Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 277
Filter
1.
Cells ; 11(23)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36496996

ABSTRACT

The most commonly used treatment for Parkinson's disease (PD) is levodopa, prescribed in conjunction with carbidopa. Virtually all patients with PD undergo dopamine replacement therapy using levodopa during the course of the disease's progression. However, despite the fact that levodopa is the "gold standard" in PD treatments and has the ability to significantly alleviate PD symptoms, it comes with side effects in advanced PD. Levodopa replacement therapy remains the current clinical treatment of choice for Parkinson's patients, but approximately 80% of the treated PD patients develop levodopa-induced dyskinesia (LID) in the advanced stages of the disease. A better understanding of the pathological mechanisms of LID and possible means of improvement would significantly improve the outcome of PD patients, reduce the complexity of medication use, and lower adverse effects, thus, improving the quality of life of patients and prolonging their life cycle. This review assesses the recent advancements in understanding the underlying mechanisms of LID and the therapeutic management options available after the emergence of LID in patients. We summarized the pathogenesis and the new treatments for LID-related PD and concluded that targeting pathways other than the dopaminergic pathway to treat LID has become a new possibility, and, currently, amantadine, drugs targeting 5-hydroxytryptamine receptors, and surgery for PD can target the Parkinson's symptoms caused by LID.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Humans , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/complications , Dyskinesia, Drug-Induced/therapy , Dyskinesia, Drug-Induced/drug therapy , Antiparkinson Agents/adverse effects , Quality of Life , Dopamine
3.
Eur J Neurol ; 28(5): 1574-1580, 2021 05.
Article in English | MEDLINE | ID: mdl-33511741

ABSTRACT

BACKGROUND AND PURPOSE: The purpose was to assess the effect of bilateral subthalamic nucleus deep brain stimulation (STN DBS) on diphasic dyskinesia in patients with Parkinson disease (PD) and to assess the factors associated with the remission of diphasic dyskinesia. METHODS: Medical records for PD patients who underwent bilateral STN DBS at the Movement Disorder Center of Seoul National University Hospital from March 2005 to November 2016 were reviewed. Patients were evaluated preoperatively and at 3, 6 and 12 months after surgery, and annually thereafter. The presence of peak-dose dyskinesia and diphasic dyskinesia is based on the interview and examination of patients at baseline and at each follow-up. RESULTS: Amongst 202 patients who underwent STN DBS, 66 patients who had diphasic dyskinesia preoperatively were included in the analysis. Diphasic dyskinesia disappeared in 49 (74%) after surgery. In 27 (55.1%) patients whose diphasic dyskinesia disappeared after DBS, peak-dose and diphasic dyskinesia disappeared persistently from as early as 3 months postoperatively. Age at onset was younger and disease duration at surgery was longer in patients whose diphasic dyskinesia persisted compared with patients whose diphasic dyskinesia disappeared. Multivariate Cox regression analysis demonstrated that patients with greater postoperative decrease of dopaminergic medications were more likely to have remission of diphasic dyskinesia. CONCLUSION: This study showed that bilateral STN DBS is effective in controlling diphasic dyskinesia and should be considered in PD patients with diphasic dyskinesia.


Subject(s)
Deep Brain Stimulation , Dyskinesia, Drug-Induced , Subthalamic Nucleus , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/therapy , Humans , Levodopa/adverse effects , Treatment Outcome
5.
Ann Clin Transl Neurol ; 7(1): 59-68, 2020 01.
Article in English | MEDLINE | ID: mdl-31813194

