The major efforts to selectively deliver drugs to the brain in the past decade have relied on smart molecular techniques to penetrate the blood-brain barrier, whereas intraarterial drug delivery has drawn relatively little attention. Meanwhile, rapid progress has been made in the field of endovascular surgery. Modern endovascular procedures can permit highly targeted drug delivery by the intracarotid route. Intracarotid drug delivery can be the primary route of drug delivery or it could be used to facilitate the delivery of smart neuropharmaceuticals. There have been few attempts to systematically understand the kinetics of intracarotid drugs. Anecdotal data suggest that intracarotid drug delivery is effective in the treatment of cerebral vasospasm, thromboembolic strokes, and neoplasms. Neuroanesthesiologists are frequently involved in the care of such high-risk patients. Therefore, it is necessary to understand the applications of intracarotid drug delivery and the unusual kinetics of intracarotid drugs.

FOR more than 50 yr, intracarotid anesthetic drugs have been used in diagnostic neuroradiology to locate brain functions.1In the 1980s, intracarotid drugs were extensively investigated for the treatment of malignant brain tumors.2–6However, the attempts at intracarotid chemotherapy of brain tumors proved disappointing because of unexplained neurotoxicity and a relatively modest impact on the clinical outcome of the disease.7–9Therefore, the enthusiasm for intracarotid drug delivery waned by the early 1990s. Intracarotid injections can rapidly generate high concentrations of drug within a region of interest at a fraction of the total systemic dose.2,10Consequently, intracarotid delivery can serve as the primary method of drug delivery, or the technique could be used to increase the effectiveness of other methods of brain-selective drug delivery that target specific characteristics of the blood–brain barrier (BBB).11Currently, intracarotid drugs are used for localizing neurologic functions in the brain and for the treatment of intractable cerebral vasospasm, ischemic strokes, and intracranial malignancies.12 

In the past decade, rapid advances have been made in endovascular neurosurgery. These include the development of microcatheters and small balloon occluding catheters, which can be floated into distal regions of the brain. These devices can selectively deliver drugs or manipulate blood flows, in relatively small vascular territories consisting of 40–100 g of brain tissue.13The feasibility of intraarterial interventions on a limited scale decreases the risks of neurologic complications. Improved techniques of intraarterial delivery have also recently emerged that, compared with conventional infusions, can significantly augment tissue drug deposition.14–16Magnetic resonance imaging with temporal resolution sufficient to guide catheter placement, described as “interventional magnetic resonance imaging,” is also rapidly advancing and could lead to novel intraarterial interventions guided by fast magnetic resonance imaging and spectroscopy.17Collectively, such technological advances compel us to reevaluate intracarotid drug delivery and ascertain its role in the current and future treatments of brain diseases.

An understanding of intracarotid drug delivery is important to anesthesiologists who are often involved in the care of patients receiving intracarotid drugs. This review rationalizes the use of intracarotid drugs and describes their present and future applications. It also discusses anesthetic management of the current therapeutic interventions using intracarotid drugs.

Some key anatomical features of the cerebral circulation affect intracarotid drug delivery to the brain. These include the size of the brain relative to the body weight, anatomical configuration of cerebral arteries, the state of the BBB, and the anatomical compartments within the brain.

The Size of the Brain Relative to the Body Weight Varies across Animal Species

In general, relative to body size, primates have a much larger brain weight than nonprimate animals, and even among primates, humans have a much greater brain-to-body weight ratio.18As a result, primates can tolerate greater intracarotid doses of drugs based on body weight. For example, an intracarotid dose of 8 mg/kg of an antineoplastic drug, carmustine, produces severe hemorrhaging necrotizing lesions in dogs, but the same dose is well tolerated in rhesus monkeys and in humans.19–21 

The Configuration of Cerebral Arteries

Anatomical variations of the cerebral arteries can influence regional distribution and concentration of drugs after intracarotid delivery. The configuration of cerebral arteries varies (1) across animal species,22(2) within individuals of the same species, and (3) even in an individual over time.

The extent of communication between the external carotid artery and the internal carotid artery (ICA) greatly varies across animal species. Rabbits and primates show a clear separation of the intracranial and extracranial irrigations.23–25On the other hand, many animals, such as goats and pigs, have extensive collateral communications between the intracranial and extracranial arterial irrigations.22Even within animals of the same species, there can be significant differences in the origin and the size of the ICAs.24Smaller animals have larger external carotid arteries, and the sizes of the ICAs, relative to the vertebral arteries, differ across species. Therefore, ICA occlusion is fairly well tolerated in rodents and causes a transient decrease in blood flow in rabbits.26However, ICA occlusion often causes neurologic symptoms in humans.27,28The ability of human subjects to tolerate ICA occlusion depends largely on the configuration of the circle of Willis. Only 18% of human subjects have a balanced, symmetrical circle of Willis (fig. 1); therefore, there is a need to clinically test whether a patient can tolerate ICA occlusion before ICA sacrifice.29–31The configuration of the circle of Willis also determines the resistance of each arterial segment that could influence the distribution of intracarotid drugs.32 

Fig. 1. Some variations in the anatomy of the circle of Willis in human subjects. A complete balanced circle is seen in 18% of the general population. Hypoplastic segment of the posterior cerebral artery (PCoA) is seen in 22%. Hypoplastic segment of the anterior cerebral artery (A1) is seen in 25%. A fetal posterior cerebral artery (fetal PCA) with hypoplastic posterior cerebral artery is seen in 15%.  29,ACA = anterior cerebral artery; BA = basilar artery; MCA = middle cerebral artery; PCA = posterior cerebral artery. From Duong  et al. 29; used with permission. 

Fig. 1. Some variations in the anatomy of the circle of Willis in human subjects. A complete balanced circle is seen in 18% of the general population. Hypoplastic segment of the posterior cerebral artery (PCoA) is seen in 22%. Hypoplastic segment of the anterior cerebral artery (A1) is seen in 25%. A fetal posterior cerebral artery (fetal PCA) with hypoplastic posterior cerebral artery is seen in 15%.  29,ACA = anterior cerebral artery; BA = basilar artery; MCA = middle cerebral artery; PCA = posterior cerebral artery. From Duong  et al. 29; used with permission. 

Close modal

Furthermore, the blood flow in a given artery can change with time, which could affect the regional tissue concentrations of intracarotid drugs. It has been shown that the pharmacologic effects of intracarotid drugs, such as anesthetics, are directly related to cerebral blood flow (CBF).33,34If changes in blood flow have a significant effect on tissue drug concentrations, the arterial concentrations generated after injection of the same dose of a drug might be different over time even in the same individual. Consider, for example, superselective intracarotid infusion of papaverine for cerebral vasospasm. Initially, when blood flow is low, the concentration of the drug is high because of minimal dilution with arterial blood. As vasospasm resolves, blood flow increases and arterial concentration declines because of greater dilution.35In theory, the vasodilatory effects of intracarotid drugs that augment local blood flow may paradoxically decrease regional deposition of the drugs.33 

The Blood–Brain Barrier

The BBB is primarily constituted by tight junctions between the endothelial cells that severely restrict migration of molecules from the blood into the brain tissue. In other capillary beds, there are clefts between the endothelial cells that permit passage of molecules. However, in the brain, the tight endothelial junctions permit only transcellular migration of molecules by diffusion, by carrier transport, or rarely by pinocytosis.36,37Abnormalities of endothelial tight junctions are seen with brain tumors and could be responsible for tumor-related brain edema.38Morphologically, endothelial cells of brain arteries, compared with other regions of the body, are rich in mitochondria. These mitochondria are also larger in size. This suggests that endothelial cells are metabolically very active.39–41Endothelial cells also are rich in metabolic enzymes such as catecholamine methyltransferase, monoamine oxidase, and adenosine deaminase,42which create a chemical barrier to drugs, such as catecholamines and adenosine.43In general, small nonpolar lipid soluble drugs can easily penetrate the BBB by passive diffusion. In contrast, the effective delivery of macromolecules that include antineoplastic drugs and nucleotides is possible only when the BBB is disrupted.44 

Physiologic Compartments in the Brain

The brain is best described in terms of three compartments: blood, cerebral spinal fluid, and brain parenchyma. The brain tissue itself can be further divided into gray and white matter. Additional compartments might be created within the brain parenchyma that have their own kinetic characteristics in pathologic states (fig. 2).10Consequently, under physiologic conditions, there are three interphases between the compartments in the brain of which the BBB is the most significant.44The BBB has been regarded as the gatekeeper to the brain. Compared with the blood–cerebrospinal fluid interphase, the BBB has 5,000-fold greater area and has much lower permeability.45 

Fig. 2. Schematic representations of the compartments in the brain and the complex kinetics of intracarotid drug delivery. Brain parenchyma is not a homogenous entity and is separated into gray and white matter.  Bidirectional arrows represent the “diffusivity of the drugs” within the brain parenchyma. Pathologic lesion (Pathology) may create additional tissue compartments. Three barriers separate the compartments: blood–brain, blood–cerebral spinal fluid (CSF), and CSF–brain. Of the three, the blood–brain barrier is the most significant. From Joshi  et al. 71; used with permission. 

Fig. 2. Schematic representations of the compartments in the brain and the complex kinetics of intracarotid drug delivery. Brain parenchyma is not a homogenous entity and is separated into gray and white matter.  Bidirectional arrows represent the “diffusivity of the drugs” within the brain parenchyma. Pathologic lesion (Pathology) may create additional tissue compartments. Three barriers separate the compartments: blood–brain, blood–cerebral spinal fluid (CSF), and CSF–brain. Of the three, the blood–brain barrier is the most significant. From Joshi  et al. 71; used with permission. 

Close modal

Figure 3shows the simple kinetic framework described by Dedrick10demonstrating the advantage of intracarotid injection. If C1and C2are the regional and systemic drug concentrations, the pharmacokinetic advantage of intracarotid (ic) over intravenous (iv) infusion can be defined as

Fig. 3. Model of intracarotid drug delivery. C1, C2and V1, V2are the concentrations and volumes in a brain (1) and the remaining body (2), respectively. Q is the arterial blood flow and venous return. CL1and CL2are the cerebral and the remaining body clearances. ICA = internal carotid artery. From Dedrick  10; used with permission. 

Fig. 3. Model of intracarotid drug delivery. C1, C2and V1, V2are the concentrations and volumes in a brain (1) and the remaining body (2), respectively. Q is the arterial blood flow and venous return. CL1and CL2are the cerebral and the remaining body clearances. ICA = internal carotid artery. From Dedrick  10; used with permission. 

Close modal

In figure 3, I is the rate of drug infusion, Q is the regional blood flow, E is the first-pass extraction of the drug from the region, and CL1and CL2are the clearances from the region and from the rest of the body. CLTBis clearance from the whole body (CL1+ CL2). R can also be derived as

If CL1= 0, then

Conversely, if CL2= 0, then

Equation 1and its derivatives 1a and 1b show that the maximum advantage of intracarotid drug infusion is evident (1) with drugs that have high CLTB, (2) when the regional arterial flow (Q) is low, or (3) when the regional extraction is high. For example, we can illustrate the advantage of intracarotid delivery of propofol using equation 1a. Assuming an ICA flow to be 200 ml/min and CLTBof propofol to be 2,100 ml/min for a 7-kg individual, R propofol will be 1 + 10.5 = 11.5. This theoretical number is fairly close to the observed experimental results showing a 10-fold reduction in dose of propofol required to produce electrocerebral silence with intracarotid delivery compared with intravenous infusion.46Conversely, the possibility of achieving high intracarotid concentrations also suggests that intracarotid infusion might result in locally toxic drug concentrations without any evidence of systemic toxicity.

Pitfalls in Kinetic Modeling of Intracarotid Drug Infusions

There are several assumptions in the kinetic model proposed by Dedrick.10These include uniform mixing of drug in the blood, determining free drug concentration based on conventional steady state drug–protein interactions, constant clearances over time, constant regional blood flow, homogenous distribution within the arterial irrigation, and homogenous behavior of the brain compartment. Two of these assumptions merit close scrutiny:

Estimation of the Free Drug Concentration after Intracarotid Injection.

According to the “free-drug hypothesis,” it is the free drug in the plasma that is available for diffusion across the BBB. However, in vivo  during intracarotid injections, the tissue brain concentrations are higher than those predicted by the free-drug hypothesis. Jones et al.  47compared the uptake of different benzodiazepines after intracarotid bolus injection in rats. They found that the brain uptake of this class of drugs was largely determined by their lipophilicities. However, in vitro  modeling grossly underestimated the in vivo  uptake of these drugs. They determined that observed in vivo  data deviated from the more theoretical model with higher doses of albumin that were injected along with the drug. The free drug concentration seemed to be 5- to 25-fold greater in vivo  than predicted by in vitro  observations.47Several possible explanations have been offered for this observation. First is the “free-intermediate hypothesis,” which states that rapid uptake of a drug into the brain enhances the release of drug bound to plasma proteins.48,49The second possibility is that other proteins in the blood have higher drug affinity, such that in vitro  estimates of free drug concentrations based on albumin binding alone underestimate the blood concentrations.49,50The third possibility is that there might be some drug–protein binding competitor in the capillaries that increases the drug–protein dissociation rate.51Fourth, when drugs bind to proteins, they are absorbed by a peptide transporter system, which may increase transfer across the BBB.52 

Heterogenous Nature of Brain Compartments.

It is best to consider the brain parenchyma in terms of separate compartments with distinct units as described previously: (1) gray matter, (2) white matter, and (3) pathologic compartments, such as tumor or ischemic regions, within the brain parenchyma.53,54These compartments will differ with respect to their blood flow, BBB functions, and ability to extract and metabolize drugs. For example, the blood flow to the gray matter (50 ml · 100 g−1· min−1) is approximately twofold greater than in the white matter (25 ml · 100 g−1· min−1) and fourfold greater than in the ischemic regions (12 ml · 100 g−1· min−1).55Baseline variability in regional blood flow could result in differences in drug concentrations in the brain tissue compartments after intraarterial injection.10Eventually, effective modeling of intracarotid drug delivery will require an understanding of the micropharmacokinetic factors that determine drug distribution within the brain tissue (fig. 4).56 

Fig. 4. Micropharmacokinetics of drug delivery to the brain: potential factors that could affect drug concentrations at it specific site of action. BBB = blood–brain barrier. From Wang and Joshi  56; used with permission. 

Fig. 4. Micropharmacokinetics of drug delivery to the brain: potential factors that could affect drug concentrations at it specific site of action. BBB = blood–brain barrier. From Wang and Joshi  56; used with permission. 

