1
|
Jacobson PB, Mothe A, Levy A, Krakovsky M, Hooker BA, Zhang X, Mollon J, Mordashova Y, Droescher M, Weiss S, Barghorn S, Dreher I, Awwad K, Nimmrich V, Huang L, Fung E, Buck WR, Pfleeger K, Ziemann A, Smith E, Fox GB, Tator CH, Gold M. Neutralizing RGMa with Elezanumab Promotes Cerebroprotection and Recovery in Rabbit Middle Cerebral Artery Occlusion. Transl Stroke Res 2024; 15:805-817. [PMID: 37326791 PMCID: PMC11226526 DOI: 10.1007/s12975-023-01164-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/17/2023]
Abstract
Repulsive guidance molecule A (RGMa) is an inhibitor of neuronal growth and survival which is upregulated in the damaged central nervous system following acute spinal cord injury (SCI), traumatic brain injury, acute ischemic stroke (AIS), and other neuropathological conditions. Neutralization of RGMa is neuroprotective and promotes neuroplasticity in several preclinical models of neurodegeneration and injury including multiple sclerosis, AIS, and SCI. Given the limitations of current treatments for AIS due to narrow time windows to intervention (TTI), and restrictive patient selection criteria, there is significant unmet need for therapeutic agents that enable tissue survival and repair following acute ischemic damage for a broader population of stroke patients. In this preclinical study, we evaluated whether elezanumab, a human anti-RGMa monoclonal antibody, could improve neuromotor function and modulate neuroinflammatory cell activation following AIS with delayed intervention times up to 24 h using a rabbit embolic permanent middle cerebral artery occlusion model (pMCAO). In two replicate 28-day pMCAO studies, weekly intravenous infusions of elezanumab, over a range of doses and TTIs of 6 and 24 h after stroke, significantly improved neuromotor function in both pMCAO studies when first administered 6 h after stroke. All elezanumab treatment groups, including the 24 h TTI group, had significantly less neuroinflammation as assessed by microglial and astrocyte activation. The novel mechanism of action and potential for expanding TTI in human AIS make elezanumab distinct from current acute reperfusion therapies, and support evaluation in clinical trials of acute CNS damage to determine optimal dose and TTI in humans.
Collapse
Affiliation(s)
- Peer B Jacobson
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd, North Chicago, IL, 60064, USA.
| | - Andrea Mothe
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, ON, M5T 0S8, Canada
| | | | | | - Bradley A Hooker
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Xiaomeng Zhang
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Jennifer Mollon
- Data and Statistical Sciences, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, 67061, KnollstrasseLudwigshafen, Germany
| | - Yulia Mordashova
- Data and Statistical Sciences, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, 67061, KnollstrasseLudwigshafen, Germany
| | - Mathias Droescher
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Sabine Weiss
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Stefan Barghorn
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Ingeborg Dreher
- Department of Drug Metabolism, Pharmacokinetics and Bioanalysis, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Khader Awwad
- Department of Drug Metabolism, Pharmacokinetics and Bioanalysis, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Volker Nimmrich
- Department of Drug Metabolism, Pharmacokinetics and Bioanalysis, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Lili Huang
- AbbVie Biologics, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Emma Fung
- AbbVie Biologics, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Wayne R Buck
- Preclinical Safety, AbbVie Inc, 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Kimberly Pfleeger
- Department of Neuroscience Development, AbbVie Inc, 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Adam Ziemann
- Department of Neuroscience Development, AbbVie Inc, 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Elaine Smith
- Department of Neuroscience Development, AbbVie Inc, 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Gerard B Fox
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - Charles H Tator
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, ON, M5T 0S8, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Michael Gold
- Department of Neuroscience Development, AbbVie Inc, 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| |
Collapse
|
2
|
Singh D, Wasan H, Reeta KH. Preclinical Stroke Research and Translational Failure: A Bird's Eye View on Preventable Variables. Cell Mol Neurobiol 2022; 42:2003-2017. [PMID: 33786698 PMCID: PMC11421600 DOI: 10.1007/s10571-021-01083-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/18/2021] [Indexed: 02/08/2023]
Abstract
Despite achieving remarkable success in understanding the cellular, molecular and pathophysiological aspects of stroke, translation from preclinical research has always remained an area of debate. Although thousands of experimental compounds have been reported to be neuro-protective, their failures in clinical setting have left the researchers and stakeholders in doldrums. Though the failures described have been excruciating, they also give us a chance to refocus on the shortcomings. For better translational value, evidences from preclinical studies should be robust and reliable. Preclinical study design has a plethora of variables affecting the study outcome. Hence, this review focusses on the factors to be considered for a well-planned preclinical study while adhering to guidelines with emphasis on the study design, commonly used animal models, their limitations with special attention on various preventable attritions including comorbidities, aged animals, time of dosing, outcome measures and physiological variables along with the concept of multicentric preclinical randomized controlled trials. Here, we provide an overview of a panorama of practical aspects, which could be implemented, so that a well-defined preclinical study would result in a neuro-protectant with better translational value.
Collapse
Affiliation(s)
- Devendra Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Himika Wasan
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
3
|
Lourbopoulos A, Mourouzis I, Xinaris C, Zerva N, Filippakis K, Pavlopoulos A, Pantos C. Translational Block in Stroke: A Constructive and "Out-of-the-Box" Reappraisal. Front Neurosci 2021; 15:652403. [PMID: 34054413 PMCID: PMC8160233 DOI: 10.3389/fnins.2021.652403] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Why can we still not translate preclinical research to clinical treatments for acute strokes? Despite > 1000 successful preclinical studies, drugs, and concepts for acute stroke, only two have reached clinical translation. This is the translational block. Yet, we continue to routinely model strokes using almost the same concepts we have used for over 30 years. Methodological improvements and criteria from the last decade have shed some light but have not solved the problem. In this conceptual analysis, we review the current status and reappraise it by thinking "out-of-the-box" and over the edges. As such, we query why other scientific fields have also faced the same translational failures, to find common denominators. In parallel, we query how migraine, multiple sclerosis, and hypothermia in hypoxic encephalopathy have achieved significant translation successes. Should we view ischemic stroke as a "chronic, relapsing, vascular" disease, then secondary prevention strategies are also a successful translation. Finally, based on the lessons learned, we propose how stroke should be modeled, and how preclinical and clinical scientists, editors, grant reviewers, and industry should reconsider their routine way of conducting research. Translational success for stroke treatments may eventually require a bold change with solutions that are outside of the box.
