throbber
Hindawi Publishing Corporation
`Iournal of Biomedicine and Biotechnology
`Volume 2010, Article ID 479364, 18 pages
`d0i:10.1155/2010/479364
`
`Review Article
`
`Molecular and Therapeutic Potential and Toxicity of Valproic Acid
`
`Sébastien Chateauvieux, Franck Morceau, Mario Dicato, and Marc Diederich
`
`Laboratoire de Biologie Moléculaire et Cellulaire du Cancer {LBMCC), “Fondation de Recherche Cancer ct Sang’:
`Hépital Kirchberg, Kirchberg 2540, Luxembourg
`
`Correspondence should be addressed to Marc Diederich, marc.diederich@lbmcc.lu
`
`Received 7 January 2010; Revised 3 May 2010; Accepted 6 June 2010
`
`Academic Editor: Ronald E. Baynes
`
`Copyright © 2010 Sébastien Chateauvieux et al. This is an open access article distributed under the Creative Commons Attribution
`License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
`cited.
`
`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 Historic 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.
`
`1. Introduction
`
`Valproic acid (2—propylvaleric acid, 2—propylpentanoic acid
`or n—dipropylacetic acid)
`(see Figure 1(a)), derived from
`valeric acid (Figure 1(b)) (naturally produced by Valerian,
`Valericma oflicinalis) (see Figure 1(c)), was first synthesized
`in 1882 by Burton [1]. It is a branched short—chain fatty
`acid, forming a clear liquid at room temperature, and
`whose half—life is 9 to 16 hours. For nearly a century,
`this molecule was used as a “physiologically inert” solvent
`for organic compounds. It was in 1963, during a study
`focused on molecules with potential anti—convulsive activity,
`in which VPA was used as a molecular carrier, that the
`pharmacological activity of VPA was demonstrated: VPA
`prevented pentylenetetrazol—induced convulsions in rodents
`[2—4].
`In the human brain, VPA alters the activity of the neu-
`rotransmitter Gamma Amino Butyrate (GABA) by poten-
`tialising the inhibitory activity of GABA through several
`mechanisms,
`including inhibition of GABA degradation,
`inhibition of GABA Transaminobutyratre (ABAT), increased
`GABA synthesis, and decreased turnover [5]. Moreover, VPA
`attenuates N—Methyl—D—Aspartate—mediated excitation [6, 7]
`and blocks Na+ channels, Ca2+ channels (voltage—dependent
`
`L type CACNA1 type C, D, N, and F), and voltage—gated K+
`channels (SCN) [8].
`Besides its clinical use as an anticonvulsant and mood-
`
`stabilizing drug [9], VPA presents beneficial effects in clinical
`depression [10], absence seizures
`[11, 12],
`tonic—clonic
`seizures, complex partial seizures [13], juvenile myoclonic
`epilepsy [14], seizures associated with LennoX—Gastaut syn-
`drome [15], migraine headaches, and schizophrenia. VPA as
`a therapeutic agent is commercially available as Depakote,
`Depakote ER, Depakene, Depacon, Stavzor, Mylproin,
`Ergenyl, Dipropylacetic acid, Myproic Acid, Dipropylacetate,
`and Convulex.
`
`More recently VPA has been described as an HDAC
`inhibitor, resulting in an increased interest for its use in
`cancer therapy. Chromatin is formed of DNA packaged in
`nucleosome structures, constituted by 146 base—pair DNA
`sequence winding around an octamere of histones (two
`copies of each histone: H2A, H2B, H3, and H4) held in
`place by histone H1. The condensed form of chromatin
`(heterochromatin)
`is inactive in terms of transcription
`whereas the decondensed form (euchromatin) corresponds
`to an active form. The transition between euchromatin
`
`and heterochromatin is dependent upon two families of
`proteins: histone acetyl transferases (HATS), and histone
`
`Ranbaxy Ex. 1040
` Ranbaxy Ex. 1040
`IPR Petition - USP 9,050,302
`IPR Petition — USP 9,050,302
`
`

`
`deacetylases (HDACs). It has been established that histone
`acetylation leads to relaxation of the nucleosome structure,
`releasing the DNA and allowing transcription. Inhibition of
`HDAC promotes decondensed chromatin formation, thereby
`promoting the expression of genes.
