Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.246
1.
Clin Transl Sci ; 17(5): e13796, 2024 May.
Article En | MEDLINE | ID: mdl-38712716

Apomorphine, used to treat OFF episodes in patients with Parkinson's disease (PD), is typically administered via subcutaneous injections. Administration of an oromucosal solution could offer a non-invasive and user-friendly alternative. This two-part clinical study evaluated the safety, tolerability, pharmacokinetics (PK), and dose proportionality of a novel apomorphine hydrochloride oromucosal solution, as well as its relative bioavailability to subcutaneous apomorphine injection and apomorphine sublingual film. In part A of the study, 12 patients with PD received 2 mg oromucosal apomorphine (4% weight/volume) and 2 mg subcutaneous apomorphine in a randomized order, followed by 4 and 8 mg oromucosal apomorphine. In part B of the study, 13 patients with PD received 7 mg oromucosal apomorphine (7% weight/volume) and 30 mg sublingual apomorphine in a randomized order, followed by 14 mg oromucosal apomorphine. Washout between dose administrations in both study parts was at least 2 days. Safety, tolerability, and PK were assessed pre- and post-dose. Both study parts showed that oromucosal apomorphine was generally well-tolerated. Observed side effects were typical for apomorphine administration and included asymptomatic orthostatic hypotension, yawning, fatigue, and somnolence. Oromucosal apomorphine exposure increased with dose, although less than dose proportional. The mean (SD) maximum exposure reached with 14 mg oromucosal apomorphine was 753.0 (298.6) ng*min/mL (area under the plasma concentration-time curve from zero to infinity) and 8.0 (3.3) ng/mL (maximum plasma concentration). This was comparable to exposure reached after 2 mg subcutaneous apomorphine and approximately half of the exposure observed with 30 mg sublingual apomorphine. In summary, clinically relevant plasma concentrations could be reached in PD patients without tolerability issues.


Apomorphine , Parkinson Disease , Humans , Apomorphine/administration & dosage , Apomorphine/pharmacokinetics , Apomorphine/adverse effects , Parkinson Disease/drug therapy , Male , Middle Aged , Female , Aged , Administration, Sublingual , Injections, Subcutaneous , Dose-Response Relationship, Drug , Administration, Oral , Biological Availability , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/adverse effects , Cross-Over Studies
2.
N Engl J Med ; 390(13): 1176-1185, 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38598572

BACKGROUND: Lixisenatide, a glucagon-like peptide-1 receptor agonist used for the treatment of diabetes, has shown neuroprotective properties in a mouse model of Parkinson's disease. METHODS: In this phase 2, double-blind, randomized, placebo-controlled trial, we assessed the effect of lixisenatide on the progression of motor disability in persons with Parkinson's disease. Participants in whom Parkinson's disease was diagnosed less than 3 years earlier, who were receiving a stable dose of medications to treat symptoms, and who did not have motor complications were randomly assigned in a 1:1 ratio to daily subcutaneous lixisenatide or placebo for 12 months, followed by a 2-month washout period. The primary end point was the change from baseline in scores on the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) part III (range, 0 to 132, with higher scores indicating greater motor disability), which was assessed in patients in the on-medication state at 12 months. Secondary end points included other MDS-UPDRS subscores at 6, 12, and 14 months and doses of levodopa equivalent. RESULTS: A total of 156 persons were enrolled, with 78 assigned to each group. MDS-UPDRS part III scores at baseline were approximately 15 in both groups. At 12 months, scores on the MDS-UPDRS part III had changed by -0.04 points (indicating improvement) in the lixisenatide group and 3.04 points (indicating worsening disability) in the placebo group (difference, 3.08; 95% confidence interval, 0.86 to 5.30; P = 0.007). At 14 months, after a 2-month washout period, the mean MDS-UPDRS motor scores in the off-medication state were 17.7 (95% CI, 15.7 to 19.7) with lixisenatide and 20.6 (95% CI, 18.5 to 22.8) with placebo. Other results relative to the secondary end points did not differ substantially between the groups. Nausea occurred in 46% of participants receiving lixisenatide, and vomiting occurred in 13%. CONCLUSIONS: In participants with early Parkinson's disease, lixisenatide therapy resulted in less progression of motor disability than placebo at 12 months in a phase 2 trial but was associated with gastrointestinal side effects. Longer and larger trials are needed to determine the effects and safety of lixisenatide in persons with Parkinson's disease. (Funded by the French Ministry of Health and others; LIXIPARK ClinicalTrials.gov number, NCT03439943.).