Close modal

Intracarotid infusion of drugs offers the following theoretical advantages:

High Regional Arterial Concentration

Intracarotid drug infusion can restrict the initial volume of distribution to one cerebral hemisphere. The volume of blood flow through the ICA in humans has been estimated to be approximately 200 ml/min, whereas the cardiac output is 5–6 l/min. Consequently, drugs that are infused into the small volume of ICA flow achieve relatively high initial arterial concentrations at low total doses, thereby decreasing systemic toxicity.46,57,58 

High Free Drug Concentrations

Although the exact nature of the interactions between the drug and binding proteins after intracarotid infusions remains to be fully understood, limited data suggest that intracarotid administration of drugs results in disproportionately higher free drug concentrations than those predicted by steady state kinetic models.47Recent videomicroscopic images have revealed that bolus injections of drugs transiently overwhelm the blood flow and may deliver virtually undiluted drug to the brain.59Such injections, in theory, would considerably attenuate the decrease in free drug concentration due to protein binding or the uptake by blood cells. Manipulating the key parameters of bolus injection, volume, concentration, and frequency can have a significant effect on the tissue concentrations of intracarotid drugs.59Bolus injections, when combined with regional blood flow manipulation, could therefore significantly enhance intracarotid drug delivery.34 

Rapid Onset of Action

Intracarotid delivery can achieve rapid, virtually instantaneous, high drug concentrations in the brain. Such an ability to instantaneously generate high tissue concentrations may be critical in some clinical situations, such as preventing reperfusion injury after intracarotid thrombolysis.60–62 

The theoretical disadvantages of intracarotid infusions can be summarized as follows:

The High Resting CBF as a Disadvantage

As the target organ, the brain, with a relatively high resting blood flow that amounts to 15–20% of the cardiac output, is at a disadvantage compared with some other less-well-perfused organs.63High blood flow decreases the peak drug concentrations due to greater dilution by the arterial blood. Increased blood flow to the brain could decrease the drug transit time through cerebral circulation if the cerebral blood volume was not proportionately increased. Transit time seems to have a direct effect on tissue concentrations of highly lipid-soluble anesthetic drugs after intracarotid injections.56In addition, high blood flow will augment drug efflux from the brain tissue.

Kinetics of Intracarotid Drugs Are Difficult to Model

For example, under physiologic conditions, the brain has at least three compartments: gray matter, white matter, and cerebrospinal fluid. These compartments demonstrate unique kinetic characteristics. Pathologic tissue, such as cerebral edema or brain tumor, may have significantly different kinetic properties. These factors may make it difficult to model drug kinetics in physiologic and pathologic states and has led to the failure of theoretical models of intracarotid drug delivery.64 

Loss of Intracarotid Dose Advantage over Time

The advantages of intracarotid versus  intravenous drug delivery decrease with prolonged infusions due to lower peak regional concentrations and the eventual redistribution of the drug. Computer simulations suggest that intracarotid drug infusion over 2 h versus  the same dose infusion over 10 h results in regional concentrations that are 5.5- and 3.1-fold greater than intravenous delivery, respectively.10Experimental data also suggest that the comparative dose advantages of intracarotid versus  intravenous drug delivery decrease when infusions are used for prolonged periods of time. In rabbits, intracarotid propofol achieves transient electrocerebral silence at one tenth the intravenous dose. On the other hand, to maintain electrocerebral silence for 1 h, intracarotid delivery is only 5-fold as effective as the intravenous injection.46 

Brain Tissue Drug Concentration Measurements Are Challenging

Measurements of drug concentrations are particularly challenging with intracarotid drug delivery to the brain. Postmortem samples do not provide time histories.65Multiple tissue biopsies, though feasible, tend to injure the preparation and are not site specific.66Insights into the field have been limited to a few magnetic resonance imaging positron emission tomography studies and a few radiolabeled drug–based studies.47,67–70The α half-life of many lipid-soluble drugs, such as carmustine and propofol, is exceedingly short, ranging between 1.4 and 7 min.68,71Microdialysis requires time to obtain sufficient sample for analysis, is invasive, and may alter tissue characteristics.72,73Therefore, the exceedingly rapid changes in tissue drug concentrations, particularly after bolus injections, are beyond the time resolution of microdialysis. Novel optical techniques, such as diffuse reflectance spectroscopy, can noninvasively measure tissue drug concentrations in virtual real time, which could provide better understanding of intracarotid drug kinetics.74,75 

From the practical standpoint, intracarotid drug delivery could be improved by the following means:

Disrupting the Blood–Brain Barrier

Controlled disruption of the BBB can be achieved by either hyperosmotic agents or chemical means.76,77 

Hyperosmotic Disruption

Intracarotid injection of hypertonic substances such as mannitol and arabinose leads to a transient loss of BBB functions (fig. 5).78Hypertonic agents cause vasodilation and shrinkage of the endothelial cells, resulting in increased diffusivity and bulk flow across the BBB.77Cell shrinkage, along with the contraction of the endothelial cytoskeleton, results in widening of the tight junction to approximately 20 nm, which results in a 10-fold increase in permeability of some compounds. Alternately, some believe that mechanisms other than cell shrinkage, such as alterations in Na+–Ca2+exchange, might play a role during osmotic disruption of the BBB.79The effects of hypertonic mannitol were previously thought to be exceedingly transient, although data suggest that in humans, hypertonic BBB disruption lasts for 30 min and BBB functions remain impaired for several hours afterward.80Zunkeler et al.  showed that the healthy BBB was almost 15-fold more susceptible to osmotic disruption than the BBB in glioma tumors.81,82Biomechanical factors such as systemic hypotension seem to mitigate osmotic disruption of the BBB, whereas hypertension enhances disruption.83,84In experimental settings, the efficacy of hypertonic mannitol can also be enhanced either by cooling the solution to 4°C or with a Na+–Ca2+exchange blocking drug, KB-R7943.85,86There is evidence to suggest that treatment with steroids, anesthetics, and magnesium can mitigate BBB disruption by intracarotid mannitol.87–90Restoring BBB functions after intracarotid mannitol may help in limiting the complications of hypertonic mannitol therapy.88,91–93 

Fig. 5. Disruption of the blood–brain barrier with intracarotid mannitol. The figure shows the disruption of the blood–brain barrier by intracarotid injection of mannitol (25%) in New Zealand White rabbits as evidenced by the distribution of 2% Evans blue dye: 2-ml (  A ), 4-ml (  B ), 6-ml (  C ), and 8-ml (  D ) mannitol bolus injected through the carotid artery in 1 min. From Wang  et al. 83; used with permission. 

Fig. 5. Disruption of the blood–brain barrier with intracarotid mannitol. The figure shows the disruption of the blood–brain barrier by intracarotid injection of mannitol (25%) in New Zealand White rabbits as evidenced by the distribution of 2% Evans blue dye: 2-ml (  A ), 4-ml (  B ), 6-ml (  C ), and 8-ml (  D ) mannitol bolus injected through the carotid artery in 1 min. From Wang  et al. 83; used with permission. 

Close modal

Chemical Disruption of the Blood–Brain Barrier

Chemical disruption of BBB by intracarotid injection of bradykinins such as Cereport (RMP-7) or leukotrienes (LTC-4) results in transient opening of the BBB.76Contrary to the disruption by mannitol, disruption of the BBB by Cereport and leukotrienes seems to be limited to pathologic lesions and not normal brain. The effects of the drug are reversible within 30–60 min. Cereport can increase the uptake of intraarterially delivered antineoplastic drugs.94,95 

Superselective Intracarotid Delivery of Drugs

One of the ways to minimize the side effects of intracarotid drugs is to restrict their delivery to pathologic lesions by superselective cannulation of the feeding arteries. With the development of highly flexible flow directed catheters with external diameters of 1.5–2 mm, it is possible to cannulate distal branches of the carotid artery without the risk of obstructing the blood flow (fig. 6).96The problem with distal cannulation is the increased risk of streaming (see Streaming of Drugs section).97However, side port catheters and diastolic pulse delivery can minimize the chances of streaming.98The theoretical advantages of superselective delivery of drugs include minimizing the total dose, decreasing the risk of regional toxicity, and limiting potential vascular complications to the pathologic regions of the brain. In case of intracarotid vasodilators, superselective infusions will decrease the chances of cerebral steal or the likelihood of an increase in intracranial pressure (ICP). A key concept recently introduced by Gobin et al.  14was to determine the dose of intracarotid drugs based on a spatial fractionation algorithm. The algorithm calculates the intracarotid dose requirements during superselective drug injections based on the volume of tissue perfused by a given intracranial artery. Such an approach decreases the risks of neurologic complications during intracarotid chemotherapy.

Fig. 6. Superselective placement of microcatheters in the distal regions of the brain. Microcatheters of 1.2- to 1.8-French sizes can be navigated into the distal brain for delivering drugs; measuring physiologic parameters, such as pressure and blood flow; and repairing blood vessels. Typically, two to three coaxial catheters are used. The first catheter, not shown in the figure, is located in the femoral artery (the introducer sheath). The second catheter is placed in the internal carotid artery (ICA) guiding catheter, and the third catheter (microcatheter) is guided in the distal regions of the brain (  A ). In this example, catheters are being used for measuring change in arterial blood pressure, cerebral blood flow, and cerebral blood flow velocity to determine the site of effect of intracarotid vasodilators.  B and  C show the placement of detectors. The model can be used to determine changes in proximal arterial resistance and can estimate changes in the cerebral arteries that lie distally beyond the tip of the microcatheter (  D ). MCA = middle cerebral artery; TCD = transcranial Doppler; Xe = xenon. From Joshi  et al. 96 

Fig. 6. Superselective placement of microcatheters in the distal regions of the brain. Microcatheters of 1.2- to 1.8-French sizes can be navigated into the distal brain for delivering drugs; measuring physiologic parameters, such as pressure and blood flow; and repairing blood vessels. Typically, two to three coaxial catheters are used. The first catheter, not shown in the figure, is located in the femoral artery (the introducer sheath). The second catheter is placed in the internal carotid artery (ICA) guiding catheter, and the third catheter (microcatheter) is guided in the distal regions of the brain (  A ). In this example, catheters are being used for measuring change in arterial blood pressure, cerebral blood flow, and cerebral blood flow velocity to determine the site of effect of intracarotid vasodilators.  B and  C show the placement of detectors. The model can be used to determine changes in proximal arterial resistance and can estimate changes in the cerebral arteries that lie distally beyond the tip of the microcatheter (  D ). MCA = middle cerebral artery; TCD = transcranial Doppler; Xe = xenon. From Joshi  et al. 96 

Close modal

Localizing the Drug Effects

Intracarotid infusions can be further targeted to the brain by enhancing their removal from the systemic circulation, by extracorporeal removal, by forced alkaline diuresis, or by neutralizing the effects of a drug by the coadministration of a systemic antidote.3,99The use of drugs, such as adenosine, with exceedingly short biologic half-lives greatly minimizes the risks of systemic side effects and could potentially lead to “fire-and-forget” types of intraarterial drug interventions.58 

Manipulating CBF to Increase Drug Delivery

Early computer simulations showed that intracarotid drug delivery to the brain is particularly useful in low-blood-flow states.10In a series of experiments, CBF was altered by changing ventilation, by augmenting blood flow with intracarotid verapamil pretreatment, or by producing transient flow arrest. The dose of an intracarotid anesthetic, propofol, required to produce electroencephalographic silence, was directly related to the blood flow.33Both hypercapnia and verapamil pretreatment, which increased CBF, increased the dose requirements of intracarotid anesthetics, whereas flow arrest significantly increased the duration of electroencephalographic silence after bolus anesthetic injection (figs. 7 and 8).33,34 

Fig. 7. Effect of blood flow on the dose requirements of intracarotid propofol. Intraarterial verapamil is a potent cerebral vasodilator. (  A ) The dose of intracarotid propofol required to produce 10 min of electrocerebral (electroencephalographic [EEG]) silence significantly increases with verapamil pretreatment (n = 14,  P = 0.004). (  B ) The dose requirement of propofol is linearly related to the increase in blood flow after verapamil pretreatment (  r = 0.75,  P = 0.0021). (  C ) Increase in cerebral blood flow with changes in ventilation directly affects the dose of propofol required for electrocerebral silence. HYO = hypocapnia; HYR = hypercapnia; NOR = normocapnia. Adapted from Joshi  et al. 33 

Fig. 7. Effect of blood flow on the dose requirements of intracarotid propofol. Intraarterial verapamil is a potent cerebral vasodilator. (  A ) The dose of intracarotid propofol required to produce 10 min of electrocerebral (electroencephalographic [EEG]) silence significantly increases with verapamil pretreatment (n = 14,  P = 0.004). (  B ) The dose requirement of propofol is linearly related to the increase in blood flow after verapamil pretreatment (  r = 0.75,  P = 0.0021). (  C ) Increase in cerebral blood flow with changes in ventilation directly affects the dose of propofol required for electrocerebral silence. HYO = hypocapnia; HYR = hypercapnia; NOR = normocapnia. Adapted from Joshi  et al. 33 

Close modal

Fig. 8. Increase in the duration of electroencephalographic (EEG) silence with intracarotid anesthetic drugs injected during transient cerebral hypoperfusion (CHP). The duration of EEG silence after intracarotid injection of thiopental (3 mg as 1% solution,  A ) and propofol (3 mg as 1% solution,  B ). In thiopental-1 and -3 and propofol-1 and -3, injections were undertaken with normal blood flow; whereas the second injections (thiopental-2 and propofol-2) were made during transient CHP. During transient CHP, the blood flow decreased to 10–20% of baseline for less than 60 s. The effects of the anesthetics were significantly prolonged when they were injected during CHP. Adapted from data in Joshi  et al. 33,34 

Fig. 8. Increase in the duration of electroencephalographic (EEG) silence with intracarotid anesthetic drugs injected during transient cerebral hypoperfusion (CHP). The duration of EEG silence after intracarotid injection of thiopental (3 mg as 1% solution,  A ) and propofol (3 mg as 1% solution,  B ). In thiopental-1 and -3 and propofol-1 and -3, injections were undertaken with normal blood flow; whereas the second injections (thiopental-2 and propofol-2) were made during transient CHP. During transient CHP, the blood flow decreased to 10–20% of baseline for less than 60 s. The effects of the anesthetics were significantly prolonged when they were injected during CHP. Adapted from data in Joshi  et al. 33,34 

Close modal

There are several significant practical concerns with regard to intracarotid drug delivery to the brain:

The Risk of Embolism and Hemorrhage during Endovascular Interventions

Recent review suggests that the incidence of neurologic complications after endovascular procedures varies a great deal, from 5.6% during the placement of a carotid stent to 19% during occlusion of a parent artery in the treatment of cerebral aneurysms.100A significant factor contributing to this morbidity is cerebral embolism due to thrombi, dislodged atheromatous plaques, or air emboli. A number of strategies are being developed to interrupt the thromboembolic pathway. Soft, flow-guided catheters can decrease the risk of endothelial injury. Adhesion, activation and aggregation of platelets can be inhibited with agents such as aspirin, ticlopidine, and clopidogrel. The coagulation cascade can be blocked by inhibiting the activation of fibrinogen by thrombin. The antithrombin effects (as well as the potential adverse effects) of heparin are well known, can easily be monitored by measuring the activated clotting time, and can be easily reversed with protamine. Therefore, heparin provides convenient thromboembolic prophylaxis in clinical settings. Although heparin has been the mainstay of thromboembolic prophylaxis during endovascular surgery, newer strategies, such as hirudin analog or platelet receptor antibodies, may find wider application in the future.101Endovascular interventions in the background of significant anticoagulation carry a greater risk of hemorrhagic stroke as a complication. Catastrophic bleeds during the procedure necessitate immediate and aggressive endovascular and surgical interventions. Rapid reversal of the anticoagulant effects of heparin with protamine is particularly useful in such situations. A well-thought-out management strategy should be in place before anticoagulation.102 

The risk of thromboembolism is also directly related to the duration of endovascular catheterization. Prolonged catheterization of the cerebral arteries can permit repeated cerebral angiography to monitor response or facilitate delivery of drugs. Retrograde cannulation of the superficial temporal arteries was used to permit cerebral angiography over a week without any complication in patients with intracranial aneurysms.103Kallmes et al.  104have used a swine model to demonstrate that catheters with hydrophilic surfaces were less thrombogenic than those with hydrophobic surfaces. The risk of thromboembolism also depended on the catheter material not just surface coating. In this model, long-term implantation of a microcatheter was well tolerated for the longest duration of the study, 35 days.

