. Journal of Biological Chemistry. 2008;283(7):3791–3798.
    1. Hallas J, Friis S, Bjerrum L, et al. Cancer risk in long-term users of valproate: a population-based case-control study. Cancer Epidemiology Biomarkers and Prevention. 2009;18(6):1714–1719.
    1. Citrome L, Shope CB, Nolan KA, Czobor P, Volavka J. Risperidone alone versus risperidone plus valproate in the treatment of patients with schizophrenia and hostility. International Clinical Psychopharmacology. 2007;22(6):356–362.
    1. Glick ID, Bosch J, Casey DE. A double-blind randomized trial of mood stabilizer augmentation using lamotrigine and valproate for patients with schizophrenia who are stabilized and partially responsive. Journal of Clinical Psychopharmacology. 2009;29(3):267–271.
    1. Ashrafi MR, Shabanian R, Zamani GR, Mahfelati F. Sodium Valproate versus Propranolol in paediatric migraine prophylaxis. European Journal of Paediatric Neurology. 2005;9(5):333–338.
    1. Lonergan E, Luxenberg J. Valproate preparations for agitation in dementia. Cochrane Database of Systematic Reviews. 2009;(3) Article ID CD003945.
    1. Novick D, Gonzalez-Pinto A, Haro JM, et al. Translation of randomised controlled trial findings into clinical practice: comparison of olanzapine and valproate in the EMBLEM study. Pharmacopsychiatry. 2009;42(4):145–152.
    1. Smith LA, Cornelius VR, Azorin JM, et al. Valproate for the treatment of acute bipolar depression: systematic review and meta-analysis. Journal of Affective Disorders. 2009;122(1):1–9.
    1. Piepers S, Veldink JH, de Jong SW, et al. Randomized sequential trial of valproic acid in amyotrophic lateral sclerosis. Annals of Neurology. 2009;66(2):227–234.
    1. Swoboda KJ, Scott CB, Reyna SP, et al. Phase II open label study of valproic acid in spinal muscular atrophy. PLoS ONE. 2009;4(5) Article ID e5268.
    1. Agrawal RP, Goswami J, Jain S, Kochar DK. Management of diabetic neuropathy by sodium valproate and glyceryl trinitrate spray: a prospective double-blind randomized placebo-controlled study. Diabetes Research and Clinical Practice. 2009;83(3):371–378.
    1. Moog C, Kuntz-Simon G, Caussin-Schwemling C, Obert G. Sodium valproate, an anticonvulsant drug, stimulates human immunodeficiency virus type 1 replication independently of glutathione levels. Journal of General Virology. 1996;77, part 9:1993–1999.
    1. Witvrouw M, Schmit J-C, Van Remoortel B, et al. Cell type-dependent effect of sodium valproate on human immunodeficiency virus type 1 replication in vitro. AIDS Research and Human Retroviruses. 1997;13(2):187–192.
    1. Shaw RN, Arbiser JL, Offermann MK. Valproic acid induces human herpesvirus 8 lytic gene expression in BCBL-1 cells. AIDS. 2000;14(7):899–902.
    1. Lechowicz M, Dittmer DP, Lee JY, et al. Molecular and clinical assessment in the treatment of AIDS Kaposi sarcoma with valproic acid. Clinical Infectious Diseases. 2009;49(12):1946–1949.
    1. Khanim FL, Bradbury CA, Arrazi J, et al. Elevated FOSB-expression; a potential marker of valproate sensitivity in AML. British Journal of Haematology. 2009;144(3):332–341.
    1. Kuendgen A, Strupp C, Aivado M, et al. Treatment of myelodysplastic syndromes with valproic acid alone or in combination with all-trans retinoic acid. Blood. 2004;104(5):1266–1269.
    1. Bug G, Ritter M, Wassmann B, et al. Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer. 2005;104(12):2717–2725.
    1. Inoue Y, Suzuki T, Takimoto M, et al. Treatment with valproic acid for myelofibrosis with myeloid metaplasia. Annals of Hematology. 2005;84(12):833–834.