Antiparkinson Agents , Glucagon-Like Peptide-1 Receptor Agonists , Parkinson Disease , Peptides , Humans , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Antiparkinson Agents/therapeutic use , Disabled Persons , Double-Blind Method , Motor Disorders/drug therapy , Parkinson Disease/drug therapy , Peptides/administration & dosage , Peptides/adverse effects , Peptides/therapeutic use , Treatment Outcome , Glucagon-Like Peptide-1 Receptor Agonists/administration & dosage , Glucagon-Like Peptide-1 Receptor Agonists/adverse effects , Glucagon-Like Peptide-1 Receptor Agonists/therapeutic use , Disease Progression , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , Injections, Subcutaneous
3.
Mol Pharm ; 21(5): 2512-2533, 2024 May 06.
Article En | MEDLINE | ID: mdl-38602861

Parkinson's disease (PD) is a debilitating neurodegenerative disease primarily impacting neurons responsible for dopamine production within the brain. Pramipexole (PRA) is a dopamine agonist that is currently available in tablet form. However, individuals with PD commonly encounter difficulties with swallowing and gastrointestinal motility, making oral formulations less preferable. Microneedle (MN) patches represent innovative transdermal drug delivery devices capable of enhancing skin permeability through the creation of microconduits on the surface of the skin. MNs effectively reduce the barrier function of skin and facilitate the permeation of drugs. The work described here focuses on the development of polymeric MN systems designed to enhance the transdermal delivery of PRA. PRA was formulated into both dissolving MNs (DMNs) and directly compressed tablets (DCTs) to be used in conjunction with hydrogel-forming MNs (HFMNs). In vivo investigations using a Sprague-Dawley rat model examined, for the first time, if it was beneficial to prolong the application of DMNs and HFMNs beyond 24 h. Half of the patches in the MN cohorts were left in place for 24 h, whereas the other half remained in place for 5 days. Throughout the entire 5 day study, PRA plasma levels were monitored for all cohorts. This study confirmed the successful delivery of PRA from DMNs (Cmax = 511.00 ± 277.24 ng/mL, Tmax = 4 h) and HFMNs (Cmax = 328.30 ± 98.04 ng/mL, Tmax = 24 h). Notably, both types of MNs achieved sustained PRA plasma levels over a 5 day period. In contrast, following oral administration, PRA remained detectable in plasma for only 48 h, achieving a Cmax of 159.32 ± 113.43 ng/mL at 2 h. The HFMN that remained in place for 5 days demonstrated the most promising performance among all investigated formulations. Although in the early stages of development, the findings reported here offer a hopeful alternative to orally administered PRA. The sustained plasma profile observed here has the potential to reduce the frequency of PRA administration, potentially enhancing patient compliance and ultimately improving their quality of life. This work provides substantial evidence advocating the development of polymeric MN-mediated drug delivery systems to include sustained plasma levels of hydrophilic pharmaceuticals.


Administration, Cutaneous , Drug Delivery Systems , Needles , Parkinson Disease , Pramipexole , Rats, Sprague-Dawley , Pramipexole/administration & dosage , Pramipexole/pharmacokinetics , Animals , Rats , Parkinson Disease/drug therapy , Drug Delivery Systems/methods , Male , Skin Absorption/drug effects , Skin/metabolism , Skin/drug effects , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacokinetics , Dopamine Agonists/administration & dosage , Dopamine Agonists/pharmacokinetics , Hydrogels/chemistry
4.
Xenobiotica ; 54(4): 201-210, 2024 Apr.
Article En | MEDLINE | ID: mdl-38563808

The novel anti-Parkinson disease drug, FLZ, had a complicated drug absorption and metabolise process reported in single-dose studies. A multi-peak absorption peak phenomenon was found.This study focused on the multi-dose pharmacokinetics (PK) characteristics of FLZ, T1, and T2 in cynomolgus monkeys and raised discussion on its multi-peak absorption situation. Different doses of FLZ ranging from 75 to 300 mg/kg were administered orally to 16 cynomolgus monkeys. The whole treatment period lasted for 42 days with FLZ once a day.The primary metabolites of FLZ were Target1 (T1) and Target2 (T2), which had plasma exposure (calculated as AUC0-24, day 42) approximately 2 and 10 times higher than the parent drug. The proportion of plasma exposure increase was lower than the proportion of dose increase in FLZ, T1, and T2.Gender influenced its exposure (AUC0-24) with approximately 3-fold higher in males than females. There was no significant accumulation of T1 and T2. Enterohepatic Circulation (EHC) and gastrointestinal (GI) tract absorption may be involved in the mechanism of multi-peak characteristics.


Antiparkinson Agents , Macaca fascicularis , Animals , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/administration & dosage , Male , Female , Administration, Oral , Dose-Response Relationship, Drug
5.
Expert Opin Drug Deliv ; 21(3): 437-456, 2024 Mar.
Article En | MEDLINE | ID: mdl-38507231

INTRODUCTION: The current treatment modalities available for Parkinson's disease (PD) prove inadequate due to the inherent constraints in effectively transporting bioactive compounds across the blood-brain barrier. The utilization of synergistic combinations of multiple drugs in conjunction with advanced nanotechnology, emerges as a promising avenue for the treatment of PD, offering potential breakthroughs in treatment efficacy, targeted therapy, and personalized medicine. AREAS COVERED: This review provides a comprehensive analysis of the efficacy of multifactorial interventions for PD, simultaneously addressing the primary challenges of conventional therapies and highlighting how advanced technologies can help overcome these limitations. Part II focuses on the effectiveness of nanotechnology for improving pharmacokinetics of conventional therapies, through the synergistic use of dual or multiple therapeutic agents into a single nanoformulation. Significant emphasis is laid on the advancements toward innovative integrations, such as CRISPR/Cas9 with neuroprotective agents and stem cells, all effectively synergized with nanocarriers. EXPERT OPINION: By using drug combinations, we can leverage their combined effects to enhance treatment efficacy and mitigate side effects through lower dosages. This article is meant to give nanocarrier-mediated co-delivery of drugs and the strategic incorporation of CRISPR/Cas9, either as an independent intervention or synergized with a neuroprotective agent.


