1
|
Filling the gaps on stroke research: Focus on inflammation and immunity. Brain Behav Immun 2021; 91:649-667. [PMID: 33017613 PMCID: PMC7531595 DOI: 10.1016/j.bbi.2020.09.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
For the last two decades, researchers have placed hopes in a new era in which a combination of reperfusion and neuroprotection would revolutionize the treatment of stroke. Nevertheless, despite the thousands of papers available in the literature showing positive results in preclinical stroke models, randomized clinical trials have failed to show efficacy. It seems clear now that the existing data obtained in preclinical research have depicted an incomplete picture of stroke pathophysiology. In order to ameliorate bench-to-bed translation, in this review we first describe the main actors on stroke inflammatory and immune responses based on the available preclinical data, highlighting the fact that the link between leukocyte infiltration, lesion volume and neurological outcome remains unclear. We then describe what is known on neuroinflammation and immune responses in stroke patients, and summarize the results of the clinical trials on immunomodulatory drugs. In order to understand the gap between clinical trials and preclinical results on stroke, we discuss in detail the experimental results that served as the basis for the summarized clinical trials on immunomodulatory drugs, focusing on (i) experimental stroke models, (ii) the timing and selection of outcome measuring, (iii) alternative entry routes for leukocytes into the ischemic region, and (iv) factors affecting stroke outcome such as gender differences, ageing, comorbidities like hypertension and diabetes, obesity, tobacco, alcohol consumption and previous infections like Covid-19. We can do better for stroke treatment, especially when targeting inflammation following stroke. We need to re-think the design of stroke experimental setups, notably by (i) using clinically relevant models of stroke, (ii) including both radiological and neurological outcomes, (iii) performing long-term follow-up studies, (iv) conducting large-scale preclinical stroke trials, and (v) including stroke comorbidities in preclinical research.
Collapse
|
2
|
Bath PM, Appleton JP, England T. The Hazard of Negative (Not Neutral) Trials on Treatment of Acute Stroke: A Review. JAMA Neurol 2020; 77:114-124. [PMID: 31790551 DOI: 10.1001/jamaneurol.2019.4107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Importance While there are a limited number of beneficial treatments for acute stroke (eg, stroke units, reperfusion, aspirin, hemicraniectomy), there are more negative (as opposed to neutral) interventions spanning multiple different mechanisms of action. To reduce the risk of future negative studies, it is vital to understand why previous interventions appeared to cause harm. Observations The limited number of beneficial treatments for acute ischemic stroke are far outnumbered by negative (not neutral) interventions that worsened outcomes in randomized clinical trials (RCTs), including those with putative neuroprotectant, anticoagulant, anti-inflammatory, free radical-scavenging, hemorrhagic, or vasoactive activity. Other agents reduced thrombolytic efficiency or exhibited neuropsychiatric or cardiac toxicity. In intracerebral hemorrhage, platelet transfusion was hazardous. Although reperfusion treatments should be given as soon as possible, very early intervention with other strategies may instead be hazardous, as has been seen with physical therapy and vasodepressors. Conclusions and Relevance The lessons learned from negative stroke RCTs are vital for designing future studies. Multicenter preclinical studies are necessary, and animals that die must be included in analyses. Randomized clinical trials must assess multiple neurological, vascular, cardiac, and general safety effects, whether these are on target or off target. All preclinical trials and RCTs must be published in full. Learning from the past will help to reduce the number of negative stroke RCTs in the future.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, England.,Stroke, Nottingham University Hospitals NHS Trust, Nottingham, England
| | - Jason P Appleton
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, England.,Stroke, Nottingham University Hospitals NHS Trust, Nottingham, England
| | - Timothy England
- Vascular Medicine, Division of Medical Sciences and Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, England
| |
Collapse
|
3
|
Ma J, Ma Y, Shuaib A, Winship IR. Improved collateral flow and reduced damage after remote ischemic perconditioning during distal middle cerebral artery occlusion in aged rats. Sci Rep 2020; 10:12392. [PMID: 32709950 PMCID: PMC7381676 DOI: 10.1038/s41598-020-69122-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/07/2020] [Indexed: 02/05/2023] Open
Abstract
Circulation through cerebral collaterals can maintain tissue viability until reperfusion is achieved. However, collateral circulation is time limited, and failure of collaterals is accelerated in the aged. Remote ischemic perconditioning (RIPerC), which involves inducing a series of repetitive, transient peripheral cycles of ischemia and reperfusion at a site remote to the brain during cerebral ischemia, may be neuroprotective and can prevent collateral failure in young adult rats. Here, we demonstrate the efficacy of RIPerC to improve blood flow through collaterals in aged (16-18 months of age) Sprague Dawley rats during a distal middle cerebral artery occlusion. Laser speckle contrast imaging and two-photon laser scanning microscopy were used to directly measure flow through collateral connections to ischemic tissue. Consistent with studies in young adult rats, RIPerC enhanced collateral flow by preventing the stroke-induced narrowing of pial arterioles during ischemia. This improved flow was associated with reduced early ischemic damage in RIPerC treated aged rats relative to controls. Thus, RIPerC is an easily administered, non-invasive neuroprotective strategy that can improve penumbral blood flow via collaterals. Enhanced collateral flow supports further investigation as an adjuvant therapy to recanalization therapy and a protective treatment to maintain tissue viability prior to reperfusion.
Collapse
Affiliation(s)
- Junqiang Ma
- Neurochemical Research Unit, Department of Psychiatry, 12-127 Clinical Sciences Building, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yonglie Ma
- Neurochemical Research Unit, Department of Psychiatry, 12-127 Clinical Sciences Building, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Ashfaq Shuaib
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neurochemical Research Unit, Department of Psychiatry, 12-127 Clinical Sciences Building, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Shibeko AM, Chopard B, Hoekstra AG, Panteleev MA. Redistribution of TPA Fluxes in the Presence of PAI-1 Regulates Spatial Thrombolysis. Biophys J 2020; 119:638-651. [PMID: 32653051 DOI: 10.1016/j.bpj.2020.06.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022] Open
Abstract
The fibrin clot is gelatinous matter formed upon injury to stop blood loss and is later destroyed by fibrinolysis, an enzymatic cascade with feedback. Pharmacological fibrinolysis stimulation is also used to destroy pathological, life-threatening clots and thrombi (thrombolysis). The regulation of the nonlinear spatially nonuniform fibrinolytic process in thrombolysis is not currently well understood. We developed a reaction-diffusion-advection model of thrombolysis by tissue plasminogen activator (TPA) in an occluded vessel with a pressure gradient. Sensitivity-analysis-based model reduction was used to reveal the critical processes controlling different steps of thrombolysis. The propagation of thrombolysis in the system without flow was predominantly controlled by TPA diffusion, whereas transport of other active components was rendered nonessential either by their high fibrin-binding parameters and short lifetimes or their initial uniform distribution. The concentration of the main TPA inhibitor plasminogen activator inhibitor 1 (PAI-1) controlled both the extent of lysis propagation and the shape of fibrin spatial distribution during lysis. Interestingly, PAI-1 remained important even when its concentration was an order of magnitude below that of TPA because of its role at the edge of the diffusing TPA front. The system was robust to reaction rate constant perturbations. Using these data, a reduced model of thrombolysis was proposed. In the presence of flow, convection of TPA was the critical controlling process; although the role of PAI-1 concentration was much less in the presence of flow, its influence became greater in the presence of collateral bypassing vessels, which sufficiently reduced TPA flux through the thrombus. Flow bypass through the collateral vessel caused a decrease in TPA flux in the clotted vessel, which increased the PAI-1/TPA ratio, thus making PAI-1-induced inhibition relevant for the regulation of spatial lysis up to its arrest.
Collapse
Affiliation(s)
- Alexey M Shibeko
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Bastien Chopard
- Computer Science Department, University of Geneva, Carouge, Switzerland
| | - Alfons G Hoekstra
- Computational Science Lab, Institute for Informatics, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia.
| |
Collapse
|
5
|
Kaiser EE, West FD. Large animal ischemic stroke models: replicating human stroke pathophysiology. Neural Regen Res 2020; 15:1377-1387. [PMID: 31997796 PMCID: PMC7059570 DOI: 10.4103/1673-5374.274324] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The high morbidity and mortality rate of ischemic stroke in humans has led to the development of numerous animal models that replicate human stroke to further understand the underlying pathophysiology and to explore potential therapeutic interventions. Although promising therapeutics have been identified using these animal models, with most undergoing significant testing in rodent models, the vast majority of these interventions have failed in human clinical trials. This failure of preclinical translation highlights the critical need for better therapeutic assessment in more clinically relevant ischemic stroke animal models. Large animal models such as non-human primates, sheep, pigs, and dogs are likely more predictive of human responses and outcomes due to brain anatomy and physiology that are more similar to humans-potentially making large animal testing a key step in the stroke therapy translational pipeline. The objective of this review is to highlight key characteristics that potentially make these gyrencephalic, large animal ischemic stroke models more predictive by comparing pathophysiological responses, tissue-level changes, and model limitations.