    1. Kuendgen A, Knipp S, Fox F, et al. Results of a phase 2 study of valproic acid alone or in combination with all-trans retinoic acid in 75 patients with myelodysplastic syndrome and relapsed or refractory acute myeloid leukemia. Annals of Hematology, Supplement. 2005;84(supplement 1):61–66.
    1. Kuendgen A, Schmid M, Schlenk R, et al. The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer. 2006;106(1):112–119.
    1. Soriano AO, Yang H, Faderl S, et al. Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood. 2007;110(7):2302–2308.
    1. Voso MT, Santini V, Finelli C, et al. Valproic acid at therapeutic plasma levels may increase 5-azacytidine efficacy in higher risk myelodysplastic syndromes. Clinical Cancer Research. 2009;15(15):5002–5007.
    1. Stamatopoulos B, Meuleman N, De Bruyn C, et al. Antileukemic activity of valproic acid in chronic lymphocytic leukemia B cells defined by microarray analysis. Leukemia. 2009;23:2281–2289.
    1. Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C, Perez-Cardenas E, de la Cruz-Hernandez E, Herrera LA. Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors. Cancer Treatment Reviews. 2008;34(3):206–222.
    1. Chavez-Blanco A, Perez-Plasencia C, Perez-Cardenas E, et al. Antineoplastic effects of the DNA methylation inhibitor hydralazine and the histone deacetylase inhibitor valproic acid in cancer cell lines. Cancer Cell International. 2006;6, article 2
    1. Chen C-L, Sung J, Cohen M, et al. Valproic acid inhibits invasiveness in bladder cancer but not in prostate cancer cells. Journal of Pharmacology and Experimental Therapeutics. 2006;319(2):533–542.
    1. Thelen P, Schweyer S, Hemmerlein B, Wuttke W, Seseke F, Ringert RH. Expressional changes after histone deacetylase inhibition by valproic acid in LNCaP human prostate cancer cells. International Journal of Oncology. 2004;24(1):25–31.
    1. Xia Q, Sung J, Chowdhury W, et al. Chronic administration of valproic acid inhibits prostate cancer cell growth in vitro and in vivo. Cancer Research. 2006;66(14):7237–7244.
    1. Huang X, Guo B. Adenomatous polyposis coli determines sensitivity to histone deacetylase inhibitor-induced apoptosis in colon cancer cells. Cancer Research. 2006;66(18):9245–9251.
    1. Friedmann I, Atmaca A, Chow KU, Jäger E, Weidmann E. Synergistic effects of valproic acid and mitomycin C in adenocarcinoma cell lines and fresh tumor cells of patients with colon cancer. Journal of Chemotherapy. 2006;18(4):415–420.
    1. Olsen CM, Meussen-Elholm ETM, Røste LS, Taubøll E. Antiepileptic drugs inhibit cell growth in the human breast cancer cell line MCF7. Molecular and Cellular Endocrinology. 2004;213(2):173–179.
    1. Mongan NP, Gudas LJ. Valproic acid, in combination with all-trans retinoic acid and 5-aza-2’-deoxycytidine, restores expression of silenced RARβ2 in breast cancer cells. Molecular Cancer Therapeutics. 2005;4(3):477–486.
    1. Hodges-Gallagher L, Valentine CD, Bader SE, Kushner PJ. Inhibition of histone deacetylase enhances the anti-proliferative action of antiestrogens on breast cancer cells and blocks tamoxifen-induced proliferation of uterine cells. Breast Cancer Research and Treatment. 2007;105(3):297–309.
    1. Pathil A, Armeanu S, Venturelli S, et al. HDAC inhibitor treatment of hepatoma cells induces both TRAIL-independent apoptosis and restoration of sensitivity to TRAIL. Hepatology. 2006;43(3):425–434.
    1. Cinatl J, Jr., Cinatl J, Scholz M, et al. Antitumor activity of sodium valproate in cultures of human neuroblastoma cells. Anti-Cancer Drugs. 1996;7(7):766–773.