Antiparkinson Agents , Drug Carriers , Nanoparticles , Nanotechnology , Neuroprotective Agents , Parkinson Disease , Humans , Parkinson Disease/drug therapy , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/therapeutic use , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/pharmacology , Drug Carriers/chemistry , Animals , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Drug Delivery Systems , Precision Medicine , Drug Therapy, Combination , CRISPR-Cas Systems , Drug Combinations , Combined Modality Therapy , Drug Development , Drug Design
6.
Expert Opin Drug Deliv ; 21(3): 423-435, 2024 Mar.
Article En | MEDLINE | ID: mdl-38481172

INTRODUCTION: Parkinson's disease (PD) is a neurological condition defined by a substantial reduction in dopamine-containing cells in the substantia nigra. Levodopa (L-Dopa) is considered the gold standard in treatment. Recent research has clearly shown that resistance to existing therapies can develop. Moreover, the involvement of multiple pathways in the nigrostriatal dopaminergic neuronal loss suggests that modifying the treatment strategy could effectively reduce this degeneration. AREAS COVERED: This review summarizes the key concerns with treating PD patients and the combinations, aimed at effectively managing PD. Part I focuses on the clinical diagnosis at every stage of the disease as well as the pharmacological treatment strategies that are applied throughout its course. It methodically elucidates the potency of multifactorial interventions in attenuating the disease trajectory, substantiating the rationale for co-administration of dual or multiple therapeutic agents. Significant emphasis is laid on evidence-based pharmacological combinations for PD management. EXPERT OPINION: By utilizing multiple drugs in a combination fashion, this approach can leverage the additive or synergistic effects of these agents, amplify the spectrum of treatment, and curtail the risk of side effects by reducing the dose of each drug, demonstrating significantly greater efficacy.


Antiparkinson Agents , Drug Therapy, Combination , Levodopa , Parkinson Disease , Parkinson Disease/drug therapy , Humans , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/therapeutic use , Antiparkinson Agents/pharmacology , Levodopa/administration & dosage , Levodopa/therapeutic use , Animals , Drug Carriers/chemistry , Nanoparticles , Drug Synergism
7.
Parkinsonism Relat Disord ; 122: 106088, 2024 May.
Article En | MEDLINE | ID: mdl-38461688

INTRODUCTION: Dystonia is a painful OFF-related complication in Parkinson's disease (PD) with limited treatment options. METHODS: Post-hoc analysis using pooled data from two extended-release amantadine pivotal trials and follow-on open-label extension. Dystonia was assessed using the Unified Dyskinesia Rating Scale (UDysRS) Part 2 and the Movement Disorder Society-Unified PD Rating Scale (MDS-UPDRS) item 4.6. RESULTS: Of 196 participants, 119 (60.7%) reported OFF-related dystonia at baseline per UDysRS. Twelve-week treatment with extended-release amantadine improved OFF dystonia (treatment differences vs placebo: UDysRS Part 2, -1.0 [-1.9,-0.1]; p = 0.03 and MDS-UPDRS Item 4.6, -0.3 [-0.6,-0.05]; p = 0.02). There was no correlation between changes in OFF time and changes in OFF dystonia. Double-blind improvements in OFF dystonia were sustained throughout the 2-year follow-up. CONCLUSIONS: Extended-release amantadine yielded a sustained reduction in OFF-related dystonia in PD patients that was independent from a reduction in OFF time. A randomized controlled trial is warranted to confirm these findings.


Amantadine , Antiparkinson Agents , Delayed-Action Preparations , Dystonia , Parkinson Disease , Humans , Amantadine/administration & dosage , Parkinson Disease/complications , Parkinson Disease/drug therapy , Male , Female , Dystonia/drug therapy , Dystonia/etiology , Aged , Middle Aged , Antiparkinson Agents/administration & dosage , Double-Blind Method
8.
Mov Disord Clin Pract ; 11(5): 534-542, 2024 May.
Article En | MEDLINE | ID: mdl-38470011

BACKGROUND: PD causes striatal dopaminergic denervation in a posterior/dorsal to anterior/ventral gradient, leaving motor and associative cortico-striato-pallido-thalamic loops differentially susceptible to hyperdopaminergic effects with treatment. As the choice and titration of symptomatic PD medications are guided primarily by motor symptoms, it is important to understand their cognitive implications. OBJECTIVE: To investigate the effects of acute dopaminergic medication administration on executive function in Parkinson's disease (PD). METHODS: Participants with idiopathic PD were administered the oral Symbol Digit Modalities Test (SDMT; n = 181) and the Stroop test (n = 172) in the off-medication and "best on" medication states. ANCOVA was used to test for differences between off-medication and on-medication scores corrected for age and years of education. RESULTS: After administration of symptomatic medications, scores worsened on the SDMT (F = 11.70, P < 0.001, d = -0.13), improved on the Stroop color (F = 26.89, P < 0.001, d = 0.184), word (F = 6.25, P = 0.013, d = 0.09), and color-word (F = 13.22, P < 0.001, d = 0.16) test components, and the Stroop difference and ratio-based interference scores did not significantly change. Longer disease duration correlated with lower scores on the SDMT, Stroop color, word, and color-word scores; however, longer disease duration and higher levodopa-equivalents correlated with higher Stroop difference-based interference scores. CONCLUSIONS: Symptomatic medication differentially affects performance on two cognitive tests in PD. After acute treatment, core Stroop measures improved, Stroop interference was unchanged, and SDMT performance worsened, likely reflecting complex changes in processing speed and executive function related to acute treatment. When considering motor symptom therapies in PD, an individual's cognitive demands and expectations, especially regarding executive function, should be considered.