ABSTRACT

OBJECTIVE: To compare the efficacy of subthalamic nucleus (STN) and globus pallidus internus (GPi) deep brain stimulation (DBS) on reducing levodopa-induced dyskinesia (LID) in Parkinson's disease, and to explore the potential underlying mechanisms. METHODS: We retrospectively assessed clinical outcomes in 43 patients with preoperative LID who underwent DBS targeting the STN (20/43) or GPi (23/43). The primary clinical outcome was the change from baseline in the Unified Dyskinesia Rating Scale (UDysRS) and secondary outcomes included changes in the total daily levodopa equivalent dose, the drug-off Unified Parkinson Disease Rating Scale Part Ⅲ at the last follow-up (median, 18 months), adverse effects, and programming settings. Correlation analysis was used to find potential associated factors that could be used to predict the efficacy of DBS for dyskinesia management. RESULTS: Compared to baseline, both the STN group and the GPi group showed significant improvement in LID with 60.73 ± 40.29% (mean ± standard deviation) and 93.78 ± 14.15% improvement, respectively, according to the UDysRS score. Furthermore, GPi-DBS provided greater clinical benefit in the improvement of dyskinesia (P < 0.05) compared to the STN. Compared to the GPi group, the levodopa equivalent dose reduction was greater in the STN group at the last follow-up (43.81% vs. 13.29%, P < 0.05). For the correlation analysis, the improvement in the UDysRS outcomes were significantly associated with a reduction in levodopa equivalent dose in the STN group (r = 0.543, P = 0.013), but not in the GPi group (r = -0.056, P = 0.801). INTERPRETATION: Both STN and GPi-DBS have a beneficial effect on LID but GPi-DBS provided greater anti-dyskinetic effects. Dyskinesia suppression for STN-DBS may depend on the reduction of levodopa equivalent dose. Unlike the STN, GPi-DBS might exert a direct and independent anti-dyskinesia effect.


Subject(s)
Antiparkinson Agents/adverse effects , Deep Brain Stimulation , Dyskinesia, Drug-Induced/therapy , Globus Pallidus , Levodopa/adverse effects , Outcome Assessment, Health Care , Parkinson Disease/drug therapy , Subthalamic Nucleus , Aged , Antiparkinson Agents/administration & dosage , Deep Brain Stimulation/methods , Dyskinesia, Drug-Induced/etiology , Female , Follow-Up Studies , Humans , Levodopa/administration & dosage , Male , Middle Aged , Retrospective Studies
6.
Neurosci Lett ; 716: 134653, 2020 01 18.
Article in English | MEDLINE | ID: mdl-31778767

ABSTRACT

Patients with Parkinson's Disease (PD) experience bothersome motor fluctuations and Levodopa-induced Dyskinesias (LIDs). Cerebellar continuous theta burst stimulation (cTBS) was used as an inhibitory protocol of repetitive transcranial magnetic stimulation (rTMS) to reduce LIDs in PD patients. The influence of Val66Met polymorphism of Brain Derived Neurotrophic Factor (BDNF) gene on the therapeutic response to cTBS was investigated and the serum levels of BDNF were measured before and after treatment. Eleven patients were exposed to a session of cTBS and sham stimulation (one week apart) after the administration of 125 % of their usual morning dose of Levodopa and LIDs were video-recorded and evaluated at different time points (0, 15, 30, 45, 60, 90 min after Levodopa). Cerebellar cTBS significantly reduced LIDs with respect to sham stimulation and decreased serum BDNF levels. These effects were evident in the Val66Val group (7 subjects) but not in the Val66Met group (4 subjects). These data confirm the efficacy of cerebellar cTBS in reducing LIDs in PD patients and show that the clinical effect is accompanied by a decrease in serum BDNF levels. Moreover, they suggest that BDNF Val66Met polymorphism may influence the clinical and biological response to cTBS.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Cerebellum/physiopathology , Dyskinesia, Drug-Induced/therapy , Parkinson Disease/therapy , Transcranial Magnetic Stimulation/methods , Aged , Antiparkinson Agents/adverse effects , Brain-Derived Neurotrophic Factor/genetics , Female , Humans , Levodopa/adverse effects , Male , Middle Aged , Parkinson Disease/blood , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Theta Rhythm
7.
Neurochem Int ; 131: 104543, 2019 12.
Article in English | MEDLINE | ID: mdl-31491493

