3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

download 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

of 12

Transcript of 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    1/12

    Neuroprotection for ischaemic stroke: Translation fromthe bench to the bedside

    Brad A. Sutherland1†, Jens Minnerup2†, Joyce S. Balami3, Francesco Arba1,Alastair M. Buchan1*‡, and Christoph Kleinschnitz4‡

    Neuroprotection seeks to restrict injury to the brain paren-

    chyma following an ischaemic insult by preventing salvage-

    able neurons from dying. The concept of neuroprotection

    has shown promise in experimental studies, but has failed to

    translate into clinical success. Many reasons exist for this

    including the heterogeneity of human stroke and the lack ofmethodological agreement between preclinical and clinical

    studies. Even with the proposed Stroke Therapy Academic

    Industry Roundtable criteria for preclinical development of

    neuroprotective agents for stroke, we have still seen limited

    success in the clinic, an example being NXY-059, which ful-

    filled nearly all the Stroke Therapy Academic Industry

    Roundtable criteria. There are currently a number of ongoing

    trials for neuroprotective strategies including hypothermia

    and albumin, but the outcome of these approaches remains

    to be seen. Combination therapies with thrombolysis also

    need to be fully investigated, as restoration of oxygen and

    glucose will always be the best therapy to protect against

    cell death from stroke. There are also a number of promising

    neuroprotectants in preclinical development including hae-

    matopoietic growth factors, and inhibitors of the nicotina-

    mide adenine dinucleotide phosphate oxidases, a source of

    free radical production which is a key step in the pathophysi-

    ology of acute ischaemic stroke. For these neuroprotectants

    to succeed, essential quality standards need to be adhered

    to; however, these must remain realistic as the evidence that

    standardization of procedures improves translational success

    remains absent for stroke.

    Key words: acute stroke therapy, ischaemic cascade, ischaemicstroke, neuroprotection, STAIR, translation

    Introduction to neuroprotection

    Currently, the only approved measures for the treatment of 

    acute ischaemic stroke are thrombolysis and antiplatelet

    therapy. However, the concept of neuroprotection has received

    significant attention over the past 30 years, with many experi-

    mental neuroprotectants being trialled preclinically and clini-

    cally. Where thrombolysis aims to break down the occluding

    clot to restore blood flow to the ischaemic brain, neuroprotec-

    tion seeks to limit ischaemic injury by preventing the salvage-able neurons in the penumbra that surrounds the core from

    dying. Rapid restoration of oxygen and glucose by thromboly-

    sis will always provide the most effective neuroprotection, but

    directly targeting the brain parenchyma to confer neuropro-

    tection may be a viable method, particularly in conjunction

    with thrombolysis. Many well-defined molecular targets

    (Fig. 1) now exist within the ischaemic cascade that can, in

    theory, be pharmacologically altered to produce neuroprotec-

    tion (1). Neuroprotective agents aim to salvage ischaemic

    tissue, limit infarct size, prolong the time window for throm-

    bolytic therapy or minimize post-ischaemic reperfusion

    injury or inflammation. Over 1000 neuroprotective agents

    have been tested in basic stroke studies (2) with many showing

    promise. Despite this, neuroprotection in the clinic has failed

    to eventuate, disappointing clinicians, researchers, and stroke

    patients alike. Nearly 200 neuroprotection clinical trials are

    ongoing or have been completed, with none achieving success-

    ful translation to clinical practice so far (3).

    This review attempts to define neuroprotection and outline

    the current status of the neuroprotection field both preclini-

    cally and clinically, while identifying the problems associated

    with translating neuroprotection from the bench to bedside. It

    also describes some promising neuroprotectants, the use of 

    Correspondence: Alastair M Buchan*, Acute Stroke Programme, Level 7

    John Radcliffe Hospital, Oxford Biomedical Research Centre, Nuffield

    Department of Clinical Medicine, University of Oxford, Headley Way,

    Oxford, OX3 9DU, UK.

    E-mail: [email protected] 1Acute Stroke Programme, Nuffield Department of Clinical Medicine,

    University of Oxford, Oxford, UK2Department of Neurology, University of Münster, Münster, Germany 3Acute Stroke Programme, Department of Medicine and Clinical

    Geratology, Oxford University NHS Trust, Oxford, UK4Department of Neurology, University of Würzburg, Würzburg,

    Germany 

    Conflict of interest: The authors declare no conflict of interest with this

    manuscript.

    †Equal contribution;

    ‡ joint senior authors.

    DOI: 10.1111/j.1747-4949.2012.00770.x

    Review

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke OrganizationVol 7, July 2012, 407–418   407

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    2/12

    neuroprotection alongside thrombolysis, and finally some

    concerns with the current criteria for preclinical neuroprotec-

    tion studies.

    How to define and measure neuroprotection

    As described earlier, neuroprotection is designed to restrict

    injury to the brain following an injurious ischaemic insult by 

    preventing neuronal cell death, especially in the salvageable

    penumbral region. This leads to the working definition of 

    neuroprotection as ‘any strategy, or combination of strate-

    gies, that antagonizes, interrupts, or slows the sequence of 

    injurious biochemical and molecular events that, if left

    unchecked, would eventuate in irreversible ischemic injury’

    (4). According to this definition, protection from injury 

    originates at the neuron itself (endogenous or direct neuro-

    protection). Consequently, it does not include treatment

    approaches that primarily target the cerebral vasculature,

    such as thrombolytics, antithrombotics, and antiplatelet

    drugs (extrinsic or indirect neuroprotection) (4). Even

    though these agents do protect the brain by restoring blood

    flow and preventing clot formation, their mechanisms of 

    action are vascular-based and do not target the brain paren-

    chyma itself. Nevertheless, given the wide array of biochemi-

    Fig. 1   The cascade of biochemical events leading to apoptosis or necrosis following cerebral ischaemia. Vascular occlusion in a blood vessel initiates a

    complex signalling cascade that leads to neuronal cell death. The reduction in blood flow produces ionic pump failure and anoxic depolarization leading

    to enhanced glutamate release and a sudden increase in intracellular calcium. This rise in calcium triggers mitochondrial collapse, free radical production,

    cytotoxic oedema, and increased NO generation. Reperfusion also produces injury by augmenting BBB breakdown, inflammation, and free radical

    production leading to apoptosis. Red borders signify important events in the cascade. The blue border indicates reperfusion. This figure has been adapted

    from Durukan & Tatlisumak (1). AA, arachidonic acid; AMPA,  a -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; BBB, blood-brain barrier; iNOS,

    inducible nitric oxide synthase; NMDA, N-methyl-D-aspartate; nNOS, neuronal nitric oxide synthase; NO, nitric oxide; PLA 2, phospholipase A2.

    Review   B. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke Organization408   Vol 7, July 2012, 407–418

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    3/12

    cal pathways that have been elucidated to play a role inischaemic cell death (Fig. 1), this working definition still

    covers an extensive variety of potential neuroprotective

    agents.

    In the scope of published neuroprotective preclinical

    studies, there is significant variability in the quantity of neu-

    roprotection achieved. Some agents produced substantial pro-

    tection of the brain following ischaemia (e.g. NXY-059), while

    others showed minimal neuroprotection (e.g. edaravone) (2).

    Also, based on the Stroke Therapy Academic Industry Round-

    table (STAIR) criteria (5,6), many neuroprotective studies

    exhibit low methodological quality (2) with a wide heteroge-

    neity in the methodology used. This has led to increasingly variable results meaning many neuroprotective agents pro-

    ceeded to clinical investigation with only weak preclinical evi-

    dence and so were doomed to fail. There are many other

    reasons why clinical neuroprotection has not eventuated given

    the preclinical success, which are described in Table 1 and in

    many other reports (7–9).

    It is hard to gauge from the experimental evidence and the

    low methodological quality whether there are really any 

    drugs that can induce ‘true’ neuroprotection. It is possible

    that many of the neuroprotective effects observed could be

    due to a manifestation of physiological/pathophysiological

    changes following ischaemia. These changes could include

    modulating temperature (hypothermia), cerebral blood flow 

    (CBF; hyperperfusion), inflammation (anti-inflammatory 

    effects), and blood-brain barrier (BBB) damage (reducing

    BBB disruption and vascular permeability). One example is

    the N-methyl-D-aspartate (NMDA) receptor antagonist

    MK-801, which induced hypothermia to produce neuropro-

    tection instead of directly targeting the neuron (10). Inter-

    estingly, the same compound also raised CBF in the

    ischaemic region which may have contributed to its neuro-

    protective effects (11). The concept of neuroprotection pro-

    duced by enhancing CBF rather than inhibiting the

    ischaemic cascade has recently been discussed (12). Sophis-ticated imaging tools might help to delineate genuine neu-

    roprotection from within the neuron from secondary or

    non-specific effects in the future. This was demonstrated in a

    mouse model of multiple sclerosis where a new form of early 

    but reversible axonal damage caused by oxidative stress and

    successive mitochondrial dysfunction was visualized  in vivo

    by serial two-photon microscopy (13).