Cognition , Executive Function , Parkinson Disease , Humans , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Male , Aged , Female , Middle Aged , Cognition/drug effects , Executive Function/drug effects , Levodopa/therapeutic use , Levodopa/administration & dosage , Levodopa/pharmacology , Neuropsychological Tests/statistics & numerical data , Antiparkinson Agents/therapeutic use , Antiparkinson Agents/pharmacology , Antiparkinson Agents/administration & dosage , Stroop Test , Dopamine Agents/therapeutic use , Dopamine Agents/pharmacology , Dopamine Agents/administration & dosage
9.
Am J Ther ; 31(3): e209-e218, 2024.
Article En | MEDLINE | ID: mdl-38460175

BACKGROUND: For Parkinson disease (PD) patients who have been diagnosed with advanced disease that can no longer be effectively controlled with optimized oral or transdermal medications, a range of device-aided therapies (DAT) are available, comprising either deep brain stimulation or infusion therapies providing continuous dopaminergic stimulation. Levodopa-entacapone-carbidopa intestinal gel (LECIG) infusion is the latest DAT for advanced PD (APD) that was approved in Romania in 2021. STUDY QUESTION: What is the experience to date in real-world clinical practice in Romania regarding the efficacy and tolerability of LECIG in APD? STUDY DESIGN: A retrospective evaluation of 74 APD patients treated with LECIG at 12 specialized APD centers in Romania. MEASURES AND OUTCOMES: Demographic data and various clinical parameters were recorded, including Mini Mental State Evaluation score or Montreal Cognitive Assessment Test score. Levodopa-equivalent daily dose and the administered doses of levodopa and other PD medications were evaluated at baseline and after starting LECIG treatment. The efficacy of LECIG in reducing daily hours of off time, motor fluctuations, and dyskinesias were assessed. Any percutaneous endoscopic gastrojejunostomy system or device complications after starting LECIG treatment were noted. RESULTS: At baseline, patients were taking oral levodopa for a mean of 5.3 times per day, with a high proportion also taking concomitant add-on therapies (dopamine agonists, 86%, monoamine oxidase type-B inhibitors, 53%; catechol-O-methyltransferase inhibitors, 64%). LECIG treatment significantly reduced daily off time versus baseline from 5.7 h/d to 1.7 hours per day ( P < 0.01). Duration and severity of dyskinesias was also significantly reduced versus baseline, and improvements were observed in Hoehn and Yahr Scale scores. LECIG treatment also allowed a significant reduction in the use of concomitant oral medications. CONCLUSIONS: These findings suggest that LECIG treatment is an effective DAT option in APD that can simplify the treatment regimen.


Antiparkinson Agents , Carbidopa , Catechols , Drug Combinations , Gels , Levodopa , Nitriles , Parkinson Disease , Humans , Parkinson Disease/drug therapy , Levodopa/administration & dosage , Levodopa/therapeutic use , Levodopa/adverse effects , Carbidopa/administration & dosage , Carbidopa/therapeutic use , Carbidopa/adverse effects , Male , Female , Retrospective Studies , Aged , Catechols/administration & dosage , Catechols/therapeutic use , Catechols/adverse effects , Middle Aged , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/therapeutic use , Antiparkinson Agents/adverse effects , Nitriles/administration & dosage , Nitriles/therapeutic use , Nitriles/adverse effects , Treatment Outcome , Romania
10.
Neurol Sci ; 45(6): 2661-2670, 2024 Jun.
Article En | MEDLINE | ID: mdl-38183553

INTRODUCTION: The acute levodopa challenge test (ALCT) is an important and valuable examination but there are still some shortcomings with it. We aimed to objectively assess ALCT based on a depth camera and filter out the best indicators. METHODS: Fifty-nine individuals with parkinsonism completed ALCT and the improvement rate (IR, which indicates the change in value before and after levodopa administration) of the Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale part III (MDS-UPDRS III) was calculated. The kinematic features of the patients' movements in both the OFF and ON states were collected with an Azure Kinect depth camera. RESULTS: The IR of MDS-UPDRS III was significantly correlated with the IRs of many kinematic features for arising from a chair, pronation-supination movements of the hand, finger tapping, toe tapping, leg agility, and gait (rs = - 0.277 ~ - 0.672, P < 0.05). Moderate to high discriminative values were found in the selected features in identifying a clinically significant response to levodopa with sensitivity, specificity, and area under the curve (AUC) in the range of 50-100%, 47.22%-97.22%, and 0.673-0.915, respectively. The resulting classifier combining kinematic features of toe tapping showed an excellent performance with an AUC of 0.966 (95% CI = 0.922-1.000, P < 0.001). The optimal cut-off value was 21.24% with sensitivity and specificity of 94.44% and 87.18%, respectively. CONCLUSION: This study demonstrated the feasibility of measuring the effect of levodopa and objectively assessing ALCT based on kinematic data derived from an Azure Kinect-based system.