Collapse
Affiliation(s)
- Erin E Kaiser
- Regenerative Bioscience Center; Neuroscience Program, Biomedical and Health Sciences Institute; Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Franklin D West
- Regenerative Bioscience Center; Neuroscience Program, Biomedical and Health Sciences Institute; Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, USA
| |
Collapse
|
6
|
Abstract
Novel therapeutic intervention that aims to enhance the endogenous recovery potential of the brain during the subacute phase of stroke has produced promising results. The paradigm shift in treatment approaches presents new challenges to preclinical and clinical researchers alike, especially in the functional endpoints domain. Shortcomings of the "neuroprotection" era of stroke research are yet to be fully addressed. Proportional recovery observed in clinics, and potentially in animal models, requires a thorough reevaluation of the methods used to assess recovery. To this end, this review aims to give a detailed evaluation of functional outcome measures used in clinics and preclinical studies. Impairments observed in clinics and animal models will be discussed from a functional testing perspective. Approaches needed to bridge the gap between clinical and preclinical research, along with potential means to measure the moving target recovery, will be discussed. Concepts such as true recovery of function and compensation and methods that are suitable for distinguishing the two are examined. Often-neglected outcomes of stroke, such as emotional disturbances, are discussed to draw attention to the need for further research in this area.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Burke Neurological Research Institute, White Plains, NY, USA
| | - Sunghee Cho
- Burke Neurological Research Institute, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Neurological Research Institute, White Plains, NY, USA
| |
Collapse
|
7
|
Lapchak PA, Boitano PD, Bombien R, Chou D, Knight M, Muehle A, Winkel MT, Khoynezhad A. CNB-001 reduces paraplegia in rabbits following spinal cord ischemia. Neural Regen Res 2019; 14:2192-2198. [PMID: 31397359 PMCID: PMC6788235 DOI: 10.4103/1673-5374.262598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Spinal cord ischemia associated with trauma and surgical procedures including thoraco-abdominal aortic aneurysm repair and thoracic endovascular aortic repair results in devastating clinical deficits in patients. Because spinal cord ischemia is inadequately treated, we studied the effects of [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl) vinyl)-2-methoxy-phenol)] (CNB-001), a novel curcumin-based compound, in a rabbit SCI model. CNB-001 is known to inhibit human 5-lipoxygenase and 15-lipoxygenase and reduce the ischemia-induced inflammatory response. Moreover, CNB-001 can reduce the level of oxidative stress markers and potentiate brain-derived neurotrophic factor and brain-derived neurotrophic factor receptor signaling. The Tarlov scale and quantal analysis technique results revealed that CNB-001 administered as an intravenous dose (bolus) 30 minutes prior to spinal cord ischemia improved the behaviors of female New Zealand White rabbits. The improvements were similar to those produced by the uncompetitive N-methyl-D-aspartate receptor antagonist memantine. At 48 hours after aortic occlusion, there was a 42.7% increase (P < 0.05) in tolerated ischemia duration (n = 14) for rabbits treated with CNB-001 (n = 16), and a 72.3% increase for rabbits treated with the positive control memantine (P < 0.05) (n = 23) compared to vehicle-treated ischemic rabbits (n = 22). CNB-001 is a potential important novel treatment for spinal cord ischemia induced by aortic occlusion. All experiments were approved by the CSMC Institutional Animal Care and Use Committee (IACUC #4311) on November 1, 2012.
Collapse
Affiliation(s)
| | - Paul D Boitano
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| | - Rene Bombien
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| | - Daisy Chou
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| | - Margot Knight
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| | - Anja Muehle
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| | - Mihaela Te Winkel
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| | - Ali Khoynezhad
- Department of Surgery, Memorial Care Health System, Long Beach, CA, USA
| |
Collapse
|
8
|
Lapchak PA, Boitano PD, Bombien R, Cook DJ, Doyan S, Lara JM, Schubert DR. CNB-001, a pleiotropic drug is efficacious in embolized agyrencephalic New Zealand white rabbits and ischemic gyrencephalic cynomolgus monkeys. Exp Neurol 2018; 313:98-108. [PMID: 30521790 DOI: 10.1016/j.expneurol.2018.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/16/2018] [Accepted: 11/30/2018] [Indexed: 01/10/2023]
Abstract
Ischemic stroke is an acute neurodegenerative disease that is extremely devastating to patients, their families and society. Stroke is inadequately treated even with endovascular procedures and reperfusion therapy. Using an extensive translational screening process, we have developed a pleiotropic cytoprotective agent with the potential to positively impact a large population of brain ischemia patients and revolutionize the process used for the development of new drugs to treat complex brain disorders. In this unique translational study article, we document that the novel curcumin-based compound, CNB-001, when administered as a single intravenous dose, has significant efficacy to attenuate clinically relevant behavioral deficits following ischemic events in agyrencephalic rabbits when administered 1 h post-embolization and reduces infarct growth in gyrencephalic non-human primates, when administered 5 min after initiation of middle cerebral artery occlusion. CNB-001 is safe and does not increase morbidity or mortality in either research species. Mechanistically, CNB-001 inhibits human 5- and 15-lipoxygenase in vitro, and can attenuate ischemia-induced inflammatory markers, and oxidative stress markers, while potentially promoting synaptic plasticity mediated by enhanced brain-derived neurotrophic factor (BDNF).
Collapse
Affiliation(s)
- Paul A Lapchak
- Neurocore LLC, Western University of Health Sciences, Pomona, CA 91766, USA.
| | | | | | - Douglas J Cook
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | | | | | - David R Schubert
- Cellular Neurobiology Laboratories, The Salk Institute, La Jolla, CA, USA
| |
Collapse
|
9
|
Preclinical Evidence and Mechanism of Xingnaojing Injection for Cerebral Ischemia: A Systematic Review and Meta-Analysis of Animal Studies. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:9624175. [PMID: 30581490 PMCID: PMC6276459 DOI: 10.1155/2018/9624175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 11/06/2018] [Indexed: 01/21/2023]
Abstract
Objectives Cerebral ischemia can cause severe harm to people's health with the characteristics of high incidence, high disability, and high mortality. Xingnaojing injection (XNJI) is widely used in the treatment of cerebral ischemia. The aim of this review is to evaluate the efficacy and mechanism of XNJI in animal models of cerebral ischemia. Methods Total seven electronic databases in English or Chinese (CNKI, Wanfang, VMIS, PubMed, MEDLINE, Embase, and the Cochrane Library) about most experiments and studies which came out before June 2018 of XNJI for cerebral ischemia have been searched. Data extraction, quality assessment, and meta-analysis are conducted according to the Cochrane standards and RevMan 5.3 software. Results We have identified 23 eligible studies and made a meta-analysis based on these studies. Meta-analysis shows that XNJI contributes significantly to reduction in neurological deficit score (P = 0.0002, MD = −1.25, 95% CI: −1.92, −0.58) compared with the control group of cerebral ischemia. Subgroup analytic results demonstrate that XNJI has been more effective in animal model of cerebral ischemia-reperfusion injury (P = 0.009, MD = −1.35, 95%CI: −2.36, −0.34) than that of permanent cerebral ischemia (P = 0.0002, MD = −1.08, 95%CI: −1.66, −0.51). Compared with control group, XNJI could remarkably reduce cerebral infarction area (P < 0.00001, MD = −14.98, 95%CI: −21.36, −8.59), brain edema (P < 0.00001, MD = −4.64, 95%CI: −5.38, −3.90), and neuronal cell apoptosis (P < 0.0001, MD = −12.21, 95%CI: 18.05, −6.37). Meanwhile, the meta-analysis shows that XNJI has a significant anti-inflammatory effect, and the levels of TNF-α, IL-6, and IL-1β are significantly reduced by XNJI (P = 0.001, MD = −4.13, 95%CI:−6.68, −1.58; P < 0.00001, MD = −119.23, 95%CI: −138.04, −100.43; P = 0.21, MD = −228.69, 95% CI: −586.20, 128.83). Additionally, XNJI could raise the body's antioxidant function and the level of SOD and GSH-Px (P = 0.002, MD = 53.02, 95% CI: −20.52, 85.78; P = 0.01, MD = 8.65, 95% CI: 1.77, 15.48) and decrease the level of MDA (P < 0.00001, MD = −4.16, 95% CI: −5.50, −2.82). Conclusion XNJI might be effective in cerebral ischemia by regulating oxidative stress and inflammatory reaction.
Collapse
|
10
|
Webb RL, Kaiser EE, Jurgielewicz BJ, Spellicy S, Scoville SL, Thompson TA, Swetenburg RL, Hess DC, West FD, Stice SL. Human Neural Stem Cell Extracellular Vesicles Improve Recovery in a Porcine Model of Ischemic Stroke. Stroke 2018; 49:1248-1256. [PMID: 29650593 PMCID: PMC5916046 DOI: 10.1161/strokeaha.117.020353] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/28/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Recent work from our group suggests that human neural stem cell-derived extracellular vesicle (NSC EV) treatment improves both tissue and sensorimotor function in a preclinical thromboembolic mouse model of stroke. In this study, NSC EVs were evaluated in a pig ischemic stroke model, where clinically relevant end points were used to assess recovery in a more translational large animal model. METHODS Ischemic stroke was induced by permanent middle cerebral artery occlusion (MCAO), and either NSC EV or PBS treatment was administered intravenously at 2, 14, and 24 hours post-MCAO. NSC EV effects on tissue level recovery were evaluated via magnetic resonance imaging at 1 and 84 days post-MCAO. Effects on functional recovery were also assessed through longitudinal behavior and gait analysis testing. RESULTS NSC EV treatment was neuroprotective and led to significant improvements at the tissue and functional levels in stroked pigs. NSC EV treatment eliminated intracranial hemorrhage in ischemic lesions in NSC EV pigs (0 of 7) versus control pigs (7 of 8). NSC EV-treated pigs exhibited a significant decrease in cerebral lesion volume and decreased brain swelling relative to control pigs 1-day post-MCAO. NSC EVs significantly reduced edema in treated pigs relative to control pigs, as assessed by improved diffusivity through apparent diffusion coefficient maps. NSC EVs preserved white matter integrity with increased corpus callosum fractional anisotropy values 84 days post-MCAO. Behavior and mobility improvements paralleled structural changes as NSC EV-treated pigs exhibited improved outcomes, including increased exploratory behavior and faster restoration of spatiotemporal gait parameters. CONCLUSIONS This study demonstrated for the first time that in a large animal model novel NSC EVs significantly improved neural tissue preservation and functional levels post-MCAO, suggesting NSC EVs may be a paradigm changing stroke therapeutic.