    1. Michaelis M, Suhan T, Cinatl J, Driever PH, Cinatl J., Jr. Valproic acid and interferon-alpha synergistically inhibit neuroblastoma cell growth in vitro and in vivo. International Journal of Oncology. 2004;25(6):1795–1799.
    1. Bacon CL, O’Driscoll E, Regan CM. Valproic acid suppresses G1 phase-dependent sialylation of a 65 kDa glycoprotein in the C6 glioma cell cycle. International Journal of Developmental Neuroscience. 1997;15(6):777–784.
    1. Das CM, Aguilera D, Vasquez H, et al. Valproic acid induces p21 and topoisomerase-II (α/β) expression and synergistically enhances etoposide cytotoxicity in human glioblastoma cell lines. Journal of Neuro-Oncology. 2007;85(2):159–170.
    1. Catalano MG, Fortunati N, Pugliese M, et al. Valproic acid induces apoptosis and cell cycle arrest in poorly differentiated thyroid cancer cells. Journal of Clinical Endocrinology and Metabolism. 2005;90(3):1383–1389.
    1. Atmaca A, Al-Batran S-E, Maurer A, et al. Valproic acid (VPA) in patients with refractory advanced cancer: a dose escalating phase I clinical trial. British Journal of Cancer. 2007;97(2):177–182.
    1. Arce C, Pérez-Plasencia C, González-Fierro A, et al. A proof-of-principle study of epigenetics therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer. PLoS One. 2006;1(1, article e98)
    1. Munster P, Marchion D, Bicaku E, et al. Clinical and biological effects of valproic acid as a histone deacetylase inhibitor on tumor and surrogate tissues: phase I/II trial of valproic acid and epirubicin/FEC. Clinical Cancer Research. 2009;15(7):2488–2496.
    1. Daud AI, Dawson J, DeConti RC, et al. Potentiation of a topoisomerase I inhibitor, karenitecin, by the histone deacetylase inhibitor valproic acid in melanoma: translational and phase l/ll clinical trial. Clinical Cancer Research. 2009;15(7):2479–2487.
    1. Rocca A, Minucci S, Tosti G, et al. A phase I-II study of the histone deacetylase inhibitor valproic acid plus chemoimmunotherapy in patients with advanced melanoma. British Journal of Cancer. 2009;100(1):28–36.
    1. Sharma S, Symanowski J, Wong B, et al. A phase II clinical trial of oral valproic acid in patients with castration-resistant prostate cancers using an intensive biomarker sampling strategy. Translational Oncology. 2008;1(3):141–147.
    1. Yu H-Y, Shen Y-Z, Sugiyama Y, Hanano M. Drug interaction. Effects of salicylate on pharmacokinetics of valproic acid in rats. Drug Metabolism and Disposition. 1990;18(1):121–126.
    1. Battino D, Estienne M, Avanzini G. Clinical pharmacokinetics of antiepileptic drugs in paediatric patients. Part I: phenobarbital, primidone, valproic acid, ethosuximide and mesuximide. Clinical Pharmacokinetics. 1995;29(4):257–286.
    1. Yamamura N, Imura K, Naganuma H, Nishimura K. Panipenem, a carbapenem antibiotic, enhances the glucuronidation of intravenously administered valproic acid in rats. Drug Metabolism and Disposition. 1999;27(6):724–730.
    1. Yokogawa K, Iwashita S, Kubota A, et al. Effect of meropenem on disposition kinetics of valproate and its metabolites in rabbits. Pharmaceutical Research. 2001;18(9):1320–1326.
    1. Rambeck B, Wolf P. Lamotrigine clinical pharmacokinetics. Clinical Pharmacokinetics. 1993;25(6):433–443.
    1. May TW, Rambeck B, Jürgens U. Serum concentrations of lamotrigine in epileptic patients: the influence of dose and comedication. Therapeutic Drug Monitoring. 1996;18(5):523–531.
