Clinical Trials for Huntington Disease - Practical Neurology

Page created by Arnold Hartman
 
CONTINUE READING
Clinical Trials for Huntington Disease - Practical Neurology
S P E C I A L R E P OR T

Clinical Trials for
Huntington Disease
The promise for potential disease‑modifying research is strong, although
continued discovery uncovers new questions and challenges to overcome.
By Natalia P. Rocha, PharmD, MSc, PhD; Gabriela D. Colpo, PhD;
Antonio L. Teixeira, MD, PhD, MSc; and Erin Furr Stimming, MD

                                Huntington disease (HD) is an        improving function and quality of life for people with HD.
                                inherited neurodegenerative          Pharmacologic options for the motor symptoms include
                                disease characterized by a clini‑    neuroleptics, gabaergics, and antiglutaminergics, all of which
                                cal triad of motor, cognitive, and   are used off‑label. The vesicular monoamine transporter 2
                                psychiatric symptoms. Common         (VMAT2) inhibitors, tetrabenazine and deutetrabenazine, are
                                motor symptoms include cho‑          the only 2 medications currently approved by the Food and
                                rea, dystonia, and incoordina‑       Drug Administration (FDA) for the treatment of chorea in HD.
                                tion. The cognitive symptoms         Antidepressants, neuroleptics, and mood stabilizers are often
                                are primarily subcortical, which     used to treat neuropsychiatric symptoms. Interdisciplinary,
                                results in a dysexecutive syn‑       nonpharmacologic treatments are also important, including
                                drome. Common neuropsychi‑           physical, occupational, and speech therapy, psychotherapy,
                                atric symptoms include apathy,       genetic counseling, nutrition and social work services.4
anosognosia, irritability, impulsivity, depression, and anxiety.1
   The autosomal dominant inheritance pattern of HD has              Investigational Disease‑Modifying Treatments
been known for decades. The specific mutation, however, was              Initial attempts to develop DMTs for HD focused on cel‑
described over 26 years ago and is a highly penetrant CAG            lular mechanisms associated with neuronal death, such as glu‑
trinucleotide repeat expansion in the huntingtin (HTT) gene          tamate‑mediated excitotoxicity, and apoptosis. Subsequently,
(chromosome 4 (4p16.3)]).2 The CAG repeat expansion in               other neurodegenerative mechanisms were targeted, includ‑
exon 1 is translated into an expanded polyglutamine resulting        ing inflammatory and immune mechanisms, oxidative stress,
in a mutant HTT protein (mHTT). Ubiquitous at the tissue             and mitochondrial dysfunction. Despite promising results
and subcellular level, wild‑type HTT (wtHTT) has numerous            in preclinical studies, none of these have yet demonstrated
functions, not all of which are understood, including tissue         efficacy as DMTs for HD. Several factors may have influenced
maintenance, cell morphology and survival, and control of            these disappointing results, including the lack of appropriate
organelle and vesicle transport. In the nucleus, HTT may act         clinical and biologic markers for selecting participants and
as a scaffold for transcriptional complexes. Of note, the HTT        tracking disease progression and modification. In addition, the
gene is thought to be necessary for embryonic development.3          power of the studies and clinical scales used as primary end‑
   The identification of the huntingtin gene, previously known       points may not have been sensitive enough to detect changes
as interesting transcript 15 (IT15) has been a major milestone       in disease progression. More recently, strategies have shifted
in HD research and several disease‑modifying therapies               from nondisease specific neurodegeneration to interventions
(DMTs) have been studied since its discovery, although none          targeting core upstream disease‑specific processes.5 This revo‑
have yet proven efficacy or been approved.4 Various com‑             lutionary change has been made possible by rapid advances
pounds have been tested, including novel approaches target‑          in gene‑modifying technology. Current and future trials are
ing immune dysregulation in HD, mitochondrial‑based pro‑             focused on reducing the amount of mHTT (Figure).
tective strategies, and gene‑modifying technologies (Table).
                                                                     Lowering Mutant Huntingtin Levels
Symptomatic Treatments                                                  Lowering mHTT levels is among the most promising
  Current treatments are purely symptomatic, aimed at                strategies for developing DMTs for HD. These strategies are