Collapse
Affiliation(s)
- Robin L Webb
- From the ArunA Biomedical, Athens, GA (R.L.W., S.L. Scoville, T.A.T., R.L.S).,Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| | - Erin E Kaiser
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice).,Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences (E.E.K., F.D.W.)
| | - Brian J Jurgielewicz
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| | - Samantha Spellicy
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| | - Shelley L Scoville
- From the ArunA Biomedical, Athens, GA (R.L.W., S.L. Scoville, T.A.T., R.L.S)
| | - Tyler A Thompson
- From the ArunA Biomedical, Athens, GA (R.L.W., S.L. Scoville, T.A.T., R.L.S)
| | | | - David C Hess
- University of Georgia, Rhodes Center for Animal and Dairy Science, Athens; and Department of Neurology, Augusta University, GA (D.C.H.)
| | - Franklin D West
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice).,Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences (E.E.K., F.D.W.)
| | - Steven L Stice
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| |
Collapse
|
11
|
Smith CJ, Hulme S, Vail A, Heal C, Parry-Jones AR, Scarth S, Hopkins K, Hoadley M, Allan SM, Rothwell NJ, Hopkins SJ, Tyrrell PJ. SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke): A Randomized Controlled Phase 2 Trial. Stroke 2018; 49:1210-1216. [PMID: 29567761 DOI: 10.1161/strokeaha.118.020750] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE The proinflammatory cytokine IL-1 (interleukin-1) has a deleterious role in cerebral ischemia, which is attenuated by IL-1 receptor antagonist (IL-1Ra). IL-1 induces peripheral inflammatory mediators, such as interleukin-6, which are associated with worse prognosis after ischemic stroke. We investigated whether subcutaneous IL-1Ra reduces the peripheral inflammatory response in acute ischemic stroke. METHODS SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke) was a single-center, double-blind, randomized, placebo-controlled phase 2 trial of subcutaneous IL-1Ra (100 mg administered twice daily for 3 days) in patients presenting within 5 hours of ischemic stroke onset. Randomization was stratified for baseline National Institutes of Health Stroke Scale score and thrombolysis. Measurement of plasma interleukin-6 and other peripheral inflammatory markers was undertaken at 5 time points. The primary outcome was difference in concentration of log(interleukin-6) as area under the curve to day 3. Secondary outcomes included exploratory effect of IL-1Ra on 3-month outcome with the modified Rankin Scale. RESULTS We recruited 80 patients (mean age, 72 years; median National Institutes of Health Stroke Scale, 12) of whom 73% received intravenous thrombolysis with alteplase. IL-1Ra significantly reduced plasma interleukin-6 (P<0.001) and plasma C-reactive protein (P<0.001). IL-1Ra was well tolerated with no safety concerns. Allocation to IL-1Ra was not associated with a favorable outcome on modified Rankin Scale: odds ratio (95% confidence interval)=0.67 (0.29-1.52), P=0.34. Exploratory mediation analysis suggested that IL-1Ra improved clinical outcome by reducing inflammation, but there was a statistically significant, alternative mechanism countering this benefit. CONCLUSIONS IL-1Ra reduced plasma inflammatory markers which are known to be associated with worse clinical outcome in ischemic stroke. Subcutaneous IL-1Ra is safe and well tolerated. Further experimental studies are required to investigate efficacy and possible interactions of IL-1Ra with thrombolysis. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: ISRCTN74236229.
Collapse
Affiliation(s)
- Craig J Smith
- From the Greater Manchester Comprehensive Stroke Centre, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S., A.R.P.-J., P.J.T.)
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Sharon Hulme
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Andy Vail
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, United Kingdom (A.V., C.H.)
| | - Calvin Heal
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, United Kingdom (A.V., C.H.)
| | - Adrian R Parry-Jones
- From the Greater Manchester Comprehensive Stroke Centre, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S., A.R.P.-J., P.J.T.)
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Sylvia Scarth
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Karen Hopkins
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Margaret Hoadley
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, University of Manchester, United Kingdom (S.M.A., N.J.R.)
| | - Nancy J Rothwell
- Division of Neuroscience and Experimental Psychology, University of Manchester, United Kingdom (S.M.A., N.J.R.)
| | - Stephen J Hopkins
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Pippa J Tyrrell
- From the Greater Manchester Comprehensive Stroke Centre, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S., A.R.P.-J., P.J.T.)
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| |
Collapse
|
12
|
Yang W, Paschen W. Is age a key factor contributing to the disparity between success of neuroprotective strategies in young animals and limited success in elderly stroke patients? Focus on protein homeostasis. J Cereb Blood Flow Metab 2017; 37:3318-3324. [PMID: 28752781 PMCID: PMC5624400 DOI: 10.1177/0271678x17723783] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neuroprotection strategies to improve stroke outcome have been successful in the laboratory but not in clinical stroke trials, and thus have come under scrutiny by the medical community. Experimental stroke investigators are therefore under increased pressure to resolve this problem. Acute ischemic stroke represents a severe form of metabolic stress that activates many pathological processes and thereby impairs cellular functions. Traditionally, neuroprotection strategies were designed to improve stroke outcome by interfering with pathological processes triggered by ischemia. However, stroke outcome is also dependent on the brain's capacity to restore cellular functions impaired by ischemia, and this capacity declines with age. It is, therefore, conceivable that this age-dependent decline in the brain's self-healing capacity contributes to the disparity between the success of neuroprotective strategies in young animals, and limited success in elderly stroke patients. Here, prosurvival pathways that restore protein homeostasis impaired by ischemic stress should be considered, because their capacity decreases with increasing age, and maintenance of proteome fidelity is pivotal for cell survival. Boosting such prosurvival pathways pharmacologically to restore protein homeostasis and, thereby, cellular functions impaired by ischemic stress is expected to counterbalance the compromised self-healing capacity of aged brains and thereby help to improve stroke outcome.
Collapse
Affiliation(s)
- Wei Yang
- 1 Laboratory of Molecular Neurobiology, Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Wulf Paschen
- 1 Laboratory of Molecular Neurobiology, Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,2 Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
13
|
Gribkoff VK, Kaczmarek LK. The need for new approaches in CNS drug discovery: Why drugs have failed, and what can be done to improve outcomes. Neuropharmacology 2017; 120:11-19. [PMID: 26979921 PMCID: PMC5820030 DOI: 10.1016/j.neuropharm.2016.03.021] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/14/2016] [Accepted: 03/11/2016] [Indexed: 12/31/2022]
Abstract
An important goal of biomedical research is to translate basic research findings into useful medical advances. In the field of neuropharmacology this requires understanding disease mechanisms as well as the effects of drugs and other compounds on neuronal function. Our hope is that this information will result in new or improved treatment for CNS disease. Despite great progress in our understanding of the structure and functions of the CNS, the discovery of new drugs and their clinical development for many CNS disorders has been problematic. As a result, CNS drug discovery and development programs have been subjected to significant cutbacks and eliminations over the last decade. While there has been recent resurgence of interest in CNS targets, these past changes in priority of the pharmaceutical and biotech industries reflect several well-documented realities. CNS drugs in general have higher failure rates than non-CNS drugs, both preclinically and clinically, and in some areas, such as the major neurodegenerative diseases, the clinical failure rate for disease-modifying treatments has been 100%. The development times for CNS drugs are significantly longer for those drugs that are approved, and post-development regulatory review is longer. In this introduction we review some of the reasons for failure, delineating both scientific and technical realities, some unique to the CNS, that have contributed to this. We will focus on major neurodegenerative disorders, which affect millions, attract most of the headlines, and yet have witnessed the fewest successes. We will suggest some changes that, when coupled with the approaches discussed in the rest of this special volume, may improve outcomes in future CNS-targeted drug discovery and development efforts. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Valentin K Gribkoff
- Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
14
|
Shinoda N, Hori S, Mikami K, Bando T, Shimo D, Kuroyama T, Kuramoto Y, Matsumoto M, Hirai O, Ueno Y. Utility of relative ADC ratio in patient selection for endovascular revascularization of large vessel occlusion. J Neuroradiol 2017; 44:185-191. [DOI: 10.1016/j.neurad.2016.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/19/2016] [Accepted: 12/30/2016] [Indexed: 10/19/2022]
|
15
|
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
|
16
|
Stroke Cytoprotection: Can Repeating History with New Expectations Really Be the Path to Success in Stroke Research? Transl Stroke Res 2017; 8:104-106. [DOI: 10.1007/s12975-017-0528-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 02/16/2017] [Indexed: 01/05/2023]
|
17
|
Hainsworth AH, Allan SM, Boltze J, Cunningham C, Farris C, Head E, Ihara M, Isaacs JD, Kalaria RN, Lesnik Oberstein SAMJ, Moss MB, Nitzsche B, Rosenberg GA, Rutten JW, Salkovic-Petrisic M, Troen AM. Translational models for vascular cognitive impairment: a review including larger species. BMC Med 2017; 15:16. [PMID: 28118831 PMCID: PMC5264492 DOI: 10.1186/s12916-017-0793-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 01/12/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Disease models are useful for prospective studies of pathology, identification of molecular and cellular mechanisms, pre-clinical testing of interventions, and validation of clinical biomarkers. Here, we review animal models relevant to vascular cognitive impairment (VCI). A synopsis of each model was initially presented by expert practitioners. Synopses were refined by the authors, and subsequently by the scientific committee of a recent conference (International Conference on Vascular Dementia 2015). Only peer-reviewed sources were cited. METHODS We included models that mimic VCI-related brain lesions (white matter hypoperfusion injury, focal ischaemia, cerebral amyloid angiopathy) or reproduce VCI risk factors (old age, hypertension, hyperhomocysteinemia, high-salt/high-fat diet) or reproduce genetic causes of VCI (CADASIL-causing Notch3 mutations). CONCLUSIONS We concluded that (1) translational models may reflect a VCI-relevant pathological process, while not fully replicating a human disease spectrum; (2) rodent models of VCI are limited by paucity of white matter; and (3) further translational models, and improved cognitive testing instruments, are required.
Collapse
Affiliation(s)
- Atticus H Hainsworth
- Clinical Neurosciences (J-0B) Molecular and Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK. .,Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK.
