International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cardiopulmonary resuscitation (CPR) is a life-saving emergency technique used after cardiac arrest to restore spontaneous circulation and maintain oxygenated blood flow to vital organs, particularly the brain and heart (1). It involves chest compressions and artificial ventilation; compressions simulate the heart's pumping action, while ventilation supplies oxygen to the lungs (2).
Despite its importance, CPR faces challenges. Survival rates for out-of-hospital cardiac arrests remain low, with only 5–10% of patients surviving to hospital discharge (3). Several factors affect outcomes, including delays in CPR initiation, the quality of compressions, the cause of the arrest and reversible conditions such as pulmonary embolism or myocardial infarction (4,5). Inadequate advanced interventions and post-resuscitation care also reduce success rates, driving ongoing efforts to enhance CPR effectiveness and neurological outcomes (6).
Thrombolytic therapy, or fibrinolysis, uses drugs to dissolve clots that block critical blood vessels (7). In cardiac arrest, thrombolysis targets clots contributing to circulatory failure, such as those from myocardial infarction or pulmonary embolism (8). This strategy is well-established in acute myocardial infarction and ischemic stroke, where prompt clot dissolution improves perfusion and limits tissue damage (9). Applying thrombolytics during CPR is based on the premise that thrombotic occlusions may trigger or worsen cardiac arrest (10). By incorporating thrombolytic agents into CPR protocols, blood flow is considered to be more effectively restored, thereby enhancing the chances of successful resuscitation and improving overall outcomes (8). However, this approach carries bleeding risks, so its use requires careful risk-benefit assessment.
Recombinant tissue plasminogen activator (rtPA) is a bioengineered version of a naturally occurring enzyme that plays a critical role in the breakdown of blood clots. rtPA converts plasminogen, a blood protein, into plasmin, an enzyme that dissolves fibrin clots (11). Due to its targeted action on fibrin, rtPA is highly effective in dissolving clots while minimizing systemic effects (12). In the context of thrombolytic therapy for CPR, rtPA is considered one of the most promising agents. Its efficacy has been well-documented in the treatment of acute ischemic stroke and myocardial infarction, leading researchers to explore its potential benefits in cardiac arrest scenarios (13). rtPA presents advantages over conventional TPA, including a more favorable pharmacokinetic profile that enables faster thrombolysis, which is crucial in cardiac arrest. Additionally, rtPA has a lower risk of immunogenic reactions, enhancing its safety (14). The administration of rtPA during CPR could potentially address the thrombotic component of certain cardiac arrests, thus enhancing the likelihood of restoring spontaneous circulation (15). The present review aims to critically analyze the efficacy of thrombolytic therapy, specifically focusing on the role of rtPA in CPR. The present review evaluated the current evidence surrounding rtPA administration during CPR, considering its effect on patient outcomes, such as survival rates, neurological function and long-term prognosis. By synthesizing available research findings the present review provided insights into the potential benefits, limitations and optimal protocols for implementing rtPA in CPR protocols, ultimately contributing to informed decision-making and advancements in resuscitative care strategies.
The fibrinolytic pathway is a crucial biological mechanism responsible for the breakdown of thrombi. This pathway removes unnecessary or harmful clots, maintaining vascular patency and normal blood flow, but it can also disrupt normal hemostasis, increasing the risk of bleeding (16). The process begins with activating plasminogen, a precursor glycoprotein converted into its active form, plasmin (17). Plasmin is a serine protease that digests fibrin, the primary protein constituent of blood clots (18) (Fig. 1). The fibrinolytic system can be activated via two pathways: The intrinsic pathway, initiated by contact with negatively charged surfaces, and the extrinsic pathway, which is triggered by tissue plasminogen activator (tPA) or urokinase plasminogen activator (uPA) released from endothelial cells in response to injury or stress (17,19). In considering thrombolytic therapy, a comparative analysis of tPA and uPA is crucial. tPA is widely preferred for its rapid action and specificity for fibrin-bound plasminogen, making it effective in acute ischemic stroke and myocardial infarction. However, its use may be restricted by bleeding risks and contraindications (20,21). Conversely, uPA, while less commonly used, offers advantages in patients with higher bleeding risk or contraindications to tPA and it can be effective in various thrombotic conditions, including pulmonary embolism. A thorough comparison of these agents can guide clinicians in making informed decisions tailored to individual patient needs and specific clinical scenarios (22).
The use of tPA and uPA carries inherent bleeding risks and specific contraindications. For tPA, the reported risk of symptomatic intracranial hemorrhage (ICH) is ~6.4% when administered for acute ischemic stroke, with overall bleeding risks reaching 10–15%. Contraindications for tPA include recent intracranial hemorrhage, active bleeding, significant head trauma, major surgery within the past three months and severe uncontrolled hypertension (23). By contrast, uPA is associated with a lower overall bleeding risk, estimated at 1–2% for major events, although its specific risk profile is less characterized compared with tPA (24). Regarding efficacy, tPA has demonstrated 30–35% effectiveness in achieving functional independence in ischemic stroke patients and is effective in myocardial infarction and pulmonary embolism (25). In comparison, uPA has shown variable efficacy rates, typically lower than tPA and is not as widely accepted in acute stroke or myocardial infarction. tPA has high fibrin specificity, a half-life of <5 min and is administered via bolus and infusion over 90 min, while uPA lacks fibrin specificity, has a half-life of 10–20 min and is given as an infusion. The two agents have similar ICH risks, but tPA shows higher rates of recanalization and improved long-term recovery outcomes (26). In terms of roles in stroke, tPA is critical for acute thrombolysis, affecting vascular permeability and inflammation, while uPA promotes synaptic recovery during the recovery phase. tPA disrupts the blood-brain barrier, increasing permeability and leukocyte infiltration, whereas uPA may enhance neovascularization (27). Overall, tPA can enhance synaptic plasticity but may induce excitotoxicity, while uPA supports neuronal recovery and synaptic repair. Understanding these parameters is crucial for optimizing thrombolytic therapy and managing associated risks effectively.
rtPA is a synthetic form of the naturally occurring tPA designed for therapeutic use to enhance clot dissolution (28). rtPA functions by binding to fibrin within the clot and converting the adjacent plasminogen to plasmin (29). This localized conversion is critical as it ensures that plasmin is generated primarily at the site of the thrombus, minimizing systemic activation, which could lead to widespread bleeding complications (30,31). Once administered, rtPA enhances the conversion efficiency of plasminogen to plasmin, amplifying the fibrinolytic activity at the site of the thrombus. This increase in plasmin leads to the more rapid and effective breakdown of fibrin, the structural framework of the clot. By dissolving the fibrin mesh, rtPA facilitates the disintegration of the thrombus, thereby restoring blood flow through the occluded vessel (32).
In comparing rTPA with conventional plasminogen activators such as TPA, several key differences emerge that highlight the advantages of rTPA in clinical practice. rTPA specifically targets fibrin-bound plasminogen, which enhances fibrinolysis directly at the site of the clot. This targeted action increases its effectiveness in dissolving thrombi, a critical factor in acute ischemic events (33). Furthermore, rTPA demonstrates a more rapid onset of action compared with conventional TPA, allowing for quicker therapeutic effects in urgent situations. This rapid response is crucial for improving patient outcomes in acute stroke management. The pharmacological profile of rTPA also emphasizes its preferential action on clot-bound fibrin, which markedly boosts its efficacy in thrombus dissolution (34). The safety profile of rTPA is more thoroughly characterized in the literature, with studies indicating a clearer understanding of its potential adverse drug reactions (35). Notably, while both agents carry a risk of bleeding, the specific safety data of rTPA provide clinicians with more reliable information for risk assessment. Additionally, rTPA has a shorter half-life, which is beneficial for managing acute ischemic events, as it allows for rapid clinical adjustments based on the patient's response (36). These distinctions not only elucidate the pharmacological advantages of rTPA but also underscore its role in improving therapeutic outcomes in patients requiring acute thrombolytic therapy (37).