                                                                                                    JUNE 2020 PRACTICAL NEUROLOGY 69
Clinical Trials for Huntington Disease - Practical Neurology
SP E C I A L R EPO R T

          TABLE. CLINICAL TRIALS OF POTENTIAL DISEASE‑MODIFYING THERAPIES FOR HUNTINGTON’S DISEASE
       Phase/designa       Intervention/treatment                       Study status; Outcome measures                            NCT
                                                      Huntingtin‑Lowering Therapies
   Phase 1/2a 13‑wk with   Intrathecal second‑gener‑     Completed; showed a dose‑dependent reduction in CSF levels of         NCT02519036
   14‑month OLE (n=46)     ation 2’‑O‑meth‑oxyethyl      mutant HTT; no serious adverse events were reported6                  NCT03342053
                           chimeric ASOs (10, 30, 60,
                           90, 120 mg) every 4 or 8 wks
   Phase 3 5‑year OLE      Tominersen every 8 or         Recruiting; long‑term safety/tolerability of tominersen in people     NCT03842969
   (n=1,050)               16 wks                        with HD who were in prior studies
   Phase 3 (n=909)         Tominersen every 8 or         Recruiting; efficacy and safety of tominersen in manifest HD          NCT03761849
                           16 wks
   Phase 1b/2a (n=48)      Intrathecal stereopure ASO Recruiting; safety, tolerability, pharmacokinetics, and pharmacody‑      NCT03225833
                           (single and multiple doses) namics of single/multiple doses of stereopure ASO in adults with
                                                         early HD with single nucleotide polymorphism (SNP) rs362307
   Phase 1b/2a (n=48)      Intrathecal injections of     Recruiting; safety, tolerability, pharmacokinetics, and pharmacody‑   NCT03225846
                           stereopure ASO (single and namics of single/multiple doses of stereopure ASO in adults with
                           multiple doses)               early HD with SNP rs362331
   Phase 1/2 5‑year with   Striatally‑administered       Recruiting; evaluate the safety and potential efficacy                NCT04120493
   3.5‑year OLE (n=26)     recombinant HTT
                           (rAAV5‑miHTT)
                                             Huntingtin Lowering and Modulation Therapies
   Phase 2 26‑week         Second‑generation             Completed; showed improvement in executive function with              NCT01590888
   (n=109)                 8‑hydroxyquinoline analog,    250 mg treatment vs placebo; well tolerated and safe15
                           100 or 250 mg/d
   Phase 1 14‑day (n=55)   Selisistat 10 or 100 mg/d     Completed; showed safety and tolerability in adults with early  NCT01485952
                                                         stage HD at plasma concentrations beneficial in animal models18
   Phase 2 12‑week         Selisistat 50 or 200 mg/d     Completed; showed safety and tolerability with a trend toward NCT01521585
   (n=144)                                               modulated plasma levels of soluble mutant huntingtin19
   Phase 1b 14‑day         Selisistat 100 mg/d           Completed; no results published or posted                       NCT01485965
   open‑label paral‑                                     Groups are fasting vs nonfasting
   lel‑group (n=26)
                                      Immunomodulatory and Anti‑inflammatory Therapies
   Phase 2 12‑month        Laquinimod 0.5, 1.0 or Completed; reduced caudate and whole‑brain atrophy (most                     NCT02215616
   (n=352)                 1.5 mg/d               evident in participants with early HD) but no change in UHDRS
   Phase 2 (n=301)         Pepinemab (humanized     Active, not recruiting; safety, tolerability, immunogenicity, and          NCT02481674
                           antiSEMA4D MAb)          efficacy of pepinemab in prodromal and early manifest HD
                                    Metabolic‑ and Mitochondrial‑Based Protective Strategies
   Phase 3 60‑month        Coenzyme Q10 (CoQ10)     Terminated; futility analysis failed to show likelihood of benefit;        NCT00608881
   (n=609)                 240 mg/d                 generally safe and well tolerated24
   Phase 1/2 16‑week       Creatine 8 g/d                Completed; showed safety and tolerability, reduced levels of        NCT00026988
   (n=64)                                                serum 8‑hydroxy‑2’‑deoxyguanosine levels (indicatory of DNA
                                                         oxidative injury), increased creatine (serum/brain) that returned
                                                         to baseline after washout, but no change in UHDRS
   Phase 2 6‑month        Creatine monohydrate           Completed; showed safety and tolerability; slowed cortical and      NCT00592995
   (n=64); 12‑ (n=38) and 30 g/d                         striatal atrophy at 6 and 18 months but no clinical improvement NCT01411150
   24‑month OLEs (n=24)                                                                                                      NCT01411163
                                                                                                                 Table continued on page 71