`VPA, as well as other HDAC inhibitors (HDACi), is able
`to alter expression of many genes. Corresponding proteins
`were described to play important roles in cellular activity and
`could influence several important pathways such as cell cycle
`control, differentiation, DNA repair, and apoptosis [16—19].
`VPA specifically targets 2 of the 4 classes of HDACs:
`class I, subclasses Ia and Ib, and class II, subclass IIa. Within
`
`subclass IIa, HDAC9 is an exception to this modulation,
`being activated by VPA, which is also true for HDAC11 [20].
`HDAC 6, 8, and 10 are not modulated. It is interesting to
`mention that HDAC classes I and II have been reported to
`be strongly implicated in neuronal function, which could
`partially explain the action of VPA in neural pathologies.
`DNA methylation also contributes to the regulation of
`gene expression. Hypermethylation of the promoter, usually
`corresponding to inhibition of gene expression,
`is con-
`trolled by DNA methytransferase (DNMT). Demethylation
`of nucleic acid has been commonly associated with pas-
`sive processes corresponding to inhibition of maintenance
`methylation during S—Phase of the cell cycle. The existence
`of DNA demethylase was shown a decade ago, resulting
`in a demethylated active DNA form [21, 22]. HDACi have
`been associated with demethylation of DNA, and since 2001,
`were associated with the active demethylated form. The
`exact mechanism is not yet known, but it seems that VPA
`does not directly enhance the enzymatic activity of DNA
`demethylase. However, through HDACi activity, VPA enables
`methylated DNA to be more accessible, which is confirmed
`by the observation that inhibition of HAT diminishes the
`demethylation effect triggered by VPA [23, 24]. In addition, it
`has been shown that valproic acid downregulates expression
`of proteins essential for chromatin maintenance: SMCs 1-
`6 (Structural Maintenance of chromatin 1 to 6), DNMT1
`(DNA methyl transferase—1), and HP1 (Heterochromatin
`Protein—1) [25]. The effects upon transcription are observed
`after less than 24 hours, while 48 hours are needed to see
`the effects upon protein levels, which correlates with DNA
`decondensation (shown in breast cancer cell lines).
`Recently,
`it has been shown that VPA is also able to
`induce mono—, di—, or tri—methylation of histone 3, particu-
`larly at lysine 9 (H3K4) [20, 26-28]. Methylation of histones
`at this lysine is associated with increased transcriptional
`activity. However, this phenomenon and its purpose are not
`currently clear, considering the specific site of methylation,
`and the fact that it only occurs on already hyperacetylated
`histones, and near—demethylated genes. It is assumed that
`this modification could serve to stabilize the transiently
`released form of chromatin, mediated by histone acetylation
`[28].
`Among many drugs named as “molecular therapies,”
`epigenetic drugs are between the most encouraging, because
`in contrast to other drugs that target the expression of a
`molecule or a family of molecules, they target chromatin
`through associated proteins (HDAC, DNMT, HP1, and
`
`Iournal of Biomedicine and Biotechnology
`
`HO
`
`OH
`
`photo by Maarten.@2007 Erowid.org
`
`111111 o_fj‘lL.11zail5 flower
`
`.
`
`structure of
`(b)
`(a) Structure of Valproic acid,
`FIGURE 1:
`(c) valeriana officinalis
`(valeriana officinalis
`in
`valeric acid,
`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).
`
`SMCs). Thus, epigenetic drugs affect the expression of many
`proteins and therefore may be applicable to a wide range of
`pathologies, especially cancer, where multiple antioncogens
`are repressed during carcinogenesis. Epigenetic drugs could
`particularly target these repressed tumor suppressor genes.
