1
|
Kung TFC, Wilkinson CM, Dirks CA, Jickling GC, Colbourne F. Glibenclamide does not improve outcome following severe collagenase-induced intracerebral hemorrhage in rats. PLoS One 2021; 16:e0252584. [PMID: 34081746 PMCID: PMC8174736 DOI: 10.1371/journal.pone.0252584] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/18/2021] [Indexed: 11/26/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating insult with few effective treatments. Edema and raised intracranial pressure contribute to poor outcome after ICH. Glibenclamide blocks the sulfonylurea 1 transient receptor potential melastatin 4 (Sur1-Trpm4) channel implicated in edema formation. While glibenclamide has been found to improve outcome and reduce mortality in animal models of severe ischemic stroke, in ICH the effects are less clear. In our previous study, we found no benefit after a moderate-sized bleed, while others have reported benefit. Here we tested the hypothesis that glibenclamide may only be effective in severe ICH, where edema is an important contributor to outcome. Glibenclamide (10 μg/kg loading dose, 200 ng/h continuous infusion) was administered 2 hours post-ICH induced by collagenase injection into the striatum of adult rats. A survival period of 24 hours was maintained for experiments 1-3, and 72 hours for experiment 4. Glibenclamide did not affect hematoma volume (~81 μL) or other safety endpoints (e.g., glucose levels), suggesting the drug is safe. However, glibenclamide did not lessen striatal edema (~83% brain water content), ionic dyshomeostasis (Na+, K+), or functional impairment (e.g., neurological deficits (median = 10 out of 14), etc.) at 24 hours. It also did not affect edema at 72 h (~86% brain water content), or overall mortality rates (25% and 29.4% overall in vehicle vs. glibenclamide-treated severe strokes). Furthermore, glibenclamide appears to worsen cytotoxic edema in the peri-hematoma region (cell bodies were 46% larger at 24 h, p = 0.0017), but no effect on cell volume or density was noted elsewhere. Overall, these findings refute our hypothesis, as glibenclamide produced no favorable effects following severe ICH.
Collapse
Affiliation(s)
- Tiffany F. C. Kung
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Christine A. Dirks
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
2
|
Lapchak PA, Lara JM, Boitano PD. Cytoprotective Drug-Tissue Plasminogen Activator Protease Interaction Assays: Screening of Two Novel Cytoprotective Chromones. Transl Stroke Res 2017; 8:10.1007/s12975-017-0533-7. [PMID: 28405804 DOI: 10.1007/s12975-017-0533-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/21/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Tissue plasminogen activator (tPA) is currently used in combination with endovascular procedures to enhance recanalization and cerebral reperfusion and is also currently administered as standard-of-care thrombolytic therapy to patients within 3-4.5 h of an ischemic stroke. Since tPA is not neuroprotective or cytoprotective, adjuvant therapy with a neuroprotective or an optimized cytoprotective compound is required to provide the best care to stroke victims to maximally promote clinical recovery. In this article, we describe the use of a sensitive standardized protease assay with CH3SO2-D-hexahydrotyrosine-Gly-Arg-p-nitroanilide•AcOH, a chromogenic protease substrate that is cleaved to 4-nitroaniline (p-nitroaniline) and measured spectrophotometrically at 405 nm (OD405 nm), and how the assay can be used as an effective screening assay to study drug-tPA interactions. While we focus on two compounds of interest in our drug development pipeline, the assay is broadly applicable to all small molecule neuroprotective or cytoprotective compounds currently being discovered and developed worldwide. In this present study, we found that the specific tPA inhibitor, plasminogen activator inhibitor-1 (PAI-1; 0.25 μM), significantly (p < 0.0001) inhibited 4-nitroaniline release, by 97.74% during the 10-min duration of the assay, which is indicative of tPA protease inhibition. In addition, two lead chromone cytoprotective candidates, 2-(3',4',5'-trihydroxyphenyl)chromen-4-one (3',4',5'-trihydroxyflavone) (CSMC-19) and 3-hydroxy-2-[3-hydroxy-4-(pyrrolidin-1-yl)phenyl]benzo[h]chromen-4-one (CSMC-140), also significantly (p < 0.05) reduced 4-nitroaniline accumulation, but to a lesser extent. The reduction was 68 and 45%, respectively, at 10 μM, and extrapolated IC50 values were 4.37 and >10 μM for CSMC-19 and CSMC-140, respectively. Using bonafide 4-nitroaniline, we then demonstrated that the reduction of 4-nitroaniline detection was not due to drug-4-nitroaniline quenching of signal detection at OD405 nm. In conclusion, the results suggest that high concentrations of both cytoprotectives reduced 4-nitroaniline production in vitro, but the inhibition only occurs with concentrations 104-1025-fold that of EC50 values in an efficacy assay. Thus, CSMC-19 and CSMC-140 should be further developed and evaluated in embolic stroke models in the absence or presence of a thrombolytic. If necessary, they could be administered once effective tPA thrombolysis has been confirmed to avoid the possibility that the chromone will reduce the efficacy of tPA in patients. Stroke investigator developing new cytoprotective small molecules should consider adding this sensitive assay to their development and screening repertoire to assess possible drug-tPA interactions in vitro as a de-risking step.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neurology, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Suite 8318, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.
