scholarly journals Predict drug permeability to blood–brain-barrier from clinical phenotypes: drug side effects and drug indications

2016 ◽  
pp. btw713 ◽  
Author(s):  
Zhen Gao ◽  
Yang Chen ◽  
Xiaoshu Cai ◽  
Rong Xu
2020 ◽  
Vol 26 (37) ◽  
pp. 4721-4737 ◽  
Author(s):  
Bhumika Kumar ◽  
Mukesh Pandey ◽  
Faheem H. Pottoo ◽  
Faizana Fayaz ◽  
Anjali Sharma ◽  
...  

Parkinson’s disease is one of the most severe progressive neurodegenerative disorders, having a mortifying effect on the health of millions of people around the globe. The neural cells producing dopamine in the substantia nigra of the brain die out. This leads to symptoms like hypokinesia, rigidity, bradykinesia, and rest tremor. Parkinsonism cannot be cured, but the symptoms can be reduced with the intervention of medicinal drugs, surgical treatments, and physical therapies. Delivering drugs to the brain for treating Parkinson’s disease is very challenging. The blood-brain barrier acts as a highly selective semi-permeable barrier, which refrains the drug from reaching the brain. Conventional drug delivery systems used for Parkinson’s disease do not readily cross the blood barrier and further lead to several side-effects. Recent advancements in drug delivery technologies have facilitated drug delivery to the brain without flooding the bloodstream and by directly targeting the neurons. In the era of Nanotherapeutics, liposomes are an efficient drug delivery option for brain targeting. Liposomes facilitate the passage of drugs across the blood-brain barrier, enhances the efficacy of the drugs, and minimize the side effects related to it. The review aims at providing a broad updated view of the liposomes, which can be used for targeting Parkinson’s disease.


Pharmaceutics ◽  
2020 ◽  
Vol 12 (2) ◽  
pp. 138 ◽  
Author(s):  
Paolo Giunchedi ◽  
Elisabetta Gavini ◽  
Maria Cristina Bonferoni

Nose-to-brain delivery represents a big challenge. In fact there is a large number of neurological diseases that require therapies in which the drug must reach the brain, avoiding the difficulties due to the blood–brain barrier (BBB) and the problems connected with systemic administration, such as drug bioavailability and side-effects. For these reasons the development of nasal formulations able to deliver the drug directly into the brain is of increasing importance. This Editorial regards the contributions present in the Special Issue “Nose-to-Brain Delivery”.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5012-5012
Author(s):  
Catherine Gordon ◽  
Alan Nolan ◽  
Wanda Dorsett-Martin ◽  
Gail C. Megason ◽  
Laree Hiser

