Abstract
Introduction:
Research has advanced our understanding of the molecular and cellular mechanisms of cerebral edema and has propelled the development of novel anti-edema therapeutics. Current evidence supports aberrant neuro-glial ion transport as a central mechanism that underlies pathological fluid accumulation after central nervous system injury.
Areas covered:
Novel agents in clinical development show potential in altering the natural history and treatment of cerebral edema. Using the PubMed and Google Scholar databases, we review recent advances in our understanding of cerebral edema and describe agents under active investigation, their mechanism, and their application in recent and ongoing clinical trials.
Expert Opinion:
Pharmacotherapies that target molecular mechanisms underlying the compensatory post-injury response of ion channels and transporters that lead to pathological alteration of osmotic gradients, are the most promising therapeutic strategies. Repurposing of drugs such as glyburide that inhibit the aberrant upregulation of ion channels such as SUR1-TRPM4, and novel agents, such as ZT-a1, which re-establish physiological regulation of ion channels such as NKCC1/KCC, could be useful adjuvants to prevent and even reverse fluid accumulation in the brain parenchyma.
1. Introduction
Cerebral edema, the pathological accumulation of fluid in the brain parenchyma, is caused by multiple traumatic and pathological insults, including traumatic brain injury (TBI), stroke (both hemorrhagic and ischemic), infection, primary and metastatic tumors, and inflammatory disease. Even some systemic diseases, including acute liver failure and diabetic ketoacidosis, can lead to brain swelling. Regardless of the inciting event, a common consequence of cerebral edema is the elevation of intracranial pressure (ICP). Swelling brain tissue and increasing ICPs result in compromised cerebral blood flow, ischemia, cell death, and neurological deficits.1 Although the severity, location, and extent of swelling determine the specific downstream consequences, the side effects are often severe, frequently affecting functional outcome and increasing mortality to upwards of 80%.1,2
Here, we review recent advancements in our understanding of the pathology of cerebral edema and highlight novel or repurposed therapeutic agents that are being explored in animal models of edema, as well as preclinical and clinical human trials. This was accomplished through a comprehensive literature review using both PubMed and Google Scholar databases, limiting searches to include only studies published after the year 2000 so that the most up-to-date information was reported. Clinicaltrails.gov was utilized for all ongoing or recently completed clinical trials.
2. Management of Cerebral Edema
The Monro-Kellie doctrine, first described by Dr. Alexander Monro and Dr. George Kellie more than two centuries ago, is a well-accepted principle in neurological disease. This principle states that the sum of the volumes of intracerebral blood, cerebrospinal fluid, and brain tissue, consisting of interstitial and intracellular fluid, is constant given the constraints of a rigid skull.3 An increase in one must cause a reciprocal decrease in one or more of the others. Therefore, current management of cerebral edema focuses on temporizing tissue swelling and decreasing ICPs to prevent brain herniation and ischemia resulting from high ICPs and loss of cerebral blood flow.
Osmotherapy, a common first line management, consists of intravenous administration of hypertonic solution (typically mannitol or hypertonic saline). Following Starling’s principle, creating an osmotic gradient across blood vessels causes water to move from the intra- and extra-cellular compartments of the brain into the vasculature, decreasing parenchymal fluid volume. This movement of water along an imposed ionic gradient serves to decrease intracranial volume, and as a result, pressures. In addition to osmotherapy, the glucocorticoid Decadron is commonly used, especially in the setting of tumor-induced edema.4 Other less commonly used medications for edema include loop diuretics (typically furosemide), anti-inflammatory agents, and barbiturates.5
Although often first-line, medical management is largely temporizing and exposes patients to significant side effects, as well as rebound swelling after therapy is discontinued. Surgical management with decompressive craniectomy is often required if medical therapy fails, or as initial management if the edema is too extensive, resulting in dramatically increased ICP, and urgent intervention is needed. Interestingly, these strategies to reduce cerebral swelling and increasing ICP have been used for over a century.
3. Pathophysiology of Cerebral Edema
Cerebral edema is traditionally classified as cytotoxic or vasogenic, based on location of fluid accumulation.6,7 Vasogenic edema causes increased extracellular fluid due to blood-brain barrier (BBB) breakdown, while cytotoxic edema occurs when water accumulates in the intracellular space, resulting in cell swelling.6,7 Ionic cerebral edema, more recently defined as a subset of cytotoxic edema, consists of vessel leakage into the extracellular space through an intact BBB.6
While historical classifications remain useful, current research is advancing our understanding of the mechanisms underlying cerebral edema. Intracranial water balance is based on intra- and extra-cellular and vascular ionic gradients dictated primarily by cellular channels and transporters. Accordingly, current theories ascribe the pathophysiology of cerebral edema to alterations in intracranial ion transport, and new pharmacological targets in this framework are under investigation.1
4. Anti-edema Drugs in Development
As our understanding of the pathophysiology of cerebral edema increases, pharmacological treatments are being developed to target specific, underlying molecular mechanisms. New candidates are showing promise in attenuating tissue swelling and improving functional outcomes in in vivo models and clinical trials. Table 1 summarizes these novel therapeutic agents.
