A Comprehensive Report on the Plasminogen-Plasmin System: From Molecular Biology to Therapeutic Intervention
1.0 Executive Summary
This report provides a comprehensive analysis of the plasminogen-plasmin system, a critical enzymatic cascade responsible for fibrinolysis and broader physiological processes including tissue remodeling and inflammation. A foundational clarification is made between plasminogen, the inactive zymogen identified by DrugBank ID DB16701 and CAS Number 9001-91-6, and its active form, plasmin, a potent serine protease. The system's central function is the degradation of fibrin clots, a process tightly regulated by a balance of physiological activators, such as tissue plasminogen activator (tPA), and inhibitors, like α2-antiplasmin. Dysregulation of this system leads to severe pathological states, including thrombosis from insufficient activity and hemorrhage from excessive activity.
Therapeutic intervention targeting this system is categorized into three distinct strategies. The most established approach involves the administration of plasminogen activators (e.g., Alteplase) for acute thrombolytic therapy in conditions like ischemic stroke and myocardial infarction. A second, more recent strategy is plasminogen replacement therapy (e.g., Ryplazim) for the chronic management of congenital plasminogen deficiency. The third is the investigational use of direct-acting plasmin, which aims to provide localized thrombolysis with a potentially improved safety profile. Across all pro-fibrinolytic therapies, hemorrhage remains the principal and mechanism-based safety concern, dictating stringent patient selection and risk management. Key knowledge gaps persist, most notably the clinical pharmacokinetics of exogenously administered plasmin. Future advancements in the field are trending towards precision medicine, focusing on developing agents with enhanced fibrin-specificity, novel delivery methods for localized effects, and curative approaches like gene therapy for congenital deficiencies.
2.0 Molecular and Biochemical Profile: Plasminogen and Plasmin
A precise understanding of the molecular entities within the plasminogen-plasmin system is fundamental to comprehending its physiological regulation and pharmacological manipulation. The initial query's identifiers correspond to the zymogen, plasminogen, which must be clearly distinguished from the active enzyme, plasmin.
2.1 Nomenclature and Identification
The primary components of the system are the precursor protein, plasminogen, and its activated enzymatic form, plasmin. Their distinct identifiers are crucial for accurate scientific and clinical discourse.
Plasminogen (The Zymogen): This is the inactive precursor that circulates in the blood. The identifiers provided in the user query correspond to this molecule.[1]
- DrugBank ID: DB16701 [1]
- CAS Number: 9001-91-6 [4]
- Synonyms: Profibrinolysin, Plasma Trypsinogen [6]
Plasmin (The Active Enzyme): This is the functional serine protease responsible for fibrinolysis.
- CAS Number: 9001-90-5 [9]
- Synonyms: Fibrinolysin [1]
- EC Number: 3.4.21.7 (Enzyme Commission number for serine endopeptidases) [11]
The following table provides a concise comparison of these two key proteins.
Table 1: Identification and Key Properties of Plasminogen and Plasmin
Characteristic | Plasminogen | Plasmin |
---|
Common Name | Plasminogen | Plasmin |
DrugBank ID | DB16701 | DB05254 (Fibrinolysin) |
CAS Number | 9001-91-6 | 9001-90-5 |
EC Number | N/A (Zymogen) | 3.4.21.7 |
Synonyms | Profibrinolysin, Plasma Trypsinogen | Fibrinolysin |
Molecular Weight | ~92-93 kDa | ~85 kDa |
Structure | Single-chain glycoprotein | Two-chain enzyme (Heavy A, Light B) |
Primary Function | Inactive precursor of plasmin | Active serine protease; degrades fibrin |
2.2 Structure and Physicochemical Properties of Plasminogen
Plasminogen is a single-chain glycoprotein synthesized predominantly by the liver, which circulates in human plasma at a concentration of approximately 100–200 mg/L.[14] It has a molecular weight of about 92-93 kDa and is composed of 791 amino acids with a carbohydrate content of approximately 2%.[14]
The protein's structure is modular, comprising seven distinct domains that dictate its function and regulation. These include an N-terminal Pan Apple (PAp) domain, five homologous "Kringle" domains (KR1-5), and a C-terminal serine protease (SP) domain.[11] The Kringle domains, each containing about 80 amino acids stabilized by three disulfide bonds, are of paramount importance as they contain lysine-binding sites (LBS).[14] These sites mediate the binding of plasminogen to lysine residues present on its primary substrate, fibrin, as well as on cell surface receptors and inhibitors like α2-antiplasmin.[14] This binding is the biochemical basis for localizing fibrinolytic activity, ensuring that plasmin generation is concentrated at the site of a thrombus rather than occurring systemically. The PAp domain plays a critical role in maintaining plasminogen in its inactive state.[11]
This intricate structure allows for a sophisticated regulatory mechanism based on conformational plasticity. In circulation, plasminogen adopts a "closed," compact, and activation-resistant conformation.[11] Upon binding to lysine residues on a fibrin clot or a cell surface, it undergoes a significant conformational change to a more linear, "open" form. This open conformation exposes the Arg561-Val562 activation bond, making it a much more efficient substrate for plasminogen activators.[18] This conformational shift represents a state of heightened "activation potential," a dynamic condition regulated by the local microenvironment. It is an elegant physiological solution to prevent widespread, uncontrolled proteolysis, and its circumvention by certain therapeutic agents explains the risk of a systemic lytic state.[20]
Further complexity arises from different isoforms. The native circulating form is Glu-plasminogen, named for its N-terminal glutamic acid.[14] Limited proteolytic cleavage by plasmin itself can remove an N-terminal peptide, yielding Lys-plasminogen, which has a more open conformation and is activated more rapidly.[15] Additionally, two major glycoforms exist: Type I plasminogen, with both N-linked and O-linked glycosylation, is preferentially recruited to blood clots, whereas Type II, with only an O-linked sugar, is more readily recruited to cell surfaces.[11]
2.3 Structure and Physicochemical Properties of Plasmin
Plasmin is formed from its zymogen precursor through the specific proteolytic cleavage of the peptide bond between Arginine-561 (Arg561) and Valine-562 (Val562).[11] This reaction is catalyzed by physiological plasminogen activators like tPA and uPA.