Air Embolism

One of the less-recognized risks of intracarotid drug therapy is the accidental injection of air, sometimes dissolved in fluids. Studies with transcranial Doppler flow measurements have revealed a frequent occurrence of microscopic air emboli during angiography. Microscopic air entrainment is likely to occur at two times during the angiographic procedure: during the aspiration of contrast into the syringe and during the injection of contrast. Increased viscosity of contrast increases the risk of air entrainment. The chances of air emboli being injected are decreased if the syringes are allowed to stand. The risk of air embolism is directly related to the rate of injection and inversely related to its viscosity. Therefore, decreasing the rate of injection decreases the chances of air embolism.105 

Streaming of Drugs

Streaming of drugs  refers to the uneven distribution of drugs in the arterial stream that results in different drug concentrations within that arterial distribution.106Streaming has been observed in vivo  and in vitro  and has been invoked to explain focal drug toxicity of chemotherapeutic agents within arterial distribution (figs. 9 and 10). Using a polystyrene cast of human cerebral arteries, it has been shown that streaming depends on the rate of drug infusion, the type of catheter used for drug delivery, and the position of the catheter in relation to the arterial branching. This in vitro  model suggests that there can be an almost 5-fold difference in drug concentrations due to streaming.107In a human study using O15positron emission tomography, Saris et al.  98demonstrated that there can be an 11-fold variation in regional drug concentration within the arterial irrigation of the supraophthalmic segment of the ICA.

Fig. 9. Factors that influence the extent of streaming in an  in vitro model of human cerebral arteries.  A shows streaming of the dye (2 ml/min) from a P-100 catheter in a supraophthalmic position  (bold vertical arrow ). Note that there is a clear stream of dye in the anterior choroidal artery (Ach A) and that there is no flow of the dye to the anterior cerebral artery (ACA).  B shows a more homogeneous dispersion of the dye when it is injected in an infraophthalmic location at a rate of 17 ml/min. A proximal site and a high rate volume rate of infusion, decrease the variability in drug concentration due to streaming. From Lutz  et al. 107; used with permission. 

Fig. 9. Factors that influence the extent of streaming in an  in vitro model of human cerebral arteries.  A shows streaming of the dye (2 ml/min) from a P-100 catheter in a supraophthalmic position  (bold vertical arrow ). Note that there is a clear stream of dye in the anterior choroidal artery (Ach A) and that there is no flow of the dye to the anterior cerebral artery (ACA).  B shows a more homogeneous dispersion of the dye when it is injected in an infraophthalmic location at a rate of 17 ml/min. A proximal site and a high rate volume rate of infusion, decrease the variability in drug concentration due to streaming. From Lutz  et al. 107; used with permission. 

Close modal

Fig. 10. Effect of the method of drug injection on the distribution of intracarotid drugs. Reconstructed autoradiographic images show distribution of tracer at two levels of a nonhuman primate brain, fronto-parietal cortex (level 1) and through basal ganglia and the temporal lobe (level 2).  (A ) Injection of intracarotid tracer as pulses during diastole shows homogenous tissue drug concentrations.  (B ) Intravenous infusion of drugs also produces similar homogenous concentration.  (C ) Intracarotid infusion of the tracer produces marked variations in tissue tracer concentrations due to streaming. These results suggest that injection of intracarotid drugs as diastole-phase pulses considerably decreases variations in tissue drug concentrations due to streaming. From Saris  et al. 157; used with permission. 

Fig. 10. Effect of the method of drug injection on the distribution of intracarotid drugs. Reconstructed autoradiographic images show distribution of tracer at two levels of a nonhuman primate brain, fronto-parietal cortex (level 1) and through basal ganglia and the temporal lobe (level 2).  (A ) Injection of intracarotid tracer as pulses during diastole shows homogenous tissue drug concentrations.  (B ) Intravenous infusion of drugs also produces similar homogenous concentration.  (C ) Intracarotid infusion of the tracer produces marked variations in tissue tracer concentrations due to streaming. These results suggest that injection of intracarotid drugs as diastole-phase pulses considerably decreases variations in tissue drug concentrations due to streaming. From Saris  et al. 157; used with permission. 

Close modal

Local versus  Systemic Toxicity

The delivery of drugs directly into the brain has been associated with regional toxicity. There is an increased incidence of white matter lesions with intracarotid chemotherapy compared with intravenous therapy.108,109Streaming is also thought to be responsible for retinal injury after intracarotid infusion of antineoplastic drugs.110Therefore, in defining toxicity, one has to factor in the total dose of the intraarterial drug, as well as the highest possible concentration that might result from inadequate mixing. In addition, possibility of injury to blood vessels, such as to the vascular endothelium, should also be considered.111,112 

Standardizing the Dose of Intracarotid Drugs

One of the major problems in intracarotid drug therapy is the standardization of doses within individuals of the same species or during the extrapolation of data between animals of different species. Doses have been described in terms of weight/unit body surface area,113total milligram dose, or infusion rates in mg/min, and dose/kg body weight. Investigators have also assessed the response to intracarotid drugs as a function of estimated molar concentrations.114–116In the absence of any consistent way to present dose data, it is sometimes difficult to compare drug response in different studies. We recommend that to optimally interpret intracarotid dose–response data, one has to consider all three infusion parameters: (1) The rate of drug infusion (mg/min, or mol/min), which, along with the blood flow, determines peak concentrations. (2) Estimated concentration in arterial blood, i.e. , dose of drug/volume of blood flow in the infused artery. With superselective infusions, drug concentrations may be estimated by the volume of tissue infused based on angiographic measurements.117(3) The total dose when it is sufficient to cause systemic toxicity.

The exact description of intracarotid doses is particularly important in extrapolating the doses from one animal species to another. Because of the differences in relative sizes of various organs, scaling of intracarotid doses merely on the basis of body weight or body surface area can lead to errors. For example, within individuals of a given species, parameters such as blood flow in the ICA can be assumed to be relatively constant, but across animal species, those assumptions may not be valid. Therefore, the same amount of drug will generate different arterial blood concentrations. Because of the relatively large size of the brain and higher carotid arterial blood flow, primates tolerate a much higher dose of intracarotid drugs compared with rodents or dogs. To undertake intraspecies comparisons, Dedrick118described kinetic models based on organ size and surface areas. These relative sizes of organs differed between species but their function and surface characteristics were similar. Even when such allometric corrections are made, it is still difficult to project intracarotid kinetic data from one animal species to another.

Intracarotid Anesthetics for the Localization of Brain Functions

More than 50 yr ago, Wada developed the technique of injecting sodium amytal into the carotid artery.1The procedure was originally developed to permit unilateral electroconvulsive therapy but subsequently became a standard method for localizing language function and memory. Other anesthetic drugs that have been used for Wada testing include methohexitone and, recently, propofol.1,119–122Compared with amytal, intracarotid methohexitone seems to have a shorter duration of effect, thereby making it possible to test multiple arterial territories in the same setting.122However, there seems to be poor justification for the selection of intracarotid doses.71Most radiologists use a graded dose of 75–125 mg amytal mixed with contrast media. After an initial dose of 75 mg, additional boluses of 25 mg are injected until there is an upper limb drift. Speech, language, and memory functions are assessed as the deficit resolves. Intracarotid amytal in doses as high as 3 mg/kg or 200 mg has been used during the Wada test. Such high intracarotid doses of anesthetic drugs on recirculation suppress the contralateral hemisphere. Any additional baseline sedation provided by an anesthesiologist in attendance would further complicate the interpretation of the Wada test. Therefore, it has been recommended that when high doses of amytal are used, the contralateral hemisphere should be tested on a subsequent day.123 

In recent years, there has been considerable interest in the use of propofol for Wada testing.120In a recent study, however, as many as 19 of 58 patients developed transient neurologic symptoms. These consisted of tonic movement, confusion, and pain. The symptoms were seen when more than 10 mg of the drug was rapidly injected in patients older than 55 yr. None of these patients had any permanent complication, but the authors recommend limiting the dose of propofol to 10 mg and careful monitoring in patients older than 55 yr.124Despite these recent reports with propofol, the safety of the Wada test is underscored by the fact that the test has been widely used for a very long time. Although neurologic complications have been reported due to arterial emboli, they do not seem to be frequent.125 

Superselective Wada testing is also used before embolization of cerebral arteriovenous malformations (fig. 11). Because embolization of the lesion could injure both gray and white matter, injection of amytal is combined with injection of lidocaine to test for both the gray and the white matter. Amytal is injected first to suppress gray matter activity, followed by the injection of lidocaine to suppress nerve conduction through the white matter. Anesthesiologists involved in the care of patients undergoing Wada tests should use judicious amounts of sedation. Both propofol and dexmedetomidine have been used for the purpose. Midazolam may impair memory testing and therefore has to be used cautiously.

Fig. 11. Superselective Wada testing: a case report. A 32-yr-old woman with seizures and left hemicranial headaches was found to harbor a subcortical arteriovenous malformation (AVM) in the left parietal lobe. (  A ) Left carotid arteriography confirmed the presence of the AVM with multiple nidal aneurysms (  arrow ). (  B ) During local anesthesia, only a 0.5-mm diameter microcatheter was placed in close proximity of the AVM for the injection of anesthetic drugs, amytal and lidocaine, for superselective Wada testing. The testing is performed in conjunction with members of the neuropsychological service to determine the risk of causing neurologic deficits both at embolization or surgical resection. In this case, no changes in function were identified. (  C ) Fluorangiography of the cranium after the occlusion of the AVM with n-butyl-cyanoacrylate shows radio-opaque glue filling the sinusoids within the AVM nidus (  arrow ). (  D ) Control angiography after occlusion of the AVM shows complete obliteration of the AVM nidus and no other branch vessel occlusion (  arrow ). The patient tolerated embolization and resection without any complication and returned to work in 9 days. Images and case history are provided by Phillip M. Meyers, M.D. 

Fig. 11. Superselective Wada testing: a case report. A 32-yr-old woman with seizures and left hemicranial headaches was found to harbor a subcortical arteriovenous malformation (AVM) in the left parietal lobe. (  A ) Left carotid arteriography confirmed the presence of the AVM with multiple nidal aneurysms (  arrow ). (  B ) During local anesthesia, only a 0.5-mm diameter microcatheter was placed in close proximity of the AVM for the injection of anesthetic drugs, amytal and lidocaine, for superselective Wada testing. The testing is performed in conjunction with members of the neuropsychological service to determine the risk of causing neurologic deficits both at embolization or surgical resection. In this case, no changes in function were identified. (  C ) Fluorangiography of the cranium after the occlusion of the AVM with n-butyl-cyanoacrylate shows radio-opaque glue filling the sinusoids within the AVM nidus (  arrow ). (  D ) Control angiography after occlusion of the AVM shows complete obliteration of the AVM nidus and no other branch vessel occlusion (  arrow ). The patient tolerated embolization and resection without any complication and returned to work in 9 days. Images and case history are provided by Phillip M. Meyers, M.D. 

Close modal

Cerebral Vasospasm

Cerebral vasospasm  is a pathologic narrowing of the cerebral arteries. The fundamental method to overcome this added arterial resistance, whether proximal or distal, is to increase the cerebral perfusion pressure and the cardiac output by using hypervolemic hemodilution with induced hypertension (triple-H treatment).126A significant number of patients with cerebral vasospasm do not respond to triple-H treatment. Furthermore, triple-H treatment has to be carefully applied to patients with untreated or multiple aneurysms. The alternate strategy in such cases is to decrease cerebrovascular resistance by superselective intracarotid or intracarotid infusion of vasodilators.58,117,127,128Intracarotid papaverine has been the mainstay of such a treatment, although a variety of drugs such as calcium channel blockers, mannitol, and prostaglandin E have also been used intraarterially for treating vasospasm (fig. 12). Other drugs, such as adenosine, have been proposed for the treatment of vasospasm and, despite their significant vasodilator effects, have not yet been used in clinical settings, probably because of their short duration of effect and poor penetration of the BBB (fig. 13).

Fig. 12. Treatment of cerebral vasospasm with intracarotid verapamil. A 57-yr-old woman developed sudden severe headache and collapsed to the floor. On admission to the hospital emergency department, she was comatose with decorticate posturing. (  A ) Computed tomographic brain scan without contrast demonstrates diffuse subarachnoid hemorrhage (  arrow ). (  B and  C ) Cerebral arteriography demonstrates a 7-mm left posterior communicating artery aneurysm (  arrow ). There is no spasm in the cerebral arteries at the time of initial presentation. (  D ) Surveillance arteriography on day 7 after the hemorrhage shows moderate to severe, flow-limiting spasm of the cerebral arteries (  arrows ). (  E ) To prevent stroke, endovascular treatment including balloon angioplasty (  arrow ) followed by vasodilator infusion therapy with 5 mg verapamil was performed. (  F ) Cerebral arteriography after treatment shows both improved vessel dimension and flow in the cerebral vasculature. Images and case history are provided by Phillip M. Meyers, M.D. 

Fig. 12. Treatment of cerebral vasospasm with intracarotid verapamil. A 57-yr-old woman developed sudden severe headache and collapsed to the floor. On admission to the hospital emergency department, she was comatose with decorticate posturing. (  A ) Computed tomographic brain scan without contrast demonstrates diffuse subarachnoid hemorrhage (  arrow ). (  B and  C ) Cerebral arteriography demonstrates a 7-mm left posterior communicating artery aneurysm (  arrow ). There is no spasm in the cerebral arteries at the time of initial presentation. (  D ) Surveillance arteriography on day 7 after the hemorrhage shows moderate to severe, flow-limiting spasm of the cerebral arteries (  arrows ). (  E ) To prevent stroke, endovascular treatment including balloon angioplasty (  arrow ) followed by vasodilator infusion therapy with 5 mg verapamil was performed. (  F ) Cerebral arteriography after treatment shows both improved vessel dimension and flow in the cerebral vasculature. Images and case history are provided by Phillip M. Meyers, M.D. 