Table 1:
Drug | Target | Experimental Investigation | Preclinical Findings | Clinical Trials | Clinical Trial Patient Population | Clinical Trial Results |
---|---|---|---|---|---|---|
ZT-1a | SPAK | Zhang et al. 20204 | ZT-1a reduced NKCC1 and KCC2 phosphorylation, mitigating cerebral edema and improving functional outcomes in a rat model of stroke | N/A | N/A | N/A |
Glyburide | SUR1-TRPM4 | Simard et al. 20065 | Glyburide attenuated brain water volume and decreased 7-day mortality from 65% to 24% in a rat model of middle cerebral artery occlusion | NCT01268683; Phase 2a; Completed in 2013 | 10 patients with a 82–210 mL acute MCA or MCA/ACA ischemic stroke | Improved clinical outcomes and attenuated vasogenic edema |
NCT01794182; Phase 2; Completed in 2016 | 83 patients with a 82–300 mL acute MCA ischemic stroke | Attenuated NIH stroke scale scores and reduced 30-day mortality rates, however, the primary and secondary outcome goals were not met | ||||
NCT02864953; Phase 3; Ongoing | Aims to recruit 680 patients with 80–300 mL acute MCA ischemic stroke | N/A | ||||
Bevacizumab | VEGF-A | Folkins et al. 20078 | Bevacizumab reduced BBB permeability and normalized tumoral and peritumoral vasculature | No identifier; Phase 2; Completed in 2007 | 32 patients with progressive or recurrent grade III/IV gliomas who were treated with radiation and not surgical intervention | Reduced tumor size and edema resulting in improved neurological outcomes |
NCT00943826; Phase 3; Completed in 2015 | 921 patients with stable or decreasing glucocorticoid use and a newly diagnosed, untreated glioblastoma | Improved length of progression-free survival and maintenance of baseline performance, but failed to improve overall survival | ||||
Cerdiranib | VEGFR | Kamoun et al. 200916 | Cerdiranib reduced tumor vasculature permeability and size, as well as expansion of edema | NCT00305656; Phase 2; Completed in 2012 | 31 patients with a confirmed diagnosis of glioblastoma | More typical vascular development around the tumor and mitigated development of cerebral edema |
Celecoxib | COX2 | Chu et al. 200411 | Celecoxib treatment in rats with induced cerebral hemorrhage reduced edema, inflammation, and cell death, leading to increased functional recovery | NCT00526214; Pilot Trial; Completed in 2009 | 44 patients with diagnosed intracerebral hemorrhage not caused by trauma, aneurysmal bleeding, or anticoagulation | Mitigated hematoma and perihematomal edema progression and expansion |
Fingolimod | S1P Receptors | Wei et al. 201113 | Fingolimod reduced infarct volume, neuronal cell death, edema, and neurological dysfunction in a mouse model of middle cerebral artery occlusion | NCT02002390; Phase 2; Completed in 2014 | 22 patients with ischemic stroke or intracerebral hemorrhage not caused by coagulopathy, trauma, or thrombocytopenia | Reduced edema and improved neurological outcomes in hemorrhage patients and reduced lesional expansion and improved neurological functioning in stroke patients |
Conivaptan | AVP A1A/A2 | Can et al. 201917 | Conivaptan was shown to be a more potent diuretic than mannitol in a rat model of ischemic brain injury | NCT03000283; Pilot Trial; Ongoing | Goal of 7 patients with an intracerebral hemorrhage of >20 mL not due to thrombolysis, infection, trauma, or tumor | N/A |
Xerecept | Unknown | Tjuvajev et al. 199614 | Xerecept directly acted on tumor microvasculature, reducing permeability and vasogenic edema in rats with RG2 cell-derived gliomas | No identifier; Phase 1; Completed in 1998 | 17 patients with primary brain tumors and radiographic evidence of edema | 10 of 17 patients in the clinical trial had improved neurological outcomes |
NCT00088166; Phase 3; Completed in 2008 | 200 patients with a brain tumor and complications from prior steroid use | Reduced steroid requirements and steroid-associated side effects such as Cushing’s Syndrome and myopathy |
