Stroke remains one of the leading causes of mortality and disability worldwide, with ischemic stroke constituting the majority of cases. Despite significant advances in our understanding of its pathophysiology and the development of acute interventions, stroke continues to pose substantial health and socioeconomic burdens globally. This comprehensive review examines the fundamental pathophysiological mechanisms underlying different stroke types, explores the cellular and molecular pathways involved in stroke progression, and evaluates the current landscape of evidence-based interventions versus those that lack robust clinical validation. The pathophysiology of stroke involves complex cascades of excitotoxicity, oxidative stress, and inflammatory processes that ultimately lead to neuronal death and functional impairment. While some interventions like tissue plasminogen activator (tPA) and endovascular thrombectomy have shown significant efficacy for ischemic stroke, many neuroprotective agents that showed promise in preclinical studies have failed to translate into clinical success. The emerging approaches in stroke management aim to target multiple phases of the ischemic cascade, emphasizing prevention of core expansion, optimization of reperfusion, and mitigation of secondary injury.
Stroke Definition and Classification
Stroke is characterized by a sudden disruption of blood flow to the brain, resulting in neurological deficits and potential long-term disability. It is generally accepted as the leading cause of adult disability globally, with profound health and socioeconomic implications1. Stroke can be broadly classified into two major types: ischemic stroke, which accounts for approximately 85% of all cases, and hemorrhagic stroke, which comprises the remaining 15%48. Ischemic stroke occurs when blood flow to a region of the brain is interrupted due to vessel occlusion, while hemorrhagic stroke results from the rupture of cerebral blood vessels, leading to bleeding into the brain parenchyma or surrounding spaces1012.
The etiology of ischemic stroke varies, including thrombotic, embolic, and hypoperfusion mechanisms, each with distinct pathophysiological characteristics. Thrombotic stroke results from local formation of a clot within a cerebral blood vessel, often due to atherosclerosis. Embolic stroke, in contrast, occurs when a clot forms elsewhere in the body and travels to the cerebral circulation. Hypoperfusion stroke develops when systemic circulation fails to provide adequate blood flow to the brain, commonly seen during cardiac arrest or severe hypotension48. Hemorrhagic stroke encompasses intracerebral hemorrhage (ICH), where bleeding occurs directly into the brain tissue, and subarachnoid hemorrhage (SAH), where bleeding occurs in the space between the brain and the surrounding membranes210.
The distinction between these stroke types is crucial not only for understanding their underlying pathophysiology but also for determining appropriate therapeutic interventions. While ischemic stroke management focuses on restoring blood flow and salvaging vulnerable tissue, hemorrhagic stroke treatment aims to control bleeding, reduce intracranial pressure, and prevent secondary injury cascades212.
Pathophysiology of Ischemic Stroke
Ischemic stroke initiates a complex cascade of pathophysiological events triggered by the reduction or complete cessation of cerebral blood flow. This reduction leads to a critical shortage of oxygen and glucose, essential substrates for brain metabolism, resulting in a sequence of cellular and molecular changes that ultimately lead to neuronal injury and death14. The brain's high metabolic demands make it particularly vulnerable to ischemic insults, with irreversible damage occurring within minutes in severely affected regions.
The ischemic cascade begins with energy depletion, manifested by a dramatic reduction in adenosine triphosphate (ATP) production. This energy failure leads to the dysfunction of energy-dependent membrane ion pumps, particularly the Na⁺/K⁺-ATPase, resulting in membrane depolarization4. The subsequent influx of calcium into neurons triggers the excessive release of excitatory neurotransmitters, primarily glutamate, initiating a process known as excitotoxicity418. Glutamate binds to N-methyl-D-aspartate (NMDA) receptors and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, causing further calcium influx, which activates various calcium-dependent enzymes that degrade essential cellular components18.
Concurrent with excitotoxicity, oxidative stress emerges as a significant contributor to ischemic injury. The production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) overwhelms the brain's antioxidant defense mechanisms, leading to oxidative damage to proteins, lipids, and nucleic acids4. Mitochondrial dysfunction further exacerbates this oxidative stress, creating a vicious cycle of cellular damage4. The increased production of free radicals also activates inflammatory pathways, attracting leukocytes to the site of injury and promoting the release of pro-inflammatory cytokines and chemokines8.