The cleavage event transforms the single-chain plasminogen into a two-chain active enzyme with a molecular weight of approximately 85 kDa.[13] The resulting structure consists of a heavy chain A (comprising the N-terminal portion and the five Kringle domains) connected via two disulfide bonds to a light chain B (the C-terminal serine protease domain).[12]
As a serine protease, plasmin exhibits trypsin-like activity, preferentially cleaving peptide bonds on the C-terminal side of lysine and arginine residues, though it is more selective than trypsin.[11] Its optimal enzymatic activity occurs at a pH of approximately 8.9.[13] For research and manufacturing purposes, plasmin is often supplied as a lyophilized powder or as a suspension in 3.2 M ammonium sulfate for stability.[13] Studies have shown that human plasmin is more resistant to autodigestion at alkaline pH than bovine trypsin and that its stability can be enhanced by the presence of glycerol, a property not shared by related diols, suggesting a specific structural interaction.[22]
3.0 The Fibrinolytic System: Physiology and Pathophysiology
Plasmin does not operate in isolation; it is the central effector enzyme of the fibrinolytic system, a dynamic cascade crucial for maintaining vascular homeostasis and participating in a wide array of other biological processes.
3.1 The Central Role of Plasmin in Fibrinolysis
The primary and most well-characterized function of plasmin is fibrinolysis: the enzymatic dissolution of fibrin polymers that form the structural matrix of blood clots.[11] By degrading fibrin into soluble fragments, known as fibrin degradation products (FDPs), plasmin restores blood flow in vessels after hemostasis and tissue repair are complete, thereby maintaining vascular patency.[18]
This process is meticulously regulated by a balance of activators and inhibitors.
- Activators: The generation of plasmin from plasminogen is initiated by specific activators.
- Tissue Plasminogen Activator (tPA): Synthesized and released by vascular endothelial cells, tPA is the principal initiator of intravascular fibrinolysis. Its enzymatic efficiency increases by several orders of magnitude when both tPA and its substrate, plasminogen, are co-localized on the surface of a fibrin clot. This fibrin-cofactor dependence is a key mechanism for ensuring that fibrinolysis is a localized event.[18]
- Urokinase Plasminogen Activator (uPA): While also present in blood, uPA's primary role is in the extracellular space, where it facilitates processes like cell migration and tissue remodeling by activating plasminogen on cell surfaces.[14]
- Inhibitors: To prevent premature or excessive clot lysis and systemic proteolysis, the fibrinolytic system is tightly controlled by several inhibitors.
- Plasminogen Activator Inhibitor-1 (PAI-1): This is the main physiological inhibitor of both tPA and uPA, controlling the rate of plasmin generation.[19]
- α2-Antiplasmin: This is the most important and rapid inhibitor of plasmin itself. It circulates in plasma and quickly forms a stable, inactive 1:1 complex with any free plasmin that escapes the confines of the clot, effectively preventing unwanted systemic fibrinogenolysis.[11]
- α2-Macroglobulin: This acts as a secondary, slower-acting inhibitor of plasmin, serving as a backup mechanism to α2-antiplasmin.[11]
3.2 Non-Fibrinolytic Functions of Plasmin
Plasmin's proteolytic activity extends far beyond fibrin, positioning it as a powerful modulator of the extracellular environment. Its diverse substrate profile reveals its function as a "master switch" in numerous physiological and pathological processes, where its activity can initiate cascades of tissue alteration.
- Extracellular Matrix (ECM) Degradation: Plasmin is a broad-spectrum protease capable of degrading key components of the ECM, including fibronectin, laminin, thrombospondin, and some collagen types.[11]
- Tissue Remodeling and Cell Migration: Through its ECM-degrading activity, plasmin is integral to processes that require tissue restructuring and cell movement. This includes embryogenesis, angiogenesis (the formation of new blood vessels), wound healing, and ovulation, where it weakens the wall of the Graafian follicle to allow for egg release.[11]
- Inflammation and Immunity: Plasmin is deeply involved in the inflammatory response. It can activate mediators of the complement system and modulate immune cell trafficking by cleaving tissue-tethered chemokines, such as CCL21, to create a soluble gradient that attracts other immune cells.[11]
- Pathological Roles: In pathological contexts, dysregulated plasmin activity is a key factor in cancer progression. By degrading the ECM and activating latent matrix metalloproteinases (MMPs), plasmin facilitates tumor cell invasion and metastasis.[14]
3.3 Pathophysiology of Dysregulation
An imbalance in the fibrinolytic system can have severe consequences, leading to either bleeding or thrombosis.
- Hyperfibrinolysis (Excessive Activity): An overactive fibrinolytic state leads to premature lysis of hemostatic plugs, resulting in bleeding. This is a key component of the coagulopathy seen in severe trauma, where an initial surge in fibrinolysis contributes to massive hemorrhage.[25] Therapeutically, this state is intentionally induced by thrombolytic drugs, and bleeding is their primary adverse effect.[29]
- Hypofibrinolysis (Insufficient Activity): A deficient fibrinolytic system results in inadequate clot clearance, predisposing an individual to thrombotic events.[11] The most severe form is
Type I Plasminogen Deficiency (hypoplasminogenemia), a rare autosomal recessive disorder. The profound lack of plasminogen leads to the accumulation of extravascular fibrin deposits on mucous membranes, forming characteristic "ligneous" (wood-like) pseudomembranes. These lesions most commonly affect the conjunctiva of the eye (ligneous conjunctivitis) but can occur throughout the body, causing significant morbidity.[12]
The pathophysiology of severe trauma reveals a particularly complex, biphasic dysregulation. An initial, transient phase of hyperfibrinolysis, which contributes to acute hemorrhage, is often followed by a state of "fibrinolytic shutdown".[25] This subsequent hypofibrinolytic state is associated with an increased risk of thrombosis and multi-organ failure. This dynamic shift illustrates that the problem is not simply "too much" or "too little" fibrinolysis but a profound temporal dysregulation. This complexity poses a significant challenge for therapeutic intervention; for example, an antifibrinolytic agent that is life-saving in the early hemorrhagic phase could be detrimental if it exacerbates the later thrombotic phase, highlighting a critical need for real-time diagnostics to guide therapy.