Antiparkinson Agents , Feasibility Studies , Levodopa , Parkinsonian Disorders , Humans , Levodopa/administration & dosage , Levodopa/therapeutic use , Levodopa/pharmacology , Male , Female , Aged , Middle Aged , Antiparkinson Agents/therapeutic use , Antiparkinson Agents/administration & dosage , Biomechanical Phenomena/physiology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/diagnosis , Severity of Illness Index
11.
J Neurol ; 271(5): 2412-2422, 2024 May.
Article En | MEDLINE | ID: mdl-38214756

BACKGROUND: The long-term prognosis of impulsive compulsive disorders (ICD) remains poorly studied in Parkinson's disease (PD). OBJECTIVE: Evaluating the natural history of ICD and its impact on PD symptoms including cognition and treatment adjustments. MATERIALS AND METHODS: We assessed PD patients at baseline (BL) with (BL-ICD+) or without (BL-ICD-) ICD despite dopamine agonist (DA) exposure of > 300 mg levodopa-equivalent daily dose for > 12 months at baseline and after more than two years of follow-up. ICD were assessed using the Ardouin's Scale of Behaviors in PD (ASBPD), cognition using the Mattis scale, and PD symptoms using the UPDRS score. Treatment adjustments, DA withdrawal-associated symptoms, and ICDs social consequences were recorded. RESULTS: 149 patients were included (78 cases and 71 controls), mean duration of follow-up was 4.4 ± 1 years. At baseline, psychiatric disorders were more common among BL-ICD + (42.3 vs 12.3% among BL-ICD-, p < 0.01). At follow-up, 53.8% of BL-ICD + were not ICD-free while 21.1% of BL-ICD- had developed ICD. BL-ICD + more frequently experienced akinesia (21.8 vs 8.5%, p = 0.043) and rigidity worsening (11.5 vs 1.4%, p = 0.019) following therapeutic modifications. Decision to decrease > 50% DA doses (12.8 vs 1.4%, p = 0.019) or to withdraw DA (19.2 vs 5.6%, p = 0.025) was more frequently considered among BL-ICD+ . At follow-up, the prevalence of cognitive decline was lower among BL-ICD + (19.2 vs 37.1%, p = 0.025). CONCLUSION: ICDs were associated with increased psychiatric burden at baseline and better cognitive prognosis. Most patients were still showing ICDs at the follow-up visit, suggesting ICD to be considered as a chronic, neuropsychiatric disorder.


Disruptive, Impulse Control, and Conduct Disorders , Parkinson Disease , Humans , Parkinson Disease/complications , Parkinson Disease/drug therapy , Male , Disruptive, Impulse Control, and Conduct Disorders/etiology , Female , Middle Aged , Aged , Prognosis , Prospective Studies , Dopamine Agonists/administration & dosage , Dopamine Agonists/adverse effects , Follow-Up Studies , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects
12.
Clin Pharmacol Drug Dev ; 13(5): 549-559, 2024 May.
Article En | MEDLINE | ID: mdl-38178727

KW-6356 is a selective antagonist and inverse agonist of the adenosine A2A receptor. The primary aim of the present analysis was to characterize the pharmacokinetics (PK) of KW-6356 and its active metabolite M6 in healthy subjects and patients with Parkinson's disease (PD). We pooled concentration-time data from healthy subjects and patients with PD who were administered KW-6356. Using these data, we developed a population PK model by sequentially fitting the KW-6356 parameters followed by the M6 parameters. A first-order absorption with a 1-compartment model for KW-6356 and a 1-compartment model for M6 best described the profiles. The covariates included in the final models were food status (fed/fasted/unknown) on first-order absorption rate constant, baseline serum albumin level on apparent clearance of KW-6356, and baseline body weight on apparent volume of distribution of KW-6356 and apparent clearance of M6. No covariate had a clinically meaningful impact on KW-6356 or M6 exposure.