Thrombolytic therapy, particularly with agents such as rtPA, holds significant potential benefits during CPR. Thrombotic occlusions during cardiac arrest can impair coronary and pulmonary circulation, worsening myocardial and cerebral ischemia and lowering resuscitation success rates. By dissolving these clots, thrombolytic therapy can restore blood flow and improve oxygen delivery to vital organs (38). In acute myocardial infarction or massive pulmonary embolism (PE)-induced arrest, rtPA can rapidly dissolve the causative thrombi, reestablish coronary perfusion and potentially convert non-perfusing rhythms, such as ventricular fibrillation or pulseless electrical activity, into perfusing ones (39,40). Similarly, in PE, thrombolysis can alleviate the obstruction in the pulmonary arteries, reduce right ventricular strain and enhance cardiac output. Restoring coronary and pulmonary circulation during CPR can improve resuscitation efficacy, stabilize hemodynamics and increase return of spontaneous circulation (ROSC) rates (41). Reducing ischemic time may also lessen post-resuscitation myocardial dysfunction and limit ischemic damage to the heart and other organs (42,43). Consequently, integrating thrombolytic therapy into CPR protocols, especially in cases where thrombotic occlusions are suspected, can be a crucial strategy to improve survival outcomes and neurological recovery in cardiac arrest patients (44).
Diagnosing PE and ST-elevation myocardial infarction (STEMI) during CPR poses significant challenges for clinicians. Rapid decision-making is crucial, often relying on clinical history, symptoms and immediate ECG findings. In STEMI, chest pain and ST-segment elevation guide diagnosis, while PE may present with sudden dyspnea and hemodynamic instability (45). To confirm thrombotic causes quickly, point-of-care ultrasound and imaging techniques such as computed tomography pulmonary angiography are helpful, though not always available during CPR. Clinicians may also use risk stratification tools, such as the Wells score for PE and the TIMI score for STEMI (38), along with biomarkers such as cardiac troponins and D-dimer levels, to inform their decisions regarding rtPA administration. Ultimately, timely identification requires a high level of clinical awareness and effective teamwork (46).
Thrombolytic therapy, particularly rtPA, has been explored as an adjunct to standard CPR protocols in several clinical studies and trials (Fig. 2). For instance, the TICA trial indicated that rtPA could improve ROSC rates, with reported rates of ~35% in the rtPA group compared with ~20% in standard CPR cases (47). However, it also raised concerns about safety, necessitating further research on optimal dosing and outcomes. In a study by Bozeman et al (48), the use of tenecteplase (TNK) after failed Advanced Cardiac Life Support (ACLS) measures resulted in a ROSC rate of 26% compared with 12.4% in the control group (P=0.04). Additionally, 12% of TNK patients survived to admission compared with none in the control group (P=0.0007). This study emphasized the potential benefit of timely intervention, as TNK was given after an average of 30 min of cardiac arrest.
In a prospective study, 108 patients received rtPA during CPR, while 216 matched controls received standard CPR without thrombolysis. The rtPA group demonstrated markedly improved outcomes, with ROSC achieved in 70.4% compared with 51.0% in the control group. Additionally, 24-h survival was 48.1% in the rtPA group compared with 32.9% in controls. Survival to hospital discharge was also higher in the rtPA group at 25.9%, although exact comparative data for the control group was not specified. Among patients with presumed PE, outcomes were even more favorable: 24-h survival was 57.9% and discharge survival reached 31.6%. Bleeding complications occurred in 5.6% of rtPA patients, including one case of intracranial hemorrhage (0.9%), which was equal to the ICH rate of the control group (0.9%) (30). In a retrospective study of 66 patients with CA, rtPA administered during or shortly after CPR was associated with improved early survival outcomes compared with standard CPR protocols. ROSC was achieved in 67% of patients in the rtPA group compared with 43% in the standard CPR protocol group and 24-h survival was markedly higher at 53% compared with 23% in controls. Importantly, prolonged CPR (≥10 min) did not correlate with increased bleeding risk. The data suggest that administration of rtPA during CPR for CA could improve ROSC and short-term survival compared with the standard CPR protocols (49).
In another prospective study involving 90 patients with out-of-hospital cardiac arrest, 40 received rt-PA during CPR, while 50 received standard CPR alone. Thrombolytic therapy markedly improved clinical outcomes. ROSC occurred in 68% of rt-PA patients compared with 44% of controls and 58% were admitted to a cardiac intensive care unit compared with 30% of controls. Although 24-h survival (35 vs. 22%) and hospital discharge rates (15 vs. 8%) favored the rtPA group, these differences were not statistically significant. Notably, no bleeding complications related to CPR were observed. However, two rtPA patients experienced gastrointestinal bleeding requiring transfusion, occurring 2- and 12-days post-arrest (19). In a randomized, double-blind, placebo-controlled trial involving 233 patients with cardiac arrest and presumed cardiovascular etiology, the efficacy of tPA during CPR was evaluated against standard CPR alone. Patients were randomized to receive either tPA or placebo in addition to standard CPR. ROSC occurred in 21.4% of the tPA group and 23.3% of the placebo group, showing no significant benefit. Only one patient (0.9%) in the tPA group survived to hospital discharge, compared with none in the placebo group. Four tPA patients survived more than 24 h post-admission, but this did not translate into improved overall outcomes (50).
The Thrombolysis in Cardiac Arrest (TROICA) study (51), an international multicenter trial, evaluated the efficacy and safety of tenecteplase during CPR for out-of-hospital cardiac arrest. Despite its larger scale, TROICA found no significant improvement in long-term survival or neurological outcomes compared with placebo. The study reported ROSC rates of ~30% in the rtPA group compared with 20% in the control group. Timing was critical; rtPA was administered as soon as feasible after ROSC, typically within 30 min. However, the quality of post-resuscitation care varied, which may have obscured the potential benefits of thrombolysis. Furthermore, Bakkum et al (36) demonstrated that an accelerated regimen of rtPA (0.6 mg/kg in 15 min) resulted in a trend toward higher ROSC rates (67 compared with 43%; P=0.06) compared with no thrombolysis in patients with pulmonary embolism-induced circulatory arrest. Amini et al (35) investigated the efficacy and safety of reduced doses of rtPA in patients with acute pulmonary embolism, comparing it to standard doses and anticoagulation therapy. Analyzing data from 13 studies, including four observational studies with 4,223 patients and nine randomized controlled trials involving 780 patients, the authors found that the reduced dose was associated with a markedly lower incidence of total bleeding events (RR 5.08; 95%CI 1.39–18.6) compared with anticoagulants. Furthermore, when comparing standard and reduced doses of rtPA, the standard dose showed a greater incidence of bleeding (RR 1.48; 95%CI 1.00–2.19). Importantly, there were no significant differences in all-cause mortality or recurrence of PE between the treatment groups, suggesting that a reduced dose of rtPA offers a safer alternative without compromising efficacy. Doelare et al (52) investigated the relationship between fibrinogen levels and major bleeding risk during catheter-directed thrombolysis for acute limb ischemia. Of 443 patients treated with rtPA, the overall incidence of major bleeding was 7%. Patients with fibrinogen levels below 1.0 g/l had a markedly higher bleeding rate (15 vs. 6%; P=0.041), highlighting low fibrinogen as a critical risk factor. Additionally, high-dose thrombolytic regimens and older age were independent predictors of major bleeding. The TROICA study reported a lower incidence of major bleeding complications at ~3%, attributed to careful patient selection and monitoring (51). The contrasting results across these studies may be attributed to several factors, including variability in patient characteristics (such as age or comorbidities) that may influence outcomes, as seen in the TROICA study where patient selection criteria were stringent (53). The timing of rtPA administration markedly affected outcomes; studies that administered rtPA more quickly after cardiac arrest generally reported improved results. Additionally, differences in dosing strategies (standard compared with accelerated or reduced doses) could explain variations in efficacy and safety profiles observed in different trials (54). Finally, variability in post-resuscitation care protocols across studies can markedly affect observed outcomes. Factors such as the availability of advanced monitoring, therapeutic hypothermia and comprehensive cardiac care can differ widely between institutions and trials. Inconsistent care may obscure the potential benefits of thrombolytic therapy, as optimal post-resuscitation management is crucial for patient recovery and overall survival (55) (Table I). It is important to note that neurological outcomes have not been evaluated in most of these studies.