Collapse
Affiliation(s)
- Athanasios Lourbopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurointensive Care Unit, Schoen Klinik Bad Aibling, Bad Aibling, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilian University, Munich, Germany
| | - Iordanis Mourouzis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christodoulos Xinaris
- IRCCS – Istituto di Ricerche Farmacologiche ‘Mario Negri’, Centro Anna Maria Astori, Bergamo, Italy
- University of Nicosia Medical School, Nicosia, Cyprus
| | - Nefeli Zerva
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Filippakis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelos Pavlopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Pantos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
4
|
Kim Y, Lee YB, Bae SK, Oh SS, Choi JR. Development of a photochemical thrombosis investigation system to obtain a rabbit ischemic stroke model. Sci Rep 2021; 11:5787. [PMID: 33707580 PMCID: PMC7970995 DOI: 10.1038/s41598-021-85348-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/25/2021] [Indexed: 11/15/2022] Open
Abstract
Photochemical thrombosis is a method for the induction of ischemic stroke in the cerebral cortex. It can generate localized ischemic infarcts in the desired region; therefore, it has been actively employed in establishing an ischemic stroke animal model and in vivo assays of diagnostic and therapeutic techniques for stroke. To establish a rabbit ischemic stroke model and overcome the shortcoming of previous studies that were difficult to build a standardized photothrombotic rabbit model, we developed a photochemical thrombosis induction system that can produce consistent brain damage on a specific area. To verify the generation of photothrombotic brain damage using the system, longitudinal magnetic resonance imaging, 2,3,5-triphenyltetrazolium chloride staining, and histological staining were applied. These analytical methods have a high correlation for ischemic infarction and are appropriate for analyzing photothrombotic brain damage in the rabbit brain. The results indicated that the photothrombosis induction system has a main advantage of being accurately controlled a targeted region of photothrombosis and can produce cerebral hemisphere lesions on the target region of the rabbit brain. In conjugation with brain atlas, it can induce photochemical ischemic stroke locally in the part of the brain that is responsible for a particular brain function and the system can be used to develop animal models with degraded specific functions. Also, the photochemical thrombosis induction system and a standardized rabbit ischemic stroke model that uses this system have the potential to be used for verifications of biomedical techniques for ischemic stroke at a preclinical stage in parallel with further performance improvements.
Collapse
Affiliation(s)
- Yoonhee Kim
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Yoon Bum Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Seung Kuk Bae
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu, 41566, Korea
| | - Sung Suk Oh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea.
| | - Jong-Ryul Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea.
| |
Collapse
|
5
|
Bach A, Clausen BH, Kristensen LK, Andersen MG, Ellman DG, Hansen PB, Hasseldam H, Heitz M, Özcelik D, Tuck EJ, Kopanitsa MV, Grant SG, Lykke-Hartmann K, Johansen FF, Lambertsen KL, Strømgaard K. Selectivity, efficacy and toxicity studies of UCCB01-144, a dimeric neuroprotective PSD-95 inhibitor. Neuropharmacology 2019; 150:100-111. [DOI: 10.1016/j.neuropharm.2019.02.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/17/2019] [Accepted: 02/26/2019] [Indexed: 01/09/2023]
|
6
|
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
|
7
|
Intravenous xenogeneic human cardiosphere-derived cell extracellular vesicles (exosomes) improves behavioral function in small-clot embolized rabbits. Exp Neurol 2018; 307:109-117. [DOI: 10.1016/j.expneurol.2018.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022]
|
8
|
Hou H, Khan N, Gohain S, Eskey CJ, Moodie KL, Maurer KJ, Swartz HM, Kuppusamy P. Dynamic EPR Oximetry of Changes in Intracerebral Oxygen Tension During Induced Thromboembolism. Cell Biochem Biophys 2017; 75:285-294. [PMID: 28434138 DOI: 10.1007/s12013-017-0798-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Cerebral tissue oxygenation (oxygen tension, pO2) is a critical parameter that is closely linked to brain metabolism, function, and pathophysiology. In this work, we have used electron paramagnetic resonance oximetry with a deep-tissue multi-site oxygen-sensing probe, called implantable resonator, to monitor temporal changes in cerebral pO2 simultaneously at four sites in a rabbit model of ischemic stroke induced by embolic clot. The pO2 values in healthy brain were not significantly different among the four sites measured over a period of 4 weeks. During exposure to 15% O2 (hypoxia), a sudden and significant decrease in pO2 was observed in all four sites. On the other hand, brief exposure to breathing carbogen gas (95% O2 + 5% CO2) showed a significant increase in the cerebral pO2 from baseline value. During ischemic stroke, induced by embolic clot in the left brain, a significant decline in the pO2 of the left cortex (ischemic core) was observed without any change in the contralateral sites. While the pO2 in the non-infarct regions returned to baseline at 24-h post-stroke, pO2 in the infarct core was consistently lower compared to the baseline and other regions of the brain. The results demonstrated that electron paramagnetic resonance oximetry with the implantable resonator can repeatedly and simultaneously report temporal changes in cerebral pO2 at multiple sites. This oximetry approach can be used to develop interventions to rescue hypoxic/ischemic tissue by modulating cerebral pO2 during hypoxic and stroke injury.
Collapse
Affiliation(s)
- Huagang Hou
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Nadeem Khan
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Sangeeta Gohain
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Clifford J Eskey
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Karen L Moodie
- Center for Comparative Medicine and Research, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Kirk J Maurer
- Center for Comparative Medicine and Research, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Harold M Swartz
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA
| | - Periannan Kuppusamy
- Department of Radiology, The Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive,, Lebanon, 03756, NH, USA.
| |
Collapse
|
9
|
Translational Stroke Research Opportunities and a Strategy to Develop Effective Cytoprotection. Transl Stroke Res 2017; 8:318-321. [DOI: 10.1007/s12975-017-0529-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 12/21/2022]
|
10
|
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]
|
11
|
|
12
|
Kent TA, Mandava P. Embracing Biological and Methodological Variance in a New Approach to Pre-Clinical Stroke Testing. Transl Stroke Res 2016; 7:274-83. [PMID: 27018014 PMCID: PMC5425098 DOI: 10.1007/s12975-016-0463-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/08/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Abstract
High-profile failures in stroke clinical trials have discouraged clinical translation of neuroprotectants. While there are several plausible explanations for these failures, we believe that the fundamental problem is the way clinical and pre-clinical studies are designed and analyzed for heterogeneous disorders such as stroke due to innate biological and methodological variability that current methods cannot capture. Recent efforts to address pre-clinical rigor and design, while important, are unable to account for variability present even in genetically homogenous rodents. Indeed, efforts to minimize variability may lessen the clinical relevance of pre-clinical models. We propose a new approach that recognizes the important role of baseline stroke severity and other factors in influencing outcome. Analogous to clinical trials, we propose reporting baseline factors that influence outcome and then adapting for the pre-clinical setting a method developed for clinical trial analysis where the influence of baseline factors is mathematically modeled and the variance quantified. A new therapy's effectiveness is then evaluated relative to the pooled outcome variance at its own baseline conditions. In this way, an objective threshold for robustness can be established that must be overcome to suggest its effectiveness when expanded to broader populations outside of the controlled environment of the PI's laboratory. The method is model neutral and subsumes sources of variance as reflected in baseline factors such as initial stroke severity. We propose that this new approach deserves consideration for providing an objective method to select agents worthy of the commitment of time and resources in translation to clinical trials.
