1
|
Karunakaran KB, Jain S, Widera D, Cottrell GS. Spatial and functional profiles distinguish target sets of Parkinson's disease and antipsychotic drugs with different clinical effects. Transl Psychiatry 2025; 15:124. [PMID: 40185727 PMCID: PMC11971416 DOI: 10.1038/s41398-025-03351-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Several studies have examined the genetic factors shared between Parkinson's disease (PD) and schizophrenia (SZ), but the biological themes underlying their clinical relationships remain less explored. We employed systematic transcriptomic and network analyses to examine the genes targeted by two sets of antipsychotic drugs (APDs) - first-generation APDs inducing Parkinsonism and second-generation APDs typically effective against psychotic symptoms in PD - and two sets of PD drugs, one at risk of psychosis and the other with a lower risk of psychosis. Although global brain expression patterns did not effectively differentiate between the targets of the two sets of APDs, they did differentiate the targets of the two PD drug sets. However, both APD and PD target sets showed differences in mean expression levels in specific brain regions. Moreover, they showed significant enrichment for genes highly expressed in distinct adult and prenatal brain structures relative to the overall distribution of such genes among all brain-expressed genes. Specific neurotransmitter systems, either individually or in combinations, appeared to underlie the clinically informed drug categories, indicating their differential roles in inducing or not inducing PD and psychosis. Additionally, the target sets formed distinct network modules representing different biological mechanisms and exhibited differential proximity to putative PD and SZ risk genes in the human interactome. In summary, our study identified specific spatial and functional features that distinguish the target sets of PD and antipsychotic drugs with different clinical effects.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- School of Pharmacy, University of Reading, Reading, UK.
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| | - Sanjeev Jain
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Darius Widera
- School of Pharmacy, University of Reading, Reading, UK
| | | |
Collapse
|
2
|
Ozawa M, Shiraishi T, Murakami H, Yoshimaru D, Onda A, Matsuno H, Komatsu T, Sakuta K, Sakai K, Umehara T, Mitsumura H, Okano HJ, Iguchi Y. Structural MRI study of Pareidolia and Visual Hallucinations in Drug-Naïve Parkinson's disease. Sci Rep 2024; 14:31293. [PMID: 39733021 PMCID: PMC11682137 DOI: 10.1038/s41598-024-82707-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Visual hallucinations (VH) and pareidolia, a type of minor hallucination, share common underlying mechanisms. However, the similarities and differences in their brain regions remain poorly understood in Parkinson's disease (PD). A total of 104 drug-naïve PD patients underwent structural MRI and were assessed for pareidolia using the Noise Pareidolia Test (NPT) were enrolled. Subcortical gray matter volume and cortical surface volume were analyzed using the FreeSurfer software. Structural analyses revealed associations between NPT scores and atrophy in the right thalamus, right hippocampus, right temporal cortex, and right orbitofrontal cortex in all PD participants. These results were almost the same after adjusting for right-handed 97 patients with PD. It is considered that hallucinations in patients with PD are related to altered integration of sensory input (bottom-up) and prior knowledge (top-down) within the visual system. Our findings indicate that pareidolia in PD involves both bottom-up (thalamus and temporal cortex) and top-down (orbitofrontal cortex) processing disturbances; in contrast, VH predominantly involves bottom-up but not top-down regions. Understanding these distinctions could aid in the development of targeted interventions for hallucinations in patients with PD.
Collapse
Affiliation(s)
- Masakazu Ozawa
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | - Tomotaka Shiraishi
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hidetomo Murakami
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Department of Neurology, Showa University School of Medicine, Tokyo, Japan
| | - Daisuke Yoshimaru
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Asako Onda
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiromasa Matsuno
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Teppei Komatsu
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenichi Sakuta
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kenichiro Sakai
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Tadashi Umehara
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hidetaka Mitsumura
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
3
|
Wu J, Jin X, Xie W, Liu L, Wang F, Zhu L, Shen Y, Qiu L. Global research trends and hotspots in Parkinson's disease psychosis: a 25-year bibliometric and visual analysis. Front Aging Neurosci 2024; 16:1480234. [PMID: 39649718 PMCID: PMC11621064 DOI: 10.3389/fnagi.2024.1480234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/07/2024] [Indexed: 12/11/2024] Open
Abstract
Background Parkinson's disease psychosis (PDP) is one of the most severe and disabling non-motor symptoms in the progression of Parkinson's disease (PD), significantly impacting the prognosis of PD patients. In recent years, there has been an increase in literature on PDP. However, bibliometrics has rarely been applied to PDP research. This study provides an overview of the current state of PDP research and predicts future trends in this field. Methods The literature search was conducted using the Web of Science Core Collection, with the search terms (Parkinson* AND (psychotic* OR hallucination* OR illusion* OR delusion* OR misperception* OR psychosis OR psychoses)). VOSviewer and CiteSpace software were employed to perform bibliometric analysis and visual representation of the search results. Results A total of 603 articles were effectively included. Since 2017, there has been a significant upward trend in publications related to PDP. The United States, the United Kingdom, and Canada were the top three contributing countries in terms of publication volume, with France also having a strong influence in this field. Movement Disorders and King's College London included and published the most articles on PDP. The paper titled "Hallucinations in Parkinson's Disease: Prevalence, Phenomenology, and Risk Factors" received the highest number of citations and average citations. Cluster analysis results identified brain, prevalence, connectivity, and atypical antipsychotics as key hotspots in this field. High-frequency keywords were grouped into three themes: neurobiology, therapeutic strategies, and symptom research. Among them, pimavanserin, risk, and functional connectivity have been the most studied areas in the past 7 years and are likely to remain key topics in future research. Conclusion Research on PDP has garnered increasing attention. This study visualizes PDP research over the past 25 years to analyze global hotspots and trends. It offers researchers a valuable perspective for identifying key topics and understanding research trajectories in this expanding field.
Collapse
Affiliation(s)
- Jianhong Wu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Xin Jin
- Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu, China
| | - Weiming Xie
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Liang Liu
- Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fei Wang
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Ling Zhu
- Jiangyin People's Hospital, Wuxi, Jiangsu, China
| | - Yuan Shen
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Linghe Qiu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
4
|
Lipari N, Galfano A, Venkatesh S, Grezenko H, Sandoval IM, Manfredsson FP, Bishop C. The effects of chemogenetic targeting of serotonin-projecting pathways on L-DOPA-induced dyskinesia and psychosis in a bilateral rat model of Parkinson's disease. Front Neural Circuits 2024; 18:1463941. [PMID: 39634948 PMCID: PMC11615880 DOI: 10.3389/fncir.2024.1463941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/07/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Parkinson's disease (PD) is commonly characterized by severe dopamine (DA) depletion within the substantia nigra (SN) leading to a myriad of motor and non-motor symptoms. One underappreciated and prevalent non-motor symptom, Parkinson's disease-associated psychosis (PDAP), significantly erodes patient and caregiver quality of life yet remains vastly understudied. While the gold standard pharmacotherapy for motor symptoms Levodopa (LD) is initially highly effective, it can lead to motor fluctuations like LD-induced dyskinesia (LID) and non-motor fluctuations such as intermittent PDAP. One source of these fluctuations could be the serotonergic raphe nuclei and their projections. Serotonin (5-HT) neurons possess the machinery necessary to convert and release DA from exogenous LD. In DA-depleted brain regions these 5-HT projections can act as surrogates to the DA system initially compensating but chronically leading to aberrant neuroplasticity which has been linked to LID and may also contribute to non-motor fluctuations. In support, recent work from our lab established a positive relationship between LID and PDAP in parkinsonian rats. Therefore, it was hypothesized that normalizing 5-HT forebrain input would reduce the co-expression of LID and PDAP. Methods To do so, we expressed 5-HT projection specific inhibitory designer receptor exclusively activated by designer drugs (DREADDs) using Cre-dependent AAV9-hM4di in tryptophan hydroxylase 2 (TPH2)-Cre bilaterally 6-OHDA-lesioned rats. Thereafter we used the designer drug Compound 21 to selectively inhibit 5-HT raphe projections during LD treatment to modulate the expression of PDAP, assayed by prepulse inhibition (PPI) and LID, quantified by the abnormal involuntary movements (AIMs) test. Results Our results suggest that chemogenetic inhibition of 5-HT raphe-projecting cells significantly reduces LID without affecting stepping ability or established sensorimotor gating deficits. Discussion Overall, this study provides further evidence for the complex influence of 5-HT raphe-projecting neurons on LD's neurobehavioral effects.
Collapse
Affiliation(s)
- Natalie Lipari
- Department of Psychology, Binghamton University, Binghamton, NY, United States
| | - Ashley Galfano
- Department of Psychology, Binghamton University, Binghamton, NY, United States
| | - Shruti Venkatesh
- Department of Psychology, Binghamton University, Binghamton, NY, United States
| | - Han Grezenko
- Barrow Neurological Institute, Phoenix, AZ, United States
| | | | | | - Christopher Bishop
- Department of Psychology, Binghamton University, Binghamton, NY, United States
| |
Collapse
|
5
|
Stamenović J, Živadinović B, Đurić V. Clinical characteristics and treatment of psychosis in Parkinson's disease: A narrative review. J Chin Med Assoc 2024; 87:972-979. [PMID: 39118220 DOI: 10.1097/jcma.0000000000001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a chronic, progressive, neurodegenerative disorder whose clinical presentation consists of motor and non-motor signs and symptoms. Among the non-motor symptoms, psychosis can occur in the later stages of the disease. Psychosis in PD (PDP) is a common, complex, and significantly disabling disorder associated with poorer quality of life, accelerated cognitive decline, need for hospitalization or institutionalization, and mortality. Hallucinations are a significant symptom of PDP, sporadic at first but more frequent in the later course of the disease, and significantly disrupt daily activities. Appropriate and timely screening of psychotic manifestations is necessary for adequate therapeutic procedures. After the exclusion of comorbid conditions as a possible cause of psychosis, correction of antiparkinsonian therapy may be required, and if necessary, the introduction of antipsychotics. The latest therapeutic recommendations include the use of pimavanserin, if available, otherwise second-generation or atypical antipsychotics. Although PDP has long been recognized as a possible complication in the course of the disease, further clinical studies are needed to fully understand its etiopathogenesis and pathophysiological mechanisms.
Collapse
Affiliation(s)
- Jelena Stamenović
- Medical Faculty, Department of Neurology, University of Niš, Niš, Serbia
- Clinic of Neurology, University Clinical Center of Niš, Niš, Serbia
| | - Biljana Živadinović
- Medical Faculty, Department of Neurology, University of Niš, Niš, Serbia
- Clinic of Neurology, University Clinical Center of Niš, Niš, Serbia
| | | |
Collapse
|
6
|
Imlay RK, Alsayed M, Starcher M, Tager A, Griffith J. Combined Pimavanserin and Maintenance Electroconvulsive Therapy: A Novel Approach to Parkinson's Disease Psychosis. Cureus 2024; 16:e64316. [PMID: 39130906 PMCID: PMC11316412 DOI: 10.7759/cureus.64316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Parkinson's disease (PD) is among the most common neurodegenerative diseases. Parkinson's disease psychosis (PDP) is a potential psychiatric manifestation of PD that is associated with increased morbidity and mortality. The treatment of PD with concomitant PDP is challenging as standard-of-care medication to improve motor symptoms can cause or exacerbate PDP. In this case report, we present an atypical presentation of a 70-year-old female who developed PDP only four years after her initial PD diagnosis, much earlier than the established average. Treatment was particularly complex as her PDP symptoms were refractory to PD medication reduction and oral antipsychotics, yet her PD motor symptoms were well controlled with a deep brain stimulator (DBS). We discuss a combination of pimavanserin and maintenance electroconvulsive therapy (ECT) as a safe and efficacious treatment modality which has led to remission of her PDP while DBS continues to provide adequate management of her PD symptoms. This case improves upon the early recognition of PDP and outlines a unique treatment modality not well described in the literature. This is the only case that demonstrates the efficacy of combining pimavanserin and ECT for refractory PDP in a patient with a DBS.
Collapse
Affiliation(s)
- Riley K Imlay
- Psychiatry, West Virginia University School of Medicine, Charleston, USA
| | - Majd Alsayed
- Psychiatry, Charleston Area Medical Center, Charleston, USA
| | - Madison Starcher
- Psychiatry, West Virginia University School of Medicine, Charleston, USA
| | - Alfred Tager
- Internal Medicine and Psychiatry, Charleston Area Medical Center, Charleston, USA
| | - James Griffith
- Psychiatry, Charleston Area Medical Center, Charleston, USA
| |
Collapse
|
7
|
Isaacson SH, Pahwa R, Pagan F, Abler V, Truong D. Retrospective analyses evaluating the mortality risk associated with pimavanserin or other atypical antipsychotics in patients with Parkinson disease psychosis. Clin Park Relat Disord 2024; 10:100256. [PMID: 38770047 PMCID: PMC11103413 DOI: 10.1016/j.prdoa.2024.100256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/24/2024] [Accepted: 05/05/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction Parkinson's disease (PD) is associated with increased mortality risk (MR), reflecting progression of motor and nonmotor symptoms. PD psychosis (PDP), a common nonmotor symptom, increases with prolonged disease and elevates the MR of PD even further. Pimavanserin is the only FDA-approved treatment for PDP. This review summarizes real-world evidence around the MR of patients with PDP treated with pimavanserin versus off-label atypical antipsychotics. Methods A PubMed search was conducted using the following search terms: pimavanserin AND antipsychotic AND mortality AND Parkinson's disease AND psychosis. Inclusion criteria specified the entry of retrospective, observational, and open-label studies comparing pimavanserin to atypical antipsychotics or untreated controls. Results A total of 10 of the 32 articles met inclusion criteria. Among five comparisons of pimavanserin with atypical antipsychotics, two were large (n = 21,719; n = 21,975), representative, Medicare-database studies, which demonstrated comparable or lower all-cause pimavanserin MR. Among three pimavanserin versus control studies, two reported lower or comparable pimavanserin MR and one, long-term care study reported higher MR for pimavanserin versus non-pimavanserin treated patients with unknown PDP status. Two open-label extensions reported pimavanserin mortality rates of 6.45 and 18.8 deaths per 100 patient-years, which are comparable to, or lower than, mortality rates for PD, PDP, and other atypical antipsychotics. Most studies (70 %; 7 of 10) demonstrated pimavanserin's MR was lower than or similar to other atypical antipsychotics or untreated controls. Conclusions Pimavanserin did not increase the MR in PDP. Pimavanserin's MR appears to be comparable to or lower than other atypical antipsychotics prescribed for PDP, including quetiapine.