    Another problem in animal studies is how neuroprotection

    is assessed. Infarct volume is most commonly used as the

    primary end-point and is quantified by using a histological

    stain. The majority of experiments claim that neuroprotection

    has been achieved when there is a reduction in infarct volume.However, this is difficult to translate to human studies where

    neuroprotection would be reached if stroke patients received

    sustainable functional benefit. So what does neuroprotection

    really mean if infarct volume is reduced but functional

    improvement is lacking? Clearly, functional assessment

    including mortality rates in preclinical studies should be a

    mandatory outcome parameter for the investigation of any 

    neuroprotective drug, as outlined by STAIR (5,6). Unfortu-

    nately, meaningful functional testing in small laboratory 

    animals, especially mice, is frequently hampered by stroke

    severity and the limited correlation with higher brain func-

    tioning in humans, and so representative tests must be care-

    fully selected.

    The current status of neuroprotection

    Even after 30 years of neuroprotection research, no neuro-

    protective therapy has been brought into clinical practice.

    However, there are some exciting new developments in the

    neuroprotection field, and this section will describe the

    current status of neuroprotection in both preclinical animal

    research and clinical human research.

    Table 1  Reasons for translational failure of neuroprotective agents from pre-clinical to clinical studies

    Animal models Human studies

    Highly control led, homogeneous population Variable, heterogeneous population

    Younger animals Older patients

    Limited comorbidities Numerous comorbidities

    Induced onset of stroke Spontaneous onset of strokeUniform aetiology Variable aetiologies

    Ischaemic territory usually from middle cerebral artery Ischaemic territory not restricted to middle cerebral artery

    Control over therapeutic time window (usually early treatment) Less control over therapeutic time window (usually delayed treatment)

    Controlled occlusion duration Variable occlusion duration

    Adequate sample size Inadequate sample size

    Wide scope for dose optimization Reduced scope for dose optimization

    Multiple routes of administration Limited routes of administration

    Rapid availability of the drugs to the target area Slow availability of the drugs to the target area

    Infarct volume as outcome Function as outcome

    ReviewB. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke OrganizationVol 7, July 2012, 407–418   409

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    4/12

    Preclinical animal research

    The development of new neuroprotective therapies for stroke

    involves the evaluation of candidate drugs from   in vitro

    models to animal experiments and, finally, testing in clinical

    trials. Animal studies not only allow the determination of a

    drug’s efficacy but also can elucidate its underlying mecha-

    nisms in stroke pathophysiology. Up to now, numerous drugstargeting different aspects of the ischaemic cascade (Fig. 1)

    were tested in animal models of focal cerebral ischaemia. Drug

    mechanisms that were found to be successful in experimental

    studies regarding both infarct size reduction and improved

    functional outcome include impeding excitotoxicity, local

    inflammation, neuronal apoptosis, free radical damage, and

    calcium influx into cells. As reviewed by O’Collins  et al . (2),

    some of these mechanisms of brain injury were targeted by 

    more than 10 different agents in hundreds of experiments. For

    example, the authors identified 277 studies of 21 drugs aiming

    to attenuate excitotoxicity in experimental stroke and 114

    studies relating to nine drugs with anti-inflammatory activi-ties (2). Overall, 1026 candidate stroke drugs have been iden-

    tified in the period covering 1957 to 2003 (2). A more recent

    survey of Pubmed-referenced publications showed that the

    number of experimental studies of candidate neuroprotective

    drugs for stroke therapy has particularly increased over the last

    15 years (4). Nearly two-thirds of the published studies report

    an improved outcome with a neuroprotective compound

    compared to control treatment in animal models of focal cer-

    ebral ischaemia (2). However, there appears to be significant

    publication bias in preclinical stroke studies which may 

    account for approximately one-third of the efficacy reported

    in meta-analyses, leading to an overstatement of efficacy (14).

    In contrast, in clinical trials, drugs targeting only one key 

    mechanism of cerebral ischaemia have failed to improve

    outcome as discussed later in this review. One plausible reason

    for this failure might be the multiplicity of mechanisms

    involved in causing neuronal damage following stroke (Fig. 1).

    Therefore, a novel approach for the development of neuropro-

    tective drugs includes the evaluation of compounds with a

    multimodal mode of action. This concept also considers the

    use of agents with recovery-enhancing properties in addition

    to neuroprotective actions (15,16). A recent trend for judging

    the potency of neuroprotectants is to pool results from differ-

    ent animal studies for meta-analysis. This method, originally 

    applied for clinical trials in humans, was recently used toobtain further information on the efficacy, the dose–response

    relationship, and the therapeutic time window of promising

    stroke drugs for potential guidance of clinical trials (17–19).

    Clinical human research

    A considerable number of neuroprotection clinical trials for

    ischaemic stroke are ongoing or have been completed. Viewing

    the Internet Stroke Centre Stroke Trials Registry (3), a number

    of agents possessing a wide variety of mechanisms of action

    have been tried. Unfortunately, none of these agents have

    achieved clinical success for neuroprotection. However, there

    are some promising ongoing studies that, among others,

    include the use of hypothermia, albumin, magnesium, mino-

    cycline, and statins as potential approaches to neuroprotection

    in the clinical setting. The ongoing clinical trials investigating

    neuroprotection for acute ischaemic stroke have been outlinedin Table 2. A list of neuroprotective strategies that have under-

    gone completed studies for acute ischaemic stroke and that

    have all failed to show neuroprotection in the clinic is shown

    in Supporting Information Table S1.

    Hypothermia

    Hypothermia is one of the most promising neuroprotective

    approaches, which has consistently shown benefit in animal

    models of cerebral ischaemia, reducing infarct volume by 

    more than 40% (21). Hypothermia is thought to be neuropro-

    tective through several mechanisms including decreasing exci-

    tatory amino acid release, reducing free radical formation,enhancing small ubiquitin-related modifier (SUMO)-related

    pathways, attenuating protein kinase C activity, and slowing

    cellular metabolism (22–24).

    The Cooling for Acute Ischaemic Brain Damage (COOL-

    AID) studies, COOL-AID I (using surface cooling) (25), and

    COOL-AID II (using endovascular cooling) (26) showed that

    mild therapeutic hypothermia for acute ischaemic stroke was

    feasible, but with no change in clinical outcome. The recent

    Intravascular Cooling in the Treatment of Stroke – Longer

    recombinant tissue plasminogen activator (rtPA) window 

    study showed that catheter-based cooling within 6 h of 

    symptom onset of acute stroke was well tolerated in patients

    given rtPA, but there were no differences in 90-day outcomes

    (27). Other safety and efficacy clinical studies such as Control-

    led Hypothermia in Large Infarction and Cooling in Acute

    Stroke are ongoing (3).

    Despite the encouraging results from hypothermia studies

    in humans, there are a number of limitations in applying

    hypothermia to stroke patients. Stroke patients are generally 

    awake and do not tolerate cooling in contrast to cardiac arrest

    and brain injury patients. Attaining target temperature and

    prolonging or maintaining that temperature stroke patients

    while awake is challenging. There are frequent complications

    such as pneumonia, hypotension, cardiac arrhythmias, elec-

    trolyte derangements, and infections (27–29). A number of patients also experience shivering during cooling which can

    be controlled with anti-shivering agents such as buspirone

    and meperidine (28). Another problem is the rebound

    increase in intracranial pressure experienced during re-

    warming; a phenomenon that is not well studied in laboratory 

    models (30).

    Albumin

    Albumin, a protein involved in the transport of small mol-

    ecules in the blood, plays a key role in restricting fluid leaking

    Review   B. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke Organization410   Vol 7, July 2012, 407–418

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    5/12

    from the vasculature into the tissue (31). In animal studies,

    albumin was shown to diminish infarct volume significantly 

    with a therapeutic time window of four-hours poststroke (32).

    Albumin produces its neuroprotective effect through several

    mechanisms including ameliorating brain swelling, enhancing

    blood flow to sub-occlusive microvascular lesions, maintain-

    ing vascular patency, and preventing re-occlusion after suc-

    cessful thrombolysis (4).

    The pilot study Albumin in Acute Stroke (ALIAS) demon-

    strated that high-dose human albumin therapy is safe and may 

    confer a neuroprotective effect within five-hours after acute

    ischaemic stroke (33,34). These encouraging results have led

    to a large placebo-controlled randomized multicentre phase lll

    trial of albumin therapy in acute ischaemic stroke – ALIAS-

    Part 2 – which is ongoing (35).