4.1. Cation-Chloride Cotransporter (CCC) Regulation
Intracranial ionic homeostasis is, in part, maintained by cation-Cl− cotransporters (CCC), specifically NKCC1 and the KCCs. Through phosphorylation, SPAK (SPS1-related proline/alanine-rich kinase) is the master regulator of both, stimulating NKCC1 and inhibiting KCCs.8 As electroneutral cotransporters, NKCC1 imports and KCC1–4 export Cl− by utilizing the transmembrane gradients of Na+ and/or K+. Their coordinated regulation is required to effect appropriate cell volume changes while preventing pathological volume changes in response to altered osmotic gradients. SPAK, in combination with OSR1 (oxidative stress-responsive kinase 1), which functions similarly to SPAK in regard to modulating NKCC1 and KCCs activity, ensures control over cellular Cl− concentrations, and consequently, water movement and cell volume.8 In experimental models, enhanced SPAK activity has been found in ischemia-induced cerebral edema.9
A newly developed, selective SPAK inhibitor, ZT-1a (5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide), is a modulator of both NKCC1 and KCC, inhibiting and activating these cotransporters, respectively.9 SPAK inhibition with ZT-1a stimulates K+-dependent Cl− export, and improves regulation of cellular volume after insult. In animal models, ZT-a1 reduces ischemia-induced CCC phosphorylation, and results in reduced infarct volume, decreased cerebral edema, and improved functional outcomes.9 Although further research and clinical trials are needed to better understand the therapeutic benefit and potential side effects, given the target specificity and promising in vivo results, ZT-1a has significant therapeutic potential for treatment of cerebral edema.
4.2. SUR1-TRPM4 Inhibition
The sulfonylurea receptor 1 (Sur1) is an ion channel important in cerebral ion homeostasis that is upregulated in neurons, astrocytes, microglia, oligodendrocytes, and microvascular endothelial cells under ischemic conditions in both humans10,11 and animal models.4,12–14 Sur1 associates with transient receptor potential melastatin 4 (Trpm4), which is concurrently upregulated with Sur1 after cerebral ischemia,15 to create SUR1-TRPM4, a non-selective cation channel.4 SUR1-TRPM4 can complex with AQP4, increasing influx of cations and water into cells, particularly astrocytes. In addition, SUR1 expression contributes to vascular damage that may play a role in vasogenic edema.4 Glyburide, a second-generation sulfonylurea developed for type 2 diabetes mellitus, targets the SUR1-TRPM4 channel and inhibits its upregulation after central nervous system (CNS) injury. In vivo studies show that glyburide-mediated inhibition of SUR1/TRPM4 after ischemic injury reduces brain swelling and death.12
Glyburide, which is used clinically to lower blood glucose levels in diabetic patients, does not penetrate the intact blood-brain barrier. However, after ischemic, which results in BBB breakdown, significant levels of the drug accumulate in the injured brain tissue. Therefore, low doses of glyburide appear to allow therapeutic effects on ischemia-related edema. Although glyburide’s system mechanism of action does present dose-limiting side effect of hypoglycemia, treatment with low doses appear to have therapeutic effects on the post-ischemic brain with minimal effect on blood glucose.4 With the conclusion of phase 2 clinical trials, evidence supports administration of intravenous glyburide; the greatest benefit was observed in stroke patients with large hemispheric infarcts, where glyburide treatment resulted in reduced parenchymal swelling, improved functional outcomes, and reduced mortality (ClinicalTrials.gov Identifiers: NCT01268683; NCT01794182). A phase 3 clinical trial is currently underway (ClinicalTrials.gov Identifiers: NCT02864953).