Collapse
Affiliation(s)
- Stuart H. Isaacson
- Parkinson’s Disease and Movement Disorders of Boca Raton, 951 NW 13th Street, Bldg. 5-E, Boca Raton, FL 33486, USA
| | - Rajesh Pahwa
- Department of Neurology, University of Kansas Medical Center, 2060 W 39th Ave, Kansas City, KS 66103, USA
| | - Fernando Pagan
- Department of Neurology, Georgetown University Medical Center, 3900 Reservoir Rd NW, Washington, DC 20007, USA
| | - Victor Abler
- Acadia Pharmaceuticals Inc, 12830 El Camino Real, San Diego, CA 92130, USA
| | - Daniel Truong
- The Parkinson and Movement Disorder Institute, 9940 Talbert Ave #100, Fountain Valley, CA 92708, USA
- Department of Psychiatry and Neuroscience, University of California Riverside, 900 University Ave, Riverside, CA 92521, USA
| |
Collapse
|
8
|
Karunakaran KB, Ganapathiraju MK, Jain S, Brahmachari SK, Balakrishnan N. Drug contraindications in comorbid diseases: a protein interactome perspective. NETWORK MODELING ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2024; 13:10. [DOI: 10.1007/s13721-023-00440-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/17/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2025]
Abstract
AbstractAdverse drug reactions (ADRs) are leading causes of death and drug withdrawals and frequently co-occur with comorbidities. However, systematic studies on the effects of drugs on comorbidities are lacking. Drug interactions with the cellular protein–protein interaction (PPI) network give rise to ADRs. We selected 6 comorbid disease pairs, identified the drugs used in the treatment of the individual diseases ‘A’ and ‘B’– 44 drugs in anxiety and depression, 128 in asthma and hypertension, 48 in chronic obstructive pulmonary disease and heart failure, 58 in type 2 diabetes and obesity, 58 in Parkinson’s disease and schizophrenia, and 84 in rheumatoid arthritis and osteoporosis—and categorized them based on whether they aggravate the comorbid condition. We constructed drug target networks (DTNs) and examined their enrichment among genes in disease A/B PPI networks, expressed across 53 tissues and involved in ~ 1000 pathways. To characterize the biological features of the DTNs, we performed principal component analysis and computed the Euclidean distance between DTN component scores and feature loading values. DTNs of disease A drugs not contraindicated in B were affiliated with proteins common to A/B networks or uniquely found in the B network, similarly regulated common pathways, and disease-B specific pathways and tissues. DTNs of disease A drugs contraindicated in B were affiliated with common proteins or those uniquely found in the A network, differentially regulated common pathways, and disease A-specific pathways and tissues. Hence, DTN enrichment in pathways, tissues, and PPI networks of comorbid diseases will help identify drug contraindications in comorbidities.
Collapse
|
9
|
Brinker D, Smilowska K, Paschen S, Antonini A, Moro E, Deuschl G. How to Use the New European Academy of Neurology/Movement Disorder Society European Section Guideline for Invasive Therapies in Parkinson's Disease. Mov Disord Clin Pract 2024; 11:209-219. [PMID: 38214401 DOI: 10.1002/mdc3.13962] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/28/2023] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND The decision to choose invasive treatments for Parkinson's disease (PD) is complex and needs careful consideration. OBJECTIVES Although the recommendations of the European Academy of Neurology/Movement Disorder Society European Section guideline for invasive therapies of PD are useful, the different clinical profiles of people with PD who seek advice for possible invasive therapy need further attention. METHODS AND RESULTS Here we describe 8 clinical standard situations of people with PD unsatisfied with their current oral treatment where invasive therapies may be considered. These are PD patients presenting with the following symptoms: (1) severe motor fluctuations, (2) beginning of levodopa-responsive fluctuations, severe tremor at (3) young or (4) advanced age, (5) impulse control disorders and related behavioral disorders, (6) hallucinations and psychosis, (7) minimal cognitive impairment or mild dementia, and (8) patients in need of palliative care. For some of these conditions, evidence at lower level or simple clinical considerations exist. CONCLUSIONS There are no one-fits-all answers, but physician and patient should discuss each option carefully considering symptom profile, psychosocial context, availability of therapy alternatives, and many other factors. The current paper outlines our proposed approach to these circumstances.
Collapse
Affiliation(s)
- Dana Brinker
- Department of Neurology, UKSH, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Katarzyna Smilowska
- Department of Neurology, UKSH, Christian-Albrechts-University Kiel, Kiel, Germany
- Department of Neurology, Regional Specialist Hospital im. Św. Barbary, Sonowiec, Poland
| | - Steffen Paschen
- Department of Neurology, UKSH, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Study Center for Neurodegenerative Diseases (CESNE), Department of Neuroscience, University of Padua, Padua, Italy
| | - Elena Moro
- Grenoble Alpes University, Chu of Grenoble, Division of Neurology, Grenoble Institute of Neurosciences, Grenoble, France
| | - Günther Deuschl
- Department of Neurology, UKSH, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
10
|
Karunakaran KB, Jain S, Brahmachari SK, Balakrishnan N, Ganapathiraju MK. Parkinson's disease and schizophrenia interactomes contain temporally distinct gene clusters underlying comorbid mechanisms and unique disease processes. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:26. [PMID: 38413605 PMCID: PMC10899210 DOI: 10.1038/s41537-024-00439-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024]
Abstract
Genome-wide association studies suggest significant overlaps in Parkinson's disease (PD) and schizophrenia (SZ) risks, but the underlying mechanisms remain elusive. The protein-protein interaction network ('interactome') plays a crucial role in PD and SZ and can incorporate their spatiotemporal specificities. Therefore, to study the linked biology of PD and SZ, we compiled PD- and SZ-associated genes from the DisGeNET database, and constructed their interactomes using BioGRID and HPRD. We examined the interactomes using clustering and enrichment analyses, in conjunction with the transcriptomic data of 26 brain regions spanning foetal stages to adulthood available in the BrainSpan Atlas. PD and SZ interactomes formed four gene clusters with distinct temporal identities (Disease Gene Networks or 'DGNs'1-4). DGN1 had unique SZ interactome genes highly expressed across developmental stages, corresponding to a neurodevelopmental SZ subtype. DGN2, containing unique SZ interactome genes expressed from early infancy to adulthood, correlated with an inflammation-driven SZ subtype and adult SZ risk. DGN3 contained unique PD interactome genes expressed in late infancy, early and late childhood, and adulthood, and involved in mitochondrial pathways. DGN4, containing prenatally-expressed genes common to both the interactomes, involved in stem cell pluripotency and overlapping with the interactome of 22q11 deletion syndrome (comorbid psychosis and Parkinsonism), potentially regulates neurodevelopmental mechanisms in PD-SZ comorbidity. Our findings suggest that disrupted neurodevelopment (regulated by DGN4) could expose risk windows in PD and SZ, later elevating disease risk through inflammation (DGN2). Alternatively, variant clustering in DGNs may produce disease subtypes, e.g., PD-SZ comorbidity with DGN4, and early/late-onset SZ with DGN1/DGN2.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India.
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan.
| | - Sanjeev Jain
- National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore, India.
| | | | - N Balakrishnan
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | - Madhavi K Ganapathiraju
- Department of Computer Science, Carnegie Mellon University Qatar, Doha, Qatar.
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Kim DD, Procyshyn RM, Jones AA, Gicas KM, Jones PW, Petersson AM, Lee LHN, McLellan-Carich R, Cho LL, Panenka WJ, Leonova O, Lang DJ, Thornton AE, Honer WG, Barr AM. Relationship between drug-induced movement disorders and psychosis in adults living in precarious housing or homelessness. J Psychiatr Res 2024; 170:290-296. [PMID: 38185074 DOI: 10.1016/j.jpsychires.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
BACKGROUND Studies have reported positive associations between drug-induced movement disorders (DIMDs) and symptoms of psychosis in patients with schizophrenia. However, it is not clear which subtypes of symptoms are related to each other, and whether one symptom precedes another. The current report assessed both concurrent and temporal associations between DIMDs and symptoms of psychosis in a community-based sample of homeless individuals. METHODS Participants were recruited in Vancouver, Canada. Severity of DIMDs and psychosis was rated annually, allowing for the analysis of concurrent associations between DIMDs and Positive and Negative Syndrome Scale (PANSS) five factors. A brief version of the PANSS was rated monthly using five psychotic symptoms, allowing for the analysis of their temporal associations with DIMDs. Mixed-effects linear and logistic regression models were used to assess the associations. RESULTS 401 participants were included, mean age of 40.7 years (SD = 11.2) and 77.4% male. DIMDs and symptoms of psychosis were differentially associated with each other, in which the presence of parkinsonism was associated with greater negative symptoms, dyskinesia with disorganized symptoms, and akathisia with excited symptoms. The presence of DIMDs of any type was not associated with depressive symptoms. Regarding temporal associations, preceding delusions and unusual thought content were associated with parkinsonism, whereas dyskinesia was associated with subsequent conceptual disorganization. CONCLUSIONS The current study found significant associations between DIMDs and symptoms of psychosis in individuals living in precarious housing or homelessness. Moreover, there were temporal associations between parkinsonism and psychotic symptoms (delusions or unusual thought content), and the presence of dyskinesia was temporally associated with higher odds of clinically relevant conceptual disorganization.
Collapse
Affiliation(s)
- David D Kim
- Department of Anesthesiology, Pharmacology & Therapeutics, 2176 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada
| | - Ric M Procyshyn
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Andrea A Jones
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Kristina M Gicas
- Department of Psychology, University of the Fraser Valley, Abbotsford, BC, Canada
| | - Paul W Jones
- Department of Psychology, Simon Fraser University, Burnaby, BC, Canada
| | - Anna M Petersson
- Department of Psychology, Simon Fraser University, Burnaby, BC, Canada
| | - Lik Hang N Lee
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Rachel McLellan-Carich
- Department of Anesthesiology, Pharmacology & Therapeutics, 2176 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada
| | - Lianne L Cho
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - William J Panenka
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Olga Leonova
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Donna J Lang
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Allen E Thornton
- Department of Psychology, Simon Fraser University, Burnaby, BC, Canada
| | - William G Honer
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Alasdair M Barr
- Department of Anesthesiology, Pharmacology & Therapeutics, 2176 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Ahizoune A, Ait Berri M. Delusional Infestation in Parkinson's Disease Secondary to Piribedil Escalation: An Uncommon Case Report. Cureus 2024; 16:e53631. [PMID: 38449964 PMCID: PMC10917070 DOI: 10.7759/cureus.53631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2024] [Indexed: 03/08/2024] Open
Abstract
Delusional infestation (DI) is characterized by delusions of being infested by small microorganisms or even inanimate objects without any medical or microbiological evidence. The pathophysiology of DI is not well understood, and there are two types of DI: the primary form, where there is no underlying cause, and the secondary form, which is related to an associated psychiatric disorder, medical condition, or substance use. DI in Parkinson's disease (PD) is rarely reported, and most published cases are due to antiparkinsonian drugs. Piribedil is a dopaminergic agonist used for the symptomatic treatment of PD either as monotherapy or as adjuvant therapy with other antiparkinsonian treatments. We report the case of an 81-year-old man followed for PD at our institution who developed DI after piribedil dose escalation. When DI is secondary to an antiparkinsonian drug, the treatment of choice is based on withdrawing the implicated drug.
Collapse
Affiliation(s)
- Aziz Ahizoune
- Department of Neurology, Military Hospital Moulay Ismail of Meknès, Sidi Mohamed Ben Abdellah University, Fez, MAR
| | - Maha Ait Berri
- Department of Neurology, Military Hospital Moulay Ismail of Meknès, Sidi Mohamed Ben Abdellah University, Fez, MAR
| |
Collapse
|
13
|
Weintraub D, Espay AJ, Sharma VD, Tariot PN, Abler V, Pathak S, Stankovic S. Pimavanserin for psychosis in Parkinson's disease dementia: Subgroup analysis of the HARMONY Trial. Parkinsonism Relat Disord 2024; 119:105951. [PMID: 38113700 DOI: 10.1016/j.parkreldis.2023.105951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Pimavanserin is FDA-approved to treat Parkinson's disease (PD) psychosis. We analyzed the effect of pimavanserin on psychosis in the PD dementia (PDD) subgroup from the phase 3 HARMONY trial. METHODS This subgroup analysis included PDD patients enrolled in an international, multicenter, randomized discontinuation study of pimavanserin for dementia-related psychosis. PDD patients with moderate-to-severe psychosis, age 50-90 years, received pimavanserin 34 mg/day for 12 weeks (open-label period). Those with a sustained psychosis response to pimavanserin at weeks 8 and 12 were randomized during the double-blind period to continue pimavanserin or receive placebo. Primary efficacy endpoint was time to psychosis relapse as measured by the SAPS-H + D and CGI-I. Safety was assessed, as were effects on motor symptoms and cognitive abilities using the ESRS-A and MMSE. RESULTS 392 patients were enrolled in HARMONY (mean age: 72.6 years; 38.8 % female): 59 had PDD; 49/59 remained on pimavanserin during the open-label period (safety analysis set), and 36/49 were randomized to pimavanserin (n = 16) or placebo (n = 20) in the double-blind phase (intent-to-treat analysis set). Risk of psychosis relapse was lower with pimavanserin 34 mg compared with placebo in the double-blind phase (HR = 0.052; 95 % CI 0.016-0.166; 1-sided nominal p < 0.001). During the open-label period, 46.9 % experienced a treatment-emergent adverse event; event incidence was similar across arms in the double-blind period. Pimavanserin did not adversely affect motor or cognitive function in either treatment phase. CONCLUSIONS Pimavanserin significantly reduced risk of psychosis relapse in patients with PDD, was well tolerated, and did not worsen motor or cognitive function.