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Johannes Boltze
- Department of Translational Medicine and Cell Technology, University of Lübeck, Lübeck, Germany.,Neurovascular Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Catriona Cunningham
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Chad Farris
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Elizabeth Head
- Department of Pharmacology & Nutritional Sciences, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Masafumi Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Jeremy D Isaacs
- Clinical Neurosciences (J-0B) Molecular and Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK.,Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Raj N Kalaria
- Institute of Neuroscience, University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, UK
| | | | - Mark B Moss
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Björn Nitzsche
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.,Clinic for Nuclear Medicine, University of Leipzig, Leipzig, Germany.,Institute for Anatomy, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Gary A Rosenberg
- Department of Neurology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - Julie W Rutten
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Aron M Troen
- Institute of Biochemistry Food and Nutrition Science, Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
18
|
Thomas A, Detilleux J, Flecknell P, Sandersen C. Impact of Stroke Therapy Academic Industry Roundtable (STAIR) Guidelines on Peri-Anesthesia Care for Rat Models of Stroke: A Meta-Analysis Comparing the Years 2005 and 2015. PLoS One 2017; 12:e0170243. [PMID: 28122007 PMCID: PMC5266292 DOI: 10.1371/journal.pone.0170243] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/30/2016] [Indexed: 11/19/2022] Open
Abstract
Numerous studies using rats in stroke models have failed to translate into successful clinical trials in humans. The Stroke Therapy Academic Industry Roundtable (STAIR) has produced guidelines on the rodent stroke model for preclinical trials in order to promote the successful translation of animal to human studies. These guidelines also underline the importance of anaesthetic and monitoring techniques. The aim of this literature review is to document whether anaesthesia protocols (i.e., choice of agents, mode of ventilation, physiological support and monitoring) have been amended since the publication of the STAIR guidelines in 2009. A number of articles describing the use of a stroke model in adult rats from the years 2005 and 2015 were randomly selected from the PubMed database and analysed for the following parameters: country where the study was performed, strain of rats used, technique of stroke induction, anaesthetic agent for induction and maintenance, mode of intubation and ventilation, monitoring techniques, control of body temperature, vascular accesses, and administration of intravenous fluids and analgesics. For each parameter (stroke, induction, maintenance, monitoring), exact chi-square tests were used to determine whether or not proportions were significantly different across year and p values were corrected for multiple comparisons. An exact p-test was used for each parameter to compare the frequency distribution of each value followed by a Bonferroni test. The level of significant set at < 0.05. Results show that there were very few differences in the anaesthetic and monitoring techniques used between 2005 and 2015. In 2015, significantly more studies were performed in China and significantly fewer studies used isoflurane and nitrous oxide. The most striking finding is that the vast majority of all the studies from both 2005 and 2015 did not report the use of ventilation; measurement of blood gases, end-tidal carbon dioxide concentration, or blood pressure; or administration of intravenous fluids or analgesics. The review of articles published in 2015 showed that the STAIR guidelines appear to have had no effect on the anaesthetic and monitoring techniques in rats undergoing experimental stroke induction, despite the publication of said guidelines in 2009.
Collapse
MESH Headings
- Analgesics/administration & dosage
- Anesthesia/methods
- Anesthesia/standards
- Anesthesia/veterinary
- Anesthetics/administration & dosage
- Anesthetics/classification
- Animals
- Guideline Adherence
- Infarction, Middle Cerebral Artery
- Infusions, Intravenous/methods
- Infusions, Intravenous/standards
- Infusions, Intravenous/veterinary
- Intubation, Intratracheal/methods
- Intubation, Intratracheal/standards
- Intubation, Intratracheal/veterinary
- Models, Animal
- Monitoring, Intraoperative/methods
- Monitoring, Intraoperative/standards
- Monitoring, Intraoperative/veterinary
- Perioperative Care/methods
- Perioperative Care/standards
- Perioperative Care/veterinary
- Practice Guidelines as Topic
- Rats
- Respiration, Artificial/methods
- Respiration, Artificial/standards
- Respiration, Artificial/veterinary
- Sampling Studies
- Species Specificity
- Stroke
Collapse
Affiliation(s)
- Aurelie Thomas
- University of Liège, Faculty of Veterinary Medicine, Liege, Belgium
| | - Johann Detilleux
- University of Liège, Faculty of Veterinary Medicine, Liege, Belgium
| | - Paul Flecknell
- University of Newcastle, Comparative Biology Centre, Newcastle, United Kingdom
| | | |
Collapse
|
19
|
Lapchak PA, Zhang JH. The High Cost of Stroke and Stroke Cytoprotection Research. Transl Stroke Res 2016; 8:307-317. [PMID: 28039575 DOI: 10.1007/s12975-016-0518-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 10/20/2022]
Abstract
Acute ischemic stroke is inadequately treated in the USA and worldwide due to a lengthy history of neuroprotective drug failures in clinical trials. The majority of victims must endure life-long disabilities that not only affect their livelihood, but also have an enormous societal economic impact. The rapid development of a neuroprotective or cytoprotective compound would allow future stroke victims to receive a treatment to reduce disabilities and further promote recovery of function. This opinion article reviews in detail the enormous costs associated with developing a small molecule to treat stroke, as well as providing a timely overview of the cell-death time-course and relationship to the ischemic cascade. Distinct temporal patterns of cell-death of neurovascular unit components provide opportunities to intervene and optimize new cytoprotective strategies. However, adequate research funding is mandatory to allow stroke researchers to develop and test their novel therapeutic approach to treat stroke victims.
Collapse
Affiliation(s)
- Paul A Lapchak
- Director of Translational Research, Department of Neurology & Neurosurgery, Advanced Health Sciences Pavilion, Suite 8305, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd, Los Angeles, CA, 90048, USA.
| | - John H Zhang
- Director, Center for Neuroscience Research, Loma Linda University School of Medicine, 11175 Campus St, Loma Linda, CA, 92350, USA
| |
Collapse
|
20
|
Urodilatin reverses the detrimental influence of bradykinin in acute ischemic stroke. Exp Neurol 2016; 284:1-10. [DOI: 10.1016/j.expneurol.2016.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/15/2016] [Accepted: 07/14/2016] [Indexed: 02/03/2023]
|
21
|
Carey LM, Seitz RJ. Functional Neuroimaging in Stroke Recovery and Neurorehabilitation: Conceptual Issues and Perspectives. Int J Stroke 2016; 2:245-64. [DOI: 10.1111/j.1747-4949.2007.00164.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background In stroke, functional neuroimaging has become a potent diagnostic tool; opened new insights into the pathophysiology of ischaemic damage in the human brain; and made possible the assessment of functional–structural relationships in postlesion recovery. Summary of review Here, we give a critical account on the potential and limitation of functional neuroimaging and discuss concepts related to the use of neuroimaging for exploring the neurobiological and neuroanatomical mechanisms of poststroke recovery and neurorehabilitation. We identify and provide evidence for five hypotheses that functional neuroimaging can provide new insights into: adaptation occurs at the level of functional brain systems; the brain–behaviour relationship varies with recovery and over time; functional neuroimaging can improve our ability to predict recovery and select individuals for rehabilitation; mechanisms of recovery reflect different pathophysiological phases; and brain adaptation may be modulated by experience and specific rehabilitation. The significance and application of this new evidence is discussed, and recommendations made for investigations in the field. Conclusion Functional neuroimaging is an important tool to explore the mechanisms underlying brain plasticity and, thereby, to guide clinical research in neurorehabilitation.
Collapse
Affiliation(s)
- Leeanne M. Carey
- National Stroke Research Institute, Neurosciences Building, Heidelberg Heights, Vic., Australia
- School of Occupational Therapy, LaTrobe University, Bundoora, Vic., Australia
| | - Rüdiger J. Seitz
- National Stroke Research Institute, Neurosciences Building, Heidelberg Heights, Vic., Australia
- Institute of Advanced Study, La Trobe University, Bundoora, Vic., Australia
- Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
22
|
Fisher M, Cheung K, Howard G, Warach S. New Pathways for Evaluating Potential Acute Stroke Therapies. Int J Stroke 2016; 1:52-8. [DOI: 10.1111/j.1747-4949.2006.00025.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pharmacological therapy for acute ischemic stroke remains limited to one successful, approved treatment: tissue plasminogen activator within 3 h of stroke onset. Many neuroprotective drugs and a few other thrombolytics were evaluated in clinical trials, but none demonstrated unequivocal success and were approved by regulatory agencies. The development paradigm for such therapies needs to provide convincing evidence of efficacy and safety to obtain approval by the Food and Drug Administration (FDA). The FDA modernization act of 1997 stated that such evidence could be derived from one large phase III trial with a clinical endpoint and supportive evidence. Drugs being developed for acute ischemic stroke can potentially be approved under this act by coupling a major phase III trial with supportive evidence provided by a phase IIB trial demonstrating an effect on a relevant biomarker such as magnetic resonance imaging or computed tomography assessment of ischemic lesion growth. Statistical approaches have been developed to optimize the design of such an imaging-based phase IIB study, for example approaches that modify randomization probabilities to assign larger proportions of patients to the ‘winning’ strategy (i.e. ‘pick the winner’ strategies) with an interim assessment to reduce the sample size requirement. Demonstrating a treatment effect on a relevant imaging-based biomarker should provide supportive evidence for a new drug application, if a subsequent phase III trial with a clinical outcome demonstrates a significant treatment effect.
Collapse
Affiliation(s)
- Marc Fisher
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Kenneth Cheung
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | | | - Steven Warach
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| |
Collapse
|
23
|
Jolkkonen J, Kwakkel G. Translational Hurdles in Stroke Recovery Studies. Transl Stroke Res 2016; 7:331-42. [PMID: 27000881 DOI: 10.1007/s12975-016-0461-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/11/2016] [Accepted: 03/09/2016] [Indexed: 12/20/2022]
Abstract
Emerging understanding of brain plasticity has opened new avenues for the treatment of stroke. The promising preclinical evidence with neuroprotective drugs has not been confirmed in clinical trials, thus nowadays, researchers, pharmaceutical companies, and funding bodies hesitate to initiate these expensive trials with restorative therapies. Since many of the previous failures can be traced to low study quality, a number of guidelines such as STAIR and STEPS were introduced to rectify these shortcomings. However, these guidelines stem from the study design for neuroprotective drugs and one may question whether they are appropriate for restorative approaches, which rely heavily on behavioral testing. Most of the recovery studies conducted in stroke patients have been small-scale, proof-of-concept trials. Consequently, the overall effect sizes of pooled phase II trials have proved unreliable and unstable in most meta-analyses. Although the methodological quality of trials in humans is improving, most studies still suffer from methodological flaws and do not meet even the minimum of evidence-based standards for reporting randomized controlled trials. The power problem of most phase II trials is mostly attributable to a lack of proper stratification with robust prognostic factors at baseline as well as the incorrect assumption that all patients will exhibit the same proportional amount of spontaneous neurological recovery poststroke. In addition, most trials suffer from insufficient treatment contrasts between the experimental and control arm and the outcomes have not been sufficiently responsive to detect small but clinically relevant changes in neurological impairments and activities. This narrative review describes the main factors that bias recovery studies, both in experimental animals and stroke patients.