Adenosine A2 Receptor Antagonists , Healthy Volunteers , Models, Biological , Parkinson Disease , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Adenosine A2 Receptor Agonists/pharmacokinetics , Adenosine A2 Receptor Agonists/administration & dosage , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacokinetics , Adenosine A2 Receptor Antagonists/administration & dosage , Adenosine A2 Receptor Antagonists/pharmacology , Administration, Oral , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/administration & dosage , Drug Administration Schedule , Parkinson Disease/drug therapy , Receptor, Adenosine A2A/metabolism
13.
Brain Res ; 1809: 148349, 2023 06 15.
Article En | MEDLINE | ID: mdl-36972837

Overactivity of the corticostriatal glutamatergic pathway is documented in Parkinson's disease (PD) and stimulation of presynaptic metabotropic glutamate (mGlu) receptors 4 on these striatal afferents inhibits glutamate release normalizing neuronal activity in the basal ganglia. Moreover, mGlu4 receptors are also expressed in glial cells and are able to modulate glial function making this receptor a potential target for neuroprotection. Hence, we investigated whether foliglurax, a positive allosteric modulator of mGlu4 receptors with high brain exposure after oral administration, has neuroprotective effects in MPTP mice to model early PD. Male mice were treated daily from day 1 to 10 with 1, 3 or 10 mg/kg of foliglurax and administered MPTP on the 5th day then euthanized on the 11th day. Dopamine neuron integrity was assessed with measures of striatal dopamine and its metabolites levels, striatal and nigral dopamine transporter (DAT) binding and inflammation with markers of striatal astrocytes (GFAP) and microglia (Iba1). MPTP lesion produced a decrease in dopamine, its metabolites and striatal DAT specific binding that was prevented by treatment with 3 mg/kg of foliglurax, whereas 1 and 10 mg/kg had no beneficial effect. MPTP mice had increased levels of GFAP; foliglurax treatment (3 mg/kg) prevented this increase. Iba1 levels were unchanged in MPTP mice compared to control mice. There was a negative correlation between dopamine content and GFAP levels. Our results show that positive allosteric modulation of mGlu4 receptors with foliglurax provided neuroprotective effects in the MPTP mouse model of PD.


1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Antiparkinson Agents , Dopaminergic Neurons , Neuroprotective Agents , Receptors, Metabotropic Glutamate , Animals , Male , Mice , Allosteric Regulation/drug effects , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Basal Ganglia/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Astrocytes/metabolism , Microglia/metabolism , Neostriatum/drug effects , Neostriatum/metabolism
14.
N Engl J Med ; 387(22): 2045-2055, 2022 12 01.
Article En | MEDLINE | ID: mdl-36449420

BACKGROUND: Iron content is increased in the substantia nigra of persons with Parkinson's disease and may contribute to the pathophysiology of the disorder. Early research suggests that the iron chelator deferiprone can reduce nigrostriatal iron content in persons with Parkinson's disease, but its effects on disease progression are unclear. METHODS: We conducted a multicenter, phase 2, randomized, double-blind trial involving participants with newly diagnosed Parkinson's disease who had never received levodopa. Participants were assigned (in a 1:1 ratio) to receive oral deferiprone at a dose of 15 mg per kilogram of body weight twice daily or matched placebo for 36 weeks. Dopaminergic therapy was withheld unless deemed necessary for symptom control. The primary outcome was the change in the total score on the Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS; range, 0 to 260, with higher scores indicating more severe impairment) at 36 weeks. Secondary and exploratory clinical outcomes at up to 40 weeks included measures of motor and nonmotor disability. Brain iron content measured with the use of magnetic resonance imaging was also an exploratory outcome. RESULTS: A total of 372 participants were enrolled; 186 were assigned to receive deferiprone and 186 to receive placebo. Progression of symptoms led to the initiation of dopaminergic therapy in 22.0% of the participants in the deferiprone group and 2.7% of those in the placebo group. The mean MDS-UPDRS total score at baseline was 34.3 in the deferiprone group and 33.2 in the placebo group and increased (worsened) by 15.6 points and 6.3 points, respectively (difference, 9.3 points; 95% confidence interval, 6.3 to 12.2; P<0.001). Nigrostriatal iron content decreased more in the deferiprone group than in the placebo group. The main serious adverse events with deferiprone were agranulocytosis in 2 participants and neutropenia in 3 participants. CONCLUSIONS: In participants with early Parkinson's disease who had never received levodopa and in whom treatment with dopaminergic medications was not planned, deferiprone was associated with worse scores in measures of parkinsonism than those with placebo over a period of 36 weeks. (Funded by the European Union Horizon 2020 program; FAIRPARK-II ClinicalTrials.gov number, NCT02655315.).


Antiparkinson Agents , Deferiprone , Iron Chelating Agents , Iron , Parkinson Disease , Substantia Nigra , Humans , Deferiprone/administration & dosage , Deferiprone/adverse effects , Deferiprone/pharmacology , Deferiprone/therapeutic use , Iron/analysis , Iron/metabolism , Levodopa/therapeutic use , Neutropenia/chemically induced , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Substantia Nigra/chemistry , Substantia Nigra/diagnostic imaging , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Disease Progression , Double-Blind Method , Administration, Oral , Brain/diagnostic imaging , Brain Chemistry , Dopamine Agents/administration & dosage , Dopamine Agents/adverse effects , Dopamine Agents/pharmacology , Dopamine Agents/therapeutic use , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use
15.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article En | MEDLINE | ID: mdl-35197281