Abstract Abstract 5012 Background: Current combination chemotherapy regimens are highly effective for treating acute lymphoblastic leukemia (ALL). Unfortunately, vincristine, a component used in current regimens, generally causes significant neuropathy. The neuropathy is usually reversible, but it can have a negative effect on the treatment of ALL by delaying therapy or requiring reduced drug doses. The focus of this research is on exploring pharmaceutical solutions for minimizing the neurotoxicity of vincristine. We selected noscapine as a good candidate for use with vincristine. Noscapine, an FDA-approved antitussive, has no known significant side effects and good oral bioavailability, making it relatively easy to administer to children. Noscapine also demonstrates anticancer properties that are currently being studied, with encouraging preliminary results, in a variety of solid and hematological malignancies. Noscapine has been shown to protect against vincristine-induced demyelination of axons in vitro. In addition, vincristine and noscapine are synergistic in their antiproliferative effects in two leukemic cell lines. While noscapine has been used as a single anticancer agent, no preclinical or clinical studies examining it in combination with vincristine have been reported. Objectives: Methods: Animal studies: C57BL/6 female mice were used to test the safety and efficacy of noscapine as a protective agent against vincristine-induced neuropathy. Behavioral assays (tail-flick and rotarod) were used to assess neuropathy. Control groups receiving only saline, vincristine, or noscapine were compared to groups of mice receiving a combination of vincristine and noscapine. Independent experiments were performed that varied in drug dose and delivery. Vincristine was given as an intraperitoneal injection twice a week with doses varying from 0.1 to 1.5 mg/kg. Noscapine was administered along with vincristine by injection or ad libitum in the drinking water. Data regarding neuropathy and other observable side effects, including weight loss, were collected. Brain and peripheral organs were collected from euthanized mice, quick-frozen in liquid nitrogen and stored at −80 degrees C. Mass spectroscopy experiments: Tissues were homogenized, extracted with methanol and separated using Isolute C2EC SPC columns. Columns were conditioned with methanol, loaded and washed with 5% methanol and eluted with 100% methanol. Separation with liquid chromatography was followed by electrospray ionization and MS/MS analysis using a triple quadrupole-linear ion trap mass spectrometer. Results: The combination of vincristine and noscapine resulted in significant toxic effects including ataxia, seizures, and death in our mouse model. Side effects were not observed in mice given only noscapine. Mice given only vincristine had expected neuropathy as measured by the tail-flick and rotarod assays, but no other significant adverse effects. Noscapine had some protective effect against vincristine-induced neuropathy, but the effect was not statistically significant. Given the adverse neurologic reactions seen, it was hypothesized that noscapine permitted vincristine to cross the blood-brain barrier. Previous studies have shown that noscapine freely crosses the blood-brain barrier, and vincristine is fatal in humans when given intrathecally. Our hypothesis was not supported by analysis of whole brains collected one hour after intraperitoneal injection of vincristine and noscapine (or controls). Mass spectroscopy results showed a decreased uptake of noscapine into brain and peripheral organs in the mice that received noscapine and vincristine in combination. Accumulation of vincristine was unchanged. Conclusion: Noscapine has a significant drug-drug interaction with vincristine in mice. The exact mechanism responsible for this interaction remains undetermined, but several hypotheses including effects on transport mechanisms are currently being investigated. Mass spectroscopy data suggests that vincristine alters the pharmacokinetic or pharmacodynamic properties of noscapine. This may limit the use of noscapine as a neuroprotective agent. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Krishnapriya Madhu Varier ◽  
Sumathi Thangarajan ◽  
Arulvasu Chinnasamy ◽  
Gopalsamy Balakrishnan ◽  
Renjith Paulose

<p><strong>Objective: </strong>Parkinson’s disease (PD) is a leading cause of mental disability and death worldwide. Even though there are many advances in drug development against PD, a potent low dosage drug with fewer side effects are still in its nursery. This is a pioneer <em>in silico</em> attempt to test the anti-PD actions of esculin and hinokitol to act novel drugs.</p><p><strong>Methods: </strong>In this study, using Auto dock tools 4.2, esculin and hinokitol (β-Thujaplicin) were predicted for its inhibitory actions with Alpha-Synuclein (AS) Apo site, Dopamine D3 Receptor (D3R), Glycogen Synthase Kinase-3 Beta (GSK3β), Mono Oxidase B (MAO-B), Parkin and Tyrosine 3-Hydroxylase (TH) with levodopa standard. The reliability of the 3D predicted model of these proteins were analysed using RAMPAGE. Further, the blood-brain barrier (BBB) crossing ability of the natural compounds were analysed using cbligand. The <em>In silico </em>ADME (Absorption, Distribution, Metabolism, Excretion) properties of esculin and hinokitol were compared with that of levodopa using molinspiration and admetSAR @ LMMD software.<strong></strong></p><p><strong>Results: </strong>The predictions were that hinokitol, being blood-brain barrier positive (BBB+) with fewer side effects could be a potent anti-PD drug than esculin as it proved to be blood-brain barrier negative (BBB-). Hinokitol was predicted to be good inhibitors of AS, MAO-B and Parkin.</p><p><strong>Conclusion: </strong>The study revealed that hinokitol could be a potent anti-PD drug, being BBB+. Hinokitol was additionally predicted as a good inhibitor of AS, MAO-B and Parkin than levodopa standard.</p><p> </p>


Sign in / Sign up

Export Citation Format

Share Document