ABSTRACT

Prolonged administration of Levodopa (L-dopa) therapy can generate L-dopa-induced dyskinesia (LID). Accumulating evidence indicates that hyper-activation of the dopamine D1 receptor (D1R) and the cAMP signaling cascade in the medium spiny neurons (MSNs) of the striatum are involved in LID. Previous studies have shown that striatal ß-arrestin2 overexpression significantly reduces LID severity and have indicated that ß-arrestin2 may play a causal role in the dyskinesia sensitization process. L-dopa-induced changes in the expression of signaling molecules and other proteins in the striatum were examined immunohistochemically and by western blot. A rAAV (recombinant adeno-associated virus) vector was used to overexpress and ablate ß-arrestin2. We found that striatal overexpression of AAV-mediated ß-arrestin2 produced less severe AIMs (abnormal involuntary movements) in response to L-dopa, whereas selective deletion of ß-arrestin2 in the striatal neurons dramatically enhanced the severity of dyskinesia induced by L-dopa. Furthermore, no significant improvements in motor behavior (FFT: forelimb functional test) were seen with the inhibition or overexpression of ß-arrestin2. Finally, overexpression of ß-arrestin2 diminished L-dopa-induced D1R and phosphor-DARPP32/ERK levels. Viral deletion of ß-arrestin2 markedly enhanced the key biochemical markers in the direct pathway. We found that increased availability of ß-arrestin2 ameliorated dyskinesia severity with no influence on the anti-Parkinsonian action of L-dopa, suggesting a promising approach for controlling LID in Parkinson's disease. In addition, overexpression of ß-Arrestin2 prevented the development of LID by inhibiting G protein-dependent D1R and phosphor-DARPP32/ERK signaling in dyskinetic rats.


Subject(s)
Antiparkinson Agents , Dyskinesia, Drug-Induced/therapy , Levodopa , Neostriatum/metabolism , Parkinson Disease, Secondary/therapy , beta-Arrestin 2/biosynthesis , beta-Arrestin 2/genetics , Adenoviridae/genetics , Animals , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dyskinesia, Drug-Induced/psychology , Gene Deletion , Genetic Therapy , Genetic Vectors , MAP Kinase Signaling System/drug effects , Male , Neostriatum/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/psychology , Phosphoproteins/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function , Transcription Factors/drug effects
8.
J Neural Transm (Vienna) ; 126(11): 1479-1483, 2019 11.
Article in English | MEDLINE | ID: mdl-31494731

ABSTRACT

Bilateral subthalamic nucleus deep brain stimulation (STN DBS) improves motor fluctuations and dyskinesias in patients with Parkinson's disease (PD). Dyskinesia improvement with STN DBS is believed to result entirely from levodopa reduction. However, some studies suggest that STN DBS may also directly suppress dyskinesias. To determine whether bilateral STN DBS improves dyskinesias beyond what would be expected from levodopa reduction alone, we analyzed pre-operative and post-operative dyskinesia scores (sum of MDS-UPDRS items 4.1 and 4.2) from 61 PD patients with bilateral STN DBS. A multiple regression model (adjusted for disease severity, disease duration, active contacts above the STN, use of amantadine, high pre-operative levodopa-equivalent dose (LED), sex, and interaction between active contacts above the STN and amantadine use) was created to describe the relationship between dyskinesia scores and LED prior to DBS. Using this model, a post-operative dyskinesia score was estimated from post-operative LED and compared to the actual post-operative dyskinesia score. The regression model was statistically significant overall (p = 0.003, R2 = 0.34, adjusted R2 = 0.24). The actual post-operative dyskinesia score (1.0 ± 1.4) was significantly lower than the score predicted by the model (3.0 ± 1.1, p < 0.0001). Dyskinesias after STN DBS improved more than predicted by levodopa reduction alone. Our data support the idea that STN stimulation may directly improve dyskinesias.