70 PRACTICAL NEUROLOGY JUNE 2020
Clinical Trials for Huntington Disease - Practical Neurology
S P E C I A L R E P OR T

     TABLE. CLINICAL TRIALS OF POTENTIAL DISEASE‑MODIFYING THERAPIES FOR HUNTINGTON’S DISEASE (CONT.)
      Phase/designa                        Intervention/treatment                                       Study status/outcome measures                             NCT
                            Metabolic‑ and Mitochondrial‑Based Protective Strategies (cont.)
Phase 2 44‑month OLE Creatine monohydrate      Completed ; showed safety and tolerability in adults with HD                                                  NCT01412151
(n=10)               ≤30 g/d
Phase 3 48‑month                         Creatine monohydrate                             Terminated; interim analysis showed lack of promise for slow‑      NCT00712426
(n=553)                                  ≤40 g/d                                          ing disease progression in patients with early‑HD27
Phase 3 6‑month study Ethyl‑EPA, 2 g/d                                                    Completed; showed that ethyl‑EPA did not improve motor             NCT00146211
(n=316)                                                                                   function, global functioning cognition, or global impression28
Abbreviations: ASO, antisense oligonucleotide; EPA, eicosapentaenoic acid; HD, Huntington disease; HTT, huntingtin; MAb, monoclo‑
nal antibody; OLE, open‑label extension; SEMA4D, semaphorin 4D; UHDRS, Unified Huntington Disease Rating Scale.
a Studies are double‑blind randomized placebo‑ or sham‑controlled unless otherwise noted.

