Skip to main content
Erschienen in: Translational Stroke Research 5/2013

Open Access 01.10.2013 | Review Article

Therapeutic Antibodies in Stroke

verfasst von: Chye Yun Yu, Gandi Ng, Ping Liao

Erschienen in: Translational Stroke Research | Ausgabe 5/2013

Abstract

Immunotherapy represents an active area of biomedical research to treat cancer, autoimmune diseases, and neurodegenerative disorders. In stroke, recanalization therapy is effective in reducing brain tissue damage after acute ischemic stroke. However, the narrow time window restricts its application for the majority of stroke patients. There is an urgent need to develop adjuvant therapies such as immunotherapy, stem cell replacement, and neuroprotective drugs. A number of molecules have been targeted for immunotherapy in stroke management, including myelin-associated proteins and their receptors, N-methyl-d-aspartic acid receptors, cytokines, and cell adhesion molecules. Both active vaccination and passive antibodies were tested in animal models of acute ischemic stroke. However, the mechanisms underlying the efficacy of immunotherapy are different for each target protein. Blocking myelin-associated proteins may enhance neuroplasticity, whereas blocking adhesion molecules may yield neuroprotection by suppressing the immune response after stroke. Although results from animal studies are encouraging, clinical trials using therapeutic antibodies failed to improve stroke outcome due to severe side effects. It remains a challenge to generate specific therapeutic antibodies with minimal side effects on other organs and systems.

Introduction

Prophylactic vaccination is widely used and is proven effective against infectious diseases. More recently, much attention has been paid to immunotherapy for the treatment of other diseases such as cancer [1], autoimmune diseases [2], and neurodegenerative disorders [3]. Immunotherapy has great potential to be an effective adjuvant therapy. Due to the specificity of the immune response, harnessing the immune system to block specific signaling pathways provides a powerful tool for the treatment of disease.
Stroke is one of the most common causes of death worldwide and is a heavy burden on the health care system. Ischemic strokes constitute the majority of all strokes. Inflammation triggered after stroke is characterized by an orderly sequence of events involving different components of the brain. Right after arterial occlusion, release of reactive oxygen species triggers the coagulation cascade and activates complement, platelet, and endothelial cells. The white blood cell count and cytokines and inflammatory markers are increased within hours, followed by a marked immunodepression within 1–2 days, particularly in large strokes. Such changes in the systemic immunity lead to higher occurrence of infection in respiratory and urinary systems. As the ischemia progresses, toxic molecules such as excessive ATP and neurotransmitters are released into the extracellular space to trigger innate and adaptive immunity. With the increased permeability of the blood–brain barrier, autoimmunity is induced against the dead brain cells. Circulating T cells are sensitized to produce antibodies against antigens in central nervous system. Antigen-presenting cells are mobilized from the periphery to the ischemic brain and contribute to the destruction of brain tissues at the site of ischemic lesion. Autoimmunity may have long-term consequences on stroke survivors including dementia and brain atrophy. On the other hand, the immunosuppression after stroke may reduce the autoimmune attach on the brain by limiting the development of T cells. The detailed immunology after stroke was best reviewed in ref [4].
The most effective treatments for acute ischemic stroke are revascularization by thrombolysis, the dissolving of the clot, and embolectomy, the surgical removal of the clot. Tissue plasminogen activator (tPA), a thrombolytic agent approved by the FDA, is widely used to treat acute embolic or thrombotic stroke. However, the narrow therapeutic time window (<4.5 h post-stroke) benefits only a minority of stroke patients. Reperfusion after this time window causes damage to brain tissue as deleterious biochemical events are triggered that antagonize the beneficial effects. Thus, the challenge for reperfusion therapy is to both protect brain tissue and extend the therapeutic time window [5].
Immunotherapy for stroke treatment attracts significant scientific attention. Numerous signaling pathways are altered after stroke insult. Blocking certain deleterious pathways may delay brain tissue damage and even widen the time window for revascularization therapy. Immunotherapy provides a novel type of adjuvant stroke therapy. The interaction of antibodies with cytotoxic molecules and their receptors could rescue cell viability or delay cell death. Current investigations of stroke immunotherapy include active immunization by inoculation with peptides and passive immunization by direct injection of antibody into the animals. Many molecules have been targeted for stroke therapy, and a number of antibodies have been developed. These molecules are primarily on the cell membrane or in the extracellular space where they are accessible to the antibodies. Middle cerebral artery occlusion (MCAO) is the most common animal model of focal ischemia. The efficacy of the blocking antibodies is evaluated in either transient or permanent MCAO models. Although most of the antibodies were effective in reducing brain damage in animal models of stroke, clinical trials for several antibodies failed due to poor patient outcomes. Here, we review the current understanding of immunotherapy, particularly the use of therapeutic antibodies, for stroke management.

Myelin-Associated Proteins

Myelin in the adult central nervous system (CNS) contains abundant growth-inhibitory molecules, including proteoglycans, Nogo-A, myelin-associated glycoprotein (MAG), versican V2, and oligodendrocyte myelin glycoprotein (Omgp) [6]. Disrupted myelin is often associated with deposits of immunoglobulin and activated complement, which may cause autoimmune diseases such as multiple sclerosis [7].
Nogo-A, a member of the reticulon family, is produced by oligodendrocytes [8] and is a potent inhibitor of axonal remodeling. Nogo-A interacts with the Nogo-66 receptor and inhibits neurite outgrowth after CNS injury [9]. The monoclonal antibody (IN-1) against Nogo-A was generated several years prior to the identification of Nogo-A [10]. Because Nogo-A plays an important role after CNS injury, a number of studies have aimed to block its functions by infusion of antibodies against Nogo-A or the Nogo-66 receptor after stroke.
Immunotherapy against Nogo-A in animal models of stroke was shown to enhance functional recovery by improving neuroplasticity. Application of anti-Nogo-A antibody via intracerebroventricular [11] or intrathecal routes [12] at 1 week after operation could hasten the recovery from neurological damage. In a permanent rat MCAO model, delayed treatment with anti-Nogo-A antibody (9 weeks after operation) enhanced sprouting and midline crossing of corticorubral axons to innervate the deafferented red nucleus [13]. Following MCAO, IN-1-treated animals showed increased dendritic arborization and spine density in the lesioned hemisphere [14]. IN-1 also promoted formation of new efferent cortical projections [15].
As stroke commonly occurs in hypertensive or aging patients, anti-Nogo-A antibodies were tested in animals with hypertension or in aged animals. Again, the antibody exhibited great potential to enhance neuroplasticity after stroke, significant functional improvement was observed in hypertensive rats receiving anti-Nogo-A antibody after permanent MCAO [16]. In aged rats (18 months old), anti-Nogo-A therapy given 1 week after MCAO improved reference memory in the Morris water maze task [17]. Similar to anti-Nogo-A therapy, antibody against the Nogo-66 receptor was proven effective in reducing stroke-induced brain damage. The impaired forelimb function of rats that underwent MCAO was ameliorated after the implantation of a hydrogel carrying polyclonal antibodies against the Nogo-66 receptor. Neurons and neurofibers were also found within the gel providing evidence of neuronal migration and regeneration [18].
In contrast to the promising results of blocking Nogo-A or the Nogo-66 receptor, mice with a Nogo-A deletion displayed increased mortality and exacerbated neurological deficits after transient MCAO. A similar observation was made in rats that received anti-Nogo-A antibody immediately following stroke, indicating that Nogo-A may play an opposite role in cerebral damage during the hyperacute phase of stroke [19]. It is possible that in the hyperacute phase, tissue damage is more prominent than tissue repair. Thus, the time point to block Nogo-A function is critical, antibodies that inhibit Nogo-A function may yield better effect during neural regeneration in the later stages of stroke recovery. It is critical to identify the proper time point for anti-Nogo-A treatment in human patients, as the rodents have a different life span than humans. A human monoclonal anti-Nogo-A antibody (ATI355) from Novartis has been approved for clinical trials to treat acute spinal cord injury (Table 1. ClinicalTrials.gov Identifier: NCT00406016). The phase I clinical trial has been completed and no severe side effects were observed from more than 50 subjects. Thus, this antibody may potentially be used for the treatment of ischemic stroke in the future.
Table 1
Clinical trials for stroke immunotherapy
Target
Condition
Intervention
Phase
Description
E-selectin
Ischemic stroke
E-selectin nasal spray
II
Terminated (NCT00012454)
E-selectin
Ischemic stroke
Recombinant human E-selectin
I
Not recruited (NCT00069069)
Nogo-A
Acute spinal cord injury
ATI355 (anti-Nogo-A)
I
Completed (NCT00406016)
ICAM-1
Acute stroke
Enlimomab (anti-ICAM-1)
III
Completed (Enlimomab Acute Stroke Trial)
MAG is another molecule found in myelin that can inhibit neurite outgrowth after injury. In a MCAO rat model, both intracerebral and systemic administration of MAG neutralization antibody reduced the lesion area and improved motor function. The effect was attributed not only to the blockade of MAG protein but also to the protection of oligodendrocytes from oxidative stress-induced cell death [20].
Omgp is important for nerve myelination in the CNS. Autoantibody against Omgp is believed to cause multiple sclerosis [21]. Active immunization was applied to test the role of Omgp in stroke progress. Nasal vaccination with Omgp peptides before MCAO induced an anti-inflammatory response against CNS myelin, a process that involves the induction of CD4+ T cells producing IL-10. As a result, the infarct volume decreased and functional recovery was enhanced [22].

NMDA Receptor

Glutamate is a major excitatory neurotransmitter in the brain. In ischemic stroke, cessation of cerebral blood flow depletes the brain of glucose and oxygen, leading to neuronal membrane depolarization and release of large amount of glutamate into the extracellular space. Glutamate binds to glutamate receptors, such as N-methyl-d-aspartic acid (NMDA) receptors and dl-α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors causing influx of Na+ and Ca2+ ions into the cell. Excessive activation of glutamate receptor can result in cell death. Accordingly, glutamate receptors are a major target for neuroprotection in a wide range of CNS disorders. Extensive studies have been conducted on glutamate receptors to improve stroke therapy.
tPA is a serine protease that converts plasminogen into active plasmin, leading to fibrin blood clot degradation. Endogenous tPA is produced after a thrombus forms. Due to its ability to recanalize cerebral blood vessels, recombinant tPA is used for thrombolysis within 4.5 h after stroke onset. It is the only FDA-approved pharmacological agent for acute ischemic stroke therapy [23]. However, tPA treatment has severe side effects such as cerebral edema and hemorrhage. tPA interacts with and cleaves the GluN1 (NR1) subunit of NMDA receptor, resulting in Ca2+ influx that activates a cascade of cell death signals [24]. Accordingly, blocking the interaction of tPA with the NMDA receptor would reduce the side effects of tPA treatment. Immunotherapy with antibodies targeting the tPA binding site on the NMDA receptor was able to prevent neuronal death both in vitro and in vivo. Using the adeno-associated virus as a carrier, autoantibodies were generated against the GluN1 subunit of the NMDA receptor in rats. After vaccination, antibodies were able to pass through the blood–brain barrier (BBB) and reduced total infarct volume by 70 % in a permanent MCAO model [25]. Importantly, the antibodies persisted for over 5 months in the rats, indicating that vaccination can potentially prevent stroke. In a separate study, direct injection of a fragment of the GluN1 subunit prevented tPA cleavage of the NMDA receptor, successfully reducing the infarct volume after permanent MCAO [26].
The NMDA receptor was also targeted for passive immunization to treat ischemic stroke. Antibodies were produced in vitro against the GluN1 subunit and injected into animals after MCAO. The antibodies successfully diffused into the infarct region and reduced the brain tissue injury and BBB leakage in combination with tPA treatment. Importantly, a longer therapeutic time window for tPA thrombolysis was achieved in the animal model [27].
These experiments suggest the possibility of using active immunization against the NMDA receptor as a preventive vaccination and passive immunization to extend the time window for tPA thrombolysis [28]. Unfortunately, immunotherapy targeting NMDA receptors has not been successful in clinical practice. Although a wealth of evidence indicates that blockade of glutamate receptors could lead to dramatic neuroprotection in animal models, clinical trials of glutamate receptor blockers have not achieved therapeutic outcome [29]. A possible reason for this failure could be that the beneficial effects of glutamate receptor block are overcome by the deleterious effects on healthy cells. In patients with anti-NMDAR encephalitis, autoantibodies against GluN1 subunit of NMDA receptor are associated with a number of psychiatric symptoms within a few days after onset [30]. These antibodies bind, cross-link, and internalize NMDA receptors, leading to a decrease of synaptic NMDA currents [31]. Thus, blocking NMDA receptors indiscriminately may cause severe damage to the healthy neurons in other regions of the brain.

Cell Adhesion Molecules and Cytokines

Inflammation has been suggested to participate in stroke pathophysiology. Inflammatory signaling is triggered immediately following the ischemic attack and plays a dynamic role in all stages of stroke. This process is initiated with an orderly inflammation response that leads to the activation of complement, platelets, and endothelial cells within the injured vasculature. The subsequent release of cytokines leads to leukocytes infiltration through the BBB and contributes to the tissue damage after stroke. Proper modulation of the immune response after stroke insult can help protect the injured tissue and promote functional recovery.
Recruitment of leukocytes and platelets to injured cerebral blood vessels is a highly coordinated process that involves various adhesion molecules on the surface of endothelial cells, leukocytes, and platelets. These adhesion molecules include the selectin family, intercellular adhesion molecule-1 (ICAM-1, also known as CD54), and β2-integrins. Numerous studies have demonstrated that disruption of leukocyte function yielded beneficial effects on stroke pathology. For example, depletion of neutrophils by an anti-neutrophil monoclonal antibody significantly reduced free radical generation [32] and attenuated brain edema after ischemic reperfusion injury [33]. In this way, immunotherapy could serve a beneficial role by inhibiting leukocyte functions. Neutralization of the adhesion molecules by therapeutic antibodies can disrupt the interaction of leukocytes with the injured vascular wall, thereby reducing leukocyte infiltration. Numerous antibodies have been developed to target these adhesion molecules.
The selectin family is composed of E-, L-, and P-selectin. They are involved in the adhesion of leukocytes to endothelial cells and subsequent transmigration into the brain parenchyma. Upregulation of E- and P-selectin on the surface of activated endothelial cells occurs soon after ischemic onset [34]. Endothelial selectins bind to leukocytes with low affinity via glycoproteins expressed on leukocytes. Surface expression of P-selectin on platelets and L-selectin on leukocytes also facilitates platelet–leukocyte aggregation.
In a permanent MCAO rat model, pretreatment with monoclonal anti-P-selectin antibody reduced leukocyte infiltration. Regional cerebral blood flow was elevated in association with attenuation of cerebral infarction [35] and brain edema [36]. In a murine transient MCAO model, neutralization of E-selectin with a monoclonal antibody led to increased cerebral blood flow, reduced neutrophil accumulation at the injured site, and improved neuroprotection [37]. Active immunization targeting E-selectin was also effective in stroke management. In spontaneously hypertensive rats, immune tolerance by repeated nasal instillation of E-selectin antigen successfully prevented the development of both ischemic and hemorrhagic strokes by suppressed endothelial activation [38, 39]. A phase II clinical trial was conducted with E-selectin nasal spray in patients with a stroke history to test the safety and effectiveness of E-selectin in preventing the formation of blood clots that can cause stroke. This trial was terminated (Table 1. NCT00012454) and another phase I trial using recombinant human E-selectin to prevent secondary stroke has not yet opened for participant recruitment since 2006 (Table 1. NCT00069069).
The effects of blocking L-selectin are different from blocking E- or P-selectin. In a rabbit transient MCAO model, a humanized anti-L-selectin antibody failed to prevent neutrophil migration into the injured site and did not alleviate neuronal damage [40]. However, when used in combination with tPA in a similar animal model, anti-L-selectin antibody demonstrated a moderate reduction of brain damage, which was absent in animals treated with tPA alone [41]. Thrombolysis with tPA alters the coagulation status in stroke patients and could lead to hemorrhage. It is possible that the beneficial effects of blocking L-selectin depend on the altered coagulation environment after tPA treatment.
ICAM-1 is a member of the immunoglobulin superfamily that is expressed on the surface of endothelial cells. Various cytokines can stimulate the expression of ICAM-1 [42, 43]. β2-Integrins are expressed on the surface of leukocytes; ICAM-1 is the ligand for β2-integrins LFA-1 (CD11a/CD18) and Mac-1 (CD11b/CD18). Interaction between β2-integrins (LFA-1 and Mac-1) and ICAM-1 provides a firm adhesion between leukocytes and endothelium [43, 44]. In various animal models of transient ischemic stroke, blocking of ICAM-1 with antibodies reduced polymorphonuclear leukocyte infiltration and neurological damage [4547]. Together with tPA treatment, application of anti-ICAM-1 enhanced the thrombolytic effect and extended the therapeutic window for tPA [48].
Interestingly, anti-ICAM-1 treatment showed beneficial effects in transient ischemic stroke models but not in permanent MCAO models [49]. A commercial murine monoclonal anti-ICAM-1 antibody (Enlimomab) has been used for the treatment of ischemic stroke. However, the phase III clinical trial showed that the fatality rate was significantly higher in the Enlimomab patient group than in the placebo group [50]. The negative effect was apparent on day 5 after treatment. The serious adverse events include fever, myocardial infarction, pulmonary edema, pneumonia, stroke-related deterioration, cardiac arrest, meningitis, cerebral edema, and intracranial hemorrhage.
Immunoblocking of the β2-integrin Mac-1 effectively alleviated ischemic reperfusion damage in the liver and heart by reducing neutrophil infiltration [5153]. A monoclonal antibody targeting Mac-1 (CD11b) achieved similar effects with decreased intraparenchymal neutrophils after transient ischemia–reperfusion [54, 55]. In a rabbit thromboembolic stroke model, monoclonal antibody against CD18 successfully reduced intracranial pressure but had no effect on cerebral blood flow and infarction [56].
The production and release of pro-inflammatory cytokines within the CNS are hallmarks of tissue damage following stroke. They are intimately involved in both innate and adaptive immunity at various stages of stroke [4]. IL-8 is a potent chemoattractant for polymorphonuclear neutrophils [57, 58] that is upregulated after stroke [59, 60]. The rat equivalent of IL-8, cytokine-induced neutrophil chemoattractant (CINC), can be blocked by antibodies [61]. In a transient ischemic reperfusion stroke model, application of anti-CINC antibodies alleviated cerebral edema and reduced the infarct size 7 days after reperfusion [62].
CD 95L and tumor necrotic factor (TNF) interact with TNF receptor 1 and trigger apoptosis. As members of the cytokine family, they are involved in systematic inflammation. Humanized monoclonal antibodies specific for TNF were used in clinical practice to treat diseases such as rheumatoid arthritis and Crohn’s disease [63]. These antibodies include infliximab (Remicade) from Centocor (Philadelphia, PA, US) and adalimumab (Humira) from Abbott (North Chicago, IL, US). TNF is involved in both innate and adaptive immune response after stroke [4]. TNF was found in the brain 30 min after MCAO [64] and its levels were increased early after transient ischemia attack [65]. Neutralization antibody against TNF was found to ameliorate brain damage in both permanent and reperfusion MCAO models [66, 67]. When both TNF and CD95L were blocked by antibodies, an additive effect was observed on the reduction of the infarct volume after transient MCAO [68]. It is worthwhile to investigate whether infliximab or adalimumab could be helpful in stroke management, as they have already been proven safe in clinical practice.

Side Effect, Safety, and Future Perspectives

General side effects could arise from immunotherapy. Constitutional symptoms include transient low-grade fever, malaise, nausea, dyspnea, diarrhea, fatigue, hoarseness, palpitations, and headache. Aseptic meningitis could also occur due to a non-immune response triggered by formation of macro-aggregates or due to local inflammation caused by antibody interaction with endothelial antigens on the meningeal vessels. Other severe side effects include acute renal failure and anaphylaxis. It should be noted that thromboembolic events could occur with the increased plasma viscosity that develops after immunoglobulin infusion [69]. Myocardial infarction, pulmonary embolism, venous thrombosis, or even secondary stroke might occur. Animal experiments need to be carefully designed and evaluated to fully understand the possible side effects that may arise after immunotherapy.
The failure of clinical trials of Enlimomab provides valuable information on how to apply immunotherapy properly for stroke management. Preclinical studies on anti-ICAM-1 antibody showed beneficial effects in transient ischemic animal models but not in permanent stroke models [4549]. However, the patients enrolled in the Enlimomab trial may not have reperfusion. Furthermore, Enlimomab is a murine monoclonal antibody, which triggered the patients’ own immune responses [70]. Human anti-mouse antibody was found in patients receiving Enlimomab therapy [71], which could lead to an increased infection rate. Patients with fever generally had a poor outcome or died; such outcomes are not oddities in these trails. In animal models, stimulation of the self-immune system was observed with the activation of circulating neutrophilic granulocytes, increased β2-integrin CD11b, and E- and P-selectins. Additionally, ICAM-1 was upregulated by anti-ICAM-1 treatment [72, 73]. To avoid the side effects from murine antibodies, humanized monoclonal antibodies were shown to be effective in treating other diseases without severe side effects [63].
Modern technology has greatly improved the production of high quality antibodies, including humanized antibodies, thus reducing the general side effects of immunotherapy. However, attention must be paid to several important factors throughout research for stroke immunotherapy. First, preclinical studies are performed on animals. Because the pathophysiology of stroke in humans and animal models is different, a comparison of the progression of stroke between species would help to improve the outcome of immunotherapy. Second, the target molecule should be chosen carefully. A molecule that is expressed highly in other tissues and has important functions should be targeted with caution, as severe side effects are more likely to occur. A deleterious molecule that is upregulated or expressed exclusively in the infarct region after stroke could be an ideal target for immunotherapy. Third, the stroke development is a dynamic process. Some molecules could play opposite roles during different phases of stroke. For example, blocking Nogo-A may be successful only at a later stage when neural regeneration occurs. Thus, choosing a time point for a particular immunotherapy is critical to avoid side effects. Fourth, because various molecules play different roles in stroke, a combination of multiple antibodies might achieve a better therapeutic outcome. Finally, current recanalization is effective in stroke management. Interaction of immunotherapy with current reperfusion methods needs to be carefully investigated to achieve a synergistic effect while avoiding antagonism.
In summary, numerous signaling pathways are altered after stroke insult. It is critical to determine the relative hierarchical importance, time course, and crosstalk among these signaling pathways to rationally plan targets for immunotherapy and points of intervention.

Acknowledgments

This research is supported by the Singapore Ministry of Health’s National Medical Research Council.

Conflict of Interest

This article does not contain any studies with human or animal subjects. Chye Yun Yu, Gandi Ng, and Ping Liao declare that they have no conflicts of interest.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
2.
Zurück zum Zitat Iorio R, Lennon VA. Neural antigen-specific autoimmune disorders. Immunol Rev. 2012;248:104–21.PubMedCrossRef Iorio R, Lennon VA. Neural antigen-specific autoimmune disorders. Immunol Rev. 2012;248:104–21.PubMedCrossRef
3.
Zurück zum Zitat Villoslada P, Moreno B, Melero I, Pablos JL, Martino G, Uccelli A, et al. Immunotherapy for neurological diseases. Clin Immunol. 2008;128:294–305.PubMedCrossRef Villoslada P, Moreno B, Melero I, Pablos JL, Martino G, Uccelli A, et al. Immunotherapy for neurological diseases. Clin Immunol. 2008;128:294–305.PubMedCrossRef
4.
Zurück zum Zitat Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med. 2011;17:796–808.PubMedCrossRef Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med. 2011;17:796–808.PubMedCrossRef
5.
Zurück zum Zitat Donnan GA, Davis SM. Stroke: expanded indications for stroke thrombolysis—what next? Nat Rev Neurol. 2012;8:482–3.PubMedCrossRef Donnan GA, Davis SM. Stroke: expanded indications for stroke thrombolysis—what next? Nat Rev Neurol. 2012;8:482–3.PubMedCrossRef
6.
7.
Zurück zum Zitat Franklin RJ, Ffrench-Constant C, Edgar JM, Smith KJ. Neuroprotection and repair in multiple sclerosis. Nat Rev Neurol. 2012;8:624–34.PubMedCrossRef Franklin RJ, Ffrench-Constant C, Edgar JM, Smith KJ. Neuroprotection and repair in multiple sclerosis. Nat Rev Neurol. 2012;8:624–34.PubMedCrossRef
8.
Zurück zum Zitat GrandPre T, Nakamura F, Vartanian T, Strittmatter SM. Identification of the Nogo inhibitor of axon regeneration as a reticulon protein. Nature. 2000;403:439–44.PubMedCrossRef GrandPre T, Nakamura F, Vartanian T, Strittmatter SM. Identification of the Nogo inhibitor of axon regeneration as a reticulon protein. Nature. 2000;403:439–44.PubMedCrossRef
9.
Zurück zum Zitat Fournier AE, GrandPre T, Strittmatter SM. Identification of a receptor mediating Nogo-66 inhibition of axonal regeneration. Nature. 2001;409:341–6.PubMedCrossRef Fournier AE, GrandPre T, Strittmatter SM. Identification of a receptor mediating Nogo-66 inhibition of axonal regeneration. Nature. 2001;409:341–6.PubMedCrossRef
10.
Zurück zum Zitat Caroni P, Schwab ME. Antibody against myelin-associated inhibitor of neurite growth neutralizes nonpermissive substrate properties of CNS white matter. Neuron. 1988;1:85–96.PubMedCrossRef Caroni P, Schwab ME. Antibody against myelin-associated inhibitor of neurite growth neutralizes nonpermissive substrate properties of CNS white matter. Neuron. 1988;1:85–96.PubMedCrossRef
11.
Zurück zum Zitat Seymour AB, Andrews EM, Tsai SY, Markus TM, Bollnow MR, Brenneman MM, et al. Delayed treatment with monoclonal antibody IN-1 1 week after stroke results in recovery of function and corticorubral plasticity in adult rats. J Cereb Blood Flow Metab. 2005;25:1366–75.PubMedCrossRef Seymour AB, Andrews EM, Tsai SY, Markus TM, Bollnow MR, Brenneman MM, et al. Delayed treatment with monoclonal antibody IN-1 1 week after stroke results in recovery of function and corticorubral plasticity in adult rats. J Cereb Blood Flow Metab. 2005;25:1366–75.PubMedCrossRef
12.
Zurück zum Zitat Tsai SY, Markus TM, Andrews EM, Cheatwood JL, Emerick AJ, Mir AK, et al. Intrathecal treatment with anti-Nogo-A antibody improves functional recovery in adult rats after stroke. Exp Brain Res. 2007;182:261–6.PubMedCrossRef Tsai SY, Markus TM, Andrews EM, Cheatwood JL, Emerick AJ, Mir AK, et al. Intrathecal treatment with anti-Nogo-A antibody improves functional recovery in adult rats after stroke. Exp Brain Res. 2007;182:261–6.PubMedCrossRef
13.
Zurück zum Zitat Tsai SY, Papadopoulos CM, Schwab ME, Kartje GL. Delayed anti-Nogo-a therapy improves function after chronic stroke in adult rats. Stroke. 2011;42:186–90.PubMedCrossRef Tsai SY, Papadopoulos CM, Schwab ME, Kartje GL. Delayed anti-Nogo-a therapy improves function after chronic stroke in adult rats. Stroke. 2011;42:186–90.PubMedCrossRef
14.
Zurück zum Zitat Papadopoulos CM, Tsai SY, Cheatwood JL, Bollnow MR, Kolb BE, Schwab ME, et al. Dendritic plasticity in the adult rat following middle cerebral artery occlusion and Nogo-a neutralization. Cereb Cortex. 2006;16:529–36.PubMedCrossRef Papadopoulos CM, Tsai SY, Cheatwood JL, Bollnow MR, Kolb BE, Schwab ME, et al. Dendritic plasticity in the adult rat following middle cerebral artery occlusion and Nogo-a neutralization. Cereb Cortex. 2006;16:529–36.PubMedCrossRef
15.
Zurück zum Zitat Papadopoulos CM, Tsai SY, Alsbiei T, O’Brien TE, Schwab ME, Kartje GL. Functional recovery and neuroanatomical plasticity following middle cerebral artery occlusion and IN-1 antibody treatment in the adult rat. Ann Neurol. 2002;51:433–41.PubMedCrossRef Papadopoulos CM, Tsai SY, Alsbiei T, O’Brien TE, Schwab ME, Kartje GL. Functional recovery and neuroanatomical plasticity following middle cerebral artery occlusion and IN-1 antibody treatment in the adult rat. Ann Neurol. 2002;51:433–41.PubMedCrossRef
16.
Zurück zum Zitat Wiessner C, Bareyre FM, Allegrini PR, Mir AK, Frentzel S, Zurini M, et al. Anti-Nogo-A antibody infusion 24 hours after experimental stroke improved behavioral outcome and corticospinal plasticity in normotensive and spontaneously hypertensive rats. J Cereb Blood Flow Metab. 2003;23:154–65.PubMedCrossRef Wiessner C, Bareyre FM, Allegrini PR, Mir AK, Frentzel S, Zurini M, et al. Anti-Nogo-A antibody infusion 24 hours after experimental stroke improved behavioral outcome and corticospinal plasticity in normotensive and spontaneously hypertensive rats. J Cereb Blood Flow Metab. 2003;23:154–65.PubMedCrossRef
17.
Zurück zum Zitat Gillani RL, Tsai SY, Wallace DG, O’Brien TE, Arhebamen E, Tole M, et al. Cognitive recovery in the aged rat after stroke and anti-Nogo-A immunotherapy. Behav Brain Res. 2010;208:415–24.PubMedCrossRef Gillani RL, Tsai SY, Wallace DG, O’Brien TE, Arhebamen E, Tole M, et al. Cognitive recovery in the aged rat after stroke and anti-Nogo-A immunotherapy. Behav Brain Res. 2010;208:415–24.PubMedCrossRef
18.
Zurück zum Zitat Ma J, Tian WM, Hou SP, Xu QY, Spector M, Cui FZ. An experimental test of stroke recovery by implanting a hyaluronic acid hydrogel carrying a Nogo receptor antibody in a rat model. Biomed Mater. 2007;2:233–40.PubMedCrossRef Ma J, Tian WM, Hou SP, Xu QY, Spector M, Cui FZ. An experimental test of stroke recovery by implanting a hyaluronic acid hydrogel carrying a Nogo receptor antibody in a rat model. Biomed Mater. 2007;2:233–40.PubMedCrossRef
19.
Zurück zum Zitat Kilic E, ElAli A, Kilic U, Guo Z, Ugur M, Uslu U, et al. Role of Nogo-A in neuronal survival in the reperfused ischemic brain. J Cereb Blood Flow Metab. 2010;30:969–84.PubMedCrossRef Kilic E, ElAli A, Kilic U, Guo Z, Ugur M, Uslu U, et al. Role of Nogo-A in neuronal survival in the reperfused ischemic brain. J Cereb Blood Flow Metab. 2010;30:969–84.PubMedCrossRef
20.
Zurück zum Zitat Irving EA, Vinson M, Rosin C, Roberts JC, Chapman DM, Facci L, et al. Identification of neuroprotective properties of anti-MAG antibody: a novel approach for the treatment of stroke? J Cereb Blood Flow Metab. 2005;25:98–107.PubMedCrossRef Irving EA, Vinson M, Rosin C, Roberts JC, Chapman DM, Facci L, et al. Identification of neuroprotective properties of anti-MAG antibody: a novel approach for the treatment of stroke? J Cereb Blood Flow Metab. 2005;25:98–107.PubMedCrossRef
21.
Zurück zum Zitat Derfuss T, Meinl E. Identifying autoantigens in demyelinating diseases: valuable clues to diagnosis and treatment? Curr Opin Neurol. 2012;25:231–8.PubMedCrossRef Derfuss T, Meinl E. Identifying autoantigens in demyelinating diseases: valuable clues to diagnosis and treatment? Curr Opin Neurol. 2012;25:231–8.PubMedCrossRef
22.
Zurück zum Zitat Frenkel D, Huang Z, Maron R, Koldzic DN, Hancock WW, Moskowitz MA, et al. Nasal vaccination with myelin oligodendrocyte glycoprotein reduces stroke size by inducing IL-10-producing CD4+ T cells. J Immunol. 2003;171:6549–55.PubMed Frenkel D, Huang Z, Maron R, Koldzic DN, Hancock WW, Moskowitz MA, et al. Nasal vaccination with myelin oligodendrocyte glycoprotein reduces stroke size by inducing IL-10-producing CD4+ T cells. J Immunol. 2003;171:6549–55.PubMed
23.
Zurück zum Zitat The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. Tissue plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333:1581–7.CrossRef The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. Tissue plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333:1581–7.CrossRef
24.
Zurück zum Zitat Nicole O, Docagne F, Ali C, Margaill I, Carmeliet P, MacKenzie ET, et al. The proteolytic activity of tissue-plasminogen activator enhances NMDA receptor-mediated signaling. Nat Med. 2001;7:59–64.PubMedCrossRef Nicole O, Docagne F, Ali C, Margaill I, Carmeliet P, MacKenzie ET, et al. The proteolytic activity of tissue-plasminogen activator enhances NMDA receptor-mediated signaling. Nat Med. 2001;7:59–64.PubMedCrossRef
25.
Zurück zum Zitat During MJ, Symes CW, Lawlor PA, Lin J, Dunning J, Fitzsimons HL, et al. An oral vaccine against NMDAR1 with efficacy in experimental stroke and epilepsy. Science. 2000;287:1453–60.PubMedCrossRef During MJ, Symes CW, Lawlor PA, Lin J, Dunning J, Fitzsimons HL, et al. An oral vaccine against NMDAR1 with efficacy in experimental stroke and epilepsy. Science. 2000;287:1453–60.PubMedCrossRef
26.
Zurück zum Zitat Benchenane K, Castel H, Boulouard M, Bluthe R, Fernandez-Monreal M, Roussel BD, et al. Anti-NR1 N-terminal-domain vaccination unmasks the crucial action of tPA on NMDA-receptor-mediated toxicity and spatial memory. J Cell Sci. 2007;120:578–85.PubMedCrossRef Benchenane K, Castel H, Boulouard M, Bluthe R, Fernandez-Monreal M, Roussel BD, et al. Anti-NR1 N-terminal-domain vaccination unmasks the crucial action of tPA on NMDA-receptor-mediated toxicity and spatial memory. J Cell Sci. 2007;120:578–85.PubMedCrossRef
27.
Zurück zum Zitat Macrez R, Obiang P, Gauberti M, Roussel B, Baron A, Parcq J, et al. Antibodies preventing the interaction of tissue-type plasminogen activator with N-methyl-d-aspartate receptors reduce stroke damages and extend the therapeutic window of thrombolysis. Stroke. 2011;42:2315–22.PubMedCrossRef Macrez R, Obiang P, Gauberti M, Roussel B, Baron A, Parcq J, et al. Antibodies preventing the interaction of tissue-type plasminogen activator with N-methyl-d-aspartate receptors reduce stroke damages and extend the therapeutic window of thrombolysis. Stroke. 2011;42:2315–22.PubMedCrossRef
28.
Zurück zum Zitat Macrez R, Bezin L, Le Mauff B, Ali C, Vivien D. Functional occurrence of the interaction of tissue plasminogen activator with the NR1 subunit of N-methyl-d-aspartate receptors during stroke. Stroke. 2010;41:2950–5.PubMedCrossRef Macrez R, Bezin L, Le Mauff B, Ali C, Vivien D. Functional occurrence of the interaction of tissue plasminogen activator with the NR1 subunit of N-methyl-d-aspartate receptors during stroke. Stroke. 2010;41:2950–5.PubMedCrossRef
29.
Zurück zum Zitat Muir KW. Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Curr Opin Pharmacol. 2006;6:53–60.PubMedCrossRef Muir KW. Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Curr Opin Pharmacol. 2006;6:53–60.PubMedCrossRef
30.
Zurück zum Zitat Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol. 2011;10:63–74.PubMedCrossRef Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol. 2011;10:63–74.PubMedCrossRef
31.
Zurück zum Zitat Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci. 2010;30:5866–75.PubMedCrossRef Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci. 2010;30:5866–75.PubMedCrossRef
32.
Zurück zum Zitat Matsuo Y, Kihara T, Ikeda M, Ninomiya M, Onodera H, Kogure K. Role of neutrophils in radical production during ischemia and reperfusion of the rat brain: effect of neutrophil depletion on extracellular ascorbyl radical formation. J Cereb Blood Flow Metab. 1995;15:941–7.PubMedCrossRef Matsuo Y, Kihara T, Ikeda M, Ninomiya M, Onodera H, Kogure K. Role of neutrophils in radical production during ischemia and reperfusion of the rat brain: effect of neutrophil depletion on extracellular ascorbyl radical formation. J Cereb Blood Flow Metab. 1995;15:941–7.PubMedCrossRef
33.
Zurück zum Zitat Matsuo Y, Onodera H, Shiga Y, Nakamura M, Ninomiya M, Kihara T, et al. Correlation between myeloperoxidase-quantified neutrophil accumulation and ischemic brain injury in the rat. Effects of neutrophil depletion. Stroke. 1994;25:1469–75.PubMedCrossRef Matsuo Y, Onodera H, Shiga Y, Nakamura M, Ninomiya M, Kihara T, et al. Correlation between myeloperoxidase-quantified neutrophil accumulation and ischemic brain injury in the rat. Effects of neutrophil depletion. Stroke. 1994;25:1469–75.PubMedCrossRef
34.
Zurück zum Zitat Zhang R, Chopp M, Zhang Z, Jiang N, Powers C. The expression of P- and E-selectins in three models of middle cerebral artery occlusion. Brain Res. 1998;785:207–14.PubMedCrossRef Zhang R, Chopp M, Zhang Z, Jiang N, Powers C. The expression of P- and E-selectins in three models of middle cerebral artery occlusion. Brain Res. 1998;785:207–14.PubMedCrossRef
35.
Zurück zum Zitat Suzuki H, Abe K, Tojo SJ, Kitagawa H, Kimura K, Mizugaki M, et al. Reduction of ischemic brain injury by anti-P-selectin monoclonal antibody after permanent middle cerebral artery occlusion in rat. Neurol Res. 1999;21:269–76.PubMed Suzuki H, Abe K, Tojo SJ, Kitagawa H, Kimura K, Mizugaki M, et al. Reduction of ischemic brain injury by anti-P-selectin monoclonal antibody after permanent middle cerebral artery occlusion in rat. Neurol Res. 1999;21:269–76.PubMed
36.
Zurück zum Zitat Suzuki H, Hayashi T, Tojo SJ, Kitagawa H, Kimura K, Mizugaki M, et al. Anti-P-selectin antibody attenuates rat brain ischemic injury. Neurosci Lett. 1999;265:163–6.PubMedCrossRef Suzuki H, Hayashi T, Tojo SJ, Kitagawa H, Kimura K, Mizugaki M, et al. Anti-P-selectin antibody attenuates rat brain ischemic injury. Neurosci Lett. 1999;265:163–6.PubMedCrossRef
37.
Zurück zum Zitat Huang J, Choudhri TF, Winfree CJ, McTaggart RA, Kiss S, Mocco J, et al. Postischemic cerebrovascular E-selectin expression mediates tissue injury in murine stroke. Stroke. 2000;31:3047–53.PubMedCrossRef Huang J, Choudhri TF, Winfree CJ, McTaggart RA, Kiss S, Mocco J, et al. Postischemic cerebrovascular E-selectin expression mediates tissue injury in murine stroke. Stroke. 2000;31:3047–53.PubMedCrossRef
38.
Zurück zum Zitat Ishibashi S, Maric D, Mou Y, Ohtani R, Ruetzler C, Hallenbeck JM. Mucosal tolerance to E-selectin promotes the survival of newly generated neuroblasts via regulatory T-cell induction after stroke in spontaneously hypertensive rats. J Cereb Blood Flow Metab. 2009;29:606–20.PubMedCrossRef Ishibashi S, Maric D, Mou Y, Ohtani R, Ruetzler C, Hallenbeck JM. Mucosal tolerance to E-selectin promotes the survival of newly generated neuroblasts via regulatory T-cell induction after stroke in spontaneously hypertensive rats. J Cereb Blood Flow Metab. 2009;29:606–20.PubMedCrossRef
39.
Zurück zum Zitat Takeda H, Spatz M, Ruetzler C, McCarron R, Becker K, Hallenbeck J. Induction of mucosal tolerance to E-selectin prevents ischemic and hemorrhagic stroke in spontaneously hypertensive genetically stroke-prone rats. Stroke. 2002;33:2156–63.PubMedCrossRef Takeda H, Spatz M, Ruetzler C, McCarron R, Becker K, Hallenbeck J. Induction of mucosal tolerance to E-selectin prevents ischemic and hemorrhagic stroke in spontaneously hypertensive genetically stroke-prone rats. Stroke. 2002;33:2156–63.PubMedCrossRef
40.
Zurück zum Zitat Yenari MA, Sun GH, Kunis DM, Onley D, Vexler V. L-selectin inhibition does not reduce injury in a rabbit model of transient focal cerebral ischemia. Neurol Res. 2001;23:72–8.PubMedCrossRef Yenari MA, Sun GH, Kunis DM, Onley D, Vexler V. L-selectin inhibition does not reduce injury in a rabbit model of transient focal cerebral ischemia. Neurol Res. 2001;23:72–8.PubMedCrossRef
41.
Zurück zum Zitat Bednar MM, Gross CE, Russell SR, Fuller SP, Ellenberger CL, Schindler E, et al. Humanized anti-L-selectin monoclonal antibody DREG200 therapy in acute thromboembolic stroke. Neurol Res. 1998;20:403–8.PubMed Bednar MM, Gross CE, Russell SR, Fuller SP, Ellenberger CL, Schindler E, et al. Humanized anti-L-selectin monoclonal antibody DREG200 therapy in acute thromboembolic stroke. Neurol Res. 1998;20:403–8.PubMed
42.
Zurück zum Zitat Rieckmann P, Nunke K, Burchhardt M, Albrecht M, Wiltfang J, Ulrich M, et al. Soluble intercellular adhesion molecule-1 in cerebrospinal fluid: an indicator for the inflammatory impairment of the blood–cerebrospinal fluid barrier. J Neuroimmunol. 1993;47:133–40.PubMedCrossRef Rieckmann P, Nunke K, Burchhardt M, Albrecht M, Wiltfang J, Ulrich M, et al. Soluble intercellular adhesion molecule-1 in cerebrospinal fluid: an indicator for the inflammatory impairment of the blood–cerebrospinal fluid barrier. J Neuroimmunol. 1993;47:133–40.PubMedCrossRef
43.
Zurück zum Zitat Smith CW, Marlin SD, Rothlein R, Toman C, Anderson DC. Cooperative interactions of LFA-1 and Mac-1 with intercellular adhesion molecule-1 in facilitating adherence and transendothelial migration of human neutrophils in vitro. J Clin Invest. 1989;83:2008–17.PubMedCrossRef Smith CW, Marlin SD, Rothlein R, Toman C, Anderson DC. Cooperative interactions of LFA-1 and Mac-1 with intercellular adhesion molecule-1 in facilitating adherence and transendothelial migration of human neutrophils in vitro. J Clin Invest. 1989;83:2008–17.PubMedCrossRef
44.
Zurück zum Zitat Frijns CJ, Kappelle LJ. Inflammatory cell adhesion molecules in ischemic cerebrovascular disease. Stroke. 2002;33:2115–22.PubMedCrossRef Frijns CJ, Kappelle LJ. Inflammatory cell adhesion molecules in ischemic cerebrovascular disease. Stroke. 2002;33:2115–22.PubMedCrossRef
45.
Zurück zum Zitat Bowes MP, Zivin JA, Rothlein R. Monoclonal antibody to the ICAM-1 adhesion site reduces neurological damage in a rabbit cerebral embolism stroke model. Exp Neurol. 1993;119:215–9.PubMedCrossRef Bowes MP, Zivin JA, Rothlein R. Monoclonal antibody to the ICAM-1 adhesion site reduces neurological damage in a rabbit cerebral embolism stroke model. Exp Neurol. 1993;119:215–9.PubMedCrossRef
46.
Zurück zum Zitat Chopp M, Li Y, Jiang N, Zhang RL, Prostak J. Antibodies against adhesion molecules reduce apoptosis after transient middle cerebral artery occlusion in rat brain. J Cereb Blood Flow Metab. 1996;16:578–84.PubMedCrossRef Chopp M, Li Y, Jiang N, Zhang RL, Prostak J. Antibodies against adhesion molecules reduce apoptosis after transient middle cerebral artery occlusion in rat brain. J Cereb Blood Flow Metab. 1996;16:578–84.PubMedCrossRef
47.
Zurück zum Zitat Zhang RL, Chopp M, Li Y, Zaloga C, Jiang N, Jones ML, et al. Anti-ICAM-1 antibody reduces ischemic cell damage after transient middle cerebral artery occlusion in the rat. Neurology. 1994;44:1747–51.PubMedCrossRef Zhang RL, Chopp M, Li Y, Zaloga C, Jiang N, Jones ML, et al. Anti-ICAM-1 antibody reduces ischemic cell damage after transient middle cerebral artery occlusion in the rat. Neurology. 1994;44:1747–51.PubMedCrossRef
48.
Zurück zum Zitat Bowes MP, Rothlein R, Fagan SC, Zivin JA. Monoclonal antibodies preventing leukocyte activation reduce experimental neurologic injury and enhance efficacy of thrombolytic therapy. Neurology. 1995;45:815–9.PubMedCrossRef Bowes MP, Rothlein R, Fagan SC, Zivin JA. Monoclonal antibodies preventing leukocyte activation reduce experimental neurologic injury and enhance efficacy of thrombolytic therapy. Neurology. 1995;45:815–9.PubMedCrossRef
49.
Zurück zum Zitat Zhang RL, Chopp M, Jiang N, Tang WX, Prostak J, Manning AM, et al. Anti-intercellular adhesion molecule-1 antibody reduces ischemic cell damage after transient but not permanent middle cerebral artery occlusion in the Wistar rat. Stroke. 1995;26:1438–42.PubMedCrossRef Zhang RL, Chopp M, Jiang N, Tang WX, Prostak J, Manning AM, et al. Anti-intercellular adhesion molecule-1 antibody reduces ischemic cell damage after transient but not permanent middle cerebral artery occlusion in the Wistar rat. Stroke. 1995;26:1438–42.PubMedCrossRef
50.
Zurück zum Zitat Enlimomab Acute Stroke Trial Investigators. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57:1428–34.CrossRef Enlimomab Acute Stroke Trial Investigators. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57:1428–34.CrossRef
51.
Zurück zum Zitat Jaeschke H, Farhood A, Bautista AP, Spolarics Z, Spitzer JJ, Smith CW. Functional inactivation of neutrophils with a Mac-1 (CD11b/CD18) monoclonal antibody protects against ischemia–reperfusion injury in rat liver. Hepatology. 1993;17:915–23.PubMedCrossRef Jaeschke H, Farhood A, Bautista AP, Spolarics Z, Spitzer JJ, Smith CW. Functional inactivation of neutrophils with a Mac-1 (CD11b/CD18) monoclonal antibody protects against ischemia–reperfusion injury in rat liver. Hepatology. 1993;17:915–23.PubMedCrossRef
52.
Zurück zum Zitat Simpson PJ, Todd 3rd RF, Fantone JC, Mickelson JK, Griffin JD, Lucchesi BR. Reduction of experimental canine myocardial reperfusion injury by a monoclonal antibody (anti-Mo1, anti-CD11b) that inhibits leukocyte adhesion. J Clin Invest. 1988;81:624–9.PubMedCrossRef Simpson PJ, Todd 3rd RF, Fantone JC, Mickelson JK, Griffin JD, Lucchesi BR. Reduction of experimental canine myocardial reperfusion injury by a monoclonal antibody (anti-Mo1, anti-CD11b) that inhibits leukocyte adhesion. J Clin Invest. 1988;81:624–9.PubMedCrossRef
53.
Zurück zum Zitat Simpson PJ, Todd 3rd RF, Mickelson JK, Fantone JC, Gallagher KP, Lee KA, et al. Sustained limitation of myocardial reperfusion injury by a monoclonal antibody that alters leukocyte function. Circulation. 1990;81:226–37.PubMedCrossRef Simpson PJ, Todd 3rd RF, Mickelson JK, Fantone JC, Gallagher KP, Lee KA, et al. Sustained limitation of myocardial reperfusion injury by a monoclonal antibody that alters leukocyte function. Circulation. 1990;81:226–37.PubMedCrossRef
54.
Zurück zum Zitat Chen H, Chopp M, Zhang RL, Bodzin G, Chen Q, Rusche JR, et al. Anti-CD11b monoclonal antibody reduces ischemic cell damage after transient focal cerebral ischemia in rat. Ann Neurol. 1994;35:458–63.PubMedCrossRef Chen H, Chopp M, Zhang RL, Bodzin G, Chen Q, Rusche JR, et al. Anti-CD11b monoclonal antibody reduces ischemic cell damage after transient focal cerebral ischemia in rat. Ann Neurol. 1994;35:458–63.PubMedCrossRef
55.
Zurück zum Zitat Chopp M, Zhang RL, Chen H, Li Y, Jiang N, Rusche JR. Postischemic administration of an anti-Mac-1 antibody reduces ischemic cell damage after transient middle cerebral artery occlusion in rats. Stroke. 1994;25:869–75.PubMedCrossRef Chopp M, Zhang RL, Chen H, Li Y, Jiang N, Rusche JR. Postischemic administration of an anti-Mac-1 antibody reduces ischemic cell damage after transient middle cerebral artery occlusion in rats. Stroke. 1994;25:869–75.PubMedCrossRef
56.
Zurück zum Zitat Bednar MM, Wright SD, Raymond-Russell SJ, Kohut JJ, Gross CE. IB4, a monoclonal antibody against the CD18 leukocyte adhesion protein, reduces intracranial pressure following thromboembolic stroke in the rabbit. Neurol Res. 1996;18:171–5.PubMed Bednar MM, Wright SD, Raymond-Russell SJ, Kohut JJ, Gross CE. IB4, a monoclonal antibody against the CD18 leukocyte adhesion protein, reduces intracranial pressure following thromboembolic stroke in the rabbit. Neurol Res. 1996;18:171–5.PubMed
57.
Zurück zum Zitat Colditz I, Zwahlen R, Dewald B, Baggiolini M. In vivo inflammatory activity of neutrophil-activating factor, a novel chemotactic peptide derived from human monocytes. Am J Pathol. 1989;134:755–60.PubMed Colditz I, Zwahlen R, Dewald B, Baggiolini M. In vivo inflammatory activity of neutrophil-activating factor, a novel chemotactic peptide derived from human monocytes. Am J Pathol. 1989;134:755–60.PubMed
58.
Zurück zum Zitat Huber AR, Kunkel SL, Todd 3rd RF, Weiss SJ. Regulation of transendothelial neutrophil migration by endogenous interleukin-8. Science. 1991;254:99–102.PubMedCrossRef Huber AR, Kunkel SL, Todd 3rd RF, Weiss SJ. Regulation of transendothelial neutrophil migration by endogenous interleukin-8. Science. 1991;254:99–102.PubMedCrossRef
59.
Zurück zum Zitat Al-Bahrani A, Taha S, Shaath H, Bakhiet M. TNF-alpha and IL-8 in acute stroke and the modulation of these cytokines by antiplatelet agents. Curr Neurovasc Res. 2007;4:31–7.PubMedCrossRef Al-Bahrani A, Taha S, Shaath H, Bakhiet M. TNF-alpha and IL-8 in acute stroke and the modulation of these cytokines by antiplatelet agents. Curr Neurovasc Res. 2007;4:31–7.PubMedCrossRef
60.
Zurück zum Zitat Kostulas N, Pelidou SH, Kivisakk P, Kostulas V, Link H. Increased IL-1beta, IL-8, and IL-17 mRNA expression in blood mononuclear cells observed in a prospective ischemic stroke study. Stroke. 1999;30:2174–9.PubMedCrossRef Kostulas N, Pelidou SH, Kivisakk P, Kostulas V, Link H. Increased IL-1beta, IL-8, and IL-17 mRNA expression in blood mononuclear cells observed in a prospective ischemic stroke study. Stroke. 1999;30:2174–9.PubMedCrossRef
61.
Zurück zum Zitat Wittwer AJ, Carr LS, Zagorski J, Dolecki GJ, Crippes BA, De Larco JE. High-level expression of cytokine-induced neutrophil chemoattractant (CINC) by a metastatic rat cell line: purification and production of blocking antibodies. J Cell Physiol. 1993;156:421–7.PubMedCrossRef Wittwer AJ, Carr LS, Zagorski J, Dolecki GJ, Crippes BA, De Larco JE. High-level expression of cytokine-induced neutrophil chemoattractant (CINC) by a metastatic rat cell line: purification and production of blocking antibodies. J Cell Physiol. 1993;156:421–7.PubMedCrossRef
62.
Zurück zum Zitat Yamasaki Y, Matsuo Y, Zagorski J, Matsuura N, Onodera H, Itoyama Y, et al. New therapeutic possibility of blocking cytokine-induced neutrophil chemoattractant on transient ischemic brain damage in rats. Brain Res. 1997;759:103–11.PubMedCrossRef Yamasaki Y, Matsuo Y, Zagorski J, Matsuura N, Onodera H, Itoyama Y, et al. New therapeutic possibility of blocking cytokine-induced neutrophil chemoattractant on transient ischemic brain damage in rats. Brain Res. 1997;759:103–11.PubMedCrossRef
63.
Zurück zum Zitat Bachmann MF, Dyer MR. Therapeutic vaccination for chronic diseases: a new class of drugs in sight. Nat Rev Drug Discov. 2004;3:81–8.PubMedCrossRef Bachmann MF, Dyer MR. Therapeutic vaccination for chronic diseases: a new class of drugs in sight. Nat Rev Drug Discov. 2004;3:81–8.PubMedCrossRef
64.
Zurück zum Zitat Buttini M, Appel K, Sauter A, Gebicke-Haerter PJ, Boddeke HW. Expression of tumor necrosis factor alpha after focal cerebral ischaemia in the rat. Neuroscience. 1996;71:1–16.PubMedCrossRef Buttini M, Appel K, Sauter A, Gebicke-Haerter PJ, Boddeke HW. Expression of tumor necrosis factor alpha after focal cerebral ischaemia in the rat. Neuroscience. 1996;71:1–16.PubMedCrossRef
65.
Zurück zum Zitat Saito K, Suyama K, Nishida K, Sei Y, Basile AS. Early increases in TNF-alpha, IL-6 and IL-1 beta levels following transient cerebral ischemia in gerbil brain. Neurosci Lett. 1996;206:149–52.PubMedCrossRef Saito K, Suyama K, Nishida K, Sei Y, Basile AS. Early increases in TNF-alpha, IL-6 and IL-1 beta levels following transient cerebral ischemia in gerbil brain. Neurosci Lett. 1996;206:149–52.PubMedCrossRef
66.
Zurück zum Zitat Lavine SD, Hofman FM, Zlokovic BV. Circulating antibody against tumor necrosis factor-alpha protects rat brain from reperfusion injury. J Cereb Blood Flow Metab. 1998;18:52–8.PubMedCrossRef Lavine SD, Hofman FM, Zlokovic BV. Circulating antibody against tumor necrosis factor-alpha protects rat brain from reperfusion injury. J Cereb Blood Flow Metab. 1998;18:52–8.PubMedCrossRef
67.
Zurück zum Zitat Meistrell 3rd ME, Botchkina GI, Wang H, Di Santo E, Cockroft KM, Bloom O, et al. Tumor necrosis factor is a brain damaging cytokine in cerebral ischemia. Shock. 1997;8:341–8.PubMedCrossRef Meistrell 3rd ME, Botchkina GI, Wang H, Di Santo E, Cockroft KM, Bloom O, et al. Tumor necrosis factor is a brain damaging cytokine in cerebral ischemia. Shock. 1997;8:341–8.PubMedCrossRef
68.
Zurück zum Zitat Martin-Villalba A, Hahne M, Kleber S, Vogel J, Falk W, Schenkel J, et al. Therapeutic neutralization of CD95-ligand and TNF attenuates brain damage in stroke. Cell Death Differ. 2001;8:679–86.PubMedCrossRef Martin-Villalba A, Hahne M, Kleber S, Vogel J, Falk W, Schenkel J, et al. Therapeutic neutralization of CD95-ligand and TNF attenuates brain damage in stroke. Cell Death Differ. 2001;8:679–86.PubMedCrossRef
69.
Zurück zum Zitat Dalakas MC. High-dose intravenous immunoglobulin and serum viscosity: risk of precipitating thromboembolic events. Neurology. 1994;44:223–6.PubMedCrossRef Dalakas MC. High-dose intravenous immunoglobulin and serum viscosity: risk of precipitating thromboembolic events. Neurology. 1994;44:223–6.PubMedCrossRef
70.
Zurück zum Zitat Cosimi AB, Conti D, Delmonico FL, Preffer FI, Wee SL, Rothlein R, et al. In vivo effects of monoclonal antibody to ICAM-1 (CD54) in nonhuman primates with renal allografts. J Immunol. 1990;144:4604–12.PubMed Cosimi AB, Conti D, Delmonico FL, Preffer FI, Wee SL, Rothlein R, et al. In vivo effects of monoclonal antibody to ICAM-1 (CD54) in nonhuman primates with renal allografts. J Immunol. 1990;144:4604–12.PubMed
71.
Zurück zum Zitat Schneider D, Berrouschot J, Brandt T, Hacke W, Ferbert A, Norris SH, et al. Safety, pharmacokinetics and biological activity of enlimomab (anti-ICAM-1 antibody): an open-label, dose escalation study in patients hospitalized for acute stroke. Eur Neurol. 1998;40:78–83.PubMedCrossRef Schneider D, Berrouschot J, Brandt T, Hacke W, Ferbert A, Norris SH, et al. Safety, pharmacokinetics and biological activity of enlimomab (anti-ICAM-1 antibody): an open-label, dose escalation study in patients hospitalized for acute stroke. Eur Neurol. 1998;40:78–83.PubMedCrossRef
72.
Zurück zum Zitat Furuya K, Takeda H, Azhar S, McCarron RM, Chen Y, Ruetzler CA, et al. Examination of several potential mechanisms for the negative outcome in a clinical stroke trial of enlimomab, a murine anti-human intercellular adhesion molecule-1 antibody: a bedside-to-bench study. Stroke. 2001;32:2665–74.PubMedCrossRef Furuya K, Takeda H, Azhar S, McCarron RM, Chen Y, Ruetzler CA, et al. Examination of several potential mechanisms for the negative outcome in a clinical stroke trial of enlimomab, a murine anti-human intercellular adhesion molecule-1 antibody: a bedside-to-bench study. Stroke. 2001;32:2665–74.PubMedCrossRef
73.
Zurück zum Zitat Vuorte J, Lindsberg PJ, Kaste M, Meri S, Jansson SE, Rothlein R, et al. Anti-ICAM-1 monoclonal antibody R6.5 (Enlimomab) promotes activation of neutrophils in whole blood. J Immunol. 1999;162:2353–7.PubMed Vuorte J, Lindsberg PJ, Kaste M, Meri S, Jansson SE, Rothlein R, et al. Anti-ICAM-1 monoclonal antibody R6.5 (Enlimomab) promotes activation of neutrophils in whole blood. J Immunol. 1999;162:2353–7.PubMed
Metadaten
Titel
Therapeutic Antibodies in Stroke
verfasst von
Chye Yun Yu
Gandi Ng
Ping Liao
Publikationsdatum
01.10.2013
Verlag
Springer US
Erschienen in
Translational Stroke Research / Ausgabe 5/2013
Print ISSN: 1868-4483
Elektronische ISSN: 1868-601X
DOI
https://doi.org/10.1007/s12975-013-0281-2

Weitere Artikel der Ausgabe 5/2013

Translational Stroke Research 5/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.