Collapse
Affiliation(s)
- Daniel Weintraub
- Departments of Psychiatry and Neurology, Perelman School of Medicine at the University of Pennsylvania, 3615 Chestnut Street, #330, Philadelphia, PA, 19104, USA.
| | - Alberto J Espay
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, 3113 Bellevue Ave, Cincinnati, OH, 45219, USA.
| | - Vibhash D Sharma
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA.
| | - Pierre N Tariot
- Banner Alzheimer's Institute and University of Arizona College of Medicine, 901 E Willetta St, Phoenix, AZ, 85006, USA.
| | - Victor Abler
- Acadia Pharmaceuticals Inc., 12830 El Camino Real #400, San Diego, CA, 92130, USA.
| | - Sanjeev Pathak
- Acadia Pharmaceuticals Inc., 12830 El Camino Real #400, San Diego, CA, 92130, USA.
| | - Srdjan Stankovic
- Acadia Pharmaceuticals Inc., 12830 El Camino Real #400, San Diego, CA, 92130, USA.
| |
Collapse
|
14
|
Zhu J, Zhao Y, Jiang Y, Pan Y, Jiang X, Wang Y, Li D, Zhang L. The relationship between obstructive sleep apnea and visual hallucinations in PD patients: a polysomnography study. Front Neurol 2024; 14:1275660. [PMID: 38274875 PMCID: PMC10810068 DOI: 10.3389/fneur.2023.1275660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Purpose Parkinson's disease (PD) patients frequently experience visual hallucinations (VHs) and obstructive sleep apnea (OSA). The aim of this study was to describe the prevalence and clinical correlates of VHs and OSA in the Chinese population with PD. Materials and methods A sample of 489 PD patients was recruited for the present study. Patients were categorized as having formed VHs (FVHs) or minor VHs (MVHs) or as non-hallucinators (NVHs) according to the Unified Parkinson's Disease Rating Scale (UPDRS) and an initial questionnaire. Polysomnography (PSG) was used for objective assessment of sleep. Results VHs were observed in 143 (29.2%) patients. Among them, 75 of the hallucinators experienced MVHs, and 68 experienced FVHs. The disease duration, UPDRS Part III score, Hoehn and Yahr (H-Y) stage, Pittsburgh Sleep Quality Index (PSQI) score and rapid eye movement (REM) sleep behavior disorder (RBD) score of hallucinators were significantly greater than those of non-hallucinators (P < 0.05). We also observed OSA in 38.7, 54.7, and 63.3% of the NVH, MVH, and FVH groups, respectively. PSG showed that the VH groups had a lower total sleep time, lower sleep efficiency, higher arousal index, lower sleep latency, lower N1%, higher apnea-hypopnea index (AHI), higher average duration of apnea, higher respiratory-related arousal (RRA), and lower values of the lowest O2 and mean O2. The forward binary logistic regression model showed that AHI, N1%, RRA and lowest O2 were independently associated with VHs in PD patients. Conclusions Our results confirm the high prevalence of VHs and OSA as well as their relationship in patients with PD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Li Zhang
- Department of Geriatric Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
Khommyatov MR, Smolentseva IG, Golubev SA, Goryunov AV, Samushiya MA. [Clinical presentation and diagnosis of Parkinson's disease in patients with schizophrenia spectrum disorders]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:73-80. [PMID: 39690554 DOI: 10.17116/jnevro202412411173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
OBJECTIVE To study the features of the clinical picture and diagnosis of Parkinson's disease (PD) in patients with schizophrenia spectrum disorders (SSD). MATERIAL AND METHODS The analysis of databases of four psychoneurological dispensaries in Moscow with the allocation of groups of patients with diagnoses of SSD (F20-F25 according to ICD-10) was carried out. Among these groups, a targeted search for patients with an established diagnosis of PD (G20) was conducted. Among this category of patients, the incidence of PD, the therapy taken, the premotor symptoms of PD and the parameters characterizing the features of the clinical picture and diagnosis of PD were analyzed. RESULTS A total of 9124 patients with SSD were identified, among them 11 people diagnosed with PD, while the final incidence of PD was 0.1% (95% CI 0-0.2%). The average age at onset of SSD and PD was 33.18±8.72 years and 58.9±8.1 years, respectively. The average time between the appearance of the first signs of PD and the final diagnosis was 5.09±1.92 years. The most commonly used antipsychotic in the group of patients with PD was quetiapine (54.5%); clozapine and some typical antipsychotics were used less frequently. The most common premotor symptoms were apathy, depression, anxiety, constipation and orthostatic hypotension. Hyposmia and behavioral disorders in the REM sleep phase were less common. CONCLUSION The results confirm the difficulty of managing patients with comorbidity of PD and SSD. Psychiatrists should pay close attention to the features of motor symptoms in patients with SSD and carefully identify clinical signs that may indicate the presence of PD or other neurodegenerative diseases with parkinsonism.
Collapse
Affiliation(s)
| | - I G Smolentseva
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - S A Golubev
- Central State Medical Academy, Moscow, Russia
- Gannushkin Psychiatric Clinical Hospital No. 4, Moscow, Russia
- Mental Health Research Center, Moscow, Russia
| | | | | |
Collapse
|
16
|
Alva G, Cubała WJ, Berrio A, Coate B, Abler V, Pathak S. Safety Profile of Pimavanserin Therapy in Elderly Patients with Neurodegenerative Disease-Related Neuropsychiatric Symptoms: A Phase 3B Study. J Alzheimers Dis 2024; 98:265-274. [PMID: 38427485 PMCID: PMC10977351 DOI: 10.3233/jad-231167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2024] [Indexed: 03/03/2024]
Abstract
Background Pimavanserin, a 5-HT2A receptor inverse agonist/antagonist, is the only medication approved by the FDA for the treatment of hallucinations and delusions associated with Parkinson's disease psychosis (PDP). Further expanding knowledge of the safety profile of pimavanserin in PDP and neurodegenerative diseases (NDD) such as Alzheimer's disease is of great interest for informing its use in patients with PDP (with or without dementia), given this population is highly sensitive to adverse effects following antipsychotic use. Objective This trial evaluated the effects of pimavanserin compared to placebo in frail older adults and elderly patients with neuropsychiatric symptoms related to NDD, such as hallucinations and delusions, to better understand the safety of pimavanserin in this population. Methods This was a phase 3b, 8-week treatment (study duration of up to 16 weeks), multicenter, randomized, double-blind, placebo-controlled, two-arm parallel-group trial (NCT03575052). The primary endpoint was safety and tolerability, measured by treatment-emergent adverse events (TEAEs). Secondary safety endpoints were change from baseline in motor and cognitive function; exploratory endpoints included suicidality, sleep quality, and neuropsychiatric symptoms. Results Incidences of TEAEs were similar between treatment groups; 29.8% reported ≥1 TEAE (pimavanserin: 30.4%; placebo: 29.3%), and 1.8% reported serious TEAEs (pimavanserin: 2.0%; placebo: 1.5%). Pimavanserin did not impact motor- or cognitive-related function. Conclusions Pimavanserin was well tolerated and not associated with motor or cognitive impairment. Together, these findings highlight the manageable and generally favorable safety profile of pimavanserin in patients with NDD, contributing to our knowledge on the safety of pimavanserin as it generalizes to patients with PDP.
Collapse
Affiliation(s)
- Gus Alva
- Department of Psychiatry and Neuroscience, ATP Clinical Research, University of California at Riverside, Riverside, CA, USA
| | - Wiesław J. Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ana Berrio
- ACADIA Pharmaceuticals Inc., Princeton, NJ, USA
| | - Bruce Coate
- ACADIA Pharmaceuticals Inc., Princeton, NJ, USA
| | | | | |
Collapse
|
17
|
Aldred J, Freire-Alvarez E, Amelin AV, Antonini A, Bergmans B, Bergquist F, Bouchard M, Budur K, Carroll C, Chaudhuri KR, Criswell SR, Danielsen EH, Gandor F, Jia J, Kimber TE, Mochizuki H, Robieson WZ, Spiegel AM, Standaert DG, Talapala S, Facheris MF, Fung VSC. Continuous Subcutaneous Foslevodopa/Foscarbidopa in Parkinson's Disease: Safety and Efficacy Results From a 12-Month, Single-Arm, Open-Label, Phase 3 Study. Neurol Ther 2023; 12:1937-1958. [PMID: 37632656 PMCID: PMC10630297 DOI: 10.1007/s40120-023-00533-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/07/2023] [Indexed: 08/28/2023] Open
Abstract
INTRODUCTION Foslevodopa/foscarbidopa, a soluble formulation of levodopa/carbidopa (LD/CD) prodrugs for the treatment of Parkinson's disease (PD), is administered as a 24-hour/day continuous subcutaneous infusion (CSCI) with a single infusion site. The efficacy and safety of foslevodopa/foscarbidopa versus oral immediate-release LD/CD was previously demonstrated in patients with PD in a 12-week, randomized, double-blind, phase 3 trial (NCT04380142). We report the results of a separate 52-week, open-label, phase 3 registrational trial (NCT03781167) that evaluated the safety/tolerability and efficacy of 24-hour/day foslevodopa/foscarbidopa CSCI in patients with advanced PD. METHODS Male and female patients with levodopa-responsive PD and ≥ 2.5 hours of "Off" time/day received 24-hour/day foslevodopa/foscarbidopa CSCI at individually optimized therapeutic doses (approximately 700-4250 mg of LD per 24 hours) for 52 weeks. The primary endpoint was safety/tolerability. Secondary endpoints included changes from baseline in normalized "Off" and "On" time, percentage of patients reporting morning akinesia, Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS), Parkinson's Disease Sleep Scale-2 (PDSS-2), 39-item Parkinson's Disease Questionnaire (PDQ-39), and EuroQol 5-dimension questionnaire (EQ-5D-5L). RESULTS Of 244 enrolled patients, 107 discontinued, and 137 completed treatment. Infusion site events were the most common adverse events (AEs). AEs were mostly nonserious (25.8% of patients reported serious AEs) and mild/moderate in severity. At week 52, "On" time without troublesome dyskinesia and "Off" time were improved from baseline (mean [standard deviation (SD)] change in normalized "On" time without troublesome dyskinesia, 3.8 [3.3] hours; normalized "Off" time, -3.5 [3.1] hours). The percentage of patients experiencing morning akinesia dropped from 77.7% at baseline to 27.8% at week 52. Sleep quality (PDSS-2) and quality of life (PDQ-39 and EQ-5D-5L) also improved. CONCLUSION Foslevodopa/foscarbidopa has the potential to provide a safe and efficacious, individualized, 24-hour/day, nonsurgical alternative for patients with PD. TRIAL REGISTRATION NUMBER ClinicalTrials.gov identifier NCT03781167.
Collapse
Affiliation(s)
- Jason Aldred
- Selkirk Neurology and Inland Northwest Research, 610 S Sherman St, Spokane, WA, 99202, USA.
| | - Eric Freire-Alvarez
- Neurology Department, University General Hospital of Elche, Carrer Almazara, 11, 03203, Elche, Spain
| | - Alexander V Amelin
- Department of Neurology and Neurosurgery, Pavlov First Saint Petersburg State Medical University, Ulitsa L'va Tolstogo, 6-8, St. Petersburg, 197022, Russia
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Padua University, Via VIII Febbraio, 2, 35122, Padua, Italy
| | - Bruno Bergmans
- Department of Neurology, AZ St-Jan Brugge-Oostende AV, Ruddershove 10, 8000, Brugge, Belgium
- Department of Neurology, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Filip Bergquist
- Department of Pharmacology, University of Gothenburg, Universitetsplatsen 1, 405 30, Gothenburg, Sweden
| | - Manon Bouchard
- Clinique Neuro-Lévis, 1190 A Rue de Courchevel #301, Lévis, QC, G6W 0M5, Canada
| | - Kumar Budur
- AbbVie Inc., 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - Camille Carroll
- Faculty of Health, University of Plymouth, Drake Circus, Plymouth, PL4 8AA, UK
| | - K Ray Chaudhuri
- Parkinson's Foundation International Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
- King's College Institute of Psychiatry, Psychology and Neuroscience, 16 De Crespigny Park, London, SE5 8AF, UK
| | - Susan R Criswell
- Department of Neurology, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO, 63130, USA
| | - Erik H Danielsen
- Department of Neurology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Florin Gandor
- Movement Disorders Hospital, Straße Nach Fichtenwalde 16, 14547, Beelitz-Heilstätten, Germany
- Department of Neurology, Otto-Von-Guericke University Magdeburg, Universitätspl. 2, 39106, Magdeburg, Germany
| | - Jia Jia
- AbbVie Inc., 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - Thomas E Kimber
- Department of Neurology, Royal Adelaide Hospital, Port Road, Adelaide, SA, 5000, Australia
- Department of Medicine, University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | | - Amy M Spiegel
- AbbVie Inc., 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - David G Standaert
- Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, 35233, USA
| | | | | | - Victor S C Fung
- Movement Disorders Unit, Westmead Hospital, Cnr Hawkesbury Road and Darcy Rd, Westmead, NSW, 2145, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
18
|
Bhansali S, Assaedi E, Yu JRT, Mandava N, Sonneborn C, Hogue O, Walter BL, Samala RV, Margolius A. End of life care of hospitalized patients with Parkinson disease: a retrospective analysis and brief review. Front Aging Neurosci 2023; 15:1265156. [PMID: 37744391 PMCID: PMC10511646 DOI: 10.3389/fnagi.2023.1265156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Background Towards the end of life (EOL), persons with parkinsonism (PwP) have complex needs and can present with unique palliative care (PC) challenges. There are no widely accepted guidelines to aid neurologists, hospitalists, or PC clinicians in managing the symptoms of PwP at EOL. We examined a population of PwP at EOL, aiming to describe trends of in-hospital management and utilization of PC services. Methods All PwP admitted to two hospitals during 2018 (N = 727) were examined retrospectively, assessing those who died in hospital or were discharged with hospice (EOL group, N = 35) and comparing them to the main cohort. Their demographics, clinical data, engagement of multidisciplinary and palliative services, code status changes, invasive care, frequency of admissions, and medication administration were assessed. Results Among the EOL group, 8 expired in hospital, and 27 were discharged to hospice. Forty-six percent of EOL patients received a PC consultation during their admission. The median interval from admission to death was 37 days. Seventy-seven percent had a full code status on admission. Compared to hospice patients, those who expired in hospital had higher rates of invasive procedures and intensive care unit transfers (41% vs. 75%, in both variables), and lower rates of PC involvement (52% vs. 25%). The transition of code status change for the EOL group from Full code to Do Not Resuscitate (DNR) occurred at a median 4-5 days from admission. For patients that passed in the hospital, the median days from transition of code status to death was 0(IQR 0-1). Levodopa dose deviations were frequent in both EOL and non-EOL group, but contraindicated medications were infrequently administered (11% in EOL group vs. 9% in non-EOL group). Conclusion Our data suggest a low utilization of PC services and delayed discussions of goals of care. More work is needed to raise awareness of inpatient teams managing PwP regarding the unique but common challenges facing PwP with advanced disease. A brief narrative review summarizing the suggested management of symptoms common to hospitalized PwP near EOL is provided.