focused on inhibiting messenger RNA (mRNA) synthesis by                                                  conserved protein with several important cellular functions,3
blocking transcription with zinc finger motif proteins, avoid‑                                           it is unclear if wtHTT reduction has long‑term safety. In this
ing posttranscriptional processing to increase mRNA degra‑                                               regard, the use of an allele‑specific ASO is an interesting alterna‑
dation with antisense oligonucleotides (ASOs), and inhibit‑                                              tive. There are 2 randomized double‑blind placebo‑controlled
ing mRNA translation with small interfering RNA (siRNA).5                                                phase 1a/2b trials studying safety, tolerability, pharmacokinetics,
   Antisense Oligonucleotides. Designed to target mRNA, ASOs                                             and pharmacodynamics of intrathecally administered allele‑spe‑
are synthetic, short, single‑stranded DNA sequences that form                                            cific ASOs in people with early manifest HD (Table). Allele
a hybridized complex with specific mRNAs causing them to be                                              specificity has been achieved by developing stereopure ASOs
degraded through an RNAse‑H1 mechanism. The first human                                                  directed at specific single nucleotide polymorphisms (SNPs).
clinical trial with ASO therapy for lowering HTT evaluated the                                           Both trials are enrolling participants with specific HTT SNPs on
safety, tolerability, pharmacokinetics, and pharmacodynam‑                                               the same allele as the pathogenic CAG expansion.
ics of multiple ascending doses of tominersen administered                                                   Small Interfering RNAs. The siRNA and artificial microRNA
intrathecally to 46 participants with early manifest HD at                                               (miRNA) neutralize mRNA molecules to inhibiting mRNA
4‑week intervals for 13 weeks. Tominersen is nonallele‑specific                                          translation, a process known as RNA interference (RNAi).7,8
and thus targets both wtHTT and mHTT. Preliminary results                                                Using the adeno‑associated viral (AAV) vector to deliver
suggest that tominersen is safe, and well tolerated. At the                                              an siRNA targeting mHTT mRNA in a mouse model of HD,
90‑mg and 120‑mg doses, tominersen was associated with an                                                more than 80% of the cells in the striatum were successfully
approximately 40% reduction of mHTT levels in cerebrospinal                                              transduced (ie, expressed the siRNA). Of note, there were also
fluid (CSF), which may correspond to a 55% to 85% reduction                                              significant improvements in HD‑associated behavioral deficits
in cortical mHTT levels. There was also a correlation between                                            and the reduction of striatal HTT aggregates in the transfected
reduced CSF mHTT levels and improved composite Unified                                                   mice.9 A cholesterol‑conjugated siRNA targeting HTT (cc‑siR‑
Huntington Disease Rating Scale (cUHDRS) scores.6                                                        NA‑HTT) ameliorated HD‑related neuropathology and motor
   There are 2 open‑label extensionsa underway for partici‑                                              deficits in a different mouse model.8 An artificial miRNA deliv‑
pants who have previously received tominersen and a phase 3                                              ered via the AAV vector to a sheep model of HD also reduced
double‑blind placebo‑controlled interventional trial evaluat‑                                            50% to 80% of HTT mRNA and protein in the striatum and
ing safety and efficacy in individuals with manifest HD along                                            prevented subsequent neuron loss at 1 and 6 months after
with a natural history study. The primary outcome measure                                                injection. The sheep model results show that effective silencing
for the pivotal trial is the total functional capacity (TFC) in                                          of mHTT by AAV‑delivered artificial miRNA can be achieved
the US and the cUHDRS outside the US. Secondary outcomes                                                 and sustained in a large‑animal brain.10 Another mouse model
include additional components of the UHDRS, including cog‑                                               injected with an AAV‑delivered microRNA (miRNA) for
nitive and behavioral assessments, the clinical global impres‑                                           human HTT mRNA selectively knocks down expression and is
sion score, adverse events, pharmacokinetic markers, CSF                                                 most effectively delivered to the putamen and thalamus with
mHTT, neurofilament (NfL) levels, and brain volume on MRI.                                               MRI‑guided injection and reduced HTT levels in deep brain tis‑
   Tominersen has demonstrated reduction in CSF HTT levels,                                              sues, such as the caudate, putamen, and thalamus, in addition
and efficacy of the treatment is under investigation. It is unclear,                                     to the cortex. This treatment was well tolerated by the animals
however, if residual mHTT levels and lowering both wtHTT and                                             for up to 5 weeks and phase 1 clinical trials in humans with HD
mHTT levels in the CSF are clinically relevant. HTT is a highly                                          are expected to begin soon.11
a. See table for NCT identifiers of all clinical trials mentioned without published results

                                                                                                                                          JUNE 2020 PRACTICAL NEUROLOGY 71
Clinical Trials for Huntington Disease - Practical Neurology
SP E C I A L R EPO R T

                                                                         ed HTT and reversed the HD‑associated neuropathology and
                                                                         behavioral phenotypes. Decreased mHTT expression did not
                                                                         affect cell viability, but alleviated motor deficits.13 Despite the
                                                                         positive results in preclinical studies, however, several issues
                                                                         must be addressed before bringing CRISPR/Cas9 technologies
                                                                         into humans, including that it is irreversible with ethical con‑
                                                                         cerns regarding germline alteration and that there are delivery
                                                                         problems with viral vectors and concerns about the possible
                                                                         immunogenicity of bacterial proteins.14