Collapse
Affiliation(s)
- Jukka Jolkkonen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland. .,Neurocenter, Neurology, University Hospital of Kuopio, Kuopio, Finland.
| | - Gert Kwakkel
- Department of Rehabilitation Medicine, VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands.,Neurorehabilitation, Amsterdam Rehabilitation Research Center, Reade, Amsterdam, The Netherlands.,Neuroscience Campus Amsterdam, VU University Amsterdam, Amsterdam, The Netherlands.,Department of Physical Therapy and Human Movement Sciences, Northwestern University, Evanston, IL, USA
| |
Collapse
|
24
|
Tiwari YV, Jiang Z, Sun Y, Du F, Rodriguez P, Shen Q, Duong TQ. Effects of stroke severity and treatment duration in normobaric hyperoxia treatment of ischemic stroke. Brain Res 2016; 1635:121-9. [PMID: 26826010 PMCID: PMC4779399 DOI: 10.1016/j.brainres.2016.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 11/26/2022]
Abstract
In order to improve clinical trial design and translation of normobaric oxygen (NBO) treatment of ischemic stroke, NBO treatment parameters need to be better understood. This study investigated NBO treatment efficacy at two different stroke severities and two NBO treatment durations in rats. For the 60-min middle cerebral artery occlusion (MCAO), NBO treatment for 25 min and 150 min were studied. For the 90-min MCAO, NBO treatment for 55 min and 150 min were studied. Cerebral blood flow (CBF), apparent diffusion coefficients (ADC) and T2 MRI were acquired during occlusion prior to treatment, after reperfusion, and 48h after MCAO. The effects of NBO treatment on lesion volumes, and CBF, ADC and T2 of ischemic core, perfusion-diffusion mismatch and normal tissue were analyzed longitudinally. The major findings were: i) NBO treatment was effective in both groups of stroke severities, salvaging similar percentage of initial abnormal ADC tissue, and ii) NBO treatments continued after reperfusion were more beneficial than NBO treatment during occlusion alone for both MCAO groups. These findings underscore the importance of the effects of NBO duration and stroke severity on treatment outcomes.
Collapse
Affiliation(s)
- Yash Vardhan Tiwari
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA; Department of Biomedical Engineering, University of Texas, San Antonio, TX, USA
| | - Zhao Jiang
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Yuhao Sun
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Fang Du
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Pavel Rodriguez
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Qiang Shen
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Timothy Q Duong
- Research Imaging Institute, University of Texas Health Science Center, 8403 Floyd Curl Dr, San Antonio, TX 78229, USA.
| |
Collapse
|
25
|
Winship IR. Cerebral collaterals and collateral therapeutics for acute ischemic stroke. Microcirculation 2015; 22:228-36. [PMID: 25351102 DOI: 10.1111/micc.12177] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/22/2014] [Indexed: 11/29/2022]
Abstract
Cerebral collaterals are vascular redundancies in the cerebral circulation that can partially maintain blood flow to ischemic tissue when primary conduits are blocked. After occlusion of a cerebral artery, anastomoses connecting the distal segments of the MCA with distal branches of the ACA and PCA (known as leptomeningeal or pial collaterals) allow for partially maintained blood flow in the ischemic penumbra and delay or prevent cell death. However, collateral circulation varies dramatically between individuals, and collateral extent is significant predictor of stroke severity and recanalization rate. Collateral therapeutics attempt to harness these vascular redundancies by enhancing blood flow through pial collaterals to reduce ischemia and brain damage after cerebral arterial occlusion. While therapies to enhance collateral flow remain relatively nascent neuroprotective strategies, experimental therapies including inhaled NO, transient suprarenal aortic occlusion, and electrical stimulation of the parasympathetic sphenopalatine ganglion show promise as collateral therapeutics with the potential to improve treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Ian R Winship
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
26
|
Blondeau N. The nutraceutical potential of omega-3 alpha-linolenic acid in reducing the consequences of stroke. Biochimie 2015; 120:49-55. [PMID: 26092420 DOI: 10.1016/j.biochi.2015.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023]
Abstract
Stroke is a worldwide major cause of mortality and morbidity. Preclinical studies have identified over 1000 molecules with brain-protective properties. More than 200 clinical trials have evaluated neuroprotective candidates for ischemic stroke yet, to date almost all failed, leading to a re-analysis of treatment strategies against stroke. An emerging view is to seek combinatory therapy, or discovering molecules able to stimulate multiple protective and regenerative mechanisms. A pertinent experimental approach to identify such candidates is the study of brain preconditioning, which refers to how the brain protects itself against ischemia and others stress-inducing stimuli. The recent discovery that nutrients like alpha-linolenic acid (ALA is an essential omega-3 polyunsaturated fatty acid required as part of our daily diet), may be an efficient brain preconditionner against stroke fosters the novel concept of brain preconditioning by nutraceuticals. This review stresses the underestimated role of nutrition in preventing and combating stroke. Although there is a consensus that increased consumption of salt, fatty foods and alcoholic beverages may promote pathologies like hypertension, obesity and alcoholism - all of which are well known risk factors of stroke - few risk factors are attributed to a deficiency in an essential nutrient in the diet. The ALA deficiency observed in the Western modern diets may itself constitute a risk factor. This review outlines how ALA supplementation by modification of the daily diet prevented mortality and cerebral damage in a rodent model of ischemic stroke. It also describes the pleiotropic ability of ALA to trigger responses that are multicellular, mechanistically diverse, resulting in neuronal protection, stimulation of neuroplasticity, and brain artery vasodilation. Overall, this review proposes a promising therapeutic opportunity by integrating a nutritional-based approach focusing on enriching the daily diet in ALA to prevent the devastating damage caused by stroke.
Collapse
Affiliation(s)
- Nicolas Blondeau
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France.
| |
Collapse
|
27
|
Jickling GC, Sharp FR. Improving the translation of animal ischemic stroke studies to humans. Metab Brain Dis 2015; 30:461-7. [PMID: 24526567 PMCID: PMC4186910 DOI: 10.1007/s11011-014-9499-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 01/28/2014] [Indexed: 12/18/2022]
Abstract
Despite testing more than 1,026 therapeutic strategies in models of ischemic stroke and 114 therapies in human ischemic stroke, only one agent tissue plasminogen activator has successfully been translated to clinical practice as a treatment for acute stroke. Though disappointing, this immense body of work has led to a rethinking of animal stroke models and how to better translate therapies to patients with ischemic stroke. Several recommendations have been made, including the STAIR recommendations and statements of RIGOR from the NIH/NINDS. In this commentary we discuss additional aspects that may be important to improve the translational success of ischemic stroke therapies. These include use of tissue plasminogen activator in animal studies; modeling ischemic stroke heterogeneity in terms of infarct size and cause of human stroke; addressing the confounding effect of anesthesia; use of comparable therapeutic dosage between humans and animals based on biological effect; modeling the human immune system; and developing outcome measures in animals comparable to those used in human stroke trials. With additional study and improved animal modeling of factors involved in human ischemic stroke, we are optimistic that new stroke therapies will be developed.
Collapse
Affiliation(s)
- Glen C Jickling
- Department of Neurology, MIND Institute Wet Labs Room 2415, University of California at Davis Medical Center, 2805 50th Street, Sacramento, CA, 95817, USA,
| | | |
Collapse
|
28
|
Smith CJ, Denes A, Tyrrell PJ, Di Napoli M. Phase II anti-inflammatory and immune-modulating drugs for acute ischaemic stroke. Expert Opin Investig Drugs 2015; 24:623-43. [PMID: 25727670 DOI: 10.1517/13543784.2015.1020110] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Stroke is the second leading cause of death worldwide and the leading cause of adult neurological disability. Despite advances in stroke unit care, and increasing use of thrombolysis, there remains an urgent need for safe and effective treatments for acute ischaemic stroke. However, this is against a backdrop of multiple failures in translational drug development. Cerebral ischaemia initiates a complex cascade of immune and inflammatory pathways in the brain microvasculature and periphery, which contribute to the evolution of cerebral injury, resolution and repair. Targeting specific inflammatory or immune pathways, therefore, represents an attractive treatment strategy in acute ischaemic stroke. Although anti-inflammatory drugs have already failed in clinical trial development, several are currently at the Phase II developmental stage. AREAS COVERED The authors highlight several candidate drugs, which modulate a range of inflammatory and immune pathways, and have been investigated in pre-clinical and Phase II studies to date. EXPERT OPINION Drugs targeting inflammatory and immune pathways offer theoretical advantages including potentially longer therapeutic time windows and effects complementary to thrombolysis (ameliorating reperfusion injury). Fundamental changes in the approach to pre-clinical and clinical drug development are required to facilitate successful translation of promising candidate drugs into clinical practice.