    The preclinical evidence for the validity of albumin as a

    neuroprotective agent is limited in that albumin efficacy in

    focal cerebral ischaemia was mainly described by only one

    group and independent confirmation by others is pending.

    Magnesium

    Magnesium may have significant neuroprotective properties

    in stroke, with preclinical evidence revealing a 25% level of 

    protection (36). Magnesium produces this protection through

    a number of mechanisms including antagonism of calcium

    channels, noncompetitive antagonism of NMDA receptors,

    inhibition of excitatory neurotransmitter release, and vascular

    smooth muscle relaxation (37). However, magnesium can

    also produce post-ischaemic hypothermia which could con-

    tribute to its neuroprotective effects in studies that were not

    temperature-controlled (38). Looking at studies that were

    temperature-controlled, magnesium was largely ineffective

    (38) suggesting that magnesium may only produce neuropro-

    tection in concert with hypothermia.

    Much of the failure of previous neuroprotective trials may 

    be due to the delayed delivery of agents to stroke patients. The

    Field Administration of Stroke Therapy–Magnesium (FAST-

    MAG) Pilot Trial attempted to overcome this by having para-

    medics initiate magnesium sulphate therapy in acute stroke

    patients in the field before arrival to the hospital (39). The

    field-based magnesium intervention was feasible and safe with

    no serious adverse effects, and was associated with a beneficial

    functional outcome at three-months. Based on these positive

    results, a large phase lll clinical trial is already in progress

    (FAST-MAG) (3).

    Although magnesium might act pleiotropically on ischae-

    mic neurons, powerful effects of this naturally occurring elec-

    trolyte on stroke outcome in clinical practice may be

    surprising, especially when given as a monotherapy. A combi-

    natory approach with other neuroprotective or clot-breakingagents may be more promising.

    Minocycline

    Minocycline is a tetracycline antibiotic, which has been shown

    to produce a 30% reduction in infarct size in models of cer-

    ebral ischaemia (36). The proposed mechanisms of action of 

    minocycline include anti-inflammatory effects, reduction of 

    microglial activation, matrix metalloproteinase activity, and

    nitric oxide (NO) production, and inhibition of apoptosis

    (40). Moreover, via its antibacterial properties, minocycline

    Table 2  Neuroprotective compounds currently undergoing clinical trials*

    Category Name(s) Mechanism

    Clinical

    phase Manufacturer

    Antioxidant Ebselen Free radical scavenger III Daiichi Pharmaceutical Co., LTD

    Edaravone (MCI-186) Free radical scavenger III Mitsubishi Pharma Corporation

    Anti-apoptotic/ regeneration AX200 (filgrastim, G-CSF analogue) Growth factor II Sygnis Bioscience GmbH & Co KGHuman Chorionic Gonadotropin

    (hCG)/Erythropoietin (NTx-265)

    Growth factors, oxygen delivery I I Stem Cel l Therapeutics Corp.

    Excitotoxicity Magnesium sulphate NMDA ion channel blocker III Many manufacturers (Abbott

    Laboratories for FAST-MAG)

    Fluid regulators Albumin Haemodiluting agent III Baxter Bioscience

    Others Citicoline (CDP choline) Membrane stabilizer III Ferrer Grupo

    Deferoxamine mesylate Iron chelator II Novartis Pharma

    DP-b99 Metal ion chelator III D-Pharm Ltd

    Hemicraniectomy Reduce cerebral oedema and intracranial

    pressure

    III None

    Hypothermia Reduce cerebral oxygen metabolism,

    synaptic inhibitor

    III None

    Insulin Reduce glucose and brain damage III Eli Lilly

    Lovastatin HMG CoA reductase inhibitor, antioxidant II Many manufacturers

    Minocycline Antibiotic, pleiotropic protective effects III WyethSimvastatin HMG CoA reductase inhibitor, antioxidant III Many manufacturers

    *Information gathered from Stroke Trials Registry (3), O’Collins  et al . (2), and Cochrane Clinical Trials Database (20).

    ReviewB. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke OrganizationVol 7, July 2012, 407–418   411

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    6/12

    could reduce infections such as pneumonia or urinary tract

    infections resulting from stroke-induced immunosuppression

    (41).

    In an open-label evaluator study, minocycline administra-

    tion led to a significantly better outcome in acute stroke

    patients compared to placebo (40). Similarly, in Minocycline

    to Improve Neurological Outcome in Stroke, minocycline wassafe and well tolerated alone and in combination with rtPA

    (42). Encouraging results from these trials have led to the

    ongoing Phase III Neuroprotection with Minocycline Therapy 

    for Acute Stroke Recovery Trial (43).

    While the efficacy and neuroprotective potential of mino-

    cycline in acute ischaemic stroke still need to be established,

    this antibiotic has been used in clinical practice for many 

     years without serious safety concerns. Nevertheless, wide-

    spread and uncritical application of anti-infective agents

    could promote the occurrence of multiresistant and invasive

    pathogens especially in the setting of intensive care units or

    stroke wards.

    Statins

    Hydroxymethylglutaryl–coenzyme A (HMG-CoA) reductase

    inhibitors (statins) are the most widely used cholesterol-

    lowering drugs. In addition to their well-established role for

    stroke prevention, statins may also be protective in acute

    ischaemic stroke (2,44). The main proposed mechanism of 

    action is due to an increase in NO bioavailability that regulates

    cerebral perfusion and improves endothelial function (45).

    Other possible mechanisms include antioxidant properties,

    atherosclerotic plaque stabilization, and anti-inflammatory 

    effects (45).

    Neuroprotection with Statin Therapy for Acute Recovery 

    Trial (NeuSTART) was a phase 1B dose-escalation study that

    showed that lovastatin administration was safe and feasible up

    to three-days after an acute ischaemic stroke (46). Now, a

    phase ll trial (NeuSTART II) is in progress to confirm lovas-

    tatin safety and efficacy in improving functional outcome after

    stroke (3).

    Although HMG-CoA reductase inhibitors clearly act

    beyond their sole lipid-lowering properties, the concept of 

    statins as powerful neuroprotectants or anti-inflammatory 

    drugs was recently called into question in another frequent

    neurological disease. Surprisingly, patients suffering from

    multiple sclerosis and treated with statins in combination withinterferon-b showed a trend towards increased disease activity 

    and lesion size compared with patients receiving placebo plus

    interferons (47).

    DP-b99

    DP-b99 is a novel therapeutic that chelates membrane-

    activated divalent metal ions such as calcium and zinc (48). As

    cell death following cerebral ischaemia is in part mediated by 

    these toxic metals, DP-b99 administration was shown to

    provide significant neuroprotection in animal models of 

    stroke (48). This promising compound underwent a phase II

    trial showing that patients receiving DP-b99 had improve-

    ments in a number of secondary end-points following acute

    ischaemic stroke (49). Now, the phase III Membrane Activator

    Chelator Stroke Intervention trial is underway investigating

    the capacity of DP-b99 to improve functional outcome follow-

    ing acute ischaemic stroke (3).

    Difficulties in translation intoclinical practice

    Examples of neuroprotective therapies frompreclinical to clinical

    As outlined earlier, a wide variety of neuroprotective drugs

    have been tested in preclinical animal studies with about 100

    of these being trialled in human studies (2). Even in cases

    where the drug showed neuroprotection in animal experi-

    ments, all have failed to achieve the primary end-point of neuroprotection in humans. Described later are two case

    examples, tirilazad and NXY-059, which are drugs that have

    shown a good level of protection preclinically, that have pro-

    ceeded into clinical trials with limited success.

    Tirilazad

    Tirilazad (U74006F) is a 21-aminosteroid that can inhibit

    lipid peroxidation by acting as a free radical scavenger (50). In

    transient focal ischaemia, Xue et al . (51) showed that tirilazad

    reduced cortical infarct size in rats, but this was not observed

    in permanent ischaemia. An overall analysis of all tirilazad

    preclinical studies showed that tirilazad reduced injury by 29% and had more significant effects following transient

    occlusion compared to permanent ischaemia (52). This sug-

    gests that tirilazad required revascularization so that it could

    reach the ischaemic penumbra to achieve neuroprotection.

    The efficacy of tirilazad was greater when given pre-ischaemia,

    but some efficacy was observed in delayed treatment out to

    six-hours post-ischaemia (52).