Apomorphine, a dopamine agonist, is a highly effective therapeutic to prevent intermittent off episodes in advanced Parkinson's disease. However, its short systemic half-life necessitates three injections per day. Such a frequent dosing regimen imposes a significant compliance challenge, especially given the nature of the disease. Here, we report a deep eutectic-based formulation that slows the release of apomorphine after subcutaneous injection and extends its pharmacokinetics to convert the current three-injections-a-day therapy into an every-other-day therapy. The formulation comprises a homogeneous mixture of a deep eutectic solvent choline-geranate, a cosolvent n-methyl-pyrrolidone, a stabilizer polyethylene glycol, and water, which spontaneously emulsifies into a microemulsion upon injection in the subcutaneous space, thereby entrapping apomorphine and significantly slowing its release. Ex vivo studies with gels and rat skin demonstrate this self-emulsification process as the mechanism of action for sustained release. In vivo pharmacokinetics studies in rats and pigs further confirmed the extended release and improvement over the clinical comparator Apokyn. In vivo pharmacokinetics, supported by a pharmacokinetic simulation, demonstrate that the deep eutectic formulation reported here allows the maintenance of the therapeutic drug concentration in plasma in humans with a dosing regimen of approximately three injections per week compared to the current clinical practice of three injections per day.


Antiparkinson Agents/administration & dosage , Apomorphine/administration & dosage , Delayed-Action Preparations , Drug Implants , Emulsions , Parkinson Disease/drug therapy , Subcutaneous Tissue , Animals , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/therapeutic use , Apomorphine/pharmacokinetics , Apomorphine/therapeutic use , Area Under Curve , Half-Life , Humans , Rats , Swine
16.
Drugs ; 82(2): 169-197, 2022 Feb.
Article En | MEDLINE | ID: mdl-35076890

There has been exponential growth in the awareness and understanding of gastrointestinal (GI) dysfunction in Parkinson's disease (PD) over the past 3 decades. The clinical features of GI dysfunction in PD have been clearly identified and innovative research has demonstrated the presence of pathology within the enteric nervous system (ENS) in individuals with PD, leading to suggestions that the GI system may be ground zero for the genesis and the portal of entry of PD pathology, which then ascends via the vagus nerve to the central nervous system (CNS). This theory, as well as the more recent recognition of the association of PD with dysbiosis within the gut microbiota, has been the object of intense study and scrutiny. Since most PD medications are absorbed through the GI system, the need for better understanding of changes within the GI tract that may potentially affect the pattern of response to medications has become evident. In this review, current knowledge of the pathophysiology of changes within the GI tract and the gut microbiome of individuals with PD, including changes that occur with progression of the disease, will be addressed. We focus on common clinical GI problems in PD that can arise from different segments of the GI tract. Relevant diagnostic evaluations and treatment options for each of these problems will be reviewed.


Antiparkinson Agents/therapeutic use , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/therapy , Gastrointestinal Motility/physiology , Parkinson Disease/physiopathology , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Deglutition Disorders/physiopathology , Diet , Enteric Nervous System/physiopathology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Transit/physiology , Humans , Oral Health , Weight Loss/physiology
17.
Expert Opin Drug Saf ; 21(2): 137-147, 2022 Feb.
Article En | MEDLINE | ID: mdl-34597253

INTRODUCTION: While levodopa is still the most effective treatment for Parkinson's disease, concerns about long-term complications such as wearing-off and dyskinesia with levodopa usage remain. AREAS COVERED: Safinamide is a highly selective and reversible monoamine oxidase B inhibitor introduced in the European Union, Japan, and the United States as an adjunctive agent to levodopa in PD patients with motor fluctuation. This review outlines the pharmacological properties, therapeutic effects, and tolerability of safinamide as an adjunct to levodopa in patients with advanced PD. Efficacy and safety findings from double-blind and placebo-controlled clinical trials for safinamide as an adjunct therapy to levodopa for PD are summarized. EXPERT OPINION: Safinamide was well tolerated as a treatment for PD, and there was no significant difference in the frequency and severity of adverse events between the safinamide and placebo groups. It was also suggested that safinamide had a beneficial effect on the accompanying non-motor symptoms such as PD-related pain. Safinamide may exhibit neuroprotective effects through antioxidant and anti-glutamate effects, and research on the disease-modifying effect of PD is desired in the future.


Alanine/analogs & derivatives , Benzylamines/administration & dosage , Levodopa/administration & dosage , Parkinson Disease/drug therapy , Alanine/administration & dosage , Alanine/adverse effects , Animals , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Benzylamines/adverse effects , Drug Therapy, Combination , Humans , Levodopa/adverse effects , Parkinson Disease/physiopathology , Treatment Outcome
18.
Neuropharmacology ; 204: 108869, 2022 02 15.
Article En | MEDLINE | ID: mdl-34742740

BACKGROUND: Motor complications, characterized by "off" periods - when anti-parkinsonian medications are ineffective - and dyskinesia, are the hallmark of advanced Parkinson's disease (PD). While levodopa is the gold standard PD medication in terms of efficacy, its short duration of effect coupled with progressive loss of dopaminergic neurons leads to motor complications and fails to treat off periods. PURPOSE OF REVIEW: This review focuses on novel dopaminergic therapies that were recently made clinically available or are currently in development for the treatment of motor complications. First, it will discuss rescue therapies for the treatment of off episodes, including novel apomorphine and levodopa formulations. Second, it will highlight adjunctive dopaminergic medications approved to reduce total daily off time. Third, it will discuss longer-acting levodopa formulations in development and introduce a novel selective dopamine agonist under study. Finally, it will cover novel dopaminergic delivery mechanisms, with specific focus on continuous subcutaneous infusions in development. SUMMARY: The breadth of dopaminergic therapies recently approved or in development for motor complications, and specifically off time reduction, evokes cautious optimism. Gains in reducing off time with rescue therapies, adjunctive medications or longer-acting levodopa formulations are modest, and underscore the need for more continuous dopaminergic delivery to address the underlying pathophysiology and translate to clinically meaningful improvement in motor complications.