`Moreover, given that
`the balance of acetylation and
`deacetylation, under the control of HAT and HDAC,
`is
`not restricted to histones [29], it can be hypothesized that
`VPA, like other HDACi, could modulate molecular activity
`in addition to transcription. Targeted genes could be Ku
`(releasing BAX), STAT3, HSP90, p53, and various transcrip-
`tion factors. Candidates have already been mentioned in a
`preliminary study in 2005 [30].
`it has not
`For all the signaling pathways modulated,
`been established if VPA acts through epigenetic regulation,
`inhibition of acetylation of molecules other than histones, or
`by other molecular mechanisms.
`
`

`
`Iournal of Biomedicine and Biotechnology
`
`2. VPA Targets a Wide Range of Pathologies
`
`Valproic acid used in therapy is available in many formu-
`lations: syrup, suppositories, tablets, or locale injection; the
`different formulations can affect the bioavailability and rate
`of absorption of the molecule. Classically age or weight
`has no influence in VPA serum concentration. There is no
`
`proportional relationship between the dose administrated
`and the serum concentration [31]. Diet plays also a role in the
`rate of absorption as VPA is more rapidly bioavailable when
`ingested before feeding. Finally,
`the serum concentration
`can be strongly influenced by combination with pheny-
`toin (49.5%), carbamazepine (66.2%), or phenobarbital
`(76.3%) than when given alone (100%). On average, 250 mg
`VPA ingested induce a serum concentration of 54.6 ug/mL
`(0.34 mM) [32]. Overdosage of VPA results in somnolence,
`heart block or deep coma.
`
`2.1. Neurological Diseases. Many of the neurodegenerative
`diseases identified to day have genetic causes. Spinal muscular
`atrophy (SMA) is caused by the homozygous loss of the
`SMN1 gene (survival motor neuron protein). The effects of
`the loss of this gene could be modulated by expression of
`the Fl—SMN2 protein (Full Length), which determines the
`severity of the disease [33]. VPA is a promising candidate
`for Fl—SMN2—overexpression therapies, because it has been
`shown that it is able to increase both SMN transcript and
`protein levels in SMA patients. However, the specificity of
`target of several HDACi members decreases their effective-
`ness and could lead to the choice of less specific molecules,
`such as SAHA [28].
`Parkinson’s disease (PD) is caused by the degeneration of
`nigrostriatal dopaminergic neurons. To date, several scien-
`tific papers have shown no effect of VPA on PD. However,
`recent in vitro studies have shown positive effects of VPA
`in models mimicking PD at different levels: VPA treatment
`prevents apoptosis induced by rotenone (inducing PD—like
`neurodegeneration); protects neurons from dopaminergic
`toxin 1—methyl—phenylpyridinium (MPP+) by stimulating
`the release of trophic substances from glia; protects cul-
`tured dopaminergic neurons from over—activated microglia—
`induced degeneration by promoting microglia apoptosis and
`protects neurons by increasing oc—synuclein expression and
`preventing its monoubiquitination and nuclear transloca—
`tion [34—37]. In rodents, experiments show that selective
`alterations of 0c—synuclein caused by rotenone (decrease of
`the native protein and an increase in monoubiquitination in
`substantia nigra and striatum) are counteracted by long—term
`administration of VPA [38].
`Past clinical trials had showed a lack of VPA activity in
`patients with Huntington’s disease (HD) [39], which is sur-
`prising given that the molecular explanation for this disease
`involves excitotoxicity and reduced gene transcription due
`to decreased levels of histone acetylation. However, a recent
`study showed that using high doses of VPA (100 mg/kg/ days)
`is capable of prolonging life expectancy and improving
`traction [40]. This study, based on tests conducted on the
`N171—82Q transgenic mouse model of HD, showed that
`following chronic intraperitoneal daily administration, VPA
`
`significantly improved the condition of mice, increasing the
`number of the surviving mice and reducing the decrease in
`motor activity, and this without exerting any noteworthy side
`effects upon behavior or striatal dopamine content at the
`dose administered. This study therefore recommends further
`clinical trials, based on increasing doses of VPA administered
`in mono— or polytherapy.