Collapse
Affiliation(s)
- Sakhi Bhansali
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Ekhlas Assaedi
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jeryl Ritzi T. Yu
- Institute for Neurosciences, St. Luke’s Medical Center, Quezon City, Philippines
- University of the East Ramon Magsaysay Memorial Medical Center, Quezon City, Philippines
| | - Nymisha Mandava
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Claire Sonneborn
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Olivia Hogue
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | | | - Renato V. Samala
- Department of Palliative and Supportive Care, Cleveland Clinic, Cleveland, OH, United States
| | - Adam Margolius
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
19
|
Spetsieris PG, Eidelberg D. Parkinson's disease progression: Increasing expression of an invariant common core subnetwork. Neuroimage Clin 2023; 39:103488. [PMID: 37660556 PMCID: PMC10491857 DOI: 10.1016/j.nicl.2023.103488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Notable success has been achieved in the study of neurodegenerative conditions using reduction techniques such as principal component analysis (PCA) and sparse inverse covariance estimation (SICE) in positron emission tomography (PET) data despite their widely differing approach. In a recent study of SICE applied to metabolic scans from Parkinson's disease (PD) patients, we showed that by using PCA to prespecify disease-related partition layers, we were able to optimize maps of functional metabolic connectivity within the relevant networks. Here, we show the potential of SICE, enhanced by disease-specific subnetwork partitions, to identify key regional hubs and their connections, and track their associations in PD patients with increasing disease duration. This approach enabled the identification of a core zone that included elements of the striatum, pons, cerebellar vermis, and parietal cortex and provided a deeper understanding of progressive changes in their connectivity. This subnetwork constituted a robust invariant disease feature that was unrelated to phenotype. Mean expression levels for this subnetwork increased steadily in a group of 70 PD patients spanning a range of symptom durations between 1 and 21 years. The findings were confirmed in a validation sample of 69 patients with up to 32 years of symptoms. The common core elements represent possible targets for disease modification, while their connections to external regions may be better suited for symptomatic treatment.
Collapse
Affiliation(s)
- Phoebe G Spetsieris
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, United States
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, United States; Molecular Medicine and Neurology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, United States.
| |
Collapse
|
20
|
Shabir G, Saeed A, Zahid W, Naseer F, Riaz Z, Khalil N, Muneeba, Albericio F. Chemistry and Pharmacology of Fluorinated Drugs Approved by the FDA (2016-2022). Pharmaceuticals (Basel) 2023; 16:1162. [PMID: 37631077 PMCID: PMC10458641 DOI: 10.3390/ph16081162] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Fluorine is characterized by high electronegativity and small atomic size, which provide this molecule with the unique property of augmenting the potency, selectivity, metabolic stability, and pharmacokinetics of drugs. Fluorine (F) substitution has been extensively explored in drug research as a means of improving biological activity and enhancing chemical or metabolic stability. Selective F substitution onto a therapeutic or diagnostic drug candidate can enhance several pharmacokinetic and physicochemical properties such as metabolic stability and membrane permeation. The increased binding ability of fluorinated drug target proteins has also been reported in some cases. An emerging line of research on F substitution has been addressed by using 18F as a radiolabel tracer atom in the extremely sensitive methodology of positron emission tomography (PET) imaging. This review aims to report on the fluorinated drugs approved by the US Food and Drug Administration (FDA) from 2016 to 2022. It cites selected examples from a variety of therapeutic and diagnostic drugs. FDA-approved drugs in this period have a variety of heterocyclic cores, including pyrrole, pyrazole, imidazole, triazole, pyridine, pyridone, pyridazine, pyrazine, pyrimidine, triazine, purine, indole, benzimidazole, isoquinoline, and quinoline appended with either F-18 or F-19. Some fluorinated oligonucleotides were also authorized by the FDA between 2019 and 2022.
Collapse
Affiliation(s)
- Ghulam Shabir
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan;
| | - Aamer Saeed
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan;
| | - Wajeeha Zahid
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Fatima Naseer
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Zainab Riaz
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Nafeesa Khalil
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Muneeba
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
21
|
Nieto-Escamez F, Obrero-Gaitán E, Cortés-Pérez I. Visual Dysfunction in Parkinson's Disease. Brain Sci 2023; 13:1173. [PMID: 37626529 PMCID: PMC10452537 DOI: 10.3390/brainsci13081173] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Non-motor symptoms in Parkinson's disease (PD) include ocular, visuoperceptive, and visuospatial impairments, which can occur as a result of the underlying neurodegenerative process. Ocular impairments can affect various aspects of vision and eye movement. Thus, patients can show dry eyes, blepharospasm, reduced blink rate, saccadic eye movement abnormalities, smooth pursuit deficits, and impaired voluntary and reflexive eye movements. Furthermore, visuoperceptive impairments affect the ability to perceive and recognize visual stimuli accurately, including impaired contrast sensitivity and reduced visual acuity, color discrimination, and object recognition. Visuospatial impairments are also remarkable, including difficulties perceiving and interpreting spatial relationships between objects and difficulties judging distances or navigating through the environment. Moreover, PD patients can present visuospatial attention problems, with difficulties attending to visual stimuli in a spatially organized manner. Moreover, PD patients also show perceptual disturbances affecting their ability to interpret and determine meaning from visual stimuli. And, for instance, visual hallucinations are common in PD patients. Nevertheless, the neurobiological bases of visual-related disorders in PD are complex and not fully understood. This review intends to provide a comprehensive description of visual disturbances in PD, from sensory to perceptual alterations, addressing their neuroanatomical, functional, and neurochemical correlates. Structural changes, particularly in posterior cortical regions, are described, as well as functional alterations, both in cortical and subcortical regions, which are shown in relation to specific neuropsychological results. Similarly, although the involvement of different neurotransmitter systems is controversial, data about neurochemical alterations related to visual impairments are presented, especially dopaminergic, cholinergic, and serotoninergic systems.
Collapse
Affiliation(s)
- Francisco Nieto-Escamez
- Department of Psychology, University of Almeria, 04120 Almeria, Spain
- Center for Neuropsychological Assessment and Rehabilitation (CERNEP), 04120 Almeria, Spain
| | - Esteban Obrero-Gaitán
- Department of Health Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23071 Jaen, Spain;
| | - Irene Cortés-Pérez
- Department of Health Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23071 Jaen, Spain;
| |
Collapse
|
22
|
d’Angremont E, Begemann MJH, van Laar T, Sommer IEC. Cholinesterase Inhibitors for Treatment of Psychotic Symptoms in Alzheimer Disease and Parkinson Disease: A Meta-analysis. JAMA Neurol 2023; 80:813-823. [PMID: 37358841 PMCID: PMC10294019 DOI: 10.1001/jamaneurol.2023.1835] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/17/2023] [Indexed: 06/27/2023]
Abstract
Importance Psychotic symptoms greatly increase the burden of disease for people with neurodegenerative disorders and their caregivers. Cholinesterase inhibitors (ChEIs) may be effective treatment for psychotic symptoms in these disorders. Previous trials only evaluated neuropsychiatric symptoms as a secondary and an overall outcome, potentially blurring the outcomes noted with ChEI use specifically for psychotic symptoms. Objective To quantitatively assess the use of ChEIs for treatment of individual neuropsychiatric symptoms, specifically hallucinations and delusions, in patients with Alzheimer disease (AD), Parkinson disease (PD), and dementia with Lewy bodies (DLB). Data Sources A systematic search was performed in PubMed (MEDLINE), Embase, and PsychInfo, without year restrictions. Additional eligible studies were retrieved from reference lists. The final search cutoff date was April 21, 2022. Study Selection Studies were selected if they presented the results of placebo-controlled randomized clinical trials, including at least 1 donepezil, rivastigmine, or galantamine treatment arm in patients with AD, PD, or DLB; if they applied at least 1 neuropsychiatric measure including hallucinations and/or delusions; and if a full-text version of the study was available in the English language. Study selection was performed and checked by multiple reviewers. Data Extraction and Synthesis Original research data were requested on eligible studies. A 2-stage meta-analysis was then performed, using random-effects models. Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines were followed for extracting data and assessing the data quality and validity. Data extraction was checked by a second reviewer. Main Outcomes and Measures Primary outcomes were hallucinations and delusions; secondary outcomes included all other individual neuropsychiatric subdomains as well as the total neuropsychiatric score. Results In total, 34 eligible randomized clinical trials were selected. Individual participant data on 6649 individuals (3830 [62.6%] women; mean [SD] age, 75.0 [8.2] years) were obtained from 17 trials (AD: n = 12; PD: n = 5; individual participant data were not available for DLB). An association with ChEI treatment was shown in the AD subgroup for delusions (-0.08; 95% CI, -0.14 to -0.03; P = .006) and hallucinations (-0.09; 95% CI, -0.14 to -0.04; P = .003) and in the PD subgroup for delusions (-0.14; 95% CI, -0.26 to -0.01; P = .04) and hallucinations (-0.08, 95% CI -0.13 to -0.03; P = .01). Conclusions and Relevance The results of this individual participant data meta-analysis suggest that ChEI treatment improves psychotic symptoms in patients with AD and PD with small effect sizes.
Collapse
Affiliation(s)
- Emile d’Angremont
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Groningen, the Netherlands
| | - Marieke J. H. Begemann
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Groningen, the Netherlands
| | - Teus van Laar
- Department of Neurology, University Medical Center Groningen, Groningen, the Netherlands
| | - Iris E. C. Sommer
- Department of Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
23
|
Isaacson SH, Goldstein M, Pahwa R, Singer C, Klos K, Pucci M, Zhang Y, Crandall D, Koblan KS, Navia B. Ulotaront, a Trace Amine-Associated Receptor 1/Serotonin 5-HT 1A Agonist, in Patients With Parkinson Disease Psychosis: A Pilot Study. Neurol Clin Pract 2023; 13:e200175. [PMID: 37273942 PMCID: PMC10238151 DOI: 10.1212/cpj.0000000000200175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 04/04/2023] [Indexed: 06/06/2023]
Abstract
Background and Objectives Ulotaront (SEP-363856) is a trace amine-associated receptor 1 agonist with 5-HT1A receptor agonist activity currently in phase 3 clinical development for the treatment of schizophrenia. In this exploratory, flexibly dosed study, ulotaront was evaluated for the treatment of Parkinson disease psychosis (PDP). Methods Patients with PDP requiring antipsychotic therapy were randomized, double-blind to ulotaront (25, 50, or 75 mg/d) or placebo. Mixed Model for Repeated Measures was used to assess change from baseline in the Scale for the Assessment of Positive Symptoms for Parkinson Disease (SAPS-PD) at 6 weeks (primary end point). Results The efficacy analysis sample comprised 38 patients (ulotaront, n = 24; placebo, n = 14). SAPS-PD total scores were numerically reduced in ulotaront-treated vs placebo-treated patients from week 1 to week 6: Least squares mean (95% confidence interval) difference in change from baseline at week 6 was -1.1 (-6.5, 4.3, p = 0.681). PDP symptom complete remission (≥100% improvement [reduction] from baseline in SAPS-PD total score) was observed in 25% of ulotaront-treated vs 0% of placebo-treated patients. SAPS-PD and Neuropsychiatric Inventory hallucinations subscales were numerically reduced vs placebo, and SAPS-PD total scores were reduced in patients with greater cognitive impairment (baseline Mini-Mental State Examination [MMSE] scores ≤24). Ulotaront improved Scales for Outcomes in Parkinson Disease Sleep Scale - Daytime Sleepiness scores (p = 0.022). There was no worsening of Unified Parkinson Disease Rating Scale Part III motor score, MMSE, or vital signs. Adverse events (≥10%) with ulotaront vs placebo included hallucinations (24% vs 14%), confusional state (20% vs 14%), dizziness (16% vs 7%), nausea (12% vs 7%), and falls (12% vs 21%). Discussion In this exploratory pilot study, ulotaront may decrease PDP symptoms without worsening motor function, particularly in patients with cognitive impairment. Trial Registration Information ClinicalTrials.gov identifier: NCT02969369; submitted: November 17, 2016; study start date: December 31, 2016. Classification of Evidence This Class II study was an exploratory pilot study that was underpowered to detect a statistically significant difference between ulotaront and placebo in the treatment of patients with Parkinson disease psychosis without worsening motor function.