    The preclinical evidence earlier convincingly showed pro-

    tection, while the clinical evidence failed to reproduce these

    results. The Randomized Trial of Tirilazad mesylate in patients

    with Acute Stroke (RANTTAS) trial (53) was a multicentre,

    randomized, double-blinded, vehicle-controlled trial investi-

    gating tirilazad in acute stroke patients. Patients were not

    thrombolysed, and so were not stratified between transient

    and permanent ischaemia, although preclinical evidence sug-

    gested that this was important. Tirilazad was administered

    within-six hours (median time of 4·3 h) with subsequent

    administrations every six-hours for 11 additional doses. The

    study was prematurely terminated after inclusion of 556

    patients due to lack of any functional benefit at three-months.

    Further systematic analysis of all clinical trials investigating

    tirilazad showed that tirilazad actually increased disability and

    death in acute stroke patients (54). This is at odds with the

    Review   B. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke Organization412   Vol 7, July 2012, 407–418

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    7/12

    preclinical data and may be due to not using patients that had

    recanalization, and administering the treatments much later

    compared to the preclinical setting (median over all studies:

    five-hours clinical vs. 10 mins preclinical) (52). In order for

    neuroprotection to translate, methodologies between animal

    and human studies need to be more consistent and tightly 

    controlled.

    NXY-059

    NXY-059 is a nitrone that exhibits free radical scavenging

    properties and inhibits many stages of the ischaemic cascade

    (55). NXY-059 has shown significant neuroprotective effects

    in animal models of both transient and permanent occlusion

    of the middle cerebral artery (MCA) (56), with an overall

    reduction in infarct volume of 43% (18). The protective effects

    of NXY-059 in rodents were confirmed in nonhuman pri-

    mates (marmosets), which follow the STAIR criteria (57). The

    time course of effects of NXY-059 is similar to rtPA with

    efficacy within four-hours of occlusion (58). Therefore, due to

    their distinct mechanisms, NXY-059 could potentially be usedin concert with thrombolytic treatment for acute ischaemic

    stroke in humans.

    Two trials were performed to assess the neuroprotective

    activity of NXY-059 on human stroke: SAINT I and SAINT II.

    The SAINT I trial (59) was a phase III double-blinded, rand-

    omized, placebo-controlled trial that revealed a small but sig-

    nificant improvement in disability (modified Rankin scale)

    with NXY-059 three-months following stroke, but it did not

    improve neurological outcome (National Institute of Health

    Stroke Scale).   Post hoc   analysis showed that patients who

    underwent thrombolysis and treatment with NXY-059 had

    reduced incidence of haemorrhagic transformation. SAINT II(60) was a statistically more powerful study but disappoint-

    ingly failed to confirm the data reported in SAINT I. In SAINT

    II, there was no difference in disability at three-months, and

    the reduction in haemorrhagic transformation with rtPA by 

    NXY-059 could not be reproduced. There was a higher fre-

    quency of rtPA use in SAINT II (44% vs. 28%) (61) which may 

    have contributed to the difference in results. Only a small

    proportion of NXY-059 may cross the BBB, and the neuropro-

    tective action of NXY-059 may be mediated in the endothe-

    lium and neurovascular unit (58,62). This would also explain

    the reduced risk of haemorrhage in thrombolysed patients

    (62). However, there was no preclinical evidence that NXY-059

    exerted its effects by altering CBF in experimental models

    (63).

    There were a number of methodological weaknesses in the

    preclinical NXY-059 studies that potentially affected the

    human trials. Methodological quality was low (18) and only 

    9% of studies with NXY-059 measured CBF (64). The lack of 

    infarction observed in many NXY-059 studies may be due to

    not confirming MCA occlusion with CBF measurements,

    rather than a neuroprotective effect of NXY-059 (65). Many 

    studies were not blinded or randomized (18), and further

    analysis suggests that there may have been significant publica-

    tion bias (56). Interestingly, out of all the current drugs in

    phase II/III trials,only NXY-059 fulfilled the STAIR criteria for

    adequate translation into clinical trials (64), even with these

    methodological weaknesses.

    A potential reason for clinical failure of the SAINT trials is

    the difference in methodology compared to animal studies.

    The SAINT trials enrolled patients up to six-hours post-ischaemia onset, while a maximum of four-hours time

    window was chosen in animal studies (66). In animal studies,

    only occlusion of the MCA was performed, while the SAINT

    trials enrolled patients with different types of stroke, such as

    posterior or lacunar strokes. The SAINT trials perhaps should

    have selected stroke patients that more closely resembled what

    had previously worked in animal experiments, e.g. patients

    with MCA occlusion (66).

    Both tirilazad and NXY-059 are antioxidants that were sup-

    posedly neuroprotective by scavenging free radicals and pre-

    venting oxidative stress. Although oxidative stress has been

    suggested for many years to cause tissue damage and neuronal

    death, there is still no successful therapeutic application. Todate, all clinical attempts to scavenge reactive oxygen or nitro-

    gen species (ROS/RNS) by applying antioxidants have not

    resulted in clinical benefit and have even caused harm. Given

    that ROS/RNS are extremely short-lived molecules that form

    at multiple sites of the brain upon ischaemia, this is in fact not

    surprising. However, the characterization of the relevant enzy-

    matic sources of oxidative stress such as nicotinamide adenine

    dinucleotide phosphate (NADPH) oxidases (see later) may 

    allow therapeutic targeting of oxidative stress by preventing

    the formation of ROS initially instead of scavenging ROS after

    they have been formed (67).

    Clinical neuroprotection – why it has failed

    The two case examples of preclinical to clinical neuroprotec-

    tion assessment outlined the problems associated with trans-

    lation. The reasons for translational failure are numerous

    (Table 1), ranging from flaws in clinical trial design, delayed

    treatment time window, small sample sizes, different

    outcome measures, insufficient dosing, and failure to achieve

    adequate plasma levels of study medications (7,68,69). Also,

    the heterogeneous nature of human stroke is at odds with

    the homogeneous animal models currently used (70).

    Animal models of stroke mimic at best less than 25% of all

    strokes, with the rat model of MCA occlusion probably 

    reflecting the Total Anterior Circulation Stroke Syndrome in

    humans (71). Therefore, there is a need to create new animal

    models that better reflect the heterogeneity of ischaemic

    stroke in humans. In addition, preclinical studies are usually 

    performed on young healthy animals, whereas patients are

    mostly elderly with possible comorbidities. Ageing is associ-

    ated with significant structural and functional changes in the

    brain, which affects outcome and the ability to recover after

    an ischaemic event (72).

    ReviewB. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke OrganizationVol 7, July 2012, 407–418   413

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    8/12

    Experimental studies require a more rigorous design with

    higher quality standard levels to avoid bias, and a careful

    control of physiological variables to distinguish genuine drug

    mode of action from other non-specific effects (4,56). In clini-

    cal studies, the treatment time window should be restricted to

    a period similar to that shown to be effective experimentally.

    Unfortunately, most neuroprotective agents target early eventsin the ischaemic cascade, which require rapid administration

    following stroke onset, which is challenging in acute ischaemic

    stroke patients. Therefore, agents that have long therapeutic

    time windows are optimal.

    Combining neuroprotection with thrombolysis

    A crucial protective strategy following stroke is the early reca-

    nalization of the blood vessel to restore flow back into the

    ischaemic region of the brain. This is usually achieved by 

    thrombolysis, and rtPA is currently the only United States

    Food and Drug Administration-approved thrombolytic

    therapy. Unfortunately, less than 15% of patients actually receive this therapy (73) because of the short time window of 

    treatment (4·5 h) and the risk of haemorrhagic transforma-

    tion. Therefore, treatment strategies that can improve post-

    ischaemic CBF, reduce cerebral injury, restrict adverse effects,

    and extend the therapeutic time window may prove useful to

    improve rtPA therapy. Thrombolysis may improve delivery of 

    the neuroprotectant to the penumbral region increasing the

    chances of a beneficial effect. Many neuroprotective agents

    exhibit synergistic effects with rtPA preclinically including

    matrix metalloprotease inhibitors (74), free radical scaven-

    gers (75,76), NMDA receptor antagonists (77),  a -amino-3-

    hydroxy-5-methyl-4-isoxazolepropionic acid receptor antago-nists (78), antioxidant agents (79), anti-inflammatory agents

    (80), and antiplatelet agents (80–83).

    The concept of thrombolysis combined with neuroprotec-

    tive therapy has not been extensively explored in human

    clinical trials. As outlined earlier, SAINT I showed that NXY-

    059 administration in stroke patients who had received rtPA

    reduced haemorrhagic transformation compared to NXY-059

    alone (59). However, there was no additive benefit observed

    with lubeluzole (sodium channel blocker) and rtPA, even

    though combination therapy did not increase adverse effects

    (84). Other combination studies with rtPA include clom-

    ethiazole (g -aminobutyric acid agonist that showed no

    benefit) (85) and UK-279276 (neutrophil inhibitory factor

    that showed no benefit) (86). Much like neuroprotection

    itself, the promise of combination therapy of thrombolysis

    and neuroprotection is tempting, but this needs further

    investigation in clinical trials before the viability of this strat-

    egy can be confirmed.