                                                                         Huntingtin Protein Modulation
                                                                            Strategies for HTT protein modulation include clearance
                                                                         and inhibition of protein aggregation. Excessive metal concen‑
                                                                         trations induce mHTT aggregation and promote formation of
                                                                         reactive oxygen species (ROS). Therefore, it has been hypoth‑
                                                                         esized that 8‑hydroxyquinoline transition metal ligand, which
                                                                         redistributes metals (eg, copper, zinc, and iron) from locations
                                                                         where they are abundant to subcellular locations where they
                                                                         might be deficient, could attenuate mHTT effects by inhib‑
                                                                         iting metal‑induced aggregation.15 In preclinical studies in
                                                                         Caenorhabditis elegans and mouse models of HD, 8‑hydroxy‑
                                                                         quinoline transition metal ligand was effective in ameliorat‑
                                                                         ing the HD phenotype.16 In a 26‑week phase 2 randomized
                                                                         double‑blind placebo‑controlled clinical trial, 109 participants
                                                                         with HD had significant improvement in cognition (Trail
                                                                         Making Test Part B) with a 250‑mg dose, but no effects in
                                                                         other domains were seen compared with the 100‑mg dose
                                                                         or placebo. This isolated finding was thought to be of limited
   Fig. 1: Strategies for Lowering Mutant Huntingtin Levels. Different   clinical relevance.15 Although the treatment was considered
   approaches for lowering mutant huntingtin (mHTT) include              safe and well tolerated, the FDA issued a partial clinical hold
   using clustered regularly interspaced short palindromic repeats       because of safety concerns for the 250‑mg dose and required
   (CRISPR)‑Cas9 to edit DNA so mHTT is not transcribed, antisense       more neurotoxicity data. There have been no recent updates.
   oligonucleotides (ASOs) to reduce production of mRNA during              Selisistat has been studied for promotion of HTT protein
   processing (splicing) of pre-mRNA, microscopic or small interfering   clearance. Selisistat is an inhibitor of the silent information reg‑
   RNA (miRNA or siRNA) to reduce production of mHTT protein dur‑        ulator T1 (SirT1), which is a member of the sirtuin deacetylase
   ing mRNA translation, and protein modulation to reduce aggrega‑       family that can remove acetyl groups from mHTT. Inhibition
   tion or increase clearance.                                           of SirT1 alleviated pathology in animal and cell models of HD.17
                                                                         Clinical trials of selisistat demonstrated that it is safe and well
      Similarly, an engineered miRNA delivered via AAV vector            tolerated but failed to show efficacy.18,19 Another trial has been
   serotype 5 (AAV5‑miHTT) in a single intrastriatal injection into      registered that will assess selisistat in fasted vs fed conditions
   a minipig model of HD resulted in a strong, widespread and            (ie, the effect of food on the pharmacokinetics of selisistat in
   sustained (up to 12 months) reduction of mHTT levels and a            patients with HD). The study was designed to explore poten‑
   phase 1 trial of this agent in humans with HD is also beginning.      tial biomarkers for use in subsequent phase 2/3 studies, but no
      Genome Editing. The clustered regularly interspaced short          results have been published to date. There are no phase 3 trials
   palindromic repeats (CRISPR)‑associated (Cas)9 system has             planned; further development appears to be on hold.
   emerged as a promising option because of allele‑specificity and
   potency. The CRISPR/Cas9 technology was used in fibroblasts           Neuroprotective Approaches
   of an HD patient to delete the promoter regions, transcription           Immune dysregulation and mitochondrial dysfunction are
   start site, and the CAG mutation expansion of the mHTT gene.          regarded as important contributors to neurodegeneration in
   Complete inactivation of the mutant allele occurred without           HD and have been targets for HD‑modifying therapies.
   effects on the normal allele.12 The CRISPR/Cas9 method has               Immunomodulatory Agents. The immunomodulatory
   also been tested in an HD rodent model and efficiently deplet‑        strategies for HD include laquinimod, an immunomodulatory