Blood-brain barrier (BBB) disruption represents another critical aspect of ischemic stroke pathophysiology. The BBB, a specialized interface between the cerebral circulation and the central nervous system, undergoes a multiphasic pattern of permeability changes throughout the different stages of stroke6. In the hyperacute stage, sudden hypoxia damages the BBB, leading to cytotoxic edema and increased permeability. In the acute stage, neuroinflammatory responses further compromise BBB integrity, heightening the risk of hemorrhagic transformation, particularly following reperfusion therapy6. In the subacute stage (1-3 weeks), repair mechanisms, especially neoangiogenesis, commence, though the newly formed vessels exhibit leaky characteristics. Interestingly, this controlled permeability has been associated with improved clinical recovery6.
Cerebral edema emerges as a serious complication of ischemic stroke, contributing significantly to morbidity and mortality. Two primary forms of edema develop: cytotoxic edema, resulting from cellular swelling due to ion pump failure, and vasogenic edema, stemming from BBB disruption and the extravasation of fluid into the extracellular space13. The development of cerebral edema increases intracranial pressure, potentially leading to herniation and secondary injury to previously unaffected brain regions13.
Pathophysiology of Hemorrhagic Stroke
Hemorrhagic stroke encompasses intracerebral hemorrhage (ICH) and subarachnoid hemorrhage (SAH), both characterized by the rapid accumulation of blood within or surrounding the brain tissue. The pathophysiology of hemorrhagic stroke differs significantly from ischemic stroke, though some overlapping mechanisms exist, particularly in the secondary injury phase212. This distinct pathophysiology necessitates different therapeutic approaches and has implications for clinical outcomes.
The initial insult in hemorrhagic stroke involves the rupture of cerebral blood vessels, which may result from various etiologies, including hypertension, cerebral amyloid angiopathy, arteriovenous malformations, aneurysms, or coagulopathies1012. The sudden accumulation of blood creates a mass effect, directly compressing adjacent brain tissue and potentially obstructing cerebrospinal fluid circulation, leading to increased intracranial pressure12. This primary mechanical injury is followed by a cascade of secondary injury mechanisms that evolve over hours to days.
Inflammatory responses play a central role in the pathophysiology of hemorrhagic stroke. The extravasated blood triggers a robust inflammatory reaction involving both resident brain cells and infiltrating immune cells1014. Microglia, the brain's primary immune cells, become activated and release cytokines and chemokines, contributing to neuroinflammation14. The NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome, predominantly released by microglia, has been identified as a major contributor to neuroinflammation following hemorrhagic stroke14. This inflammasome activation leads to the production of pro-inflammatory cytokines, particularly interleukin-1β (IL-1β) and IL-18, exacerbating tissue damage14.
Blood components, particularly hemoglobin and its breakdown products, initiate several pathophysiological processes. As red blood cells lyse, hemoglobin is released and subsequently broken down into heme and iron, potent neurotoxins that induce oxidative stress and cellular damage12. Iron-induced injury contributes significantly to secondary neuronal death through lipid peroxidation, DNA damage, and disruption of the BBB12. Moreover, thrombin, a serine protease present in extravasated blood, activates the coagulation cascade and stimulates protease-activated receptors on various cell types, triggering inflammatory responses and potentially contributing to edema formation12.
Programmed cell death pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis, have been implicated in the pathophysiology of hemorrhagic stroke17. The P2X7 receptor, an ATP-gated non-selective cation channel, plays a pivotal role in mediating programmed cell death following hemorrhagic stroke17. Activation of this receptor can regulate cell death through multiple mechanisms, including exocytosis of secretory lysosomes, exocytosis of autophagosomes or autophagolysosomes, and direct release of cytosolic IL-1β secondary to regulated cell death by pyroptosis or necroptosis17. Genetic deletion and pharmacological blockade of the P2X7 receptor have shown neuroprotective effects in experimental models of hemorrhagic stroke, highlighting its potential as a therapeutic target17.
Cerebral edema development following hemorrhagic stroke shares some similarities with ischemic stroke but also exhibits distinct features. Perihematomal edema evolves through several phases, influenced by thrombin activation, inflammatory mediators, hemoglobin breakdown products, and BBB disruption13. This edema contributes to increased intracranial pressure and neurological deterioration, representing a significant cause of morbidity and mortality13.