Subject(s)
Deep Brain Stimulation , Dopamine Agents/administration & dosage , Dyskinesia, Drug-Induced/therapy , Levodopa/administration & dosage , Outcome Assessment, Health Care , Parkinson Disease/therapy , Subthalamic Nucleus , Aged , Dopamine Agents/adverse effects , Female , Humans , Levodopa/adverse effects , Male , Middle Aged , Parkinson Disease/drug therapy
9.
Lancet Neurol ; 18(9): 880-890, 2019 09.
Article in English | MEDLINE | ID: mdl-31279747

ABSTRACT

A large and ever-growing number of medications can induce various movement disorders. Drug-induced movement disorders are disabling but are often under-recognised and inappropriately managed. In particular, second generation antipsychotics, like first generation agents, are associated with potentially debilitating side-effects, most notably tardive syndromes and parkinsonism, as well as potentially fatal acute syndromes. Appropriate, evidence-based management is essential as these drugs are being prescribed to a growing population vulnerable to these side-effects, including children and elderly people. Prevention of the development of drug-induced movement disorders is an important consideration when prescribing medications that can induce movement disorders. Recent developments in diagnosis, such as the use of dopamine transporter imaging for drug-induced parkinsonism, and treatment, with the approval of valbenazine and deutetrabenazine, the first drugs indicated for tardive syndromes, have improved outcomes for many patients with drug-induced movement disorders. Future research should focus on development of safer antipsychotics and specific therapies for the different tardive syndromes and the treatment of drug-induced parkinsonism.


Subject(s)
Antipsychotic Agents/adverse effects , Dyskinesia, Drug-Induced/diagnosis , Disease Management , Dyskinesia, Drug-Induced/prevention & control , Dyskinesia, Drug-Induced/therapy , Humans
10.
Mov Disord ; 34(12): 1818-1830, 2019 12.
Article in English | MEDLINE | ID: mdl-31335998

ABSTRACT

BACKGROUND: Thalidomide and closely related analogues are used clinically for their immunomodulatory and antiangiogenic properties mediated by the inhibition of the proinflammatory cytokine tumor necrosis factor α. Neuroinflammation and angiogenesis contribute to classical neuronal mechanisms underpinning the pathophysiology of l-dopa-induced dyskinesia, a motor complication associated with l-dopa therapy in Parkinson's disease. The efficacy of thalidomide and the more potent derivative 3,6'-dithiothalidomide on dyskinesia was tested in the 6-hydroxydopamine Parkinson's disease model. METHODS: Three weeks after 6-hydroxydopamine infusion, rats received 10 days of treatment with l-dopa plus benserazide (6 mg/kg each) and thalidomide (70 mg/kg) or 3,6'-dithiothalidomide (56 mg/kg), and dyskinesia and contralateral turning were recorded daily. Rats were euthanized 1 hour after the last l-dopa injection, and levels of tumor necrosis factor-α, interleukin-10, OX-42, vimentin, and vascular endothelial growth factor immunoreactivity were measured in their striatum and substantia nigra reticulata to evaluate neuroinflammation and angiogenesis. Striatal levels of GLUR1 were measured as a l-dopa-induced postsynaptic change that is under tumor necrosis factor-α control. RESULTS: Thalidomide and 3,6'-dithiothalidomide significantly attenuated the severity of l-dopa-induced dyskinesia while not affecting contralateral turning. Moreover, both compounds inhibited the l-dopa-induced microgliosis and excessive tumor necrosis factor-α in the striatum and substantia nigra reticulata, while restoring physiological levels of the anti-inflammatory cytokine interleukin-10. l-Dopa-induced angiogenesis was inhibited in both basal ganglia nuclei, and l-dopa-induced GLUR1 overexpression in the dorsolateral striatum was restored to normal levels. CONCLUSIONS: These data suggest that decreasing tumor necrosis factor-α levels may be useful to reduce the appearance of dyskinesia, and thalidomide, and more potent derivatives may provide an effective therapeutic approach to dyskinesia. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/therapy , Immunologic Factors/therapeutic use , Levodopa/adverse effects , Parkinson Disease/complications , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Animals , Cytokines/metabolism , Dyskinesia, Drug-Induced/psychology , Interleukin-10/metabolism , Male , Neostriatum/metabolism , Oxidopamine , Parkinson Disease/drug therapy , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Ugeskr Laeger ; 181(17)2019 Apr 22.
Article in Danish | MEDLINE | ID: mdl-31036137

ABSTRACT

In this review, we discuss respiratory dyskinesia, which is a rare adverse reaction to antipsychotic medications. The condition may mimic psychogenic hyperventilation syndrome or other respiratory or cardiac disorder. Respiratory dyskinesia is mostly seen in patients with tardive dyskinesia but may precede manifestations of tardive dyskinesia. If a patient receiving antipsychotic medication presents with symptoms of tachypnoea or acute respiratory distress, the possibility of respiratory dyskinesia should be considered, since it is a potentially reversible condition.