4.3. Vascular Endothelial Growth Factor (VEGF) Inhibition
Intracranial malignancies, both primary and metastatic, cause significant peri-lesional edema. The chronic progression of tumor-associated edema can be difficult to manage and often causes significant morbidity and mortality. Vascular endothelial growth factor (VEGF), a glycoprotein upregulated in intracranial malignancies, contributes to tumor angiogenesis and formation of inter-endothelial gaps, fragmentations, and fenestrations in the brain endothelium; thus, overexpression and aberrant activity of VEGF may result in compromised BBB function.16,17 Bevacizumab, a monoclonal immunoglobulin G humanized antibody against VEGF-A, and cediranib, a VEGFR tyrosine kinase antagonist, have emerged as promising anti-angiogenic and anti-edema therapies.18,19 Although VEGF inhibition by these drugs does not improve overall patient survival, animal studies and clinical trials demonstrate normalization of tumor vasculature, reduction in the severity of peritumoral edema, and improvement of progression-free survival in both animal studies and clinical trials (ClinicalTrials.gov NCT00943826; NCT00305656).20–22
VEGF inhibition has been investigated in stroke-induced cerebral edema as well, however, findings remain contradictory and time-dependent, including a possible protective effect of VEGF in the early post-stroke brain. Ongoing investigation may further clarify the role of VEGF and the consequences of its inhibition in a stroke setting, which may allow introduction of clinical trials in the future that are likely focused on delayed VEGF inhibiton.23,24 However, researchers and physicians advocating for the use of antiangiogenic agents such as bevacizumab and cediranib in tumor or stoke pathology should be aware that they are not without risks and adverse events. Their use has been found to cause increased risk of thromboembolic complications, hypertension, hemorrhage, gastrointestinal perforation, rebound edema, and surgical wound dehiscence. Additionally, given an increased bleeding risk, once treatment is started, surgery is contraindicated for at least 28 days after last dose, preventing any further potentially necessary surgical intervention.19,25
4.4. Arginine Vasopressin (AVP) Receptor Inhibition
Arginine vasopressin (AVP), also known as antidiuretic hormone (ADH), a peptide produced in the posterior pituitary, has been implicated in intracerebral volume regulation. AVP exerts homeostatic effects via signaling through G protein-coupled receptors expressed on vasculature (V1A), the anterior pituitary gland (V1B/V3), and in renal collecting duct principal cells (V2), conferring integrated control of body fluid volume. Physiologically present in non-pathological CSF, AVP demonstrates an ability to increase brain water content,1 and plasma concentrations can be significantly increased in stroke patients.26 Indeed, the hyponatremia characteristic of the syndrome of inappropriate antidiuretic hormone secretion (SIADH) is present in a significant proportion of TBI and SAH patients, indirectly exacerbating brain edema.27
Vaptans, small-molecule vasopressin receptor inhibitors, can modestly ameliorate cerebral edema. Administration of the AVP A1A/A2 receptor inhibitor, conivaptan, demonstrated reduction of brain edema in a rodent model,28 and is now being investigated in an ongoing phase 1 clinical trial (ClinicalTrials.gov Identifier: NCT03000283). However, while conivaptan effects on renal water excretion have been well characterized, the drug’s mechanism of action in the brain remains poorly understood, warranting further molecular and clinical research. Furthermore, the side effect profile of conivaptan includes hypernatremia, dehydration, and infusion site reaction. Conivaptan may also precipitate renal failure and ischemic organ damage; thus, given the aforementioned considerations, it is classified as a category C drug in pregnancy, and it is contraindicated in lactating women. These risks will need to be taken into account if proven effective for cerebral edema in a clinical setting.29
4.5. Inflammatory Cascade Inhibition
Proinflammatory cascades induced by CNS injury often contribute to widespread cerebral edema in patients. Cyclooxygenase (COX) enzymes propagate this inflammatory signaling by converting arachidonic acid into proinflammatory prostaglandins.30 This action plays a key pathological role in amplifying tissue injury, especially in the setting of intracerebral hemorrhage (ICH). COX2 is upregulated in endothelium and leukocytes in rodent models of ICH, exacerbating progression of neuronal cell death, infarct volume, and brain edema.1,31 In a retrospective study, ICH patients treated with the non-steroidal COX2 inhibitor, celecoxib, exhibited attenuated hematoma expansion and decreased edema.32 A 2009 pilot clinical trial of 44 patients demonstrated similar findings, showing reduced hematoma expansion and less peri-hematomal edema in patients treated with celecoxib as compared with standard management (ClinicalTrials.gov Identifier: NCT00526214).33 Although celecoxib treatment following cerebral hemorrhage failed to show sustained improvements in functional outcomes, COX inhibitors of increased specificity are being developed and investigated.33
Sphingosine-1-phosphate (S1P) and its G protein-coupled receptors S1P1–5 propagate inflammatory responses.34 Expressed by all cell types in the CNS, S1P receptors are upregulated in neuroinflammatory conditions such as stroke.35 Each receptor type demonstrates a unique function determined by its localization. S1P1, S1P2, and S1P3 are expressed on neuro-endothelial cells, where they regulate vascular and BBB permeability. The prominent role of S1P2 in disrupting intercellular adherens junctions and increasing vascular permeability makes it of particular interest as a potential therapeutic target in brain edema.36 The S1P receptor modulator Fingolimod, originally approved to treat multiple sclerosis, is being repurposed for treatment of brain edema, with promising preliminary and clinical data. In rodent models of ICH, fingolimod mitigated onset and progression of cerebral edema.37 Similarly, in a phase 2 clinical trial of patients with either ischemic or hemorrhagic stroke, fingolimod reduced peri-hematomal edema and lesional growth, and improved neurological outcomes (ClinicalTrials.gov Identifier: NCT02002390). However, the mechanism by which fingolimod exerts its effects via the S1Ps remains elusive and requires further research. Furthermore, the side effects associated with fingolimod include bradycardia and slowing of atrioventricular conduction, requiring close observation upon initiation, increase risk of infections due to its immunosuppressive effects, and has been associated with a few cases of progressive multifocal leukoencephalopathy.38,39 Development of a specific S1P2 inhibitor may allow direct inhibition of vasogenic edema, mitigating the off-target effects responsible for adverse treatment outcomes.