    One major drawback of testing combination therapies is

    that any observed effect or side effect often cannot be

    unequivocally assigned to one or the other partner. Moreover,

    it is difficult to foresee whether the two (or more) compounds

    will act synergistically (multiplicative effect), independently 

    from each other (additive effect) or even in an antagonistic

    manner (neutralizing effect). Even though rtPA is a thrombo-

    lytic, it has many effects independent of thrombolysis, which

    may be due to the L-arginine present in the rtPA formulation

    (87). Every stroke patient receiving rtPA also receives

    L-arginine, which is a substrate for NO synthesis, and may 

    affect outcome following ischaemia (87). Therefore, it will bedifficult to elicit individual effects of compounds, even if rtPA

    is part of the combination therapy.

    Promising neuroprotectants

    There are still many potential neuroprotective compounds

    that are currently being investigated for the treatment of acute

    ischaemic stroke. Discussed later are two groups of neuropro-

    tective agents that have shown promising effects.

    Haematopoietic growth factors

    Haematopoietic growth factors are a group of regulatory molecules that are responsible for the mobilization, prolif-

    eration, maturation, and survival of bone marrow-derived

    cells (88). Receptors of several haematopoietic growth factors

    are expressed on neurons. Moreover, functions of growth

    factors paralleling those in the haematopoietic system were

    identified in the brain (89,90). Among the haematopoietic

    growth factors, the granulocyte-colony-stimulating factor

    (G-CSF) and erythropoietin (EPO) were particularly well

    investigated for their effects in cerebral ischaemia. Both

    G-CSF and EPO reduced glutamate-induced neuronal cell

    death  in vitro   and prevented apoptosis of neurons in vivo  by 

    activating several anti-apoptotic pathways (89,91). G-CSFand EPO also demonstrated anti-inflammatory actions after

    ischaemia (92,93). Besides having neuroprotective properties,

    G-CSF and EPO facilitate functional recovery poststroke by 

    enhancing neurogenesis and angiogenesis (89,94,95). Meta-

    analyses of EPO and G-CSF in animal experimental stroke

    showed that both factors reduced infarcts and improved

    functional outcomes (19,96–98). However, when the impact

    of common sources of bias, e.g. unblinded outcome assess-

    ment, was considered for analysis, the efficacy of EPO was

    lowered, suggesting that the benefit of EPO might be over-

    estimated (97).

    A small clinical trial showed that EPO is safe and might be

    beneficial in acute ischaemic stroke (99). However, these

    promising results were not confirmed by a larger phase II/III

    German multicentre stroke trial which investigated stroke

    patients that had received either EPO or placebo within six-

    hours of symptom onset (100). The primary end-point,

    change in Barthel Index on day 90, and all secondary out-

    comes failed to show any benefit of EPO. Moreover, an

    increased mortality rate was observed after EPO treatment.

    Potential reasons for the differences between preclinical

    studies and the clinical trial could be due to an overestimated

    efficacy of EPO in animal studies through neglected quality 

    Review   B. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke Organization414   Vol 7, July 2012, 407–418

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    9/12

    characteristics and unexpected side effects of EPO in patients,

    i.e. an intrinsic stroke-inducing capacity of EPO (101). A

    recent phase IIa clinical trial demonstrated that G-CSF is well

    tolerated even at high doses in stroke patients (102). Explora-

    tory analysis revealed a dose-dependent beneficial effect of 

    G-CSF in patients with large infarcts (102). A phase II trial on

    AX200 (filgrastim, a G-CSF analogue) for ischaemic strokepatients (AXIS-2) is currently ongoing, and results are

    expected soon (103).

    NADPH oxidase inhibitors

    The concept that free radicals including ROS are involved in

    the pathophysiology of acute ischaemic stroke and account for

    secondary infarct growth is over 30 years old (104) but still

    valid and based on solid experimental data (105,106). If ROS

    are the trigger that causes neurotoxicity in the ischaemic brain,

    the question regarding the main sources of ROS arises. Some

    of the most attractive candidates are NADPH oxidases, the

    only known enzyme family solely dedicated to ROS produc-

    tion. Many other enzymes are able to form ROS, e.g. xanthineoxidase, uncoupled NO synthase, and cytochrome P450

    enzymes. However, in all these cases, ROS formation requires

    an initial oxidation step to occur and none of them forms ROS

    natively (107). The structure and function of the NADPH

    oxidases were initially characterized in neutrophils, where the

    enzyme plays a pivotal role in immunological host defence.

    Recently, it has been discovered that the catalytic sub-unit of 

    the phagocytic NADPH-oxidase is only one member of a

    family of four homologous proteins known as NOX1-4 (for

    NADPH-oxidase).

    By strictly adhering to current quality standards in experi-

    mental stroke research (5), we could demonstrate thatNOX4-derived oxidative stress is a crucial player in the

    pathophysiology of cerebral ischaemia (67,108). NOX4 was

    massively induced in neurons and brain vessels in human

    stroke patients and mice subjected to transient MCAO. Mice

    deficient in NOX4, but not those deficient for NOX1 or

    NOX2, were largely protected from oxidative stress and neu-

    ronal apoptosis, after both transient and permanent cerebral

    ischaemia. This was independent of gender and age as elderly 

    mice were equally protected. Interestingly from a transla-

    tional perspective, application of the only validated pharma-

    cological NADPH oxidase inhibitor, VAS2870, several hours

    after ischaemia had the same beneficial effect as deleting

    NOX4 (67). The extent of neuroprotection was exceptional

    (~70% reduction of stroke volumes), resulting in signifi-

    cantly improved long-term neurological function and

    reduced mortality.

    Targeting the right enzymatic source of ROS rather than

    applying non-specific antioxidants after radicals have already 

    been generated may represent an attractive treatment option

    in acute ischaemic brain damage and other disease states

    related to oxidative stress (109). Novel and sub-type-specific

    NADPH oxidase inhibitors on a small molecule base with

    improved pharmacological properties are currently under

    development and bear a realistic chance to enter clinical trials

    within the next few years.

    Conclusions – the future of neuroprotectionin ischaemic stroke

    The translational disappointments have created a great deal of 

    pessimism regarding the future of neuroprotection trials in

    humans and have cast doubt on the neuroprotection hypoth-

    esis. Some have even suggested that the initially favourable

    results of several of the trials, e.g. SAINT I, were likely chance

    findings and that the idea of neuroprotection as a form of 

    treatment for acute stroke should be abandoned. However,

    looking into the past, despite the initial scepticism about

    stroke care, the landmark National Institute of Neurological

    Disorders and Stroke alteplase trial (110) not only revolution-

    ized stroke treatment but also reinvigorated enthusiasm in

    stroke care and research. While recent progress in stroke trials

    has not directly yielded new clinical drugs, they have providedimportant mechanistic insights into the complex pathophysi-

    ology of ischaemic stroke which will pave the way for upcom-

    ing studies. Future neuroprotection experiments must be

    methodologically sound and learn from previous failed

    attempts, while clinical trials must take into account the

    success achieved in preclinical studies.

    With this respect, the implementation of essential quality 

    standards in experimental stroke research is without doubt a

    meaningful measure. However, one has to keep the balance

    between high quality on the one hand, and practicability in an

    academic laboratory environment on the other hand. The

    postulation to validate rodent findings in primates, forexample, is easily spoken but nearly impossible to realize given

    that only a few primate facilities are available across Europe

    (111). Ethical aspects have to be carefully considered as well

    when using higher animal species in injuring and disabling

    disease models. In order to provide proof-of-principle evi-

    dence, it is in most cases neither necessary nor feasible to plan

    and perform basic stroke studies like large controlled rand-

    omized clinical trials. This is also frequently prevented by the

    increasing numbers of budget cuts the scientific community 

    currently has to face, limited animal housing and breeding

    space, lack of qualified staff, rough scientific competition, and

    the strict domestic and international regulations for animal

    care and use. Finally, more than 10 years after the first STAIR 

    recommendations were published, the ultimate proof that

    plain standardization of procedures in fact increases the rate

    of successful translation from bench to bedside in stroke

    research is still missing. Some critics even raise the provocative

    question of whether excessive methodological uniformity 

    counteracts innovation and only prevents promising drug

    candidates from entering into clinical trials and renowned

    scientific journals. Positive experiences from other neurologi-

    cal diseases like multiple sclerosis should teach the stroke com-

    munity that even imperfect animal models can serve as a basis

    ReviewB. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke OrganizationVol 7, July 2012, 407–418   415

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    10/12

    for hypothesis-driven research that ultimately facilitates the

    development of new drugs.