Antiparkinson Agents/therapeutic use , Apomorphine/therapeutic use , Dopamine Agents/therapeutic use , Dopamine Agonists/therapeutic use , Dyskinesias/drug therapy , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Alanine/analogs & derivatives , Antiparkinson Agents/administration & dosage , Apomorphine/administration & dosage , Benzylamines , Delayed-Action Preparations , Disease Progression , Dopamine Agents/administration & dosage , Dopamine Agonists/administration & dosage , Drug Compounding , Drug Delivery Systems , Dyskinesias/etiology , Humans , Levodopa/administration & dosage , Oxadiazoles , Parkinson Disease/complications
19.
Biomed Pharmacother ; 145: 112389, 2022 Jan.
Article En | MEDLINE | ID: mdl-34775235

Parkinson's disease (PD) is a multifactorial neurodegenerative disease with damages to mitochondria and endoplasmic reticulum (ER), followed by neuroinflammation. We previously reported that a triple herbal extract DA-9805 in experimental PD toxin-models had neuroprotective effects by alleviating mitochondrial damage and oxidative stress. In the present study, we investigated whether DA-9805 could suppress ER stress and neuroinflammation in vitro and/or in vivo. Pre-treatment with DA-9805 (1 µg/ml) attenuated upregulation of glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and cleaved caspase-3 in SH-SY5Y neuroblastoma cells treated with thapsigargin (1 µg/ml) or tunicamycin (2 µg/ml). In addition, DA-9805 prevented the production of IL-1ß, IL-6, TNF-α and nitric oxide through inhibition of NF-κB activation in BV2 microglial cells stimulated with lipopolysaccharides (LPS). Intraperitoneal injection of LPS (10 mg/kg) into mice can induce acute neuroinflammation and dopaminergic neuronal cell death. Oral administration of DA-9805 (10 or 30 mg/kg/day for 3 days before LPS injection) prevented loss of dopaminergic neurons and activation of microglia and astrocytes in the substantia nigra in LPS-injected mouse models. Taken together, these results indicate that DA-9805 can effectively prevent ER stress and neuroinflammation, suggesting that DA-9805 is a multitargeting and disease-modifying therapeutic candidate for PD.


Antiparkinson Agents , Endoplasmic Reticulum Stress , Inflammation , Plant Extracts , Animals , Humans , Male , Mice , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Inflammation/drug therapy , Lipopolysaccharides , Mice, Inbred C57BL , Microglia/drug effects , Neuroblastoma/metabolism , Neuroinflammatory Diseases/drug therapy , Parkinson Disease/drug therapy , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Substantia Nigra/drug effects
20.
Brain Res ; 1775: 147727, 2022 01 15.
Article En | MEDLINE | ID: mdl-34788638

BACKGROUND: Our understanding of how balance control responds to levodopa over the course of a single day in people with Parkinson's disease (PD) is limited with the majority of studies focused on isolated comparisons of ON vs. OFF levodopa medication. OBJECTIVE: To evaluate the temporal dynamics of postural control following the first levodopa dose of the day during a challenging standing task in a group of people with PD. METHODS: Changes in postural control were evaluated by monitoring cortical activity (covering frontal, motor, parietal and occipital areas), body sway parameters (force platform), and lower limb muscle activity (tibialis anterior and gastrocnemius medialis) in 15 individuals with PD during a semi-tandem standing task. Participants were assessed during two 60 second trials every 30 minutes (ON-30 ON-60 etc.) for 3 hours after the first matinal dose (ON-180). RESULTS: Compared to when tested OFF-medication, cortical activity was increased across all four regions from ON-60 to ON-120 with early increases in alpha and beta band activity observed at ON-30. Levodopa was associated with increased gastrocnemius medialis activity (ON-30 to ON-120) and ankle co-contraction (ON-60 to ON-120). Changes in body sway outcomes (particularly in the anterior-posterior direction) were evident from ON-60 to ON-120. CONCLUSIONS: Our results reveal a 60-minute window within which postural control outcomes may be obtained that are different compared to OFF-state and remain stable (from 60-minutes to 120-minutes after levodopa intake). Identifying a window of opportunity for measurement when individuals are optimally medicated is important for observations in a clinical and research setting.


Antiparkinson Agents/administration & dosage , Cerebral Cortex/drug effects , Levodopa/administration & dosage , Parkinson Disease/drug therapy , Postural Balance/drug effects , Aged , Aged, 80 and over , Antiparkinson Agents/therapeutic use , Cerebral Cortex/physiopathology , Female , Humans , Levodopa/therapeutic use , Male , Middle Aged , Parkinson Disease/physiopathology , Postural Balance/physiology
...