Evidence-Based Interventions for Stroke
The management of stroke has evolved significantly over recent decades, with several interventions demonstrating robust evidence of efficacy. For ischemic stroke, the therapeutic focus centers on rapid restoration of cerebral blood flow to salvage vulnerable tissue in the penumbra, the region surrounding the irreversibly damaged core that remains potentially salvageable415. This approach is encapsulated by the adage "time is brain," emphasizing the critical importance of prompt intervention.
Intravenous thrombolysis with tissue plasminogen activator (tPA) represents the cornerstone of acute ischemic stroke treatment. Administered within a therapeutic window (traditionally 4.5 hours from symptom onset, though newer evidence supports extended windows in selected patients), tPA dissolves the occluding clot and restores blood flow4. Despite its efficacy, tPA has significant limitations, including the narrow therapeutic window, risk of hemorrhagic transformation, and contraindications in patients with recent surgery, coagulopathies, or prior stroke34. Moreover, tPA is less effective for large vessel occlusions, prompting the development of mechanical thrombectomy.
Endovascular thrombectomy has emerged as a revolutionary intervention for acute ischemic stroke caused by large vessel occlusion. Multiple pivotal clinical trials have demonstrated its substantial benefit when performed within 6-24 hours of symptom onset in selected patients15. Thrombectomy achieves higher rates of recanalization than intravenous thrombolysis alone, particularly for proximal occlusions, and has expanded the therapeutic window for eligible patients15. The success of thrombectomy has shifted the paradigm of stroke management, emphasizing the importance of rapid triage and transfer to comprehensive stroke centers capable of performing the procedure.
For hemorrhagic stroke, evidence-based interventions focus on preventing hematoma expansion, controlling intracranial pressure, and minimizing secondary injury12. Hemostatic therapy, particularly with recombinant activated factor VII, has shown promise in limiting hematoma growth in selected patients, though its overall benefit remains controversial12. Blood pressure management represents another crucial aspect of hemorrhagic stroke care, with guidelines recommending careful reduction of severely elevated blood pressure to balance the risks of ongoing bleeding against those of cerebral hypoperfusion12.
Surgical interventions for hemorrhagic stroke have evolved substantially, with a shift toward minimally invasive approaches. Traditional craniotomy for clot evacuation has shown limited benefit in unselected patients, while minimally invasive surgery with stereotactic or endoscopic techniques has demonstrated more promising results in recent trials12. For subarachnoid hemorrhage caused by aneurysmal rupture, early securing of the aneurysm through either endovascular coiling or surgical clipping is recommended to prevent rebleeding2.
The concept of "multiphase adjuvant neuroprotection" has emerged as a promising paradigm for enhancing outcomes in stroke patients undergoing reperfusion therapies15. This approach recognizes the diverse and potentially non-overlapping pathophysiological mechanisms triggered before, during, and after intervention. Before thrombectomy, strategies focus on preventing the growth of the ischemic core; during thrombectomy, efforts aim to improve recanalization while reducing distal embolization and maximizing reperfusion; after reperfusion, interventions target the reduction of secondary reperfusion injury15. This multi-targeted approach seeks to minimize final infarct size and improve functional outcomes in patients treated with thrombectomy.
Controversial and Emerging Therapies
Despite significant advances in stroke management, many promising interventions have failed to demonstrate efficacy in clinical trials, highlighting the challenges in translating preclinical findings to patient care. The quest for effective neuroprotective agents, in particular, has been marked by numerous disappointments, with over 1,000 agents showing promise in experimental models but failing in clinical trials7. This bench-to-bedside translation gap has prompted a reevaluation of research methodologies and a search for novel therapeutic targets.
Neuroprotective strategies aim to interrupt the ischemic cascade, preserving vulnerable neurons in the penumbra. Despite robust preclinical evidence, prominent examples of substances that failed in clinical trials include tirilazad and disodium 2,4-sulphophenyl-N-tert-butylnitrone (NXY-059)7. The NXY-059 trial, initially meeting the recommendations of the Stroke Therapy Academic Industry Roundtable (STAIR) committee, was retrospectively shown to have serious flaws in study design, quality, and statistical analysis7. These failures have led to more rigorous criteria for preclinical evidence and improved clinical trial design, with current trials such as ESCAPE-NA implementing lessons learned from past disappointments7.