72 PRACTICAL NEUROLOGY JUNE 2020
Clinical Trials for Huntington Disease - Practical Neurology
S P E C I A L R E P OR T

drug originally studied for the treatment of relapsing‑remit‑       eration, immune/inflammatory mechanisms, glutamate‑medi‑
ting multiple sclerosis; minocycline, a second‑generation tetra‑    ated excitotoxicity, the dopamine pathway, and mitochondrial
cycline that has been in therapeutic use for over 30 years; and     dysfunction. None have yet been successful in identifying a
pepinemab, an investigational antibody to semaphorin 4D             drug with proven efficacy to modify HD progression; however,
(SEM4D). Neuroprotective and HD‑ameliorating effects of             pending results may be positive. Several factors might influence
laquinimod in preclinical studies20‑22 motivated clinical trials    the previous disappointing results, including underpowered
in participants with HD. In randomized placebo‑controlled           studies and the lack of appropriate clinical and biological
double‑blind trials, neither 12 weeks of laquinimod or 8 weeks      markers capable of tracking disease modification. The clini‑
of minocycline vs placebo showed efficacy as measured by            cal scales currently used as primary endpoints might not be
change in the UHDRS or cognitive test scores. The laquini‑          sensitive enough at detecting signs and symptoms of disease
mod treatment, however, was correlated with a significant           progression. New innovative tools to objectively assess disease
reduction in caudate and whole‑brain atrophy that was most          signs and symptoms and the development of neuroimag‑
evident in people with early manifest HD. At this time, it is       ing‑based or biological fluid‑based markers of disease progres‑
unclear how a positive MRI‑based secondary outcome in               sion will be imperative to the success of clinical trials in HD.
the setting of a negative clinical primary outcome should be           The recent advances in therapeutic strategies promise an
interpreted. It is possible that laquinimod would be more           exciting era for clinical trials in HD. Current and future trials
efficient if it were initiated years (or even decades) before       are focused on lowering mHTT by targeting mHTT produc‑
disease onset, thus preventing neurodegeneration. Regarding         tion, aggregation, misfolding, and removal. The ASOs are
minocycline, the short‑period treatment could explain the           the furthest along in clinical development in the realm of
lack of efficacy; however, no results were seen in another trial    huntingtin‑lowering strategies; however, many questions
of 18 months of minocycline treatment.23                            remain, particularly long‑term safety and the effect of decreas‑
   Pepinemab is an antibody against SEM4D thought to                ing wtHTT levels. The results of studies using CRISPR/Cas9
decrease central nervous system inflammation, increase              technologies in preclinical models of HD are very encouraging,
neuronal outgrowth, and enhance oligodendrocyte matura‑             yet these techniques are distant from use in humans. In sum‑
tion. Pepinemab is being evaluated in a phase 2 multicenter         mary, the landscape is changing for many inherited condi‑
randomized double‑blind placebo‑controlled study assessing          tions due to recent technological advances in gene modifying
safety, tolerability, pharmacokinetics, and efficacy in partici‑    approaches. The future for HD disease‑modifying research is
pants with prodromal or early manifest HD.                          promising, although with continued discovery, new questions
   Mitochondrial‑Based Agents. Several clinical trials have eval‑   and challenges emerge. n
uated antioxidant treatment with coenzyme Q10 (CoQ10)               1. Ross CA, Aylward EH, Wild EJ, et al. Huntington disease: natural history, biomarkers and prospects for therapeutics. Nat Rev
and creatinine as potential DMTs for HD. Although both                   Neurol. 2014;10(4):204‑216.
                                                                    2. The Huntington’s Disease Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and
CoQ10 and creatinine were generally safe and well toler‑                 unstable on Huntington’s disease chromosomes. Cell. 1993;72(6):971‑983.
ated, neither slowed functional decline in HD and the trials        3. Saudou F, Humbert S. The biology of huntingtin. Neuron. 2016;89(5):910‑926.
                                                                    4. Testa CM, Jankovic J. Huntington disease: a quarter century of progress since the gene discovery. J Neurol Sci. 2019;396:52‑68.