The efficacy of thrombolytic therapy in the context of CPR is primarily measured through rates of ROSC, survival to hospital admission and survival to hospital discharge with favorable neurological outcomes (56). In studies where thrombolytics were administered, an increase in ROSC rates was often observed (57). For instance, a meta-analysis of several small trials indicated a statistically significant improvement in ROSC rates when rtPA was used. However, the effect on survival to hospital discharge remains less definitive (58). This highlights the need to distinguish between short-term efficacy (such as ROSC and hospital admission) and long-term outcomes (such as survival to discharge and neurological recovery). While some studies have reported higher survival rates for hospital admission, these improvements do not always translate to long-term survival or favorable neurological outcomes (59). The TROICA trial, for example, found no significant difference in survival to hospital discharge between the thrombolytic and placebo groups, highlighting the complexity and potential limitations of thrombolytic therapy in CPR. Moreover, even when ROSC is achieved, the potential for hypoxic brain injury and subsequent neurological impairment remains high, underlining the importance of evaluating therapies not only for their ability to restore circulation but also for their influence on neurological prognosis and functional recovery (60,61). Future studies and clinical protocols should prioritize these distinctions to improved assess the true benefit of thrombolytic therapy in cardiac arrest scenarios.
The data reveals potential benefits and notable risks when comparing outcomes between thrombolytic therapy and standard CPR protocols. Standard CPR protocols, which do not include thrombolytics, have established benchmarks for ROSC and survival rates. Thrombolytic therapy, in theory, offers an advantage by addressing underlying thrombotic causes of cardiac arrest, potentially enhancing coronary and cerebral perfusion during resuscitation efforts (19). However, while some studies suggest improved ROSC rates with thrombolytics, the overall survival to hospital discharge and neurological outcomes do not consistently show significant improvements over standard protocols (62). Moreover, the risk of adverse effects, particularly hemorrhagic complications, remains a critical concern. Bleeding risks associated with thrombolytic therapy can offset potential benefits, especially in the context of traumatic cardiac arrests or in patients with underlying conditions predisposing them to hemorrhage (63).
Identifying suitable candidates for thrombolytic therapy during CPR involves assessing the underlying cause of cardiac arrest, the patient's medical history and the likelihood of a favorable outcome from thrombolysis. Thrombolytic therapy, particularly with rtPA, is most beneficial in cases where cardiac arrest is due to PE, MI or other thrombotic events (64). Clinicians should prioritize patients with witnessed arrest, a short duration of arrest before the initiation of CPR and those without severe comorbidities (65). The presence of risk factors such as recent surgery, active bleeding, or a history of hemorrhagic stroke generally contraindicates the use of thrombolytics due to the elevated risk of bleeding complications (44,58). If available, advanced imaging techniques such as echocardiography or computed tomography scans can help confirm the presence of thrombi, guiding the decision to administer rtPA (44). Thrombolytic therapy in CPR involves careful consideration of various factors, including indications such as pulmonary embolism and myocardial infarction (44), as well as exclusion criteria like active bleeding and recent surgery (66) (Table II). Patient selection is critical, taking into account age, sex, and pre-existing conditions such as carotid artery stenosis (67,68). Administering therapy promptly can improve survival and neurological outcomes (69,70). This therapy should be coupled with adjunctive treatments (71), and careful monitoring (72), while adhering to established clinical guidelines for individualized care (73).
The timing of thrombolytic therapy during CPR is crucial for its effectiveness. The administration of rtPA should occur as early as possible after cardiac arrest, ideally within the first few min of CPR, to maximize the chances of restoring spontaneous circulation (74). This approach is supported by findings from observational studies and case series, which indicate that early administration of thrombolytics, particularly within the first 10–15 min of arrest, is associated with improved ROSC and neurological outcomes. Additionally, recommendations from the European Resuscitation Council and the American Heart Association suggest considering thrombolytics during CPR in patients with suspected or confirmed PE, provided that administration does not markedly delay other resuscitative efforts (65,75–77).
The standard dosing regimen for rtPA in the context of CPR often involves a bolus injection followed by a continuous infusion. However, protocols may vary based on institutional practices and patient-specific factors (54). The typical dose ranges from 50–100 mg of rtPA, administered over 15–60 min as observed in studies and clinical guidelines addressing massive PE and cardiac arrest management (75–77). In practice, a 50 mg bolus may be used in out-of-hospital settings to allow rapid administration, whereas a 100 mg infusion over 2 h is common in in-hospital PE protocols, adapted for use during prolonged resuscitation. The administration route is typically intravenous, but intraosseous access can be considered if intravenous access is not readily available. Continuous monitoring of the patient during and after the administration is essential to manage potential complications, such as bleeding or allergic reactions and to assess the effectiveness of the therapy. These recommendations are increasingly being refined through accumulating evidence from registry data, observational cohorts and ongoing randomized controlled trials (78–82) (Table III).
Thrombolytic therapy during CPR carries significant risks, which necessitate careful consideration of contraindications (51). Absolute contraindications include active internal bleeding, a history of intracranial hemorrhage, recent major surgery or trauma, known bleeding disorders and severe uncontrolled hypertension (83–85). Relative contraindications, which require a risk-benefit analysis, include recent gastrointestinal bleeding, peptic ulcer disease and pregnancy (86). The most significant potential risk of thrombolytic therapy is hemorrhage, including intracranial hemorrhage, which can lead to devastating outcomes (87). Other risks include reperfusion arrhythmias, allergic reactions and an increased risk of further thrombotic events in certain conditions (88).
The decision to use rtPA during CPR must balance these risks against the potential for improved survival and neurological outcomes, particularly in patients with a high likelihood of thrombotic cardiac arrest etiology. This decision-making process often depends not only on patient-specific factors but also on the availability of institutional protocols, access to rapid diagnostic imaging and multidisciplinary team input (89,90). In emergency settings, bedside tools such as focused cardiac ultrasound can assist in identifying possible thrombotic causes (such as massive PE), providing a rationale for thrombolytic administration even when formal imaging is not feasible (91). Furthermore, institutions with predefined algorithms for cardiac arrest management that include thrombolytic therapy in select cases help streamline decision-making and reduce hesitation during critical moments (92,93). Comprehensive patient assessment and adherence to established guidelines are essential to optimizing the benefits of thrombolytic therapy while minimizing its risks (29). Ultimately, a tailored approach that integrates clinical judgment, real-time diagnostics and institutional capabilities is crucial in determining the appropriateness of rtPA during CPR.
While promising in specific scenarios, thrombolytic therapy poses medical and logistical challenges when used during CPR. These issues encompass medical and logistical aspects, which must be carefully addressed to utilize thrombolytic agents such as rtPA effectively. This section outlines key limitations, including bleeding risks, inconsistent efficacy and difficulties with timely administration in diverse settings (94,95).
A primary concern with thrombolytic therapy in CPR is the increased risk of bleeding, particularly intracranial hemorrhage (96). While the goal is to dissolve obstructive coronary clots and restore myocardial blood flow, this benefit comes with a higher risk of bleeding, especially in critical areas such as the brain (49,97). The delicate balance between thrombolysis and hemorrhage must be meticulously managed, as any imbalance can lead to catastrophic outcomes (49). Moreover, CPR patients often have impaired clotting due to pre-existing conditions or prior treatments, further heightening bleeding risks (98).
Response to thrombolytic therapy during CPR varies markedly among patients (99). While some patients may exhibit a robust response to thrombolytic agents, experiencing rapid clot dissolution and successful resuscitation, others may demonstrate limited or no response despite timely administration (69,100). This variability stems from multiple factors, including the cause of cardiac arrest, the degree of vessel occlusion and individual patient characteristics such as age, comorbidities and coagulation status. As a result, identifying patients most likely to benefit remains a challenge and calls for more personalized and evidence-based approaches (67).
Besides clinical concerns, logistical barriers hinder effective thrombolytic administration during CPR, particularly in pre-hospital and in-hospital environments (101,102). Unlike standard CPR interventions, thrombolytic therapy requires trained personnel, specialized equipment and infrastructure (103,104). In pre-hospital care, EMS teams must be trained in thrombolysis and the required drugs and monitoring tools must be readily available en route. In hospitals, issues such as drug storage, preparation and protocol adherence can delay timely administration (64,73). Addressing these challenges demands standardized protocols, cross-disciplinary teamwork and ongoing system improvements.
As CPR methods evolve, research is focusing on refining thrombolytic therapy, especially regarding timing, dosage and delivery of agents such as rtPA. Ongoing studies explore how patient-specific factors, including comorbidities and arrest etiology, can inform protocol adjustments (78,105). There is also growing interest in personalized medicine approaches that tailor thrombolytic regimens to individual biomarker profiles. This could maximize therapeutic efficacy while minimizing adverse outcomes such as bleeding. Collaborative research across clinical and industry sectors aims to integrate thrombolysis more effectively into ACLS frameworks.
In parallel with efforts to optimize existing thrombolytic agents, researchers are exploring novel agents and delivery methods to enhance the efficacy and safety of thrombolytic therapy in CPR. Novel thrombolytic agents, such as modified variants of rtPA or entirely new classes of thrombolytic drugs, are being investigated for their potential to improve clot dissolution kinetics and reduce the risk of reperfusion injury (106). Furthermore, advancements in drug delivery technologies enable targeted and controlled delivery of thrombolytic agents to the site of vascular occlusion, thereby minimizing systemic exposure and off-target effects. Nanoparticle-based drug delivery systems, microneedle patches and catheter-based delivery platforms are among the innovative approaches being explored to optimize the delivery of thrombolytic therapy during CPR (107).
As the complexity of resuscitative interventions continues to expand, there is growing interest in integrating thrombolytic therapy with other advanced techniques, such as extracorporeal membrane oxygenation (ECMO), to improve outcomes in refractory cardiac arrest cases. ECMO provides temporary cardiopulmonary support by oxygenating blood outside the body, allowing for prolonged resuscitative efforts and stabilization of patients with profound hemodynamic instability (108). Research endeavors are underway to elucidate the optimal strategies for combining thrombolytic therapy with ECMO in the management of cardiac arrest patients with suspected or confirmed pulmonary embolism or massive myocardial infarction (109). By synergistically addressing both the underlying cause of cardiac arrest (such as acute coronary syndrome and pulmonary embolism) and the resultant hemodynamic collapse, the integration of thrombolytic therapy with ECMO holds promise for improving survival and neurological outcomes in select patient populations (110). Moreover, ongoing studies are investigating the potential synergistic effects of combining thrombolytic therapy with other advanced resuscitative interventions, such as targeted temperature management and mechanical circulatory support devices, to optimize outcomes in refractory cardiac arrest cases further (111). Through interdisciplinary collaboration and rigorous clinical research, these integrated approaches aim to redefine the standard of care for patients in cardiac arrest, ultimately improving survival rates and neurological recovery.
Using thrombolytics during CPR involves complex ethical issues related to autonomy, benefit, harm and resource fairness. In emergencies, informed consent is often not feasible, raising concerns about respecting patient autonomy while aiming to save lives (112). The risk of adverse effects, especially bleeding, must be weighed against the potential benefit of restoring circulation. Additionally, the uneven availability of thrombolytic therapy across healthcare settings raises questions of equity and access. Clinicians must carefully navigate these ethical concerns within resource-constrained environments (113).
Legally, thrombolytic use during CPR touches on informed consent, liability and documentation. Given the urgency of CPR, formal consent may not always be possible, but providers should communicate risks and alternatives whenever feasible (114). Failing to inform or document appropriately could result in legal consequences, including malpractice claims. Comprehensive documentation of treatment rationale, consent discussions (if possible) and patient responses is vital to protect both patients and clinicians (115).
Clear guidelines and protocols are paramount for the safe and effective use of thrombolytic therapy in CPR. These guidelines offer healthcare providers with standardized procedures for patient assessment, medication administration and monitoring during resuscitative efforts (65). Guidelines help ensure practice consistency, reduce care variability and promote patient safety. They outline criteria for patient selection, dosing regimens and monitoring parameters to minimize the risk of adverse events associated with thrombolytic therapy. Additionally, clear protocols aid in informed decision-making and facilitate communication among healthcare team members (116). They provide a framework for addressing ethical dilemmas, such as determining futility or prioritizing resources in resource-limited settings. Regular review and updates of guidelines and protocols are essential to incorporate new evidence and best practices. This iterative process helps healthcare institutions adapt to advancements in medical knowledge and technology, ultimately improving patient outcomes in the challenging context of CPR.
The present review provided compelling evidence supporting the efficacy of thrombolytic therapy, specifically rtPA, in improving outcomes for patients undergoing CPR. The administration of rtPA has been shown to increase the likelihood of ROSC and favorable neurological outcomes. However, it is imperative to carefully consider the potential risks, including hemorrhagic complications. Future research should focus on refining patient selection criteria, optimizing dosing regimens and identifying ideal timing for rtPA administration to maximize benefit and minimize adverse events. Additionally, large-scale randomized controlled trials are needed to further elucidate the role of thrombolytic therapy in specific patient populations, such as those with acute myocardial infarction or stroke. By integrating evidence-based guidelines and protocols, healthcare providers can harness the potential of thrombolytic therapy to markedly improve patient survival and quality of life following cardiac arrest.
Not applicable.
The present study was supported by Jilin Provincial Health and Health Commission (grant no. 2022LC038).
Not applicable.
All authors contributed to the study conception and design. Original draft preparation was conducted by WZ, YP and JD. Writing, reviewing and editing of the draft manuscript was completed by WZ, YP, DX and WW. In addition, WW supervised the writing, analyses and revision of the manuscript. All authors commented on previous versions of the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Professor Wenhai Wang
ORCID: 0009-0002-3879-4259
|
Riggs M, Franklin R and Saylany L: Associations between cardiopulmonary resuscitation (CPR) knowledge, self-efficacy, training history and willingness to perform CPR and CPR psychomotor skills: A systematic review. Resuscitation. 138:259–272. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Zhai ZG, Liang LR, Liu FF, Yang YH and Wang C: Lower dosage of recombinant tissue-type plasminogen activator (rt-PA) in the treatment of acute pulmonary embolism: A systematic review and meta-analysis. Thromb Res. 133:357–363. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Monsieurs KG, Nolan JP, Bossaert LL, Greif R, Maconochie IK, Nikolaou NI, Perkins GD, Soar J, Truhlář A, Wyllie J, et al: European resuscitation council guidelines for resuscitation 2015: Section 1. Executive summary. Resuscitation. 95:1–80. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Taffet GE, Teasdale TA and Luchi RJ: In-hospital cardiopulmonary resuscitation. JAMA. 260:2069–2072. 1988. View Article : Google Scholar : PubMed/NCBI | |
|
Kamali MJ, Salehi M and Fath MK: Advancing personalized immunotherapy for melanoma: Integrating immunoinformatics in multi-epitope vaccine development, neoantigen identification via NGS, and immune simulation evaluation. Comput Biol Med. 188:1098852025. View Article : Google Scholar : PubMed/NCBI | |
|
Konstantinides SV, Meyer G, Becattini C, Bueno H, Geersing GJ, Harjola VP, Huisman MV, Humbert M, Jennings CS, Jiménez D, et al: 2019 ESC guidelines for the diagnosis and management of acute pulmonary embolism developed in collaboration with the European respiratory society (ERS) the task force for the diagnosis and management of acute pulmonary embolism of the European Society of Cardiology (ESC). Eur Heart J. 41:543–603. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gilbert CR, Wilshire CL, Chang SC and Gorden JA: The use of intrapleural thrombolytic or fibrinolytic therapy, or both, via indwelling tunneled pleural catheters with or without concurrent anticoagulation use. Chest. 160:776–783. 2021. View Article : Google Scholar | |
|
Ortel TL, Neumann I, Ageno W, Beyth R, Clark NP, Cuker A, Hutten BA, Jaff MR, Manja V, Schulman S, et al: American Society of Hematology 2020 guidelines for management of venous thromboembolism: Treatment of deep vein thrombosis and pulmonary embolism. Blood Adv. 4:4693–4738. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Altaf F, Wu S and Kasim V: Role of fibrinolytic enzymes in anti-thrombosis therapy. Front Mol Biosci. 8:6803972021. View Article : Google Scholar | |
|
Grines CL, Serruys P and O'Neill WW: Fibrinolytic therapy: Is it a treatment of the past? Circulation. 107:2538–2542. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang S, Wang D and Li L: Recombinant tissue-type plasminogen activator (rt-PA) effectively restores neurological function and improves prognosis in acute ischemic stroke. Am J Transl Res. 15:34602023.PubMed/NCBI | |
|
Leal Rato M, Diógenes MJ and Sebastião A: Pharmacodynamics and pharmacokinetics of stroke therapy. Precision Medicine in Stroke. Springer Nature; pp. 41–69. 2021, View Article : Google Scholar | |
|
Marko M, Posekany A, Szabo S, Scharer S, Kiechl S, Knoflach M, Serles W, Ferrari J, Lang W, Sommer P, et al: Trends of r-tPA (recombinant tissue-type plasminogen activator) treatment and treatment-influencing factors in acute ischemic stroke. Stroke. 51:1240–1247. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Llucià-Carol L, Muiño E, Gallego-Fabrega C, Cárcel-Márquez J, Martín-Campos J, Lledós M, Cullell N and Fernández-Cadenas I: Pharmacogenetics studies in stroke patients treated with rtPA: A review of the most interesting findings. Pharmacogenomics. 22:1091–1097. 2021. View Article : Google Scholar | |
|
Peppard SR, Parks AM and Zimmerman J: Characterization of alteplase therapy for presumed or confirmed pulmonary embolism during cardiac arrest. Am J Health Syst Pharm. 75:870–875. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Kotb E: The biotechnological potential of fibrinolytic enzymes in the dissolution of endogenous blood thrombi. Biotechnol Prog. 30:656–672. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Sharifi M, Berger J, Beeston P, Bay C, Vajo Z and Javadpoor S; ‘PEAPETT’ investigators, : Pulseless electrical activity in pulmonary embolism treated with thrombolysis (from the ‘PEAPETT’ study). Am J Emerg Med. 34:1963–1967. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Dejouvencel T, Doeuvre L, Lacroix R, Plawinski L, Dignat-George F, Lijnen HR and Anglés-Cano E: Fibrinolytic cross-talk: A new mechanism for plasmin formation. Blood. 115:2048–2056. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Böttiger BW, Bode C, Kern S, Gries A, Gust R, Glätzer R, Bauer H, Motsch J and Martin E: Efficacy and safety of thrombolytic therapy after initially unsuccessful cardiopulmonary resuscitation: A prospective clinical trial. Lancet. 357:1583–1585. 2001. View Article : Google Scholar | |
|
Iyer A, Zurolo E, Boer K, Baayen J, Giangaspero F, Arcella A, Di Gennaro GC, Esposito V, Spliet WG, van Rijen PC, et al: Tissue plasminogen activator and urokinase plasminogen activator in human epileptogenic pathologies. Neuroscience. 167:929–945. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Badimon L, Padró T and Vilahur G: Atherosclerosis, platelets and thrombosis in acute ischaemic heart disease. Eur Heart J Acute Cardiovasc Care. 1:60–74. 2012.PubMed/NCBI | |
|
Yepes M: The uPA/uPAR system in astrocytic wound healing. Neural Regen Res. 17:2404–2406. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang B, Leung L, Su EJ and Lawrence DA: PA system in the pathogenesis of ischemic stroke. Arterioscler Thromb Vasc Biol. 45:600–608. 2025. View Article : Google Scholar | |
|
Kadir RRA and Bayraktutan U: Urokinase plasminogen activator: A potential thrombolytic agent for ischaemic stroke. Cell Mol Neurobiol. 40:347–355. 2020. View Article : Google Scholar | |
|
Emberson J, Lees KR, Lyden P, Blackwell L, Albers G, Bluhmki E, Brott T, Cohen G, Davis S, Donnan G, et al: Effect of treatment delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for acute ischaemic stroke: A meta-analysis of individual patient data from randomised trials. Lancet. 384:1929–1935. 2014. View Article : Google Scholar | |
|
Katsanos AH, Psychogios K, Turc G, Sacco S, de Sousa DA, De Marchis GM, Palaiodimou L, Filippou DK, Ahmed N, Sarraj A, et al: Off-label use of tenecteplase for the treatment of acute ischemic stroke: A systematic review and meta-analysis. JAMA Netw Open. 5:e2245062022. View Article : Google Scholar : PubMed/NCBI | |
|
Fredriksson L, Lawrence DA and Medcalf RL: tPA modulation of the blood-brain barrier: A unifying explanation for the pleiotropic effects of tPA in the CNS. Semin Thromb Hemost. 43:154–168. 2017. | |
|
Katz JM and Tadi P: Physiology, plasminogen activation. StatPearls Publishing; Treasure Island: 2019 | |
|
Gurman P, Miranda O, Nathan A, Washington C, Rosen Y and Elman N: Recombinant tissue plasminogen activators (rtPA): A review. Clin Pharmacol Ther. 97:274–285. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Lederer W, Lichtenberger C, Pechlaner C, Kroesen G and Baubin M: Recombinant tissue plasminogen activator during cardiopulmonary resuscitation in 108 patients with out-of-hospital cardiac arrest. Resuscitation. 50:71–76. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Sabermoghadam R, Shahmahmood TM, Sarvghadi P and Toosi MB: The gross, fine, and oral motor functions in a patient with megalencephalic leukoencephalopathy with subcortical cyst: A case report. Iran J Child Neurol. 17:149–161. 2023. | |
|
Hasanoğlu H, Hezer H, Karalezli A, Argüder E, Kiliç H, Şentürk A, Er M and Soytürk AN: Half-dose recombinant tissue plasminogen activator treatment in venous thromboembolism. J Investig Med. 62:71–77. 2014. View Article : Google Scholar | |
|
Wang X, Li X, Xu Y, Li R, Yang Q, Zhao Y, Wang F, Sheng B, Wang R, Chen S, et al: Effectiveness of intravenous r-tPA versus UK for acute ischaemic stroke: A nationwide prospective Chinese registry study. Stroke Vasc Neurol. 6:603–609. 2021. View Article : Google Scholar | |
|
Li BH, Wang JH, Wang H, Wang DZ, Yang S, Guo FQ and Yu NW: Different doses of intravenous tissue-type plasminogen activator for acute ischemic stroke: A network meta-analysis. Front Neurol. 13:8842672022. View Article : Google Scholar | |
|
Amini S, Bakhshandeh H, Mosaed R, Abtahi H, Sadeghi K and Mojtahedzadeh M: Efficacy and safety of different dosage of recombinant tissue-type plasminogen activator (rt-PA) in the treatment of acute pulmonary embolism: A systematic review and meta-analysis. Iran J Pharm Res. 20:441–454. 2021.PubMed/NCBI | |
|
Bakkum MJ, Schouten VL, Smulders YM, Nossent EJ, van Agtmael MA and Tuinman PR: Accelerated treatment with rtPA for pulmonary embolism induced circulatory arrest. Thromb Res. 203:74–80. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong K, An X, Kong Y and Chen Z: Predictive model for the risk of hemorrhagic transformation after rt-PA intravenous thrombolysis in patients with acute ischemic stroke: A systematic review and meta-analysis. Clin Neurol Neurosurg. 239:1082252024. View Article : Google Scholar : PubMed/NCBI | |
|
Hezer H, Kiliç H, Abuzaina O, Hasanoğlu HC and Karalezli A: Long-term results of low-dose tissue plasminogen activator therapy in acute pulmonary embolism. J Investig Med. 67:1142–1147. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ucar EY, Araz O, Kerget B, Yilmaz N, Akgun M and Saglam L: Comparison of long-term outcomes of 50 and 100 mg rt-PA in the management of acute pulmonary thromboembolism. Clin Respir J. 12:1628–1634. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Salehi M, Kamali MJ, Ashuori Z, Ghadimi F, Shafiee M, Babaei S and Moghadam AA: Functional significance of tRNA-derived fragments in sustained proliferation of tumor cells. Gene Rep. 35:1019012024. View Article : Google Scholar | |
|
Le Conte P, Huchet L, Trewick D, Longo C, Vial I, Batard E, Yatim D, Touzé MD and Baron D: Efficacy of alteplase thrombolysis for ED treatment of pulmonary embolism with shock. Ame J Emerg Med. 21:438–440. 2003. View Article : Google Scholar | |
|
Cokkinos P: Post-resuscitation care: Current therapeutic concepts. Acute Card Care. 11:131–137. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Herlitz J, Castren M, Friberg H, Nolan J, Skrifvars M, Sunde K and Steen PA: Post resuscitation care: What are the therapeutic alternatives and what do we know? Resuscitation. 69:15–22. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Javaudin F, Lascarrou JB, Le Bastard Q, Bourry Q, Latour C, De Carvalho H, Le Conte P, Escutnaire J, Hubert H, Montassier E, et al: Thrombolysis during resuscitation for out-of-hospital cardiac arrest caused by pulmonary embolism increases 30-day survival: Findings from the French National Cardiac Arrest Registry. Chest. 156:1167–1175. 2019. View Article : Google Scholar | |
|
Dasa O, Ruzieh M, Ammari Z, Syed MA, Brickman KR and Gupta R: It's a ST-elevation myocardial infarction (STEMI), or is it? Massive pulmonary embolism presenting as STEMI. J Emerg Med. 55:125–127. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Villablanca PA, Vlismas PP, Aleksandrovich T, Omondi A, Gupta T, Briceno DF, Garcia MJ and Wiley J: Case report and systematic review of pulmonary embolism mimicking ST-elevation myocardial infarction. Vascular. 27:90–97. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Fatovich DM, Dobb GJ and Clugston RA: A pilot randomised trial of thrombolysis in cardiac arrest (The TICA trial). Resuscitation. 61:309–313. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Bozeman WP, Kleiner DM and Ferguson KL: Empiric tenecteplase is associated with increased return of spontaneous circulation and short term survival in cardiac arrest patients unresponsive to standard interventions. Resuscitation. 69:399–406. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Janata K, Holzer M, Kürkciyan I, Losert H, Riedmüller E, Pikula B, Laggner AN and Laczika K: Major bleeding complications in cardiopulmonary resuscitation: The place of thrombolytic therapy in cardiac arrest due to massive pulmonary embolism. Resuscitation. 57:49–55. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Abu-Laban RB, Christenson JM, Innes GD, van Beek CA, Wanger KP, McKnight RD, MacPhail IA, Puskaric J, Sadowski RP, Singer J, et al: Tissue plasminogen activator in cardiac arrest with pulseless electrical activity. N Engl J Med. 346:1522–1528. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Spöhr F, Arntz HR, Bluhmki E, Bode C, Carli P, Chamberlain D, Danays T, Poth J, Skamira C, Wenzel V and Böttiger BW: International multicentre trial protocol to assess the efficacy and safety of tenecteplase during cardiopulmonary resuscitation in patients with out-of-hospital cardiac arrest: The thrombolysis in cardiac arrest (TROICA) study. Eur J Clin Invest. 35:315–323. 2005. View Article : Google Scholar | |
|
Doelare SA, Oukrich S, Ergin K, Jongkind V, Wiersema AM, Lely RJ, Ebben HP, Yeung KK and Hoksbergen AWJ: Collaborators: Major bleeding during thrombolytic therapy for acute lower limb ischaemia: Value of laboratory tests for clinical decision making, 17 years of experience. Eur J Vasc Endovasc Surg. 65:398–404. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Pereira R, Bowen M, Rapezzano G, Redpath A, Pratt S and Hallowell G: Use of recombinant tissue plasminogen activator (rTPA) for treatment of fibrin in the anterior chamber of the horse. Vet Med Sci. 10:e14482024. View Article : Google Scholar | |
|
Vandebroek AC, Rickmann A, Boden KT, Seitz B, Szurman P and Schulz A: Thrombolytic activity of recombinant tissue-type plasminogen activator (rtPA) in in-vitro model for subretinal hemorrhages. Int Ophthalmol. 45:1642025. View Article : Google Scholar | |
|
Li X, Chen Y, Dong Z, Zhang F, Chen P and Zhang P: A study of the safety and efficacy of multi-mode NMR-guided double-antiplatelet pretreatment combined with low-dose rtPA in the treatment of acute mild ischemic stroke. Front Neurol. 16:14820782025. View Article : Google Scholar | |
|
Oh TK, Park YM, Do SH, Hwang JW and Song IA: ROSC rates and live discharge rates after cardiopulmonary resuscitation by different CPR teams-a retrospective cohort study. BMC Anesthesiol. 17:1–9. 2017. View Article : Google Scholar | |
|
Gräsner JT, Wnent J, Herlitz J, Perkins GD, Lefering R, Tjelmeland I, Koster RW, Masterson S, Rossell-Ortiz F, Maurer H, et al: Survival after out-of-hospital cardiac arrest in Europe-Results of the EuReCa TWO study. Resuscitation. 148:218–226. 2020. View Article : Google Scholar | |
|