Hyperbaric oxygen therapy (HBOT) represents another intervention with promising preclinical data but limited clinical validation. HBOT has demonstrated significant pre-clinical effectiveness for acute ischemic stroke, with reported benefits including reductions in oxidative stress, inflammation, neural apoptosis, and improvements in physiological parameters such as edema and oxygen perfusion11. However, clinical translation has been hampered by inconsistencies in treatment protocols and study design. While the medical community works to standardize clinical protocols for HBOT in acute stroke, pre-clinical investigations continue to explore novel applications, including HBOT preconditioning based on the premise of mild oxidative stress priming the brain to tolerate the full-blown oxidative stress inherent in stroke11.
Stem cell therapies have emerged as a promising avenue for stroke recovery, targeting the chronic phase where few effective interventions exist. Various stem cell types, including mesenchymal stem cells, neural stem cells, and induced pluripotent stem cells, have shown potential to enhance recovery through multiple mechanisms, including secretion of trophic factors, modulation of inflammation, promotion of angiogenesis, and neurogenesis4. Despite encouraging results in preclinical studies and early-phase clinical trials, definitive evidence of efficacy from large randomized controlled trials remains pending4. Questions regarding optimal cell type, delivery route, timing, and dose continue to be investigated.
Triple neuroprotection (TNP), encompassing targeted hypothermia, controlled induced hypertension, and barbiturate infusion, represents an innovative approach for patients with acute stroke who have failed tPA or present beyond the therapeutic window3. This strategy has been employed during emergency carotid endarterectomy with promising results in selected cases3. A pilot protocol demonstrated favorable clinical outcomes in patients despite being in therapeutic inertia with significant carotid stenosis3. While this approach shows promise, larger studies are needed to establish its efficacy and safety.
The management of stroke in the context of comorbidities presents additional challenges and opportunities. Cancer-associated stroke, for instance, involves unique pathophysiological mechanisms, including direct cancer effects and coagulation disorders establishing a state of hypercoagulation19. Treatment approaches must balance the risk of hemorrhage against the need for anticoagulation, with low-molecular-weight heparin remaining the first choice despite the promise of new oral anticoagulants19. Similarly, coronavirus disease-2019 (COVID-19) has been associated with an increased risk for both ischemic and hemorrhagic stroke, necessitating tailored management strategies that address the virus-specific mechanisms16.
Current Challenges and Future Directions
The field of stroke research and management faces several persistent challenges that influence both therapeutic development and clinical outcomes. Understanding and addressing these challenges is crucial for advancing the field and improving patient care. Simultaneously, emerging approaches and technologies offer promising directions for future research and intervention strategies.
A significant challenge in stroke management is the limited therapeutic window for current interventions. Despite advances in extending eligibility criteria for reperfusion therapies, many patients still present beyond the established time frames or have contraindications to these treatments415. This limitation underscores the need for interventions effective in delayed presentations and alternative approaches for patients ineligible for current therapies. Additionally, the heterogeneity of stroke presentations and underlying pathophysiology complicates therapeutic development, as interventions successful in specific stroke subtypes may not generalize to the broader stroke population8.
The translation gap between preclinical promise and clinical efficacy represents another substantial challenge. Many interventions demonstrating robust effects in animal models fail to show benefit in human trials7. This discrepancy has prompted a reevaluation of preclinical models and methodologies, with increased emphasis on replicating the complexity of human stroke, including comorbidities, age-related factors, and genetic diversity78. Improved alignment between preclinical studies and clinical trial designs, along with more rigorous reporting standards, may enhance translational success rates.
Access to advanced stroke care remains uneven globally, with significant disparities in stroke outcomes based on geographical location, socioeconomic status, and healthcare system resources8. Strategies to address these disparities include telemedicine initiatives, development of stroke care networks, education programs for healthcare providers, and simplified protocols that can be implemented in resource-limited settings8. The importance of stroke prevention through management of modifiable risk factors also cannot be overstated, as it represents the most cost-effective approach to reducing the global burden of stroke8.
Looking forward, several promising directions merit exploration. The concept of multimodal imaging for patient selection has already enhanced the precision of reperfusion therapy decisions and may further refine patient stratification for experimental therapies415. Advanced technologies, including artificial intelligence, hold potential for improving diagnostic accuracy, predicting outcomes, and personalizing treatment regimens based on individual patient characteristics8.