Table 2: Major Physiological Regulators of the Plasminogen-Plasmin System
Regulator | Type | Primary Source | Function/Target |
---|
tPA | Activator | Endothelial Cells | Activates fibrin-bound plasminogen |
uPA | Activator | Leukocytes, Fibroblasts, etc. | Activates cell surface-bound plasminogen |
PAI-1 | Inhibitor | Endothelial Cells | Inhibits tPA and uPA |
α2-Antiplasmin | Inhibitor | Liver | Rapidly inhibits free plasmin |
α2-Macroglobulin | Inhibitor | Liver | Slowly inhibits free plasmin |
4.0 Pharmacology of Fibrinolytic and Antifibrinolytic Agents
Therapeutic manipulation of the plasminogen-plasmin system is achieved through several distinct pharmacological strategies, each with unique mechanisms, pharmacokinetic profiles, and interaction risks.
4.1 Pharmacodynamics (Mechanism of Action)
The mechanisms by which drugs modulate fibrinolysis can be broadly categorized as indirect activation, direct action, replacement, or inhibition.
- Indirect-Acting Agents (Plasminogen Activators): This is the most common class of thrombolytic drugs. They function by catalytically converting the body's endogenous plasminogen into active plasmin.[20]
- Examples: Alteplase (a recombinant form of human tPA), Reteplase, Tenecteplase, Streptokinase, and Urokinase.[19]
- Mechanism: These agents mimic physiological activators by cleaving the Arg561-Val562 bond of plasminogen.[18] A key distinction exists between "fibrin-specific" and "non-specific" agents. Fibrin-specific agents like Alteplase possess a high affinity for fibrin, which co-localizes the activator with its substrate (plasminogen) on the clot surface. This dramatically enhances catalytic efficiency at the target site while minimizing the activation of plasminogen circulating freely in the plasma.[20] In contrast, non-specific agents like Streptokinase form a complex with plasminogen that activates both clot-bound and circulating plasminogen indiscriminately, leading to a more profound systemic lytic state characterized by fibrinogen degradation.[20]
- Direct-Acting Agents (Plasmin): This is an investigational approach that involves the administration of the active enzyme, plasmin, itself.
- Mechanism: This strategy bypasses the entire plasminogen activation step, delivering the fibrinolytic effector directly to the thrombus. The theoretical advantage is a more direct, potentially more controllable, and activation-independent thrombolytic effect.[13]
- Replacement Therapy (Plasminogen): This strategy is used to treat deficiency states.
- Example: Ryplazim® (plasminogen, human-tvmh).[30]
- Mechanism: Ryplazim administration temporarily increases the plasma concentration of plasminogen. This restores the substrate pool, allowing the patient's own endogenous activators to generate plasmin where needed (i.e., at sites of extravascular fibrin deposition), thereby correcting the functional deficit of the fibrinolytic system.[30]
- Antifibrinolytic Agents (Inhibitors): These drugs are used to prevent or treat bleeding by blocking fibrinolysis.
- Examples: Tranexamic acid (TXA), aminocaproic acid.[4]
- Mechanism: These molecules are synthetic analogs of the amino acid lysine. They competitively bind to the lysine-binding sites within the Kringle domains of both plasminogen and plasmin. This action has a dual effect: it prevents plasminogen from binding to fibrin, thereby inhibiting its activation by tPA, and it prevents already-formed plasmin from binding to and degrading its fibrin substrate.[28]
A critical and counterintuitive pharmacodynamic consideration is the potential for a paradoxical procoagulant effect. Studies have demonstrated that high concentrations of plasmin can activate components of the intrinsic coagulation pathway, specifically Factor XII, leading to the generation of thrombin.[37] This is supported by observations that lysis rates can paradoxically decrease at very high concentrations of tPA.[38] This suggests that excessive thrombolytic therapy might not only increase bleeding risk but could also, under certain conditions, promote the formation of new clots. This creates a complex therapeutic window where dosing must be sufficient to achieve lysis but not so high as to trigger these paradoxical effects.
4.2 Pharmacokinetics (Absorption, Distribution, Metabolism, and Excretion)
The pharmacokinetic profiles of these agents differ dramatically and are the primary determinant of their clinical utility, creating a "PK/PD mismatch" that defines their therapeutic applications.
- Plasminogen Activators: These are characterized by extremely rapid clearance and short half-lives, suiting them for acute, emergency interventions where a powerful but transient effect is desired.
- Alteplase (rtPA): Has a very short initial half-life of approximately 5 minutes.[20] It is primarily cleared from circulation by the liver.[39] This necessitates administration as a continuous intravenous infusion following an initial bolus.
- Reteplase and Tenecteplase: These are genetically engineered variants of tPA, modified to have longer half-lives and greater fibrin affinity. This improved pharmacokinetic profile allows for more convenient administration as one or two intravenous boluses.[20]
- Plasminogen (Ryplazim): As a replacement therapy for a chronic condition, this plasma-derived protein exhibits a much longer half-life, suitable for maintaining sustained physiological levels.
- Half-life: The elimination half-life is approximately 34 hours after the first dose, which extends to around 39.2 hours by week 12 of therapy.[35]
- Distribution: The volume of distribution (Vd) is relatively small at approximately 63.3 mL/kg, suggesting it is largely confined to the plasma compartment.[35]
- Clearance: The clearance rate appears to decrease with prolonged administration.[8]
- Exogenous Plasmin: A critical knowledge gap exists regarding the clinical pharmacokinetics of directly administered plasmin. As a large, active protease, its disposition cannot be reliably extrapolated from its zymogen or activators. Based on principles of large molecule and cell therapy pharmacokinetics, one might hypothesize rapid clearance from the blood and significant uptake by reticuloendothelial organs like the liver, spleen, and lungs, but this remains speculative without clinical data.[41] The absence of this fundamental PK data is a major challenge for the rational design of dosing regimens and the overall development of direct plasmin therapeutics.