Subject(s)
Antipsychotic Agents , Dyskinesia, Drug-Induced , Antipsychotic Agents/adverse effects , Dyskinesia, Drug-Induced/diagnosis , Dyskinesia, Drug-Induced/therapy , Dyspnea , Humans
12.
Mov Disord ; 34(5): 697-707, 2019 05.
Article in English | MEDLINE | ID: mdl-31002755

ABSTRACT

BACKGROUND: Levodopa-induced dyskinesias are an often debilitating side effect of levodopa therapy in Parkinson's disease. Although up to 90% of individuals with PD develop this side effect, uniformly effective and well-tolerated antidyskinetic treatment remains a significant unmet need. The pathognomonic loss of striatal dopamine in PD results in dysregulation and disinhibition of striatal CaV1.3 calcium channels, leading to synaptopathology that appears to be involved in levodopa-induced dyskinesias. Although there are clinically available drugs that can inhibit CaV1.3 channels, they are not adequately potent and have only partial and transient impact on levodopa-induced dyskinesias. METHODS: To provide unequivocal target validation, free of pharmacological limitations, we developed a CaV1.3 shRNA to provide high-potency, target-selective, mRNA-level silencing of striatal CaV1.3 channels and examined its ability to impact levodopa-induced dyskinesias in severely parkinsonian rats. RESULTS: We demonstrate that vector-mediated silencing of striatal CaV1.3 expression in severely parkinsonian rats prior to the introduction of levodopa can uniformly and completely prevent induction of levodopa-induced dyskinesias, and this antidyskinetic benefit persists long term and with high-dose levodopa. In addition, this approach is capable of ameliorating preexisting severe levodopa-induced dyskinesias. Importantly, motoric responses to low-dose levodopa remained intact in the presence of striatal CaV1.3 silencing, indicating preservation of levodopa benefit without dyskinesia liability. DISCUSSION: The current data provide some of the most profound antidyskinetic benefit reported to date and suggest that genetic silencing of striatal CaV1.3 channels has the potential to transform treatment of individuals with PD by allowing maintenance of motor benefit of levodopa in the absence of the debilitating levodopa-induced dyskinesia side effect. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Calcium Channels/genetics , Dyskinesia, Drug-Induced/prevention & control , Levodopa/adverse effects , Neostriatum/metabolism , Parkinsonian Disorders/drug therapy , Adrenergic Agents/toxicity , Animals , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/therapy , Green Fluorescent Proteins , Luminescent Agents , Medial Forebrain Bundle , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , RNA Interference , RNA, Small Interfering , Rats , Substantia Nigra , Tyrosine 3-Monooxygenase/metabolism
14.
Fortschr Neurol Psychiatr ; 87(4): 217-224, 2019 Apr.
Article in German | MEDLINE | ID: mdl-29996156

ABSTRACT

Tardive dyskinesias (TDs) are still common long-term sequelae of antipsychotic treatment. They are generally irreversible and associated with cognitive deficits, a decrease in quality of life and increased mortality. Furthermore, they potentially contribute to further stigmatization of the affected patients. However due to limited treatment options, antipsychotic drugs are still one of the cornerstones in treatment of most severe mental illnesses. Therefore, knowledge about risk factors and prevention of TDs is crucial. If TDs occur, the immediate optimization of the antipsychotic drug regimen is required. Targeted medical treatments such as VMAT - 2 inhibitors can be considered. The novel VMAT-2 inhibitors are not yet approved in Germany. Other drugs that are currently used to treat TDs include clonazepam and gingko biloba. This review summarizes the current evidence of treatment options of TDs and seeks to formulate clinical recommendations for the prevention and management of TDs.