Combinatorial approaches that target multiple aspects of the ischemic cascade simultaneously may overcome the limitations of single-target interventions. The multiphase adjuvant neuroprotection paradigm exemplifies this strategy, recognizing the diverse mechanisms operating at different stages of stroke progression15. Similarly, combining established reperfusion methods with neuroprotective agents, anti-inflammatory interventions, or regenerative therapies may yield synergistic benefits415.
Conclusion
Stroke continues to pose significant challenges to global health, despite substantial progress in understanding its pathophysiology and developing interventions. The complex cascade of events following ischemic or hemorrhagic insults to the brain involves multiple interacting pathways, including excitotoxicity, oxidative stress, inflammation, blood-brain barrier disruption, and programmed cell death. This complexity has both impeded therapeutic development and offered numerous potential targets for intervention.
Current evidence-based interventions, notably intravenous thrombolysis and endovascular thrombectomy for ischemic stroke, have revolutionized acute stroke management, significantly improving outcomes for eligible patients. However, their limitations, including narrow therapeutic windows and selection criteria, leave many patients without effective treatment options. For hemorrhagic stroke, management strategies focusing on hemostasis, blood pressure control, and surgical evacuation have shown varying degrees of efficacy, highlighting the need for further refinement and novel approaches.
The path forward requires addressing persistent challenges, including the translation gap between preclinical studies and clinical trials, access disparities to advanced stroke care, and the heterogeneity of stroke presentations. Promising directions include multimodal imaging for enhanced patient selection, artificial intelligence applications, combinatorial therapeutic approaches, and the multiphase adjuvant neuroprotection paradigm. These strategies, coupled with continued emphasis on stroke prevention and public education, offer hope for reducing the global burden of this devastating disease.
As research advances and our understanding of stroke pathophysiology deepens, the integration of personalized medicine approaches, recognizing individual genetic, demographic, and comorbidity profiles, may further optimize stroke prevention, acute intervention, and recovery strategies. By building on established evidence while embracing innovation, the stroke community can continue to make strides toward improving outcomes for the millions affected by this condition worldwide.
Citations:
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10968326/
- https://pubmed.ncbi.nlm.nih.gov/19026899/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7607101/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497522/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10241149/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7756029/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9140731/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7589849/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9978764/
- https://pubmed.ncbi.nlm.nih.gov/36601948/
- https://pubmed.ncbi.nlm.nih.gov/30079404/
- https://pubmed.ncbi.nlm.nih.gov/35420918/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9434007/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6712291/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7045534/
- https://pubmed.ncbi.nlm.nih.gov/35762309/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6251042/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5851248/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6365034/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5809704/
- https://pubmed.ncbi.nlm.nih.gov/28830757/
- https://pubmed.ncbi.nlm.nih.gov/22463492/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8636795/
- https://pubmed.ncbi.nlm.nih.gov/33260055/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11324503/
- https://www.semanticscholar.org/paper/0acfa0e1e1a8887b559450d95cc01520ec805609
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5903066/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11721500/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11640599/
- https://pubmed.ncbi.nlm.nih.gov/33772503/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7059570/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10414072/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10472948/
- https://pubmed.ncbi.nlm.nih.gov/38051175/
- https://pubmed.ncbi.nlm.nih.gov/36529857/
- https://pubmed.ncbi.nlm.nih.gov/28792246/
- https://www.semanticscholar.org/paper/276ec52f1e92cfe83235608f9cbb0fe1de77a339
- https://pubmed.ncbi.nlm.nih.gov/36050815/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9940155/
- https://pubmed.ncbi.nlm.nih.gov/35524026/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8586130/
- https://www.semanticscholar.org/paper/73384f9ab790eaeba4a449088a738baf56d95667
- https://www.semanticscholar.org/paper/c0f299eda459399227702d224a43b9bb1b61eb49
- https://pubmed.ncbi.nlm.nih.gov/27896592/
- https://www.semanticscholar.org/paper/985dd2e319ccc3568118ffad20fd0f4836925850
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11408949/
- https://pubmed.ncbi.nlm.nih.gov/38290814/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9738261/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10483231/