Close modal

Fig. 13. Effects of adenosine on human cerebral blood flow (CBF). Adenosine is rapidly metabolized by the vascular endothelium and the erythrocytes with an elimination half-life of 1–10 s. An intracarotid bolus of 1–2 mg adenosine results in significant change in shape of the 133xenon washout curve. Compare the washouts during injection of adenosine, nicardipine, and saline (  A ). The effects of adenosine bolus seem to last for less than 30 s. The conventional CBF measurements (standard initial slope analysis [standard ISI]) that are measured between 20 and 80 s after 133xenon injection does not show an increase in CBF (  solid circles , B and  C ) with intracarotid adenosine. However, early initial slope analysis (early ISI) that is measured between 5 and 25 s after 133xenon (  solid squares , B and  C ) seem to have a dramatic effect on CBF. In case of nicardipine, a longer-acting drug, the early ISI and standard ISI yield similar results. Effects of intracarotid adenosine on CBF are exceedingly transient, whereas nicardipine has a sustained effect. From Joshi  et al. 58; used with permission. 

Fig. 13. Effects of adenosine on human cerebral blood flow (CBF). Adenosine is rapidly metabolized by the vascular endothelium and the erythrocytes with an elimination half-life of 1–10 s. An intracarotid bolus of 1–2 mg adenosine results in significant change in shape of the 133xenon washout curve. Compare the washouts during injection of adenosine, nicardipine, and saline (  A ). The effects of adenosine bolus seem to last for less than 30 s. The conventional CBF measurements (standard initial slope analysis [standard ISI]) that are measured between 20 and 80 s after 133xenon injection does not show an increase in CBF (  solid circles , B and  C ) with intracarotid adenosine. However, early initial slope analysis (early ISI) that is measured between 5 and 25 s after 133xenon (  solid squares , B and  C ) seem to have a dramatic effect on CBF. In case of nicardipine, a longer-acting drug, the early ISI and standard ISI yield similar results. Effects of intracarotid adenosine on CBF are exceedingly transient, whereas nicardipine has a sustained effect. From Joshi  et al. 58; used with permission. 

Close modal

Table 1shows the published outcomes of intracarotid vasodilator therapy. Clinically, approximately 30–60% patients seem to benefit from intracarotid vasodilators. Angiographic improvement, however, is seen much more frequently. Although there is a suggestion that intracarotid vasodilators may augment CBF, improve cerebral oxygenation, and reverse metabolic acidosis, the effect of vasodilator therapy seems to be transient and resolves completely over the next 24 h. By measuring the time it takes for radiocontrast to transit through the cerebral arteries and the capillary bed, one can estimate the differential effects of intracarotid vasodilators on the proximal and distal cerebral circulation. In the case of papaverine, the resistance of both the proximal and the distal cerebral arteries decreases during drug infusion.35,128 

Table 1. Selected Reports of Intracarotid Vasodilators for the Treatment of Human Cerebral Vasospasm 

Table 1. Selected Reports of Intracarotid Vasodilators for the Treatment of Human Cerebral Vasospasm 
Table 1. Selected Reports of Intracarotid Vasodilators for the Treatment of Human Cerebral Vasospasm 

Intracarotid vasodilator therapy carries with it two general risks: cerebral steal and increase in ICP. However, neurologic complications can also result from specific drugs, e.g. , neurotoxicity with papaverine.129 

Cerebral Steal.

Cerebral steal has been invoked to explain the decrease in regional blood flow in proximity to focal cerebral pathology after the administration of intravenous or intracarotid papaverine.130,131Any decrease in hemispheric cerebrovascular resistance, whether pathologic or iatrogenic, can cause a redistribution of blood flow away from those ischemic areas where there is already maximal vasodilation. In recent years, the clinical significance of steal has been challenged as with regard to large arteries.132There are, however, examples of microcirculatory steal during hypercapnia or during administration of volatile anesthetic agents, such as halothane, in the setting of focal cerebral ischemia.133,134Unless vasodilator therapy is restricted to the arteries afflicted by cerebral vasospasm, intracarotid vasodilator therapy carries the theoretical risk of cerebral steal, due to vasodilation in the normal vascular beds.

Increase in Intracranial Pressure.

Increase in ICP has been reported with intracarotid infusion of papaverine. Controversy surrounds the effect of intracarotid vasodilators on ICP. Experiments in dogs and monkeys that do not have cerebral vasospasm suggest that intracarotid vasodilators such as sodium nitroprusside increase ICP but may not increase CBF, implying that nitroprusside affects cerebral capacitance arterioles to increase cerebral blood volume with no effect on the resistance arterioles.135On the other hand, intracarotid infusion of adenosine has a relatively benign effect on ICP but augments CBF, suggesting that adenosine primarily affects the resistance arterioles.136It remains to be seen whether intracarotid therapy could selectively target proximal or distal cerebral arteries to minimize the risk of increased ICP.

Cross et al.  137measured changes in ICP during intracarotid infusion of papaverine in 28 patients. The increase in ICP ranged from 0 to 60 mmHg. A baseline ICP of 15 mm or greater was associated with a greater risk of increasing ICP. However, Hunt and Hess scores, Fisher grades, age, and Glasgow Coma Scale scores on admission and immediately before treatment did not correlate with ICP increases. ICP increases result in a decrease in cerebral perfusion pressure. Therefore, hypertensive-hypervolemic interventions need to be continued during intracarotid vasodilator infusions. ICP should be monitored during intracarotid vasodilator therapy whenever feasible.

Intracarotid Papaverine.

Papaverine is perhaps the most potent cerebral vasodilator. Papaverine acts through several intracellular pathways, which include cyclic guanosine monophosphate, cyclic adenosine monophosphate, phosphodiesterase inhibition, Ca2+channel blockade, and histamine release. Table 1summarizes the efficacy of papaverine and other vasodilators in the settings of cerebral vasospasm. Intracarotid papaverine can relieve angiographic narrowing, augment CBF, and mitigate neurologic symptoms in patients with cerebral vasospasm resistant to medical treatment. Typically, during the treatment of cerebral vasospasm, a dose of 300 mg papaverine is infused over 1 h.138Unfortunately, intracarotid papaverine infusion also results in significant neurologic complications,139attributable to several factors: cerebral steal, increased ICP,137,140microembolization of papaverine crystals,141,142neurotoxicity of the preservative (chlorobutanol),142proconvulsive properties of the drug,143and paradoxical vasoconstriction.141 

Cerebral steal, i.e. , redistribution of blood flow away from the ischemic region, has often been implicated in the etiology of neurologic symptoms after intravenous or intracarotid administration of papaverine.130,144There is evidence that intravenous papaverine increases blood flow in ischemic regions of patients with cerebrovascular insufficiency.145However, experiments in cats with middle cerebral artery occlusion suggest that steal does not always occur with cerebral vasodilation. Intravenous papaverine in the feline middle cerebral artery occlusion model did not decrease regional CBF to ischemic areas as long as the blood pressure is maintained.146 

Intracarotid Calcium Channel Blockers.

Intracarotid calcium channel blockers have been used to treat cerebral vasospasm for more than a decade; however, their intracarotid administration is still not considered to be the mainstay for the treatment of cerebral vasospasm. Intracarotid verapamil, nicardipine, and nimodipine have been used for the treatment of vasospasm (table 1). There are no human studies as yet that compare the relative potencies of calcium channel blockers against papaverine. However, studies in similar groups of patients who do not have cerebral vasospasm suggest that intracarotid verapamil, compared with papaverine, seems to be less efficacious in augmenting CBF. Verapamil in small doses (3 mg) results in angiographic improvement without systemic hypotension, although only 5 of 29 patients in this retrospective study improved clinically.147The modest efficacy of the calcium channel blocker might still be advantageous because it is likely to decrease the risk of cerebral steal or increasing ICP. Experimental vasospasm triggered by topical application of endothelin 1 seems to be more responsive to intracarotid nicardipine than verapamil.148Intracarotid nicardipine is effective in reversing cerebral vasospasm, but no study has as yet compared the potency of the various intracarotid calcium channel blockers.149 

Intracarotid Prostaglandins.

Recently, intracarotid infusions of liposomes with papaverine and prostaglandin E2 have been used for the treatment of cerebral vasospasm. Two thirds of the patients demonstrated an increase in CBF after such a treatment.150 

To enhance the clinical efficacy of intracarotid vasodilators and decrease the risks of complications, a number of adjuvant strategies have been suggested. First, transluminal angioplasty has been used to treat cerebral vasospasm when it affects the ICA or the proximal segment of the middle cerebral artery. Angioplasty, compared with intracarotid drugs, results in a more profound and persistent increase in blood flow but it can only be used in the proximal arteries. Drugs such as papaverine are therefore useful in treating distal spasm and could be of benefit even after angioplasty.128Hypothermia has also been used to provide brain protection and increase the time window for intracarotid vasodilator therapy.151Techniques of selective brain cooling either by the infusion of cold saline or by endovascular cooling devices are currently being developed to minimize injury after vasospasm and ischemic stroke.152–154Finally, cocktails of vasodilators have been used so as to increase both the potency and duration of effects of intracarotid vasodilators. Therefore, papaverine has been used in conjunction with nimodipine, nicardipine, prostaglandins to enhance its safety and efficacy.150 

The key in the anesthetic management of patients for intracarotid vasodilator treatment is to recognize the potential for exacerbating neurologic injury. These patients may present either with untreated ruptured aneurysms or after clipping of the lesion. At many institutions, triple-H therapy is instituted before intracarotid vasodilator therapy. Patients with subarachnoid hemorrhage may require inotropic support because of impaired myocardial functions secondary to hemorrhage. Ideally, management of these cases requires monitoring of the ICP so as to treat any increase by mechanical drainage, hyperventilation, or supplemental intravenous anesthesia. Furthermore, because of recirculation of intracarotid vasodilators, there may be significant systemic hypotension. Therefore, anesthesiologists should be prepared to induce hypertension to ensure adequate perfusion pressure.

Cancer Chemotherapy

The major driving force for the development of intracarotid therapies in the 1980s was the treatment of brain neoplasms. However, it soon became evident that the BBB prevented effective transfer of chemotherapeutic agents.2Therefore, attempts to disrupt the BBB began fairly early on.77,94Currently, intracarotid chemotherapy is used for the treatment of primary central nervous system lymphoma, primitive neuroectodermal tumor, germ cell tumor, cancer metastasis to the brain, and low- or high-grade glioma (fig. 14).111The initial response to intracarotid chemotherapy is impressive, with as many as 70–81% tumors regressing, occasionally with a significant prolongation of survival.155,156However, most studies have been anecdotal or uncontrolled case series. The major concern of intracarotid therapy is unexplained local toxicity, likely attributed to unusually high concentrations of the drug due to streaming. One strategy that has minimized streaming is using pulsed intracarotid infusion, which delivers the drug in the diastolic phase of cardiac cycle to increase mixing.106,157 

Fig. 14. Response of a lymphoid tumor to intracarotid chemotherapy. A 62-yr-old right-handed woman presented with three symptomatic white chorioretinal lesions in the right eye. A vitreal biopsy showed small lymphoid cells. Six months later, the patient started experiencing problems while walking. Magnetic resonance imaging revealed contrast-enhancing lesion in the brain (  A   D ). A stereotactic brain biopsy showed large B-cell lymphoma, CD20 positive. Cerebrospinal fluid contained 20 atypical cells/ml. The patient underwent first treatment with blood–brain barrier disruption with intracarotid mannitol and intraarterial methotrexate, and carboplatin that was supplemented with intravenous rituximab. She was admitted every 4 weeks for 2 consecutive days of the same treatment with barrier opening and intraarterial chemotherapy in one of the major brain-feeding arteries, the left or the right internal carotid or the vertebral artery. She was also treated with intraocular rituximab and methotrexate. After eight courses of treatment, there was radiologic evidence of complete tumor regression (  E   H ) without any neurologic symptoms or evidence of intraocular disease. Images and case history are courtesy of Edward A. Neuwelt, M.D. (Professor of Neurology and Neurosurgery, Oregon Health and Science University, Portland, Oregon). 

Fig. 14. Response of a lymphoid tumor to intracarotid chemotherapy. A 62-yr-old right-handed woman presented with three symptomatic white chorioretinal lesions in the right eye. A vitreal biopsy showed small lymphoid cells. Six months later, the patient started experiencing problems while walking. Magnetic resonance imaging revealed contrast-enhancing lesion in the brain (  A   D ). A stereotactic brain biopsy showed large B-cell lymphoma, CD20 positive. Cerebrospinal fluid contained 20 atypical cells/ml. The patient underwent first treatment with blood–brain barrier disruption with intracarotid mannitol and intraarterial methotrexate, and carboplatin that was supplemented with intravenous rituximab. She was admitted every 4 weeks for 2 consecutive days of the same treatment with barrier opening and intraarterial chemotherapy in one of the major brain-feeding arteries, the left or the right internal carotid or the vertebral artery. She was also treated with intraocular rituximab and methotrexate. After eight courses of treatment, there was radiologic evidence of complete tumor regression (  E   H ) without any neurologic symptoms or evidence of intraocular disease. Images and case history are courtesy of Edward A. Neuwelt, M.D. (Professor of Neurology and Neurosurgery, Oregon Health and Science University, Portland, Oregon). 

Close modal

The role of regional blood flow in enhancing intracarotid delivery of chemotherapeutic drugs is ill understood at this time. Some groups have suggested an increase in drug doses in proportion to the regional blood flow to safely achieve higher total doses and better tumor regressions.14,156At the same time, it has recently been shown that transient decrease in CBF could significantly increase delivery of anticancer drugs to the brain in a rabbit model.15Therefore, the exact role for the manipulation of CBF in enhancing cancer drug delivery remains to be fully understood at this time. The fundamental problem confronting investigators is the inability to rapidly measure tissue concentrations of chemotherapeutic drugs. Insights into intracarotid delivery of some anticancer drugs could be generated by optically tracking their concentrations in the brain tissue by novel spectroscopic techniques.65,74,75 

The disruption of the BBB with intracarotid mannitol that is often needed to improve regional delivery of anticancer drugs also carries significant clinical risks.158Because of pain associated with the procedure, the disruption is undertaken during general anesthesia. In a recent report involving 17 patients and 210 treatment cycles, focal seizures occurred in 9 patients and in 10% of the treatment cycles. In some cases, these seizures were generalized (2 patients and 3% of treatment cycles). Chances of seizure were higher with intracarotid than with vertebral artery injections. Impaired consciousness was seen in 7.6% of patients with recovery in 24 h. One patient in this series had an intractable increase in ICP necessitating decompressive hemispherectomy a day after treatment. Attempts to augment CBF to enhance drug delivery with intravenous atropine probably contributed to ST-segment changes seen in 4.3% of the cases, requiring β-blockade. Other complications included transient neurologic deficits (6%), postoperative nausea and vomiting despite 20 mg odansetron (11.9%), and headache (4%). Management of these patients requires a well-thought-out plan to include close monitoring of the patient’s neurologic and hemodynamic status and to address significant complications that arise in the postoperative periods.159Patients with brain tumors may require preoperative embolization with n-butyl-cyanoacrylate to decrease blood loss during surgery (fig. 15). These procedures are relatively painless and can usually be performed with minimal sedation.

