Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Innov Clin Neurosci ; 21(1-3): 52-60, 2024.
Article in English | MEDLINE | ID: mdl-38495603

ABSTRACT

The 1983 Orphan Drug Act in the United States (US) changed the landscape for development of therapeutics for rare or orphan diseases, which collectively affect approximately 300 million people worldwide, half of whom are children. The act has undoubtedly accelerated drug development for orphan diseases, with over 6,400 orphan drug applications submitted to the US Food and Drug Administration (FDA) from 1983 to 2023, including 350 drugs approved for over 420 indications. Drug development in this population is a global and collaborative endeavor. This position paper of the International Society for Central Nervous System Clinical Trials and Methodology (ISCTM) describes some potential best practices for the involvement of key stakeholder feedback in the drug development process. Stakeholders include advocacy groups, patients and caregivers with lived experience, public and private research institutions (including academia and pharmaceutical companies), treating clinicians, and funders (including the government and independent foundations). The authors articulate the challenges of drug development in orphan diseases and propose methods to address them. Challenges range from the poor understanding of disease history to development of endpoints, targets, and clinical trials designs, to finding solutions to competing research priorities by involved parties.

2.
J Clin Psychopharmacol ; 42(3): 234-237, 2022.
Article in English | MEDLINE | ID: mdl-35489028

ABSTRACT

PURPOSE/BACKGROUND: Drug trials of the central nervous system(CNS) have been plagued with uninformative failures, often because of the difficulties of knowing definitively whether dosing achieved was sufficient to modulate the intended CNS target at adequate concentrations to produce pharmacodynamic or dose-related changes in readouts of brain function. Key design elements can be introduced into early-stage trials to get at this issue. METHODS/PROCEDURES: This commentary builds on a review of earlier clinical studies in Fragile X syndrome to explore the extent to which the chain of evidence is in place to allow for interpretation of the results as ruling in or out the utility of modulating one or another molecular target to treat this disorder. Recent and current biomarker studies in Fragile X syndrome occurring subsequent to the clinical studies are reviewed to see if they might address any chain of evidence gaps. FINDINGS/RESULTS: Despite the strong preclinical basis for targeting molecular mechanisms, the lack of efficacy seen in clinical studies remains uninterpretable, with regard to ruling in or out the utility of targeting the mechanism in a clinical population, given the absence of studies, which address whether doses of administered drug impacted the targeted brain mechanism. IMPLICATIONS/CONCLUSIONS: The value of pursuing clinical studies of compounds targeted to novel mechanisms in the absence of clinical pharmacological evidence of some anticipated mediating pharmacokinetic/pharmacodynamic signals is questionable. One or more biomarkers of a drug effect on brain function are needed to establish dose dependent CNS effects that allow one to interpret clinical results as ruling in or out a mechanism and providing a firm basis for continuing or not, as well as informing dose selection in any clinical efficacy trials. Initiatives to address this general need in pediatric psychopharmacology are highlighted.


Subject(s)
Fragile X Syndrome , Brain , Central Nervous System , Child , Clinical Trials as Topic , Fragile X Syndrome/drug therapy , Humans
4.
Pharmaceut Med ; 34(4): 233-245, 2020 08.
Article in English | MEDLINE | ID: mdl-32705569

ABSTRACT

In 2012, the US National Institute of Mental Health launched three clinical trial contracts under a new FAST initiative. The overall goal for these contracts (Fast-Fail Trials) was to focus early-stage trials, testing novel pharmacologic agents that target the central nervous system, on pharmacologic-based designs to objectively identify doses that produce central nervous system effects. The three contracts targeted different psychiatric populations: psychotic (FAST-PS), mood and anxiety (FAST-MAS), and autism spectrum disorders (FAST-AS). The FAST initiative was a first attempt for the National Institute of Mental Health to adapt an experimental medicine approach to its clinical trial portfolio. As the Fast-Fail trials implemented this new approach for the field, we present the rationale for each trial, design considerations, results, and how each one contributed new knowledge to the field of psychopharmacology; important lessons for pharma and biotech. Under the FAST initiative, the National Institute of Mental Health assembled research teams with a broad range of expertise, who developed and validated the outcome measures and study protocol, and conducted multi-site clinical trials, testing candidate compounds. In the FAST-PS contract, the team validated an imaging-based pharmacodynamic biomarker of the effect of ketamine in the brain that could be utilized in subsequent clinical trials. The initial FAST-AS study was an important first step in the design of early-stage target-engagement trials in autism spectrum disorder, suggesting that a resting electroencephalogram can be used as a pharmacodynamic measure in future studies. The FAST-MAS study showed that blocking the kappa-opioid receptor significantly affects functional magnetic resonance imaging ventral striatal activation in the monetary incentive delay task in anticipation of gain. Together, the outcomes of the FAST-FAIL trials demonstrated the importance of rigorously designed and informative central nervous system trials, including the value of pharmacodynamic measures in early-stage trials. Use of these measures furthered our knowledge about the relationship between specific molecular mechanisms, brain effects, and therapeutic effects in patients with mental illnesses.