`
`2.2. Addiction. The GABAergic system is involved in psy-
`chostimulant sensitization, and VPA can modulate central
`
`GABAergic neurotransmission. An initial study, conducted
`on mice, showed that multiple injections of VPA, admin-
`istered consecutively after methamphetamine, could reduce
`the addictive behavior induced by this drug. However,
`this effect is not reproducible for cocaine—induced behavior
`[41]. These initial results indicated that
`these forms of
`addiction should not involve the same neural mechanisms.
`
`A clinical study of VPA use in treating addiction to various
`substances particularly targeted the stage of detoxification
`prior to treatment and rehabilitation. It was shown that
`a combination of Buprenorphine and VPA seems to be
`the most appropriate for detoxification, compared to the
`traditional combination Clonidine and Carbamazepine [42].
`Several addictions, obsessive—compulsive disorders, and
`compulsive sexual behaviors have similarities in their pro-
`cesses, and, in the use of VPA, may find a possible therapeutic
`treatment.
`
`(HDAC1) has
`2.3. HIV Infection. Histone deacetylase 1
`been implicated in maintaining HIV in infected cells. The
`inhibitory action of VPA upon this protein makes it a
`good candidate for AIDS therapy. A study published in
`2005 undertaken with four patients, showed that
`three
`of them treated with valproic acid in addition to highly
`active antiretroviral therapy (HAART), showed a reduction
`in latent HIV infection [43]. However, a subsequent study
`showed that VPA, alone and in a long—term treatment, is not
`sufficient to reduce the size of the HIV-1 reserve [44].
`
`2.4. Other Pathologies. The possibility of using VPA in
`treatment of Duchenne Muscular Dystrophy, a skeletal
`muscle degeneration disease, was recently demonstrated in
`vitro and in vivo on mice mdx/utrn’/’ [45]. This study
`showed that VPA is able to induce the Akt/mTOR/p70S6K
`pathway, through the induction of phosphatidylinositol 3-
`OH kinase (PI3K), resulting in lower collagen content and
`fibrosis, a decrease of hind limb contractures, an increase of
`
`sarcolemmal integrity, a decrease of CD8—positive inflamma-
`tory cells and higher levels of activated Akt in muscles.
`
`3. VPA Targets Signaling Pathways in
`Cancer Cells
`
`Epigenetic processes, such as histone deacetylation and DNA
`methylation, are known to contribute to the cancerous
`transformation of cells by silencing critical genes, leading
`to chemotherapy resistance. It has been hypothesized that
`HDACi act
`through a derepression or a stimulation of
`
`

`
`[46—48]. Several data
`silenced tumor suppressor genes
`demonstrate the ability of these epigenetic drugs to modulate
`global gene expression in tumors (Figure 2).
`VPA modulates expression of p21WAF1/CDKNIA [49],
`a CDK (cyclin dependent kinase) generally associated with
`cell cycle arrest
`in G1/S phase. VPA appears to induce
`an increase in the amount of p21WAF1 protein over 48
`hours in AML cells, which is independent of p53 levels
`(whose expression is not modulated by VPA), but appears
`to be dependent on c—myc, whose mRNA and protein levels
`decrease in a dose—dependent manner during VPA treatment.
`Under VPA treatment, transcription of c—myc is dependent
`upon INK/SAPK, and ectopic expression of c—myc, leading to
`more resistant cells in cell cycle arrest, shows the importance
`of this protein in VPA—mediated activity. In addition to this,
`while VPA treatment in the cell lines is followed by a decrease
`of c—myc and an induction of p21, it has been observed that
`in primary AML cells, only the systematic decrease of c—myc
`is maintained, without upregulation of p21 expression.
`VPA—induced apoptosis, via the extrinsic pathway involv-
`ing engagement of the caspase—8—dependent cascade, sensi-
`tizes cells to TRAIL/Apo2L—mediated apoptosis by increasing
`expression of DR4 and DR5 by 3- and 14-fold, respectively.