Subject(s)
Antipsychotic Agents/adverse effects , Dyskinesia, Drug-Induced , Tardive Dyskinesia , Dyskinesia, Drug-Induced/prevention & control , Dyskinesia, Drug-Induced/therapy , Germany , Humans , Quality of Life , Tardive Dyskinesia/chemically induced , Tardive Dyskinesia/prevention & control , Tardive Dyskinesia/therapy
16.
Mol Neurobiol ; 56(4): 2408-2423, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30030752

ABSTRACT

Although L-3,4-dihydroxyphenylalanine (L-DOPA) is currently the most effective medication for treating Parkinson's disease (PD) motor symptoms, its prolonged administration causes several adverse effects, including dyskinesia. To identify the mechanisms underlying the effects of acupuncture on L-DOPA-induced dyskinesia (LID), antidyskinetic effects of acupuncture were investigated in two mouse models of PD. Acupuncture stimulation at GB34 alleviated abnormal involuntary movements (AIMs) in Pitx3-deficient aphakia mice (ak/ak) following L-DOPA administration and these effects were reproduced in 6-hydroxydopamine (6-OHDA)-lesioned mice with LID. A transcriptome analysis of the hypothalamus revealed pro-melanin-concentrating hormone (Pmch) gene was highly expressed in acupuncture-treated mouse from ak/ak model of LID as well as 6-OHDA model of LID. Acupuncture combined with the administration of MCH receptor antagonist did not have any beneficial effects on dyskinesia in L-DOPA-injected ak/ak mice, but the intranasal administration of MCH attenuated LID to the same degree as acupuncture in both ak/ak and 6-OHDA mice with LID. A gene expression profile with a hierarchical clustering analysis of the dyskinesia-induced ak/ak mouse brain revealed an association between the mechanisms underlying acupuncture and MCH. Additionally, altered striatal responses to L-DOPA injection were observed after prolonged acupuncture and MCH treatments, which suggests that these treatment modalities influenced the compensatory mechanisms of LID. In summary, present study demonstrated that acupuncture decreased LID via hypothalamic MCH using L-DOPA-administered ak/ak and 6-OHDA mouse models and that MCH administration resulted in novel antidyskinetic effects in these models. Thus, acupuncture and MCH might be valuable therapeutic candidates for PD patients suffering from LID.


Subject(s)
Acupuncture Therapy , Aphakia/complications , Dyskinesia, Drug-Induced/complications , Dyskinesia, Drug-Induced/therapy , Hypothalamic Hormones/metabolism , Levodopa/adverse effects , Melanins/metabolism , Pituitary Hormones/metabolism , Transcription Factors/deficiency , Animals , Aphakia/genetics , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/pathology , Gene Expression Regulation , Homeodomain Proteins , Hypothalamus/pathology , Mice, Inbred C57BL , Neostriatum/metabolism , Neostriatum/pathology , Oxidopamine , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Up-Regulation
17.
Drugs Aging ; 35(11): 959-971, 2018 11.
Article in English | MEDLINE | ID: mdl-30357723

ABSTRACT

Drug-induced parkinsonism (DIP) and tardive dyskinesia (TD) are iatrogenic consequences of antidopaminergic drugs. Both are particularly prevalent among the elderly and those with dementia. However, despite their prevalence, these disorders are often overlooked. Both entities share risk factors, physiopathological mechanisms and, to some degree, therapeutic approaches. Withdrawing the causal agent, reducing the dose or switching to a less potent antidopaminergic drug should be the first therapeutic options. Here we review both entities and emerging therapies including the recently approved drugs deutetrabenazine and valbenazine. We discuss relevant aspects for clinical practice such as new diagnostic techniques and the latest advances in the understanding of DIP and TD.