Table 3: Comparative Pharmacokinetic Profiles of Fibrinolytic Agents
Agent | Class | Half-Life | Volume of Distribution | Clearance Mechanism | Typical Administration |
---|
Alteplase | Plasminogen Activator | ~5 min | N/A | Hepatic | IV Bolus + 60-min Infusion |
Reteplase | Plasminogen Activator | Longer than Alteplase | N/A | Hepatic/Renal | IV Double Bolus |
Tenecteplase | Plasminogen Activator | Longer than Alteplase | N/A | Hepatic | IV Single Bolus |
Plasminogen (Ryplazim) | Replacement Therapy | ~39 hours | ~63 mL/kg | N/A | IV Infusion (q2-4 days) |
Investigational Plasmin | Direct-Acting Enzyme | Unknown (Critical Gap) | Unknown | Unknown | Catheter-Directed Infusion |
4.3 Drug Interactions
Given their profound effects on hemostasis, agents modulating the plasmin system have numerous clinically significant drug interactions, primarily related to bleeding risk.
- Agents that Increase Bleeding Risk (Pharmacodynamic Synergism): The most significant interactions involve the concomitant use of drugs that interfere with any aspect of hemostasis. The risk of hemorrhage is substantially increased.
- Anticoagulants: Co-administration with heparin, warfarin, direct thrombin inhibitors (e.g., bivalirudin), and direct Factor Xa inhibitors (e.g., rivaroxaban, apixaban, edoxaban) is a major interaction.[43]
- Antiplatelet Agents: Concurrent use of aspirin, P2Y12 inhibitors (e.g., clopidogrel, prasugrel, ticagrelor), and glycoprotein IIb/IIIa inhibitors (e.g., abciximab, eptifibatide) significantly elevates bleeding risk.[43]
- Nonsteroidal Anti-inflammatory Drugs (NSAIDs): Drugs like ibuprofen, naproxen, and ketorolac, which inhibit platelet function, also increase the risk of bleeding when combined with fibrinolytic therapy.[29]
- Agents that Decrease Efficacy (Pharmacodynamic Antagonism):
- Antifibrinolytic Agents: Tranexamic acid and aminocaproic acid directly oppose the action of thrombolytics and are contraindicated for concurrent use unless specifically employed to manage a bleeding complication.[29]
- Other Notable Interactions:
- Angiotensin-Converting Enzyme (ACE) Inhibitors: Concomitant use with Alteplase may increase the risk of developing orolingual angioedema.[33]
Table 4: Clinically Significant Drug Interactions
Interacting Drug Class | Example Drugs | Effect on Plasmin System Therapy | Clinical Recommendation |
---|
Anticoagulants | Heparin, Warfarin, Rivaroxaban | Major increase in bleeding risk | Generally contraindicated or requires extremely cautious use with intensive monitoring. |
Antiplatelet Agents | Aspirin, Clopidogrel, Abciximab | Major increase in bleeding risk | Often used adjunctively in AMI, but significantly increases hemorrhage risk. Requires careful risk-benefit assessment. |
NSAIDs | Ibuprofen, Ketorolac | Moderate increase in bleeding risk | Avoid concomitant use when possible, especially during active thrombolytic therapy. |
Antifibrinolytic Agents | Tranexamic Acid, Aminocaproic Acid | Direct antagonism, decreased efficacy | Contraindicated unless used to reverse life-threatening hemorrhage caused by the fibrinolytic agent. |
ACE Inhibitors | Lisinopril, Enalapril | Increased risk of angioedema (with Alteplase) | Monitor patient closely for signs of orolingual swelling. |
5.0 Clinical Development and Therapeutic Applications
The pharmacological principles described above have been translated into three distinct therapeutic strategies, each addressing a different clinical problem: acute thrombus removal, chronic zymogen deficiency, and localized clot dissolution.
5.1 Plasminogen Activators (Thrombolytic Therapy)
These agents form the backbone of pharmacological reperfusion therapy for acute thrombotic events. Their use is time-critical and aimed at rapidly restoring blood flow to ischemic tissue.
- Approved Indications:
- Acute Ischemic Stroke (AIS): Alteplase is the standard of care, approved by the FDA for administration within 3 hours of symptom onset, with an extended window of up to 4.5 hours for certain eligible patients.[39]
- Acute Myocardial Infarction (AMI): Thrombolytics are indicated for ST-segment elevation myocardial infarction (STEMI), particularly in settings where timely primary percutaneous coronary intervention (PCI) is unavailable (delay >120 minutes).[20]
- Massive Pulmonary Embolism (PE): Alteplase is approved for the treatment of massive PE that causes hemodynamic instability (e.g., hypotension).[39]
- Other Uses: A low-dose formulation of Alteplase (Cathflo Activase®) is used to restore patency to occluded central venous access devices.[40] Off-label, catheter-directed thrombolysis is used for severe deep vein thrombosis (DVT) and peripheral arterial occlusion (PAO).[20]
- Dosing and Administration: Regimens are highly specific to the indication and are often weight-based to balance efficacy and bleeding risk. For AIS, the standard Alteplase dose is 0.9 mg/kg (not to exceed a total dose of 90 mg), with 10% of the dose given as an intravenous bolus over one minute, followed by an infusion of the remaining 90% over 60 minutes.[47] This complex administration protocol stands in contrast to newer agents like Tenecteplase, which can be given as a single bolus for AMI, and is being investigated off-label for AIS. This off-label use, while potentially more convenient, introduces new patient safety challenges related to medication familiarity and the potential for dosing errors.[49]
5.2 Plasminogen Replacement Therapy
This therapy is not for thrombolysis but for the chronic management of a rare genetic disorder.
- Approved Indication: Ryplazim® (plasminogen, human-tvmh) is the first and only therapy approved by the FDA for the treatment of Plasminogen Deficiency Type 1 (hypoplasminogenemia).[30]
- Clinical Efficacy: The approval of Ryplazim was based on clinical trials demonstrating its ability to restore physiological plasminogen levels and resolve the clinical manifestations of the disease. In a pivotal study, regular administration of Ryplazim resulted in a complete or partial resolution of lesions in all patients who had them at baseline. Specifically, 78% of external lesions and 75% of internal lesions had resolved by week 48, with no new or recurrent lesions observed during the study period.[30] This demonstrates significant clinical benefit in a disease with no other approved treatments.
- Dosing and Administration: The recommended starting dose is 6.6 mg/kg administered intravenously. The dosing frequency is highly individualized, typically ranging from every 2 to 4 days, and is guided by monitoring trough plasminogen activity levels and assessing the clinical status of the patient's lesions.[35]
5.3 Investigational Direct Plasmin Therapy
This approach represents a potential third wave of fibrinolytic therapy, aiming to improve upon the safety profile of plasminogen activators.