`In the absence of TRAIL/Apo2L, VPA is able to reduce the
`expression of antiapoptotic factors acting on both extrinsic
`and intrinsic apoptotic pathways, including c—FLIPs, proteins
`associated with DISC and Bcl—2/Bcl—X(L). When applied
`with TRAIL/Apo2L, VPA increased cell death and caspase-
`3 activity. These results were reproduced in AML, CLL,
`thoracic cancers, and hepatocellular carcinoma [50—53].
`VPA induces Caspase—11 and FAS —L at the transcriptomic
`level, and Caspase—3 at the proteomic level during exposition
`[54]. Long treatment periods result in enhancement of Fas-
`dependent apoptosis associated with an overexpression of
`Fas and Fas ligand, and a central role in Bcl—2 inhibition [55].
`It has been reported that VPA increased fi—catenin levels
`through concentration—dependent
`inhibition of glycogen
`synthase kinase 3—fi (and 30:) activity, and tau phosphory—
`lation [56]. On one hand, this resulted in the inhibition of
`ubiquitination of fi—catenin and on the other hand, in the
`inhibition of c—jun phosphorylation leading to an increase
`in the DNA binding activity of AP—1. Inhibition of fi—catenin
`ubiquitination led to its translocation into the nucleus, and
`resultant upregulation of c—myc transcription.
`The PI3—kinase/Akt pathway and Sp1 are involved in
`HSP70 induction by HDAC inhibitors, and induction of
`HSP70 by VPA in cortical neurons may contribute to its
`neuroprotective and therapeutic effects [57].
`In medullary thyroid cancer (MTC) cells, VPA is able
`to modify the expression of Notchl, by increasing its
`expression, resulting in inhibition of the growth of these cells
`and the induction of apoptosis. These results along with the
`apparent tolerance of patients to VPA treatment have led to
`the suggestion to involve VPA being in future clinical trials
`on advanced medullary thyroid cancer [58].
`PPAR signaling is involved in several biochemical regula-
`tion processes, including lipidic metabolism, differentiation,
`insulin sensitivity, and cell survival. VPA appeared to be
`a pan—activator of PPAR (both PPARoc, also PPAR5 and
`
`Iournal of Biomedicine and Biotechnology
`
`[59, 60]. This mechanism could be one factor
`PPAR)/)
`favoring the teratogenic effect of VPA [60]. In contrast, it
`was reported that in neuronal cells VPA induced a significant
`decrease in PPARy signaling [61]. These results highlight
`potential tissue—specific effects of VPA, as observed in several
`publications, and the difficulties in predicting its effects prior
`to clinical trials.
`
`In similar fashion, HDAC3 is involved in inhibition
`of STAT3 phosphorylation [62]. This inhibition led to a
`decrease in the dimerisation of STAT3 and its translocation
`
`into the nucleus for transcriptional activation of many genes
`in a wide range of biological processes, including induction
`of immune response, oncogenesis, cell cycle control, develop-
`ment, cell adhesion, and differentiation [63]. HDAC3 is one
`of the direct targets of VPA, but until now no papers present
`data concerning the effect of VPA on the STAT3 pathway.
`
`4. Clinical Trials with VPA
`
`As previously mentioned, several reports have demonstrated
`the ability of epigenetic drugs to modulate global gene
`expression in tumors. In many cases, such drugs have moved
`into the first or second phase of clinical trials for treatment
`of various solid cancers or leukemia, and in cotreatment with
`
`several chemotherapeutic agents (Table 1).
`VPA is used for many years in the treatment of convulsive
`seizures and in chronic treatment of epilepsy. Highlighting its
`HDAC inhibitory property and therefore its potential action
`on some cancers has made this molecule, already well known,
`a top candidate in new clinical trials.