Collapse
Affiliation(s)
- Craig J Smith
- Greater Manchester Comprehensive Stroke Centre, Department of Medical Neurosciences, Salford Royal Foundation Trust , Salford , UK
| | | | | | | |
Collapse
|
29
|
Wali B, Ishrat T, Won S, Stein DG, Sayeed I. Progesterone in experimental permanent stroke: a dose-response and therapeutic time-window study. Brain 2014; 137:486-502. [PMID: 24374329 PMCID: PMC3914469 DOI: 10.1093/brain/awt319] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/10/2013] [Accepted: 09/23/2013] [Indexed: 11/14/2022] Open
Abstract
Currently, the only approved treatment for ischaemic stroke is tissue plasminogen activator, a clot-buster. This treatment can have dangerous consequences if not given within the first 4 h after stroke. Our group and others have shown progesterone to be beneficial in preclinical studies of stroke, but a progesterone dose-response and time-window study is lacking. We tested male Sprague-Dawley rats (12 months old) with permanent middle cerebral artery occlusion or sham operations on multiple measures of sensory, motor and cognitive performance. For the dose-response study, animals received intraperitoneal injections of progesterone (8, 16 or 32 mg/kg) at 1 h post-occlusion, and subcutaneous injections at 6 h and then once every 24 h for 7 days. For the time-window study, the optimal dose of progesterone was given starting at 3, 6 or 24 h post-stroke. Behavioural recovery was evaluated at repeated intervals. Rats were killed at 22 days post-stroke and brains extracted for evaluation of infarct volume. Both 8 and 16 mg/kg doses of progesterone produced attenuation of infarct volume compared with the placebo, and improved functional outcomes up to 3 weeks after stroke on locomotor activity, grip strength, sensory neglect, gait impairment, motor coordination and spatial navigation tests. In the time-window study, the progesterone group exhibited substantial neuroprotection as late as 6 h after stroke onset. Compared with placebo, progesterone showed a significant reduction in infarct size with 3- and 6-h delays. Moderate doses (8 and 16 mg/kg) of progesterone reduced infarct size and improved functional deficits in our clinically relevant model of stroke. The 8 mg/kg dose was optimal in improving motor, sensory and memory function, and this effect was observed over a large therapeutic time window. Progesterone shows promise as a potential therapeutic agent and should be examined for safety and efficacy in a clinical trial for ischaemic stroke.
Collapse
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
30
|
Oligogenesis and oligodendrocyte progenitor maturation vary in different brain regions and partially correlate with local angiogenesis after ischemic stroke. Transl Stroke Res 2013; 2:366-75. [PMID: 22022343 DOI: 10.1007/s12975-011-0078-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Oligogenesis plays an important role in functional recovery after ischemic stroke. We tested the hypothesis that oligogenesis and the maturation of oligodendrocyte progenitor cells (OPCs) vary in different brain regions using a rat transient middle cerebral artery occlusion (tMCAO) model. Compared to Day 1, olig2(+) OPCs and oligodendrocytes (OLGs) increased in the peri-infarct basal ganglia (BG) 7 (44%) and 14 (61%) days after 2 hours of MCAO; OPCs (PDGFRα(+)) and OLGs (CC1(+)) increased in this region 14 days after tMCAO by 139% and 126%, respectively. Although the olig2(+) cells and OLGs did not increase significantly in the peri-infarct cortex (CTX), the OPCs increased in this region by 95% at Day 14 vs. Day 1 after tMCAO. The numbers of OPCs and OLGs remained low after an initial reduction at Day 1 in the peri-infarct corpus callosum (CC). Correlation analyses showed that the numbers of olig2(+) cells (r=0.73, P=0.03) and OLGs (r=0.74, P=0.02) correlated with local vessel density; however, the number of OPCs did not correlate with vessel density (r=0.43, P=0.24). Our data show that oligogenesis and the maturation of OPCs differ in various brain regions and the difference in regional angiogenic response is one of the potential reasons.
Collapse
|
31
|
|
32
|
Hyun Lee J, Zheng Y, von Bornstadt D, Wei Y, Balcioglu A, Daneshmand A, Yalcin N, Yu E, Herisson F, Atalay YB, Kim MH, Ahn YJ, Balkaya M, Sweetnam P, Schueller O, Poyurovsky MV, Kim HH, Lo EH, Furie KL, Ayata C. Selective ROCK2 Inhibition In Focal Cerebral Ischemia. Ann Clin Transl Neurol 2013; 1:2-14. [PMID: 24466563 PMCID: PMC3900310 DOI: 10.1002/acn3.19] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVE Rho-associated kinase (ROCK) is a key regulator of numerous processes in multiple cell types relevant in stroke pathophysiology. ROCK inhibitors have improved outcome in experimental models of acute ischemic or hemorrhagic stroke. However, the relevant ROCK isoform (ROCK1 or ROCK2) in acute stroke is not known. METHODS We characterized the pharmacodynamic and pharmacokinetic profile, and tested the efficacy and safety of a novel selective ROCK2 inhibitor KD025 (formerly SLx-2119) in focal cerebral ischemia models in mice. RESULTS KD025 dose-dependently reduced infarct volume after transient middle cerebral artery occlusion. The therapeutic window was at least 3 hours from stroke onset, and the efficacy was sustained for at least 4 weeks. KD025 was at least as efficacious in aged, diabetic or female mice, as in normal adult males. Concurrent treatment with atorvastatin was safe, but not additive or synergistic. KD025 was also safe in a permanent ischemia model, albeit with diminished efficacy. As one mechanism of protection, KD025 improved cortical perfusion in a distal middle cerebral artery occlusion model, implicating enhanced collateral flow. Unlike isoform-nonselective ROCK inhibitors, KD025 did not cause significant hypotension, a dose-limiting side effect in acute ischemic stroke. INTERPRETATION Altogether, these data show that KD025 is efficacious and safe in acute focal cerebral ischemia in mice, implicating ROCK2 as the relevant isoform in acute ischemic stroke. Data suggest that selective ROCK2 inhibition has a favorable safety profile to facilitate clinical translation.
Collapse
Affiliation(s)
- Jeong Hyun Lee
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Yi Zheng
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Daniel von Bornstadt
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Ying Wei
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Aygul Balcioglu
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Ali Daneshmand
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Nilufer Yalcin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Esther Yu
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Fanny Herisson
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Yahya B Atalay
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Maya H Kim
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Yong-Joo Ahn
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Mustafa Balkaya
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | | | | | | | - Hyung-Hwan Kim
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Eng H Lo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
| | - Karen L Furie
- Department of Neurology, Rhode Island HospitalProvidence, Rhode Island, 02903
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, Massachusetts, 02129
- Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical SchoolBoston, Massachusetts, 02114
- Correspondence Cenk Ayata, Neurovascular Research Laboratory, Massachusetts General Hospital, 149 13th Street, Room 6403, Charlestown, MA 02129. Tel: (617) 726-8021; Fax: (617) 726-2547; E-mail:
| |
Collapse
|
33
|
Abstract
Neuroprotective drugs have so far failed clinical trials, at high cost, and intravenous tissue plasminogen activator (i.v. tPA) remains the only FDA-approved acute stroke therapy. Hyperoxia, acting via multiple direct and indirect mechanisms, may be a powerful neuroprotective strategy to salvage acutely ischemic brain tissue and extend the time window for acute stroke treatment. Of the available oxygen delivery methods, hyperbaric oxygen therapy (HBO) appears to be the most potent, while even normobaric oxygen therapy (NBO) may be effective if started promptly after stroke onset. HBO has so far failed to show efficacy in three clinical trials. The failure of these trials is probably attributable to factors such as delayed time to therapy, inadequate sample size and use of excessive chamber pressures. Previous trials did not assess long-term benefit in patients with tissue reperfusion. In this modern era of stroke thrombolysis and advanced neuroimaging, oxygen therapy may have renewed significance. If applied within the first few hours after stroke onset or in patients with imaging evidence of salvageable brain tissue, oxygen therapy could be used to 'buy time' for the administration of thrombolytic or neuroprotective drugs. This article reviews the history and current rationale for using oxygen therapy in stroke, the mechanisms of action of HBO and the results of animal and human studies of hyperoxia in cerebrovascular diseases.
Collapse
Affiliation(s)
- Aneesh B Singhal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
34
|
Lapchak PA, Zhang JH, Noble-Haeusslein LJ. RIGOR guidelines: escalating STAIR and STEPS for effective translational research. Transl Stroke Res 2013; 4:279-85. [PMID: 23658596 PMCID: PMC3644408 DOI: 10.1007/s12975-012-0209-2] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 08/14/2012] [Indexed: 01/10/2023]
Abstract
Stroke continues to be a serious and significant health problem in the USA and worldwide. This article will emphasize the need for good laboratory practices, transparent scientific reporting, and the use of translational research models representative of the disease state to develop effective treatments. This will allow for the testing and development of new innovative strategies so that efficacious therapies can be developed to treat ischemic and hemorrhagic stroke. This article recommends guidelines for effective translational research, most importantly, the need for study blinding, study group randomization, power analysis, accurate statistical analysis, and a conflict of interest statement. Additional guidelines to ensure reproducibility of results and confirmation of efficacy in multiple species are discussed.
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
| | - John H. Zhang
- Department of Neurosurgery, Anesthesiology, Neurology, and Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350 USA
| | - Linda J. Noble-Haeusslein
- Department of Neurological Surgery, Physical Therapy and Rehabilitation Science, and Brain and Spinal Injury Center (BASIC), University of California, San Francisco, Box 0112, 513 Parnassus Avenue, HSE-722, San Francisco, CA 94143-0112 USA
| |
Collapse
|
35
|
Abstract
There have been only 3 positive Phase III randomized clinical trials in acute ischemic stroke, all reperfusion therapies (NINDS; PROACT II; ECASS III). The only approved acute stroke therapy is <3-hour IV tPA. Although numerous compounds have shown benefit in animal models of brain infarction, there has never been a positive Phase III randomized clinical trial of a neuroprotectant in acute ischemic stroke. There are many challenges to acute stroke clinical trials but chief among these are the very short therapeutic window ("time is brain") and the issue of stroke heterogeneity. Stroke is a syndrome and only a very small percentage of all stroke patients present to hospitals in time to consider reperfusion therapy. Many drugs have been rushed to trial prematurely based on inadequate preclinical testing. Many trials have been seriously underpowered due to overly optimistic treatment expectations and the risk of brain hemorrhage has precluded aggressive multimodal treatment strategies. Rather than simply relying on a clock, new imaging methods are being developed to identify patients with "tissue at risk" and "salvageable brain" regardless of time of stroke onset. The 7 STAIR conferences have been convened to address these and other challenges to acute ischemic stroke trial design and completion.