    Acknowledgements

    The authors were supported by Fondation Leducq (BAS and

    AMB), Medical Research Council UK (AMB), the National

    Institute for Health Research Biomedical Research Centre

    (JSB, FA and AMB), the Deutsche Forschungsgemeinschaft

    (SFB 688, TPA13 and KL 2323/4-1, CK), the Wilhelm-Sander

    Stiftung (2009·017·1, CK), the European Union (Seventh

    Framework Programme FP7, HEALTH-F2-2009-241778,

    CK), and the Bundesministerium für Bildung und Forschung

    (BMBF, 01GN0980, JM).

    References

    1 Durukan A, Tatlisumak T. Acute ischemic stroke: overview of major

    experimental rodent models, pathophysiology, and therapy of focal

    cerebral ischemia. Pharmacol Biochem Behav  2007; 87:179–97.

    2 O’Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp

    BH,Howells DW. 1,026 experimental treatments in acute stroke. Ann

     Neurol  2006; 59:467–77.

    3 Strokecenter.org, Stroke Trials Registry, Dallas: UT Southwestern

    Medical Center. 1997 Availableat: http://www.strokecenter.org/trials/

    (accessed April 2011).

    4 GinsbergMD. Neuroprotection for ischemic stroke: past, presentand

    future. Neuropharmacology  2008; 55:363–89.

    5 Fisher M, Feuerstein G, Howells DW  et al . Update of the stroke

    therapy academic industry roundtable preclinical recommendations.

    Stroke 2009; 40:2244–50.

    6 STAIR.Recommendations for standards regarding preclinical neuro-

    protective and restorative drug development. Stroke 1999; 30:2752–8.

    7 Cheng YD, Al-Khoury L, Zivin JA. Neuroprotection for ischemic

    stroke: two decades of success and failure. NeuroRx  2004; 1:36–45.

    8 Endres M, Engelhardt B, Koistinaho J et al . Improving outcome after

    stroke: overcoming the translational roadblock. CerebrovascDis 2008;

    25:268–78.

    9 Hoyte L, Kaur J, Buchan AM. Lost in translation: taking neuropro-

    tection from animal models to clinical trials.   Exp Neurol   2004;

    188:200–4.

    10 Buchan A, Pulsinelli WA. Hypothermia but not the N-methyl-D-

    aspartate antagonist, MK-801, attenuates neuronal damage in gerbils

    subjected to transient global ischemia. J Neurosci 1990; 10:311–6.

    11 Buchan AM, Slivka A, Xue D. The effect of the NMDA receptor

    antagonist MK-801 on cerebral blood flow and infarct volume in

    experimental focal stroke. Brain Res 1992; 574:171–7.

    12 Sutherland BA, Papadakis M, Chen R-L, Buchan AM. Cerebral blood

    flow alteration in neuroprotection following cerebral ischemia.   J 

    Physiol  2011; 589:4105–14.

    13 Nikic I, Merkler D, Sorbara C et al . A reversible form of axon damage

    in experimental autoimmune encephalomyelitis and multiple sclero-

    sis.  Nat Med  2011; 17:495–9.

    14 Sena ES, van der Worp HB, Bath PM, Howells DW, Macleod MR.

    Publication bias in reports of animal stroke studies leads to major

    overstatement of efficacy. PLoS Biol  2010; 8:e1000344.

    15 Fisher M. New approaches to neuroprotective drug development.

    Stroke 2011; 42(Suppl. 1):S24–7.

    16 Minnerup J, Schabitz WR. Multifunctional actions of approved and

    candidate stroke drugs. Neurother  2009; 6:43–52.

    17 Dirnagl U, Macleod MR. Stroke research at a road block: the streets

    from adversity should be paved with meta-analysis and good labo-

    ratory practice. Br J Pharmacol  2009; 157:1154–6.

    18 Macleod MR, van der Worp HB, Sena ES, Howells DW, Dirnagl U,

    Donnan GA. Evidence for the efficacy of NXY-059 in experimental

    focal cerebral ischaemia is confounded by study quality.  Stroke 2008;

    39:2824–9.

    19 Minnerup J, Heidrich J, Wellmann J, Rogalewski A, Schneider A,

    Schabitz WR. Meta-analysis of the efficacy of granulocyte-colony 

    stimulating factor in animal models of focal cerebral ischemia. Stroke

    2008; 39:1855–61.

    20 Thecochranelibrary.com, The Cochrane Library, Chichester, UK:

    John wiley & Sons Ltd. 1996 Available at: http://www.

    thecochranelibrary.com/ (accessed April 2011).

    21 van der Worp HB, Sena ES, Donnan GA, Howells DW,Macleod MR.

    Hypothermia in animal models of acute ischaemic stroke: a system-

    atic review and meta-analysis. Brain 2007; 130(Pt 12):3063–74.

    22  Yenari M, Kitagawa K, Lyden P, Perez-Pinzon M. Metabolic down-

    regulation: a key to successful neuroprotection?   Stroke   2008;

    39:2910–7.

    23 Berger C, Schabitz WR, Georgiadis D, Steiner T, Aschoff A,

    Schwab S. Effects of hypothermia on excitatory amino acids and

    metabolism in stroke patients: a microdialysis study.  Stroke   2002;

    33:519–24.

    24 Globus MY, Alonso O, Dietrich WD, Busto R, Ginsberg MD.

    Glutamatereleaseand free radical production following brain injury:

    effects of posttraumatic hypothermia. J Neurochem  1995;  65:1704–

    11.

    25 Krieger DW, De Georgia MA, Abou-Chebl A et al . Cooling for acute

    ischemic brain damage (cool aid): an open pilot study of induced

    hypothermia in acute ischemic stroke. Stroke 2001; 32:1847–54.

    26 De Georgia MA, Krieger DW, Abou-Chebl A et al . Cooling for Acute

    Ischemic Brain Damage (COOL AID): a feasibility trial of endovas-

    cular cooling. Neurology  2004; 63:312–7.

    27 Hemmen TM, Raman R, Guluma KZ et al . Intravenous thrombolysis

    plus hypothermia for acute treatment of ischemic stroke (ICTuS-L):

    final results. Stroke 2010; 41:2265–70.

    28 Hemmen TM, Lyden PD. Induced hypothermia for acute stroke.

    Stroke 2007; 38(Suppl. 2):794–9.

    29 Lyden PD, Krieger D, Yenari M, Dietrich WD. Therapeutic hypother-

    mia for acute stroke. Int J Stroke  2006; 1:9–19.

    30 Schwab S, Georgiadis D, Berrouschot J, Schellinger PD, Graffagnino

    C, Mayer SA. Feasibility and safety of moderate hypothermia after

    massive hemispheric infarction. Stroke 2001; 32:2033–5.

    31 He P, Curry FE. Albumin modulation of capillary permeability:

    role of endothelial cell [Ca2+]i. Am J Physiol  1993; 265(1 Pt 2):H74–

    82.

    32 Belayev L, Liu Y, Zhao W, Busto R, Ginsberg MD. Human albumin

    therapy of acute ischemic stroke: marked neuroprotective efficacy at

    moderate doses and with a broad therapeutic window. Stroke  2001;

    32:553–60.

    33 Ginsberg MD, Hill MD, Palesch YY, Ryckborst KJ, Tamariz D. The

    ALIAS Pilot Trial: a dose-escalation and safety study of albumin

    therapy for acute ischemic stroke – I: Physiological responses and

    safety results. Stroke 2006; 37:2100–6.

    34 Palesch YY, Hill MD, Ryckborst KJ, Tamariz D, Ginsberg MD. The

    ALIAS Pilot Trial: a dose-escalation and safety study of albumin

    therapy for acute ischemic stroke – II: neurologic outcome and effi-

    cacy analysis. Stroke 2006; 37:2107–14.

    35 Ginsberg MD, Palesch YY, Hill MD. The ALIAS (ALbumin In Acute

    Stroke) Phase III randomized multicentre clinical trial: design and

    progress report. Biochem Soc Trans  2006; 34(Pt 6):1323–6.

    36 O’Collins VE, Macleod MR, Cox SF et al . Preclinical drug evaluation

    for combination therapy in acute stroke using systematic review,

    meta-analysis, and subsequent experimental testing.   J Cereb Blood 

    Flow Metab 2011; 31:962–75.