- Department of Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Suite 8318, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.
| | - Jacqueline M Lara
- Department of Neurology, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Suite 8318, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| | - Paul D Boitano
- Department of Neurology, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Suite 8318, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| |
Collapse
|
3
|
Translational Stroke Research Opportunities and a Strategy to Develop Effective Cytoprotection. Transl Stroke Res 2017; 8:318-321. [DOI: 10.1007/s12975-017-0529-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 12/21/2022]
|
4
|
Mandava P, Shah SD, Sarma AK, Kent TA. An Outcome Model for Intravenous rt-PA in Acute Ischemic Stroke. Transl Stroke Res 2015; 6:451-7. [PMID: 26385545 DOI: 10.1007/s12975-015-0427-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 09/04/2015] [Indexed: 01/19/2023]
Abstract
Most early phase trials in stroke and brain trauma have failed in phase 3, including efforts to improve acute ischemic stroke outcomes beyond that achieved by intravenous recombinant tissue plasminogen activator (t-PA) (IVT). With the exception of more recent stent retriever trials, most subsequent phase 3 trials failed. We previously showed that baseline imbalances, non-linear relationships of these factors to outcome, and unrepresentative control populations invalidate traditional statistical analysis in early trials of heterogeneous diseases such as stroke. We developed an alternative approach using a pooled outcome model derived from control arms of randomized clinical trial (RCTs). This model then permits comparing treatment trials to an expected outcome of a pooled population. Here, we hypothesized we could develop such a model for IVT and tested it against outcomes without IVT. We surveyed literature for all trials involving one arm with IVT reporting baseline National Institute Stroke Scale (NIHSS), age, and outcome. A non-linear fit was performed including multi-dimensional statistical intervals (±95 %) permitting visual comparison of outcomes at their own baselines. We compared models derived from non-IVT control arms. Models from 24 IVT RCTs representing 3195 subjects were successfully generated for functional outcome, modified Rankin Scale (mRS) 0-2 (r(2) = 0. 83, p < 0.001), and mortality (r(2) = 0.54; p = 0.001). We confirmed better outcomes compared to no IVT and mixed use IVT models across the range of baseline factors. It was possible to generate an expected outcome model for IVT from existing literature. We confirmed benefit compared to placebo. This model should be useful to compare to new agents without the need for statistical manipulation.
Collapse
Affiliation(s)
- Pitchaiah Mandava
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd (127), Houston, TX, 77030, USA.
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA.