Fig. 15. Intracarotid injection of embolic materials to facilitate surgery of brain tumors. A 39-yr-old man with neurofibromatosis type II, a condition that often results in early development of multiple brain tumors, presented with hearing loss and seizures. In addition to small bilateral tumors of the vestibular nerves (not shown), the patient had developed a large extraaxial mass most compatible with a meningioma, a benign tumor that arises from the dura matter or brain covering (  A , white arrow ). Meningiomas are often highly vascular tumors. Before surgical removal of the tumor, the patient underwent cerebral angiography that demonstrated that blood supply to the tumor arose from branches of the middle meningeal artery (  B , white arrow ). With the patient awake and in a painless manner, a tiny microcatheter was advanced into the branches of the middle meningeal artery supplying the tumor (  C , black arrow ), and the tumor was embolized. Arteriography after embolization demonstrates that there is no longer any blood flowing to the tumor (  D , white circle ), indicating that the tumor could now be surgically resected with minimal blood loss. Images and case history are provided by Phillip M. Meyers, M.D. 

Fig. 15. Intracarotid injection of embolic materials to facilitate surgery of brain tumors. A 39-yr-old man with neurofibromatosis type II, a condition that often results in early development of multiple brain tumors, presented with hearing loss and seizures. In addition to small bilateral tumors of the vestibular nerves (not shown), the patient had developed a large extraaxial mass most compatible with a meningioma, a benign tumor that arises from the dura matter or brain covering (  A , white arrow ). Meningiomas are often highly vascular tumors. Before surgical removal of the tumor, the patient underwent cerebral angiography that demonstrated that blood supply to the tumor arose from branches of the middle meningeal artery (  B , white arrow ). With the patient awake and in a painless manner, a tiny microcatheter was advanced into the branches of the middle meningeal artery supplying the tumor (  C , black arrow ), and the tumor was embolized. Arteriography after embolization demonstrates that there is no longer any blood flowing to the tumor (  D , white circle ), indicating that the tumor could now be surgically resected with minimal blood loss. Images and case history are provided by Phillip M. Meyers, M.D. 

Close modal

Intracarotid Thrombolysis for Stroke

The object of intracarotid thrombolysis is to deliver high concentrations of thrombolytic drugs locally into the brain.160Intravenous thrombolysis can achieve a significant degree of recanalization within 3 h of onset of neurologic symptoms.160,161However, it is often difficult to provide thrombolytic therapy within such a narrow time window. Intracarotid delivery of thrombolytic drugs can extend the intervention time window to 6 h.162Intracarotid urokinase, streptokinase, and more recently recombinant tissue plasminogen activator have been used for the purpose (table 2).163Development of endovascular clot retrieving devices, laser or ultrasonic clot lysis techniques, and placement of endovascular stents can supplement or provide alternatives to intracarotid thrombolytic therapy.164Intracarotid thrombolysis is superior to intravenous thrombolysis in so far as restoring tissue perfusion, although as yet there is no clear-cut evidence of improved neurologic outcome with intracarotid thrombolysis in controlled trials (fig. 16).165,166Intracarotid thrombolysis today provides the most compelling reason to investigate intracarotid drug delivery to the brain. The inability to translate the results of preclinical studies with pharmacologic treatments of ischemic stroke into effective therapies compels us to develop better methods of drug delivery.167–169Intracarotid thrombolysis provides a unique opportunity to deliver drugs at the very site of reperfusion injury, and the catheter to do so is already in situ .

Table 2. Selected Original Investigations Describing Use of Intracarotid Thrombolytics for Ischemic Stroke 

Table 2. Selected Original Investigations Describing Use of Intracarotid Thrombolytics for Ischemic Stroke 
Table 2. Selected Original Investigations Describing Use of Intracarotid Thrombolytics for Ischemic Stroke 

Fig. 16. Intracarotid thrombolysis in acute stroke. A 62-yr-old woman with multiple cardiovascular risk factors presented with quadriplegia followed by loss of consciousness. (  A ) Left vertebral arteriography demonstrates distal occlusion of the basilar artery (  arrow ). (  B ) On high-magnification angiography, the filling defect resulting from the occlusive thromboembolism is evident (  arrow ). (  C ) Microcatheter arteriography in the lateral projection further delineates the site of occlusion before thrombolysis with the plasminogen activator, urokinase. (  D ) After lysis of the occlusive thrombus, complete restoration of flow in the vertebrobasilar system has been restored. Images and case history are provided by Phillip M. Meyers, M.D. 

Fig. 16. Intracarotid thrombolysis in acute stroke. A 62-yr-old woman with multiple cardiovascular risk factors presented with quadriplegia followed by loss of consciousness. (  A ) Left vertebral arteriography demonstrates distal occlusion of the basilar artery (  arrow ). (  B ) On high-magnification angiography, the filling defect resulting from the occlusive thromboembolism is evident (  arrow ). (  C ) Microcatheter arteriography in the lateral projection further delineates the site of occlusion before thrombolysis with the plasminogen activator, urokinase. (  D ) After lysis of the occlusive thrombus, complete restoration of flow in the vertebrobasilar system has been restored. Images and case history are provided by Phillip M. Meyers, M.D. 

Close modal

Because the urgency for interventions, detailed anesthetic assessment may not be feasible in stroke setting. Centers offering intracarotid thrombolysis should have an intervention suite ready to provide immediate anesthesia and should have well-established lines of communication between the stroke team and the anesthesiologists. A rapid review of medical history, last meal time, drug treatments, and allergies, particularly immediate anticoagulant treatment, has to be undertaken. Airway interventions could be challenging with background anticoagulation because of the potential for hemorrhage from even minor trauma, such as placement of a nasal airway. Wherever feasible, these interventions should be performed with minimal sedation to permit neurologic examination. However, the condition of the patient could rapidly deteriorate because of the evolution of neurologic symptoms or because of complications of treatment, such as a hemorrhagic transformation of the infarct, necessitating urgent conversion from sedation to general anesthesia. Blood pressure manipulation, particularly induced hypertension, may be required to improve perfusion.29 

Clinicians have frequently used intracarotid drugs to treat life-threatening brain diseases, but the following applications of intracarotid drugs have not been widely recognized.

Intractable Increased ICP

Yokota et al.  170used low-dose bilateral intracarotid infusion of mannitol to treat severely increased ICP in 18 human subjects with head trauma. They observed that intracarotid mannitol significantly decreased ICP and caused no hemodynamic side effects or disturbances in electrolyte status.

Severe Intracranial Infections

Medical treatment of purulent meningitis and encephalitis is sometimes complicated by the inability to deliver sufficient antibiotics to the infected site.171In experimental animals, intracarotid hyperosmolar disruption of BBB has been shown to enhance the delivery of tobramycin and vancomycin to the brain.172Even without hypertonic disruption, intracarotid antibiotics have yielded beneficial results in treating intracranial infections.171,173–175 

Intraarterial Gene Therapy

Perhaps the most potent application of intracarotid drugs in the future is to deliver genes that could alter the course of a disease or help us to understand disease processes. Intracarotid delivery of viral vectors, DNA-bearing liposomes, and stem cells have all been successfully demonstrated.176,177Selective delivery of herpes viral vectors to tumor regions by concurrent use of Cereport (RMP-7) is one such example.178Bone marrow stem cells have been successfully delivered to traumatized regions of the brain after intracarotid injection.179Although the field of gene therapy is in its infancy, intracarotid gene delivery could play a key role in the future treatment of brain diseases due to degeneration, ischemia, trauma, or neoplasia. Therefore, intracarotid drug delivery promises to play a critical role in emerging areas of molecular and restorative neurosurgery.180,181 

During the past decade, compared with the technical advances in endovascular surgery, the field of intracarotid drug therapies has remained relatively ignored. There are ample anecdotal data to suggest that intracarotid drug therapy is effective for cerebral vasospasm, thromboembolic strokes, and neoplasms. However, there have been few attempts to systematically understand the kinetics of intracarotid drugs. The major efforts to deliver drugs selectively to the brain in the past decade have relied on molecular techniques to selectively penetrate the BBB (fig. 17). These advances in brain tissue drug targeting by novel neuropharmaceuticals come at a time of rapid advances in endovascular techniques, material sciences, and brain imaging that compel us to reevaluate intracarotid drug delivery. Intracarotid drug delivery can instantaneously generate exceedingly high local concentrations of the novel neuropharmaceuticals and thereby assist their regional delivery. Intracarotid drug delivery might be the primary route of drug delivery, or it could be used in conjunction with other brain tissue targeting technologies. Therefore, it is time to better understand the kinetics of intracarotid drugs and to develop techniques that could enhance the safety and reliability of such infusions.

Fig. 17. Techniques of selective delivery of drugs to the brain that are currently under development. These techniques generally target unique features of the blood–brain barrier, such as specific molecules or transport mechanisms. Intracarotid delivery can enhance the effectiveness of these techniques by increasing the speed of onset of drug effects and delivering exceedingly high first-pass concentrations. CED = convection enhanced delivery; CSF = cerebrospinal fluid. From Joshi  et al. 11; used with kind permission of Springer Science and Business Media. 

Fig. 17. Techniques of selective delivery of drugs to the brain that are currently under development. These techniques generally target unique features of the blood–brain barrier, such as specific molecules or transport mechanisms. Intracarotid delivery can enhance the effectiveness of these techniques by increasing the speed of onset of drug effects and delivering exceedingly high first-pass concentrations. CED = convection enhanced delivery; CSF = cerebrospinal fluid. From Joshi  et al. 11; used with kind permission of Springer Science and Business Media. 

Close modal

This review is dedicated to our parents, who, despite their hardships, always encouraged us to pursue our dreams. Acknowledgments are due to Mei Wang, M.P.H. (Staff Researcher, Department of Anesthesiology, Columbia University, New York, New York), for her help in preparing this manuscript and the ongoing intracarotid research, and Edward A. Neuwelt, M.D. (Professor of Neurology and Neurosurgery, Oregon Health and Science University, Portland, Oregon), for his help with illustrating this review (fig. 14).