4.6. Corticotrophin-Releasing Factor Therapy
Although corticosteroid therapy has proven effective for management of cerebral edema in some intracranial pathologies, and particularly in peritumor edema, the significant systemic side effect profile has prompted development of “steroid-sparing” therapies.40 Of these, human corticotrophin-releasing factor (hCRF) has as shown promise clinically. Alternatively named Xerecept, this synthetic, modified hypothalamic peptide demonstrates protective effects on brain endothelium and a lower incidence of the severe side effects associated with corticosteroid treatment when given systemically. For example, administration of Xerecept in an RG2 cell-derived glioma rodent model significantly reduces vasogenic brain edema.41 In humans, a completed phase I clinical trial demonstrated improved neurological outcomes and reduced peritumoral edema in 10 of 17 primary brain tumor patients treated with Xerecept.42 A follow up phase 3 clinical trial of 200 brain tumor patients showed that Xerecept was effective in reducing steroid requirements and steroid-related side effects such as myopathy and Cushing’s Syndrome (ClinicalTrials.gov Identifier: NCT00088166).43
5. Conclusion
Cerebral edema is a significant contributor to the morbidity and mortality of many central nervous system pathologies, especially with acute injuries and disease. The currently limited options for standard of care management include temporizing intracranial pressures with the administration of hypertonic solutions, corticosteroids, and in the most severe cases, decompressive craniectomy. However, as our understanding of the molecular drivers of cerebral edema improves, several new therapeutic agents are being developed and tested in animal models and clinical trials with promising results. With novel and specific pharmacological targets to effectively treat, and even prevent brain swelling, future patients may be partially or completely spared of the brain damage associated with elevated ICP and cerebral ischemia.
6. Expert Opinion
As an underlying theme in most neurologic and neurosurgical disease, cerebral edema represents a common secondary pathology leading to increased mortality and neurological deficits in patients. Clinical management of edema largely centers on temporizing strategies to minimize consequences of acute fluid accumulation, specifically mass effect and elevated ICPs. Corticosteroids are used for longer term management; however, their side effect profile often limits their utility and chronic administration.
Fortunately, our growing understanding of the molecular and cellular mechanisms underlying the pathophysiology of cerebral edema is fostering development of targeted antiedema agents. The most promising candidates are those targeting specific molecular mechanisms controlling the compensatory post-injury response of ion channels and transporters that lead to pathological alteration of osmotic gradients. Although further clinical studies are needed, repurposing of drugs such as glyburide to inhibit the aberrant upregulation of ion channels such as SUR1-TRPM4, and novel agents, such as ZT-a1, which re-establish physiological regulation of ion channels like NKCC1/KCC, appear to restore ion gradient homeostasis to prevent and even reverse fluid accumulation in the brain parenchyma.
Although cerebral edema across all CNS pathologies has a likely common underlying pathophysiology, it is important to consider the unique characteristics of these lesions. TBI, an acute and global CNS injury, likely involves different molecular drivers than peritumoral edema or peri-lesional edema in hemorrhagic or ischemic stroke territories. Timing of drug administration is also a consideration. Injuries such as TBI and ICH stimulate processes contributing to a pro-edema environment, including upregulation of SUR1-TRPM4 channels and enhancement of peri-lesional SPAK activity. However, in chronic and evolving oncological lesions, upregulation of the VEGF pathway and inflammatory cascades through both aberrant CNS signaling, as well as pathological signaling from the tumor itself, offer alternative pathways for drug targeting. Conversely, however, the role of VEGF in post-ischemic cerebral edema remains controversial. Some studies suggest a beneficial role of VEGF inhibition post-stroke; however, many also show a beneficial and neuroprotective effect of active VEGF signaling. These apparent inconsistencies in the literature are likely a result of experimental timing and targeting of VEGF in the ischemic brain and will likely become clearer and better defined as pre-clinical and clinical research advances and elucidates further the underlying mechanism of VEGF-mediated response.