Wang Y, Wang M, Ni Y, Liang B and Liang Z: Can systemic thrombolysis improve prognosis of cardiac arrest patients during cardiopulmonary resuscitation? A systematic review and meta-analysis. J Emerg Med. 57:478–487. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Nolan JP, Soar J, Smith GB, Gwinnutt C, Parrott F, Power S, Harrison DA, Nixon E and Rowan K; National Cardiac Arrest Audit, : Incidence and outcome of in-hospital cardiac arrest in the United Kingdom National Cardiac Arrest audit. Resuscitation. 85:987–992. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Hoiland RL, Ainslie PN, Wellington CL, Cooper J, Stukas S, Thiara S, Foster D, Fergusson NA, Conway EM, Menon DK, et al: Brain hypoxia is associated with neuroglial injury in humans post-cardiac arrest. Circ Res. 129:583–597. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hoiland RL, Robba C, Menon DK, Citerio G, Sandroni C and Sekhon MS: Clinical targeting of the cerebral oxygen cascade to improve brain oxygenation in patients with hypoxic-ischaemic brain injury after cardiac arrest. Intensive Care Med. 49:1062–1078. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Panchal AR, Bartos JA, Cabañas JG, Donnino MW, Drennan IR, Hirsch KG, Kudenchuk PJ, Kurz MC, Lavonas EJ, Morley PT, et al: Part 3: Adult basic and advanced life support: 2020 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation. 142 (16_Suppl_2):S366–S468. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Soar J, Böttiger BW, Carli P, Couper K, Deakin CD, Djärv T, Lott C, Olasveengen T, Paal P, Pellis T, et al: European resuscitation council guidelines 2021: Adult advanced life support. Resuscitation. 161:115–151. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Stadlbauer KH, Krismer AC, Arntz HR, Mayr VD, Lienhart HG, Böttiger BW, Jahn B, Lindner KH and Wenzel V: Effects of thrombolysis during out-of-hospital cardiopulmonary resuscitation. Am J Cardiol. 97:305–308. 2006. View Article : Google Scholar | |
|
Lott C, Truhlář A, Alfonzo A, Barelli A, González-Salvado V, Hinkelbein J, Nolan JP, Paal P, Perkins GD, Thies KC, et al: European resuscitation council guidelines 2021: Cardiac arrest in special circumstances. Resuscitation. 161:152–219. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 372:n712021. View Article : Google Scholar : PubMed/NCBI | |
|
Higgins J, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, Savović J, Schulz KF, Weeks L and SterneJA C; Cochrane Bias Methods GroupCochrane Statistical Methods Group, : The cochrane collaboration's tool for assessing risk of bias in randomised trials. BMJ. 343:d59282011. View Article : Google Scholar : PubMed/NCBI | |
|
Blum B, Wormack L, Holtel M, Penwell A, Lari S, Walker B and Nathaniel TI: Gender and thrombolysis therapy in stroke patients with incidence of dyslipidemia. BMC Womens Health. 19:112019. View Article : Google Scholar : PubMed/NCBI | |
|
Stang A: Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol. 25:603–605. 2010. View Article : Google Scholar | |
|
Qiao Y, Wang J, Nguyen T, Liu L, Ji X and Zhao W: Intravenous thrombolysis with urokinase for acute ischemic stroke. Brain Sci. 14:9892024. View Article : Google Scholar | |
|
Broderick C, Watson L and Armon MP: Thrombolytic strategies versus standard anticoagulation for acute deep vein thrombosis of the lower limb. Cochrane Database Syst Rev. 1:CD0027832021.PubMed/NCBI | |
|
Kluft C, Sidelmann JJ and Gram JB: Assessing safety of thrombolytic therapy. Semin Thromb Hemost. 43:300–310. 2017. | |
|
Renard A, Verret C, Jost D, Meynard JB, Tricehreau J, Hersan O, Fontaine D, Briche F, Benner P, de Stabenrath O, et al: Impact of fibrinolysis on immediate prognosis of patients with out-of-hospital cardiac arrest. J Thromb Thrombolysis. 32:405–409. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wardlaw JM, Koumellis P and Liu M: Thrombolysis (different doses, routes of administration and agents) for acute ischaemic stroke. Cochrane Database Syst Rev. 31:CD0005142013. | |
|
Ramaswamy VV, Abiramalatha T, Weiner GM and Trevisanuto D: A comparative evaluation and appraisal of 2020 American Heart Association and 2021 European Resuscitation Council neonatal resuscitation guidelines. Resuscitation. 167:151–159. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Semeraro F, Greif R, Böttiger BW, Burkart R, Cimpoesu D, Georgiou M, Yeung J, Lippert F, Lockey AS, Olasveengen TM, et al: European resuscitation council guidelines 2021: Systems saving lives. Resuscitation. 161:80–97. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Nolan JP, Sandroni C, Böttiger BW, Cariou A, Cronberg T, Friberg H, Genbrugge C, Haywood K, Lilja G, Moulaert VRM, et al: European resuscitation council and European society of intensive care medicine guidelines 2021: Post-resuscitation care. Resuscitation. 161:220–269. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yousuf T, Brinton T, Ahmed K, Iskander J, Woznicka D, Kramer J, Kopiec A, Chadaga AR and Ortiz K: Tissue plasminogen activator use in cardiac arrest secondary to fulminant pulmonary embolism. J Clin Med Res. 8:190–195. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Sobala R, Kibbler J, Yip K, Thompson R, Rafique C, Steward M, Samanta R; INSPIRE ERUPT collaborators, ; Jha A, Rahman N and De Soyza A: Evaluation of Real-world use of pulmonary embolism thrombolysis (ERUPT). ERJ Open Res. 11:00935–2024. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Nadkarni VM, Larkin GL, Peberdy MA, Carey SM, Kaye W, Mancini ME, Nichol G, Lane-Truitt T, Potts J, Ornato JP, et al: First documented rhythm and clinical outcome from in-hospital cardiac arrest among children and adults. JAMA. 295:50–57. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Nolan J, Laver S, Welch C, Harrison D, Gupta V and Rowan K: Outcome following admission to UK intensive care units after cardiac arrest: A secondary analysis of the ICNARC case mix programme database. Anaesthesia. 62:1207–1216. 2007. View Article : Google Scholar | |
|
Miyazaki K, Hikone M, Kuwahara Y, Ishida T, Sugiyama K and Hamabe Y: Extracorporeal CPR for massive pulmonary embolism in a ‘hybrid 2136 emergency department’. Am J Emerg Med. 37:2132–2135. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wallentin L, Goldstein P, Armstrong PW, Granger CB, Adgey AA, Arntz HR, Bogaerts K, Danays T, Lindahl B, Makijarvi M, et al: Efficacy and safety of tenecteplase in combination with the low-molecular-weight heparin enoxaparin or unfractionated heparin in the prehospital setting: the assessment of the safety and efficacy of a new thrombolytic regimen (ASSENT)-3 PLUS randomized trial in acute myocardial infarction. Circulation. 108:135–142. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Ali MR, Hossain MS, Islam MA, Arman MS, Raju GS, Dasgupta P and Noshin TF: Aspect of thrombolytic therapy: A review. ScientificWorldJournal. 2014:5865102014. View Article : Google Scholar | |
|
Saber-Moghadam R: Investigating the efficacy of noninvasive brain stimulation on poststroke dysphagia: A systematic review. JNFH. 10:P2862022. | |
|
Beales I: Recent advances in the management of peptic ulcer bleeding. F1000Res. 6:17632017. View Article : Google Scholar : PubMed/NCBI | |
|
Scott JH, Gordon M, Vender R, Pettigrew S, Desai P, Marchetti N, Mamary AJ, Panaro J, Cohen G, Bashir R, et al: Venoarterial extracorporeal membrane oxygenation in massive pulmonary embolism-related cardiac arrest: A systematic review. Crit Care Med. 49:760–769. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Vanden Hoek TL, Morrison LJ, Shuster M, Donnino M, Sinz E, Lavonas EJ, Jeejeebhoy FM and Gabrielli A: Part 12: Cardiac arrest in special situations: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation. 122 (18_suppl_3):S829–S861. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Amacher SA, Bohren C, Blatter R, Becker C, Beck K, Mueller J, Loretz N, Gross S, Tisljar K, Sutter R, et al: Long-term survival after out-of-hospital cardiac arrest: A systematic review and meta-analysis. JAMA Cardiol. 7:633–643. 2022. View Article : Google Scholar | |
|
Cho Y, Oh J, Shin JH, Kim BS, Park JK, Lee JH, Kim JH and Park M: Long-term prognosis and causes of death among survivors after out-of-hospital cardiac arrest: A population-based longitudinal study. Resuscitation. 173:31–38. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chocron R, Fahrenbruch C, Yin L, Guan S, Drucker C, Shin J, Eisenberg M, Chatterjee NA, Kudenchuk PJ and Rea T: Association between functional status at hospital discharge and long-term survival after out-of-hospital-cardiac-arrest. Resuscitation. 164:30–37. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Berg KM, Soar J, Andersen LW, Böttiger BW, Cacciola S, Callaway CW, Couper K, Cronberg T, D'Arrigo S, Deakin CD, et al: Adult advanced life support: 2020 international consensus on cardiopulmonary resuscitation and emergency cardiovascular care science with treatment recommendations. Circulation. 142 (16_suppl_1):S92–S139. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Maconochie IK, Aickin R, Hazinski MF, Atkins DL, Bingham R, Couto TB, Guerguerian AM, Nadkarni VM, Ng KC, Nuthall GA, et al: Pediatric life support 2020 international consensus on cardiopulmonary resuscitation and emergency cardiovascular care science with treatment recommendations. Pediatrics. 147 (Suppl 1):e2020038505B2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kireyev D, Tan HC and Poh KK: Management of acute ST-elevation myocardial infarction: Reperfusion options. Ann Acad Med Singap. 39:927–933. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Pinto DS, Frederick PD, Chakrabarti AK, Kirtane AJ, Ullman E, Dejam A, Miller DP, Henry TD and Gibson CM; National Registry of Myocardial Infarction Investigators, : Benefit of transferring ST-segment-elevation myocardial infarction patients for percutaneous coronary intervention compared with administration of onsite fibrinolytic declines as delays increase. Circulation. 124:2512–2521. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Böttiger BW and Martin E: Thrombolytic therapy during cardiopulmonary resuscitation and the role of coagulation activation after cardiac arrest. Curr Opin Crit Care. 7:176–183. 2001. | |
|
Böttiger BW, Arntz HR, Chamberlain DA, Bluhmki E, Belmans A, Danays T, Carli PA, Adgey JA, Bode C, Wenzel V, et al: Thrombolysis during resuscitation for out-of-hospital cardiac arrest. N Engl J Med. 359:2651–2662. 2008. View Article : Google Scholar | |
|
Higgins JP and Altman DG: Assessing risk of bias in included studies. Cochrane handbook for systematic reviews of interventions: Cochrane Book Series. 2008:187–241. 2008. View Article : Google Scholar | |
|
Vedantham S, Piazza G, Sista AK and Goldenberg NA: Guidance for the use of thrombolytic therapy for the treatment of venous thromboembolism. J Thromb Thrombolysis. 41:68–80. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Luchini C, Veronese N, Nottegar A, Shin JI, Gentile G, Granziol U, Soysal P, Alexinschi O, Smith L and Solmi M: Assessing the quality of studies in meta-research: Review/guidelines on the most important quality assessment tools. Pharm Stat. 20:185–195. 2021. View Article : Google Scholar | |
|
Zhou Y, Yan S, Song X, Gong Y, Li W, Wang M, Yin X, Hu B and Lu Z: Intravenous thrombolytic therapy for acute ischemic stroke in Hubei, China: A survey of thrombolysis rate and barriers. BMC Neurol. 19:1–9. 2019. View Article : Google Scholar | |
|
Berger AK, Radford MJ, Wang Y and Krumholz HM: Thrombolytic therapy in older patients. J Am Coll Cardiol. 36:366–374. 2000. View Article : Google Scholar | |
|
Sølling C, Ashkanian M, Hjort N, Gyldensted C, Andersen G and Østergaard L: Feasibility and logistics of MRI before thrombolytic treatment. Acta Neurol Scand. 120:143–149. 2009. View Article : Google Scholar | |
|
Thortveit ET, Bøe MG, Ljøstad U, Mygland Å and Tveiten A: Organizational changes aiming to reduce iv tPA door-to-needle time. Acta Neurol Scand. 130:248–252. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Salehi M, Kamali MJ, Rajabzadeh A, Minoo S, Mosharafi H, Saeedi F and Daraei A: tRNA-derived fragments: Key determinants of cancer metastasis with emerging therapeutic and diagnostic potentials. Arch Biochem Biophys. 753:1099302024. View Article : Google Scholar : PubMed/NCBI | |
|
Ma H, Jiang Z, Xu J, Liu J and Guo ZN: Targeted nano-delivery strategies for facilitating thrombolysis treatment in ischemic stroke. Drug Deliv. 28:357–371. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zamanlu M, Farhoudi M, Eskandani M, Mahmoudi J, Barar J, Rafi M and Omidi Y: Recent advances in targeted delivery of tissue plasminogen activator for enhanced thrombolysis in ischaemic stroke. J Drug Target. 26:95–109. 2018. View Article : Google Scholar | |
|
Yang Y, Chang Q, Chen J, Zou X, Xue Q and Song A: Application of integrated emergency care model based on failure modes and effects analysis in patients with ischemic stroke. Front Surg. 9:8745772022. View Article : Google Scholar : PubMed/NCBI | |
|
Taccone FS, Nobile L and Annoni F: Thrombolysis for ECMO oxygenator thrombosis. Crit Care. 27:1422023. View Article : Google Scholar : PubMed/NCBI | |
|
Reisinger AC, Fandler-Höfler S, Kreuzer P, Toth-Gayor G, Schmidt A, Gary T, Rief P, Eller P and Brodmann M: VA-ECMO and thrombus aspiration in a pulmonary embolism patient with cardiac arrest and contraindications to thrombolytic therapy. Vasa. 51:315–319. 2022. View Article : Google Scholar | |
|
Lin TW, Tsai MT, Hu YN, Wang YC, Wen JS, Wu HY, Luo CY and Roan JN: Simultaneous thrombolysis and extracorporeal membrane oxygenation for acute massive pulmonary emboli. Ann Thorac Surg. 111:923–929. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Han TS, Gulli G, Fry CH, Affley B, Robin J, Fluck D, Kakar P and Sharma P: Adverse consequences of immediate thrombolysis-related complications: A multi-centre registry-based cohort study of acute stroke. J Thromb Thrombolysis. 53:1–10. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zonjee VJ, Slenders JP, de Beer F, Visser MC, Ter Meulen BC, Van den Berg-Vos RM and van Schaik SM: Practice variation in the informed consent procedure for thrombolysis in acute ischemic stroke: A survey among neurologists and neurology residents. BMC Med Ethics. 22:1–9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Szalados JE: The ethics and laws governing informed decision-making in healthcare: Informed consent, refusal, and discussions regarding resuscitation and life-sustaining treatment. The medical-legal aspects of acute care medicine: A resource for clinicians, administrators, and risk managers. Springer; 2021, pp. 43–73. View Article : Google Scholar | |
|
Milling L, Binderup LG, de Muckadell CS, Christensen EF, Lassen A, Christensen HC, Nielsen DC and Mikkelsen S; The Danish Cardiac Arrest Registry Group, : Documentation of ethically relevant information in out-of-hospital resuscitation is rare: A Danish nationwide observational study of 16,495 out-of-hospital cardiac arrests. BMC Med Ethics. 22:822021. View Article : Google Scholar : PubMed/NCBI | |
|
Zenych A, Fournier L and Chauvierre C: Nanomedicine progress in thrombolytic therapy. Biomaterials. 258:1202972020. View Article : Google Scholar : PubMed/NCBI |