Collapse
Affiliation(s)
- Thomas A Kent
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, McNair Campus, 7200 Cambridge St. 9th Floor, MS: BCM609, Houston, TX, 77030, USA.
- Michael E. DeBakey VA Medical Center Stroke Program and Center for Translational Research on Inflammatory Diseases, Houston, TX, USA.
| | - Pitchaiah Mandava
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, McNair Campus, 7200 Cambridge St. 9th Floor, MS: BCM609, Houston, TX, 77030, USA
- Michael E. DeBakey VA Medical Center Stroke Program and Center for Translational Research on Inflammatory Diseases, Houston, TX, USA
| |
Collapse
|
13
|
Lapchak PA, Boitano PD. A novel method to promote behavioral improvement and enhance mitochondrial function following an embolic stroke. Brain Res 2016; 1646:125-131. [PMID: 27180104 DOI: 10.1016/j.brainres.2016.04.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/05/2016] [Accepted: 04/16/2016] [Indexed: 10/21/2022]
Abstract
Tissue plasminogen activator (tPA) is the only FDA-approved treatment for stroke; tPA increases cerebral reperfusion, blood flow and improved behavior. Novel transcranial laser therapy (TLT) also enhances cerebral blood flow and activates mitochondrial function. Using the rabbit small clot embolic stroke model (RSCEM), we studied the effects of continuous wave TLT (7.5mW/cm(2)) alone or in combination with standardized intravenous (IV) tPA (3.3mg/kg) applied 1h post-embolization on 3 endpoints: 1) behavioral function measured 2 days [effective stroke dose (P50 in mg) producing neurological deficits in 50% of embolized rabbits], 2) intracerebral hemorrhage (ICH) rate, and 3) cortical adenosine-5'-triphosphate (ATP) content was measured 6h following embolization. TLT and tPA significantly (p<0.05) increased P50 values by 95% and 56% (p<0.05), respectively over control. TLT-tPA increased P50 by 136% over control (p<0.05). Embolization reduced cortical ATP content by 39%; decreases that were attenuated by either TLT or tPA treatment (p<0.05). TLT-tPA further enhanced cortical ATP levels 22% above that measured in naïve control. TLT and tPA both effectively and safely, without affecting ICH rate, improved behavioral outcome in embolized rabbits; and there was a trend (p>0.05) for the TLT-tPA combination to further increase P50. TLT and tPA both attenuated stroke-induced ATP deficits, and the combination of tPA and TLT produced an additive effect on ATP levels. This study demonstrates that the combination of TLT-tPA enhances ATP production, and suggests that tPA-induced reperfusion in combination with TLT neuroprotection therapy may optimally protect viable cells in the cortex measured using ATP levels as a marker.
Collapse
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center Department of Neurology & Neurosurgery, Advanced Health Sciences Pavilion Suite 8305, 127 South San Vicente Blvd, Los Angeles 90048, United States.
| | - Paul D Boitano
- Cedars-Sinai Medical Center Department of Neurology & Neurosurgery, Advanced Health Sciences Pavilion Suite 8305, 127 South San Vicente Blvd, Los Angeles 90048, United States.
| |
Collapse
|
14
|
Ma Y, Li L, Niu Z, Song J, Lin Y, Zhang H, Du G. Effect of recombinant plasminogen activator timing on thrombolysis in a novel rat embolic stroke model. Pharmacol Res 2016; 107:291-299. [DOI: 10.1016/j.phrs.2016.03.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/19/2016] [Accepted: 03/27/2016] [Indexed: 01/09/2023]
|
15
|
Lapchak PA. Critical early thrombolytic and endovascular reperfusion therapy for acute ischemic stroke victims: a call for adjunct neuroprotection. Transl Stroke Res 2015; 6:345-54. [PMID: 26314402 PMCID: PMC4568436 DOI: 10.1007/s12975-015-0419-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 08/06/2015] [Indexed: 12/28/2022]
Abstract
Today, there is an enormous amount of excitement in the field of stroke victim care due to the recent success of MR. CLEAN, SWIFT PRIME, ESCAPE, EXTEND-IA, and REVASCAT endovascular trials. Successful intravenous (IV) recombinant tissue plasminogen activator (rt-PA) clinical trials [i.e., National Institute of Neurological Disorders and Stroke (NINDS) rt-PA trial, Third European Cooperative Acute Stroke Study (ECASSIII), and Third International Stroke study (IST-3)] also need to be emphasized. In the recent endovascular and thrombolytic trials, there is statistically significant improvement using both the National Institutes of Health Stroke Scale (NIHSS) and the modified Rankin Score (mRS) scale, but neither approach promotes complete recovery in patients enrolled within any particular NIHSS or mRS score tier. Absolute improvement (mRS 0-2 at 90 days) with endovascular therapy is 13.5-31 %, whereas thrombolytics alone also significantly improve patient functional independence, but to a lesser degree (NINDS rt-PA trial 13 %). This article has 3 main goals: (1) first to emphasize the utility and cost-effectiveness of rt-PA to treat stroke; (2) second to review the recent endovascular trials with respect to efficacy, safety, and cost-effectiveness as a stroke treatment; and (3) to further consider and evaluate strategies to develop novel neuroprotective drugs. A thesis will be put forth so that future stroke trials and therapy development can optimally promote recovery so that stroke victims can return to "normal" life.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neurology & Neurosurgery, Cedars-Sinai Medical Center Advanced Healthcare Science Pavilion, 127 S. San Vicente Blvd., Suite 8305, Los Angeles, CA, 90048, USA,
| |
Collapse
|
16
|
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
|
17
|
Lapchak PA. A cost-effective rabbit embolic stroke bioassay: insight into the development of acute ischemic stroke therapy. Transl Stroke Res 2015; 6:99-103. [PMID: 25637174 PMCID: PMC4359071 DOI: 10.1007/s12975-015-0386-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 01/06/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Paul A Lapchak
- Departments of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion Suite 8305, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA,
| |
Collapse
|
18
|
Lapchak PA, Daley JT, Boitano PD. A blinded, randomized study of L-arginine in small clot embolized rabbits. Exp Neurol 2015; 266:143-6. [PMID: 25708986 PMCID: PMC4382386 DOI: 10.1016/j.expneurol.2015.02.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 02/05/2015] [Accepted: 02/11/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Tissue plasminogen activator (tPA) is administered to acute ischemic stroke victims in a vehicle formulation containing high concentrations of L-arginine (3.5g/100mg vial), a well-known nitric oxide synthase (NOS) substrate and precursor to nitric oxide (NO), as well as an enhancer of cerebral blood flow. METHODS We studied the effects of tPA vehicle compared to tPA (3.3mg/kg) formulated in the same vehicle containing L-arginine, normal saline or normal saline containing L-arginine, on behavioral function following small clot embolic strokes in rabbits using clinical rating scores and quantal analysis curves as the primary end point. Treatments were administered intravenously (1ml/kg; 20% bolus/80% infused over 30min) starting 1h following the injection of small-sized blood clots into the brain vasculature and terminal behavior was measured 2days following embolization. Behavioral rating scores were used to calculate the effective stroke dose (P50 in mg) that produces neurological deficits in 50% of the rabbits. RESULTS In this study, tPA significantly (p=0.001) improved behavior compared to all other treatments including tPA vehicle, saline and saline-L-arginine, increasing the P50 by 141% over tPA vehicle. Saline-L-arginine was not significantly different from either saline or tPA vehicle (p>0.05). CONCLUSION This study demonstrates that the L-arginine component of the tPA vehicle does not contribute to the reproducible clinical improvement observed following tPA administration in rabbits. Moreover, the administration of L-arginine was not an effective method to promote behavioral recovery following embolic strokes in the stringent rabbit small clot stroke model, nor did L-arginine exacerbate behavioral deficits or intracerebral hemorrhage in embolized rabbits.