| | - Shreyansh D Shah
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Anand K Sarma
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Thomas A Kent
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd (127), Houston, TX, 77030, USA
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Lapchak PA, Daley JT, Boitano PD. A blinded, randomized study of L-arginine in small clot embolized rabbits. Exp Neurol 2015; 266:143-6. [PMID: 25708986 PMCID: PMC4382386 DOI: 10.1016/j.expneurol.2015.02.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 02/05/2015] [Accepted: 02/11/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Tissue plasminogen activator (tPA) is administered to acute ischemic stroke victims in a vehicle formulation containing high concentrations of L-arginine (3.5g/100mg vial), a well-known nitric oxide synthase (NOS) substrate and precursor to nitric oxide (NO), as well as an enhancer of cerebral blood flow. METHODS We studied the effects of tPA vehicle compared to tPA (3.3mg/kg) formulated in the same vehicle containing L-arginine, normal saline or normal saline containing L-arginine, on behavioral function following small clot embolic strokes in rabbits using clinical rating scores and quantal analysis curves as the primary end point. Treatments were administered intravenously (1ml/kg; 20% bolus/80% infused over 30min) starting 1h following the injection of small-sized blood clots into the brain vasculature and terminal behavior was measured 2days following embolization. Behavioral rating scores were used to calculate the effective stroke dose (P50 in mg) that produces neurological deficits in 50% of the rabbits. RESULTS In this study, tPA significantly (p=0.001) improved behavior compared to all other treatments including tPA vehicle, saline and saline-L-arginine, increasing the P50 by 141% over tPA vehicle. Saline-L-arginine was not significantly different from either saline or tPA vehicle (p>0.05). CONCLUSION This study demonstrates that the L-arginine component of the tPA vehicle does not contribute to the reproducible clinical improvement observed following tPA administration in rabbits. Moreover, the administration of L-arginine was not an effective method to promote behavioral recovery following embolic strokes in the stringent rabbit small clot stroke model, nor did L-arginine exacerbate behavioral deficits or intracerebral hemorrhage in embolized rabbits.
Collapse
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Department of Neurology & Neurosurgery.
| | | | | |
Collapse
|
6
|
Wowk S, Ma Y, Colbourne F. Mild Therapeutic Hypothermia Does Not Reduce Thrombin-Induced Brain Injury. Ther Hypothermia Temp Manag 2014; 4:180-7. [DOI: 10.1089/ther.2014.0014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shannon Wowk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Yonglie Ma
- Department of Psychology, University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Psychology, University of Alberta, Edmonton, Canada
| |
Collapse
|
7
|
Lapchak PA, Boitano PD. Effect of the Pleiotropic Drug CNB-001 on Tissue Plasminogen Activator (tPA) Protease Activity in vitro: Support for Combination Therapy to Treat Acute Ischemic Stroke. JOURNAL OF NEUROLOGY & NEUROPHYSIOLOGY 2014; 5:214. [PMID: 25364620 PMCID: PMC4215513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Current state-of-the-art acute ischemic stroke clinical trials are designed to study neuroprotectants when administered following thrombolysis; tissue plasminogen activator (tPA) is administered to patients within 3-4.5 hours of an ischemic event. Thus, in order to develop a novel neuroprotectant and move it forward to a clinical trial, it is important to assess the effects of the drug on tPA's proteolytic activity in vitro, prior to extensive in vivo analysis. In this study, we determined if CNB-001 [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl)vinyl)-2-methoxy-phenol)], would affect, either enhance or inhibit tPA activity in vitro. In this tPA-inhibitor (plasminogen activator inhibitor-1; PAI-1 and 2,7-Bis-(4-Amidinobenzylidene)-Cycloheptan-1-One Dihydrochloride; tPA stop) controlled study, we used a chromogenic substrate (CH3SO2-D-hexahydrotyrosine-Gly-Arg-p-nitroanilide•AcOH) to study drug interactions in vitro, spectrophotometrically measuring protease released p-Nitroaniline from the substrate. We found that PAI-1 (0.25 μM) and tPA stop (5 μM) significantly (p<0.0001) inhibited substrate release, by 98.6% and 83.4%, respectively, thus inhibiting tPA activity in vitro. In comparison, CNB-001 (0.7-7 μM) reduced tPA activity by 28-32%, with an extrapolated IC50 value of 65.2-704 μM. Thus, although high concentrations of CNB-001 does affects tPA activity in vitro, the study supports the use of CNB-001 in combination with tPA to treat stroke, However, CNB-001 should be administered following thrombolysis to promote neuroprotection and repair.