    1. Battino D, Croci D, Granata T, Estienne M, Pisani F, Avanzini G. Lamotrigine plasma concentrations in children and adults: influence of age and associated therapy. Therapeutic Drug Monitoring. 1997;19(6):620–627.
    1. Arif H, Buchsbaum R, Weintraub D, et al. Comparison and predictors of rash associated with 15 antiepileptic drugs. Neurology. 2007;68(20):1701–1709.
    1. Levi N, Bastuji-Garin S, Mockenhaupt M, et al. Medications as risk factors of stevens-johnson syndrome and toxic epidermal necrolysis in children: a pooled analysis. Pediatrics. 2009;123(2):e297–e304.
    1. Sheorajpanday RVA, De Deyn PP. Epileptic fits and epilepsy in the elderly: general reflections, specific issues and therapeutic implications. Clinical Neurology and Neurosurgery. 2007;109(9):727–743.
    1. Longin E, Teich M, Koelfen W, König S. Topiramate enhances the risk of valproate-associated side effects in three children. Epilepsia. 2002;43(4):451–454.
    1. Gerstner T, Buesing D, Longin E, et al. Valproic acid induced encephalopathy—19 new cases in Germany from 1994 to 2003—a side effect associated to VPA-therapy not only in young children. Seizure. 2006;15(6):443–448.
    1. Nicolai J, Aldenkamp AP, Huizenga JR, Teune LK, Brouwer OF. Cognitive side effects of valproic acid-induced hyperammonemia in children with epilepsy. Journal of Clinical Psychopharmacology. 2007;27(2):221–224.
    1. Bhattacharya I, Boje KM. GHB (γ-hydroxybutyrate) carrier-mediated transport across the blood-brain barrier. Journal of Pharmacology and Experimental Therapeutics. 2004;311(1):92–98.
    1. Gerstner T, Teich M, Bell N, et al. Valproate-associated coagulopathies are frequent and variable in children. Epilepsia. 2006;47(7):1136–1143.
    1. Woyach JA, Kloos RT, Ringel MD, et al. Lack of therapeutic effect of the histone deacetylase inhibitor vorinostat in patients with metastatic radioiodine-refractory thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism. 2009;94(1):164–170.
    1. Hardy JR, Rees EAJ, Gwilliam B, Ling J, Broadley K, A’Hern R. A Phase II study to establish the efficacy and toxicity of sodium valproate in patients with cancer-related neuropathic pain. Journal of Pain and Symptom Management. 2001;21(3):204–209.
    1. Schmitt B, Martin F, Critelli H, Molinari L, Jenni OG. Effects of valproic acid on sleep in children with epilepsy. Epilepsia. 2009;50(8):1860–1867.
    1. Wirrell EC. Valproic acid-associated weight gain in older children and teens with epilepsy. Pediatric Neurology. 2003;28(2):126–129.
    1. Grosso S, Mostardini R, Piccini B, Balestri P. Body mass index and serum lipid changes during treatment with valproic acid in children with epilepsy. Annals of Pharmacotherapy. 2009;43(1):45–50.
    1. Ubeda-Martin N, Alonso-Aperte E, Achon M, Varela-Moreiras G, Puerta J, Perez de Miguelsanz J. Morphological alterations induced by valproate and its concomitant administration of folic acid or S-adenosylmethionine in pregnant rats. Nutricion Hospitalaria. 1998;13(1):41–49.
    1. Clayton-Smith J, Donnai D. Fetal valproate syndrome. Journal of Medical Genetics. 1995;32(9):724–727.
    1. Genton P, Semah F, Trinka E. Valproic acid in epilepsy: pregnancy-related issues. Drug Safety. 2006;29(1):1–21.
    1. Ornoy A. Valproic acid in pregnancy: how much are we endangering the embryo and fetus? Reproductive Toxicology. 2009;28(1):1–10.
    1. Meador KJ, Baker GA, Browning N, et al. Cognitive function at 3 years of age after fetal exposure to antiepileptic drugs. New England Journal of Medicine. 2009;360(16):1597–1605.
