You are on page 1of 11

Amyotrophic Lateral Sclerosis.

2009; 10: 8594

ORIGINAL ARTICLE

Combined riluzole and sodium phenylbutyrate therapy


in transgenic amyotrophic lateral sclerosis mice

STEVEN J. DEL SIGNORE1, DANIEL J. AMANTE1, JINHO KIM1,4,


EDWARD C. STACK1,4, SARAH GOODRICH1, KERRY CORMIER4, KAREN SMITH1,4,
MERIT E. CUDKOWICZ5 & ROBERT J. FERRANTE14

Departments of 1Neurology, 2Laboratory Medicine and Pathology, 3Psychiatry, Boston University School of Medicine,
Boston, 4Geriatric Research Education Clinical Center, New England Veterans Administration, Bedford, and 5Neurology
Clinical Trials Unit, Massachusetts General Hospital, Charlestown, Massachusetts, USA

Abstract
Recent evidence suggests that transcriptional dysregulation may play a role in the pathogenesis of amyotrophic lateral
sclerosis (ALS). The histone deacetylase inhibitor, sodium phenylbutyrate (NaPB), is neuroprotective and corrects aberrant
gene transcription in ALS mice and has recently been shown to be safe and tolerable in ALS patients while improving
hypoacetylation. Since many patients are already on riluzole, it is important to ensure that any proposed therapy does not
result in negative synergy with riluzole. The combined treatment of riluzole and NaPB significantly extended survival and
improved both the clinical and neuropathological phenotypes in G93A transgenic ALS mice beyond either agent alone.
Combination therapy increased survival by 21.5%, compared to the separate administration of riluzole (7.5%) and NaPB
(12.8%), while improving both body weight loss and grip strength. The data show that the combined treatment was
synergistic. In addition, riluzole/NaPB treatment ameliorated gross lumbar and ventral horn atrophy, attenuated lumbar
ventral horn neuronal cell death, and decreased reactive astrogliosis. Riluzole/NaPB administration increased acetylation at
H4 and increased NF-kB p50 translocation to the nucleus in G93A mice, consistent with a therapeutic effect. These data
suggest that NaPB may not interfere with the pharmacologic action of riluzole in ALS patients.

Key words: Amyotrophic lateral sclerosis, riluzole, histone deacetylase inhibition, sodium phenylbutyrate, therapy

Introduction those observed in human ALS (4). Genetic mouse


models of ALS have provided a greater understand-
Amyotrophic lateral sclerosis (ALS) is a rapidly
ing of disease pathogenesis and have helped to
developing disease characterized by degeneration of
develop potential therapeutic strategies for human
upper and lower motor neurons, progressive muscle
clinical trials (5,6). Glutamate mediated excitotoxi-
weakness and atrophy, and morbidity associated
city, transcriptional dysregulation, oxidative stress,
with failure of respiratory muscles (1). The inci-
and bioenergetic dysfunction have all been proposed
dence of ALS is 12 per 100,000 per year and may as pathophysiological mechanisms of disease in ALS,
be rising. Death usually occurs within 25 years of and each has been the focus of successful preclinical
diagnosis. Although the root cause of disease is trials in animal models with mutations in the SOD1
unclear, genetic mutations have been discovered to gene (710). Pharmacotherapies that can address
underlie 10% of cases, termed familial ALS (FALS). multiple disease pathways may provide a promising
Mutations in the enzyme Cu/Zn superoxide dismu- route to slowing disease progression.
tase (SOD1) cause about 25% of FALS cases and Since the first conceptualization of glutamate
their clinical and pathological features are indistin- excitotoxicity (11), there has been strong evidence to
guishable from those in sporadic ALS (2,3). Over- support its importance in the pathology of ALS (12).
expression of mutant SOD1 in animal models is The only drug currently approved to treat ALS is
sufficient to cause motor neuron degeneration re- riluzole, a drug with anti-excitotoxic properties
sulting in clinical and pathological features similar to (13,14). The neuroprotective effect of riluzole may

Correspondence: R. J. Ferrante, GRECC Unit 182B, Bedford VA Medical Center, 200 Springs Road, Bedford, MA 01730, USA. Fax: 781 687 3515.
E-mail: rjferr@bu.edu

(Received 28 March 2008; accepted 20 May 2008)


ISSN 1748-2968 print/ISSN 1471-180X online # 2009 Informa UK Ltd. (Informa Healthcare, Taylor & Francis AS)
DOI: 10.1080/17482960802226148
86 S. J. Del Signore et al.