Collapse
Affiliation(s)
- Stuart H Isaacson
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Mark Goldstein
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Rajesh Pahwa
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Carlos Singer
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Kevin Klos
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Michael Pucci
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Yi Zhang
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - David Crandall
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Kenneth S Koblan
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Bradford Navia
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| |
Collapse
|
24
|
Accinni T, Fanella M, Frascarelli M, Buzzanca A, Kotzalidis GD, Putotto C, Marino B, Panzera A, Moschillo A, Pasquini M, Biondi M, Di Bonaventura C, Di Fabio F. The Relationship between Motor Symptoms, Signs, and Parkinsonism with Facial Emotion Recognition Deficits in Individuals with 22q11.2 Deletion Syndrome at High Genetic Risk for Psychosis. Acta Neurol Scand 2023; 2023:1-10. [DOI: 10.1155/2023/8546610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background. The 22q11.2 Deletion Syndrome (22q11.2DS) is a genetic condition at high risk of developing both psychosis and motor disorders. Social Cognition (SC) deficits have been associated not only with schizophrenia but also with Parkinson’s disease (PD). The present study assessed SC deficits in 22q11.2DS and investigated the interaction between motor symptoms and deficits in Facial Emotion Expressions (FEE) recognition and in Theory of Mind (ToM) tasks in people with 22q11.2DS. Methods. We recruited 38 individuals with 22q11.2DS without psychosis (
, DEL) and 18 with 22q11.2DS and psychosis (
, DEL_SCZ). The Positive And Negative Syndrome Scale (PANSS), Ekman’s 60 Faces Test (EK-60F), the Awareness of Social Inference Test (TASIT EmRec), and the Movement Disorder Society-Unified Parkinson’s Disease Rating Scale part III (UPDRS III) were administered. Correlations were sought between UPDRS III and both TASIT EmRec and EK-60F scores. Analyses were conducted separately for each psychopathological subgroup. Results. Higher UPDRS III (
) and lower EK-60F (
) scores were observed in the DEL_SCZ group. We found inverse correlations between UPDRS III and both TASIT EmRec (
,
) and EK-60F (
,
) scores in the whole sample. Correlations were no longer significant in the DEL_SCZ group (UPDRS III-TASIT EmRec
; UPDRS III-EK60F
) whilst being stronger in the DEL group (TASIT EmRec,
,
; EK60F,
,
). Analyses were adjusted for CPZ Eq and IQ. Conclusions. A modulation between FEE recognition deficits and motor symptoms and signs was observed in the 22q11.2DS group, likely affecting patients’ quality of life.
Collapse
Affiliation(s)
- Tommaso Accinni
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Martina Fanella
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Marianna Frascarelli
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonino Buzzanca
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Georgios D. Kotzalidis
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Faculty of Medicine and Psychology, Via di Grottarossa 1035-1039, 00189 Rome, Italy
| | - Carolina Putotto
- Department of Pediatrics, Obstetrics and Gynecology, Sapienza University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Bruno Marino
- Department of Pediatrics, Obstetrics and Gynecology, Sapienza University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Alessia Panzera
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonella Moschillo
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Faculty of Medicine and Psychology, Via di Grottarossa 1035-1039, 00189 Rome, Italy
| | - Massimo Pasquini
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Massimo Biondi
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Carlo Di Bonaventura
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Fabio Di Fabio
- Department of Human Neurosciences, Sapienza University of Rome, Faculty of Medicine and Dentistry, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
25
|
Lipari N, Centner A, Glinski J, Cohen S, Manfredsson FP, Bishop C. Characterizing the relationship between L-DOPA-induced-dyskinesia and psychosis-like behaviors in a bilateral rat model of Parkinson's disease. Neurobiol Dis 2023; 176:105965. [PMID: 36526089 DOI: 10.1016/j.nbd.2022.105965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease associated psychosis (PDAP) is a prevalent non-motor symptom (NMS) that significantly erodes patients' and caregivers' quality of life yet remains vastly understudied. One potential source of PDAP in late-stage Parkinson's disease (PD) is the common dopamine (DA) replacement therapy for motor symptoms, Levodopa (L-DOPA). Given the high incidence of L-DOPA-induced dyskinesia (LID) in later phases of PD, this study sought to characterize the relationship between PDAP and LID in a bilateral medial forebrain bundle 6-hydroxydopamine hydrobromide (6-OHDA) lesion rat model. To assess PDAP in this model, prepulse inhibition (PPI), a well-validated assay of sensorimotor gating, was employed. First, we tested whether a bilateral lesion alone or after chronic L-DOPA treatment was sufficient to induce PPI dysfunction. Rats were also monitored for LID development, using the abnormal involuntary movements (AIMs) test, to examine PPI and LID associations. In experiment 2, Vilazodone (VZD), a serotonin transporter (SERT) blocker and 1A receptor (5-HT1A) partial agonist was administered to test its potential efficacy in reducing LID and PPI dysfunction. Once testing was complete, tissue was collected for high performance liquid chromatography (HPLC) to examine the monoamine levels in motor and non-motor circuits. Results indicate that bilateral DA lesions produced motor deficits and that chronic L-DOPA induced moderate AIMs; importantly, rats that developed more severe AIMs were more likely to display sensorimotor gating dysfunction. In addition, VZD treatment dose-dependently reduced L-DOPA-induced AIMs without impairing L-DOPA efficacy, although VZD's effects on PPI were limited. Altogether, this project established the bilateral 6-OHDA lesion model accurately portrayed LID and PDAP-like behaviors, uncovered their potential relationship, and finally, demonstrated the utility of VZD for reducing LID.
Collapse
Affiliation(s)
- Natalie Lipari
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Ashley Centner
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - John Glinski
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Sophie Cohen
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | | | | |
Collapse
|
26
|
Corcoran J, Huang AH, Miyasaki JM, Tarolli CG. Palliative care in Parkinson disease and related disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 191:107-128. [PMID: 36599503 DOI: 10.1016/b978-0-12-824535-4.00017-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although neuropalliative care is a relatively new field, there is increasing evidence for its use among the degenerative parkinsonian syndromes, including idiopathic Parkinson disease, progressive supranuclear palsy, multiple system atrophy, dementia with Lewy bodies, and corticobasal syndrome. This chapter outlines the current state of evidence for palliative care among individuals with the degenerative parkinsonian syndromes with discussion surrounding: (1) disease burden and needs across the conditions; (2) utility, timing, and methods for advance care planning; (3) novel care models for the provision of palliative care; and 4) end-of-life care issues. We also discuss currently unmet needs and unanswered questions in the field, proposing priorities for research and the assessment of implemented care models.
Collapse
Affiliation(s)
- Jennifer Corcoran
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrew H Huang
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Janis M Miyasaki
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Christopher G Tarolli
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States.
| |
Collapse
|
27
|
Rajagopalan K, Rashid N, Kumar S, Doshi D. Health care resource utilization patterns among patients with Parkinson's disease psychosis: analysis of Medicare beneficiaries treated with pimavanserin or other-atypical antipsychotics. J Med Econ 2023; 26:34-42. [PMID: 36444507 DOI: 10.1080/13696998.2022.2152600] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pimavanserin (PIM) is the only FDA-approved atypical antipsychotic (AAP) for hallucinations and delusions associated with Parkinson's disease psychosis (PDP). Comparative real-world analyses demonstrating its benefits are needed. OBJECTIVES To evaluate health care resource utilization (HCRU) outcomes among PDP patients treated with PIM vs. other-AAPs. METHODS Retrospective cohort analysis of Parts A, B, and D claims from 100% Medicare sample from 01 January 2013-31 December 2019 was conducted. PDP Patients initiating (i.e. index date) continuous monotherapy (PIM vs. other-AAPs) for ≥12-months during 01 January 2014-31 December 2018 without 12-months pre-index AAP use were selected after 1:1 propensity score matching (PSM) on 31 variables (sex, race, region, age, and 27 Elixhauser comorbidities). HCRU outcomes included: annual all-cause and psychiatric hospitalization (short-term stay, long-term stay, and SNF-stay [skilled nursing facility]) rates, annual all-cause and psychiatric-ER visit rates, mean per-patient-per-year (PPPY) hospitalizations, and average length of stay (ALOS). PIM and other-AAPs were compared using generalized linear models (GLM) controlled for demographic characteristics, comorbidities, coexisting-dementia, and coexisting insomnia. RESULTS Of 12,164 PDP patients, 48.41% (n = 5,889) were female, and mean age was 77 (±8.14) years. Among 1:1 matched patients (n = 842 in each), 37.8% (n = 319) on PIM vs. 49.8% (n = 420) on other-AAPs (p < .05) reported ≥1 all-cause hospitalizations, respectively. Specifically, short-term and SNF-stay among PIM patients vs. other-AAPs were: 34% (n = 286) vs. 46.2% (n = 389) and 20.2% (n = 170) vs. 31.8% (n = 267) (p < .05), respectively. Similarly, 9.6% (n = 81) of PIM vs. 14.6% (n = 123) of other-AAPs patients had ≥1 psychiatric hospitalization (p < .05). Furthermore, ≥1 all-cause and psychiatric ER visit among PIM vs. other-AAPs were 61.6% (n = 519) vs. 69.4% (n = 584) and 5.2% (n = 43) vs. 10.2% (n = 86) (p < .05), respectively. PIM also had significantly lower ALOS, and mean PPPY short-term hospitalization and SNF-stays. CONCLUSIONS In this analysis of PDP patients, PIM monotherapy resulted in nearly 12% and 7% lower all-cause hospitalizations and ER visits vs. other-AAPs.
Collapse
|
28
|
Should patients with Parkinson’s disease only visit a neurologist’s office? - a narrative review of neuropsychiatric disorders among people with Parkinson’s disease. CURRENT PROBLEMS OF PSYCHIATRY 2022. [DOI: 10.2478/cpp-2022-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Abstract
Introduction: Parkinson’s disease is a neurodegenerative disease that is often accompanied by disorders such as depression, psychotic disorders, cognitive disorders, anxiety disorders, sleep disorders, impulse control disorders. The aim of the study was to review the literature and present the characteristics of neuropsychiatric disorders occurring in people suffering from Parkinson’s disease, with the specification of the above-mentioned disorders.
Material and method: The literature available on the PubMed platform from 1986 to 2022 was reviewed using the following keywords: Parkinson’s disease, depression, anxiety disorders, psychotic disorders, sleep disorders, cognitive disorders, impulse control disorders. Original studies, reviews, meta-analyzes and internet sources were analyzed.
Results: The above-mentioned neuropsychiatric disorders appear with different frequency among people suffering from Parkinson’s disease and occur at different times of its duration or even precede its onset for many years. The non-motor symptoms in the form of depressed mood, energy loss or changes in the rhythm of the day may result in a delay of appropriate therapy and thus in complications. Neuropathological changes in the course of Parkinson’s disease as well as dopaminergic drugs used in its therapy influence the development of neuropsychiatric disorders.
Conclusions: In order to avoid misdiagnosis, practitioners should use, e.g. scales intended for patients with Parkinson’s disease. To prevent the consequences of the aforementioned disease entities, methods of early diagnosis, determination of risk factors and standardization of the treatment process must be determined. Consistent care for patients with Parkinson’s disease is significant, not only in the neurological field, but also in the psychiatric one.
Collapse
|
29
|
Elsibai H, Kualleny M, Fandy TE. Management of Parkinson's Disease Psychosis. Sr Care Pharm 2022; 37:339-344. [PMID: 35879841 DOI: 10.4140/tcp.n.2022.339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objectives To discuss therapeutic management of Parkinson's disease psychosis (PDP) and the burdens associated with it. Parkinson's disease (PD) is an incurable, progressive, and devastating neurodegenerative disease. The prolonged use of dopamine agonists to improve the motor symptoms of PD may result in nonmotor complications such as psychosis. These complications are as devastating as the motor symptoms that accompany PD. PDP is associated with financial burden for patients and health care organizations. Data Sources PubMed search using the phrases Parkinson's disease, Parkinson's disease psychosis, clozapine, quetiapine, and "pimavanserin." Conclusion Pimavanserin is the only US Food and Drug Administration approved drug for the management of hallucinations and delusions associated with PD psychosis. The off-label use of clozapine and quetiapine has demonstrated clinical utility; however, there is still a need for the development of novel therapeutic molecules to avoid side effects associated with current therapeutic options and reduce the burdens of patients, caregivers, and health care institutions.