- Rationale: By administering the active enzyme plasmin directly to a thrombus (typically via a catheter), this strategy bypasses the need for plasminogen activation. The hypothesis is that this could lead to effective, localized thrombolysis with less systemic activation of the fibrinolytic system, potentially reducing the risk of distant bleeding compared to systemic administration of plasminogen activators.[45]
- Clinical Development: The development of direct plasmin therapy is in early stages. A Phase I clinical trial has been completed evaluating catheter-directed administration of human plasmin at doses ranging from 25 mg to 175 mg in 83 patients with acute peripheral arterial occlusion (aPAO).[34]
- Early Results: The Phase I study reported that the therapy was generally well-tolerated with no unexpected safety concerns. The incidence of major bleeding was 4.8%, a rate consistent with that seen with existing catheter-directed therapies for this condition. Importantly, there was no trend toward increased bleeding at higher doses of plasmin, providing preliminary safety data to support further investigation into its efficacy.[34]
Table 5: Summary of Approved Indications and Dosing Regimens
Therapeutic Agent | Trade Name(s) | Indication | Typical Dosing Regimen |
---|
Alteplase | Activase®, Cathflo Activase® | Acute Ischemic Stroke (AIS) | 0.9 mg/kg (max 90 mg) IV; 10% as bolus, 90% infused over 60 min |
| | Acute Myocardial Infarction (AMI) | 100 mg total dose IV, administered over 1.5 hr (accelerated) or 3 hr |
| | Massive Pulmonary Embolism (PE) | 100 mg IV infused over 2 hr |
| | Occluded Central Venous Catheter | 2 mg instilled into catheter |
Reteplase | Retavase® | Acute Myocardial Infarction (AMI) | 10 units + 10 units IV double bolus 30 min apart |
Tenecteplase | TNKase® | Acute Myocardial Infarction (AMI) | Single weight-based IV bolus (30-50 mg) |
Plasminogen, human-tvmh | Ryplazim® | Plasminogen Deficiency Type 1 | 6.6 mg/kg IV every 2-4 days, adjusted based on trough levels |
6.0 Safety Profile and Risk Management
The therapeutic benefit of enhancing fibrinolysis is intrinsically linked to the risk of hemorrhage. Therefore, a thorough understanding of the safety profile and strict adherence to contraindications are paramount for the clinical use of these agents.
6.1 Hemorrhagic Complications
Bleeding is the most frequent and most feared complication of all pro-fibrinolytic therapies.[46] This is not an off-target side effect but rather an over-expression of the drug's intended pharmacological action. The same enzymatic activity that dissolves a pathological thrombus can also degrade fibrin within essential hemostatic plugs at sites of recent injury or vascular fragility.[21]
- Types of Bleeding: Hemorrhagic events can range from minor, such as oozing from IV sites, epistaxis (nosebleeds), or gingival bleeding, to severe, life-threatening events.[46] The most devastating is intracranial hemorrhage (ICH), but major bleeding can also occur in the gastrointestinal tract, retroperitoneal space, or genitourinary system.[52]
- Risk Factors: The risk of bleeding is not uniform across all patients. Known risk factors include advanced age, uncontrolled hypertension (systolic >185 mmHg or diastolic >110 mmHg), recent surgery, trauma, or biopsy, a history of prior ICH, and the concomitant use of other medications that impair hemostasis, such as anticoagulants and antiplatelet agents.[46]
6.2 Non-Hemorrhagic Adverse Events
While bleeding is the primary concern, other adverse events can occur.
- Hypersensitivity Reactions: As with any biologic agent, allergic reactions are possible. These can range from skin rash to severe anaphylaxis.[54] Orolingual angioedema is a specific concern with Alteplase, reported in 1-2% of stroke patients, with risk increased by concurrent use of ACE inhibitors. While often mild, it can be rapidly progressive and require emergency airway management.[46]
- Tissue Sloughing: This adverse event is unique to plasminogen replacement therapy with Ryplazim. The therapeutic goal is the dissolution of extravascular fibrin lesions. However, when these lesions are located in critical areas like the tracheobronchial tree, their breakdown and sloughing can lead to airway obstruction.[44] This represents a scenario where the desired therapeutic effect itself can cause a life-threatening complication, necessitating close monitoring, especially at the initiation of therapy.
- Cardiovascular Events: In the context of AMI treatment, successful reperfusion can be associated with arrhythmias (reperfusion arrhythmias).[46] Rarely, cholesterol embolization syndrome has been reported, where lysis of an aortic plaque can release cholesterol crystals that embolize to distal sites.[46]
- Other Adverse Events (Ryplazim): In clinical trials for Ryplazim, the most frequently reported adverse reactions (incidence ≥10%) included abdominal pain, bloating, nausea, fatigue, pain in an extremity, hemorrhage (from lesion sites), constipation, dry mouth, headache, dizziness, arthralgia, and back pain.[54]
6.3 Contraindications
Patient selection is the most critical step in mitigating the risks of fibrinolytic therapy. Contraindications are designed to exclude patients in whom the risk of bleeding is unacceptably high.
- Thrombolytic Therapy (e.g., Alteplase):
- Absolute Contraindications: Include any prior intracranial hemorrhage; known structural cerebral vascular lesion (e.g., AVM); known malignant intracranial neoplasm; ischemic stroke within the past 3 months (except the current stroke); suspected aortic dissection; active bleeding or bleeding diathesis (excluding menses); and significant closed-head or facial trauma within the past 3 months.[53]
- Relative Contraindications: These require a careful and individualized risk-benefit assessment. They include severe, uncontrolled hypertension; history of major surgery or serious trauma within the past 2-4 weeks; recent internal bleeding (within 2-4 weeks); non-compressible vascular punctures; pregnancy; and current use of anticoagulants with an INR >1.7.[53]
- Plasminogen Replacement Therapy (Ryplazim): The contraindication profile is vastly different and much narrower, reflecting its different mechanism and patient population. The only absolute contraindication listed is a known hypersensitivity to plasminogen or any other component of the Ryplazim formulation.[55]
6.4 Risk of Infectious Agent Transmission
For products derived from human plasma, such as Ryplazim, there is a theoretical risk of transmitting infectious agents, including viruses and the agents of Creutzfeldt-Jakob disease (CJD and vCJD). This risk is minimized through a multi-layered safety approach that includes stringent screening of plasma donors, testing of donated plasma for pathogens, and the incorporation of robust viral inactivation and removal steps during the manufacturing process.[54]
Table 6: Contraindications for Fibrinolytic and Plasminogen Replacement Therapies
Contraindication | Thrombolytic Therapy (e.g., Alteplase) | Plasminogen Replacement (e.g., Ryplazim) |
---|
Absolute | | |
Prior Intracranial Hemorrhage | Yes | No |
Active Internal Bleeding | Yes | No (Warning for lesion bleeding) |
Known Cerebral AVM or Neoplasm | Yes | No |
Suspected Aortic Dissection | Yes | No |
Recent (3 mo) Stroke or Head Trauma | Yes | No |
Known Hypersensitivity | Yes | Yes |
Relative | | |
Uncontrolled Severe Hypertension | Yes | No |
Recent Major Surgery/Trauma (<3 wks) | Yes | No |
Current Anticoagulant Use (INR >1.7) | Yes | No (Warning/Monitoring required) |
Pregnancy | Yes | No (Use with caution) |
7.0 Manufacturing, Formulation, and Handling
The production of these complex biologic therapies involves sophisticated biopharmaceutical processes, and their formulation dictates specific handling requirements to ensure stability, safety, and efficacy. The manufacturing method itself introduces distinct safety considerations.
7.1 Manufacturing Processes
- Recombinant Protein Production: Thrombolytic agents like Alteplase, Reteplase, and Tenecteplase, as well as investigational recombinant plasminogen, are produced via recombinant DNA technology.[33] This process typically involves expressing the human gene for the protein in a host cell line, such as Chinese Hamster Ovary (CHO) cells for Alteplase or bacterial systems like
E. coli for other proteins.[33] The manufacturing workflow includes large-scale cell culture or fermentation, followed by complex downstream purification processes to isolate the protein of interest and remove host cell proteins and other impurities. For proteins expressed in bacteria, this often includes a challenging protein refolding step to achieve the correct three-dimensional structure.[60] The primary safety concerns for recombinant products relate to potential immunogenicity and ensuring proper protein folding and activity.
- Plasma-Derived Production: Ryplazim is manufactured from large pools of donated human plasma.[30] The process involves cryoprecipitation and chromatographic fractionation techniques to separate and purify plasminogen from thousands of other plasma proteins. A critical component of this manufacturing process is the inclusion of dedicated steps for the inactivation and/or removal of potential viral contaminants to ensure the safety of the final product.[56] The primary safety concern for plasma-derived products is the theoretical risk of transmitting blood-borne pathogens.
7.2 Formulation and Presentation
- Lyophilized Powders: To ensure long-term stability, most of these protein-based therapeutics are formulated as lyophilized (freeze-dried) powders in single-dose vials.[5]
- Reconstitution and Stability: Prior to administration, these powders must be reconstituted with a specific sterile diluent, such as Sterile Water for Injection or 0.9% Sodium Chloride.[35] The reconstitution procedure requires careful aseptic technique to prevent microbial contamination and gentle swirling to avoid foaming and protein denaturation.[35] Once reconstituted, the solutions have a limited shelf-life; for example, Ryplazim must be administered within 3 hours and should not be refrigerated after reconstitution.[35] The complexity of these reconstitution and administration steps, particularly for weight-based dosing of agents like Alteplase, is a significant source of potential medication errors in the clinical setting.[47] This operational complexity drives the clinical appeal of simpler, bolus-administered alternatives.
7.3 Safety and Handling
- Biohazard Precautions: All materials derived from human plasma, including Ryplazim and research-grade plasmin/plasminogen, must be handled with universal precautions, as if they are capable of transmitting infectious agents.[10]
- Personal Protective Equipment (PPE): During preparation and administration, healthcare professionals should use appropriate PPE, including gloves and safety goggles, to prevent exposure.[9]
- Administration: Administration is typically intravenous. For agents like Alteplase, this involves a complex bolus and infusion, while Ryplazim is given as a slow infusion over 10-30 minutes.[47] Extravasation (leakage of the drug into surrounding tissue) should be avoided, as it can cause local inflammation or ecchymosis.[47]
- Disposal: All unused product, vials, syringes, and administration sets should be disposed of as biohazardous medical waste in accordance with institutional and local regulations.[19]
8.0 Synthesis and Future Outlook
The therapeutic landscape of the plasminogen-plasmin system is characterized by well-defined but distinct strategies for acute and chronic conditions, with safety—primarily the risk of hemorrhage—as the overarching challenge. Future progress will likely be driven by a move towards more precise and personalized interventions.
8.1 Current Landscape Summary
The field is currently bifurcated. On one hand, high-intensity, short-duration therapy with plasminogen activators is the standard of care for reversing acute, life-threatening thrombosis. The development of second- and third-generation activators like Reteplase and Tenecteplase has focused on optimizing pharmacokinetics for greater ease of administration (bolus dosing) and enhancing fibrin specificity, in an effort to widen the narrow therapeutic window.[20] On the other hand, low-intensity, long-term plasminogen replacement therapy has proven to be a highly effective and transformative treatment for the rare monogenic disorder of plasminogen deficiency.[30] In both cases, risk management is dominated by careful patient selection through strict adherence to contraindications to mitigate the inherent risk of bleeding.
8.2 Key Knowledge Gaps and Unmet Needs
Despite decades of clinical use, significant challenges and knowledge gaps remain.
- Pharmacokinetics of Direct Plasmin: As previously highlighted, the absence of robust clinical pharmacokinetic and biodistribution data for exogenously administered active plasmin is the single largest gap hindering the rational development of this therapeutic modality.
- Improved Risk Stratification: Current methods for patient selection rely heavily on clinical checklists of contraindications. There is a pressing need for more sophisticated tools, such as validated biomarkers or advanced imaging techniques, to more accurately predict an individual patient's risk of hemorrhage versus their potential benefit from thrombolysis.
- Therapies for Fibrinolytic Shutdown: The complex pathophysiology of trauma-induced coagulopathy, with its biphasic nature of initial hyperfibrinolysis followed by a thrombotic "shutdown," presents an unmet need.[25] Current therapies are ill-equipped to handle this dynamic shift, suggesting a need for modulatory agents rather than simple agonists or antagonists.