were terminated based on interim analyses.24‑27 The effects of      5. Mestre TA. Recent advances in the therapeutic development for Huntington disease. Parkinsonism Relat Disord. 2019;59:125‑130.
ethyl‑eicosapentaenoic acid (ethyl‑EPA) in HD have also been        6. Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, et al. Targeting huntingtin expression in patients with Huntington’s disease. N
                                                                         Engl J Med. 2019;380(24):2307‑2316.
tested but showed no beneficial effects in measures of motor        7. McBride JL, Boudreau RL, Harper SQ, et al. Artificial miRNAs mitigate shRNA‑mediated toxicity in the brain: implications for
function, global functioning cognition, or global impression.28          the therapeutic development of RNAi. Proc Natl Acad Sci U S A. 2008;105(15):5868‑5873.
                                                                    8. DiFiglia M, Sena‑Esteves M, Chase K, et al. Therapeutic silencing of mutant huntingtin with siRNA attenuates striatal and
Latrepirdine (dimebon) was also tested as a modulator of                 cortical neuropathology and behavioral deficits. Proc Natl Acad Sci U S A. 2007;104(43):17204‑17209.
mitochondrial dysfunction in HD. Treatment with latrepirdine        9. Stanek LM, Sardi SP, Mastis B, et al. Silencing mutant huntingtin by adeno‑associated virus‑mediated RNA interference amelio‑
                                                                         rates disease manifestations in the YAC128 mouse model of Huntington’s disease. Hum Gene Ther. 2014;25(5):461‑474.
vs placebo correlated with a discrete but significant improve‑      10. Pfister EL, DiNardo N, Mondo E, et al. Artificial miRNAs reduce human mutant huntingtin throughout the striatum in a
ment in the Mini‑Mental State Examination (MMSE), but not                transgenic sheep model of Huntington’s disease. Hum Gene Ther. 2018;29(6):663‑673.
                                                                    11. Zhou P, Carroll J, Chen F, et al. Robust huntingtin knockdown in cortex and putamen in large mammals using a novel
other cognitive outcome measures in a phase 2 trial.29 In a              dosing paradigm with VY‑HTT01, an AAV gene therapy targeting huntingtin for the treatment of Huntington’s disease.
subsequent multicenter 26‑week randomized double‑blind                   Hum Gene Ther. 2018;27:A83.
                                                                    12. Shin JW, Kim KH, Chao MJ, et al. Permanent inactivation of Huntington’s disease mutation by personalized allele‑specific
placebo‑controlled study, no improvement in cognition or                 CRISPR/Cas9. Hum Mol Genet. 2016;25(20):4566‑4576.
global function compared to baseline was seen in participants       13. Yang S, Chang R, Yang H, et al. CRISPR/Cas9‑mediated gene editing ameliorates neurotoxicity in mouse model of
                                                                         Huntington’s disease. J Clin Invest. 2017;127(7):2719‑2724.
with HD who had cognitive impairments,30 and an open‑label          14. Tabrizi SJ, Ghosh R, Leavitt BR. Huntingtin lowering strategies for disease modification in Huntington’s disease [published
extension was terminated.                                                correction appears in Neuron. 2019 May 22;102(4):899]. Neuron. 2019;101(5)801‑819.
                                                                    15. Huntington Study Group Reach HDI. Safety, tolerability, and efficacy of PBT2 in Huntington’s disease: a phase 2,
                                                                         randomised, double‑blind, placebo‑controlled trial. Lancet Neurol. 2015;14(1):39‑47.
Conclusions                                                         16. Cherny RA, Ayton S, Finkelstein DI, Bush AI, McColl G, Massa SM. PBT2 reduces toxicity in a C. elegans model of polyQ
                                                                         aggregation and extends lifespan, reduces striatal atrophy and improves motor performance in the R6/2 mouse model of
   Based on promising data from preclinical studies, multiple            Huntington’s disease. J Huntingtons Dis. 2012;1(2):211‑219.
clinical trials have been conducted. targeting different aspects    17. Smith MR, Syed A, Lukacsovich T, et al. A potent and selective Sirtuin 1 inhibitor alleviates pathology in multiple animal and
of HD pathology, including mHTT aggregation, neurodegen‑                                                                                              (Continued on page 76)

                                                                                                                                   JUNE 2020 PRACTICAL NEUROLOGY 73
S P E C I A L R E P OR T

                                   (Continued from page 73)
                                       cell models of Huntington’s disease. Hum Mol Genet. 2014;23(11):2995‑3007.
                                   18. Sussmuth SD, Haider S, Landwehrmeyer GB, et al. An exploratory double‑blind, randomized clinical trial with selisistat, a
                                       SirT1 inhibitor, in patients with Huntington’s disease. Br J Clin Pharmacol. 2015;79(3):465‑476.
                                   19. Reilmann R, Squitieri F, Priller J, et al. N02 Safety and tolerability of selisistat for the treatment of Huntington’s disease:
                                       results from a randomised, double‑blind, placebo‑controlled phase II trial.RJ Neurol Neurosurg Psychiatry. 2014;85:A102.
                                   20. Garcia‑Miralles M, Hong X, Tan LJ, et al. Laquinimod rescues striatal, cortical and white matter pathology and results in
                                       modest behavioural improvements in the YAC128 model of Huntington disease. Sci Rep. 2016;6:31652.
                                   21. Li C, Yuan K, Schluesener H. Impact of minocycline on neurodegenerative diseases in rodents: a meta‑analysis. Rev
                                       Neurosci. 2013;24(5):553‑562.
                                   22. Southwell AL, Franciosi S, Villanueva EB, et al. Anti‑semaphorin 4D immunotherapy ameliorates neuropathology and
                                       some cognitive impairment in the YAC128 mouse model of Huntington disease. Neurobiol Dis. 2015;76:46‑56.
                                   23. Huntington Study Group DOMINO Investigators. A futility study of minocycline in Huntington’s disease. Mov Disord.
                                       2010;25(13):2219‑2224.
                                   24. McGarry A, McDermott M, Kieburtz K, et al. A randomized, double‑blind, placebo‑controlled trial of coenzyme Q10 in
                                       Huntington disease. Neurology. 2017;88(2):152‑159.
                                   25. Ross C, Biglan K, Killoran A, et al. PREQUEL—A multicenter phase II study of coenzyme Q10 in premanifest Huntington
                                       disease. Neurotherapeutics. 2014;11:214.
                                   26. Verbessem P, Lemiere J, Eijnde BO, et al. Creatine supplementation in Huntington’s disease: a placebo‑controlled pilot trial.
                                       Neurology. 2003;61(7):925‑930.
                                   27. Hersch SM, Schifitto G, Oakes D, et al. The CREST‑E study of creatine for Huntington disease: a randomized controlled trial.
                                       Neurology. 2017;89(6):594‑601.
                                   28. Huntington Study Group. Randomized controlled trial of ethyl‑eicosapentaenoic acid in Huntington disease: the TREND‑HD
                                       study. Arch Neurol. 2008;65(12):1582‑1589.
                                   29. Kieburtz K, McDermott MP, Voss TS, et al. A randomized, placebo‑controlled trial of latrepirdine in Huntington disease.
                                       Arch Neurol. 2010;67(2):154‑160.
                                   30. Horizon Investigators of the Huntington Study Group, European Huntington’s Disease Network. A randomized, double‑blind,
                                       placebo‑controlled study of latrepirdine in patients with mild to moderate Huntington disease. JAMA Neurol. 2013;70(1):25‑33.

                                     Natalia P. Rocha, PharmD, MsC, PhD
                                     The Mitchell Center for Alzheimer’s disease and Related
                                     Brain Disorders
                                     Department of Neurology, McGovern Medical School
                                     HDSA Center of Excellence
                                     The University of Texas Health Science Center
                                     Houston, TX

                                     Gabriela D. Colpo, PhD
                                     Neuropsychiatry Program
                                     Department of Psychiatry and Behavioral Sciences
                                     McGovern Medical School
                                     The University of Texas Health Science Center
                                     Houston, TX

                                     Antonio L. Teixeira, MD, PhD, MSc
                                     Neuropsychiatry Program
                                     Department of Psychiatry and Behavioral Sciences
                                     McGovern Medical School, HDSA Center of Excellence
                                     The University of Texas Health Science Center
                                     Houston, TX

                                     Erin Furr Stimming, MD
                                     Department of Neurology, McGovern Medical School
                                     HDSA Center of Excellence
                                     The University of Texas Health Science Center
                                     Houston, TX

                                     Disclosures
                                     NPR, GDC, ALT report no disclosures
                                     EFS has disclosures at www.practicalneurology.com

76 PRACTICAL NEUROLOGY JUNE 2020
You can also read