Collapse
|
30
|
Kashihara K, Maeda T, Yoshida K. Safety and tolerability of aripiprazole in patients with psychosis associated with Parkinson's disease-Results of a multicenter open trial. Neuropsychopharmacol Rep 2022; 42:135-141. [PMID: 35226404 PMCID: PMC9216359 DOI: 10.1002/npr2.12235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 12/27/2021] [Accepted: 01/11/2022] [Indexed: 11/08/2022] Open
Abstract
Aim To evaluate the effect of aripiprazole on psychosis and motor function in Japanese Parkinson's disease patients. Methods Patients with Parkinson's disease and hallucinations and/or delusions were enrolled. They were administered aripiprazole 3 mg/day, with dosage increased or reduced as needed. Patients were evaluated using the Brief Psychiatric Rating Scale (BPRS), Clinical Global Impression‐Severity (CGI‐S) scale, and Clinical Global Impression‐Improvement scale for psychiatric response; Hoehn & Yahr staging and Unified Parkinson's Disease Rating Scale (UPDRS) part III for motor response; Mini‐Mental State Examination (MMSE) and Frontal Assessment Battery (FAB) for cognitive response; and Schwab and England Activities of Daily Living scale for daily activities of patients, before and at 2, 4, and 12 weeks after initiation of open‐label aripiprazole administration. This study was registered at the University Hospital Medical Information Network Center (registration number: UMIN000007711). Results Nine of the 24 enrolled patients discontinued the study. Among them, eight patients discontinued the trial on account of their worsening parkinsonian symptoms. There were no differences in age, disease duration, disease severity, and MMSE and FAB scores at baseline between patients who continued and discontinued the study. However, in patients who continued aripiprazole administration at 3 mg/day or less significantly improved BPRS, CGI‐S scale, and UPDRS parts I and III scores. Conclusion Significant improvements in hallucinations and delusions can be expected, although aripiprazole may aggravate parkinsonism in Parkinson's disease patients. Low‐dose use of aripiprazole may be useful for managing Parkinson's disease patients with psychosis, but only with close observation of extrapyramidal symptoms. We conducted a multicenter clinical study to determine the efficacy of aripiprazole to relieve psychosis in Parkinson's disease. We found that aripiprazole reduced psychotic symptoms without cognitive impairment (based on BPRS and CGI‐S scores), and motor symptoms also improved (based on UPDRS scores). We believe that our study makes a significant contribution to the literature because the results suggest aripiprazole may be a better alternative to the drugs that are currently being prescribed for Parkinson disease‐associated psychosis.![]()
Collapse
Affiliation(s)
- Kenichi Kashihara
- Department of Neurology, Okayama Kyokuto Hospital, Okayama, Japan.,Okayama Neurology Clinic, Okayama, Japan
| | - Tetsuya Maeda
- Department of Neurology and Movement Disorder Research, Research Institute for Brain and Blood Vessels-Akita, Akita, Japan.,Division of Neurology and Gerontology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kazuto Yoshida
- Department of Neurology, Asahikawa Red Cross Hospital, Asahikawa, Japan.,Department of Neurology, Kashiwaba Neurosurgical Hospital, Sapporo, Japan
| |
Collapse
|
31
|
Lanzetti S, Di Biase V. Small Molecules as Modulators of Voltage-Gated Calcium Channels in Neurological Disorders: State of the Art and Perspectives. Molecules 2022; 27:1312. [PMID: 35209100 PMCID: PMC8879281 DOI: 10.3390/molecules27041312] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely expressed in the brain, heart and vessels, smooth and skeletal muscle, as well as in endocrine cells. VGCCs mediate gene transcription, synaptic and neuronal structural plasticity, muscle contraction, the release of hormones and neurotransmitters, and membrane excitability. Therefore, it is not surprising that VGCC dysfunction results in severe pathologies, such as cardiovascular conditions, neurological and psychiatric disorders, altered glycemic levels, and abnormal smooth muscle tone. The latest research findings and clinical evidence increasingly show the critical role played by VGCCs in autism spectrum disorders, Parkinson's disease, drug addiction, pain, and epilepsy. These findings outline the importance of developing selective calcium channel inhibitors and modulators to treat such prevailing conditions of the central nervous system. Several small molecules inhibiting calcium channels are currently used in clinical practice to successfully treat pain and cardiovascular conditions. However, the limited palette of molecules available and the emerging extent of VGCC pathophysiology require the development of additional drugs targeting these channels. Here, we provide an overview of the role of calcium channels in neurological disorders and discuss possible strategies to generate novel therapeutics.
Collapse
Affiliation(s)
| | - Valentina Di Biase
- Institute of Pharmacology, Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Peter-Mayr Strasse 1, A-6020 Innsbruck, Austria;
| |
Collapse
|
32
|
Segal GS, Xie SJ, Paracha SUR, Grossberg GT. Psychosis in Parkinson's Disease: Current Treatment Options and Impact on Patients and Caregivers. J Geriatr Psychiatry Neurol 2021; 34:274-279. [PMID: 34219522 DOI: 10.1177/08919887211018280] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease seen in older adults after Alzheimer's disease, with increasing prevalence worldwide. Parkinson's disease psychosis (PDP) is a common, non-motor feature of PD, which increases caregiver stress and is a risk-factor for nursing home placement. In this paper we review PDP epidemiology, features, diagnosis, and treatment. PDP most often presents with sequential development of minor and then increasingly complex visual hallucinations mediated by dopaminergic-serotonergic interactions activating the mesolimbic pathway, with contributions from other structures and neurotransmitters. Appropriate evaluation of differential diagnoses for psychosis is vital before diagnosing PDP. Initial treatment should involve non-pharmacologic approaches. If these are unsuccessful and PDP symptoms significantly impact the patient's and or their caregivers' quality of life and functions, then pharmacotherapy is indicated. Pimavanserin is a recently FDA-approved pharmacologic treatment for PDP with a better profile of balanced effectiveness and safety compared to previous use of atypical antipsychotics. Early diagnosis and safer, more effective treatments for PDP should help reduce caregiver burden and enable caregivers to continue to provide care at home versus institutionalization.
Collapse
Affiliation(s)
- Gilad S Segal
- 7547Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Sophie J Xie
- 7547Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Saif-Ur-Rahman Paracha
- Department of Psychiatry and Behavioral Neuroscience, 7547Saint Louis University School of Medicine, MO, USA
| | - George T Grossberg
- Samuel W. Fordyce Professor and Director of Geriatric Psychiatry, Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, MO, USA
| |
Collapse
|
33
|
Abstract
Advanced Parkinson disease (PD) is associated with treatment-related motor fluctuations and reduced ability to perform activities of daily living. Progression of non-motor symptoms and medication-induced adverse effects complicate focused approach to motor symptom management, frequently accelerating reduced quality of life. It is thus critical for clinicians to consider disease progression versus therapeutic contributions when balancing management decisions. Such an approach requires careful recognition of inflection points resulting from therapeutic decisions and should prompt consideration of reduced pharmacologic burden and increased reliance on non-pharmacologic strategies in advanced disease. The successful approach to advanced PD requires a multidisciplinary effort focused on improving the patient's and family's quality of life, sometimes requiring sacrifice of motor symptom benefit. Here, we emphasize management strategies in advanced PD, focusing on the need to balance the therapeutic approach across advancing motor symptoms, progressive non-motor features, and potential pharmacologic adverse effects.
Collapse
Affiliation(s)
- Helen Hwang
- Department of Neurology, 7548Washington University School of Medicine, St Louis, MO, USA
| | - Scott A Norris
- Department of Neurology, 7548Washington University School of Medicine, St Louis, MO, USA
- Department of Radiology, 7548Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
34
|
Pham Nguyen TP, Abraham DS, Thibault D, Weintraub D, Willis AW. Low continuation of antipsychotic therapy in Parkinson disease - intolerance, ineffectiveness, or inertia? BMC Neurol 2021; 21:240. [PMID: 34167473 PMCID: PMC8223332 DOI: 10.1186/s12883-021-02265-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/31/2021] [Indexed: 12/25/2022] Open
Abstract
Background Antipsychotics are used in Parkinson disease (PD) to treat psychosis, mood, and behavioral disturbances. Commonly used antipsychotics differ substantially in their potential to worsen motor symptoms through dopaminergic receptor blockade. Recent real-world data on the use and continuation of antipsychotic therapy in PD are lacking. The objectives of this study are to (1) examine the continuation of overall and initial antipsychotic therapy in individuals with PD and (2) determine whether continuation varies by drug dopamine receptor blocking activity. Methods We conducted a retrospective cohort study using U.S. commercially insured individuals in Optum 2001–2019. Adults aged 40 years or older with PD initiating antipsychotic therapy, with continuous insurance coverage for at least 6 months following drug initiation, were included. Exposure to pimavanserin, quetiapine, clozapine, aripiprazole, risperidone, or olanzapine was identified based on pharmacy claims. Six-month continuation of overall and initial antipsychotic therapy was estimated by time to complete discontinuation or switching to a different antipsychotic. Cox proportional hazards models evaluated factors associated with discontinuation. Results Overall, 38.6% of 3566 PD patients in our sample discontinued antipsychotic therapy after the first prescription, 61.4% continued with overall treatment within 6 months of initiation. Clozapine use was too rare to include in statistical analyses. Overall therapy discontinuation was more likely for those who initiated medications with known dopamine-receptor blocking activity (adjusted hazard ratios 1.76 [95% confidence interval 1.40–2.20] for quetiapine, 2.15 [1.61–2.86] for aripiprazole, 2.12 [1.66–2.72] for risperidone, and 2.07 [1.60–2.67] for olanzapine), compared with serotonin receptor-specific pimavanserin. Initial antipsychotic therapy discontinuation also associated with greater dopamine-receptor blocking activity medication use – adjusted hazard ratios 1.57 (1.28–1.94), 1.88 (1.43–2.46), 2.00 (1.59–2.52) and 2.03 (1.60–2.58) for quetiapine, aripiprazole, risperidone, and olanzapine, respectively, compared with pimavanserin. Similar results were observed in sensitivity analyses. Conclusions Over one-third of individuals with PD discontinued antipsychotic therapy, especially if the initial drug has greater dopamine-receptor blocking activity. Understanding the drivers of antipsychotic discontinuation, including ineffectiveness, potentially inappropriate use, clinician inertia, patient adherence and adverse effects, is needed to inform clinical management of psychosis in PD and appropriate antipsychotic use in this population. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02265-x.
Collapse
Affiliation(s)
- Thanh Phuong Pham Nguyen
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Blockley Hall 829, Philadelphia, PA, 19104, USA. .,Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Danielle S Abraham
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Blockley Hall 829, Philadelphia, PA, 19104, USA.,Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dylan Thibault
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Blockley Hall 829, Philadelphia, PA, 19104, USA.,Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Weintraub
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Blockley Hall 829, Philadelphia, PA, 19104, USA.,Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison W Willis
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Blockley Hall 829, Philadelphia, PA, 19104, USA.,Department of Neurology Translational Center for Excellence for Neuroepidemiology and Neurological Outcomes Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
35
|
Sarangi SC, Sopory P, Reeta KH. Chronic Neurological Disorders: Genetic and Epigenetic Markers for Monitoring of Pharmacotherapy. Neurol India 2021; 69:252-259. [PMID: 33904433 DOI: 10.4103/0028-3886.314522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction Chronic neurological diseases are a major cause of mortality and morbidity in the world. With increasing life expectancy in the developing world, the incidence and prevalence of these diseases are predicted to rise even further. This has also contributed to an increase in disability-adjusted life years (DALYs) for noncommunicable diseases. Treatment for such diseases also poses a challenge with multiple genetic and epigenetic factors leading to a varied outcome. Personalization of treatment is one way that treatment outcome/prognosis of disease can be improved, and pharmacogenomics plays a significant role in this context. Methodology This article reviewed the evidence pertaining to the association of genetic and epigenetic markers with major neurological disorders like multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD), which are a major source of burden among neurological disorders. Types of studies included are peer-reviewed original research articles from the PubMed database (1999-2018). Results This study compiled data regarding specific genetic and epigenetic markers with a significant correlation between the clinical diagnosis of the disease and prognosis of therapy from 65 studies. In a single platform, this review highlights the clues to some vital questions, such as why interferon beta (IFN-β) therapy fails to improve symptoms in all MS patients? why cholinesterase inhibitors fail to improve cognitive impairment in a subset of people suffering from AD? or why some individuals on levodopa (L-DOPA) for PD suffer from side-effects ranging from dyskinesia to hallucination while others do not? Conclusion This article summarizes the genetic and epigenetic factors that may either require monitoring or help in deciding future pharmacotherapy in a patient suffering from MS, AD, and PD. As the health care system develops and reaches newer heights, we expect more and more of these biomarkers to be used as pharmacotherapeutic outcome indicators.
Collapse
Affiliation(s)
| | - Pranav Sopory
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
36
|
Terravecchia C, Mostile G, Rascunà C, Arabia G, Barone P, Marconi R, Morgante L, Quattrone A, Nicoletti A, Zappia M. Does an association between cigarette smoking and Parkinson's Disease-related psychosis exist? Insights from a large non-demented cohort. J Neurol Sci 2021; 427:117509. [PMID: 34082149 DOI: 10.1016/j.jns.2021.117509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/14/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Parkinson's Disease-related Psychosis (PDP) encompasses a spectrum of symptoms ranging from "minor" hallucinations to formed hallucinations and delusions. Notably, cognitive impairment has been recognized as the strongest risk factor for PDP. Several evidences suggest a possible role of cigarette smoking in both cognition and psychotic syndromes. OBJECTIVES To evaluate the possible independent association between cigarette smoking and PDP in a large cohort of non-demented PD patients. METHODS A cohort of non-demented PD patients was selected from the FRAGAMP study population. All participants underwent a standardised structured questionnaire to assess demographic, clinical and environmental exposure data. Clinical features were assessed using UPDRS, HY stage, AIMS, MMSE and Hamilton Rating Scale for Depression. Presence of psychotic symptoms was assessed using UPDRS-I.2 score. Diagnosis of PDP was made according to NINDS/NIMH criteria. RESULTS Four hundred eighty-five non-demented PD patients were enrolled [292 men (60.2%); mean age ± SD 65.6 ± 9.8]. Among them, 28 (5.8%) had PDP. Multivariate analysis, adjusting by HY stage, MMSE and LED, shown an independent association between PDP and "nightmares-abnormal movements during sleep" and current smoking [adjOR 7.39 (95%CI 1.45-37.69; P-value 0.016)]. CONCLUSIONS Our findings provide interesting insights about the possible role of current smoking in facilitating the occurrence of psychotic symptoms in PD.