8.3 Future Research Directions
The future of fibrinolysis therapy is trending towards greater precision in targeting, design, and application.
- Precision Targeting: The investigation of direct-acting plasmin delivered via intra-arterial catheters represents a move towards physically localizing therapy to the site of the thrombus, thereby minimizing systemic exposure and, theoretically, bleeding risk.[34] Further clinical trials are required to validate the efficacy and safety of this approach.
- Precision Engineering: The next generation of thrombolytic agents will likely continue to leverage protein engineering to create molecules with enhanced fibrin-specificity, greater resistance to inhibition by PAI-1, and finely tuned pharmacokinetic profiles that optimize the balance between efficacy and safety.
- Precision Correction: For congenital disorders like Type I Plasminogen Deficiency, gene therapy offers the ultimate form of precision medicine. By aiming to correct the underlying genetic defect, it holds the potential for a one-time, curative treatment, and early-stage investigations are underway.[31]
- Precision Dosing: The exploration of novel combination therapies, potentially using lower doses of thrombolytics in conjunction with newer, more targeted antiplatelet or anticoagulant agents, may provide a path to achieving effective thrombolysis with a reduced hemorrhagic penalty.
In essence, the evolution of therapy for the plasminogen-plasmin system is moving away from the "one-size-fits-all" approach of systemic thrombolysis. The future lies in a multi-pronged, precision-oriented strategy that tailors the intervention—be it a precisely engineered molecule, a physically targeted delivery, or a genetically corrective therapy—to the specific patient and their unique underlying pathology.
Works cited
- Physiology, Plasminogen Activation | DrugBank Online, accessed September 28, 2025, https://go.drugbank.com/articles/A236035
- Plasminogen Completed Phase 1 Trials for Type I Plasminogen Deficiency Treatment | DrugBank Online, accessed September 28, 2025, https://go.drugbank.com/drugs/DB16701/clinical_trials?conditions=DBCOND0074482&phase=1&purpose=treatment&status=completed
- Plasminogen Completed Phase 4 Trials for Acute Myocardial Infarction With ST Segment Elevation Treatment - DrugBank, accessed September 28, 2025, https://go.drugbank.com/drugs/DB16701/clinical_trials?conditions=DBCOND0039867&phase=4&purpose=treatment&status=completed
- Plasminogen from human plasma | 9001-91-6 | JAA00191 - Biosynth, accessed September 28, 2025, https://www.biosynth.com/p/JAA00191/9001-91-6-plasminogen-from-human-plasma
- Plasminogen, Human Plasma 9001-91-6 - Sigma-Aldrich, accessed September 28, 2025, https://www.sigmaaldrich.com/US/en/product/mm/528175
- Plasminogen, EACA- and Lysine-Free, Human Plasma 9001-91-6, accessed September 28, 2025, https://www.merckmillipore.com/IL/en/product/mm/528178
- Plasminogen, EACA- and Lysine-Free, Human Plasma 9001-91-6 - Sigma-Aldrich, accessed September 28, 2025, https://www.sigmaaldrich.com/BZ/en/product/mm/528178
- Plasminogen - PubChem, accessed September 28, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/9001-91-6
- Safety Data Sheet - MedchemExpress.com, accessed September 28, 2025, https://file.medchemexpress.com/batch_PDF/HY-108883/Plasmin-Human-plasma-SDS-MedChemExpress.pdf
- SAFETY DATA SHEET - Sigma-Aldrich, accessed September 28, 2025, https://www.sigmaaldrich.com/US/en/sds/SIGMA/P1867
- Plasmin - Wikipedia, accessed September 28, 2025, https://en.wikipedia.org/wiki/Plasmin
- PLG - Plasminogen - Homo sapiens (Human) | UniProtKB | UniProt, accessed September 28, 2025, https://www.uniprot.org/uniprotkb/P00747/entry
- Plasmin - from human plasma - Sigma-Aldrich, accessed September 28, 2025, https://www.sigmaaldrich.com/US/en/product/roche/10602361001
- (PDF) Structure and Function of Plasminogen/Plasmin System - ResearchGate, accessed September 28, 2025, https://www.researchgate.net/publication/287255459_Structure_and_Function_of_PlasminogenPlasmin_System
- The plasmin–antiplasmin system: structural and functional aspects ..., accessed September 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11115092/
- Plasminogen (PLG) - DiaPharma, accessed September 28, 2025, https://diapharma.com/resources/plasminogen-plg/
- New insights into the structure and function of the plasminogen/plasmin system - PubMed, accessed September 28, 2025, https://pubmed.ncbi.nlm.nih.gov/24252474/
- Physiology, Plasminogen Activation - StatPearls - NCBI Bookshelf, accessed September 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK539745/
- Plasminogen activator - Wikipedia, accessed September 28, 2025, https://en.wikipedia.org/wiki/Plasminogen_activator
- Thrombolytic (Fibrinolytic) Drugs - CV Pharmacology, accessed September 28, 2025, https://cvpharmacology.com/thrombolytic/thrombolytic
- Mechanism of Action of Plasminogen Activators - Thieme Connect, accessed September 28, 2025, https://www.thieme-connect.com/products/ejournals/pdf/10.1055/s-0037-1615941.pdf
- PROPERTIES OF HUMAN PLASMIN - Canadian Science Publishing, accessed September 28, 2025, https://cdnsciencepub.com/doi/pdf/10.1139/o62-008
- What is the mechanism of Plasmin? - Patsnap Synapse, accessed September 28, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-plasmin
- The Fibrinolytic System and Its Measurement: History, Current Uses and Future Directions for Diagnosis and Treatment - PMC - PubMed Central, accessed September 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10532028/
- The Role of Fibrinolytic System in Health and Disease - MDPI, accessed September 28, 2025, https://www.mdpi.com/1422-0067/23/9/5262
- Plasminogen and plasmin can bind to human T cells and generate truncated CCL21 that increases dendritic cell chemotactic responses - PMC, accessed September 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9270246/
- Plasminogen | DrugBank Online, accessed September 28, 2025, https://go.drugbank.com/polypeptides/P00747