stem from a number of pharmacological actions established colony maintained at the Bedford Veter-
(15). Riluzole has been shown to inhibit glutamate ans Affairs Medical Center and used for the experi-
release, possibly through inhibition of presynaptic ments. PCR assay of tail DNA was used to genotype
calcium and sodium channels (16,17) and to mod- the offspring. Animals in each experiment were from
ulate depolarizing currents at alpha-amino-3- the same ‘f’ generation, born within four days of one
hydroxy-5-methyl-4-isoxazolepropionic acid and another. Only male mice were used in the treatment
N-methyl-D-aspartic acid receptors (18,19). While studies because we have observed gender differences
several experiments in animal models have replicated in survival in the G93A transgenic ALS mouse
the therapeutic effect observed with riluzole in ALS model, with extended survival and greater variability
patients (7,20), there have also been a number of in time of death in female G93A mice (33). All
ALS rodent trials demonstrating an enhanced ther- animal experiments were performed in accordance
apeutic effect by combining one or more other drugs with the NIH Guide for the Care and Use of
with riluzole (2123). In addition, a clinical trial Laboratory Animals and were approved by the
investigating the combination of riluzole and high Veterans Administration Animal Care Committee.
doses of vitamin E has been reported (24).
In addition to excitotoxicity, transcriptional dys-
Treatment
regulation has also been shown to play a role in the
neurodegeneration observed in ALS (2528). Tran- Animals were randomly distributed into one of four
scription is regulated by complex interactions be- treatment groups (n 10) at four weeks of age:
tween many proteins. Among the most important of untreated, NaPB-treated (300 mg/kg), riluzole-
these proteins are transcription factors and histones, treated (16 mg/kg), or riluzole/NaPB-treated using
which affect the actions of RNA-Polymerase II on the above doses. NaPB (Triple Crown USA, Inc.,
individual genes. A well-recognized mechanism of Perkasie, PA) was dissolved in physiological saline at
transcriptional regulation is histone modification. a concentration of 75 mg/ml. Fresh solutions were
Acetylation of lysine 9 and 13 at histone H4 is made daily and administered via intraperitoneal
associated with an enhanced transcriptional state. injection. Riluzole was obtained as RilutekTM, 50-
Histone deacetylase (HDAC) inhibitors increase mg tablets and dissolved in drinking water at a
acetylation of histones, promoting transcriptional concentration of 100 ug/ml. Solution was made fresh
activation. Of the five classes of HDAC inhibitors, weekly and stored at 48C. Fresh solution was
the butyrates are the most developed for clinical use provided to the animals twice per week. Treatments
because they are able to penetrate the blood-brain- were started at 30 days. The untreated G93A mice
barrier (2931). Sodium phenylbutyrate (NaPB) has were injected with saline (vehicle). Separate groups
been shown to increase acetylation at H4, and (n 10) of treated and untreated mice were used for
improve the clinical, neuropathological, and mole- histological analyses.
cular phenotype in the G93A mouse model of ALS
(8). In addition, we recently reported that NaPB was
Survival
safe and tolerable in ALS patients and was ther-
apeutically effective in improving hypoacetylation Animals were assessed twice daily (early morning
levels found in this patient population (32). and late afternoon) for morbidity and mortality by
Given the multifactorial nature of disease patho- two independent observers who were blinded to the
genesis of ALS, combination therapies that address assigned treatment (RJF and KS). Motor perfor-
multiple disease pathways hold great promise for mance and ability to feed was closely monitored and
achieving maximal therapeutic effect. As riluzole is used to determine when to euthanize the mice.
the only FDA-approved drug currently available to Animals were euthanized when they were unable to
patients, it is especially important to develop com- right themselves after being placed on their back for
binatorial treatments that are safe and effective, that 30 s. Some deaths occurred overnight and were
do not negatively interfere with riluzole. We investi- recorded the following morning. Body weights
gated the effects of the combined administration of were recorded weekly on the same day and time.
riluzole and NaPB on the pathological phenotype
observed in the G93A mouse model of ALS.
Behavior analysis
Neuromuscular function was assessed using the Grip
Materials and methods Strength apparatus (Columbus Instruments, Co-
lumbus, OH). This instrument measures the peak
Experimental animals
amount of force an animal applies in grasping
Transgenic mice overexpressing human SOD1 with specially designed pull-bar forelimb and hindlimb
the G93A mutation (TgSOD1 (G93AGUR) F1/J, assemblies. Briefly, mice were held by the scruff of
Jackson Labs, Bar Harbor, ME) were bred with their back and drawn along in a straight line along
females of similar background (B6/SJL) from an the sensor assemblies to allow the mice to grasp the
Combined riluzole and sodium phenylbutyrate therapy 87