Collapse
Affiliation(s)
- Claudio Terravecchia
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Giovanni Mostile
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy; Oasi Research Institute-IRCCS, Troina, Italy
| | - Cristina Rascunà
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Gennarina Arabia
- Institute of Neurology, University "Magna Graecia", Catanzaro, Italy
| | - Paolo Barone
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | | | | | - Andrea Quattrone
- Institute of Neurology, University "Magna Graecia", Catanzaro, Italy
| | - Alessandra Nicoletti
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Mario Zappia
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy.
| |
Collapse
|
37
|
Garcia Ruiz PJ. Electroconvulsive Therapy and Movement Disorders. New Perspectives on A Time-Tested Therapy. Mov Disord Clin Pract 2021; 8:521-524. [PMID: 33981784 DOI: 10.1002/mdc3.13180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/24/2021] [Accepted: 02/14/2021] [Indexed: 11/07/2022] Open
Abstract
Electroconvulsive therapy (ECT) has been a very well known therapy in Psychiatry for over 80 years. ECT is considered useful in treating acute mania, severe depression and other psychiatric conditions. Over time, this therapy has also been used in several movement disorders including Parkinson disease (PD) and Huntington disease (HD). In this brief review, I summarize the recent History and evolution of ECT, its proven and potential applications in movement disorders as well as its potential mechanisms.
Collapse
Affiliation(s)
- Pedro J Garcia Ruiz
- Movement Disorders Unit, Department of Neurology Fundacion Jimenez Diaz Madrid Spain
| |
Collapse
|
38
|
Dhingra A, Janjua AU, Hack L, Waserstein G, Palanci J, Hermida AP. Exploring Nonmotor Neuropsychiatric Manifestations of Parkinson Disease in a Comprehensive Care Setting. J Geriatr Psychiatry Neurol 2021; 34:181-195. [PMID: 32242493 DOI: 10.1177/0891988720915525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson disease (PD) is a debilitating neurological condition that includes both motor symptoms and nonmotor symptoms (NMS). Psychiatric complaints comprise NMS and are collectively referred to as neuropsychiatric manifestations. Common findings include atypical depressive symptoms, anxiety, psychosis, impulse control disorder, deterioration of cognition, and sleep disturbances. Quality of life (QoL) of patients suffering from NMS is greatly impacted and many times can be more debilitating than motor symptoms of PD. We expand on knowledge gained from treatment models within a comprehensive care model that incorporates multidisciplinary specialists working alongside psychiatrists to treat PD. Insight into background, clinical presentations, and treatment options for patients suffering from neuropsychiatric manifestations of PD are discussed. Identifying symptoms early can help improve QoL, provide early symptom relief, and can assist tailoring treatment plans that limit neuropsychiatric manifestations.
Collapse
Affiliation(s)
- Amitha Dhingra
- Department of Psychiatry and Behavioral Sciences, 12239Emory University School of Medicine, Atlanta, GA, USA
| | - A Umair Janjua
- Department of Psychiatry and Behavioral Sciences, 12239Emory University School of Medicine, Atlanta, GA, USA
| | - Laura Hack
- Department of Psychiatry and Behavioral Sciences, 12239Emory University School of Medicine, Atlanta, GA, USA
| | - Gabriella Waserstein
- Department of Psychiatry and Behavioral Sciences, 12239Emory University School of Medicine, Atlanta, GA, USA
| | - Justin Palanci
- Department of Psychiatry and Behavioral Sciences, 12239Emory University School of Medicine, Atlanta, GA, USA
| | - Adriana P Hermida
- Department of Psychiatry and Behavioral Sciences, 12239Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
39
|
Rukavina K, Batzu L, Boogers A, Abundes-Corona A, Bruno V, Chaudhuri KR. Non-motor complications in late stage Parkinson's disease: recognition, management and unmet needs. Expert Rev Neurother 2021; 21:335-352. [PMID: 33522312 DOI: 10.1080/14737175.2021.1883428] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The burden of non-motor symptoms (NMS) is a major determinant of health-related quality of life in Parkinson's disease (PD), particularly at its late stage.Areas covered: The late stage is usually defined as the period from unstable advanced to the palliative stage, characterized by a combination of emerging treatment-resistant axial motor symptoms (freezing of gait, postural instability, falls and dysphagia), as well as both non-dopaminergic and dopaminergic NMS: cognitive decline, neuropsychiatric symptoms, aspects of dysautonomia, pain and sleep disturbances (insomnia and excessive day-time sleepiness). Here, the authors summarize the current knowledge on NMS dominating the late stage of PD and propose a pragmatic and clinically focused approach for their recognition and treatment.Expert opinion: The NMS progression pattern is complex and remains under-researched. While dopamine-dependent NMS may improve with dopamine replacement therapy, non-dopamine dependent NMS worsen progressively and culminate at the late stages of PD. Furthermore, some PD specific features could interact negatively with other comorbidities, multiple medication use and frailty - the evaluation of these aspects is important in the creation of personalized management plans in the late stage of PD.
Collapse
Affiliation(s)
- Katarina Rukavina
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience at King's College and King's College Hospital NHS Foundation Trust, London, UK.,Parkinson Foundation Centre of Excellence, King's College Hospital, London, UK
| | - Lucia Batzu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience at King's College and King's College Hospital NHS Foundation Trust, London, UK.,Parkinson Foundation Centre of Excellence, King's College Hospital, London, UK
| | - Alexandra Boogers
- Department of Neurology, University Hospital Leuven, Leuven, U.Z, Belgium
| | - Arturo Abundes-Corona
- Department of Neurology, Clinical Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Mexico City, México.,Neurology Department, American British Cowdray Medical Center IAP, Mexico City, Mexico
| | - Veronica Bruno
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - K Ray Chaudhuri
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience at King's College and King's College Hospital NHS Foundation Trust, London, UK.,Parkinson Foundation Centre of Excellence, King's College Hospital, London, UK
| |
Collapse
|
40
|
Rashid N, Shim A, Andes S, Quale S, Abler V. Treatment Patterns With Antipsychotics in Long-Term Care Patients With Parkinson's Disease Psychosis. J Appl Gerontol 2021; 41:198-206. [PMID: 33504252 DOI: 10.1177/0733464820987032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study assessed treatment change patterns in Parkinson's disease psychosis (PDP) residents receiving antipsychotic (AP) therapies in U.S. long-term care (LTC) facilities. Residents with PDP in LTC between 01/01/13 and 06/30/16 were identified with ≥1 claim of psychosis, hallucinations, or delusions after PD diagnosis. Treatment patterns were evaluated during the 12 months post index. We identified 864 PDP residents: 408 (47.2%) on AP therapy and 456 (52.8%) on no AP therapy. A total of 335 residents (82.1%) continued, 13 (3.2%) discontinued, 11 (2.7%) switched, and 49 (12.0%) augmented (used ≥2 APs) their index AP therapy. Based on the multivariate regression analysis, younger age, male gender, anemia, anxiolytic use or anxiety, sedatives/hypnotic use, bladder disorders including urinary tract infections, coronary conditions, diabetes, hypertension, and dementia were associated with a higher likelihood of treatment change. Understanding the factors associated with treatment change may inform ways to improve management of PDP in the U.S. LTC setting.
Collapse
Affiliation(s)
| | - Andrew Shim
- Acadia Pharmaceuticals Inc., San Diego, CA, USA
| | | | | | | |
Collapse
|
41
|
Pimavanserin Treatment for Parkinson's Disease Psychosis in Clinical Practice. PARKINSONS DISEASE 2021; 2021:2603641. [PMID: 33489083 PMCID: PMC7801085 DOI: 10.1155/2021/2603641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 01/04/2023]
Abstract
Background Parkinson's disease psychosis (PDP) is a common, nonmotor symptom of Parkinson's disease (PD), which may affect up to 60% of patients and is associated with impaired quality of life, increased healthcare costs, and nursing home placement, among other adverse outcomes. Characteristic symptoms of PDP include illusions; visual, auditory, tactile, and olfactory hallucinations; and delusions. PDP symptoms typically progress over its course from being mild, infrequent, and often untroubling to complex, sometimes constant, and potentially highly disturbing. PDP has traditionally been treated with atypical antipsychotics (e.g., clozapine and quetiapine) although these are not approved for this indication and clozapine requires frequent white blood cell count monitoring due to the risk of agranulocytosis. Pimavanserin is a newer atypical antipsychotic with highly selective binding to serotonergic receptors, no evidence for worsening motor symptoms in PD, and no need for white blood cell count monitoring. It is currently the only approved medication indicated for PDP treatment. However, because it was approved relatively recently (2016), clinical experience with pimavanserin is limited. Case Presentations. A wide variety of representative clinical scenarios are presented, each with distinct variables and complications. Issues addressed include distinguishing PDP from similar symptoms caused by other disorders such as dementia, coordinating pimavanserin with other PD medications and with deep brain stimulation, adapting pimavanserin dosing for optimal benefit and tolerability, and recognizing variability of PDP symptoms due to patients' changing life circumstances. Conclusions These scenarios provide multiple insights regarding PDP management and the role of pimavanserin. Effective treatment of PDP may reduce disturbing symptoms of psychosis, thus improving patient function and quality of life. In addition, effective pharmacotherapy for PDP may also facilitate the use of other medications needed to treat neurological symptoms of PD (e.g., tremor, bradykinesia, and dyskinesia), although they may also have adverse effects that contribute to symptoms of PDP.
Collapse
|
42
|
Kesserwani H. Delusional Infestation Secondary to the Dopamine Agonist Ropirinole in a Patient With Parkinson's Disease: A Case Report With an Outline of the Biology of Psychosis. Cureus 2021; 13:e12880. [PMID: 33633909 PMCID: PMC7899248 DOI: 10.7759/cureus.12880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2021] [Indexed: 11/05/2022] Open
Abstract
Delusional infestation (DI) is a thought disorder, a delusion that one is infested with pathogens. Remarkably, these patients do not typically exhibit symptoms of anxiety or depression. The role of the dopamine D2 receptor is central to the idea of psychosis. In this article, we present a case of ropirinole-induced delusional skin infestation in a patient with Parkinson's disease, that was reversible with drug discontinuation. We seize upon this opportunity to discuss the pathology of the dopamine receptors, the glutamate N-methyl D-aspartate (NMDA) receptors and the serotonin 5-hydroxytryptamine (5HT-2A) receptors in the generation of psychosis. We outline the fundamental pharmacodynamical differences between the typical and atypical anti-psychotics that will help us understand how these agents work favorably and adversely. We also briefly review the neuroradialogy of psychosis and adumbrate on the mismatch between the meso-limbic system (motivational) and the salience network (valence) as the driver of the psychotic phenomenon.
Collapse
|
43
|
Urso D, Gnoni V, Filardi M, Logroscino G. Delusion and Delirium in Neurodegenerative Disorders: An Overlooked Relationship? Front Psychiatry 2021; 12:808724. [PMID: 35115974 PMCID: PMC8804700 DOI: 10.3389/fpsyt.2021.808724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/24/2021] [Indexed: 12/04/2022] Open
Abstract
Delusions are part of the neuropsychiatric symptoms that patients suffering from neurodegenerative conditions frequently develop at some point of the disease course and are associated with an increased risk of cognitive and functional decline. Delirium is a syndrome characterized by acute onset of deficits in attention, awareness, and cognition that fluctuate in severity over a short time period. Delusions and delirium are frequently observed in the context of neurodegeneration, and their presence can easily mislead clinicians toward a misdiagnosis of psychiatric disorder further delaying the proper treatment. Risk factors for developing delusion and delirium in neurodegenerative conditions have been investigated separately while the possible interplay between these two conditions has not been explored so far. With this study, we aim to achieve a more comprehensive picture of the relationship between delusions and delirium in neurodegeneration by analyzing prevalence and subtypes of delusions in different neurodegenerative disorders; providing an overview of clinical tools to assess delusions in neurodegenerative patients and how delusions are covered by delirium assessment tools and discussing the possible common pathophysiology mechanisms between delusion and delirium in neurodegenerative patients. A more extensive characterization of the relationship between delusions and delirium may help to understand whether delusions may constitute a risk factor for delirium and may ameliorate the management of both conditions in patients with neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniele Urso
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, Pia Fondazione Cardinale G. Panico, University of Bari Aldo Moro, Bari, Italy.,Department of Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Valentina Gnoni
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, Pia Fondazione Cardinale G. Panico, University of Bari Aldo Moro, Bari, Italy.,Department of Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Marco Filardi
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, Pia Fondazione Cardinale G. Panico, University of Bari Aldo Moro, Bari, Italy.,Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, Pia Fondazione Cardinale G. Panico, University of Bari Aldo Moro, Bari, Italy.,Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
44
|
Sgambato V. Breathing new life into neurotoxic-based monkey models of Parkinson's disease to study the complex biological interplay between serotonin and dopamine. PROGRESS IN BRAIN RESEARCH 2020; 261:265-285. [PMID: 33785131 DOI: 10.1016/bs.pbr.2020.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous clinical studies have shown that the serotonergic system also degenerates in patients with Parkinson's disease. The causal role of this impairment in Parkinson's symptomatology and the response to treatment remains to be refined, in particular thanks to approaches allowing the two components DA and 5-HT to be isolated if possible. We have developed a macaque monkey model of Parkinson's disease exhibiting a double lesion (dopaminergic and serotonergic) thanks to the sequential use of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and MDMA (3,4-methylenedioxy-N-methamphetamine) (or MDMA prior MPTP). We characterized this monkey model by multimodal imaging (PET, positron emission tomography with several radiotracers; DTI, diffusion tensor imaging), behavioral assessments (parkinsonism, dyskinesia, neuropsychiatric-like behavior) and post-mortem analysis (with DA and 5-HT markers). When administrated after MPTP, MDMA damaged the 5-HT presynaptic system without affecting the remaining DA neurons. The lesion of 5-HT fibers induced by MDMA altered rigidity and prevented dyskinesia and neuropsychiatric-like symptoms induced by levodopa therapy in MPTP-treated animals. Interestingly also, prior MDMA administration aggravates the parkinsonian deficits and associated DA injury. Dystonic postures, action tremor and global spontaneous activities were significantly affected. All together, these data clearly indicate that late or early lesions of the 5-HT system have a differential impact on parkinsonian symptoms in the macaque model of Parkinson's disease. Whether MDMA has an impact on neuropsychiatric-like symptoms such as apathy, anxiety, depression remains to be addressed. Despite its limitations, this toxin-based double-lesioned monkey model takes on its full meaning and provides material for the experimental study of the heterogeneity of patients.
