1
|
You S, Zheng D, Chen X, Wang X, Ouyang M, Han Q, Cao Y, Delcourt C, Song L, Carcel C, Arima H, Liu CF, Lindley RI, Robinson T, Anderson CS, Chalmers J. Subacute Neurological Improvement Predicts Favorable Functional Recovery After Intracerebral Hemorrhage: INTERACT2 Study. Stroke 2025; 56:621-627. [PMID: 39895502 DOI: 10.1161/strokeaha.124.048847] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND The frequency and prognostic significance of subacute neurological improvement (SNI) on 90-day outcomes after acute intracerebral hemorrhage are unknown. METHODS Secondary analyses of participant data from the INTERACT2 trial (Second Intensive Blood Pressure Reduction in Acute Intracerebral Hemorrhage Trial). SNI included any, moderate, significant, and substantial neurological improvement defined as ≥1, ≥2, ≥3, and ≥4 points decrease, respectively, on the National Institutes of Health Stroke Scale from 24 hours to 7 days after intracerebral hemorrhage. Logistic regression models were used to assess associations of SNI and death or major disability (modified Rankin Scale score of 3-6), major disability (modified Rankin Scale scores, 3-5), and death alone at 90 days. Data are reported as odds ratios and 95% CIs. RESULTS Of 2571 patients included in analyses, 1492 (58.0%), 1057 (41.1%), 731 (28.4%), and 490 (19.1%) patients experienced any, moderate, significant, and substantial SNI (24 hours to 7 days) after intracerebral hemorrhage, respectively. After adjustment for key confounders, any SNI was associated with 49%, 25%, and 65% reduced odds of death or major disability (odds ratio, 0.51 [95% CI, 0.42-0.63]), major disability alone (odds ratio, 0.75 [95% CI, 0.63-0.90]), and death (odds ratio, 0.35 [95% CI, 0.24-0.50]), respectively. Moderate, significant, and substantial SNI were also significantly associated with decreased odds of death or major disability at 90 days. The relationship between any SNI and study outcomes was consistent in most subgroups, including age and baseline hematoma volume. Early intensive blood pressure-lowering treatment did not increase the odds of SNI. CONCLUSIONS SNI from 24 hours to 7 days is common after intracerebral hemorrhage and predicts a lower likelihood of death or major disability. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT00716079.
Collapse
Affiliation(s)
- Shoujiang You
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of SooChow University, Suzhou, China (S.Y., Y.C., C.-F.L.)
| | - Danni Zheng
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
| | - Xiaoying Chen
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
| | - Xia Wang
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
| | - Menglu Ouyang
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
| | - Qiao Han
- Department of Neurology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, China (Q.H.)
| | - Yongjun Cao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of SooChow University, Suzhou, China (S.Y., Y.C., C.-F.L.)
| | - Candice Delcourt
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
- Macquarie University, Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Sydney, New South Wales, Australia (C.D.)
| | - Lili Song
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
- The Institute of Science and Technology for Brain-Inspired Research, Fudan University, Shanghai, China (L.S., C.S.A.)
| | - Cheryl Carcel
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
| | - Hisatomi Arima
- Department of Preventive Medicine and Public Health, Fukuoka University, Japan (H.A.)
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of SooChow University, Suzhou, China (S.Y., Y.C., C.-F.L.)
| | - Richard I Lindley
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
- Westmead Clinical School, University of Sydney, New South Wales, Australia (R.I.L.)
| | - Thompson Robinson
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, United Kingdom (T.R.)
| | - Craig S Anderson
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
- The Institute of Science and Technology for Brain-Inspired Research, Fudan University, Shanghai, China (L.S., C.S.A.)
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (C.S.A.)
| | - John Chalmers
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (D.Z., X.C., X.W., M.O., C.D., L.S., C.C., R.I.L., C.S.A., J.C.)