Collapse
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Department of Neurology & Neurosurgery.
| | | | | |
Collapse
|
19
|
Khan N, Hou H, Eskey CJ, Moodie K, Gohain S, Du G, Hodge S, Culp WC, Kuppusamy P, Swartz HM. Deep-tissue oxygen monitoring in the brain of rabbits for stroke research. Stroke 2015; 46:e62-6. [PMID: 25613304 DOI: 10.1161/strokeaha.114.007324] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Nadeem Khan
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.).
| | - Huagang Hou
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Clifford J Eskey
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Karen Moodie
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Sangeeta Gohain
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Gaixin Du
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Sassan Hodge
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - William C Culp
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Periannan Kuppusamy
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| | - Harold M Swartz
- From the Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, NH (N.K., H.H., S.G., G.D., S.H., P.K., H.M.S.); Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH (N.K., H.H., S.G., P.K., H.M.S.); Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH (C.J.E.); Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH (K.M.); and Department of Radiology, Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock (W.C.C.)
| |
Collapse
|
20
|
Wang Y, Yoshimura R, Manabe H, Schretter C, Clarke R, Cai Y, Fitzgerald M, Lee KS. Trans-sodium crocetinate improves outcomes in rodent models of occlusive and hemorrhagic stroke. Brain Res 2014; 1583:245-54. [PMID: 25128603 DOI: 10.1016/j.brainres.2014.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/28/2014] [Accepted: 08/07/2014] [Indexed: 11/25/2022]
Abstract
Trans-sodium crocetinate (TSC) is a novel carotenoid compound capable of enhancing the diffusion of small molecules in aqueous solutions. TSC improves the diffusion of oxygen and glucose, and increases oxygenation in ischemic brain tissue. TSC also dampens the intensity of an ischemic challenge during an ongoing ischemic event. The current study examined the impact of TSC in rat models of ischemic and hemorrhagic stroke. Rat three vessel occlusion (3VO), and combined 3VO and one vessel occlusion (3VO/1VO) models of ischemic stroke were evaluated for structural and behavioral outcomes. The effects of TSC were also tested in a rat model of intracerebral hemorrhage (ICH). Delayed treatment with TSC reduced infarct volume in a rodent model of transient focal ischemia involving either 2 or 6h of ischemia. Neurological outcomes, based on a multi-scale assessment and automated gait analysis, also were improved by TSC treatment. Additionally, TSC reduced edema and hemorrhagic volume in a rat model of ICH. An optimal therapeutic candidate for early intervention in ischemic stroke should be effective when administered on a delayed basis and should not aggravate outcomes associated with hemorrhagic stroke. The current findings demonstrate that delayed TSC treatment improves outcomes in experimental models of both ischemic and hemorrhagic stroke. Together, these findings suggest that TSC may be a safe and beneficial therapeutic modality for early stroke intervention, irrespective of the type of stroke involved.
Collapse
Affiliation(s)
- Yi Wang
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Ryo Yoshimura
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Hiroaki Manabe
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Catherine Schretter
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Ryon Clarke
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
| | - Yu Cai
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Mark Fitzgerald
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
| | - Kevin S Lee
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA; Department of Neurological Surgery, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
21
|
Lapchak PA, Boitano PD. Effect of the Pleiotropic Drug CNB-001 on Tissue Plasminogen Activator (tPA) Protease Activity in vitro: Support for Combination Therapy to Treat Acute Ischemic Stroke. JOURNAL OF NEUROLOGY & NEUROPHYSIOLOGY 2014; 5:214. [PMID: 25364620 PMCID: PMC4215513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Current state-of-the-art acute ischemic stroke clinical trials are designed to study neuroprotectants when administered following thrombolysis; tissue plasminogen activator (tPA) is administered to patients within 3-4.5 hours of an ischemic event. Thus, in order to develop a novel neuroprotectant and move it forward to a clinical trial, it is important to assess the effects of the drug on tPA's proteolytic activity in vitro, prior to extensive in vivo analysis. In this study, we determined if CNB-001 [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl)vinyl)-2-methoxy-phenol)], would affect, either enhance or inhibit tPA activity in vitro. In this tPA-inhibitor (plasminogen activator inhibitor-1; PAI-1 and 2,7-Bis-(4-Amidinobenzylidene)-Cycloheptan-1-One Dihydrochloride; tPA stop) controlled study, we used a chromogenic substrate (CH3SO2-D-hexahydrotyrosine-Gly-Arg-p-nitroanilide•AcOH) to study drug interactions in vitro, spectrophotometrically measuring protease released p-Nitroaniline from the substrate. We found that PAI-1 (0.25 μM) and tPA stop (5 μM) significantly (p<0.0001) inhibited substrate release, by 98.6% and 83.4%, respectively, thus inhibiting tPA activity in vitro. In comparison, CNB-001 (0.7-7 μM) reduced tPA activity by 28-32%, with an extrapolated IC50 value of 65.2-704 μM. Thus, although high concentrations of CNB-001 does affects tPA activity in vitro, the study supports the use of CNB-001 in combination with tPA to treat stroke, However, CNB-001 should be administered following thrombolysis to promote neuroprotection and repair.
Collapse
Affiliation(s)
- Paul A Lapchak
- Departments of Neurology Advanced Health Sciences Pavilion, Los Angeles, USA,Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Los Angeles, USA,Corresponding author: Paul A Lapchak, Director of Translational Research, Cedars-Sinai Medical Center Professor, Department of Neurology & Neurosurgery, Advanced Health Sciences Pavilion, Suite 8305, 127 S. San Vicente Blvd, Los Angeles, CA 90048, USA; Tel: 310-248-8188; Fax: 310-248-7568
| | - Paul D Boitano
- Departments of Neurology Advanced Health Sciences Pavilion, Los Angeles, USA
| |
Collapse
|
22
|
Fagan SC, Lapchak PA, Liebeskind DS, Ishrat T, Ergul A. Recommendations for preclinical research in hemorrhagic transformation. Transl Stroke Res 2014; 4:322-7. [PMID: 23730351 DOI: 10.1007/s12975-012-0222-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hemorrhagic transformation (HT) is an important complication of ischemic stroke and is responsible for most of the mortality associated with acute reperfusion therapy. Although many important publications address the preclinical models of ischemic stroke, there are no current recommendations on the conduct of research aimed at understanding the mechanisms and consequences of HT. The purpose of this review is to present the various models used in HT research, the clinical correlates, and the experimental variables known to influence the quantitation of HT in preclinical investigation. Lastly, recommendations for the conduct of preclinical research in HT are provided.