1.
Wada J, Rassmussen T: Intracarotid injection of sodium amytal for the lateralization of cerebral speech dominance: Experimental and clinical observations. J Neurosurg 1960; 17:266–82
2.
Fenstermacher J, Gazendam J: Intra-arterial infusions of drugs and hyperosmotic solutions as ways of enhancing CNS chemotherapy. Cancer Treat Rep 1981; 65:27–37
3.
Dedrick RL, Oldfield EH, Collins JM: Arterial drug infusion with extracorporeal removal, I: Theoretic basis with particular reference to the brain. Cancer Treat Rep 1984; 68:373–80
4.
Neuwelt EA, Pagel M, Barnett P, Glassberg M, Frenkel EP: Pharmacology and toxicity of intracarotid Adriamycin administration following osmotic blood-brain barrier modification. Cancer Res 1981; 41:4466–70
5.
Oldfield EH, Dedrick RL, Chatterji DC, Yeager RL, Girton ME, Kornblith PL, Doppman JL: Arterial drug infusion with extracorporeal removal, II: Internal carotid carmustine in the rhesus monkey. Cancer Treat Rep 1985; 69:293–303
6.
Oldfield EH, Dedrick RL, Yeager RL, Clark WC, DeVroom HL, Chatterji DC, Doppman JL: Reduced systemic drug exposure by combining intra-arterial chemotherapy with hemoperfusion of regional venous drainage. J Neurosurg 1985; 63:726–32
7.
Fortin D, Desjardins A, Benko A, Niyonsega T, Boudrias M: Enhanced chemotherapy delivery by intraarterial infusion and blood-brain barrier disruption in malignant brain tumors: The Sherbrooke experience. Cancer 2005; 103:2606–15
8.
Miyagami M, Tsubokawa T: Chemotherapy with ACNU and radiation therapy in malignant glioma in cerebral hemisphere of adult [in Japanese]. Gan To Kagaku Ryoho 1990; 17(pt 1):1447–53
9.
Perry A, Schmidt RE: Cancer therapy-associated CNS neuropathology: An update and review of the literature. Acta Neuropathol (Berl) 2006; 111:197–212
10.
Dedrick RL: Arterial drug infusion: Pharmacokinetic problems and pitfalls. J Natl Cancer Inst 1988; 80:84–9
11.
Joshi S, Ornstein E, Bruce JN: Targeting the brain: Rationalizing the novel methods of drug delivery to the central nervous system. Neurocrit Care 2007; 6:200–12
12.
Joshi S, Emala CW, Pile-Spellman JCW: Intra-arterial drug delivery: A concise review. J Neurosurg Anesthesiol 2007; 19:111–9
13.
Pile-Spellman J, Young WL: Materials and methods for interventional neuroradiology (INR) (chapter 18.2), Neuroradiology, 3rd edition. Edited by Taveras JM. Baltimore, Williams & Wilkins, 1996, pp 1047–73Taveras JM
Baltimore
,
Williams & Wilkins
14.
Gobin YP, Cloughesy TF, Chow KL, Duckwiler GR, Sayre JW, Milanese K, Vinuela F: Intraarterial chemotherapy for brain tumors by using a spatial dose fractionation algorithm and pulsatile delivery. Radiology 2001; 218:724–32
15.
Joshi S, Wang M, Etu JJ, Suckow RF, Cooper TB, Feinmark SJ, Bruce JN, Fine RL: Transient cerebral hypoperfusion enhances intraarterial carmustine deposition into brain tissue. J Neurooncol 2007; 86:123–32
16.
Joshi S, Wang M, Etu JJ, Nishanian EV, Pile-Spellman J: Cerebral blood flow affects dose requirements of intracarotid propofol for electrocerebral silence. Anesthesiology 2006; 104:290–8
17.
Henk CB, Higgins CB, Saeed M: Endovascular interventional MRI. J Magn Reson Imaging 2005; 22:451–60
18.
Allman JM: The Evolution of Big Brains. Evolving Brains in Scientific American Library Series. New York, W.H. Freeman & Company, 2000, pp 158–208
New York
,
W.H. Freeman & Company
19.
Omojola MF, Fox AJ, Auer RN, Vinuela FV: Hemorrhagic encephalitis produced by selective non-occlusive intracarotid BCNU injection in dogs. J Neurosurg 1982; 57:791–6
20.
Crafts DC, Levin VA, Nielsen S: Intracarotid BCNU (NSC-409962): A toxicity study in six rhesus monkeys. Cancer Treat Rep 1976; 60:541–5
21.
Greenberg HS, Ensminger WD, Seeger JF, Kindt GW, Chandler F, Doan K, Dakhil SR: Intra-arterial BCNU chemotherapy for the treatment of malignant gliomas of the central nervous system: A preliminary report. Cancer Treat Rep 1981; 65:803–10
22.
Edvinsson L, MacKenzie ET: Chapter 1, General and Comparative Anatomy of the Cerebral Circulation, 2nd edition. Philadelphia, Lippincott Williams & Wilkins, 2000, pp 3–29
Philadelphia
,
Lippincott Williams & Wilkins
23.
Scremin OU, Sonnenschein RR, Rubinstein EH: Cerebrovascular anatomy and blood flow measurements in the rabbit. J Cereb Blood Flow Metab 1982; 2:55–66
24.
Lee JS, Hamilton MG, Zabramski JM: Variations in the anatomy of the rabbit cervical carotid artery. Stroke 1994; 25:501–3
25.
Roatta S, Roncari S, Micieli G, Bosone D, Passatore M: Doppler sonography to monitor flow in different cerebral arteries in the rabbit. Exp Physiol 2000; 85:431–8
26.
Visocchi M, Cuevas DC, Tartaglione T, Mastrojanni G, Cioni B: Inconsistent MRI findings in acute pre-Willisian brain ischemia in rabbit: A useful model? J Neurosurg Sci 1999; 43:93–8
27.
Marshall RS, Lazar RM, Mohr JP, Pile-Spellman J, Hacein-Bey L, Duong DH, Joshi S, Chen X, Levin B, Young WL: Higher cerebral function and hemispheral blood flow during awake carotid artery balloon test occlusions. J Neurol Neurosurg Psych 1999; 66:734–8
28.
Marshall RS, Lazar RM, Young WL, Solomon RA, Joshi S, Duong DH, Rundek T, Pile-Spellman JT: Clinical utility of quantitative cerebral blood flow measurements during internal carotid artery test occlusions. Neurosurgery 2002; 50:996–1004
29.
Duong H, Hacein-Bey L, Vang MC, Pile-Spellman J, Joshi S, Young WL: Management of cerebral arterial occlusion during endovascular treatment of cerebrovascular disease. Probl Anesth Controversies Neuroanesth 1997; 9:99–111
30.
Hillen B: The variability of the circle of Willis: Univariate and bivariate analysis. Acta Morphol Neerl Scand 1986; 24:87–101
31.
Hillen B, Hoogstraten HW, Van Overbeeke JJ, Van der Zwan A: Functional anatomy of the circulus arteriosus cerebri (Willis II). Bull Assoc Anat 1991; 75:123–6
32.
Hillen B, Drinkenburg BA, Hoogstraten HW, Post L: Analysis of flow and vascular resistance in a model of the circle of Willis. J Biomechan 1988; 21:807–14
33.
Joshi S, Wang M, Etu JJ, Nishanian EV, Pile-Spellman J: Cerebral blood flow affects dose requirements of intracarotid propofol for electrocerebral silence. Anesthesiology 2006; 104:290–8
34.
Joshi S, Wang M, Etu JJ, Pile-Spellman J: Reducing cerebral blood flow increases the duration of electroencephalographic silence by intracarotid thiopental. Anesth Analg 2005; 101:851–8
35.
Milburn JM, Moran CJ, Cross DT III, Diringer MN, Pilgram TK, Dacey RG Jr: Effect of intraarterial papaverine on cerebral circulation time. AJNR Am J Neuroradiol 1997; 18:1081–5
36.
Schlosshauer B: The blood-brain barrier: Morphology, molecules, and neurothelin. Bioessays 1993; 15:341–6
37.
Raub TJ, Kuentzel SL, Sawada GA: Permeability of bovine brain microvessel endothelial cells in vitro : Barrier tightening by a factor released from astroglioma cells. Exp Cell Res 1992; 199:330–40
38.
Takano S, Yoshii Y, Nose T: Ultrastructure of glioma vessel: Morphometric study for vascular permeability [in Japanese]. No To Shinkei 1991; 43:49–56
39.
Stewart PA, Hayakawa K, Farrell CL: Quantitation of blood-brain barrier ultrastructure. Microsc Res Tech 1994; 27:516–27
40.
Claudio L: Ultrastructural features of the blood-brain barrier in biopsy tissue from Alzheimer’s disease patients. Acta Neuropathol 1996; 91:6–14
41.
Unger RE, Oltrogge JB, von Briesen H, Engelhardt B, Woelki U, Schlote W, Lorenz R, Bratzke H, Kirkpatrick CJ: Isolation and molecular characterization of brain microvascular endothelial cells from human brain tumors. In Vitro Cell Dev Biol Anim 2002; 38:273–81
42.
Heistad DD, Kontos HA: Cerebral circulation (chapter 5), Handbook of Physiology, Section 2: The Cardiovascular System. Edited by Shepherd JT, Abboud FM. Bethesda, Maryland, American Physiological Society, 1983, pp 137–82Shepherd JT, Abboud FM
Bethesda, Maryland
,
American Physiological Society
43.
Pardridge WM, Yoshikawa T, Kang Y-S, Miller LP: Blood-brain barrier transport and brain metabolism of adenosine and adenosine analogs. J Pharmacol Exp Ther 1994; 268:14–8
44.
Jolliet-Riant P, Tillement JP: Drug transfer across the blood-brain barrier and improvement of brain delivery. Fundam Clin Pharmacol 1999; 13:16–26
45.
Goldstein GW, Betz AL: The blood-brain barrier. Sci Am 1986; 255:74–83
46.
Wang M, Joshi S, Emerson RG: Comparison of intracarotid and intravenous propofol for electrocerebral silence in rabbits. Anesthesiology 2003; 99:904–10
47.
Jones DR, Hall SD, Jackson EK, Branch RA, Wilkinson GR: Brain uptake of benzodiazepines: Effects of lipophilicity and plasma protein binding. J Pharmacol Exp Ther 1988; 245:816–22
48.
Pardridge WM, Landaw EM: Tracer kinetic model of blood-brain barrier transport of plasma protein-bound ligands: Empiric testing of the free hormone hypothesis. J Clin Invest 1984; 74:745–52
49.
Pardridge WM, Sakiyama R, Fierer G: Blood-brain barrier transport and brain sequestration of propranolol and lidocaine. Am J Physiol 1984; 247:R582–8
50.
Urien S, Albengres E, Pinquier JL, Tillement JP: Role of alpha-1 acid glycoprotein, albumin, and nonesterified fatty acids in serum binding of apazone and warfarin. Clin Pharmacol Ther 1986; 39:683–9
51.
Riant P, Bree F, Urien S, Hamberger C, Albengres E, Tillement JP: How chronically administered valproate increases chlordiazepoxide transfer through the blood-brain barrier. Fundam Clin Pharmacol 1990; 4:105–14
52.
Jolliet P, Simon N, Bree F, Urien S, Pagliara A, Carrupt PA, Testa B, Tillement JP: Blood-to-brain transfer of various oxicams: Effects of plasma binding on their brain delivery. Pharm Res 1997; 14:650–6
53.
Obrist WD, Thompson HK Jr, Wang HS, Wilkinson WE: Regional cerebral blood flow estimated by 133xenon inhalation. Stroke 1975; 6:245–56
54.
Obrist WD, Dolinskas CA, Jaggi JL, Cruz J, Steiman DL: Serial cerebral blood flow studies in acute head injury: Application of the intravenous 133 Xe method. In: Magistretti PL, ed., Functional Radionuclide Imaging of the Brain. Sereno Symposia Publication, vol 5, New York, Raven Press, 1983:145–50Serial cerebral blood flow studies in acute head injury: Application of the intravenous 133 Xe method.Magistretti PL
New York
,
Raven Press
55.
Sanelli PC, Nicola G, Tsiouris AJ, Ougorets I, Knight C, Frommer B, Veronelli S, Zimmerman RD: Reproducibility of post processing of quantitative CT perfusion maps. AJR Am J Roentgenol 2007; 188:213–8
56.
Wang M, Joshi S: Electrocerebral silence after intracarotid propofol injection is a function of transit time. Anesth Analg 2007; 104:1498–503
57.
Yamashita J, Handa H, Tokuriki Y, Ha YS, Otsuka SI, Suda K, Taki W: Intra-arterial ACNU therapy for malignant brain tumors: Experimental studies and preliminary clinical results. J Neurosurg 1983; 59:424–30
58.
Joshi S, Young WL, Pile-Spellman J, Duong DH, Vang MC, Hacein-Bey L, Lee HT, Ostapkovich N: The feasibility of intracarotid adenosine for the manipulation of human cerebrovascular resistance. Anesth Analg 1998; 87:1291–8
59.
Joshi S, Wang M, Etu JJ, Pile-Spellman J: Bolus configuration affects dose requirements of intracarotid propofol for electroencephalographic silence. Anesth Analg 2006; 102:1816–22
60.
Khatri P, Neff J, Broderick JP, Khoury JC, Carrozzella J, Tomsick T: Revascularization end points in stroke interventional trials: Recanalization versus  reperfusion in IMS-I. Stroke 2005; 36:2400–3
61.
Kidwell CS, Saver JL, Starkman S, Duckwiler G, Jahan R, Vespa P, Villablanca JP, Liebeskind DS, Gobin YP, Vinuela F, Alger JR: Late secondary ischemic injury in patients receiving intraarterial thrombolysis. Ann Neurol 2002; 52:698–703
62.
Murkin JM: Inflammatory responses and CNS injury: Implications, prophylaxis, and treatment. Heart Surg Forum 2003; 6:193–5
63.
Joshi S, Ornstein E, Young WL: Chapter 2, Cerebral and Spinal Cord Blood Flow, 4th edition. St. Louis, Mosby, 2002, pp 19–68
St. Louis
,
Mosby
64.
Donelli MG, Zucchetti M, D’Incalci M: Do anticancer agents reach the tumor target in the human brain? Cancer Chemother Pharmacol 1992; 30:251–60
65.
Mourant JR, Johnson TM, Los G, Bigio IJ: Non-invasive measurement of chemotherapy drug concentrations in tissue: Preliminary demonstrations of in vivo  measurements. Phys Med Biol 1999; 44:1397–417
66.
Hassenbusch SJ, Anderson JH, Colvin OM: Predicted and actual BCNU concentrations in normal rabbit brain during intraarterial and intravenous infusions. J Neurooncol 1996; 30:7–18
67.
Mitsuki S, Diksic M, Conway T, Yamamoto YL, Villemure JG, Feindel W: Pharmacokinetics of 11C-labelled BCNU and SarCNU in gliomas studied by PET. J Neurooncol 1991; 10:47–55
68.
Tyler JL, Yamamoto YL, Diksic M, Theron J, Villemure JG, Worthington C, Evans AC, Feindel W: Pharmacokinetics of superselective intra-arterial and intravenous [11C]BCNU evaluated by PET. J Nucl Med 1986; 27:775–80
69.
Frayne R, Omary RA, Unal O, Strother CM: Determination of optimal injection parameters for intraarterial gadolinium-enhanced MR angiography. J Vasc Interv Radiol 2000; 11:1277–84
70.
Hwang KP, Green JD, Li D, Simonetti OP, Resnick SA, Finn JP, Duerk JL, Omary RA: Minimizing contrast agent dose during intraarterial gadolinium-enhanced MR angiography: in vitro  assessment. J Magn Reson Imaging 2002; 15:55–61
71.
Joshi S, Wang M, Etu J, Nishanian EV: Comparison of intracarotid anesthetics for EEG silence. J Neurosurg Anesthesiol 2006; 18:112–8
72.
Humpel C, Ebendal T, Olson L: Microdialysis: A way to study in vivo  release of neurotrophic bioactivity: A critical summary. J Mol Med 1996; 74:523–6
73.
Layton ME, Wagner JK, Samson FE, Pazdernik TL: Redox changes in perfusates following intracerebral penetration of microdialysis probes. Neurochem Res 1997; 22:735–41