Collapse
Affiliation(s)
- Paul A Lapchak
- Departments of Neurology Advanced Health Sciences Pavilion, Los Angeles, USA,Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Los Angeles, USA,Corresponding author: Paul A Lapchak, Director of Translational Research, Cedars-Sinai Medical Center Professor, Department of Neurology & Neurosurgery, Advanced Health Sciences Pavilion, Suite 8305, 127 S. San Vicente Blvd, Los Angeles, CA 90048, USA; Tel: 310-248-8188; Fax: 310-248-7568
| | - Paul D Boitano
- Departments of Neurology Advanced Health Sciences Pavilion, Los Angeles, USA
| |
Collapse
|
8
|
Lapchak PA, Schubert DR, Maher PA. De-Risking of Stilbazulenyl Nitrone (STAZN), a Lipophilic Nitrone to Treat Stroke Using a Unique Panel of In Vitro Assays. Transl Stroke Res 2013; 2:209-17. [PMID: 22003372 DOI: 10.1007/s12975-011-0071-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the present study, we used a comprehensive panel of in vitro assays to evaluate the efficacy and safety of stilbazulenyl nitrone (STAZN) as a lead compound to treat acute ischemic stroke. First, we measured neuroprotection in vitro using two different HT22 hippocampal nerve cell assays. Secondly, to de-risk drug development, we used CeeTox analysis with the H4IIE rat hepatoma cell line to determine the acute toxicity profile of STAZN. Third, STAZN was tested in microsomes from four species for measures of metabolic stability. Last, we determined the Ames test genotoxicity profile of STAZN using Salmonella typhimurium TA989 and TA100. In vitro, STAZN was neuroprotective against toxicity induced by iodoacetic acid, and oxytosis-induced glutathione depletion was initiated by glutamate, with an EC(50) value of 1-5 μM. Secondly, using CeeTox analysis, the estimated C(Tox) value (i.e., sustained concentration expected to produce toxicity in a rat 14-day repeat dose study) for STAZN was calculated to be 260 μM. Third, the half-life of STAZN in humans, dogs, and rats was 60-78 min. Last, the genotoxicity profile showed that STAZN did not induce bacterial colony growth under any conditions tested, indicating the lack of mutagenicity with this compound. STAZN appears to be a multi-target neuroprotective compound that has an excellent safety profile in both the CeeTox and Ames mutagenicity assays. STAZN may have significant potential as a novel neuroprotective agent to treat stroke and should be pursued in clinically relevant embolic stroke models.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neurology, Cedars-Sinai Medical Center, Davis Research Building, D-2091, 110 N. George Burns Road, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
9
|
Lapchak PA. Fast neuroprotection (fast-NPRX) for acute ischemic stroke victims: the time for treatment is now. Transl Stroke Res 2013; 4:704-9. [PMID: 24323424 DOI: 10.1007/s12975-013-0303-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 01/25/2023]
Affiliation(s)
- Paul A Lapchak
- Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Rm 8305, 127 S. San Vicente Blvd, Los Angeles, CA, 90048, USA,
| |
Collapse
|
10
|
Bipyridine, an iron chelator, does not lessen intracerebral iron-induced damage or improve outcome after intracerebral hemorrhagic stroke in rats. Transl Stroke Res 2013; 4:719-28. [PMID: 24323426 DOI: 10.1007/s12975-013-0272-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 07/18/2013] [Accepted: 07/19/2013] [Indexed: 10/26/2022]
Abstract
Iron chelators, such as the intracellular ferrous chelator 2,2'-bipyridine, are a potential means of ameliorating iron-induced injury after intracerebral hemorrhage (ICH). We evaluated bipyridine against the collagenase and whole-blood ICH models and a simplified model of iron-induced damage involving a striatal injection of FeCl2 in adult rats. First, we assessed whether bipyridine (25 mg/kg beginning 12 h post-ICH and every 12 h for 3 days) would attenuate non-heme iron levels in the brain and lessen behavioral impairments (neurological deficit scale, corner turn test, and horizontal ladder) 7 days after collagenase-induced ICH. Second, we evaluated bipyridine (20 mg/kg beginning 6 h post-ICH and then every 24 h) on edema 3 days after collagenase infusion. Body temperature was continually recorded in a subset of these rats beginning 24 h prior to ICH until euthanasia. Third, bipyridine was administered (as per experiment 2) after whole-blood infusion to examine tissue loss, neuronal degeneration, and behavioral impairments at 7 days post-stroke, as well as body temperature for 3 days post-stroke. Finally, we evaluated whether bipyridine (25 mg/kg given 2 h prior to surgery and then every 12 h for 3 days) lessens tissue loss, neuronal death, and behavioral deficits after striatal FeCl2 injection. Bipyridine caused a significant hypothermic effect (maximum drop to 34.6 °C for 2-5 h after each injection) in both ICH models; however, in all experiments bipyridine-treated rats were indistinguishable from vehicle controls on all other measures (e.g., tissue loss, behavioral impairments, etc.). These results do not support the use of bipyridine against ICH.
Collapse
|
11
|
Lapchak PA. Drug-like property profiling of novel neuroprotective compounds to treat acute ischemic stroke: guidelines to develop pleiotropic molecules. Transl Stroke Res 2013; 4:328-42. [PMID: 23687519 PMCID: PMC3653324 DOI: 10.1007/s12975-012-0200-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The development of novel neuroprotective compounds to treat acute ischemic stroke (AIS) has been problematic and quite complicated, since many candidates that have been tested clinically lacked significant pleiotropic activity, were unable to effectively cross the blood brain barrier (BBB), had poor bioavailability or were toxic. Moreover, the compounds did not confer significant neuroprotection or clinical efficacy measured using standard behavioral endpoints, when studied in clinical trials in a heterogeneous population of stroke patients. To circumvent some of the drug development problems describe above, we have used a rational funnel approach to identify and develop promising candidates. Using a step-wise approach, we have identified a series of compounds based upon two different neuroprotection assays. We have then taken the candidates and determined their "drug-like" properties. This guidelines article details in vitro screening assays used to show pleiotropic activity of a series of novel compounds; including enhanced neuroprotective activity compared to the parent compound fisetin. Moreover, for preliminary drug de-risking or risk reduction during development, we used compound assessment in the CeeTox assay, ADME toxicity using the AMES test for genotoxicity and interaction with Cytochrome P450 using CYP450 inhibition analysis against a spectrum of CYP450 enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4) as a measure of drug interaction. Moreover, the compounds have been studied using a transfected Madin Darby canine kidney (MDCK) cell assay to assess blood brain barrier penetration (BBB). Using this series of assays, we have identified 4 novel molecules to be developed as an AIS treatment.
Collapse
Affiliation(s)
- Paul A Lapchak
- Director of Translational Research, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, D-2091, 110 N. George Burns Road, Los Angeles, CA 90048 USA
| |
Collapse
|
12
|
Lapchak PA. Recommendations and practices to optimize stroke therapy: developing effective translational research programs. Stroke 2013; 44:841-3. [PMID: 23391768 DOI: 10.1161/strokeaha.112.680439] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
13
|
Current world literature. Curr Opin Anaesthesiol 2012; 25:743-8. [PMID: 23147670 DOI: 10.1097/aco.0b013e32835b8a43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
A Clinically Relevant Rabbit Embolic Stroke Model for Acute Ischemic Stroke Therapy Development: Mechanisms and Targets. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Lapchak PA, Wu Q. Vascular Dysfunction in Brain Hemorrhage: Translational Pathways to Developing New Treatments from Old Targets. JOURNAL OF NEUROLOGY & NEUROPHYSIOLOGY 2011; 2011:S1-e001. [PMID: 22400125 PMCID: PMC3293216 DOI: 10.4172/2155-9562.s1-e001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hemorrhagic stroke which is a form of stroke that affects 20% of all stroke patients is a devastating condition for which new treatments must be developed. Current treatment methods are quite insufficient to reduce long term morbidity and high mortality rate, up to 50%, associated with bleeding into critical brain structures, into ventricular spaces and within the subarachnoid space. During the last 10-15 years, significant advances in the understanding of important mechanisms that contribute to cell death and clinical deficits have been made. The most important observations revolve around a key set of basic mechanisms that are altered in brain bleeding models, including activation of membrane metalloproteinases, oxidative stress and both inflammatory and coagulation pathways. Moreover, it is now becoming apparent that brain hemorrhage can activate the ischemic stroke cascade in neurons, glial cells and the vascular compartment. The activation of multiple pathways allows comes the opportunity to intervene pharmacologically using monotherapy or combination therapy. Ultimately, combination therapy or pleiotropic compounds with multi-target activities should prove to be more efficacious than any single therapy alone. This article provides a comprehensive look at possible targets for small molecule intervention as well as some new approaches that result in metabolic down-regulation or inhibition of multiple pathways simultaneously.
Collapse
Affiliation(s)
- Paul A. Lapchak
- Director of Translational Research, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, D- 2091, 110 N, George Burns Road, Los Angeles, CA 90048 USA
| | - Qiang Wu
- Project Scientist, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, D-2094E, 110 N. George Burns Road, Los Angeles, CA 90048 USA
| |
Collapse
|