Collapse
Affiliation(s)
- Susan C Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA ; Charlie Norwood VA Medical Center, Augusta, GA ; Department of Neurology, Medical College of Georgia, Augusta, GA
| | | | | | | | | |
Collapse
|
23
|
Lapchak PA, Kikuchi K, Butte P, Hölscher T. Development of transcranial sonothrombolysis as an alternative stroke therapy: incremental scientific advances toward overcoming substantial barriers. Expert Rev Med Devices 2014; 10:201-13. [DOI: 10.1586/erd.12.88] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Lapchak PA, Schubert DR, Maher PA. De-Risking of Stilbazulenyl Nitrone (STAZN), a Lipophilic Nitrone to Treat Stroke Using a Unique Panel of In Vitro Assays. Transl Stroke Res 2013; 2:209-17. [PMID: 22003372 DOI: 10.1007/s12975-011-0071-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the present study, we used a comprehensive panel of in vitro assays to evaluate the efficacy and safety of stilbazulenyl nitrone (STAZN) as a lead compound to treat acute ischemic stroke. First, we measured neuroprotection in vitro using two different HT22 hippocampal nerve cell assays. Secondly, to de-risk drug development, we used CeeTox analysis with the H4IIE rat hepatoma cell line to determine the acute toxicity profile of STAZN. Third, STAZN was tested in microsomes from four species for measures of metabolic stability. Last, we determined the Ames test genotoxicity profile of STAZN using Salmonella typhimurium TA989 and TA100. In vitro, STAZN was neuroprotective against toxicity induced by iodoacetic acid, and oxytosis-induced glutathione depletion was initiated by glutamate, with an EC(50) value of 1-5 μM. Secondly, using CeeTox analysis, the estimated C(Tox) value (i.e., sustained concentration expected to produce toxicity in a rat 14-day repeat dose study) for STAZN was calculated to be 260 μM. Third, the half-life of STAZN in humans, dogs, and rats was 60-78 min. Last, the genotoxicity profile showed that STAZN did not induce bacterial colony growth under any conditions tested, indicating the lack of mutagenicity with this compound. STAZN appears to be a multi-target neuroprotective compound that has an excellent safety profile in both the CeeTox and Ames mutagenicity assays. STAZN may have significant potential as a novel neuroprotective agent to treat stroke and should be pursued in clinically relevant embolic stroke models.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neurology, Cedars-Sinai Medical Center, Davis Research Building, D-2091, 110 N. George Burns Road, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
25
|
Lapchak PA. Fast neuroprotection (fast-NPRX) for acute ischemic stroke victims: the time for treatment is now. Transl Stroke Res 2013; 4:704-9. [PMID: 24323424 DOI: 10.1007/s12975-013-0303-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 01/25/2023]
Affiliation(s)
- Paul A Lapchak
- Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Advanced Health Sciences Pavilion, Rm 8305, 127 S. San Vicente Blvd, Los Angeles, CA, 90048, USA,
| |
Collapse
|
26
|
Yu YY, Zheng XX, Bian TT, Li YJ, Wu XW, Yang DZ, Jiang SS, Tang DQ. Development and application of a LC-MS/MS assay for the simultaneous quantification of edaravone and taurine in beagle plasma. J Sep Sci 2013; 36:3837-44. [DOI: 10.1002/jssc.201300983] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 09/30/2013] [Accepted: 10/02/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Yan-yan Yu
- Department of Pharmaceutical Analysis; Xuzhou Medical College; Xuzhou Jiangsu China
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| | - Xiao-xiao Zheng
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| | - Ting-ting Bian
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| | - Yin-jie Li
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| | - Xiao-wen Wu
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| | - Dong-zhi Yang
- Department of Pharmaceutical Analysis; Xuzhou Medical College; Xuzhou Jiangsu China
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| | - Shui-shi Jiang
- Nanjing Yoko Pharmaceutical Co. Ltd; Nanjing Jiangsu China
| | - Dao-quan Tang
- Department of Pharmaceutical Analysis; Xuzhou Medical College; Xuzhou Jiangsu China
- Key Laboratory of New Drug and Clinical Application; Xuzhou Medical College; Xuzhou Jiangsu China
| |
Collapse
|
27
|
Effects of edaravone, a free radical scavenger, on photochemically induced cerebral infarction in a rat hemiplegic model. ScientificWorldJournal 2013; 2013:175280. [PMID: 23853531 PMCID: PMC3703327 DOI: 10.1155/2013/175280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/06/2013] [Indexed: 11/17/2022] Open
Abstract
Edaravone is a free radical scavenger that protects the adjacent cortex during cerebral infarction. We created a hemiparetic model of cerebral thrombosis from a photochemically induced infarction with the photosensitive dye, rose bengal, in rats. We examined the effects of edaravone on recovery in the model. A total of 36 adult Wistar rats were used. The right sensorimotor area was irradiated with green light with a wavelength of 533 nm (10 mm diameter), and the rose bengal was injected intravenously to create an infarction. The edaravone group was injected intraperitoneally with edaravone (3 mg/kg), and the control group was injected with saline. The recovery process of the hemiplegia was evaluated with the 7-step scale of Fenny. The infarcted areas were measured after fixation. The recovery of the paralysis in the edaravone-treated group was significantly earlier than that in the untreated group. Seven days later, both groups were mostly recovered and had scores of 7, and the infarction region was significantly smaller in the edaravone-treated group. Edaravone reduced the infarction area and promoted the functional recovery of hemiparesis from cerebral thrombosis in a rat model. These findings suggest that edaravone treatment would be effective in clinical patients recovering from cerebral infarction.