| |
Collapse
|
2
|
Wang H, Dingledine RJ, Myers SJ, Traynelis SF, Fang C, Tan Y, Koszalka GW, Laskowitz DT. Clinical development of the GluN2B-selective NMDA receptor inhibitor NP10679 for the treatment of neurologic deficit after subarachnoid hemorrhage. J Pharmacol Exp Ther 2025; 392:100046. [PMID: 39892986 DOI: 10.1124/jpet.124.002334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) may be associated with cerebral vasospasm, which can lead to delayed cerebral ischemia, infarction, and worsened functional outcomes. The delayed nature of cerebral ischemia secondary to SAH-related vasculopathy presents a window of opportunity for the evaluation of well tolerated neuroprotective agents administered soon after ictus. Secondary ischemic injury in SAH is associated with increased extracellular glutamate, which can overactivate N-methyl-d-aspartate receptors (NMDARs), thereby triggering NMDAR-mediated cellular damage. In this study, we evaluated the effect of the pH-sensitive GluN2B-selective NMDAR inhibitor NP10679 on neurologic impairment after SAH. This compound demonstrates a selective increase in potency at the acidic extracellular pH levels that occur in the setting of ischemia. We found that NP10679 produced durable improvement of behavioral deficits in a well characterized murine model of SAH, and these effects were greater than those produced by nimodipine alone, the current standard of care. In addition, we observed an unexpected reduction in SAH-induced luminal narrowing of the middle cerebral artery. Neither nimodipine nor NP10679 alters each other's pharmacokinetic profile, suggesting no obvious drug-drug interactions. Based on allometric scaling of both toxicological and efficacy data, the therapeutic margin in humans should be at least 2. These results further demonstrate the utility of pH-dependent neuroprotective agents and GluN2B-selective NMDAR inhibitors as potential therapeutic strategies for the treatment of aneurysmal SAH. SIGNIFICANCE STATEMENT: This report describes the properties and utility of the GluN2B-selective pH-sensitive N-methyl-d-aspartate receptor inhibitor, NP10679, in a well characterized rodent model of subarachnoid hemorrhage. We show that the administration of NP10679 improves long-term neurological function following subarachnoid hemorrhage and that in rats, there are no drug-drug interactions between NP10679 and nimodipine, the standard of care for this indication.
Collapse
Affiliation(s)
- Haichen Wang
- Department of Neurology, Duke University, Durham, North Carolina
| | - Raymond J Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia; Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia
| | - Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia; NeurOp Inc, Atlanta, Georgia
| | - Chuan Fang
- Department of Neurology, Duke University, Durham, North Carolina; Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Yanli Tan
- Department of Neurology, Duke University, Durham, North Carolina; Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | | | | |
Collapse
|
3
|
Culkins C, Adomanis R, Phan N, Robinson B, Slaton E, Lothrop E, Chen Y, Kimmel BR. Unlocking the Gates: Therapeutic Agents for Noninvasive Drug Delivery Across the Blood-Brain Barrier. Mol Pharm 2024; 21:5430-5454. [PMID: 39324552 DOI: 10.1021/acs.molpharmaceut.4c00604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective network of various cell types that acts as a filter between the blood and the brain parenchyma. Because of this, the BBB remains a major obstacle for drug delivery to the central nervous system (CNS). In recent years, there has been a focus on developing various modifiable platforms, such as monoclonal antibodies (mAbs), nanobodies (Nbs), peptides, and nanoparticles, as both therapeutic agents and carriers for targeted drug delivery to treat brain cancers and diseases. Methods for bypassing the BBB can be invasive or noninvasive. Invasive techniques, such as transient disruption of the BBB using low pulse electrical fields and intracerebroventricular infusion, lack specificity and have numerous safety concerns. In this review, we will focus on noninvasive transport mechanisms that offer high levels of biocompatibility, personalization, specificity and are regarded as generally safer than their invasive counterparts. Modifiable platforms can be designed to noninvasively traverse the BBB through one or more of the following pathways: passive diffusion through a physio-pathologically disrupted BBB, adsorptive-mediated transcytosis, receptor-mediated transcytosis, shuttle-mediated transcytosis, and somatic gene transfer. Through understanding the noninvasive pathways, new applications, including Chimeric Antigen Receptors T-cell (CAR-T) therapy, and approaches for drug delivery across the BBB are emerging.
Collapse
Affiliation(s)
- Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Roman Adomanis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Slaton
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
4
|
Skibska A, Perlikowska R. Natural Plant Materials as a Source of Neuroprotective Peptides. Curr Med Chem 2024; 31:5027-5045. [PMID: 37403392 DOI: 10.2174/0929867331666230703145043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
In many circumstances, some crucial elements of the neuronal defense system fail, slowly leading to neurodegenerative diseases. Activating this natural process by administering exogenous agents to counteract unfavourable changes seems promising. Therefore, looking for neuroprotective therapeutics, we have to focus on compounds that inhibit the primary mechanisms leading to neuronal injuries, e.g., apoptosis, excitotoxicity, oxidative stress, and inflammation. Among many compounds considered neuroprotective agents, protein hydrolysates and peptides derived from natural materials or their synthetic analogues are good candidates. They have several advantages, such as high selectivity and biological activity, a broad range of targets, and high safety profile. This review aims to provide biological activities, the mechanism of action and the functional properties of plant-derived protein hydrolysates and peptides. We focused on their significant role in human health by affecting the nervous system and having neuroprotective and brain-boosting properties, leading to memory and cognitive improving activities. We hope our observation may guide the evaluation of novel peptides with potential neuroprotective effects. Research into neuroprotective peptides may find application in different sectors as ingredients in functional foods or pharmaceuticals to improve human health and prevent diseases.