- Recent Advances on Plasmin Inhibitors for the Treatment of Fibrinolysis-Related Disorders - PMC - PubMed Central, accessed September 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8788159/
- What is Plasmin used for? - Patsnap Synapse, accessed September 28, 2025, https://synapse.patsnap.com/article/what-is-plasmin-used-for
- Ryplazim® (Plasminogen, Human-Tvmh) – Community Plan Medical Benefit Drug Policy - UHCprovider.com, accessed September 28, 2025, https://www.uhcprovider.com/content/dam/provider/docs/public/policies/medicaid-comm-plan/ryplazim-cs.pdf
- New Plasminogen Deficiency treatments 2025 | Everyone.org, accessed September 28, 2025, https://everyone.org/explore/treatment/?id=90
- Thrombolytic (Fibrinolytic) Drugs - CV Pharmacology, accessed September 28, 2025, https://cvpharmacology.com/thrombolytic
- Alteplase: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 28, 2025, https://go.drugbank.com/drugs/DB00009
- Grifols Publishes Positive Phase I Safety Results for Plasmin, a ..., accessed September 28, 2025, https://www.prnewswire.com/news-releases/grifols-publishes-positive-phase-i-safety-results-for-plasmin-a-novel-direct-acting-thrombolytic-in-the-journal-of-thrombosis-and-haemostasis-158416785.html
- Ryplazim (plasminogen) dosing, indications, interactions, adverse effects, and more, accessed September 28, 2025, https://reference.medscape.com/drug/ryplazim-plasminogen-1000182
- Application of a plasmin generation assay to define pharmacodynamic effects of tranexamic acid in women undergoing cesarean delivery - PMC, accessed September 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7875467/
- Plasmin-Mediated Activation of Contact System in Response to Pharmacological Thrombolysis | Circulation - AHA Journals, accessed September 28, 2025, https://www.ahajournals.org/doi/10.1161/01.cir.91.1.28
- Tissue plasminogen activator (tPA) inhibits plasmin degradation of fibrin. A mechanism that slows tPA-mediated fibrinolysis but does not require alpha 2-antiplasmin or leakage of intrinsic plasminogen - PubMed, accessed September 28, 2025, https://pubmed.ncbi.nlm.nih.gov/7769094/
- Tissue Plasminogen Activator - StatPearls - NCBI Bookshelf, accessed September 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK507917/
- Tissue Plasminogen Activator Therapy - StatPearls - NCBI Bookshelf, accessed September 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK482376/
- Measurement and Quantitative Characterization of Whole-Body Pharmacokinetics of Exogenously Administered T Cells in Mice - PubMed Central, accessed September 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6382992/
- Measurement and Quantitative Characterization of Whole-Body Pharmacokinetics of Exogenously Administered T Cells in Mice - PubMed, accessed September 28, 2025, https://pubmed.ncbi.nlm.nih.gov/30622170/
- Plasminogen, human Interactions - Drugs.com, accessed September 28, 2025, https://www.drugs.com/drug-interactions/plasminogen-human.html
- Plasminogen: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed September 28, 2025, https://www.rxlist.com/plasminogen/generic-drug.htm
- Safety of plasmin in the setting of concomitant aspirin and heparin administration in an animal model of bleeding - PubMed, accessed September 28, 2025, https://pubmed.ncbi.nlm.nih.gov/14675099/
- Alteplase Side Effects: Common, Severe, Long Term - Drugs.com, accessed September 28, 2025, https://www.drugs.com/sfx/alteplase-side-effects.html
- Dosing & Administration Guidelines for Activase® (alteplase), accessed September 28, 2025, https://www.activase.com/ais/dosing-and-administration/dosing.html
- Activase, TPA (alteplase) dosing, indications, interactions, adverse effects, and more, accessed September 28, 2025, https://reference.medscape.com/drug/activase-tpa-alteplase-342287
- Indication, administration, and dosing of tissue plasminogen activators. - ResearchGate, accessed September 28, 2025, https://www.researchgate.net/figure/Indication-administration-and-dosing-of-tissue-plasminogen-activators_tbl1_330856346
- Plasminogen, Human-tmvh Monograph for Professionals - Drugs.com, accessed September 28, 2025, https://www.drugs.com/monograph/plasminogen-human-tmvh.html
- What are the side effects of Plasmin? - Patsnap Synapse, accessed September 28, 2025, https://synapse.patsnap.com/article/what-are-the-side-effects-of-plasmin
- Tissue plasminogen activator (tPA): How does it treat a stroke? - Medical News Today, accessed September 28, 2025, https://www.medicalnewstoday.com/articles/tpa-stroke
- What are the contraindications for tissue plasminogen activator (tPA)? - Dr.Oracle AI, accessed September 28, 2025, https://www.droracle.ai/articles/205669/contraindications-s-for-tpa
- Plasminogen, Human Side Effects: Common, Severe, Long Term, accessed September 28, 2025, https://www.drugs.com/sfx/plasminogen-human-side-effects.html
- Package Insert - RYPLAZIM - FDA, accessed September 28, 2025, https://www.fda.gov/media/149806/download
- The First FDA-approved Human Plasminogen Indicated for PLGD-1 - Ryplazim, accessed September 28, 2025, https://www.ryplazim.com/healthcare-provider/about-ryplazim/
- What are the contraindications of Tissue Plasminogen Activator (tPA)? - Dr.Oracle AI, accessed September 28, 2025, https://www.droracle.ai/articles/54925/what-are-the-contraindications-of-tissue-plasminogen-activator-tpa
- What are the contraindications for Tissue Plasminogen Activator (tPA)? - Dr.Oracle AI, accessed September 28, 2025, https://droracle.ai/articles/66043/what-are-the-contraindications-for-tissue-plasminogen-activator-tpa
- tPA Contraindications for Ischemic Stroke - MDCalc, accessed September 28, 2025, https://www.mdcalc.com/calc/1934/tpa-contraindications-ischemic-stroke
- US20100144622A1 - Methods for production of recombinant ..., accessed September 28, 2025, https://patents.google.com/patent/US20100144622A1/en
- Waisman Biomanufacturing: Home, accessed September 28, 2025, https://gmpbio.org/
- SAFETY DATA SHEET - BioMedica Diagnostics, accessed September 28, 2025, https://biomedicadiagnostics.com/wp-content/uploads/2019/03/400410_Human-Glu-Plasminogen-SDS_US-English_v2.pdf