Collapse
|
28
|
Lapchak PA. Drug-like property profiling of novel neuroprotective compounds to treat acute ischemic stroke: guidelines to develop pleiotropic molecules. Transl Stroke Res 2013; 4:328-42. [PMID: 23687519 PMCID: PMC3653324 DOI: 10.1007/s12975-012-0200-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The development of novel neuroprotective compounds to treat acute ischemic stroke (AIS) has been problematic and quite complicated, since many candidates that have been tested clinically lacked significant pleiotropic activity, were unable to effectively cross the blood brain barrier (BBB), had poor bioavailability or were toxic. Moreover, the compounds did not confer significant neuroprotection or clinical efficacy measured using standard behavioral endpoints, when studied in clinical trials in a heterogeneous population of stroke patients. To circumvent some of the drug development problems describe above, we have used a rational funnel approach to identify and develop promising candidates. Using a step-wise approach, we have identified a series of compounds based upon two different neuroprotection assays. We have then taken the candidates and determined their "drug-like" properties. This guidelines article details in vitro screening assays used to show pleiotropic activity of a series of novel compounds; including enhanced neuroprotective activity compared to the parent compound fisetin. Moreover, for preliminary drug de-risking or risk reduction during development, we used compound assessment in the CeeTox assay, ADME toxicity using the AMES test for genotoxicity and interaction with Cytochrome P450 using CYP450 inhibition analysis against a spectrum of CYP450 enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4) as a measure of drug interaction. Moreover, the compounds have been studied using a transfected Madin Darby canine kidney (MDCK) cell assay to assess blood brain barrier penetration (BBB). Using this series of assays, we have identified 4 novel molecules to be developed as an AIS treatment.
Collapse
Affiliation(s)
- Paul A Lapchak
- Director of Translational Research, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, D-2091, 110 N. George Burns Road, Los Angeles, CA 90048 USA
| |
Collapse
|
29
|
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: 220] [Impact Index Per Article: 18.3] [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
|
30
|
Chueh JY, Kuhn AL, Wakhloo AK, Gounis MJ. Experimental Models of Vascular Occlusions for Evaluation of Thrombectomy Devices. Cardiovasc Eng Technol 2013. [DOI: 10.1007/s13239-013-0143-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
31
|
Lapchak PA, Doyan S, Fan X, Woods CM. Synergistic Effect of AJW200, a von Willebrand Factor Neutralizing Antibody with Low Dose (0.9 mg/mg) Thrombolytic Therapy Following Embolic Stroke in Rabbits. JOURNAL OF NEUROLOGY & NEUROPHYSIOLOGY 2013; 4:10.4172/2155-9562.1000146. [PMID: 24224121 PMCID: PMC3819228 DOI: 10.4172/2155-9562.1000146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The von Willebrand factor (vWF) is an acute stroke response protein involved in platelet aggregation, adhesion, inflammation, and thrombus formation, responses that occur following an ischemic stroke. We hypothesize that administration of an anti-vWF antibody (anti-vWF-Ab) may be used as adjunctive therapy with tissue plasminogen activator (tPA) to promote behavioral improvement following an embolic stroke. In this proof-of-concept study, which used a blinded and randomized design, we studied delayed treatment with the anti-vWF-Ab, AJW200 (0.30 mg/kg), alone or in combination with a rabbit low-dose of tPA (0.9 mg/kg) using the rabbit small clot embolic stroke model (RSCEM) with behavioral function as the primary clinically relevant endpoint. To evaluate the quantitative relationship between clot burden in brain and clinical scores, so that an effective stroke dose (P50) could be calculated, logistic sigmoidal quantal analysis curves were constructed. A beneficial treatment significantly increases P50 compared to control. The effect of antibody administration, either alone or with low dose tPA was compared to a "positive control", a standard rabbit optimized dose of tPA (3.3 mg/kg), as a measure of the maximum improvement potential in the RSCEM. The anti-vWF-Ab, AJW200, or control IgG were administered IV 1 hour following embolization, and behavior was measured 48 hours later. AJW200 plus low-dose tPA significantly increased the P50 value by 74% (p<0.05, t=2.612) and 81% (p<0.05, t=2.519) compared to low dose tPA or IgG, respectively, but not the AJW200 group (p>0.05). AJW200 increased the P50 value by 28%, (p>0.05) compared to the control IgG-treated group. Standard dose tPA increased the P50 value by 154% (p<0.05). Statistically, the combination response for AJW200 plus low-dose tPA was not significantly different from standard dose tPA (p=0.26). This study shows that the concomitant administration of the anti-vWF-Ab AJW200 with low dose tPA is synergistic and results in significantly improved behavioral function following embolic stroke. We postulate that neutralization of vWF may suppress or attenuate one or more aspects of the acute phase stroke cascade response including suppression of inflammatory response and reduced leukocyte adhesion.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Sarina Doyan
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Xiaomin Fan
- AvantGen Inc, 101051 Pacific Mesa Blvd., Ste 103, San Diego, USA
| | | |
Collapse
|
32
|
Lapchak PA. Recommendations and practices to optimize stroke therapy: developing effective translational research programs. Stroke 2013; 44:841-3. [PMID: 23391768 DOI: 10.1161/strokeaha.112.680439] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
33
|
[Near-infrared laser treatment of acute stroke: from bench to bedside]. DER NERVENARZT 2013; 83:966-74. [PMID: 22801662 DOI: 10.1007/s00115-012-3567-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Near-infrared laser therapy (NIRLT) as a transcranial laser therapy (TLT) is currently being investigated as a neuroreparatory and neuroprotective treatment for acute ischemic stroke patients in a pivotal phase III trial (NEST-3). In this review we cover the theoretical background, experimental studies, translational research and the clinical trial program.
Collapse
|
34
|
Lapchak PA, Bombien R, Rajput PS. J-147 a Novel Hydrazide Lead Compound to Treat Neurodegeneration: CeeTox ™ Safety and Genotoxicity Analysis. ACTA ACUST UNITED AC 2013; 4. [PMID: 25364619 PMCID: PMC4215638 DOI: 10.4172/2155-9562.1000158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
J-147 is a broad spectrum neuroprotective phenyl hydrazide compound with significant neurotrophic properties related to the induction of brain-derived neurotrophic factor (BDNF). Because this molecule is pleiotropic, it may have substantial utility in the treatment of a wide range of neurodegenerative diseases including acute ischemic stroke (AIS), traumatic brain injury(TBI), and Alzheimer’s disease(AD) where both neuroprotection and neurotrophism would be beneficial. Because of the pleiotropic actions of J-147, we sought to determine the safety profile of the drug using multiple assay analysis. For CeeTox analyses, we used a rat hepatoma cell line (H4IIE) resulted in estimated CTox value (i.e.: sustained concentration expected to produce toxicity in a 14 day repeat dosing study) of 90 μM for J-147. The CeeTox panel shows that J-147 produced some adverse effects on cellular activities, in particular mitochondrial function, but only with high concentrations of the drug. J-147 was also not genetoxic with or without Aroclor-1254 treatment. For J-147, based upon extensive neuroprotection assay data previously published, and the CeeTox assay (CTox value of 90 μM) in this study, we estimated in vitro neuroprotection efficacy (EC50 range 0.06–0.115 μM)/toxicity ratio is 782.6–1500 fold and the neurotrophism (EC50 range 0.025 μM)/toxicity ratio is 3600, suggesting that there is a significant therapeutic safety window for J-147 and that it should be further developed as a novel neuroprotective-neurotrophic agent to treat neurodegenerative disease taking into account current National Institute of Neurological Disorders and Stroke (NINDS) RIGOR guidelines.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Rene Bombien
- Division of Cardiothoracic Surgery, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Padmesh S Rajput
- Department of Neurology, Advanced Health Sciences Pavilion, Los Angeles, USA
| |
Collapse
|
35
|
|
36
|
Lapchak PA. Transcranial near-infrared laser therapy applied to promote clinical recovery in acute and chronic neurodegenerative diseases. Expert Rev Med Devices 2012; 9:71-83. [PMID: 22145842 DOI: 10.1586/erd.11.64] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
One of the most promising methods to treat neurodegeneration is noninvasive transcranial near-infrared laser therapy (NILT), which appears to promote acute neuroprotection by stimulating mitochondrial function, thereby increasing cellular energy production. NILT may also promote chronic neuronal function restoration via trophic factor-mediated plasticity changes or possibly neurogenesis. Clearly, NILT is a treatment that confers neuroprotection or neurorestoration using pleiotropic mechanisms. The most advanced application of NILT is for acute ischemic stroke based upon extensive preclinical and clinical studies. In laboratory settings, NILT is also being developed to treat traumatic brain injury, Alzheimer's disease and Parkinson's disease. There is some intriguing data in the literature that suggests that NILT may be a method to promote clinical improvement in neurodegenerative diseases where there is a common mechanistic component, mitochondrial dysfunction and energy impairment. This article will analyze and review data supporting the continued development of NILT to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Department of Neurology, Los Angeles, CA 90048, USA.