As our understanding of the molecular mechanism and pathways underlying the development and propagation of cerebral edema improve, more signaling pathways and channels/transporters will emerge as potential targets. A recently published study showing improvement in spinal cord edema using an antipsychotic drug, trifluoperazine (TFP), identifies targeting of aquaporin water channels as a possible strategy to combat development of CNS edema.44
One important challenge that remains is how to determine when a drug target effectively reduces cerebral ischemia in the clinical population. Most studies rely on data such as imaging findings, neurological symptoms, and need for decompressive hemicraniectomy to determine drug effect and reduction in cerebral edema. However, given the complex patient presentation and clinical course that varies based on underlying pathology and progression of disease, defined and consistent endpoints would be valuable in evaluating the drug targets. Once such scoring system has been recently proposed as a standardized mechanism for determining the effect of corticosteroid use in peritumoral edema (The Response Assessment in Neuro-Oncology (RANO) proposal).45 A consistent and broadly used criteria that takes into consideration findings from previously successful clinical trials would allow more thorough evaluation of current and future drug targets and their ability to improve the neurological outcomes in this patient population.
In vivo and clinical trials are gaining momentum. Repurposing current drugs will be beneficial in shortening the translational timeline between in vivo preclinical research and eventual clinical application. Agents such as COX2 inhibitors and the VEGF inhibitor bevacizumab offer important avenues for expansion of the range of anti-edema agents available for the clinic. Although hurdles remain to be overcome in developing effective and novel therapeutics for cerebral edema, promising targets are being identified, driving new pharmaceutical development. As we identify the molecular drivers of cerebral edema and leverage these discoveries to create novel treatment strategies, clinical management of cerebral edema will become more targeted and effective.
Article Highlights.
Cerebral edema is the pathological accumulation of fluid in the brain and a potentially life-threatening complication of many central nervous system insults, including traumatic brain injury, stroke, infection, tumor, and inflammatory disease.
Due to the constraints of a rigid skull, cerebral edema often leads to increased intracranial pressure (ICP) and compromised cerebral blood flow, resulting in cell death and neurological dysfunction. Current strategies to reduce cerebral edema and ICPs have been used for over a century and rely on osmotic agents and surgery.
Historically, cerebral edema has been classified as either cytotoxic, vasogenic, or ionic, depending on location of water accumulation and blood brain barrier integrity. As our understanding of the underlying pathological mechanisms of cerebral edema expands, these classifications are being redefined to reflect the molecular mediators involved and allow development of more specific treatment targets.
Cerebral edema is dynamic and time dependent, factors that must be considered when developing and administering targeted therapies. Ongoing research is exploring the temporal expression and regulation of proteins and transporters and their role in cerebral protection and development of cerebral edema. This will guide timing and indications for newly developed targeted therapies.
Development of new drugs, and repurposing of currently used drugs, to target recently discovered pathways, such as cation-chloride cotransporters and SUR1-TRPM4, will provide more effective treatment, and potentially prevention, of cerebral edema resulting from multiple intracranial pathologies.
Given the heterogenous presentation, progression, and etiology of cerebral edema, a standardized mechanism to assessment of efficacy of treatment is needed to guide clinical implementation and further treatment development.
Funding
The work in the laboratory of the authors has been funded by U.S. Department of Health and Human Services, National Institutes of Health R01 NS109358,R01 NS111029-01A1 and Yale-National Institutes of Health (NIH) Center for Mendelian Genomics 5U54HG006504
Declaration of interest
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
Footnotes
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers
- 1.Stokum JA, Gerzanich V, Sheth KN, Kimberly WT & Simard JM Emerging Pharmacological Treatments for Cerebral Edema: Evidence from Clinical Studies. Annu. Rev. Pharmacol. Toxicol 60, 291–309 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kochanek KD, Xu J, Murphy SL, Miniño AM & Kung H-C Deaths: final data for 2009. Natl. Vital Stat. Rep 60, 1–116 (2011). [PubMed] [Google Scholar]
- 3.Mokri B The Monro-Kellie hypothesis: Applications in CSF volume depletion. Neurology 56, 1746–1748 (2001). [DOI] [PubMed] [Google Scholar]
- 4.King ZA, Sheth KN, Kimberly WT & Simard JM Profile of intravenous glyburide for the prevention of cerebral edema following large hemispheric infarction: evidence to date. Drug Des. Devel. Ther Volume 12, 2539–2552 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Important review of the basic science and clinical trials showing promise of SUR1-TRPM4 inhibiton through repurposing of the drug glyburide.