Subject(s)
Dyskinesia, Drug-Induced/therapy , Parkinsonian Disorders/chemically induced , Tardive Dyskinesia/chemically induced , Aged , Dementia/drug therapy , Humans , Risk Factors , Tetrabenazine/analogs & derivatives , Tetrabenazine/therapeutic use , Valine/analogs & derivatives , Valine/therapeutic use
18.
Ann Neurol ; 84(6): 797-811, 2018 12.
Article in English | MEDLINE | ID: mdl-30357892

ABSTRACT

Levodopa-induced dyskinesia is a common complication in Parkinson disease. Pathogenic mechanisms include phasic stimulation of dopamine receptors, nonphysiological levodopa-to-dopamine conversion in serotonergic neurons, hyperactivity of corticostriatal glutamatergic transmission, and overstimulation of nicotinic acetylcholine receptors on dopamine-releasing axons. Delay in initiating levodopa is no longer recommended, as dyskinesia development is a function of disease duration rather than cumulative levodopa exposure. We review current and in-development treatments for peak-dose dyskinesia but suggest that improvements in levodopa delivery alone may reduce its future prevalence. Ann Neurol 2018;84:797-811.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced , Levodopa/adverse effects , Parkinson Disease/drug therapy , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Dyskinesia, Drug-Induced/epidemiology , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/therapy , Humans
19.
Ther Umsch ; 75(1): 31-36, 2018 Jun.
Article in German | MEDLINE | ID: mdl-29909765

ABSTRACT

Diagnosis and treatment of motor phenomena in schizophrenia spectrum disorders Abstract. Motor abnormalities are intrinsic features of schizophrenia spectrum disorders. They may be spontaneous or antipsychotic drug-induced. The four most important symptom groups are abnormal involuntary movements or dyskinesia, Parkinsonism, catatonia and neurological soft signs. In addition, there are further motor abnormalities, which are less frequent and less operationalized. The suspected etiology of motor abnormalities is strongly associated with altered neurodevelopment. Delayed maturation in conjunction with environmental insults may give further rise to motor symptoms. For the four most relevant motor abnormalities clinical examination procedures and rating scales are available, aiding clinicians in both screening and evaluation of symptom severity. Besides these currently instrumental measures are being tested for wide spread and easy application. Treatment of motor abnormalities is necessary according to subjective well-being. Treatment options are few and remain symptomatic. The most important strategy is critical evaluation of antispychotic pharmacotherapy. Benefitial effects on motor phenomena have been noted with clozapine. Currently, specific substances against tardive dyskinesia and non-invasive brain stimulation techniques are being evaluated. However, the effeciacy of these approaches will only be available in the near future.


Subject(s)
Motor Disorders/therapy , Schizophrenia/therapy , Schizophrenic Psychology , Adult , Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Brain/drug effects , Brain/physiopathology , Child , Combined Modality Therapy , Dyskinesia, Drug-Induced/diagnosis , Dyskinesia, Drug-Induced/physiopathology , Dyskinesia, Drug-Induced/therapy , Humans , Motor Disorders/diagnosis , Motor Disorders/physiopathology , Neurologic Examination , Prodromal Symptoms , Prognosis , Risk Factors , Schizophrenia/diagnosis , Schizophrenia/physiopathology
20.
Cochrane Database Syst Rev ; 3: CD000208, 2018 03 19.
Article in English | MEDLINE | ID: mdl-29552749