| |
Collapse
|
37
|
Clinical neuroprotective drugs for treatment and prevention of stroke. Int J Mol Sci 2012; 13:7739-7761. [PMID: 22837724 PMCID: PMC3397556 DOI: 10.3390/ijms13067739] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/15/2012] [Accepted: 06/19/2012] [Indexed: 01/12/2023] Open
Abstract
Stroke is an enormous public health problem with an imperative need for more effective therapies. In therapies for ischemic stroke, tissue plasminogen activators, antiplatelet agents and anticoagulants are used mainly for their antithrombotic effects. However, free radical scavengers, minocycline and growth factors have shown neuroprotective effects in the treatment of stroke, while antihypertensive drugs, lipid-lowering drugs and hypoglycemic drugs have shown beneficial effects for the prevention of stroke. In the present review, we evaluate the treatment and prevention of stroke in light of clinical studies and discuss new anti-stroke effects other than the main effects of drugs, focusing on optimal pharmacotherapy.
Collapse
|
38
|
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
|
39
|
Animal Models of Stroke for Preclinical Drug Development: A Comparative Study of Flavonols for Cytoprotection. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_25] [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]
|
40
|
Lapchak PA. CeeTox Analysis to De-risk Drug Development: The Three Antioxidants (NXY-059, Radicut, and STAZN). Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_31] [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]
|
41
|
The Translation Procedure of Low-Level Laser Therapy in Acute Ischemic Stroke: A Nonpharmaceutics Noninvasive Method. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
A Clinically Relevant Rabbit Embolic Stroke Model for Acute Ischemic Stroke Therapy Development: Mechanisms and Targets. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
|
44
|
Macrae IM. Preclinical stroke research--advantages and disadvantages of the most common rodent models of focal ischaemia. Br J Pharmacol 2011; 164:1062-78. [PMID: 21457227 PMCID: PMC3229752 DOI: 10.1111/j.1476-5381.2011.01398.x] [Citation(s) in RCA: 173] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 03/15/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022] Open
Abstract
This review describes the most commonly used rodent models and outcome measures in preclinical stroke research and discusses their strengths and limitations. Most models involve permanent or transient middle cerebral artery occlusion with therapeutic agents tested for their ability to reduce stroke-induced infarcts and improve neurological deficits. Many drugs have demonstrated preclinical efficacy but, other than thrombolytics, which restore blood flow, none have demonstrated efficacy in clinical trials. This failure to translate efficacy from bench to bedside is discussed alongside achievable steps to improve the ability of preclinical research to predict clinical efficacy: (i) Improvements in study quality and reporting. Study design must include randomization, blinding and predefined inclusion/exclusion criteria, and journal editors have the power to ensure statements on these and mortality data are included in preclinical publications. (ii) Negative and neutral studies must be published to enable preclinical meta-analyses and systematic reviews to more accurately predict drug efficacy in man. (iii) Preclinical groups should work within networks and agree on standardized procedures for assessing final infarct and functional outcome. This will improve research quality, timeliness and translational capacity. (iv) Greater uptake and improvements in non-invasive diagnostic imaging to detect and study potentially salvageable penumbral tissue, the target for acute neuroprotection. Drug effects on penumbra lifespan studied serially, followed by assessment of behavioural outcome and infarct within in the same animal group, will increase the power to detect drug efficacy preclinically. Similar progress in detecting drug efficacy clinically will follow from patient recruitment into acute stroke trials based on evidence of remaining penumbra.
Collapse
Affiliation(s)
- I M Macrae
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
45
|
Casals JB, Pieri NCG, Feitosa MLT, Ercolin ACM, Roballo KCS, Barreto RSN, Bressan FF, Martins DS, Miglino MA, Ambrósio CE. The use of animal models for stroke research: a review. Comp Med 2011; 61:305-313. [PMID: 22330245 PMCID: PMC3155396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 01/02/2010] [Accepted: 04/22/2011] [Indexed: 05/31/2023]
Abstract
Stroke has been identified as the second leading cause of death worldwide. Stroke is a focal neurologic deficit caused by a change in cerebral circulation. The use of animal models in recent years has improved our understanding of the physiopathology of this disease. Rats and mice are the most commonly used stroke models, but the demand for larger models, such as rabbits and even nonhuman primates, is increasing so as to better understand the disease and its treatment. Although the basic mechanisms of stroke are nearly identical among mammals, we here discuss the differences between the human encephalon and various animals. In addition, we compare common surgical techniques used to induce animal models of stroke. A more complete anatomic knowledge of the cerebral vessels of various model species is needed to develop more reliable models for objective results that improve knowledge of the pathology of stroke in both human and veterinary medicine.