- 5.Shah S & Kimberly WT Today’s Approach to Treating Brain Swelling in the Neuro Intensive Care Unit. Semin. Neurol 36, 502–507 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Stokum JA, Gerzanich V & Simard JM Molecular pathophysiology of cerebral edema. J. Cereb. Blood Flow Metab 36, 513–538 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]; **Excellent review of the current understanding of the molecular and cellular mechanisms of cerebral edema.
- 7.Koenig MA Cerebral Edema and Elevated Intracranial Pressure. Contin. Lifelong Learn. Neurol 24, 1588–1602 (2018). [DOI] [PubMed] [Google Scholar]
- 8.Huang H et al. The WNK-SPAK/OSR1 Kinases and the Cation-Chloride Cotransporters as Therapeutic Targets for Neurological Diseases. Aging Dis. 10, 626 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhang J et al. Modulation of brain cation-Cl− cotransport via the SPAK kinase inhibitor ZT-1a. Nat. Commun 11, 78 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; **Important primary research article demonstrating a promising new target to attenuate cerebral edema and improve neurological outcomes, specifically by inhibiton of cation-chloride transporters; it also highlights the importance of kinome profiling and use of chemical libraries to maximize our understanding and discovery of pharmacological targets and develop useful therapies.
- 10.Mehta RI et al. Sur1-Trpm4 Cation Channel Expression in Human Cerebral Infarcts. J. Neuropathol. Exp. Neurol 74, 835–849 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mehta RI et al. Sulfonylurea Receptor 1 Expression in Human Cerebral Infarcts. J. Neuropathol. Exp. Neurol 72, 871–883 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Simard JM et al. Newly expressed SUR1-regulated NCCa-ATP channel mediates cerebral edema after ischemic stroke. Nat. Med 12, 433–440 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Simard JM et al. Glibenclamide Is Superior to Decompressive Craniectomy in a Rat Model of Malignant Stroke. Stroke 41, 531–537 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kurland DB et al. The Sur1-Trpm4 channel regulates NOS2 transcription in TLR4-activated microglia. J. Neuroinflammation 13, 130 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Loh KP et al. TRPM4 inhibition promotes angiogenesis after ischemic stroke. Pflügers Arch. - Eur. J. Physiol 466, 563–576 (2014). [DOI] [PubMed] [Google Scholar]
- 16.Jain RK Tumor angiogenesis and accessibility: Role of vascular endothelial growth factor. Semin. Oncol 29, asonc02906q0003 (2002). [DOI] [PubMed] [Google Scholar]
- 17.Dvorak HF Vascular Permeability Factor/Vascular Endothelial Growth Factor: A Critical Cytokine in Tumor Angiogenesis and a Potential Target for Diagnosis and Therapy. J. Clin. Oncol 20, 4368–4380 (2002). [DOI] [PubMed] [Google Scholar]
- 18.Folkins C et al. Anticancer Therapies Combining Antiangiogenic and Tumor Cell Cytotoxic Effects Reduce the Tumor Stem-Like Cell Fraction in Glioma Xenograft Tumors. Cancer Res. 67, 3560–3564 (2007). [DOI] [PubMed] [Google Scholar]
- 19.Gerstner ER et al. VEGF inhibitors in the treatment of cerebral edema in patients with brain cancer. Nat. Rev. Clin. Oncol 6, 229–36 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chinot OL et al. Bevacizumab plus Radiotherapy–Temozolomide for Newly Diagnosed Glioblastoma. N. Engl. J. Med 370, 709–722 (2014). [DOI] [PubMed] [Google Scholar]
- 21.Batchelor TT et al. AZD2171, a Pan-VEGF Receptor Tyrosine Kinase Inhibitor, Normalizes Tumor Vasculature and Alleviates Edema in Glioblastoma Patients. Cancer Cell 11, 83–95 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kamoun WS et al. Edema Control by Cediranib, a Vascular Endothelial Growth Factor Receptor–Targeted Kinase Inhibitor, Prolongs Survival Despite Persistent Brain Tumor Growth in Mice. J. Clin. Oncol 27, 2542–2552 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Reeson P et al. Delayed Inhibition of VEGF Signaling after Stroke Attenuates Blood-Brain Barrier Breakdown and Improves Functional Recovery in a Comorbidity-Dependent Manner. J. Neurosci 35, 5128–5143 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Greenberg DA & Jin K Vascular endothelial growth factors (VEGFs) and stroke. Cell. Mol. Life Sci 70, 1753–1761 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Abrams DA et al. Timing of surgery and bevacizumab therapy in neurosurgical patients with recurrent high grade glioma. J. Clin. Neurosci 22, 35–39 (2015). [DOI] [PubMed] [Google Scholar]