74.
Bigio IJ, Bown SG: Spectroscopic sensing of cancer and cancer therapy: Current status of translational research. Cancer Biol Ther 2004; 3:259–67
75.
Reif R, Wang M, Joshi S, A’Amar O, Bigio IJ: Optical method for real-time monitoring of drug concentrations facilitates the development of novel methods for drug delivery to brain tissue. J Biomed Opt 2007; 12:034036
76.
Kroll RA, Neuwelt EA: Outwitting the blood-brain barrier for therapeutic purposes: Osmotic opening and other means. Neurosurgery 1998; 42:1083–100
77.
Rapoport SI: Osmotic opening of the blood-brain barrier: Principles, mechanism, and therapeutic applications. Cell Mol Neurobiol 2000; 20:217–30
78.
Neuwelt EA, Minna J, Frenkel E, Barnett PA, McCormick CI: Osmotic blood-brain barrier opening to IgM monoclonal antibody in the rat. Am J Physiol 1986; 250:R875–83
79.
Nagashima T, Ikeda K, Wu S, Kondo T, Yamaguchi M, Tamaki N: The mechanism of reversible osmotic opening of the blood-brain barrier: Role of intracellular calcium ion in capillary endothelial cells. Acta Neurochir Suppl (Wien) 1997; 70:231–3
80.
Siegal T, Rubinstein R, Bokstein F, Schwartz A, Lossos A, Shalom E, Chisin R, Gomori JM: In vivo  assessment of the window of barrier opening after osmotic blood-brain barrier disruption in humans. J Neurosurg 2000; 92:599–605
81.
Zunkeler B, Carson RE, Olson J, Blasberg RG, DeVroom H, Lutz RJ, Saris SC, Wright DC, Kammerer W, Patronas NJ, Dedrick RL, Herscovitch P, Oldfield EH: Quantification and pharmacokinetics of blood-brain barrier disruption in humans. J Neurosurg 1996; 85:1056–65
82.
Sato S, Suga S, Yunoki K, Mihara B: Effect of barrier opening on brain edema in human brain tumors. Acta Neurochir Suppl (Wien) 1994; 60:116–8
83.
Wang M, Etu J, Joshi S: Enhanced disruption of the blood brain barrier by intracarotid mannitol injection during transient cerebral hypoperfusion in rabbits. J Neurosurg Anesthesiol 2007; 19:249–56
84.
Tamaki K, Sadoshima S, Heistad DD: Increased susceptibility to osmotic disruption of the blood-brain barrier in chronic hypertension. Hypertension 1984; 6:633–8
85.
Ikeda M, Bhattacharjee AK, Kondoh T, Nagashima T, Tamaki N: Synergistic effect of cold mannitol and Na(+)/Ca(2+) exchange blocker on blood-brain barrier opening. Biochem Biophys Res Commun 2002; 291:669–74
86.
Bhattacharjee AK, Nagashima T, Kondoh T, Tamaki N: The effects of the Na(+)/Ca(++) exchange blocker on osmotic blood-brain barrier disruption. Brain Res 2001; 900:157–62
87.
Chi OZ, Liu X, Weiss HR: Effects of mild hypothermia on blood-brain barrier disruption during isoflurane or pentobarbital anesthesia. Anesthesiology 2001; 95:933–8
88.
Kaya M, Gulturk S, Elmas I, Kalayci R, Arican N, Kocyildiz ZC, Kucuk M, Yorulmaz H, Sivas A: The effects of magnesium sulfate on blood-brain barrier disruption caused by intracarotid injection of hyperosmolar mannitol in rats. Life Sci 2004; 76:201–12
89.
Liu R, Wen Y, Perez E, Wang X, Day AL, Simpkins JW, Yang SH: 17β-Estradiol attenuates blood-brain barrier disruption induced by cerebral ischemia-reperfusion injury in female rats. Brain Res 2005; 1060:55–61
90.
Neuwelt EA, Barnett PA, Ramsey FL, Hellstrom I, Hellstrom KE, McCormick CI: Dexamethasone decreases the delivery of tumor-specific monoclonal antibody to both intracerebral and subcutaneous tumor xenografts. Neurosurgery 1993; 33:478–84
91.
Kozler P, Pokorny J: Altered blood-brain barrier permeability and its effect on the distribution of Evans blue and sodium fluorescein in the rat brain applied by intracarotid injection. Physiol Res 2003; 52:607–14
92.
Kozler P, Pokorny J: Effects of intracarotid injection of methylprednisolone on cellular oedema after osmotic opening of the blood-brain barrier in rats. Prague Med Rep 2004; 105:279–90
93.
Chi OZ, Chun TW, Liu X, Weiss HR: The effects of pentobarbital on blood-brain barrier disruption caused by intracarotid injection of hyperosmolar mannitol in rats. Anesth Analg 1998; 86:1230–5
94.
Black KL, Cloughesy T, Huang SC, Gobin YP, Zhou Y, Grous J, Nelson G, Farahani K, Hoh CK, Phelps M: Intracarotid infusion of RMP-7, a bradykinin analog, and transport of gallium-68 ethylenediamine tetraacetic acid into human gliomas. J Neurosurg 1997; 86:603–9
95.
Chio CC, Baba T, Black KL: Selective blood-tumor barrier disruption by leukotrienes. J Neurosurg 1992; 77:407–10
96.
Joshi S, Meyers P, Pile-Spellman J, Wang M, Sahlein D: Intracarotid verapamil decreases both proximal and distal human cerebrovascular resistance. Anesthesiology 2004; 100:774–81
97.
Agid R, Rubinstein R, Siegal T, Lester H, Bokstein F, Chisin R, Gomori JM: Does streaming affect the cerebral distribution of infraophthalmic intracarotid chemotherapy? AJNR Am J Neuroradiol 2002; 23:1732–5
98.
Saris SC, Blasberg RG, Carson RE, deVroom HL, Lutz R, Dedrick RL, Pettigrew K, Chang R, Doppman J, Wright DC: Intravascular streaming during carotid artery infusions: Demonstration in humans and reduction using diastole-phased pulsatile administration. J Neurosurg 1991; 74:763–72
99.
Oldfield EH, Clark WC, Dedrick RL, Egorin MJ, Austin HA, DeVroom HD, Joyce KM, Doppman JL: Reduced systemic drug exposure by combining intraarterial cis-diamminedichloroplatinum(II) with hemodialysis of regional venous drainage. Cancer Res 1987; 47:1962–7
100.
Qureshi AI, Luft AR, Sharma M, Guterman LR, Hopkins LN: Prevention and treatment of thromboembolic and ischemic complications associated with endovascular procedures: Part II—Clinical aspects and recommendations. Neurosurgery 2000; 46:1360–75
101.
Qureshi AI, Luft AR, Sharma M, Guterman LR, Hopkins LN: Prevention and treatment of thromboembolic and ischemic complications associated with endovascular procedures: Part I—Pathophysiological and pharmacological features. Neurosurgery 2000; 46:1344–59
102.
Young WL, Pile-Spellman J: Anesthetic considerations for interventional neuroradiology. Anesthesiology 1994; 80:427–56
103.
Zlotnik EI, Oleshkevich FV: Prolonged catheterization of the superficial temporal artery for repeated angiography [in Russian]. Vopr Neirokhir 1975; May–June:43–5
104.
Kallmes DF, McGraw JK, Evans AJ, Mathis JM, Hergenrother RW, Jensen ME, Cloft HJ, Lopes M, Dion JE: Thrombogenicity of hydrophilic and nonhydrophilic microcatheters and guiding catheters. AJNR Am J Neuroradiol 1997; 18:1243–51
105.
Markus H, Loh A, Israel D, Buckenham T, Clifton A, Brown MM: Microscopic air embolism during cerebral angiography and strategies for its avoidance. Lancet 1993; 341:784–7
106.
Saris SC, Wright DC, Oldfield EH, Blasberg RG: Intravascular streaming and variable delivery to brain following carotid artery infusions in the Sprague-Dawley rat. J Cereb Blood Flow Metab 1988; 8:116–20
107.
Lutz RJ, Dedrick RL, Boretos JW, Oldfield EH, Blacklock JB, Doppman JL: Mixing studies during intracarotid artery infusions in an in vitro  model. J Neurosurg 1986; 64:277–83
108.
Tonn JC, Roosen K, Schachenmayr W: Brain necroses after intraarterial chemotherapy and irradiation of malignant gliomas: A complication of both ACNU and BCNU? J Neurooncology 1991; 11:241–2
109.
Di Chiro G, Oldfield E, Wright DC, De Michele D, Katz DA, Patronas NJ, Doppman JL, Larson SM, Ito M, Kufta CV: Cerebral necrosis after radiotherapy and/or intraarterial chemotherapy for brain tumors: PET and neuropathologic studies. AJR Am J Roentgenol 1988; 150:189–97
110.
Millay RH, Klein ML, Shults WT, Dahlborg SA, Neuwelt EA: Maculopathy associated with combination chemotherapy and osmotic opening of the blood-brain barrier. Am J Ophthalmol 1986; 102:626–32
111.
Doolittle ND, Miner ME, Hall WA, Siegal T, Jerome E, Osztie E, McAllister LD, Bubalo JS, Kraemer DF, Fortin D, Nixon R, Muldoon LL, Neuwelt EA: Safety and efficacy of a multicenter study using intraarterial chemotherapy in conjunction with osmotic opening of the blood-brain barrier for the treatment of patients with malignant brain tumors. Cancer 2000; 88:637–47
112.
Gelman M, Chakeres DW, Newton HB: Brain tumors: Complications of cerebral angiography accompanied by intraarterial chemotherapy. Radiology 1999; 213:135–40
113.
Madajewicz S, Chowhan N, Tfayli A, Roque C, Meek A, Davis R, Wolf W, Cabahug C, Roche P, Manzione J, Iliya A, Shady M, Hentschel P, Atkins H, Braun A: Therapy for patients with high grade astrocytoma using intraarterial chemotherapy and radiation therapy. Cancer 2000; 88:2350–6
114.
Quyyumi AA, Dakak N, Husain S, Arora S, Gilligan DM, Panza JA, Cannon RO: Nitric oxide activity in the human coronary circulation: Impact of risk factors for coronary atherosclerosis. J Clin Invest 1995; 95:1747–55
115.
Quyyumi AA, Dakak N, Andrews NP, Gilligan DM, Panza JA, Cannon RO: Contribution of nitric oxide to metabolic coronary vasodilation in the human heart. Circulation 1995; 92:320–6
116.
Stewart DJ, Belanger JM, Grahovac Z, Curuvija S, Gionet LR, Aitken SE, Hugenholtz H, Benoit BG, DaSilva VF: Phase I study of intracarotid administration of carboplatin. Neurosurgery 1992; 30:512–6
117.
Fogarty-Mack P, Pile-Spellman J, Hacein-Bey L, Ostapkovich N, Joshi S, Vulliemoz Y, Young WL: Superselective intraarterial papaverine administration: Effect on regional cerebral blood flow in patients with arteriovenous malformations. J Neurosurg 1996; 85:395–402
118.
Dedrick RL: Interspecies scaling of regional drug delivery. J Pharma Sci 1986; 75:1047–52
119.
Coubes P, Baldy-Moulinier M, Zanca M, Boire JY, Child R, Bourbotte G, Frerebeau P: Monitoring sodium methohexital distribution with [99mTc]HMPAO with single photon emission computed tomography during Wada test. Epilepsia 1995; 36:1041–9
120.
Bazin JE, Picard P, Gabrillargues J, Dordain M: Propofol administered via  the carotid artery to achieve a Wada test. Can J Anaesth 1998; 45:707–8
121.
Silva TM, Hernandez-Fustes OJ, Bueno ML, Varela AM, Machado S: The Wada test with propofol in a patient with epilepsy. Arq Neuropsiquiatr 2000; 58:348–50
122.
Bacia T, Bendarzewska-Nawrocka B, Szymanska O, Dowzenko A, Sadowski Z: A comparison of Wada test for cerebral speech dominance by use of amytal or brietal. Neurol Neurochir Pol 1998; 32 (suppl 2):247–61
123.
Grote CL, Wierenga C, Smith MC, Kanner AM, Bergen DC, Geremia G, Greenlee W: Wada difference a day makes: Interpretive cautions regarding same-day injections. Neurology 1999; 52:1577–82
124.
Takayama M, Miyamoto S, Ikeda A, Mikuni N, Takahashi JB, Usui K, Satow T, Yamamoto J, Matsuhashi M, Matsumoto R, Nagamine T, Shibasaki H, Hashimoto N: Intracarotid propofol test for speech and memory dominance in man. Neurology 2004; 63:510–5
125.
Isnard J, Garde P, Fischer C, Duquesnel J, Mauguiere F: Clinical and electroencephalographic manifestations during the Wada test using intracarotid injection of methohexital (Brietal) [in French]. Rev Neurol 1994; 150:266–7
126.
Origitano TC, Wascher TM, Reichman OH, Anderson DE: Sustained increased cerebral blood flow with prophylactic hypertensive hypervolemic hemodilution (“Triple-H” Therapy) after subarachnoid hemorrhage. Neurosurgery 1990; 27:729–40
127.
Joshi S, Meyers P, Pile-Spellman J, Wang M, Sahlein D: Intracarotid verapamil decreases both proximal and distal human cerebrovascular resistance. Anesthesiology 2004; 100:774–81
128.
Oskouian RJ Jr, Martin NA, Lee JH, Glenn TC, Guthrie D, Gonzalez NR, Afari A, Vinuela F: Multimodal quantitation of the effects of endovascular therapy for vasospasm on cerebral blood flow, transcranial Doppler ultrasonographic velocities, and cerebral artery diameters. Neurosurgery 2002; 51:30–41
129.
Smith WS, Dowd CF, Johnston SC, Ko NU, DeArmond SJ, Dillon WP, Setty D, Lawton MT, Young WL, Higashida RT, Halbach VV: Neurotoxicity of intra-arterial papaverine preserved with chlorobutanol used for the treatment of cerebral vasospasm after aneurysmal subarachnoid hemorrhage. Stroke 2004; 35:2518–22
130.
Melamed E, Lavy S: Effect of papaverine on regional cerebral blood flow of the contralateral hemisphere in patients with acute cerebral infarction. Appl Neurophysiol 1979; 42:217–23
131.
Olesen J, Paulson OB: The effect of intra-arterial papaverine on the regional cerebral blood flow in patients with stroke or intracranial tumor. Stroke 1971; 2:148–59
132.
Taylor CL, Selman WR, Ratcheson RA: Steal affecting the central nervous system. Neurosurgery 2002; 50:679–88
133.
Boysen G, Engell HC, Pistolese GR, Fiorani P, Agnoli A, Lassen NA: On the critical lower level of cerebral blood flow in man with particular reference to carotid surgery. Circulation 1974; 49:1023–5
134.
Hanson E, Anderson R, Sundt T: Influence of cerebral vasoconstricting and vasodilating agents on blood flow in regions of focal ischemia. Stroke 1975; 6:642–8
135.
Candia GJ, Heros RC, Lavyne MH, Zervas NT, Nelson CN: Effect of intravenous sodium nitroprusside on cerebral blood flow and intracranial pressure. Neurosurgery 1978; 3:50–3
136.
Joshi S, Hartl R, Wang M, Feng L, Hoh D, Sciacca RR, Mangla S: The acute cerebrovascular effects of intracarotid adenosine in nonhuman primates. Anesth Analg 2003; 97:231–7
137.
Cross DT III, Moran CJ, Angtuaco EE, Milburn JM, Diringer MN, Dacey RG Jr: Intracranial pressure monitoring during intraarterial papaverine infusion for cerebral vasospasm. Am J Neurorad 1998; 19:1319–23
138.
Kassell NF, Helm G, Simmons N, Phillips CD, Cail WS: Treatment of cerebral vasospasm with intra-arterial papaverine. J Neurosurg 1992; 77:848–52
139.
Joshi S, Hashimoto T, Ostapkovich N, Pile-Spellman J, Duong DH, Hacien-Bey L, Marshall RS, James DJ, Young WL: Effect of intracarotid papaverine on human cerebral blood flow and vascular resistance during acute hemispheric arterial hypotension. J Neurosurg Anesthesiol 2001; 13:146–51