    1. Meador KJ, Penovich P, Baker GA, et al. Antiepileptic drug use in women of childbearing age. Epilepsy and Behavior. 2009;15(3):339–343.
    1. Varoglu AO. Na VPA-induced acute ischemic stroke in an epileptic patient with methylenetetrahydrofolate reductase gene polymorphism. Epilepsy Research. 2009;86(2-3):232–236.
    1. Buechler RD, Buchhalter JR. Juvenile Absence Epilepsy Exacerbated by Valproic Acid. Pediatric Neurology. 2007;36(2):121–124.
    1. Jamora D, Lim S-H, Pan A, Tan L, Tan E-K. Valproate-induced Parkinsonism in epilepsy patients. Movement Disorders. 2007;22(1):130–133.
    1. Dreifuss FE, Santilli N, Langer DH. Valproic acid hepatic fatalities: a retrospective review. Neurology. 1987;37(3):379–385.
    1. Dreifuss FE, Langer DH, Moline KA, Maxwell JE. Valproic acid hepatic fatalities. II. US experience since 1984. Neurology. 1989;39(2 I):201–207.
    1. Handoko KB, Souverein PC, van Staa TP, et al. Risk of aplastic anemia in patients using antiepileptic drugs. Epilepsia. 2006;47(7):1232–1236.
    1. Brenner S, Wolf R, Landau M, Politi Y. Psoriasiform eruption induced by anticonvulsants. Israel Journal of Medical Sciences. 1994;30(4):283–286.
    1. Baba M, Karakaş M, Aksungur VL, et al. The anticonvulsant hypersensitivity syndrome. Journal of the European Academy of Dermatology and Venereology. 2003;17(4):399–401.
    1. Harden CL, Sethi NK. Epileptic disorders in pregnancy: an overview. Current Opinion in Obstetrics and Gynecology. 2008;20(6):557–562.
    1. Alonso-Aperte E, Ubeda N, Achón M, Pérez-Miguelsanz J, Varela-Moreiras G. Impaired methionine synthesis and hypomethylation in rats exposed to valproate during gestation. Neurology. 1999;52(4):750–756.
    1. Kwan P, Brodie MJ. Neuropsychological effects of epilepsy and antiepileptic drugs. Lancet. 2001;357(9251):216–222.
    1. Easterford K, Clough P, Kellett M, Fallonand K, Duncan S. Reversible parkinsonism with normal beta-CIT-SPECT in patients exposed to sodium valproate. Neurology. 2004;62(8):1435–1437.
    1. Ristić AJ, Vojvodić N, Janković S, Sindelić A, Sokić D. The frequency of reversible parkinsonism and cognitive decline associated with valproate treatment: a study of 364 patients with different types of epilepsy. Epilepsia. 2006;47(12):2183–2185.
    1. Sechi GP, Conti M, Sau GF, Cocco GA. Valproate-induced parkinsonism, glial cells and Alexander's disease. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2008;32(5):1351–1352.
    1. Bryant III AE, Dreifuss FE. Valproic acid hepatic fatalities. III. U.S. experience since 1986. Neurology. 1996;46(2):465–469.
    1. Kassahun K, Farrell K, Abbott F. Identification and characterization of the glutathione and N-acetylcysteine conjugates of (E)-2-propyl-2,4-pentadienoic acid, a toxic metabolite of valproic acid, in rats and humans. Drug Metabolism and Disposition. 1991;19(2):525–535.
    1. Gopaul S, Farrell K, Abbott F. Effects of age and polytherapy, risk factors of valproic acid (VPA) hepatotoxicity, on the excretion of thiol conjugates of (E)-2,4-diene VPA in people with epilepsy taking VPA. Epilepsia. 2003;44(3):322–328.
    1. Kieslich M, Schwabe D, Cinatl J, Jr., Hernáiz Driever P. Increase of fetal hemoglobin synthesis indicating differentiation induction in children receiving valproic acid. Pediatric Hematology and Oncology. 2003;20(1):15–22.

Source: PubMed

3
Abonnieren