- 26.Barreca T et al. Evaluation of the Secretory Pattern of Plasma Arginine Vasopressin in Stroke Patients. Cerebrovasc. Dis 11, 113–118 (2001). [DOI] [PubMed] [Google Scholar]
- 27.Cui H et al. Inappropriate Antidiuretic Hormone Secretion and Cerebral Salt-Wasting Syndromes in Neurological Patients. Front. Neurosci 13, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Can B, Oz S, Sahinturk V, Musmul A & Alatas İO Effects of Conivaptan versus Mannitol on Post-Ischemic Brain Injury and Edema. Eurasian J. Med 51, 42–48 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ghali J Conivaptan and its role in the treatment of hyponatremia. Drug Des. Devel. Ther 253 (2009) doi: 10.2147/DDDT.S4505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen C COX-2’s new role in inflammation. Nat. Chem. Biol 6, 401–402 (2010). [DOI] [PubMed] [Google Scholar]
- 31.Chu K et al. Celecoxib Induces Functional Recovery after Intracerebral Hemorrhage with Reduction of Brain Edema and Perihematomal Cell Death. J. Cereb. Blood Flow Metab 24, 926–933 (2004). [DOI] [PubMed] [Google Scholar]
- 32.Park H-K, Lee S-H, Chu K & Roh J-K Effects of celecoxib on volumes of hematoma and edema in patients with primary intracerebral hemorrhage. J. Neurol. Sci 279, 43–46 (2009). [DOI] [PubMed] [Google Scholar]
- 33.Lee S-H et al. Effects of celecoxib on hematoma and edema volumes in primary intracerebral hemorrhage: a multicenter randomized controlled trial. Eur. J. Neurol 20, 1161–1169 (2013). [DOI] [PubMed] [Google Scholar]
- 34.Hannun YA & Obeid LM Principles of bioactive lipid signalling: lessons from sphingolipids. Nat. Rev. Mol. Cell Biol 9, 139–150 (2008). [DOI] [PubMed] [Google Scholar]
- 35.Salas-Perdomo A et al. Role of the S1P pathway and inhibition by fingolimod in preventing hemorrhagic transformation after stroke. Sci. Rep 9, 8309 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sanchez T et al. Induction of Vascular Permeability by the Sphingosine-1-Phosphate Receptor–2 (S1P2R) and its Downstream Effectors ROCK and PTEN. Arterioscler. Thromb. Vasc. Biol 27, 1312–1318 (2007). [DOI] [PubMed] [Google Scholar]
- 37.Wei Y et al. Fingolimod provides long-term protection in rodent models of cerebral ischemia. Ann. Neurol 69, 119–129 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sehr T, Akgün K, Haase R & Ziemssen T Fingolimod Leads to Immediate Immunological Changes Within 6 h After First Administration. Front. Neurol 11, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Takahashi K Effect of dosage reduction on peripheral blood lymphocyte count in patients with multiple sclerosis receiving long-term fingolimod therapy. J. Clin. Neurosci 63, 91–94 (2019). [DOI] [PubMed] [Google Scholar]
- 40.Murayi R & Chittiboina P Glucocorticoids in the management of peritumoral brain edema: a review of molecular mechanisms. Child’s Nerv. Syst 32, 2293–2302 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tjuvajev J et al. Corticotropin-releasing factor decreases vasogenic brain edema. Cancer Res. (1996). [PubMed] [Google Scholar]
- 42.Villalona-Calero MA et al. A phase I trial of human corticotropin-releasing factor (hCRF) in patients with peritumoral brain edema. Ann. Oncol 9, 71–77 (1998). [DOI] [PubMed] [Google Scholar]
- 43.Recht L, Mechtler LL, Wong ET, O’Connor PC & Rodda BE Steroid-Sparing Effect of Corticorelin Acetate in Peritumoral Cerebral Edema Is Associated With Improvement in Steroid-Induced Myopathy. J. Clin. Oncol 31, 1182–1187 (2013). [DOI] [PubMed] [Google Scholar]
- 44.Kitchen P et al. Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. Cell 181, 784–799.e19 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Arvold ND et al. Corticosteroid use endpoints in neuro-oncology: Response Assessment in Neuro-Oncology Working Group. Neuro. Oncol 20, 897–906 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]