Collapse
Affiliation(s)
- Véronique Sgambato
- Université de Lyon, CNRS UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.
| |
Collapse
|
45
|
Weil RS, Reeves S. Hallucinations in Parkinson's disease: new insights into mechanisms and treatments. ADVANCES IN CLINICAL NEUROSCIENCE & REHABILITATION 2020; 19:ONNS5189. [PMID: 33102741 PMCID: PMC7116251 DOI: 10.47795/onns5189] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hallucinations are common in Parkinson's disease and can be distressing to patients and their families. They are associated with higher rates of nursing home placement and with increased mortality. Their underlying mechanisms have been elusive, but recent advances in network imaging provides some intriguing insights into possible underlying drivers. Treatment is complicated by risk of worsening Parkinson's motor symptoms and by higher rates of mortality with antipsychotics, but new therapeutic avenues are emerging that offer potential hope.
Collapse
|
46
|
Knolle F, Garofalo S, Viviani R, Justicia A, Ermakova AO, Blank H, Williams GB, Arrondo G, Ramachandra P, Tudor-Sfetea C, Bunzeck N, Duezel E, Robbins TW, Barker RA, Murray GK. Altered subcortical emotional salience processing differentiates Parkinson's patients with and without psychotic symptoms. NEUROIMAGE-CLINICAL 2020; 27:102277. [PMID: 32540629 PMCID: PMC7298672 DOI: 10.1016/j.nicl.2020.102277] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/30/2020] [Accepted: 05/05/2020] [Indexed: 01/03/2023]
Abstract
Emotional salience processing differentiates PD patients with and without psychosis. Enhanced striatal, hippocampal and midbrain responses in PD patients with psychosis. Indication for ‘jumping to conclusions’ bias in the same PD patients with psychosis. Aberrant top-down and salience processing associated with PD psychosis. Similar deficits as proposed in ‘aberrant salience hypothesis’ of schizophrenia.
Objective Current research does not provide a clear explanation for why some patients with Parkinson’s Disease (PD) develop psychotic symptoms. The ‘aberrant salience hypothesis’ of psychosis has been influential and proposes that dopaminergic dysregulation leads to inappropriate attribution of salience to irrelevant/non-informative stimuli, facilitating the formation of hallucinations and delusions. The aim of this study is to investigate whether non-motivational salience is altered in PD patients and possibly linked to the development of psychotic symptoms. Methods We investigated salience processing in 14 PD patients with psychotic symptoms, 23 PD patients without psychotic symptoms and 19 healthy controls. All patients were on dopaminergic medication for their PD. We examined emotional salience using a visual oddball fMRI paradigm that has been used to investigate early stages of schizophrenia spectrum psychosis, controlling for resting cerebral blood flow as assessed with arterial spin labelling fMRI. Results We found significant differences between patient groups in brain responses to emotional salience. PD patients with psychotic symptoms had enhanced brain responses in the striatum, dopaminergic midbrain, hippocampus and amygdala compared to patients without psychotic symptoms. PD patients with psychotic symptoms showed significant correlations between the levels of dopaminergic drugs they were taking and BOLD signalling, as well as psychotic symptom scores. Conclusion Our study suggests that enhanced signalling in the striatum, dopaminergic midbrain, the hippocampus and amygdala is associated with the development of psychotic symptoms in PD, in line with that proposed in the ‘aberrant salience hypothesis’ of psychosis in schizophrenia.
Collapse
Affiliation(s)
- F Knolle
- Department of Psychiatry, University of Cambridge, Cambridge, UK; Department of Neuroradiology, Technical University Munich, Munich, Germany.
| | - S Garofalo
- University of Bologna, Department of Psychology, Bologna, Italy
| | - R Viviani
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria; Psychiatry and Psychotherapy Clinic III, University of Ulm, Ulm, Germany
| | - A Justicia
- Department of Psychiatry, University of Cambridge, Cambridge, UK; IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - A O Ermakova
- Faculty of Natural Sciences, Imperial College London, UK
| | - H Blank
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - G B Williams
- Department of Clinical Neuroscience and WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - G Arrondo
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - P Ramachandra
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - C Tudor-Sfetea
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - N Bunzeck
- Institute of Psychology I, University of Lübeck, Lübeck, Germany
| | - E Duezel
- Otto-von-Guericke University Magdeburg, Institute of Cognitive Neurology and Dementia Research, Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - T W Robbins
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - R A Barker
- Department of Clinical Neuroscience and WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - G K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Komendová M, Urban J. Dual-retention mechanism of dopamine-related compounds on monolithic stationary phase with zwitterion functionality. J Chromatogr A 2020; 1618:460893. [PMID: 31980263 DOI: 10.1016/j.chroma.2020.460893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 11/28/2022]
Abstract
Seven retention models have been selected to describe a dual-retention behavior of ten dopamine-related compounds on polymer-based monolithic stationary phase with zwitterion sulfobetaine functionality. Regression quality, as well as a statistical significance of individual regression parameters, have been evaluated. Better regression performance showed two four-parameter models when compared to three-parameter models. On the other hand, limited number of experimental points disqualified statistical robustness of four-parameter models. Among three-parameter models, retention description introduced by Horváth and Liang provided comparable quality of regression at significantly improved robustness. Multivariate analysis of the best three-parameter models provided the description of physicochemical properties of dopamine precursors and metabolites. Principal component analysis and logistic regression allowed structural characterization of dopamine-related compounds based solely on regression parameters extracted from an isocratic elution data. Both polarity and type of functional groups has been correctly assigned for 3-methoxytyramine that has not been part of an evaluation study. Among applied dual-retention models, Horváth´s model, initially developed to describe a retention of ionic compounds on nonpolar stationary phases, provided robust regression of experimental data and allowed an extraction of structural characteristics of dopamine-related compounds.
Collapse
Affiliation(s)
- Martina Komendová
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jiří Urban
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| |
Collapse
|
48
|
Sellers J, Darby RR, Farooque A, Claassen DO. Pimavanserin for Psychosis in Parkinson's Disease-Related Disorders: A Retrospective Chart Review. Drugs Aging 2019; 36:647-653. [PMID: 30924099 PMCID: PMC6584229 DOI: 10.1007/s40266-019-00655-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background Psychosis is common in Parkinson’s disease-related disorders and is associated with significant morbidity. Pimavanserin is a newly approved treatment for Parkinson’s disease psychosis, but real-world experience with pimavanserin has been limited by small sample sizes and limited assessment of longitudinal outcomes. Objective The aim was to summarize the clinical experience with pimavanserin in a large cohort of patients with Parkinson’s disease-related psychosis. Methods We conducted a retrospective chart review of patients who were prescribed pimavanserin at Vanderbilt University Medical Center in the southeast United States between May 2016 and July 2018. We used Chi-squared analyses to compare efficacy and tolerability of pimavanserin, considering patient diagnosis, presence of dementia or delusions, use of deep brain stimulation, and prior antipsychotic failure. Additionally, we compared the clinical characteristics of patients who started treatment and those who did not, to evaluate safety outcomes. Results We identified 107 patients prescribed pimavanserin, and 91 began treatment. Clinical improvement in psychosis was documented in 76% of patients (69/91) and did not differ based on diagnosis, presence of dementia, delusions, use of deep brain stimulation, or prior antipsychotic failure. Adverse effects were reported in 20 patients (22%), the most common of which was worsening gait instability (5/91, 5%). Side effects led to cessation of therapy in 11 of the 91 patients (12%). At current follow-up, 50 (65%) of 77 living patients remain on treatment, with a mean treatment duration of 14.6 months. Although most of these patients are on pimavanserin monotherapy (33/50, 66%), 17 patients (34%) are on a dual-antipsychotic regimen. The living patients no longer on treatment stopped pimavanserin primarily because of a lack of perceived benefit (11/77, 14%), side effects (9/77, 12%), or both (1/77, 1%), though six patients (8%) stopped for reasons unrelated to medication effects, including the desire to reduce overall medication burden and negative media reporting on pimavanserin. Conclusions Study results emphasize long-term efficacy and tolerability of pimavanserin for psychosis in Parkinson’s disease-related disorders, including patients with dementia, delusions, deep brain stimulation use, or prior antipsychotic failure.
Collapse
Affiliation(s)
- Jessie Sellers
- Department of Neurology, Division Behavioral and Cognitive Neurology, Vanderbilt University Medical Center, 1161 21st Avenue, A-0118 MCN, Nashville, TN, 37232, USA
| | - R Ryan Darby
- Department of Neurology, Division Behavioral and Cognitive Neurology, Vanderbilt University Medical Center, 1161 21st Avenue, A-0118 MCN, Nashville, TN, 37232, USA
| | - Alma Farooque
- Department of Neurology, Division Behavioral and Cognitive Neurology, Vanderbilt University Medical Center, 1161 21st Avenue, A-0118 MCN, Nashville, TN, 37232, USA
| | - Daniel O Claassen
- Department of Neurology, Division Behavioral and Cognitive Neurology, Vanderbilt University Medical Center, 1161 21st Avenue, A-0118 MCN, Nashville, TN, 37232, USA.
| |
Collapse
|
49
|
Chao L. Do Gulf War veterans with high levels of deployment-related exposures display symptoms suggestive of Parkinson’s disease? Int J Occup Med Environ Health 2019; 32:503–526. [PMID: 31309787 PMCID: PMC11892701 DOI: 10.13075/ijomeh.1896.01346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Objectives Veterans of the 1991 Gulf War (GW) were exposed to a myriad of potentially hazardous chemicals during deployment. Epidemiological data suggest a possible link between chemical exposures and Parkinson’s disease (PD); however, there have been no reliable data on the incidence or prevalence of PD among GW veterans to date. This study included the following 2 questions: 1. Do deployed GW veterans display PD-like symptoms? and 2. Is there a relationship between the occurrence and quantity of PD-like symptoms, and the levels of deployment-related exposures in GW veterans? Material and Methods Self-reports of symptoms and exposures to deployment-related chemicals were filled out by 293 GW veterans, 202 of whom had undergone 3 Tesla volumetric measurements of basal ganglia volumes. Correlation analyses were used to examine the relationship between the frequency of the veterans’ self-reported exposures to deployment-related chemicals, motor and non-motor symptoms of PD, and the total basal ganglia volumes. Results Healthy deployed GW veterans self-reported few PD-like non-motor symptoms and no motor symptoms. In contrast, GW veterans with Gulf War illness (GWI) self-reported more PD-like motor and non-motor symptoms, and more GW-related exposures. Compared to healthy deployed veterans, those with GWI also had lower total basal ganglia volumes. Conclusions Although little is known about the long-term consequences of GWI, findings from this study suggest that veterans with GWI show more symptoms as those seen in PD/prodromal PD, compared to healthy deployed GW veterans. Int J Occup Med Environ Health. 2019;32(4):503–26
Collapse
Affiliation(s)
- Linda Chao
- University of California, San Francisco, USA (Department of Radiology and Biomedical Imaging)
- University of California, San Francisco, USA (Department of Psychiatry)
- San Francisco Veterans Affairs Medical Center, San Francisco, USA (Center for Imaging of Neurodegenerative Diseases)
| |
Collapse
|
50
|
Jellinger KA. Neuropathology and pathogenesis of extrapyramidal movement disorders: a critical update-I. Hypokinetic-rigid movement disorders. J Neural Transm (Vienna) 2019; 126:933-995. [PMID: 31214855 DOI: 10.1007/s00702-019-02028-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Extrapyramidal movement disorders include hypokinetic rigid and hyperkinetic or mixed forms, most of them originating from dysfunction of the basal ganglia (BG) and their information circuits. The functional anatomy of the BG, the cortico-BG-thalamocortical, and BG-cerebellar circuit connections are briefly reviewed. Pathophysiologic classification of extrapyramidal movement disorder mechanisms distinguish (1) parkinsonian syndromes, (2) chorea and related syndromes, (3) dystonias, (4) myoclonic syndromes, (5) ballism, (6) tics, and (7) tremor syndromes. Recent genetic and molecular-biologic classifications distinguish (1) synucleinopathies (Parkinson's disease, dementia with Lewy bodies, Parkinson's disease-dementia, and multiple system atrophy); (2) tauopathies (progressive supranuclear palsy, corticobasal degeneration, FTLD-17; Guamian Parkinson-dementia; Pick's disease, and others); (3) polyglutamine disorders (Huntington's disease and related disorders); (4) pantothenate kinase-associated neurodegeneration; (5) Wilson's disease; and (6) other hereditary neurodegenerations without hitherto detected genetic or specific markers. The diversity of phenotypes is related to the deposition of pathologic proteins in distinct cell populations, causing neurodegeneration due to genetic and environmental factors, but there is frequent overlap between various disorders. Their etiopathogenesis is still poorly understood, but is suggested to result from an interaction between genetic and environmental factors. Multiple etiologies and noxious factors (protein mishandling, mitochondrial dysfunction, oxidative stress, excitotoxicity, energy failure, and chronic neuroinflammation) are more likely than a single factor. Current clinical consensus criteria have increased the diagnostic accuracy of most neurodegenerative movement disorders, but for their definite diagnosis, histopathological confirmation is required. We present a timely overview of the neuropathology and pathogenesis of the major extrapyramidal movement disorders in two parts, the first one dedicated to hypokinetic-rigid forms and the second to hyperkinetic disorders.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|