ABSTRACT

BACKGROUND: Antipsychotic (neuroleptic) medication is used extensively to treat people with chronic mental illnesses. Its use, however, is associated with adverse effects, including movement disorders such as tardive dyskinesia (TD) - a problem often seen as repetitive involuntary movements around the mouth and face. This review, one in a series examining the treatment of TD, covers miscellaneous treatments not covered elsewhere. OBJECTIVES: To determine whether drugs, hormone-, dietary-, or herb-supplements not covered in other Cochrane reviews on TD treatments, surgical interventions, electroconvulsive therapy, and mind-body therapies were effective and safe for people with antipsychotic-induced TD. SEARCH METHODS: We searched the Cochrane Schizophrenia Group's Study-Based Register of Trials including trial registers (16 July 2015 and 26 April 2017), inspected references of all identified studies for further trials and contacted authors of trials for additional information. SELECTION CRITERIA: We included reports if they were randomised controlled trials (RCTs) dealing with people with antipsychotic-induced TD and schizophrenia or other chronic mental illnesses who remained on their antipsychotic medication and had been randomly allocated to the interventions listed above versus placebo, no intervention, or any other intervention. DATA COLLECTION AND ANALYSIS: We independently extracted data from these trials and we estimated risk ratios (RR) or mean differences (MD), with 95% confidence intervals (CIs). We assumed that people who left early had no improvement. We assessed risk of bias and created 'Summary of findings' tables using GRADE. MAIN RESULTS: We included 31 RCTs of 24 interventions with 1278 participants; 22 of these trials were newly included in this 2017 update. Five trials are awaiting classification and seven trials are ongoing. All participants were adults with chronic psychiatric disorders, mostly schizophrenia, and antipsychotic-induced TD. Studies were primarily of short (three to six6 weeks) duration with small samples size (10 to 157 participants), and most (61%) were published more than 20 years ago. The overall risk of bias in these studies was unclear, mainly due to poor reporting of allocation concealment, generation of the sequence, and blinding.Nineteen of the 31 included studies reported on the primary outcome 'No clinically important improvement in TD symptoms'. Two studies found moderate-quality evidence of a benefit of the intervention compared with placebo: valbenazine (RR 0.63, 95% CI 0.46 to 0.86, 1 RCT, n = 92) and extract of Ginkgo biloba (RR 0.88, 95% CI 0.81 to 0.96, 1 RCT, n = 157), respectively. However, due to small sample sizes we cannot be certain of these effects.We consider the results for the remaining interventions to be inconclusive: Low- to very low-quality evidence of a benefit was found for buspirone (RR 0.53, 95% CI 0.33 to 0.84, 1 RCT, n = 42), dihydrogenated ergot alkaloids (RR 0.45, 95% CI 0.21 to 0.97, 1 RCT, n = 28), hypnosis or relaxation, (RR 0.45, 95% CI 0.21 to 0.94, 1 study, n = 15), pemoline (RR 0.48, 95% CI 0.29 to 0.77, 1 RCT, n = 46), promethazine (RR 0.24, 95% CI 0.11 to 0.55, 1 RCT, n = 34), insulin (RR 0.52, 95% CI 0.29 to 0.96, 1 RCT, n = 20), branched chain amino acids (RR 0.79, 95% CI 0.63 to 1.00, 1 RCT, n = 52), and isocarboxazid (RR 0.24, 95% CI 0.08 to 0.71, 1 RCT, n = 20). There was low- to very low-certainty evidence of no difference between intervention and placebo or no treatment for the following interventions: melatonin (RR 0.89, 95% CI 0.71 to 1.12, 2 RCTs, n = 32), lithium (RR 1.59, 95% CI 0.79 to 3.23, 1 RCT, n = 11), ritanserin (RR 1.00, 95% CI 0.70 to 1.43, 1 RCT, n = 10), selegiline (RR 1.37, 95% CI 0.96 to 1.94, 1 RCT, n = 33), oestrogen (RR 1.18, 95% CI 0.76 to 1.83, 1 RCT, n = 12), and gamma-linolenic acid (RR 1.00, 95% CI 0.69 to 1.45, 1 RCT, n = 16).None of the included studies reported on the other primary outcome, 'no clinically significant extrapyramidal adverse effects'. AUTHORS' CONCLUSIONS: This review has found that the use of valbenazine or extract of Ginkgo biloba may be effective in relieving the symptoms of tardive dyskinesia. However, since only one RCT has investigated each one of these compounds, we are awaiting results from ongoing trials to confirm these results. Results for the remaining interventions covered in this review must be considered inconclusive and these compounds probably should only be used within the context of a well-designed evaluative study.


Subject(s)
Dyskinesia, Drug-Induced/therapy , Adrenergic Uptake Inhibitors/therapeutic use , Adult , Anti-Anxiety Agents/therapeutic use , Antipsychotic Agents/adverse effects , Dihydroergotoxine/therapeutic use , Dyskinesia, Drug-Induced/etiology , Ginkgo biloba , Humans , Hypnosis , Plant Extracts , Randomized Controlled Trials as Topic , Relaxation Therapy , Tetrabenazine/analogs & derivatives , Tetrabenazine/therapeutic use , Valine/analogs & derivatives , Valine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...