Subject(s)
Central Nervous System Agents/administration & dosage , Clinical Trials as Topic/methods , Mental Disorders/drug therapy , Central Nervous System Agents/pharmacology , Dose-Response Relationship, Drug , Drug Development , Humans , National Institute of Mental Health (U.S.) , Psychopharmacology , Research Design , United States
6.
Prog Neurobiol ; 152: 38-57, 2017 05.
Article in English | MEDLINE | ID: mdl-27216638

ABSTRACT

Many psychiatric and behavioral disorders manifest in childhood (attention deficit hyperactivity disorder, obsessive compulsive disorder, anxiety, depression, schizophrenia, autism spectrum disorder, etc.) and the opportunity for intervening early may attenuate full development of the disorder and lessen long term disability. Yet, pediatric drug approvals for CNS indications are limited, and pediatric testing generally occurs only after establishing adult efficacy, more as an afterthought rather than with the initial goal of developing the medication for a pediatric CNS indication. With pharmaceutical companies decreasing funding of their neuroscience research divisions overall, the prospects for moving promising investigational drugs forward into pediatrics will only decline. The goal of this review is to highlight important challenges around pediatric drug development for psychiatric disorders, specifically during clinical development, and to present opportunities for filling these gaps, using new strategies for de-risking investigational drugs in new clinical trial designs/models. We will first present the current trends in pediatric drug efficacy testing in academic research and in industry trials, we will then discuss the regulatory landscape of pediatric drug testing, including policies intended to support and encourage more testing. Obstacles that remain will then be presented, followed by new designs, funding opportunities and considerations for testing investigational drugs safely.


Subject(s)
Brain Diseases/drug therapy , Brain Diseases/psychology , Drug Discovery/trends , Mental Disorders/drug therapy , Mental Disorders/psychology , Pediatrics/trends , Psychotropic Drugs/administration & dosage , Child , Clinical Trials as Topic , Evidence-Based Medicine , Humans , Psychotropic Drugs/adverse effects , Treatment Outcome
8.
Neurobiol Dis ; 15(3): 520-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15056459

ABSTRACT

The relationship between intracellular Ca(2+) ([Ca(2+)](i)) regulation and programmed cell death is not well-defined; both increases and decreases in [Ca(2+)](i) have been observed in cells undergoing apoptosis. We determined [Ca(2+)](i) in cultured murine cortical neurons undergoing apoptosis after exposure to staurosporine or following oxygen-glucose deprivation in the presence of glutamate receptor antagonists. Neuronal [Ca(2+)](i) was decreased 1-4 h after exposure to staurosporine (30 nM). A [Ca(2+)](i) decrease was also observed 1 h after the end of the oxygen-glucose deprivation period when MK-801 and 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) were added to the bathing medium during the deprivation period. A similar decrease in [Ca(2+)](i) produced by reducing extracellular Ca(2+) or chelating intracellular Ca(2+) was sufficient to induce neuronal apoptosis. Raising [Ca(2+)](i) either by activating voltage-sensitive Ca(2+) channels with (-) Bay K8644 or by application of low concentrations of kainate attenuated both staurosporine and oxygen-glucose deprivation-induced apoptosis.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Neurons/metabolism , Animals , Apoptosis/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glucose/metabolism , Intracellular Fluid/chemistry , Neurons/drug effects , Oxygen/metabolism , Receptors, Glutamate/metabolism , Staurosporine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...