`Valproic acid has been implicated in more than two
`hundred clinical trials, sometimes in association with other
`
`ranging from
`drugs and involving various pathologies,
`mood disorders to cancers, through treatment of narcotic
`addictions, muscle disorders, and the ability to reduce viral
`load in AIDS patients (http://clinicaltrials.gov/).
`However, concerning cancer prevention and therapy, a
`recent study on a population taking VPA over a long period
`(minimum 1500 g in the last 5 years) demonstrated that it
`had no significant effect upon prevention of cancer develop-
`ment [64] and thus is not eligible for chemoprevention.
`
`4.1. Clinical Trial in Noncomcerous Disorders. VPA has been
`
`used for many years as a treatment against epilepsy and
`convulsive disorders. Today, the majority of clinical trials
`involving VPA relate to neural pathology, applying to more
`than thirty different pathologies.
`One clinical study aims to compare the antiaggressive
`efficacy of risperidone monotherapy versus risperidone
`in combination with valproate in patients suffering from
`schizophrenia. The random blind test, including 33 patients
`(16 with risperidone and 17 with polytreatment), was unable
`to show any significant difference between the treatments,
`except a higher capacity of patients under combination ther-
`apy to complete the study [65]. The use of valproic acid was
`investigated for its ability to improve mood stabilization. The
`blind clinical trial aimed to assess the capacity of lamotrigine
`or VPA to increase mood stabilization for patients with
`
`

`
`Iournal of Biomedicine and Biotechnology
`
`5
`
`
`
`
`Integration to the plasmic
`membrane
`
`
`
`
`
`
`
`
`.
`C §\‘ ..“ ..
`
`\\\\\‘|\_\\\\\
`
`
`
`
`
`\
`
`\
`
`
`
`}’\'P‘’\'‘\\‘’\'P‘"‘\\‘’\'P‘\'’\\‘’\'P‘: X. --H -- -I-- --ml-- -In -g
`
`,
`Proinflammatory
`_\ c. N _. c. .\ c. .c. A c. .\ _. N >\ c. .c _.
`effect
`
`:
`
`Cell
`differentiation
`
`.
`
`.
`
`"\'P‘’\'P
`
`‘‘’\'P‘’\'‘\\‘’\'P‘i
`
`Cell
`._ _. _. .. _.
`\ _. ._ .. _.
`._ _. _.
`l proliferation
`
`‘ jg
`!_ _ _
`
`_
`
`_ _
`
`_ __ _
`
`_ _
`
`_ _
`
`_ __
`
`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 1 (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).
`
`

`
`schizophrenia who are both stabilized and partially respon-
`sive [66]. Efficiency assessment was evaluated using the Pos-
`itive and Negative Syndrome Scale, Calgary Depression Scale
`for Schizophrenia, Demoralization Scale, Clinical Global
`Impression severity and improvement scores and Lehman
`quality of life improvement scale to assess quality of life
`and social functioning. The increase of antipsychotics usage
`with mood stabilizers like lamotrigine or VPA for partially
`responsive patients with chronic schizophrenia seems to be
`an inefficient treatment strategy for improving the residual
`symptoms.
`A clinical trial to establish the difference between VPA
`
`in the treatment of pediatric migraine
`and Propranolol
`prophylaxis was undertaken on a population of 120 patients
`aged from 3 to 15 years. The results showed no significant dif-
`ference in all therapeutic effects between the two molecules,
`with a mean of 70% response and a reduction of >50% in
`headache frequency [67].
`A clinical trial concerning agitation of older people with
`dementia was based upon the use of VPA as a treatment over
`several years, without any conclusive clinical trials able to
`show the effects. The results presented were based on the
`compilation of three incomplete clinical studies and show
`that VPA does not induce any improvement in pathology but
`does present an unacceptable rate of side effects [68].
`Acute mania is a behavioral disorder outcome of bipolar
`disorder, also known as manic—depressive disorder, manic
`depression, or bipolar affective disorder. Compilation of
`data provided by European Mania in Bipolar Evaluation
`of Medication (EMBLEM) concerning the comparison of
`olanzapine and valproic acid treatments on a panel of over
`600 patients (rz : 107 valproate, n : 514 olanzapine),
`demonstrated that olanzapine monotherapy seems to be
`more effective than valproate monotherapy in the treatment
`of acute mania [69].