    37 Ovbiagele B, Kidwell CS, Starkman S, Saver JL. Neuroprotective

    agents for the treatment of acute ischemic stroke. Curr Neurol Neu-

    rosci Rep 2003; 3:9–20.

    Review   B. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke Organization416   Vol 7, July 2012, 407–418

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    11/12

    38 Meloni BP, Zhu H, Knuckey NW. Is magnesium neuroprotective

    following global and focal cerebral ischaemia? A review of published

    studies. Magnes Res 2006; 19:123–37.

    39 Saver JL, Kidwell C, Eckstein M, Starkman S. Prehospital neuropro-

    tective therapy for acute stroke: results of the Field Administration of 

    Stroke Therapy-Magnesium (FAST-MAG) pilot trial.  Stroke   2004;

    35:e106–8.

    40 Lampl Y, Boaz M, Gilad R  et al . Minocycline treatment in acute

    stroke: an open-label, evaluator-blinded study.   Neurology   2007;

    69:1404–10.

    41 Prass K, Meisel C, Hoflich C et al . Stroke-induced immunodeficiency 

    promotes spontaneous bacterial infections and is mediated by sym-

    pathetic activation reversal by poststroke T helper cell type 1-like

    immunostimulation. J Exp Med  2003; 198:725–36.

    42 Fagan SC, Waller JL, Nichols FT  et al . Minocycline to improve neu-

    rologic outcome in stroke (MINOS): a dose-finding study.  Stroke

    2010; 41:2283–7.

    43 Clinicaltrials.gov. Neuroprotection With Minocycline Therapy for

    Acute Stroke Recovery Trial (NeuMAST), Bethesda: US National

    Institutes of Health. 2009 Available at: http://clinicaltrials.gov/ct2/

    show/study/NCT00930020 (accessed April 2011).

    44 Cimino M, Balduini W, Carloni S   et al . Neuroprotective effect

    of simvastatin in stroke: a comparison between adult and neonatal

    rat models of cerebral ischemia. Neurotoxicology  2005; 26:929–33.

    45 Cimino M,Gelosa P,Gianella A,Nobili E, Tremoli E,Sironi L.Statins:

    multiple mechanisms of action in the ischemic brain. Neuroscientist 

    2007; 13:208–13.

    46 Elkind MS, Sacco RL, Macarthur RB et al . High-dose lovastatin for

    acute ischemic stroke: results of the phase I dose escalation neuro-

    protection with statin therapy for acute recovery trial (NeuSTART).

    Cerebrovasc Dis 2009; 28:266–75.

    47 Birnbaum G, Cree B, Altafullah I, Zinser M, Reder AT. Combining

    beta interferon and atorvastatin may increase disease activity in mul-

    tiple sclerosis. Neurology  2008; 71:1390–5.

    48 Angel I, Bar A, Horovitz T et al . Metal ion chelation in neurodegen-

    erative disorders. Drug Dev Res  2002; 56:300–9.

    49 Diener HC, Schneider D, Lampl Y, Bornstein NM, Kozak A,

    Rosenberg G. DP-b99, a membrane-activated metal ion chelator,

    as neuroprotective therapy in ischemic stroke.   Stroke   2008;

    39:1774–8.

    50 Braughler JM, Pregenzer JF, Chase RL, Duncan LA, Jacobsen EJ,

    McCall JM. Novel 21-amino steroids as potent inhibitors of iron-

    dependent lipid peroxidation. J Biol Chem  1987; 262:10438–40.

    51 Xue D, Slivka A, Buchan AM. Tirilazad reduces cortical infarction

    after transient but not permanent focal cerebral ischemia in rats.

    Stroke 1992; 23:894–9.

    52 Sena E, Wheble P, Sandercock P, Macleod M. Systematic review and

    meta-analysis of the efficacy of tirilazad in experimental stroke.

    Stroke 2007; 38:388–94.

    53 RANTTAS. A randomized trial of tirilazad mesylate in patients with

    acute stroke (RANTTAS). The RANTTAS Investigators.  Stroke 1996;

    27:1453–8.

    54 TISC. Tirilazad mesylate in acute ischemic stroke: A systematicreview. Tirilazad International Steering Committee.   Stroke   2000;

    31:2257–65.

    55 Green AR, Ashwood T, Odergren T, Jackson DM. Nitrones as neu-

    roprotective agents in cerebral ischemia, with particular reference to

    NXY-059.  Pharmacol Ther  2003; 100:195–214.

    56 Bath PM, Gray LJ, Bath AJ, Buchan A,Miyata T, Green AR.Effects of 

    NXY-059 in experimental stroke: an individual animal meta-analysis.

    Br J Pharmacol  2009; 157:1157–71.

    57 Marshall JW,Cummings RM, Bowes LJ, Ridley RM, Green AR. Func-

    tional and histological evidence for the protective effect of NXY-059

    in a primate model of stroke when given 4 hours after occlusion.

    Stroke 2003; 34:2228–33.

    58 Papadakis M, Nagel S, Buchan AM. Development and efficacy of 

    NXY-059 for the treatment of acute ischemic stroke.  Future Neurol 

    2008; 3:229–40.

    59 Lees KR, Zivin JA, Ashwood T  et al . NXY-059 for acute ischemic

    stroke. N Engl J Med  2006; 354:588–600.

    60 Shuaib A, Lees KR, Lyden P et al . NXY-059 for the treatment of acute

    ischemic stroke. N Engl J Med  2007; 357:562–71.

    61 Diener HC, Lees KR, Lyden P  et al . NXY-059 for the treatment of 

    acute stroke: pooled analysis of the SAINT I and II Trials. Stroke 2008;

    39:1751–8.

    62 Papadakis M, Buchan AM. NXY-059: brain or vessel protection.

    Stroke 2006; 37:2189–90.

    63 Zhao Z, Cheng M, Maples KR, Ma JY, Buchan AM. NXY-059, a novel

    free radical trapping compound, reduces cortical infarction after

    permanent focal cerebral ischemia in the rat. Brain Res 2001; 909:46–

    50.

    64 Philip M, Benatar M, Fisher M, Savitz SI. Methodological quality of 

    animal studies of neuroprotective agents currently in phase II/III

    acute ischemic stroke trials. Stroke 2009; 40:577–81.

    65 Savitz SI. Cosmic implications of NXY-059. Stroke  2009;  40(Suppl.

    3):S115–8.

    66 Savitz SI. A critical appraisal of the NXY-059 neuroprotection studies

    for acute stroke: a need for more rigorous testing of neuroprotective

    agents in animal models of stroke. Exp Neurol  2007; 205:20–5.

    67 Kleinschnitz C, Grund H, Wingler K  et al . Post-stroke inhibition of 

    induced NADPH oxidase type 4 prevents oxidative stress and neuro-

    degeneration.  PLoS Biol  2010; 8:pii:e1000479.

    68 Tymianski M. Can molecular and cellular neuroprotection be trans-

    lated into therapies for patients?: yes, but not the way we tried it

    before. Stroke 2010; 41(Suppl. 10):S87–90.

    69 Wahlgren NG, Ahmed N. Neuroprotection in cerebral ischaemia:

    facts and fancies–the need for new approaches. Cerebrovasc Dis 2004;

    17(Suppl. 1):153–66.

    70 Lyden P, WahlgrenNG. Mechanisms of action of neuroprotectants in

    stroke. J Stroke Cerebrovasc Dis  2000; 9(6 Pt 2):9–14.

    71 Small DL, Buchan AM. Animal models. Br Med Bull  2000;  56:307–

    17.

    72 Chen RL, Balami JS, Esiri MM, Chen LK, Buchan AM. Ischemic

    stroke in the elderly: an overview of evidence. Nat Rev Neurol  2010;

    6:256–65.

    73 Grotta JC, Burgin WS, El-Mitwalli A  et al . Intravenous tissue-type

    plasminogen activator therapy for ischemic stroke: Houston experi-

    ence 1996 to 2000. Arch Neurol  2001; 58:2009–13.

    74 Sumii T, Involvement LEH. of matrix metalloproteinase in

    thrombolysis-associated hemorrhagic transformation after embolic

    focal ischemia in rats.  Stroke 2002; 33:831–6.

    75 Asahi M, Asahi K, Wang X, Lo EH. Reduction of tissue plasminogen

    activator-induced hemorrhage and brain injury by free radical spin

    trapping after embolic focal cerebral ischemia in rats. J Cereb Blood 

    Flow Metab 2000; 20:452–7.

    76 Lapchak PA, Araujo DM, Song D, Wei J, Zivin JA. Neuroprotective

    effects of the spin trap agent disodium-[(tert-butylimino)

    methyl]benzene-1,3-disulfonate N-oxide (generic NXY-059) in arabbit small clot embolic stroke model: combination studies with the

    thrombolytic tissue plasminogen activator.  Stroke 2002; 33:1411–5.

    77 Lekieffre D, Benavides J, Scatton B, Nowicki JP. Neuroprotection

    afforded by a combination of eliprodil and a thrombolytic agent,

    rt-PA, in a rat thromboembolic stroke model.   Brain Res   1997;

    776:88–95.

    78 Suzuki M, Sasamata M, Miyata K. Neuroprotective effects of YM872

    coadministered with t-PA in a rat embolic stroke model.  Brain Res

    2003; 959:169–72.

    79 Lapchak PA, Zivin JA. Ebselen, a seleno-organic antioxidant, is neu-

    roprotective after embolic strokes in rabbits: synergism with low-

    dose tissue plasminogen activator.  Stroke 2003; 34:2013–8.

    ReviewB. A. Sutherland  et al .

    © 2012 The Authors.

    International Journal of Stroke © 2012 World Stroke OrganizationVol 7, July 2012, 407–418   417

  • 8/16/2019 3. Neuroprotection for Ischaemic Stroke Translation From the Bench to the Bedside

    12/12

    80 Zhang RL, Zhang ZG, Chopp M. Increased therapeutic efficacy with

    rt-PA and anti-CD18 antibody treatment of stroke in the rat.  Neu-

    rology  1999; 52:273–9.

    81 Ding G, Jiang Q, Zhang L et al . Analysis of combined treatment of 

    embolic stroke in rat with r-tPA and a GPIIb/IIIa inhibitor. J Cereb

    Blood Flow Metab 2005; 25:87–97.

    82 Shuaib A, Yang Y, Nakada MT, Li Q, Yang T. Glycoprotein IIb/IIIa

    antagonist, murine 7E3 F(ab’) 2, and tissue plasminogen activator in

    focal ischemia: evaluation of efficacy and risk of hemorrhage with

    combination therapy. J Cereb Blood Flow Metab 2002; 22:215–22.

    83 Stoll G, Kleinschnitz C, Nieswandt B. Molecular mechanisms of 

    thrombus formation in ischemic stroke: novel insights and targets for

    treatment. Blood  2008; 112:3555–62.

    84 Grotta J. Combination TherapyStroke Trial: recombinant tissue-type

    plasminogen activator with/without lubeluzole.   Cerebrovasc Dis

    2001; 12:258–63.

    85 Lyden P, Jacoby M, Schim J et al . The Clomethiazole Acute Stroke

    Study in tissue-type plasminogen activator-treated stroke (CLASS-

    T): final results. Neurology  2001; 57:1199–205.

    86 Krams M, Lees KR, Hacke W, Grieve AP, Orgogozo JM, Ford GA.

    Acute Stroke Therapy by Inhibition of Neutrophils (ASTIN): an

    adaptive dose-response study of UK-279,276 in acute ischemic

    stroke. Stroke 2003; 34:2543–8.

    87 Harston GW, Sutherland BA, Kennedy J, Buchan AM. The contribu-

    tion of L-arginine to the neurotoxicity of recombinant tissue plas-

    minogen activator following cerebral ischemia: a review of rtPA

    neurotoxicity. J Cereb Blood Flow Metab 2010; 30:1804–16.

    88 Sachs L. The molecular control of hematopoiesis: from clonal devel-

    opment in culture to therapy in the clinic.  Int J Cell Cloning  1992;

    10:196–204.

    89 Schneider A, Kruger C, Steigleder T  et al . The hematopoietic factor

    G-CSF is a neuronal ligand that counteracts programmed cell death

    and drives neurogenesis. J Clin Invest  2005; 115:2083–98.

    90 Siren AL, Knerlich F, Poser W, Gleiter CH, Bruck W, Ehrenreich H.

    Erythropoietin and erythropoietin receptor in human ischemic/

    hypoxic brain. Acta Neuropathol  2001; 101:271–6.

    91 Ruscher K, Freyer D, Karsch M  et al . Erythropoietin is a paracrine

    mediator of ischemic tolerance in thebrain:evidence from an in vitro

    model. J Neurosci 2002; 22:10291–301.

    92 Solaroglu I, Cahill J, Tsubokawa T, Beskonakli E, Zhang JH.

    Granulocyte colony-stimulating factor protects the brain against

    experimental stroke via inhibition of apoptosis and inflammation.

     Neurol Res  2009; 31:167–72.

    93 Villa P, Bigini P, Mennini T  et al . Erythropoietin selectively attenu-

    ates cytokine production and inflammation in cerebral ischemia by 

    targeting neuronal apoptosis. J Exp Med  2003; 198:971–5.

    94 Lee ST, Chu K, Jung KH et al . Granulocyte colony-stimulating factor

    enhances angiogenesis after focal cerebral ischemia. Brain Res  2005;

    1058:120–8.

    95 Wang L, Zhang Z, Wang Y, Zhang R, Chopp M. Treatment of stroke

    with erythropoietin enhances neurogenesis and angiogenesis and

    improves neurological function in rats. Stroke 2004; 35:1732–7.

    96 England TJ, Gibson CL, Bath PM. Granulocyte-colony stimulatingfactor in experimental stroke and its effects on infarct size and func-

    tional outcome: a systematic review. Brain Res Rev  2009; 62:71–82.

    97 Jerndal M, Forsberg K, Sena ES et al . A systematic review and meta-

    analysis of erythropoietin in experimental stroke. J Cereb Blood Flow 

     Metab 2010; 30:961–8.

    98 Minnerup J,Heidrich J,Rogalewski A,Schabitz WR,Wellmann J.The

    efficacy of erythropoietin and its analogues in animal stroke models:

    a meta-analysis. Stroke 2009; 40:3113–20.

    99 Ehrenreich H, Hasselblatt M, Dembowski C  et al . Erythropoietin

    therapy for acute stroke is both safe and beneficial.  Mol Med  2002;

    8:495–505.

    100 Ehrenreich H, Weissenborn K, Prange H et al . Recombinant human

    erythropoietin in the treatment of acute ischemic stroke. Stroke 2009;

    40:e647–56.

    101 Minnerup J, Wersching H, Schabitz WR. EPO for stroke therapy – Is

    there a futurefor further clinical development? ExpTranslStroke Med 

    2010; 2:10.

    102 Schabitz WR, Laage R, Vogt G  et al . AXIS: a trial of intravenous

    granulocyte colony-stimulating factor in acute ischemic stroke.

    Stroke 2010; 41:2545–51.

    103 Clinicaltrials.gov. AXIS 2: AX200 for the Treatment of Ischemic

    Stroke (AXIS-2), Bethesda: US National Institute for Health.

    2009 Available at: http://clinicaltrials.gov/ct2/show/NCT00927836

    (accessed April 2011).

    104 Flamm ES, Demopoulos HB, Seligman ML, Poser RG, Ransohoff J.

    Free radicals in cerebral ischemia.  Stroke 1978; 9:445–7.

    105 Crack PJ, Taylor JM. Reactive oxygen species and the modulation of 

    stroke. Free Radic Biol Med  2005; 38:1433–44.

    106 Dhalla NS, Temsah RM, Netticadan T. Role of oxidative stress in

    cardiovascular diseases. J Hypertens  2000; 18:655–73.

    107 Miller AA, Drummond GR, Sobey CG. Novel isoforms of NADPH-

    oxidase in cerebral vascular control.  Pharmacol Ther  2006; 111:928–

    48.

    108 Sedwick C. NOX4: a guilty party in stroke damage. PLoS Biol  2010;

    8:pii:e1000478.

    109 Sancho P, Fabregat I. The NADPH oxidase inhibitor VAS2870

    impairs cell growth and enhances TGF-beta-induced apoptosis of 

    liver tumor cells. Biochem Pharmacol  2011; 81:917–24.

    110 NINDS. Tissue plasminogen activator for acute ischemic stroke. The

    National Institute of Neurological Disorders and Stroke rt-PA Stroke

    Study Group. N Engl J Med  1995; 333:1581–7.

    111 Schiermeier Q. Primate work faces German veto.   Nature   2007;

    446:955.

    Supporting Information

    Additional Supporting Information may be found in the

    online version of this article:

    Table S1.   Neuroprotective strategies that have completed

    trials for acute ischaemic stroke.1 All neuroprotective strate-

    gies have thus far failed to show an improvement in clinical

    benefit following acute ischaemic stroke.

    Please note: Wiley-Blackwell are not responsible for the

    content or functionality of any supporting materials supplied

    by the authors. Any queries (other than missing material)

    should be directed to the corresponding author for the article.

    Review   B. A. Sutherland  et al .

    © 2012 The Authors.418 Vol 7 July 2012 407–418