Collapse
Affiliation(s)
- Anthony J Furlan
- University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
36
|
Affiliation(s)
- David W. Howells
- From the Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia (D.W.H.); and Department of Clinical Neurosciences, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom (M.R.M.)
| | - Malcolm R. Macleod
- From the Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia (D.W.H.); and Department of Clinical Neurosciences, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom (M.R.M.)
| |
Collapse
|
37
|
Neurovascular protection by targeting early blood-brain barrier disruption with neurotrophic factors after ischemia-reperfusion in rats*. J Cereb Blood Flow Metab 2013; 33:557-66. [PMID: 23299242 PMCID: PMC3618392 DOI: 10.1038/jcbfm.2012.201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The 'new penumbra' concept imbues the transition between injury and repair at the neurovascular unit with profound implications for selecting the appropriate type and timing of neuroprotective interventions. In this conceptual study, we investigated the protective effects of pigment epithelium-derived factor (PEDF) and compared them with the properties of epidermal growth factor (EGF) in a rat model of ischemia-reperfusion injury. We initiated a delayed intervention 3 hours after reperfusion using equimolar amounts of PEDF and EGF. These agents were then administered intravenously for 4 hours following reperfusion after 1 hour of focal ischemia. Magnetic resonance imaging indices were characterized, and imaging was performed at multiple time points post reperfusion. PEDF and EGF reduced lesion volumes at all time points as observed on T2-weighted images (T2-LVs). In addition PEDF selectively attenuated lesion volume expansion at 48 hours after reperfusion and persistently modulated blood-brain barrier (BBB) permeability at all time points. Intervention with peptides is suspected to cause edema formation at distant regions. The observed T2-LV reduction and BBB modulation by these trophic factors is probably mediated through a number of diverse mechanisms. A thorough evaluation of neurotrophins is still necessary to determine their time-dependent contributions against injury and their modulatory effects on repair after stroke.
Collapse
|
38
|
Zaidat OO, Liebeskind DS, Edgell RC, Amlie-Lefond CM, Kalia JS, Alexandrov AV. Clinical trial design for endovascular ischemic stroke intervention. Neurology 2012; 79:S221-33. [PMID: 23008403 DOI: 10.1212/wnl.0b013e31826992cf] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Randomized, double-blinded, placebo-controlled trials have significant impact on clinical practice. The ultimate goal of a clinical trial of therapy for acute ischemic stroke (AIS) is to compare 2 interventions. Challenges may include interventional therapy standardization, enrollment rate, patient selection, biases, data and safety monitoring, reporting, and financial and logistical support. METHOD Selected randomized and single-arm prospective AIS trial designs. Clinical trial elements and their challenges are reviewed. Innovative designs and proposed recommendations to overcome some of the specific challenges and limitations are discussed. RESULTS AIS therapy trials have specific challenges related to ethical issues, enrollment rate, outcome measures, limited time to treatment, efficacy, safety, and limited or variable operator experience with complex technology in a delicate end organ. Proposed suggestions for improving trial design include the following: incorporation of a lead-in phase; careful patient and outcome measure selection; historical, concurrent, or hybrid controls; open data access; and a Bayesian approach. An open data paradigm may facilitate creation of computerized prediction models for future trials (minimizing cost by decreasing sample size or providing futility analyses and directing resources to other trials). Collaborative, consortium, and network infrastructures may allow more effective and efficient study completion. Self-learning, self-correcting trials with intrinsic flexibility to adapt may help future clinical trial design in AIS. CONCLUSION The randomized clinical trial design in AIS endovascular therapy is challenging. Lead-in phases, careful patient selection, use of innovative outcome measures, control groups, and newer clinical trial design may enhance conduct of future trials, their validity, and their results.
Collapse
Affiliation(s)
- Osama O Zaidat
- Department of Neurology, Medical College of Wisconsin and Froedtert Hospital, Milwaukee, WI, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Zhang GS, Tian Y, Huang JY, Tao RR, Liao MH, Lu YM, Ye WF, Wang R, Fukunaga K, Lou YJ, Han F. The γ-secretase blocker DAPT reduces the permeability of the blood-brain barrier by decreasing the ubiquitination and degradation of occludin during permanent brain ischemia. CNS Neurosci Ther 2012; 19:53-60. [PMID: 23171401 DOI: 10.1111/cns.12032] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/04/2012] [Accepted: 10/08/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Tight junction protein degradation is a principal characteristic of the blood-brain barrier (BBB) damage that occurs during brain ischemia. AIMS We investigated the mechanisms of occludin degradation that underlie permanent middle cerebral artery occlusion (pMCAO) in rats. METHODS AND RESULTS Western blot and Co-immunoprecipitation data indicated ubiquitination and degradation of occludin in brain after pMCAO, which was consistent with ZO-1 degradation in penumbra regions as observed at 24 h after pMCAO. We further investigated candidate protease(s) responsible for the degradation of occludin during pMCAO. The intraventricular administration of γ-secretase blocker DAPT significantly inhibited the pMCAO-induced neurovascular damage, whereas ALLM and Batimastat, which are inhibitors of calpain and metalloproteinase proteases, respectively, were less effective. Notably, we found that DAPT significantly inhibited BBB disruption in comparison with vehicle treatment, as assessed by Evans blue excretion. Interestingly, the confocal immunostaining revealed that activation of the E3 ubiquitin ligase Itch is associated with degradation of occludin in brain microvessels following ischemia. Furthermore, our data demonstrate that the inhibition of γ-secretase signaling and the itch-mediated ubiquitination of occludin likely underlie the vasoprotective effect of DAPT after pMCAO. CONCLUSION The γ-secretase blocker DAPT reduces the permeability of the BBB by decreasing the ubiquitination and degradation of occludin during permanent brain ischemia, suggesting that γ-secretase may represent a novel therapeutic target for preventing neurovascular damage.
Collapse
Affiliation(s)
- Gen-Sheng Zhang
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lammertse DP. Clinical trials in spinal cord injury: lessons learned on the path to translation. The 2011 International Spinal Cord Society Sir Ludwig Guttmann Lecture. Spinal Cord 2012; 51:2-9. [DOI: 10.1038/sc.2012.137] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
41
|
Li J, Dong Y, Chen H, Han H, Yu Y, Wang G, Zeng Y, Xie K. Protective effects of hydrogen-rich saline in a rat model of permanent focal cerebral ischemia via reducing oxidative stress and inflammatory cytokines. Brain Res 2012; 1486:103-11. [PMID: 23010312 DOI: 10.1016/j.brainres.2012.09.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 09/16/2012] [Accepted: 09/18/2012] [Indexed: 11/17/2022]
Abstract
Hydrogen gas (H(2)) as a new medical gas exerts organ-protective effects through regulating oxidative stress, inflammation and apoptosis. In contrast to H(2), hydrogen-rich saline (HS) may be more suitable for clinical application. The present study was designed to investigate whether HS can offer a neuroprotective effect in a rat model of permanent focal cerebral ischemia and what mechanism(s) underlies the effect. Sprague-Dawley rats were subjected to permanent focal cerebral ischemia induced by permanent middle cerebral artery occlusion (pMCAO). Different doses of HS or normal saline were intraperitoneally administered at 5min after pMCAO or sham operation followed by injections at 6h, 12h and 24h. Here, we found that HS treatment significantly reduced infarct volume and improved neurobehavioral outcomes at 24h, 48h and 72h after pMCAO operation in a dose-dependent manner (P<0.05). Moreover, we found that HS treatment dose-dependently increased the activities of endogenous antioxidant enzymes (SOD and CAT) as well as decreased the levels of oxidative products (8-iso-PGF2α and MDA) and inflammatory cytokines (TNF-α and HMGB1) in injured ipsilateral brain tissues at 6h, 12h and 24h after pMCAO operation (P<0.05). Thus, hydrogen-rich saline dose-dependently exerts a neuroprotective effect against permanent focal cerebral ischemia, and its beneficial effect is at least partially mediated by reducing oxidative stress and inflammation. Molecular hydrogen may be an effective therapeutic strategy for stroke patients.
Collapse
Affiliation(s)
- Jianjun Li
- Department of Neurology, No. 323 Hospital of PLA, Xi'an 710054, Shaanxi Province, PR China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Kummer RV, Albers GW, Mori E. The Desmoteplase in Acute Ischemic Stroke (DIAS) Clinical Trial Program. Int J Stroke 2012; 7:589-96. [DOI: 10.1111/j.1747-4949.2012.00910.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background Desmoteplase is a novel, highly fibrin-specific thrombolytic agent in phase III of clinical development. In comparison to alteplase, it has high fibrin selectivity, is associated with minimal or no neurotoxicity, and has no apparent negative effect on the blood–brain barrier. The safety and efficacy of desmoteplase is being studied in the Desmoteplase in Acute Ischemic Stroke clinical trial program. Three studies (Dose Escalation Study of Desmoteplase in Acute Ischemic Stroke, Desmoteplase in Acute Ischemic Stroke, and Desmoteplase in Acute Ischemic Stroke-2) have been completed, two large randomized, double-blind, placebo-controlled, phase III trials are ongoing at >200 sites worldwide (Desmoteplase in Acute Ischemic Stroke-3 and Desmoteplase in Acute Ischemic Stroke-4, n = 800; DIAS-3 and DIAS-4), and a randomized, double-blind, placebo-controlled, dose-escalation phase II trial is ongoing in Japan (Desmoteplase in Acute Ischemic Stroke-Japan, n = 48; DIAS-J). Aims The objective of DIAS-3 and DIAS-4 is to evaluate the safety and efficacy of a single IV bolus injection of 90 μg/kg desmoteplase given three- to nine-hours after onset of ischemic stroke (National Institutes of Health Stroke Scale 4–24, age 18–85 years). The objective of DIAS-J is to evaluate the safety and tolerability of desmoteplase 70 and 90 μg/kg three- to nine-hours after ischemic stroke onset in Japanese patients. Methods Patients are included with occlusion or high-grade stenosis (thrombolysis in myocardial infarction 0–1) in proximal cerebral arteries on magnetic resonance or computed tomography angiography but excluded with extended ischemic edema on computed tomography or diffusion-weighted imaging. Conclusion Desmoteplase is the only thrombolytic agent in late-stage development for acute ischemic stroke that is now tested in patients with proven stroke pathology. The results of the Desmoteplase in Acute Ischemic Stroke clinical trial program will show whether patients with major artery occlusions but not extended ischemic brain damage can be safely and effectively treated up to nine-hours after onset.