Collapse
Affiliation(s)
- Agnieszka Skibska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University, Lodz, Poland
| | - Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University, Lodz, Poland
| |
Collapse
|
5
|
The Therapeutic Potential of Naturally Occurring Peptides in Counteracting SH-SY5Y Cells Injury. Int J Mol Sci 2022; 23:ijms231911778. [PMID: 36233079 PMCID: PMC9569762 DOI: 10.3390/ijms231911778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Peptides have revealed a large range of biological activities with high selectivity and efficiency for the development of new drugs, including neuroprotective agents. Therefore, this work investigates the neuroprotective properties of naturally occurring peptides, endomorphin-1 (EM-1), endomorphin-2 (EM-2), rubiscolin-5 (R-5), and rubiscolin-6 (R-6). We aimed at answering the question of whether well-known opioid peptides can counteract cell injury in a common in vitro model of Parkinson’s disease (PD). Antioxidant activity of these four peptides was evaluated by the 2-diphenyl-1-picrylhydrazyl radical (DPPH) scavenging activity, oxygen radical absorbance capacity (ORAC), and ferric-reducing antioxidant power (FRAP) assays, while neuroprotective effects were assessed in a neurotoxic model induced by 6-hydroxydopamine (6-OHDA) in a human neuroblastoma cell line (SH-SY5Y). The mechanisms associated with neuroprotection were investigated by the determination of mitochondrial membrane potential (MMP), reactive oxygen species (ROS) production, and Caspase-3 activity. Among the tested peptides, endomorphins significantly prevented neuronal death induced by 6-OHDA treatment, decreasing MMP (EM-1) or Caspase-3 activity (EM-2). Meanwhile, R-6 showed antioxidant potential by FRAP assay and exhibited the highest capacity to recover the neurotoxicity induced by 6-OHDA via attenuation of ROS levels and mitochondrial dysfunction. Generally, we hypothesize that peptides’ ability to suppress the toxic effect induced by 6-OHDA may be mediated by different cellular mechanisms. The protective effect caused by endomorphins results in an antiapoptotic effect (mitochondrial protection and decrease in Caspase-3 activity), while R-6 potency to increase a cell’s viability seems to be mediated by reducing oxidative stress. Our results may provide new insight into neurodegeneration and support the short peptides as a potent drug candidate to treat PD. However, further studies should be conducted on the detailed mechanisms of how tested peptides could suppress neuronal injuries.
Collapse
|
6
|
He J, Xue K, Liu J, Gu JH, Peng B, Xu L, Wang G, Jiang Z, Li X, Zhang Y. Timely and Appropriate Administration of Inhaled Argon Provides Better Outcomes for tMCAO Mice: A Controlled, Randomized, and Double-Blind Animal Study. Neurocrit Care 2022; 37:91-101. [PMID: 35137354 DOI: 10.1007/s12028-022-01448-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inhaled argon (iAr) has shown promising therapeutic efficacy for acute ischemic stroke and has exhibited impressive advantages over other inert gases as a neuroprotective agent. However, the optimal dose, duration, and time point of iAr for acute ischemic stroke are unknown. Here, we explored variable iAr schedules and evaluated the neuroprotective effects of acute iAr administration on lesion volume, brain edema, and neurological function in a mouse model of cerebral ischemic/reperfusion injury. METHODS Adult ICR (Institute of Cancer Research) mice were randomly subjected to sham, moderate (1.5 h), or severe (3 h) transient middle cerebral artery occlusion (tMCAO). One hour after tMCAO, the mice were randomized to variable iAr protocols or air. General and focal deficit scores were assessed during double-blind treatment. Infarct volume, overall recovery, and brain edema were analyzed 24 h after cerebral ischemic/reperfusion injury. RESULTS Compared with those in the tMCAO-only group, lesion volume (p < 0.0001) and neurologic outcome (general, p < 0.0001; focal, p < 0.0001) were significantly improved in the group administered iAr 1 h after stroke onset (during ischemia). Short-term argon treatment (1 or 3 h) significantly improved the infarct volume (1 vs. 24 h, p < 0.0001; 3 vs. 24 h, p < 0.0001) compared with argon inhalation for 24 h. The concentration of iAr was confirmed to be a key factor in improving focal neurological outcomes relative to that in the tMCAO group, with higher concentrations of iAr showing better effects. Additionally, even though ischemia research has shown an increase in cerebral damage proportional to the ischemia time, argon administration showed significant neuroprotective effects on infarct volume (p < 0.0001), neurological deficits (general, p < 0.0001; focal, p < 0.0001), weight recovery (p < 0.0001), and edema (p < 0.0001) in general, particularly in moderate stroke. CONCLUSIONS Timely iAr administration during ischemia showed optimal neurological outcomes and minimal infarct volumes. Moreover, an appropriate duration of argon administration was important for better neuroprotective efficacy. These findings may provide vital guidance for using argon as a neuroprotective agent and moving to clinical trials in acute ischemic stroke.
Collapse
Affiliation(s)
- Juan He
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Ke Xue
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jiayi Liu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jin-Hua Gu
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China
| | - Bin Peng
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Lihua Xu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Guohua Wang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Zhenglin Jiang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Xia Li
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yunfeng Zhang
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China.
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
7
|
Li S, Wangqin R, Meng X, Li H, Wang Y, Wang H, Laskowitz D, Chen X, Wang Y. Tolerability and Pharmacokinetics of Single Escalating and Repeated Doses of CN-105 in Healthy Participants. Clin Ther 2022; 44:744-754. [PMID: 35562205 DOI: 10.1016/j.clinthera.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/19/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE CN-105 is an IV, apolipoprotein E-mimetic pentapeptide. Preclinical studies have reported that CN-105 effectively down-regulates neuroinflammatory responses in microglia and mitigates neuronal excitotoxicity following acute brain injury. The CN-105 Phase I and II trials that have been done in the United States have demonstrated that CN-105 was well tolerated in US participants. Thus, the main objective of the present Phase I study was to investigate the tolerability and pharmacokinetic (PK) profiles of CN-105 in healthy Chinese participants. METHODS This randomized, double-blind, placebo-controlled, dose-escalation study was performed in healthy participants using sequential 30-minute IV administration of single and multiple doses of CN-105 (four times daily for 13 doses). Forty volunteers were randomly assigned, in an 8:2 ratio, to one of four dosing groups, receiving either CN-105 (0.03, 0.1, 0.3, or 1 mg/kg), or placebo. Serial blood samples were collected for the measurement of plasma concentrations of CN-105. Tolerability was also assessed. FINDINGS After single-dose administration, the plasma CN-105 concentration rapidly reached the peak by the end of infusion. The mean elimination half-life of CN-105 ranged from 2.3 to 3.6 hours. During single- and multiple-dosing paradigms, exposure to CN-105 (AUC) exhibited linear dependency on dose. Steady state was reached by the fourth dose, with minimal accumulation. The PK properties of CN-105 with single and multiple dosing were comparable to those observed in US participants. CN-105 was generally well tolerated in Chinese participants. A total of 13 adverse events were reported in 30% of subjects (12/40) at the 0.03 mg/kg (6/8), 0.1 mg/kg (1/8), 0.3 mg/kg (2/8), 1 mg/kg (0/8) doses and with placebo (3/8). All adverse events were mild or moderate in severity and self-limited, with no dose relationship observed. IMPLICATIONS CN-105 was well tolerated in these healthy Chinese participants at doses of 0.1 to 1 mg/kg with single and multiple IV administrations. The PK characteristics of CN-105 were comparable among Chinese and Western subjects. A Phase II study in patients with intracranial hemorrhage is being planned in China. CLINICALTRIALS gov identifiers: NCT02670824 and NCT03168581; Chinese Clinical Trial Registration identifier: CTR20202397.
Collapse
Affiliation(s)
- Shuya Li
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Runqi Wangqin
- Department of Neurology, Duke University Medical Center, Durham, North Carolina
| | - Xia Meng
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Haichen Wang
- Department of Neurology, Duke University Medical Center, Durham, North Carolina
| | - Daniel Laskowitz
- Department of Neurology, Duke University Medical Center, Durham, North Carolina
| | - Xia Chen
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Yongjun Wang
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Xue M, Li S, Xu M, Yan L, Laskowitz DT, Kolls BJ, Chen G, Qian X, Wang Y, Song H, Wang Y. Antagonism of nicotinic acetycholinergic receptors by CN‐105, an apoE‐mimetic peptide reduces stroke‐induced excitotoxicity. Clin Transl Med 2022; 12:e677. [PMID: 35075820 PMCID: PMC8787096 DOI: 10.1002/ctm2.677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/21/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Affiliation(s)
- Miaomiao Xue
- State Key Laboratory of Proteomics Beijing Proteome Research Center National Center for Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| | - Shuya Li
- Department of Neurology Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases Beijing China
| | - Mingzhi Xu
- State Key Laboratory of Proteomics Beijing Proteome Research Center National Center for Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| | - Li Yan
- ICE Bioscience Inc. Beijing China
| | - Daniel T. Laskowitz
- Duke Clinical Research Institute Duke University School of Medicine Durham North Carolina USA
- Department of Neurology Duke University Durham North Carolina USA
- Department of Anesthesiology Duke University Durham North Carolina USA
- Aegis‐CN, LLC Durham North Carolina USA
| | - Brad J. Kolls
- Duke Clinical Research Institute Duke University School of Medicine Durham North Carolina USA
- Department of Neurology Duke University Durham North Carolina USA
| | - Gang Chen
- Guangdong Cerebtron Biotech Ltd. Guangdong China
| | - Xiaohong Qian
- State Key Laboratory of Proteomics Beijing Proteome Research Center National Center for Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| | - Yongjun Wang
- Department of Neurology Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases Beijing China
| | - Haifeng Song
- State Key Laboratory of Proteomics Beijing Proteome Research Center National Center for Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| | - Yi Wang
- State Key Laboratory of Proteomics Beijing Proteome Research Center National Center for Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| |
Collapse
|
9
|
James ML, Troy J, Nowacki N, Komisarow J, Swisher CB, Tucker K, Hatton K, Babi MA, Worrall BB, Andrews C, Woo D, Kranz PG, Lascola C, Maughan M, Laskowitz DT. CN-105 in Participants with Acute Supratentorial Intracerebral Hemorrhage (CATCH) Trial. Neurocrit Care 2021; 36:216-225. [PMID: 34424490 DOI: 10.1007/s12028-021-01287-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/21/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Endogenous apolipoprotein (apo) E mediates neuroinflammatory responses and recovery after brain injury. Exogenously administered apoE-mimetic peptides effectively penetrate the central nervous system compartment and downregulate acute inflammation. CN-105 is a novel apoE-mimetic pentapeptide with excellent evidence of functional and histological improvement in preclinical models of intracerebral hemorrhage (ICH). The CN-105 in participants with Acute supraTentorial intraCerebral Hemorrhage (CATCH) trial is a first-in-disease-state multicenter open-label trial evaluating safety and feasability of CN-105 administration in patients with acute primary supratentorial ICH. METHODS Eligible patients were aged 30-80 years, had confirmed primary supratentorial ICH, and were able to intiate CN-105 administration (1.0 mg/kg every 6 h for 72 h) within 12 h of symptom onset. A priori defined safety end points, including hematoma volume, pharmacokinetics, and 30-day neurological outcomes, were analyzed. For clinical outcomes, CATCH participants were compared 1:1 with a closely matched contemporary ICH cohort through random selection. Hematoma volumes determined from computed tomography images on days 0, 1, 2, and 5 and ordinal modified Rankin Scale score at 30 days after ICH were compared. RESULTS In 38 participants enrolled across six study sites in the United States, adverse events occurred at an expected rate without increase in hematoma expansion or neurological deterioration. CN-105 treatment had an odds ratio (95% confidence interval) of 2.69 (1.31-5.51) for lower 30-day modified Rankin Scale score, after adjustment for ICH score, sex, and race/ethnicity, as compared with a matched contemporary cohort. CONCLUSIONS CN-105 administration represents an excellent translational candidate for treatment of acute ICH because of its safety, dosing feasibility, favorable pharmacokinetics, and possible improvement in neurological recovery.
Collapse
Affiliation(s)
- Michael L James
- Department of Anesthesiology, Duke University, Durham, NC, USA. .,Department of Neurology, Duke University, Durham, NC, USA. .,Duke Clinical Research Institute, Duke University, Durham, NC, USA.
| | - Jesse Troy
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | | | | | | | - Kristi Tucker
- Department of Neurology, Wake Forest-Baptist Health, Winston-Salem, NC, USA
| | - Kevin Hatton
- Department of Anesthesiology, University of Kentucky, Lexington, KY, USA
| | - Marc A Babi
- Departments of Neurology and Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Bradford B Worrall
- Departments of Neurology and Public Health Sciences, University of Virginia, Charlottesvile, VA, USA
| | - Charles Andrews
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel Woo
- Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - Peter G Kranz
- Department of Radiology, Duke University, Durham, NC, USA
| | | | | | - Daniel T Laskowitz
- Department of Anesthesiology, Duke University, Durham, NC, USA.,Department of Neurology, Duke University, Durham, NC, USA.,Duke Clinical Research Institute, Duke University, Durham, NC, USA.,AegisCN, LLC, Durham, NC, USA
| | | |
Collapse
|
10
|
Neuroprotective Therapies for Spontaneous Intracerebral Hemorrhage. Neurocrit Care 2021; 35:862-886. [PMID: 34341912 DOI: 10.1007/s12028-021-01311-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022]
Abstract
Patients who survive the initial ictus of spontaneous intracerebral hemorrhage (ICH) remain vulnerable to subsequent injury of the perilesional parenchyma by molecular and cellular responses to the hematoma. Secondary brain injury after ICH, which contributes to long-term functional impairment and mortality, has emerged as an attractive therapeutic target. This review summarizes preclinical and clinical evidence for neuroprotective therapies targeting secondary injury pathways following ICH. A focus on therapies with pleiotropic antiinflammatory effects that target thrombin-mediated chemotaxis and inflammatory cell migration has led to studies investigating statins, anticholinergics, sphingosine-1-phosphate receptor modulators, peroxisome proliferator activated receptor gamma agonists, and magnesium. Attempts to modulate ICH-induced blood-brain barrier breakdown and perihematomal edema formation has prompted studies of nonsteroidal antiinflammatory agents, matrix metalloproteinase inhibitors, and complement inhibitors. Iron chelators, such as deferoxamine and albumin, have been used to reduce the free radical injury that ensues from erythrocyte lysis. Stem cell transplantation has been assessed for its potential to enhance subacute neurogenesis and functional recovery. Despite promising preclinical results of numerous agents, their outcomes have not yet translated into positive clinical trials in patients with ICH. Further studies are necessary to improve our understanding of the molecular events that promote damage and inflammation of the perihematomal parenchyma after ICH. Elucidating the temporal and pathophysiologic features of this secondary brain injury could enhance the clinical efficacy of neuroprotective therapies for ICH.
Collapse
|
11
|
Perlikowska R. Whether short peptides are good candidates for future neuroprotective therapeutics? Peptides 2021; 140:170528. [PMID: 33716091 DOI: 10.1016/j.peptides.2021.170528] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/18/2021] [Accepted: 02/27/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases are a broad group of largely debilitating, and ultimately terminal conditions resulting in progressive degeneration of different brain regions. The observed damages are associated with cell death, structural and functional deficits of neurons, or demyelination. The concept of neuroprotection concerns the administration of the agent, which should reverse some of the damage or prevent further adverse changes. A growing body of evidence suggested that among many classes of compounds considered as neuroprotective agents, peptides derived from natural materials or their synthetic analogs are good candidates. They presented a broad spectrum of activities and abilities to act through diverse mechanisms of action. Biologically active peptides have many properties, including antioxidant, antimicrobial, antiinflammatory, and immunomodulatory effects. Peptides with pro-survival and neuroprotective activities, associated with inhibition of oxidative stress, apoptosis, inflammation and are able to improve cell viability or mitochondrial functions, are also promising molecules of particular interest to the pharmaceutical industries. Peptide multiple activities open the way for broad application potential as therapeutic agents or ingredients of health-promoting functional foods. Significantly, synthetic peptides can be remodeled in numerous ways to have desired features, such as increased solubility or biological stability, as well as selectivity towards a specific receptor, and finally better membrane penetration. This review summarized the most common features of major neurodegenerative disorders, their causes, consequences, and reported new neuroprotective drug development approaches. The author focused on the unique perspectives in neuroprotection and provided a concise survey of short peptides proposed as novel therapeutic agents against various neurodegenerative diseases.
Collapse
Affiliation(s)
- Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, 92-215, Lodz, Poland.
| |
Collapse
|