Collapse
Affiliation(s)
- Juliana B Casals
- Veterinary Anatomy Section, College of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo
| | - Naira CG Pieri
- Veterinary Anatomy Section, College of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo
| | - Matheus LT Feitosa
- Veterinary Anatomy Section, College of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo
| | - Anna CM Ercolin
- Department of Basic Sciences, College of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Kelly CS Roballo
- Department of Basic Sciences, College of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Rodrigo SN Barreto
- Veterinary Anatomy Section, College of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo
| | - Fabiana F Bressan
- Department of Basic Sciences, College of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Daniele S Martins
- Department of Basic Sciences, College of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Maria A Miglino
- Veterinary Anatomy Section, College of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo
| | - Carlos E Ambrósio
- Department of Basic Sciences, College of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| |
Collapse
|
46
|
Affiliation(s)
- Paul A. Lapchak
- Translational Research, Cedars-Sinai Medical Center, Department of Neurology, Burns and Allen Research Institute, Davis Research Building, Room D-2091, 110 N. George Burns Road, Los Angeles, CA 90048, USA
| |
Collapse
|
47
|
Silver JH, Lapchak PA. Continuous monitoring of changes in plasma nitrite following cerebral ischemia in a rabbit embolic stroke model. Transl Stroke Res 2011; 2:218-26. [PMID: 21625287 PMCID: PMC3103225 DOI: 10.1007/s12975-011-0073-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this proof-of-concept study, we investigated direct, continuous monitoring of plasma nitrite as an indicator of cerebral ischemia following clot embolization of rabbits via an indwelling carotid catheter. Two groups of rabbits were studied to compare the effects of embolization on nitrite levels. In the control group, blood was continuously obtained from a jugular venous catheter. The blood was immediately passed through an ultrafiltration filter; the filtrate was chemically reduced to convert free nitrite to nitric oxide (NO) and then measured using a NO-specific electrode. In the embolized group, after a baseline nitrite level was achieved, blood clots were injected into the brain via the carotid artery catheter, and then nitrite levels were continuously measured from jugular venous blood. The stroke group showed a significantly greater increase in nitrite as compared to controls (p=0.017). Using the area-under-the-curve (AUC) method, results reached statistical significance (p<0.05) within 3 min of embolization. In embolized rabbits, NO(2) levels increased 424±256% compared to baseline. This study shows that nitrite can be measured immediately following a stroke and that our system measures nitrite independent of the extent of the stroke. This study provides evidence for the feasibility of using nitrite as a marker of ischemic stroke.
Collapse
Affiliation(s)
- James H. Silver
- Silver Medical Inc., 45 Roosevelt Circle, Palo Alto, CA 94306, USA,
| | - Paul A. Lapchak
- Department of Neurology, Cedars-Sinai Medical Center, 110N George Burns Road, D-2091, Los Angeles, CA 90048, USA,
| |
Collapse
|
48
|
Turner RJ, Jickling GC, Sharp FR. Are Underlying Assumptions of Current Animal Models of Human Stroke Correct: from STAIRs to High Hurdles? Transl Stroke Res 2011; 2:138-43. [PMID: 21654913 PMCID: PMC3085747 DOI: 10.1007/s12975-011-0067-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 01/18/2011] [Accepted: 01/20/2011] [Indexed: 10/29/2022]
Abstract
Animal models of acute ischemic stroke have been criticized for failing to translate to human stroke. Nevertheless, animal models are necessary to improve our understanding of stroke pathophysiology and to guide the development of new stroke therapies. The rabbit embolic clot model is one animal model that has led to an effective therapy in human acute ischemic stroke, namely tissue plasminogen activator (tPA). We propose that potential compounds that demonstrate efficacy in non-rabbit animal models of acute ischemic stroke should also be tested in the rabbit embolic blood clot model and, where appropriate, compared to tPA prior to investigation in humans. Furthermore, the use of anesthesia needs to be considered as a major confounder in animal models of acute ischemic stroke, and death should be included as an outcome measure in animal stroke studies. These steps, along with the current STAIRs recommendations, may improve the successful translation of experimental therapies to clinical stroke treatments.
Collapse
Affiliation(s)
- Renée J. Turner
- Department of Neurology, University of California at Davis, Sacramento, CA 95817 USA
- M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817 USA
- Discipline of Pathology, The University of Adelaide, North Terrace, Adelaide, 5005 SA Australia
| | - Glen C. Jickling
- Department of Neurology, University of California at Davis, Sacramento, CA 95817 USA
- M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817 USA
| | - Frank R. Sharp
- Department of Neurology, University of California at Davis, Sacramento, CA 95817 USA
- M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817 USA
| |
Collapse
|
49
|
Lapchak PA, Zhang JH. Resolving the negative data publication dilemma in translational stroke research. Transl Stroke Res 2011; 2:1-6. [PMID: 21666822 PMCID: PMC3109983 DOI: 10.1007/s12975-010-0057-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Paul A. Lapchak
- Translational Research, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, Room D-2091, 110N. George Burns Road, Los Angeles, CA 90048, USA
| | - John H. Zhang
- Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
50
|
Lapchak PA, McKim JM. CeeTox™ Analysis of CNB-001 a Novel Curcumin-Based Neurotrophic/Neuroprotective Lead Compound to Treat Stroke: Comparison with NXY-059 and Radicut. Transl Stroke Res 2011; 2:51-9. [PMID: 21494575 PMCID: PMC3074116 DOI: 10.1007/s12975-010-0034-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the present study, we used a comprehensive cellular toxicity (CeeTox) analysis panel to determine the toxicity profile for CNB-001 [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl)vinyl)-2-methoxy-phenol)], which is a hybrid molecule created by combining cyclohexyl bisphenol A, a molecule with neurotrophic activity and curcumin, a spice with neuro-protective activity. CNB-001 is a lead development compound since we have recently shown that CNB-001 has significant preclinical efficacy both in vitro and in vivo. In this study, we compared the CeeTox profile of CNB-001 with two neuroprotective molecules that have been clinically tested for efficacy: the hydrophilic free radical spin trap agent NXY-059 and the hydrophobic free radical scavenger edaravone (Radicut). CeeTox analyses using a rat hepatoma cell line (H4IIE) resulted in estimated C(Tox) value (i.e., sustained concentration expected to produce toxicity in a rat 14-day repeat dose study) of 42 μM for CNB-001 compared with >300 μM for both NXY-059 and Radicut. The CeeTox panel suggests that CNB-001 produces some adverse effects on cellular adenosine triphosphate content, membrane toxicity, glutathione content, and cell mass (or number), but only with high concentrations of the drug. After a 24-h exposure, the drug concentration that produced a half-maximal response (TC(50)) on the measures noted above ranges from 55 to 193 μM. Moreover, all CNB-001-induced changes in the markers were coincident with loss of cell number, prior to acute cell death as measured by membrane integrity, suggesting a cytostatic effect of CNB-001. NXY-059 and Radicut did not have acute toxic effects on H4IIE cells. We also found that CNB-001 resulted in an inhibition of ethoxyresorufin-o-deethylase activity, indicating that the drug may affect cytochrome P4501A activity and that CNB-001 was metabolically unstable using a rat microsome assay system. For CNB-001, an estimated in vitro efficacy/toxicity ratio is 183-643-fold, suggesting that there is a significant therapeutic safety window for CNB-001 and that it should be further developed as a novel neuroprotective agent to treat stroke.
Collapse
Affiliation(s)
- Paul A. Lapchak
- Department of Neurology, Cedars-Sinai Medical Center, Burns and Allen Research Institute, 110 N. George Burns Road, D-2091, Los Angeles, CA 90048, USA
| | | |
Collapse
|