140.
McAuliffe W, Townsend M, Eskridge JM, Newell DW, Grady MS, Winn HR: Intracranial pressure changes induced during papaverine infusion for treatment of vasospasm. J Neurosurg 1995; 83:430–4
141.
Mathis JM, DeNardo A, Jensen ME, Scott J, Dion JE: Transient neurologic events associated with intraarterial papaverine infusion for subarachnoid hemorrhage-induced vasospasm. AJNR Am J Neuroradiol 1994; 15:1671–4
142.
Mathis JM, DeNardo AJ, Thibault L, Jensen ME, Savory J, Dion JE: In vitro  evaluation of papaverine hydrochloride incompatibilities: A simulation of intraarterial infusion for cerebral vasospasm. Am J Neuroradiol 1994; 15:1665–70
143.
Ramzan I: Proconvulsant effect of papaverine on theophylline-induced seizures in rats. Clin Exp Pharmacol Physiol 1989; 16:425–7
144.
Joshi S, Hashimoto T, Ostapkovich N, Pile-Spellman J, Duong DH, Hacien-Bey L, Marshall RS, James DJ, Young WL: Effect of intracarotid papaverine on human cerebral blood flow and vascular resistance during acute hemispheric arterial hypotension. J Neurosurg Anesthesiol 2001; 13:146–51
145.
McHenry LC Jr, Jaffe ME, Kawamura J, Goldberg HI: Effect of papaverine on regional blood flow in focal vascular disease of the brain. N Engl J Med 1970; 282:1167–70
146.
Gogolak I, Gotoh F, Tomita M, Tanahashi N, Kobari M, Shinohara T, Mihara B: No intracerebral steal phenomenon in the ischemic brain following papaverine administration. Stroke 1985; 16:114–7
147.
Feng L, Fitzsimmons BF, Young WL, Berman MF, Lin E, Aagaard BD, Duong H, Pile-Spellman J: Intraarterially administered verapamil as adjunct therapy for cerebral vasospasm: Safety and 2-year experience. AJNR Am J Neuroradiol 2002; 23:1284–90
148.
Lavine SD, Wang M, Etu JJ, Meyers PM, Joshi S: Augmentation of cerebral blood flow and reversal of endothelin-1-induced vasospasm: A comparison of intracarotid nicardipine and verapamil. Neurosurgery 2007; 60:742–8
149.
Badjatia N, Topcuoglu MA, Pryor JC, Rabinov JD, Ogilvy CS, Carter BS, Rordorf GA: Preliminary experience with intra-arterial nicardipine as a treatment for cerebral vasospasm. AJNR Am J Neuroradiol 2004; 25:819–26
150.
Kanamaru K, Waga S, Kuga Y, Nakamura F, Kamata N: Transcranial Doppler pattern after intracarotid papaverine and prostaglandin E1 incorporated in lipid microsphere in patients with vasospasm. Neurol Med Chir (Tokyo) 1998; 38 (suppl):152–5
151.
Nakamura T, Tatara N, Morisaki K, Kawakita K, Nagao S: Cerebral oxygen metabolism monitoring under hypothermia for severe subarachnoid hemorrhage: Report of eight cases. Acta Neurol Scand 2002; 106:314–8
152.
Ding Y, Li J, Rafols JA, Phillis JW, Diaz FG: Prereperfusion saline infusion into ischemic territory reduces inflammatory injury after transient middle cerebral artery occlusion in rats. Stroke 2002; 33:2492–8
153.
Ding Y, Yao B, Zhou Y, Park H, McAllister JP II, Diaz FG: Prereperfusion flushing of ischemic territory: A therapeutic study in which histological and behavioral assessments were used to measure ischemia-reperfusion injury in rats with stroke. J Neurosurg 2002; 96:310–9
154.
Wagner KR, Zuccarello M: Local brain hypothermia for neuroprotection in stroke treatment and aneurysm repair. Neurol Res 2005; 27:238–45
155.
Fontaine S, Theron J, Melanson D, Villemure JG, Emrich J, Raymond J, Ethier R: Superselective cerebral arterial infusion of BCNU in high-grade glioma: The radiologist’s point of view. Can Assoc Radiol J 1988; 39:178–81
156.
Cloughesy TF, Gobin YP, Black KL, Vinuela F, Taft F, Kadkhoda B, Kabbinavar F: Intra-arterial carboplatin chemotherapy for brain tumors: A dose escalation study based on cerebral blood flow. J Neurooncol 1997; 35:121–31
157.
Saris SC, Shook DR, Blasberg RG, Dedrick RL, Doppman JL, Bankiewicz KS, Blacklock JB, Oldfield EH: Carotid artery mixing with diastole-phased pulsed drug infusion. J Neurosurg 1987; 67:721–5
158.
Roman-Goldstein S, Mitchell P, Crossen JR, Williams PC, Tindall A, Neuwelt EA: MR and cognitive testing of patients undergoing osmotic blood-brain barrier disruption with intraarterial chemotherapy. AJNR Am J Neuroradiol 1995; 16:543–53
159.
Elkassabany N, Bhatia J, Deogaonkar A, Barnette G, Ferece S, Lotto M, Maurtua M, Ebrahim Z, Schubert A, Farga E: Anesthetic management of blood brain barrier disruption (BBBD): A retrospective review (abstract). J Neurosurg Anesthesiol 2005; 17:240
160.
Jahan R: Hyperacute therapy of acute ischemic stroke: Intraarterial thrombolysis and mechanical revascularization strategies. Tech Vasc Interv Radiol 2005; 8:87–91
161.
Jahan R, Vinuela F: Intraarterial thrombolysis for acute ischemic stroke. Adv Neurol 2003; 92:383–7
162.
Keris V, Rudnicka S, Vorona V, Enina G, Tilgale B, Fricbergs J: Combined intraarterial/intravenous thrombolysis for acute ischemic stroke. AJNR Am J Neuroradiol 2001; 22:352–8
163.
Mielke O, Wardlaw J, Liu M: Thrombolysis (different doses, routes of administration and agents) for acute ischaemic stroke. Cochrane Database Syst Rev 2004; (4):CD000514
164.
Mahon BR, Nesbit GM, Barnwell SL, Clark W, Marotta TR, Weill A, Teal PA, Qureshi AI: North American clinical experience with the EKOS MicroLysUS infusion catheter for the treatment of embolic stroke. AJNR Am J Neuroradiol 2003; 24:534–8
165.
Bourekas EC, Slivka AP, Shah R, Sunshine J, Suarez JI: Intraarterial thrombolytic therapy within 3 hours of the onset of stroke. Neurosurgery 2004; 54:39–44
166.
Lee DH, Jo KD, Kim HG, Choi SJ, Jung SM, Ryu DS, Park MS: Local intraarterial urokinase thrombolysis of acute ischemic stroke with or without intravenous abciximab: A pilot study. J Vasc Interv Radiol 2002; 13:769–74
167.
Neuroprotection: The end of an era? Lancet 2006; 368:1548
168.
Del Zoppo GJ: Why do all drugs work in animals but none in stroke patients? 1: Drugs promoting cerebral blood flow. J Intern Med 1995; 237:79–88.
169.
Grotta J: Neuroprotection is unlikely to be effective in humans using current trial designs. Stroke 2002; 33:306–7
170.
Yokota H, Nakabayashi M, Fuse A, Mashiko K, Yamamoto Y, Henmi H, Otsuka T: Continuous intracarotid infusion of mannitol in severe head injury [in Japanese]. No Shinkei Geka 1993; 21:205–11
171.
Domanskaia IA, Kukushkin AA, Mazurok II, Il’in KL: Regionarnoe primenenie antibiotikov pri lechenii cherepno-mozgovoi travmy i gnoinykh oslozhnenii. Vestn Khir Im I I Grek 1993; 150:53–6
172.
Perkins BA, Strausbaugh LJ: Effect of mannitol infusions into the internal carotid artery on entry of two antibiotics into the cerebrospinal fluid and brains of normal rabbits. Antimicrob Agents Chemother 1983; 24:339–42
173.
Kudriavtsev AE, Shikov VG: Primenenie karotidnoi infuzii lekarstvennykh veshchestv dlia lecheniia bol’nykh gnoinym meningitom. Sov Med 1976:55–7
174.
Draliuk NS, Draliuk MG: Dlitel’naia intrakarotidnaia infuziia v neirokhirurgii. Zh Vopr Neirokhir Im N N Burdenko 1986; July–August:54–7
175.
Mozhaev SV, Zubkov Iu N, Ponomarev AM, Shimchenko P: Nekotorye voprosy kliniki i kompleksnogo lecheniia septicheskikh trombozov kavernoznykh sinusov. Vestn Khir Im I I Grek 1980; 125:43–50
176.
Rainov NG, Ikeda K, Qureshi NH, Grover S, Herrlinger U, Pechan P, Chiocca EA, Breakefield XO, Barnett FH: Intraarterial delivery of adenovirus vectors and liposome-DNA complexes to experimental brain neoplasms. Hum Gene Ther 1999; 10:311–8
177.
Chauvet AE, Kesava PP, Goh CS, Badie B: Selective intraarterial gene delivery into a canine meningioma. J Neurosurg 1998; 88:870–3
178.
Barnett FH, Rainov NG, Ikeda K, Schuback DE, Elliott P, Kramm CM, Chase M, Qureshi NH, Harsh Gt, Chiocca EA, Breakefield XO: Selective delivery of herpes virus vectors to experimental brain tumors using RMP-7. Cancer Gene Ther 1999; 6:14–20
179.
Lu D, Li Y, Wang L, Chen J, Mahmood A, Chopp M: Intraarterial administration of marrow stromal cells in a rat model of traumatic brain injury. J Neurotrauma 2001; 18:813–9
180.
Amar AP, Zlokovic BV, Apuzzo ML: Endovascular restorative neurosurgery: A novel concept for molecular and cellular therapy of the nervous system. Neurosurgery 2003; 52:402–12
181.
Zlokovic BV, Apuzzo ML: Cellular and molecular neurosurgery: Pathways from concept to reality—part II: Vector systems and delivery methodologies for gene therapy of the central nervous system. Neurosurgery 1997; 40:805–12
182.
Suzuki S, Ito O, Savama T, Yamaguchi S, Goto K, Sasaki T: Intraarterial injection of colforsin daropate hydrochloride for treatment of cerebral vasospasm after aneurysmal subarachnoid hemorrhage: Preliminary report of two cases. Neuroradiology 2006; 48:50–3
183.
Fortin D, Osztie E, Neuwelt EA: Iatrogenic arterial spasm relieved by intraarterial mannitol infusion. AJNR Am J Neuroradiol 2000; 21:968–70
184.
Bejjani GK, Duong DH, Kalamarides M, Ziyal I, Sullivan BJ: Cerebral vasospasm after tumor resection: A case report. Neurochirurgie 1997; 43:164–8
185.
Fandino J, Kaku Y, Schuknecht B, Valavanis A, Yonekawa Y: Improvement of cerebral oxygenation patterns and metabolic validation of superselective intraarterial infusion of papaverine for the treatment of cerebral vasospasm. J Neurosurg 1998; 89:93–100
186.
Touho H, Furuoka N, Karasawa J: Distribution of intraarterially administered papaverine in endovascular treatment of delayed cerebral vasospasm. Neurol Med Chir (Tokyo) 1997; 37:163–71
187.
Yoshida K, Watanabe H, Nakamura S: Intraarterial injection of amrinone for vasospasm induced by subarachnoid hemorrhage. AJNR Am J Neuroradiol 1997; 18:492–6
188.
Clouston JE, Numaguchi Y, Zoarski GH, Aldrich EF, Simard JM, Zitnay KM: Intraarterial papaverine infusion for cerebral vasospasm after subarachnoid hemorrhage. AJNR Am J Neuroradiol 1995; 16:27–38
189.
Miller JA, Cross DT, Moran CJ, Dacey RG Jr, McFarland JG, Diringer MN: Severe thrombocytopenia following intraarterial papaverine administration for treatment of vasospasm. J Neurosurg 1995; 83:435–7
190.
Morgan MK, Day MJ, Little N, Grinnell V, Sorby W: The use of intraarterial papaverine in the management of vasospasm complicating arteriovenous malformation resection: Report of two cases. J Neurosurg 1995; 82:296–9
191.
Nakagawa H, Fujita T, Kubo S, Tsuruzono K, Yamada M, Tokiyoshi K, Miyawaki Y, Kanayama T, Kadota T, Hayakawa T: Selective Intra-arterial chemotherapy with a combination of etoposide and cisplatin for malignant gliomas: Preliminary report. Surg Neurol 1994; 41:19–27
192.
Kaku Y, Yonekawa Y, Tsukahara T, Kazekawa K: Superselective intra-arterial infusion of papaverine for the treatment of cerebral vasospasm after subarachnoid hemorrhage. J Neurosurg 1992; 77:842–7
193.
Boker DK, Solymosi L, Wassmann H: Immediate postangiographic intraarterial treatment of cerebral vasospasm after subarachnoid hemorrhage with nimodipine: Report on 3 cases. Neurochirurgia (Stuttg) 1985; 28 (suppl 1):118–20
194.
Abou-Chebl A, Bajzer CT, Krieger DW, Furlan AJ, Yadav JS: Multimodal therapy for the treatment of severe ischemic stroke combining GPIIb/IIIa antagonists and angioplasty after failure of thrombolysis. Stroke 2005; 36:2286–8
195.
Zaidat OO, Suarez JI, Sunshine JL, Tarr RW, Alexander MJ, Smith TP, Enterline DS, Selman WR, Landis DM: Thrombolytic therapy of acute ischemic stroke: Correlation of angiographic recanalization with clinical outcome. AJNR Am J Neuroradiol 2005; 26:880–4
196.
Li SM, Miao ZR, Zhu FS, Wang ML, Hua Y, Dong Y, Song QB, Ling F: Combined intraarterial thrombolysis and intra-cerebral stent for acute ischemic stroke institute of brain vascular diseases [in Chinese]. Zhonghua Yi Xue Za Zhi 2003; 83:9–12
197.
Rabinstein AA, Wijdicks EF, Nichols DA: Complete recovery after early intraarterial recombinant tissue plasminogen activator thrombolysis of carotid T occlusion. AJNR Am J Neuroradiol 2002; 23:1596–9
198.
Fukuda I, Gomi S, Meguro K, Wada M: Impact of immediate cerebral angiography for in-hospital cerebral thromboembolism after cardiovascular surgery. Jpn J Thorac Cardiovasc Surg 2001; 49:282–6
199.
Kase CS, Furlan AJ, Wechsler LR, Higashida RT, Rowley HA, Hart RG, Molinari GF, Frederick LS, Roberts HC, Gebel JM, Sila CA, Schulz GA, Roberts RS, Gent M: Cerebral hemorrhage after intra-arterial thrombolysis for ischemic stroke: The PROACT II trial. Neurology 2001; 57:1603–10
200.
Sylaja PN, Kuruttukulam G, Joseph S, Gupta AK, Radhakrishnan K: Selective intra arterial thrombolysis in acute carotid territory stroke. Neurol India 2001; 49:153–7
201.
Suwanwela NC, Phanthumchinda K, Suwanwela N, Tantivatana J, Janchai A: Thrombolytic treatment for acute ischemic stroke: A 2 year-experience at King Chulalongkorn Memorial Hospital. J Med Assoc Thai 2001; 84 (suppl 1):S428–36
202.
Endo S, Kuwayama N, Hirashima Y, Akai T, Nishijima M, Takaku A: Results of urgent thrombolysis in patients with major stroke and atherothrombotic occlusion of the cervical internal carotid artery. AJNR Am J Neuroradiol 1998; 19:1169–75
203.
Jahan R, Duckwiler GR, Kidwell CS, Sayre JW, Gobin YP, Villablanca JP, Saver J, Starkman S, Martin N, Vinuela F: Intraarterial thrombolysis for treatment of acute stroke: Experience in 26 patients with long-term follow-up. AJNR Am J Neuroradiol 1999; 20:1291–9
204.
Barr JD, Mathis JM, Wildenhain SL, Wechsler L, Jungreis CA, Horton JA: Acute stroke intervention with intraarterial urokinase infusion. J Vasc Interv Radiol 1994; 5:705–13
205.
Casto L, Moschini L, Camerlingo M, Gazzaniga G, Partziguain T, Belloni G, Mamoli A: Local intraarterial thrombolysis for acute stroke in the carotid artery territories. Acta Neurol Scand 1992; 86:308–11