`In contrast, a recent systematic review about the effect
`of VPA upon 142 patients with acute bipolar depression
`shows a significant effect of this molecule for the reduction
`of depressive symptoms of acute bipolar depression, as well
`as high patient tolerance [70]. The outcomes investigated
`were depression, anxiety, hypomania, attrition, and adverse
`events, and the study analyzed existing randomized control
`trial data for the efficacy and tolerability of valproate.
`Results are particularly conclusive for depression (50%
`improvement, for 22% of patients), but inconclusive for
`anxiety, and there was no evidence for an increased risk
`of mania, as
`it also induces long—term antidepressant
`effects.
`
`VPA was investigated for clinical therapy of amyotrophic
`lateral sclerosis (ALS). This prognostic neuropathy was
`studied in 163 patients who received either VPA (1500 mg)
`or placebo, daily. The end points targeted were survival and
`progression of the disease. This study concluded that VPA
`had no beneficial effect upon survival or disease progression
`in patients with ALS [71].
`A phase II study of the effect of VPA upon spinal
`muscular atrophy (SMA), with a panel of 2 SMA type I
`(ages 2-3 years), 29 SMA type II (ages 2-14 years), and
`11 type III (ages 2-31 years) was recently published. The
`
`Iournal of Biomedicine and Biotechnology
`
`investigation focused upon several factors, such as assess-
`ment of gross motor function (via the modified Hammer-
`smith Functional Motor Scale: MHFMS), electrophysiologic
`measures of innervations (with maximum ulnar compound
`muscle action potential amplitudes: CMAP) and motor unit
`number estimation (MUNE), body composition, and bone
`density via dual—energy X—ray absorptiometry (DEXA), and
`quantitative blood SMN mRNA levels, as well as carnitine
`depletion, hepatotoxicity, and increased weakness (for the
`side effects). Results revealed a trend of weight gain but
`also an increase of gross motor function in 27 patients,
`all of them with SMA type II and younger than five years
`old. There was no variation in expression of SMN protein
`(survival motor neuron protein), but bone mineral density
`and innervations increased significantly (P S .0001). In
`conclusion, the results presented by this preliminary study
`highlight
`the strengths and limitations of using a large
`cohort of patients for such a trial, as opposite results were
`obtained as functions of age and type of SMA. As such,
`no final conclusion was made concerning VPA, but it was
`suggested that additional controlled clinical trials with VPA
`targeting more restricted cohorts of subjects were needed
`[72].
`
`A phase II clinical trial, principally investigating the
`effect of glyceryl trinitrate, and incidentally that of VPA,
`on pain caused by diabetic peripheral neuropathy, showed
`that these two molecules induce significant improvement,
`whether used alone or in combination [73]. This study,
`undertaken upon 83 patients divided into four groups,
`compared visual analogue score (VAS) and present pain
`intensity (PPI) at
`the beginning of the study and after
`three months, with motor and sensory nerve conduction
`velocities measured using electrophysiological tests. These
`results show significant improvements with both drugs indi-
`vidually and in combination, except for electrophysiological
`test results with VPA treatment, which show no significant
`modification.
`
`Kaposi sarcoma (KS) is a cutaneous tumor caused by
`Human herpesvirus 8 (HHV8) and is often associated
`with coinfection by HIV (AIDS—associated Kaposi sarcoma),
`especially in African populations. Valproate stimulates HIV
`replication in some HIV—infected cell lines in culture [74, 75],
`and in the case of HHV8—infected cells, valproate induces
`expression of multiple HHV8—encoded transcripts that are
`associated with entry into the lytic phase of replication [76].