Collapse
Affiliation(s)
- Rüdiger von Kummer
- Department of Neuroradiology, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Gregory W Albers
- Department of Neurology and Neurological Sciences, Stanford University Stroke Center, Palo Alto, CA, USA
| | - Etsuro Mori
- Department of Behavioural Neurology and Cognitive Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | |
Collapse
|
43
|
Glibenclamide Administration Attenuates Infarct Volume, Hemispheric Swelling, and Functional Impairments following Permanent Focal Cerebral Ischemia in Rats. Stroke Res Treat 2012; 2012:460909. [PMID: 22988544 PMCID: PMC3440943 DOI: 10.1155/2012/460909] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/01/2012] [Accepted: 08/02/2012] [Indexed: 12/22/2022] Open
Abstract
Studies from a single laboratory have shown that in rodent models of permanent stroke, administration of the sulfonylurea glibenclamide (Glib) is highly effective in reducing edema, mortality, and lesion volume. The Stroke Therapy Academic Industry Roundtable (STAIR) recommends that new acute treatments for ischemic stroke to be replicated across different laboratories. Accordingly, we examined the effect of low-dose Glib in a permanent suture occlusion model of stroke. Male Sprague-Dawley rats underwent permanent middle cerebral artery occlusion (pMCAO) followed by an initial intraperitoneal injection of Glib (10 μg/kg) and the start of a constant infusion (200 ng/h) via miniosmotic pump at the onset of ischemia. Functional deficits were assessed by Neurological Severity Score (NSS) and grip-strength meter at 24 and 48 h after pMCAO. Glib-treated rats showed a significant reduction in infarct volume, lower NSS, and less hemispheric swelling compared to vehicle. Grip strength was decreased significantly in pMCAO rats compared to shams and significantly improved by treatment with Glib. Taken together, these data indicate that Glib has strong neuroprotective effects following ischemic stroke and may warrant further testing in future clinical trials for human stroke.
Collapse
|
44
|
O'Collins VE, Macleod MR, Donnan GA, Howells DW. Evaluation of combination therapy in animal models of cerebral ischemia. J Cereb Blood Flow Metab 2012; 32:585-97. [PMID: 22293990 PMCID: PMC3318154 DOI: 10.1038/jcbfm.2011.203] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 12/23/2011] [Indexed: 12/31/2022]
Abstract
Combination therapy has been identified as a promising strategy to improve stroke management. We conducted a systematic review and meta-analysis of evidence from animal models of ischemic stroke to determine whether combining treatments improved efficacy. Multiple databases were searched and data were extracted from focal ischemia experiments comparing control groups, single treatments, and combination treatments. Of 11,430 papers identified, 142 met the inclusion criteria; these tested 126 treatments in 373 experiments using 8,037 animals (I(2)=85 to 96%). Taken together, single treatments reduced infarct size by 20% and improved neurological score by 12% compared with control; a second therapy improved efficacy by an additional 18% and 25%, respectively. Publication bias may affect combination efficacy for infarct size but not neurological score. Combining thrombolysis with other therapies may extend the time window from 4.4 to 8 hours in animal models, although testing beyond 6 hours is required to confirm this. Benefits of additional therapy decreased as the efficacy of the primary treatment increased, with combination efficacy reaching a ceiling at 60% to 80% protection. Combining treatments may bring benefits and extend the time window for treatment. More evidence is needed due to potential publication bias and heterogeneity.
Collapse
Affiliation(s)
- Victoria E O'Collins
- Florey Neuroscience Institutes, Melbourne Brain Centre, Heidelberg, Victoria, Australia.
| | | | | | | |
Collapse
|
45
|
Ishrat T, Sayeed I, Atif F, Hua F, Stein DG. Progesterone is neuroprotective against ischemic brain injury through its effects on the phosphoinositide 3-kinase/protein kinase B signaling pathway. Neuroscience 2012; 210:442-50. [PMID: 22450229 DOI: 10.1016/j.neuroscience.2012.03.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 02/20/2012] [Accepted: 03/07/2012] [Indexed: 12/18/2022]
Abstract
We tested the hypothesis that the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) pathway mediates some of the neuroprotective effects of progesterone (PROG) after ischemic stroke. We examined whether PROG acting through the PI3K/Akt pathway could affect the expression of vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF). Rats underwent permanent focal cerebral ischemia by electrocoagulation and received intraperitoneal injections of PROG (8 mg/kg) or vehicle at 1 h post-occlusion and subcutaneous injections at 6, 24, and 48 h. PAkt/Akt levels, apoptosis and apoptosis-related proteins (phosphorylated Bcl-2-associated death promoter (pBAD), BAD, caspase-3, and cleaved caspase-3) were analyzed by TUNEL assays, Western blotting and immunohistochemistry at 24 h post-pMCAO. VEGF and BDNF were analyzed at 24, 72 h and 14 days post-pMCAO with Western blots. Following pMCAO, PROG treatment significantly (P<0.05) reduced ischemic lesion size and edema. Treatment with PROG significantly (P<0.05) decreased VEGF at 24 and 72 h but increased VEGF expression 14 days after injury. The treatment also increased BDNF, and attenuated apoptosis by increasing Akt phosphorylation compared with vehicle alone. The selective PI3K inhibitor wortmannin compromised PROG-induced neuroprotective effects and reduced the elevation of pAkt levels in the ischemic penumbra. Our findings lead us to suggest that the PI3K/Akt pathway can play a role in mediating the neuroprotective effects of PROG after stroke by altering the expression of trophic factors in the brain.
Collapse
Affiliation(s)
- T Ishrat
- Department of Emergency Medicine, Emory University School of Medicine, 1365 B Clifton Road, Suite 5100, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
46
|
Heiss WD, Brainin M, Bornstein NM, Tuomilehto J, Hong Z. Cerebrolysin in patients with acute ischemic stroke in Asia: results of a double-blind, placebo-controlled randomized trial. Stroke 2012; 43:630-6. [PMID: 22282884 DOI: 10.1161/strokeaha.111.628537] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Cerebrolysin showed neuroprotective and neurotrophic properties in various preclinical models of ischemia and small clinical trials. The aim of this large double-blind, placebo-controlled randomized clinical trial was to test its efficacy and safety in patients with acute ischemic stroke. METHODS Patients with acute ischemic hemispheric stroke were randomized within 12 hours of symptoms onset to active treatment (30 mL Cerebrolysin daily) or placebo (saline solution) given as intravenous infusion for 10 days in addition to aspirin (100 mg daily). The patients were followed up to 90 days. The primary end point was the result of a combined global directional test of modified Rankin Scale, Barthel Index, and National Institutes of Health Stroke Scale. Adverse events were documented to assess safety. RESULTS A total of 1070 patients were enrolled in this study. Five hundred twenty-nine patients were assigned to Cerebrolysin and 541 to placebo. The confirmatory end point showed no significant difference between the treatment groups. When stratified by severity however, a post hoc analysis of National Institutes of Health Stroke Scale and modified Rankin Scale showed a trend in favor of Cerebrolysin in patients with National Institutes of Health Stroke Scale >12 (National Institutes of Health Stroke Scale: OR, 1.27; CI lower bound, 0.97; modified Rankin Scale: OR, 1.27; CI lower bound, 0.90). In this subgroup, the cumulative mortality by 90 days was 20.2% in the placebo and 10.5% in the Cerebrolysin group (hazard ratio, 1.9661; CI lower bound, 1.0013). CONCLUSIONS In this study, the confirmatory end point showed neutral results between the treatment groups. However, a favorable outcome trend was seen in the severely affected patients with ischemic stroke treated with Cerebrolysin. This observation should be confirmed by a further clinical trial. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00868283.
Collapse
Affiliation(s)
- Wolf-Dieter Heiss
- Max-Planck Institut für Neurologie, Gleueler Strasse 50, 50931 Koeln, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Lapchak PA. Scientific Rigor Recommendations for Optimizing the Clinical Applicability of Translational Research. JOURNAL OF NEUROLOGY & NEUROPHYSIOLOGY 2012; 3:e111. [PMID: 24490120 PMCID: PMC3905455 DOI: 10.4172/2155-9562.1000e111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The approval of new therapies to treat neurodegenerative disease conditions by the Food and Drug administration (FDA) has been hindered by many failed clinical trials, which were based upon "significant" efficacy in preclinical or translational studies. Additional problems during drug development related to significant adverse events and unforeseen toxicity have also hampered drug development. Recent reviews of preclinical data suggests that many studies have over-estimated efficacy due to poor or inadequate study design, exclusion of important data (negative or neutral) and lack of study randomization and blinding. This article describes in detail a set of recommendations to improve the quality of science being conducted in laboratories worldwide, with the goal of documenting in the peer-reviewed literature, including Journal of Neurology and Neurophysiology, the scientific basis for the continued development of specific strategies to treat neurodegenerative diseases such as Stroke, Alzheimer's disease, Huntington's disease, Parkinson's disease, Spinal cord injury, and Amyotrophic lateral sclerosis. The minimum recommendations for effective translational research include the need for model justification, study group randomization and blinding, power analysis calculations, appropriate statistical analysis of all data sets, and a conflict of interest statement by investigators. It will also be beneficial to demonstrate reproducible efficacy in multiple species and in studies done by independent laboratories.
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
|
48
|
Sarwal A, Hussain MS, Shuaib A. Neuroprotection in Stroke. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
49
|
Hematopoietic Growth Factor Family for Stroke Drug Development. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
50
|
Ayer RE, Asgarzadie F. The incomplete picture of incomplete spinal cord injury. Transl Stroke Res 2011; 2:430-2. [PMID: 24323679 DOI: 10.1007/s12975-011-0134-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 10/27/2011] [Indexed: 11/30/2022]
Affiliation(s)
- Robert E Ayer
- Department of Neurosurgery, Loma Linda University Medical Center, 11234 Anderson Street, Rm. 2562-B, Loma Linda, CA, 92354, USA
| | | |
Collapse
|