forelimb pull-bar assembly, recording the force and subsequently stained for Nissl substance, and
applied by the forelimbs of the mouse in Newtons. immunostained for glial fibrillary acidic protein
In a continued sweep, the mouse was drawn along (GFAP) (1:100; Boehringer-Mannheim, Indianapo-
until the hindlimbs grasped and released the hin- lis, IN), acetylated histone H4 (rabbit polyclonal
dlimb assembly, again recording the force applied by antibody, 1: 1000; Upstate Biotechnology, Lake
the hindlimbs of the mouse. Forelimb and hindlimb Placid, NY, USA), and nuclear factor-kappa B
strength were measured by the same investigator (NF-kB) p50 (rabbit polyclonal antibody, 1: 500;
(SJD) three times and averaged during each weekly Cell Signaling Technology, Inc.).
assessment. It is critically vital that a single investi-
gator performs the grip strength measurements due
to inter-rater differences in tensile strength. Neuronal quantitation
Serial lumbar spinal cord tissue sections (n 20)
from L3L5 spinal cord segments were used for
Histology
gross spinal cord areas and neuronal analysis. Gross
Additional groups of 10 animals from each areas of the spinal cord sections were quantified
treatment paradigm were deeply anesthetized and from each experimental cohort using NIH Image.
transcardially perfused with 4% buffered parafor- From the same sections, the ventral horn was
maldehyde at 120 days of age. Forty mice were used delineated by a line from the central canal laterally
for neuropathological analysis. Spinal columns were and circumscribing the belly of gray matter. Abso-
removed and placed in fixative overnight. Cords lute neuronal counts of Nissl-positive neurons were
were dissected and cryoprotected in 10%, then 20%, performed in the ventral horns in the lumbar spinal
glycerol in DMSO/TBS-T. The lumbar section of cord. Only those neurons with nuclei were counted.
each cord was then blocked and cut on a freezing All counts were performed with the experimenter
sledge microtome at 50 mm. Sections were stored in (SJD) blinded to treatment conditions. Counts were
phosphate buffer with 0.01% sodium azide at 48C performed using Image J (NIH) and manually

Figure 1. Effects of riluzole and NaPB on survival, body weight, and grip strength in G93A transgenic ALS mice.
Untreated G93A mice have a median survival of 126 days (A). Treatment with either riluzole (18 mg/kg) or NaPB (300 mg/kg) resulted in a
significant survival extension of 7.5% and 12.8%, respectively. Combined riluzole/NaPB treatment resulted in the greatest survival
extension, 21.5% over untreated mice. There is a significant improvement in body weight in all treated groups, compared to untreated
control mice (B). Mice treated with combined riluzole and NaPB show the greatest improvement, with body weight significantly elevated
above all other groups. G93A mice treated with combined riluzole and NaPB resulted in a marked improvement in forelimb grip strength
compared to untreated mice (C). Starting at 91 days of age the combined therapy resulted in improved grip strength that persisted
throughout testing. In contrast, an analysis of hind limb grip strength revealed no improvement in combined riluzole and NaPB treated
G93A mice compared to untreated mice (D).
88 S. J. Del Signore et al.

verified and the data represent the average neuronal improvement in combined riluzole/NaPB treated
number from the sections used. G93A mice, compared to untreated mice (Figure
1D). It should be noted that a differential paralysis
occurs early and most severely in the hindlimbs, with
Statistical analysis less and later involvement in the forelimbs. This
Data are expressed as the mean9SEM. Statistical early and more pronounced involvement of the
comparisons of grip strength, weight data and hindlimbs may be too great for riluzole-NaPB
histological data were made by ANOVA or re- therapeutic treatment to overcome.
peated-measures ANOVA. Survival data were ana- Neuropathological analysis at 120 days revealed
lyzed using Kaplan-Meier survival curves. All other marked gross spinal cord atrophy, with increased
statistical analyses were performed using Student’s astrogliosis and neuronal loss in the ventral horns
t-test. from the lumbar spinal cord in untreated G93A
mice. These pathological findings were significantly
reduced by riluzole/NaPB administration. There
Results was significant gross atrophy of the lumbar spinal
cord in untreated G93A mice, compared to wt
Both riluzole (16 mg/kg) and NaPB (300 mg/kg) littermate control mice (wt littermate control:
treatment alone significantly extended survival in the 3.0890.18 106 mm3; untreated G93A mice:
G93A ALS mice by 7.5% and 12.8%, respectively, 2.3190.95 106 mm3, p B0.0001) with significant
compared to untreated G93A mice (Figure 1A). amelioration of gross spinal cord atrophy in riluzole/
These data are consistent with previously reported NaPB treated mice (riluzole/NaPB treated G93A
findings. The combined administration of riluzole/ mice: 2.6790.43 106 mm3, p B0.01) (Figure 2A
NaPB also significantly extended survival in the and 3AC). The gross atrophy was largely asso-
G93A mice by 21.5%, suggesting that the efficacy ciated with the ventral horn of the lumbar spinal
was synergistic without having a negative impact on cord (wt littermate control: 7.2190.45 105;
survival, compared to drug treatments alone (un-
treated G93A mice: 126.192.7 days; riluzole 16 mg/
kg per day: 135.594.4 days, p B0.05; NaPB 300
mg/kg per day: 142.295.4 days, p B0.01; riluzole/
NaPB 16 mg/kg and 300 mg/kg, respectively, per
day: 153.299.1 days, p B0.001).
There was a significant improvement in body
weight in all the treatment groups, compared to
untreated control mice (p B0.01) (Figure 1B). The
riluzole treatment arm significantly ameliorated
body weight loss, compared to untreated controls,
starting at 115 days (p B0.05). The NaPB treatment
improved body weight starting at 101 days and
continued through the end of the trial (p B0.01).
G93A mice treated with combined riluzole and
NaPB showed the greatest improvement, with body
weight significantly elevated above the untreated
G93A mice at 91 days (p B0.01) and above the
other treatment groups starting at 115 days, remain-
ing consistent through the completion of the trial
(p B0.01). Forelimb and hindlimb grip strength
were assessed as a measure of neuromuscular
performance and motor function. Others and we
have previously reported that motor performance is
improved by riluzole or NaPB treatment alone (7,8).
As such, it is of import to demonstrate that G93A
mice treated with combined riluzole and NaPB also Figure 2. Combined riluzole and NaPB treatment improves gross
area of lumbar spinal cord and ventral horn areas in G93A
improve this outcome measure. There was a marked transgenic ALS mice.
improvement in forelimb grip strength in riluzole/ Analysis of gross lumbar spinal cord area reveals a marked
NaPB treated G93A mice, compared to untreated decrease in untreated G93A mice compared to wild-type mice.
littermate G93A mice. At 91 days of age, the Treatment with riluzole and NaPB results in a significant
combined therapy resulted in significantly improved improvement in lumbar spinal cord area (A). A similar analysis
of the ventral aspect of the lumbar spinal cord revealed a marked
grip strength (p B0.01) that persisted throughout reduction in untreated G93A mice relative to wild-type control
the trial (Figure 1C). In contrast, however, an mice (B). Treatment with riluzole and NaPB significantly
analysis of hindlimb grip strength revealed no improved ventral horn area in G93A ALS mice.
Combined riluzole and sodium phenylbutyrate therapy 89

Figure 3. Neuroprotective effects of combined riluzole and NaPB in the lumbar spinal cord in G93A transgenic ALS mice. Nissl staining of
the lumbar spinal cord from (A) wild-type mice, (B) G93A mice treated with riluzole and NaPB, and (C) untreated G93A mice shows a
marked gross atrophy in untreated G93A mice. Combined treatment with riluzole and NaPB significantly improves the gross
neuropathological changes, as shown in Figure 2. Bar in A 200 mm and is the same for B and C. High magnification photomicrographs
of Nissl staining in the ventral horn from the same sections in A, B, and C of the lumbar spinal cord from (D) wild-type mice, (E) G93A
mice treated with riluzole and NaPB, and (F) untreated G93A mice demonstrates ventral neuron loss and atrophy in untreated G93A mice.
The combined treatment with riluzole and NaPB markedly improves these neuropathological changes. Bar in D50 mm and is the same for
E and F. Glial fibrillary acidic protein (GFAP) expression within the lumbar spinal cord in wild-type mice reveals low levels of reactive
astrogliosis (G). In contrast, there is marked astrogliosis, as indicated by GFAP immunostaining, in the lumbar spinal cord from untreated
G93A mice (I). Treatment with both riluzole and NaPB markedly attenuates reactive astrogliosis (H). Bar in G 50 mm and is the same in
H and I.
90 S. J. Del Signore et al.

untreated G93A mice: 5.7190.61 105, p B0.05 (13,14). Riluzole acts upon a number of physiologi-
mm3) and was significantly improved by riluzole/ cal pathways that include the inhibition of both
NaPB treatment (riluzole/NaPB treated G93A mice: glutamate release and post-synaptic glutamate re-
6.5990.37 105 mm3, p B0.01) (Figure 2B). Con- ceptor activation, as well as by inactivating voltage
sistent with the above findings, there was marked sensitive sodium channels (15). It has also been
ventral horn neuronal loss in the untreated G93A shown to have neurotrophic and anti-apoptotic
mice, compared to the wt littermate control mice effects, and reduces mitochondrial swelling. Riluzole
(wt littermate control: 93.5914.5; untreated G93A is well-tolerated, although liver function monitoring
mice: 20.495.60, p B0.0001) (Figure 3D, F), with is necessary especially during the first year of
significant reduction in neuronal loss as a conse- therapy. Since many ALS patients may well be
quence of riluzole/NaPB administration (riluzole/ placed on riluzole, it is critically vital to make certain
NaPB-treated G93A mice: 68.4917.0, p B0.01) that any prospective therapeutic agent does not
(Figure 3E). Reactive astrogliosis, a prominent negatively impact the patient or the progression of
finding in untreated G93A mice spinal cord speci- the disease.
mens, was ameliorated by riluzole/NaPB treatment, An important advance in developing drug com-
as shown by the marked reduction of glial fibrillary pounds for clinical trials in ALS patients has been
acidic protein expression in the lumbar spinal cord the introduction of transgenic mice models of ALS.
(Figure 3GI). Of the available ALS murine models, mice expres-
We have previously reported that hypoacetylation sing mutant forms of SOD1, particularly the G93A
of histone activity is present in both G93A mice and transgenic mice, have been the most widely used in
ALS patients and that histone acetylation is im- studying pathophysiological pathways and in devel-
proved by NaPB treatment (8,32). These findings oping candidate ALS therapies (5,6). There is a
demonstrate a parallelism in pathophysiological strong body of evidence suggesting that the pheno-
disease mechanism between mouse and human. types from mouse models of neurological diseases
We confirmed the hypoacetylation of histone H4 in closely correlate with human diseases and may
lumbar spinal cord of untreated G93A mice, com- validate known CNS drug targets in a therapeutically
pared to wt littermates, using immunocytochemical relevant manner. The strengths of the ALS mouse
analysis in tissue specimens and showed that rilu- models are in their utility to provide parallel patho-
zole/NaPB treatment in G93A mice significantly physiological targets that are present in ALS pa-
improves the hypoacetylation of H4 (Figure 4A tients, in their potential as sensitive predictors for
D). It has also been reported that NF-kB activation therapeutic intervention, and their promise in the
through phosphorylation of IkB or directly via development of novel drug agents (5). Although
butyrate administration increases NF-kB transloca- drug trials in mice confirm therapeutic direction, the
tion to the nucleus, improving transcription and challenge is in determining what dose might be of
inhibiting apoptosis (8,34,35). Immunoreactivity of value in patients since the pharmacokinetics of mice
NF-kB p50 was significantly reduced in the spinal and man is dissimilar. It is noteworthy that riluzole,
cords of untreated G93A mice, when compared to which extends life in humans with ALS, has a similar
wt littermates (Figure 4 EH). Riluzole/NaPB treat- survival benefit in ALS mice. While the predictive
ment significantly increased the spinal cord tissue value of therapeutic trials in the G93A ALS mice has
expression of NF-kB in ventral horn neurons from been established using riluzole, there are drug
G93A mice (Figure 4G), resulting in nuclear trans- therapies that are efficacious in ALS mouse models
location of NF-kB activity, consistent with that that do not provide benefit to patients with the
observed in littermate control mice. It is of interest disease (37). In contrast, several potential therapies
to note that among members of the NF-kB family in that have not shown efficacy in mouse models
mammals, the p50 subunit has been shown to be have also been ineffective in human trials (6,38).
neuroprotective (36). The latter scenarios may be the consequence of
insufficient fully-powered clinical trials in humans to
permit a comparison of therapeutic efficacy between
Discussion mouse and man. Alternatively, optimal therapeutic
A clear understanding of the molecular events dosing may be underestimated. Current thought
leading to motor neuron death in ALS remains to indicates that Human Equivalent Dose extrapolation
be elucidated. While a number of pathways have measurements derived from body surface area cri-
been incriminated, both glutamatergic excitotoxicity teria in animals may not accurately predict the
and transcriptional dysregulation have been sug- maximum-recommended safe dose in neurological
gested to play roles in the insidious neuronal death disorders. This is evident in human trials where
observed in ALS (8,12,32). To date, riluzole is the human equivalent dosing of bioenergetic agents
only FDA-approved ALS drug therapy that slows or comparable to that given to mice, while considered
abrogates the disease process in ALS and is asso- safe and tolerable and also effective in mice, have not
ciated with a 23 month prolongation of survival demonstrated significant efficacy in ALS patients
Combined riluzole and sodium phenylbutyrate therapy 91

Figure 4. Effects of combined riluzole and NaPB treatment on acetylated H4 and NF-kB immunoreactivity in G93A mice. Compared to
wild-type mice (A), there is a significant hypoacetylation in the untreated G93A mice (B) in the ventral horn of the lumbar spinal cord,
consistent with reduced transcription. Combined riluzole and NaPB induced a marked increase in acetylated H4 (C). Quantitative analysis
confirms the H4 findings (D). There is also a marked reduction in NF-kB expression in the lumbar spinal cord in untreated G93A mice
with little or no nuclear expression (F), compared to wild-type control mice (E). Combined riluzole and NaPB treatment results in a
notable increase in NF-kB expression in G93A mice, with nuclear translocation (arrows). Quantitative analysis confirms the NF-kB
findings (H). Bar in A 50 mm and is the same for B and C. Bar in D25 mm and is the same for E and F.
92 S. J. Del Signore et al.

(39) and in Huntington’s disease patients (40). mice. Hypoacetylation, as a potential biomarker of
Higher doses of potential therapeutic agents may manifest disease in ALS, may improve the power and
result in slowing disease progression in ALS. As cost-effectiveness of drug trials in this population.
evidence, a double-blind, randomized, controlled The current findings show that combined rilu-
study trial in Parkinson’s disease patients, using zole and NaPB treatment in G93A mice did not
CoQ10 at 1200 mg/day, slowed the rate of deteriora- adversely affect the impact of treatment and out-
tion in the UPDRS score, with higher tolerated come measures, compared to the efficacy of riluzole
doses (41). Of great interest, doses up to 3000 mg/ and NaPB alone. Interestingly, the riluzole/NaPB
day appear safe and well tolerated in ALS patients combination slowed the course of the disease,
(42). evidenced by the altered gradual slope in the
Sodium phenyl butyrate (NaPB), a histone Kaplan-Meier survival curve, compared to the un-
deacetylase (HDAC) inhibitor, is a potential candi- treated G93A mice. There was an additive improve-
date for use in ALS patients (5,8). Histones are ment in both survival and body weight, to a greater
important nuclear proteins that bind DNA in the degree than either compound alone, with a con-
formation of nucleosomes and control transcription. comitant improvement in forelimb grip strength in
Chromatin remodeling and transcription is regulated G93A mice receiving combined riluzole and NaPB.
in part by histone acetylation, which is promoted by Riluzole/NaPB also significantly ameliorated the
the opposing activities of histone acetyltransferases neuropathological sequela observed in the G93A
and HDACs. HDAC inhibitors affect histones as mice. There was reduced gross spinal cord atrophy
well as transcription factors that are influenced by and ventral horn loss, attenuated ventral horn
acetylation. As altered nucleosome dynamics may neuronal death, markedly diminished reactive astro-
play a role in the pathogenesis of ALS, the recruit- gliosis, and improved both histone H4 acetylation
ment of HDACs to DNA alters the nucleosome and NF-kB expression in the lumbar spinal cord
structure locally and inhibits transcription. HDAC from G93A mice.
inhibitors can promote either transcription activa- It must be said, however, that the treatment was
tion or suppression by relaxing DNA conformations. started prior to manifest clinical symptoms in the
Because HDAC inhibitors induce growth arrest in G93A mice and, as such, NaPB may not provide
cell proliferation models, HDAC inhibitors have also efficacy if administered after diagnosis in patients
been used as anti-cancer drugs (31). While other with sporadic ALS. While the G93A mice may not
compounds, such as trichostatin A and suberoylani- be a perfect model of sporadic disease, starting
lide hydroxamic acid, have been studied, the buty- treatment in these mice with only a few weeks
rates have been the best clinically studied remaining until their death makes any positive
compounds and are known to readily reach the brain treatment outcome very unlikely. In defense of the
(43). present treatment paradigm, the disease begins very
HDAC inhibition has been shown to be neuro- early in life in G93A mice, with motor unit number
protective in mouse models of spinal muscular decline from 40 days, N-acetyl aspartate levels
atrophy and Huntington’s disease (44,45), and is decreased by 34 days, and mitochondrial swelling
under investigation in these patient populations. We and cytoplasmic microvacuoles occurring by two
have reported that NaPB dramatically improves the weeks of age. It has been suggested that the clinical
behavioral, neuropathological, and molecular phe- and pathological features of familial ALS patients
notype in G93A ALS mice, extending survival by are indistinguishable from those in sporadic ALS
approximately 22% beyond untreated G93A mice (2,3). As such, pathophysiological mechanisms may
and restoring histone acetylation to near normal begin well before clinical symptoms, and the efficacy
levels (8). Of relevance is the finding that NaPB, in the G93A mice in this study may be proof of
either directly or post-transcriptionally, corrected concept. As stated above, NaPB administration in
gene transcription associated with apoptosis and ALS patients improved molecular pathological as-
mitochondrial dysfunction in these mice, two addi- pects of the disease (32).
tional pathophysiological mechanisms thought to ALS remains an untreatable and uniformly lethal
play a role in neuronal death in ALS. These findings disease. HDAC inhibitors target multiple pathways
provided the rationale for a safety and tolerability important in ALS pathogenesis. Our data strengthen
trial using NaPB in ALS patients. The results of this the hypothesis that transcription dysfunction may
work showed that NaPB was safe and tolerable (32). play a role in the pathogenesis of ALS, and suggest
Interestingly, a pharmacodynamic analysis showed that therapies aimed at improving histone acetyla-
that histone acetylation was decreased by approxi- tion and transcription may provide a novel treatment
mately 50% in blood buffy-coat specimens at screen- strategy that translates to clinical benefits in patients
ing from enrolled patients and was significantly with ALS. Most importantly, the combined treat-
increased after four weeks of NaPB administration ment using riluzole and NaPB shows no negative
and throughout the remainder of the 20-week trial. impact, compared to either agent alone. NaPB has
These findings parallel those observed in the ALS been used clinically to treat patients with a number
Combined riluzole and sodium phenylbutyrate therapy 93

of medical conditions and its pharmacokinetics and slices of hippocampal area CA1. Eur J Pharmacol.
side-effects are well defined in humans (46,47). 1993;250:4736.
17. Wang SJ, Wang KY, Wang WC. Mechanisms underlying
Thus, NaPB remains as a compelling candidate for riluzole inhibition of glutamate release from rat cerebral
testing in humans with ALS. cortex nerve terminals (synaptosomes). Neuroscience.
2004;125:191201.
18. Albo F, Pieri M, Zona C. Modulation of AMPA receptors in
spinal motor neurons by the neuroprotective agent riluzole.
Acknowledgements
J Neurosci Res. 2004;78:2007.
This work was supported by a Merit Review Award 19. Hubert JP, Delumeau JC, Glowinski J, Premont J, Doble A.
from the Veterans Administration (RJF and MEC). Antagonism by riluzole of entry of calcium evoked by NMDA
and veratridine in rat cultured granule cells: evidence for a
Disclosure of interests: The authors have had no dual mechanism of action. Br J Pharmacol. 1994;113:2617.
involvements that might raise the question of bias in 20. Fumagalli E, Bigini P, Barbera S, de Paola M, Mennini T.
the work reported or in the conclusions, implications Riluzole, unlike the AMPA antagonist RPR119990, reduces
or opinions stated. motor impairment and partially prevents motor neuron death
in the wobbler mouse, a model of neurodegenerative disease.
Exp Neurol. 2006;198:11428.
21. Kriz J, Gowing G, Julien JP. Efficient three-drug cocktail for
References disease induced by mutant superoxide dismutase. Ann
1. Rowland L, Schneider N. Amyotrophic lateral sclerosis. N Neurol. 2003;53:42936.
Engl J Med. 2001;344:1688700. 22. Snow RJ, Turnbull J, da Silva S, Jiang F, Tarnopolsky MA.
2. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Creatine supplementation and riluzole treatment provide
Hentati A, et al. Mutations in Cu/Zn superoxide dismutase similar beneficial effects in copper, zinc superoxide dismutase
gene are associated with familial amyotrophic lateral sclerosis. (G93A) transgenic mice. Neuroscience. 2003;119:6617.
Nature. 1993;362:5962. 23. Waibel S, Reuter A, Malessa S, Blaugrund E, Ludolph AC.
3. Andersen PM, Sims KB, Xin WW, Kiely R, O’Neill G, Ravits Rasagiline alone and in combination with riluzole prolongs
J, et al. Sixteen novel mutations in the Cu/Zn superoxide survival in an ALS mouse model. J Neurol. 2004;251:
dismutase gene in amyotrophic lateral sclerosis: a decade of 10804.
discoveries, defects and disputes. Amyotroph Lateral Scler 24. Graf M, Ecker D, Horowski R, Kramer B, Riederer P,
Other Motor Neuron Disord. 2003;4:6273. Gerlach M, et al. High dose vitamin E therapy in amyo-
4. Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, trophic lateral sclerosis as add-on therapy to riluzole: results
Alexander DD, et al. Motor neuron degeneration in mice that
of a placebo-controlled double-blind study. J Neural Transm.
express a human Cu/Zn superoxide dismutase mutation.
2005;112:64960.
Science. 1994;264:17725.
25. Malaspina A, Kaushik N, De Beleroche J. Differential
5. Ryu H, Ferrante RJ. Translational therapeutic strategies in
expression of 14 genes in amyotrophic lateral sclerosis spinal
amyotrophic lateral sclerosis. Mini Rev Med Chem.
cord detected using gridded cDNA arrays. J Neurochem.
2007;7:14150.
6. DiBernardo AB, Cudkowicz ME. Translating preclinical 2001;77:13245.
insights into effective human trials in ALS. Biochim Biophys 26. Ishigaki S, Niwa J, Ando Y, Yoshihara T, Sawada K, Doyu M,
Acta. 2006;1762:113949. et al. Differentially expressed genes in sporadic amyotrophic
7. Gurney ME, Fleck TJ, Himes CS, Hall ED. Riluzole lateral sclerosis spinal cords: screening by molecular indexing
preserves motor function in a transgenic model of familial and subsequent cDNA microarray analysis. FEBS Lett.
amyotrophic lateral sclerosis. Neurology. 1998;50:626. 2002;531:3548.
8. Ryu H, Smith K, Camelo SI, Carreras I, Lee J, Iglesias AH 27. Gonzalez de Aguilar JL, Gordon JW, Rene F, de Tapia M,
et al. Sodium phenylbutyrate prolongs survival and regulates Lutz-Bucher B, Gaiddon C, et al. Alteration of Bcl-x/Bax
expression of anti-apoptotic genes in transgenic amyotrophic ratio in a transgenic mouse model of amyotrophic lateral
lateral sclerosis mice. J Neurochem. 2005;93:108798. sclerosis: evidence for the implication of the p53 signaling
9. Wu AS, Kiaei M, Aguirre N, Crow JP, Calingasan NY, pathway. Neurobiol Dis. 2000;7:40615.
Browne SE, et al. Iron porphyrin treatment extends survival 28. Olsen MK, Roberds SL, Ellerbrock BR, Fleck TJ, McKinley
in a transgenic animal model of amyotrophic lateral sclerosis. DK, Gurney ME. Disease mechanisms revealed by transcrip-
J Neurochem. 2003;85:14250. tion profiling in SOD1-G93A transgenic mouse spinal cord.
10. Klivenyi P, Ferrante RJ, Matthews RT, Bogdanov MB, Klein Ann Neurol. 2001;50:73040.
AM, Andreassen OA, et al. Neuroprotective effects of 29. Cremer JE, Lai JC, Sarna GS. Rapid blood brain transport
creatine in a transgenic animal model of amyotrophic lateral and metabolism of butyrate and pyruvate in the rat after
sclerosis. Nat Med. 1999;5:34750. portocaval anastomosis. J Physiol. 1977;266:701.
11. Olney JW, de Gubareff T. Glutamate neurotoxicity and 30. Collins AF, Pearson HA, Giardina P, McDonagh KT,
Huntington’s chorea. Nature. 1978;271:5579. Brusilow SW, Dover GJ. Oral sodium phenylbutyrate therapy
12. van den Bosch L, van Damme P, Bogaert E, Robberecht W. in homozygous beta thalassemia: a clinical trial. Blood.
The role of excitotoxicity in the pathogenesis of amyotrophic
1995;85:439.
lateral sclerosis. Biochim Biophys Acta. 2006;1762:106882.
31. Vigushin DM, Coombes RC. Histone deacetylase inhibitors
13. Bensimon G, Lacomblez L, Meininger V. A controlled trial of
in cancer treatment. Anticancer Drugs. 2002;13:113.
riluzole in amyotrophic lateral sclerosis. N Engl J Med.
32. Cudkowicz ME, Andres PL, Schoenfeld D, Zhang H, Sher-
1994;330:58591.
man A, Yu H, et al. Safety and dose escalating study of oral
14. Lacomblez L, Bensimon G, Leigh PN, Guillet P, Meininger
sodium phenylbutyrate in subjects with ALS. Neurology.
V. Dose-ranging study of riluzole in amyotrophic lateral
sclerosis. Lancet. 1996;347:142531. 2007;68(Suppl 1):A90.
15. Doble A. The pharmacology and mechanism of action of 33. Ferrante RJ, Klein AM, Dedeoglu A, Beal MF. Therapeutic
riluzole. Neurology. 1996;47(Suppl 4):S23341. efficacy of EGb761 (Gingko biloba extract) in a transgenic
16. Martin D, Thompson MA, Nadler JV. The neuroprotective mouse of amyotrophic lateral sclerosis. J Mol Neurosci.
agent riluzole inhibits release of glutamate and aspartate from 2001;17:8996.
94 S. J. Del Signore et al.

34. Wang CY, Mayo MW, Baldwin Jr AS. TNF- and cancer evidence of slowing of the functional decline. Arch Neurol.
therapy-induced apoptosis: potentiation by inhibition of NF- 2002;59:154150.
kappa B. Science. 1996;274:7847. 42. Ferrante KL, Shefner J, Zhang H, Betensky R, O’Brien M,
35. Mayo MW, Denlinger CE, Broad RM, Yeung F, Reilly ET, Yu H, et al. Tolerance of high-dose (3000 mg/day) coenzyme
Shi Y, et al. Ineffectiveness of histone deacetylase inhibitors Q10 in ALS. Neurology. 2005;65:18346.
to induce apoptosis involves the transcriptional activation of 43. Egorin MJ, Yuan ZM, Sentz DL, Plaisance K, Eiseman JL.
NF-kB through the Akt pathway. J Biol Chem. Plasma pharmacokinetics of butyrate after intravenous ad-
2003;278:189809. ministration of sodium butyrate or oral administration of
36. Pennypacker KR, Kassed CA, Eidizadeh S, Saporta S, tributyrin or sodium butyrate to mice and rats. Cancer
Sanberg PR, Willing AE. NF-kB p50 is increased in neurons Chemother Pharmacol. 1999;43:44553.
surviving hippocampal injury. Exp Neurol. 2001;172:307 44. Chang J-G, Hsieh-Li H-M, Jong Y-J, Wang NM, Tsai CH, Li
19. H. Treatment of spinal muscular atrophy by sodium butyrate.
37. Bruijn LI, Miller TM, Cleveland DW. Unraveling the Proc Natl Acad Sci. 2001;98:980813.
mechanisms involved in motor neuron degeneration in 45. Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B,
ALS. Annu Rev Neurosci. 2004;27:72349. et al. Histone deacetylase inhibition by sodium butyrate
38. Choudry RB, Cudkowicz ME. Clinical trials in amyotrophic chemotherapy ameliorates the neurodegenerative phenotype
lateral sclerosis: the tenuous past and the promising future. in Huntington’s disease mice. J Neurosci. 2003;23:941827.
J Clin Pharmacol. 2005;45:133444. 46. Warrell RPJr, He LZ, Richon V, Calleja E, Pandolfi PP.
39. Shefner JM, Cudkowicz ME, Schoenfeld D, Conrad T, Taft Therapeutic targeting of transcription in acute promyelocytic
J, Urbinelli L, et al. A clinical trial of creatine in ALS. leukemia by use of an inhibitor of histone deacetylase. J Natl
Neurology. 2004;63:165661. Cancer Inst. 1998;90:16215.
40. Huntington Study Group. A randomized, placebo-controlled 47. Gilbert J, Baker SD, Bowling MK, Grochow L, Figg WD,
trial of coenzyme Q10 and remacemide in Huntington’s Zabelina Y, et al. A phase I dose escalation and bioavailability
disease. Neurology. 2001;57:397404. study of oral sodium phenylbutyrate in patients with refrac-
41. Shults CW, Oakes D, Kieburtz K, Beal MF, Haas R, Plumb tory solid tumor malignancies. Clin Cancer Res.
S, et al. Effects of coenzyme Q10 in early Parkinson’s disease: 2001;7:22922300.

You might also like