`Because valproate is sometimes used in individuals who
`might be infected with HHV8 and HIV, a recent study aimed
`to identify the effect of VPA in cases of AIDS—associated
`Kapo si sarcoma [77]. Their results show that treatment with
`VPA was associated with low toxicity, and that KS clinical
`response and herpesvirus lytic induction were not high
`enough to be associated with significant induction of virus
`replication.
`
`The clinical trials mentioned to date concerning treat-
`ment of nonmalignant diseases show few real benefits
`associated with use of VPA in mono— or polytreatment, and
`this despite the very encouraging preclinical data obtained so
`far.
`
`

`
`Iournal of Biomedicine and Biotechnology
`
`TABLE 1: Ongoing clinical studies implicating valproic acid in
`monotherapies.
`
`Dependences
`Alcohol abuse or dependence
`Alcoholism
`
`Cocaine dependence
`Marijuana abuse
`Substance abuse or dependence
`Substance withdrawal syndrome
`Cancer
`
`Autoimmune lymphoproliferative syndrome
`Brain and central nervous system tumor
`Breast cancer
`CLL
`
`HTLV—I associated myelopathy
`MDS risk; AML
`
`Nasopharyngeal carcinoma
`Prostate cancer
`Sarcoma
`
`Neurological disorders
`Alzheimer disease
`
`Amyotrophic lateral sclerosis
`Attention deficit hyperactivity disorder
`Autism
`
`Bipolar disorder
`Borderline personality disorder
`Cluster headache
`Dementia
`
`Depression
`Disruptive behavior
`Epilepsy
`Mania
`
`Migraine
`Mood disorder
`
`Neuralgia
`Phosphosensitive epilepsy
`Post traumatic stress disorder
`
`Progessive supranuclear palsy
`Resistant bipolar depression
`Schizophrenia
`Spinal muscular atrophy type 1
`Others
`Asthma
`
`Hypersplenism; lymphadenopathy
`Hyp oalbunemia
`Insulin resistance
`
`4.2. Clinical Trial in Solid Tumors and Leukemia
`
`4.2.1. VPA in Myeloid and Lymphoid Malignancies. The use
`of VPA in monotherapy or polytherapy seems promising for
`leukemia diseases. It has already been established that VPA
`
`exerts different effects in different cell types, and probably in
`the function of these cell types. It can induce proliferation
`in early stem cells and can inhibit angiogenesis, production
`of TNF—oc and IL-6, and activation of NF—KB. Taken together
`these properties could be beneficial in the treatment of MDS
`or leukemic pathologies.
`While HDACi are emerging as valuable new agents in the
`treatment of acute myeloid leukaemia (AML), the efficiency
`rates of these compounds in isolation are low, which requires
`them to be used in cotreatment with other anticancer
`
`drugs. Research into predictive markers of the efficiency
`of cotreatment with VPA gave rise to a study published
`in 2009, comparing results obtained by coprocessing using
`all
`trans retinoic acid (ATRA) and theophylline on 20
`patients and several cell lines, and a microarray study on
`primary AML cells and cell
`lines. Comparison between
`results obtained with cotreatment (one Complete Remission
`(CR), two Partial Remission (PR)) and those obtained in
`treatment of primary cells and cell lines allowed the authors
`to conclude that similar factors determine both in vivo and in
`
`vitro sensitivity and identified elevated FOSB—expression as a
`potential marker of VPA sensitivity [78].
`Hematological
`improvement was reported in many
`patients with different
`types of cancer
`following VPA
`treatment, with favorable responses; in patients with MDS
`(myelodystrophy syndromes) and MP8 (myeloproliferative
`syndromes)
`[79];
`in patients with AML developed from
`MPD (myeloproliferative disorders) [80]; in patients with
`myelofibrosis in myeloid metaplasia [81]. Clinical studies on
`the use of VPA have shown the beneficial effect of this drug
`especially on MDS [82, 83].
`In 2007, a phase I/II study investigated the use of a
`tritherapy combination of 5—azacitidine (5—AZA), VPA, and
`ATRA in patients with acute myeloid leukemia or high-
`risk MDS. A total of 53 patients were tr

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket