MedPath

Eculizumab Advanced Drug Monograph

Published:Jul 17, 2025

Generic Name

Eculizumab

Brand Names

Soliris, Epysqli, Bekemv

Drug Type

Biotech

CAS Number

219685-50-4

Associated Conditions

Generalized Myasthenia Gravis, Neuromyelitis Optica Spectrum Disorders, Paroxysmal Nocturnal Haemoglobinuria (PNH), Thrombotic Microangiopathies

Comprehensive Analysis of Eculizumab: A First-in-Class Terminal Complement Inhibitor

1.0 Executive Monograph: Eculizumab (Soliris) - A Definitive Analysis of a First-in-Class Complement Inhibitor

1.1 Overview

Eculizumab, marketed principally under the brand name Soliris, represents a landmark achievement in biotechnology and a paradigm shift in the treatment of a select group of rare, life-threatening diseases driven by dysregulation of the complement system.[1] As a first-in-class terminal complement inhibitor, this recombinant humanized monoclonal antibody has fundamentally altered the natural history and management of conditions such as paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), generalized myasthenia gravis (gMG), and neuromyelitis optica spectrum disorder (NMOSD).[1] Its development and approval validated the complement cascade as a viable and potent therapeutic target.

The therapeutic success of Eculizumab is rooted in its highly specific mechanism of action: it binds with high affinity to the human complement protein C5, thereby preventing its cleavage and halting the formation of the pro-inflammatory anaphylatoxin C5a and the cytolytic terminal complement complex C5b-9, also known as the Membrane Attack Complex (MAC).[1] This targeted intervention effectively arrests the downstream pathological events—such as intravascular hemolysis in PNH and thrombotic microangiopathy in aHUS—that define these diseases.[5] However, this precise mechanism is also the source of its most significant safety liability. By inhibiting the terminal complement pathway, a key component of the innate immune system's defense against encapsulated bacteria, Eculizumab confers a substantial and predictable risk of severe and potentially fatal meningococcal infections. This risk necessitated the implementation of a stringent Risk Evaluation and Mitigation Strategy (REMS) program by regulatory bodies, mandating prescriber education, patient vaccination, and continuous vigilance.[7]

Following its initial FDA approval in 2007, Eculizumab enjoyed over a decade of market exclusivity, establishing itself as a cornerstone therapy and a commercial blockbuster despite its designation as one of the world's most expensive drugs.[1] The recent expiration of its core patents has ushered in a new era characterized by the market entry of biosimilars and the strategic development of next-generation complement inhibitors by the originator, Alexion Pharmaceuticals (now Alexion AstraZeneca Rare Disease), aiming to improve upon Eculizumab's therapeutic profile, particularly regarding treatment burden and residual disease activity.[9]

1.2 Core Therapeutic Thesis

The clinical and commercial success of Eculizumab is predicated on a highly specific and potent mechanism of action. By targeting the terminal complement protein C5, it surgically disrupts the final, destructive endpoint of complement-mediated diseases while largely preserving the critical upstream functions of the immune system, such as opsonization for pathogen clearance.[4] This targeted approach provides profound therapeutic benefit in diseases where terminal complement activation is the primary driver of pathology. This very specificity, however, is intrinsically and inseparably linked to its primary safety liability—a heightened susceptibility to infections caused by encapsulated organisms, most notably

Neisseria meningitidis. Therefore, the entire clinical framework surrounding Eculizumab, from its dosing to its mandatory safety protocols, is a direct and logical consequence of this finely balanced therapeutic index.

1.3 Scope of Report

This report provides an exhaustive, expert-level analysis of Eculizumab. It integrates and synthesizes data spanning the drug's molecular engineering and discovery, its comprehensive pharmacological profile, the pivotal clinical trial evidence supporting its multiple indications, and its extensive post-marketing safety record. Furthermore, it examines the drug's global regulatory trajectory, its evolving role in clinical practice amidst the advent of biosimilars, and the forward-looking landscape of complement inhibition, which Eculizumab itself pioneered. The analysis is intended for a professional audience requiring a deep, nuanced understanding of this transformative therapeutic agent.

2.0 Molecular Profile and Physicochemical Properties

2.1 Identification and Nomenclature

Eculizumab is a biologic drug substance identified by a consistent set of names and identifiers across global regulatory and scientific databases, which is critical for accurate reporting and research.

  • Generic Name: The universally recognized non-proprietary name is Eculizumab.[1] In Japan, it may be referred to as Eculizumab (genetical recombination).[12]
  • Brand Names: The original and most widely known brand name is Soliris.[1] With the advent of biosimilars, other brand names have been approved, including Bekemv, Bkemv, and Epysqli.[5]
  • Chemical and Database Identifiers: For precise scientific and regulatory tracking, Eculizumab is assigned unique codes:
  • DrugBank ID: DB01257.[5]
  • CAS (Chemical Abstracts Service) Number: 219685-50-4.[1]
  • Unique Ingredient Identifier (UNII): A3ULP0F556.[13]
  • Synonyms and Developmental Codes: During its development and in research literature, it has been referred to by various synonyms, including Anti-Human C5 Humanized Antibody, 5G1.1, h5G1.1, and Anti-C5 monoclonal antibody 5G1-1.[5]

2.2 Chemical and Structural Characteristics

Eculizumab is a complex protein-based therapeutic, and its structure is fundamental to its function and safety profile.

  • Drug Type: It is classified as a biotech product, specifically a protein-based therapy. Its molecular class is a recombinant humanized monoclonal antibody.[1]
  • Structure: Eculizumab is an immunoglobulin G (IgG) antibody. More specifically, it is a hybrid IgG2/4κ molecule, meaning its constant region is a composite of human IgG2 and IgG4 sequences, with a kappa light chain.[1] This hybrid structure is designed to reduce effector functions like antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which are undesirable for an inhibitory antibody. The antibody is composed of two identical heavy chains, each 448 amino acids in length, and two identical light chains, each 214 amino acids long.[1]
  • Molecular Weight: The approximate molecular weight of the entire antibody is 148 kilodaltons (kDa).[1]
  • Production: The originator product, Soliris, is produced using recombinant DNA technology within a murine (mouse) myeloma NS0 cell line.[15] Some biosimilars, such as Bekemv, utilize a Chinese Hamster Ovary (CHO) cell line for production.[17]

The development of Eculizumab as a "humanized" antibody was a critical technological decision that directly underpins its viability as a long-term therapy. Early therapeutic monoclonal antibodies were entirely of murine origin. When administered to humans, these mouse proteins often provoked a significant immune response known as the Human Anti-Mouse Antibody (HAMA) response. This immune reaction could lead to hypersensitivity reactions and, more critically for chronic treatment, the generation of neutralizing antibodies that would bind to and inactivate the therapeutic agent, rendering it ineffective over time. The process of humanization addresses this challenge through sophisticated genetic engineering. It involves replacing the vast majority of the murine antibody protein sequence with corresponding human sequences. Only the small, hypervariable loops known as the complementarity-determining regions (CDRs)—the precise parts of the antibody that recognize and bind to the target antigen (in this case, C5)—are retained from the original mouse antibody.[1] By grafting these murine CDRs onto a human antibody framework, the resulting molecule is overwhelmingly human in composition, which dramatically reduces its immunogenicity. This engineering is a foundational element of Eculizumab's clinical success, minimizing the risk of neutralization and severe allergic reactions, and thereby permitting the repeated, bi-weekly infusions necessary for the chronic management of diseases like PNH and aHUS.


Table 2.1: Key Identifiers and Physicochemical Properties of Eculizumab

PropertyValueSource(s)
Generic NameEculizumab1
Primary Brand NameSoliris1
DrugBank IDDB012575
CAS Number219685-50-41
Drug TypeBiotech, Recombinant Humanized Monoclonal Antibody1
Antibody ClassIgG2/4κ1
Molecular WeightApprox. 148 kDa1
TargetComplement Protein C55

3.0 Clinical Pharmacology: Elucidation of the Terminal Complement C5 Inhibition Mechanism

3.1 Primary Target: Complement Protein C5

The therapeutic activity of Eculizumab is derived from its highly specific and high-affinity binding to a single molecular target: the human complement protein C5.[1] The complement system is a crucial part of the innate immune system, acting as a cascade of enzymatic reactions that helps clear pathogens and damaged cells. C5 is a pivotal protein positioned at the junction of the three main complement activation pathways (classical, lectin, and alternative) and the final, common terminal pathway.[4] Eculizumab's interaction is highly species-specific; it is monospecific for human C5 and does not bind to the C5 protein of other species, including mice.[4] This specificity required the use of specialized animal models or surrogate antibodies during preclinical development but ensures a highly targeted effect in humans.

3.2 Mechanism of Action (MOA)

Eculizumab functions as a terminal complement inhibitor.[1] Upon administration, it circulates in the plasma and binds to C5. This binding sterically hinders the C5 protein, preventing it from being cleaved by its activating enzyme, the C5 convertase.[1] The cleavage of C5 is the rate-limiting step for the activation of the terminal complement pathway. By blocking this single step, Eculizumab effectively prevents the generation of the two biologically active downstream fragments [4]:

  • C5a: A small peptide that is released upon cleavage. C5a is a potent anaphylatoxin and a powerful chemoattractant for immune cells. It promotes inflammation, increases vascular permeability, and has pro-thrombotic (pro-clotting) properties.[1]
  • C5b: The larger fragment that remains after cleavage. C5b is the initiating component for the assembly of the terminal complement complex (TCC). It sequentially binds to other complement proteins (C6, C7, C8, and multiple copies of C9) to form the C5b-9 complex, also known as the Membrane Attack Complex (MAC).[1]

By preventing the formation of both C5a and C5b, Eculizumab ablates the key effector functions of the terminal complement system. It neutralizes the potent pro-inflammatory and pro-thrombotic signaling of C5a and, most critically, halts the formation of the cell-lysing MAC.[4]

The decision to target C5, rather than an upstream component like C3, represents a masterful example of strategic pharmacological design aimed at maximizing efficacy while managing safety. The three major pathways of the complement system all converge at the activation of C3, making it a central node. The products of C3 activation, particularly C3b, are essential for opsonization—the process of "tagging" pathogens for phagocytosis and clearance by immune cells. This is a fundamental host defense mechanism. A therapeutic agent that blocked C3 would be a profoundly potent and broad-spectrum immunosuppressant, likely leading to an unacceptably high risk of infections from a wide array of common bacteria and fungi.

In contrast, C5 acts at a much later stage in the cascade.[4] By targeting C5, Eculizumab executes a "surgical strike" on the specific parts of the immune system driving the pathology in its target diseases, namely the lytic activity of the MAC and the intense inflammation driven by C5a. This approach leaves the critical disease-preventing functions of the proximal complement system, including C3b-mediated opsonization, largely preserved and functional.[4] This strategic precision is fundamental to the drug's therapeutic index, allowing for profound efficacy in complement-driven diseases while narrowing the scope of its immunosuppressive effect to a predictable vulnerability to a specific class of pathogens—encapsulated bacteria—which rely on complement evasion for survival.

3.3 Pathophysiological Consequences of C5 Inhibition

The clinical benefits of Eculizumab are a direct result of interrupting the disease-specific pathophysiology driven by terminal complement activation.

  • In Paroxysmal Nocturnal Hemoglobinuria (PNH): PNH is caused by a somatic mutation in the PIGA gene in a hematopoietic stem cell, leading to a deficiency of all glycosylphosphatidylinositol (GPI)-anchored proteins on the surface of its clonal progeny, including red blood cells. One of these missing proteins is CD59, a key regulator that protects cells from MAC-mediated lysis. In the absence of CD59, PNH red blood cells are exquisitely sensitive to destruction by the complement system. Eculizumab, by blocking MAC formation, directly prevents this chronic, uncontrolled intravascular hemolysis, thereby reducing anemia, transfusion dependence, and the debilitating symptoms of the disease.[1]
  • In Atypical Hemolytic Uremic Syndrome (aHUS): aHUS is a genetic disease characterized by chronic, uncontrolled activation of the alternative complement pathway, leading to systemic thrombotic microangiopathy (TMA). This process involves the formation of microthrombi (small blood clots) in small blood vessels throughout the body, particularly in the kidneys. The MAC damages endothelial cells lining these vessels, triggering platelet activation, thrombosis, and subsequent organ damage. Eculizumab inhibits this complement-mediated TMA, protecting the endothelium, allowing for platelet count recovery, and often leading to dramatic improvement in kidney function.[3]
  • In Generalized Myasthenia Gravis (gMG): In patients with AChR-antibody-positive gMG, autoantibodies bind to the acetylcholine receptor (AChR) at the neuromuscular junction. This binding activates the classical complement pathway, leading to the deposition of the C5b-9 MAC on the postsynaptic membrane. This MAC-mediated damage destroys the structural integrity of the neuromuscular junction, leading to impaired neuromuscular transmission and muscle weakness. While the precise mechanism is still under investigation, it is presumed that Eculizumab works by reducing this TCC deposition, thereby preserving the structure and function of the neuromuscular junction.[6]
  • In Neuromyelitis Optica Spectrum Disorder (NMOSD): NMOSD is a severe autoimmune disease of the central nervous system driven by autoantibodies against the aquaporin-4 (AQP4) water channel, which is highly expressed on astrocytes. The binding of these AQP4-IgG antibodies to astrocytes is a potent activator of the classical complement pathway. This leads to MAC-mediated destruction of astrocytes, which in turn causes secondary demyelination of neurons and severe neurological damage. Eculizumab is presumed to exert its therapeutic effect by inhibiting this AQP4-antibody-induced, MAC-dependent astrocyte injury, thereby preventing inflammatory relapses.[3]

4.0 Pharmacokinetic and Pharmacodynamic Profile: A Quantitative Analysis

4.1 Pharmacokinetics (PK)

The pharmacokinetic profile of Eculizumab is characteristic of a large therapeutic protein and is a key determinant of its dosing regimen, clinical utility, and safety considerations.

  • Absorption: As a monoclonal antibody, Eculizumab would be degraded in the gastrointestinal tract and therefore must be administered parenterally. It is given exclusively by intravenous (IV) infusion, which results in immediate and complete (100%) bioavailability.[5]
  • Distribution: The drug exhibits a small volume of distribution (Vd​), estimated to be between 5 and 8 Liters in a typical 70 kg patient.[1] This value approximates the volume of blood plasma, indicating that Eculizumab does not distribute extensively into peripheral tissues and remains primarily within the intravascular compartment. This is consistent with its large molecular size and its mechanism of targeting a plasma-based protein (C5).
  • Metabolism: Eculizumab is not metabolized by the hepatic cytochrome P450 (CYP450) enzyme system, which is the primary pathway for small-molecule drug metabolism. Instead, as a therapeutic protein, it is presumed to undergo catabolism through general protein degradation pathways. This involves uptake by cells of the reticuloendothelial system and breakdown into small peptides and constituent amino acids by lysosomal enzymes.[1] These amino acids are then recycled into the body's general amino acid pool.
  • Elimination: Consistent with its metabolic pathway, Eculizumab is not eliminated in any meaningful quantity through renal or biliary excretion.[5] Its clearance is primarily driven by the rate of its catabolism. The terminal elimination half-life ( t1/2​) is notably long and shows significant inter-individual variability, with a reported range of 141 to 882 hours (approximately 6 to 37 days).[6]
  • Steady State: Due to its long half-life, steady-state concentrations are achieved approximately 4 weeks after the initiation of the recommended dosing regimen. At steady state, drug levels are approximately 2-fold higher than after the first dose, indicating a moderate degree of accumulation.[6] Population pharmacokinetic analyses have demonstrated that its PK properties are dose-linear and time-independent within the clinically approved dose range of 600 mg to 1200 mg.[6]

4.2 Pharmacodynamics (PD)

The pharmacodynamic effect of Eculizumab is the direct, measurable consequence of its mechanism of action on the complement system.

  • Biomarker of Activity: The primary pharmacodynamic goal of Eculizumab therapy is to achieve and maintain complete blockade of terminal complement activity. Extensive clinical data have established that this is reliably achieved when free (unbound) plasma concentrations of C5 are suppressed to below 0.5 mcg/mL.[6] The maintenance dosing regimens for all indications are designed to keep C5 levels below this therapeutic threshold.
  • Clinical Effect in PNH: The pharmacodynamic effect translates directly to clinical biomarkers of disease activity. In pivotal clinical trials for PNH, Eculizumab administration led to a rapid and profound reduction in serum lactate dehydrogenase (LDH), a key biomarker of intravascular hemolysis. For example, in the placebo-controlled PNH Study 1, mean LDH levels in treated patients plummeted from a baseline of approximately 2200 U/L to approximately 700 U/L within the first week of treatment. This effect was sustained, with levels further decreasing to a mean of 327 U/L by the end of the 26-week study, demonstrating a rapid onset and durable pharmacodynamic response.[6]

4.3 Impact of Concomitant Procedures and Patient Factors

The pharmacokinetic profile of Eculizumab is a double-edged sword, offering both advantages and vulnerabilities that directly inform clinical management. Its independence from renal or hepatic clearance is a major clinical asset, particularly in the treatment of aHUS, a disease defined by acute kidney injury and renal failure. The ability to use a drug for a kidney disease that is not itself cleared by the kidneys provides a robust and predictable dosing profile in critically ill patients whose organ function may be compromised or fluctuating.[6]

However, the drug's nature as a large, plasma-bound protein makes it highly susceptible to removal by extracorporeal therapies that process large volumes of plasma. Procedures such as plasmapheresis (plasma exchange, PE) or plasma infusion (PI) can dramatically increase the clearance of Eculizumab. It has been shown that these procedures increase clearance by approximately 6-fold, slashing the drug's half-life to around 90 hours.[6] This rapid removal would lead to a loss of therapeutic effect and potential disease rebound. Consequently, specific supplemental dosing of Eculizumab is required for patients undergoing these procedures to maintain therapeutic concentrations.[19]

Furthermore, the drug's long half-life has significant clinical implications. While it allows for a convenient bi-weekly maintenance dosing schedule, it also means that the drug's immunosuppressive effects persist for a considerable time after treatment is stopped. The risk of meningococcal infection, for instance, continues for up to 3 months after the final dose, necessitating extended patient counseling and vigilance.[21]

Finally, the pharmacokinetics of Eculizumab are not significantly affected by patient-specific factors such as age (from 2 months to 85 years), sex, or race.[6] This consistency simplifies dosing across diverse patient populations, with the notable exception of pediatrics, where weight-based dosing is required.

5.0 Pivotal Clinical Evidence and Therapeutic Efficacy in Approved Indications

The clinical development of Eculizumab is a case study in a strategic, indication-by-indication expansion, beginning with ultra-rare diseases where the biological rationale was ironclad and progressing to more complex conditions. This phased approach first validated the drug's mechanism in settings of high unmet need, subsequently providing the clinical and financial foundation to pursue broader indications.

5.1 Paroxysmal Nocturnal Hemoglobinuria (PNH): Establishing the Standard of Care

  • Indication: Eculizumab is approved for the treatment of adult and pediatric patients with PNH to reduce hemolysis.[3]
  • Clinical Trial Evidence: The initial regulatory approvals were granted based on data from small but highly impactful clinical trials, a common path for orphan drugs targeting ultra-rare diseases.[1] The TRIUMPH study was a pivotal Phase 3, randomized, placebo-controlled trial in transfusion-dependent PNH patients. The SHEPHERD study (NCT00130000) was a crucial open-label trial that further established the drug's safety and efficacy in a broader PNH population.[22] Together, these and other studies (e.g., NCT00122317) consistently demonstrated that Eculizumab produced a rapid and sustained reduction in intravascular hemolysis, as measured by a dramatic decrease in LDH levels.[1] This biological effect translated into significant clinical benefits, most notably a marked reduction in the need for red blood cell transfusions and stabilization of hemoglobin concentrations.[8]
  • Outcomes: For patients with PNH, Eculizumab significantly improves quality of life, primarily by alleviating the profound fatigue associated with chronic hemolysis.[1] While it effectively controls the hemolytic component of the disease, long-term data suggest it does not appear to alter the underlying risk of death or the natural progression of the bone marrow failure state toward conditions like myelodysplastic syndrome (MDS) or acute myelogenous leukemia (AML).[1] Similarly, its effect on the risk of thrombosis, a major cause of mortality in PNH, is not fully established, and patients may require continued anticoagulation.[1]

5.2 Atypical Hemolytic Uremic Syndrome (aHUS): Inhibiting Complement-Mediated Thrombotic Microangiopathy

  • Indication: Eculizumab is approved for the treatment of adult and pediatric patients with aHUS to inhibit complement-mediated thrombotic microangiopathy (TMA).[3] It was granted an orphan drug designation for this use.[1]
  • Limitation of Use: It is critically important to distinguish aHUS from other forms of TMA. Eculizumab is explicitly not indicated for the treatment of patients with Shiga toxin-producing E. coli related HUS (STEC-HUS), as the underlying pathophysiology of that condition is different.[3]
  • Clinical Trial Evidence: The FDA approval in September 2011 was based on evidence from two small, prospective, single-arm trials that enrolled a total of 37 patients with aHUS (including trials NCT01193348 and NCT01194973).[1] Given the rarity and severity of the disease, a placebo-controlled trial was not feasible. These studies demonstrated that treatment with Eculizumab led to a rapid improvement in hematologic parameters characteristic of TMA, including a significant increase in platelet counts.[8] Furthermore, many patients experienced a substantial improvement in renal function, with a significant number being able to discontinue dialysis.[8]

5.3 Generalized Myasthenia Gravis (gMG): Targeting Complement at the Neuromuscular Junction

  • Indication: Eculizumab is approved for the treatment of adult and pediatric patients aged six years and older with generalized myasthenia gravis (gMG) who are positive for anti-acetylcholine receptor (AChR) antibodies.[3]
  • Clinical Trial Evidence: The cornerstone of the adult approval was the REGAIN study (NCT01997229), a Phase 3, randomized, double-blind, placebo-controlled, multicenter trial.[27] This study evaluated the efficacy and safety of Eculizumab in patients with refractory gMG, meaning they had failed prior treatment with at least two immunosuppressive agents or required chronic plasma exchange or IVIg. The results demonstrated a clinically meaningful improvement in measures of daily living and muscle strength in the Eculizumab group compared to placebo.[27] The subsequent approval for pediatric use was supported by the robust data from the adult trials, supplemented by pharmacokinetic and safety data from a small, single-arm study involving 11 adolescent patients.[26] International clinical guidelines now recommend considering Eculizumab for patients with severe, refractory anti-AChR antibody-positive gMG.[8]

5.4 Neuromyelitis Optica Spectrum Disorder (NMOSD): A Novel Approach for an Autoimmune Channelopathy

  • Indication: Eculizumab is approved for the treatment of adult patients with NMOSD who are positive for anti-aquaporin-4 (AQP4) antibodies.[1]
  • Clinical Trial Evidence: The regulatory approval for NMOSD was based on the results of the PREVENT trial (NCT02003144), a pivotal Phase 3, randomized, double-blind, placebo-controlled study.[14] The trial enrolled patients with AQP4-IgG-positive NMOSD and evaluated the efficacy of Eculizumab in preventing disease relapses, which are the primary cause of cumulative disability in this condition. The results were compelling: Eculizumab demonstrated a dramatic reduction in the risk of relapse compared to placebo. At 48 weeks, 98% of patients treated with Eculizumab remained relapse-free, establishing it as a highly effective therapy for preventing the devastating neurological attacks characteristic of NMOSD.[28]

6.0 Comprehensive Safety and Tolerability Assessment

The safety profile of Eculizumab is unique and is dominated by a single, predictable, and severe risk that is a direct consequence of its therapeutic mechanism. This has necessitated the implementation of an extensive risk management framework to ensure its benefits outweigh its risks.

6.1 The Black Box Warning: Risk of Serious Meningococcal Infections and the Soliris REMS Program

  • The Warning: Eculizumab carries a U.S. Food and Drug Administration (FDA) Boxed Warning, the agency's most stringent warning, for an increased risk of serious, life-threatening, and potentially fatal meningococcal infections caused by the bacterium Neisseria meningitidis.[3]
  • The Mechanism of Risk: This profound susceptibility to meningococcal disease is not an off-target effect but a direct and unavoidable consequence of the drug's mechanism of action. The terminal complement pathway, which Eculizumab blocks, is the human immune system's primary defense mechanism for recognizing and lysing encapsulated bacteria like N. meningitidis. By inhibiting the formation of the Membrane Attack Complex, Eculizumab effectively disables this crucial line of defense.
  • Risk Evaluation and Mitigation Strategy (REMS): Because of this severe and predictable risk, Eculizumab is available only through a restricted distribution program known as the ULTOMIRIS and SOLIRIS REMS.[7] This program imposes strict requirements on all stakeholders to mitigate the risk of meningococcal infection:
  • Prescriber Requirements: Physicians who prescribe Eculizumab must first enroll in the REMS program. This enrollment certifies that they are aware of the risks and are committed to following the program's requirements. They must counsel patients on the signs and symptoms of meningitis, provide them with specific educational materials, and ensure that patients are compliant with all vaccination and, if necessary, prophylactic antibiotic requirements.[7]
  • Patient Requirements: Patients must be vaccinated against meningococcal serogroups A, C, W, and Y, as well as serogroup B, at least two weeks prior to receiving their first dose of Eculizumab. If therapy must be initiated urgently before vaccination can take effect, the patient must receive prophylactic antibacterial drugs for the duration of the vaccination period. Crucially, patients must be given and instructed to carry a Patient Safety Card at all times during treatment and for at least 3 months following their last dose, as the risk persists due to the drug's long half-life. This card alerts any healthcare provider to the patient's increased risk, facilitating rapid diagnosis and treatment if an infection is suspected.[7]
  • Pharmacy and Healthcare Setting Requirements: Pharmacies, hospitals, and infusion centers that dispense or administer Eculizumab must be certified in the REMS program. They are responsible for verifying that the prescriber is certified before dispensing the drug.[8]

This entire safety framework—the MOA, the resulting immunodeficiency, the Black Box Warning, and the multi-layered REMS program—represents a clear causal chain. It is not a collection of disparate safety concerns but a single, central risk with a comprehensive, codified system designed to manage it. Understanding this direct link between pharmacology and public health intervention is essential for the safe and effective use of Eculizumab.

6.2 Analysis of Adverse Reactions Across Patient Populations

Beyond the risk of meningococcal disease, Eculizumab is associated with a range of other adverse reactions, the incidence of which can vary by the patient population being treated.

  • Common Adverse Effects: These are adverse reactions reported in 10% or more of patients in pivotal clinical trials.
  • In PNH: The most frequently reported adverse reactions include headache, nasopharyngitis (the common cold), back pain, and nausea.[1]
  • In aHUS: Patients in aHUS trials, who are often more acutely ill, reported a broader range of common adverse events (≥20% incidence), including headache, diarrhea, hypertension, upper respiratory infection, abdominal pain, vomiting, nasopharyngitis, anemia, cough, peripheral edema, nausea, urinary tract infections, and pyrexia (fever).[1]
  • In gMG: The most prominent adverse reaction reported at a rate of ≥10% in the placebo-controlled trial was musculoskeletal pain.[3]
  • In NMOSD: Common adverse events (≥10%) included upper respiratory infection, nasopharyngitis, diarrhea, back pain, dizziness, influenza, arthralgia (joint pain), pharyngitis (sore throat), and contusion.[3]

Table 6.1: Comparative Incidence of Common Adverse Reactions (≥10%) Across Approved Indications

Adverse ReactionPNHaHUSgMGNMOSD
Headache
Nasopharyngitis
Back Pain
Nausea
Diarrhea
Hypertension
Upper Respiratory Infection
Abdominal Pain
Vomiting
Anemia
Cough
Peripheral Edema
Urinary Tract Infections
Pyrexia (Fever)
Musculoskeletal Pain
Dizziness
Influenza
Arthralgia
Pharyngitis
Contusion
Note: This table aggregates common adverse events reported at ≥10% or ≥20% in pivotal trials for each indication, based on data from.3 A '✔' indicates the event was reported as common for that indication.

  • Other Serious Risks and Precautions:
  • Other Infections: While meningococcal disease is the most severe risk, the drug's mechanism may also increase susceptibility to other serious infections, particularly those caused by encapsulated bacteria like Streptococcus pneumoniae and Haemophilus influenzae. Fungal infections, such as Aspergillus, have also been reported, particularly in patients who are otherwise immunocompromised.[21]
  • Infusion-Related Reactions: Like all intravenous protein therapies, Eculizumab can cause infusion-related reactions, including anaphylaxis and other hypersensitivity reactions. These reactions can manifest as chest pain, dyspnea, or hemodynamic instability. The infusion may need to be slowed or stopped, and patients must be monitored for at least one hour after the infusion is complete.[1]
  • Treatment Discontinuation Risks: Discontinuation of Eculizumab therapy must be done with caution and under close medical supervision. Abruptly stopping the drug leads to the rapid return of terminal complement activity. In PNH patients, this can cause a sudden and severe rebound of intravascular hemolysis, potentially leading to thrombosis, renal failure, or death. In aHUS patients, discontinuation can lead to a recurrence of severe TMA complications. Patients must be monitored closely for disease-specific complications for an extended period after the last dose.[6]

6.3 Contraindications

The contraindications for Eculizumab are directly related to its primary safety risk:

  • Eculizumab therapy must not be initiated in patients who have an active, unresolved serious Neisseria meningitidis infection.[7]
  • It is also contraindicated in patients who are not currently vaccinated against Neisseria meningitidis, unless the treating physician determines that the risks of delaying Eculizumab therapy outweigh the risks of developing a meningococcal infection.[7]

7.0 Therapeutic Administration and Clinical Management

The administration of Eculizumab is a highly controlled process involving specific dosing regimens tailored to the indication and patient population, as well as strict protocols for preparation and infusion.

7.1 Indication-Specific Dosing Regimens

The dosing of Eculizumab varies significantly based on the indication and the patient's age and body weight. All regimens consist of an initial induction phase to rapidly achieve therapeutic drug levels, followed by a long-term maintenance phase.


Table 7.1: Comprehensive Dosing Regimens for Eculizumab by Indication and Patient Population

IndicationPatient GroupInduction PhaseMaintenance Phase
PNHAdults (≥18 years)600 mg IV weekly for 4 weeks900 mg IV at week 5, then 900 mg IV every 2 weeks
aHUS, gMG, NMOSDAdults (≥18 years)900 mg IV weekly for 4 weeks1200 mg IV at week 5, then 1200 mg IV every 2 weeks
aHUS & gMGPediatrics (≥40 kg)900 mg IV weekly for 4 doses1200 mg IV at week 5, then 1200 mg every 2 weeks
Pediatrics (30 to <40 kg)600 mg IV weekly for 2 doses900 mg IV at week 3, then 900 mg every 2 weeks
Pediatrics (20 to <30 kg)600 mg IV weekly for 2 doses600 mg IV at week 3, then 600 mg every 2 weeks
Pediatrics (10 to <20 kg)600 mg IV as a single dose300 mg IV at week 2, then 300 mg every 2 weeks
Pediatrics (5 to <10 kg)300 mg IV as a single dose300 mg at week 2, then 300 mg every 3 weeks
Source: Data compiled from FDA prescribing information.6

As discussed in Section 4.3, certain concomitant medical procedures can significantly alter the pharmacokinetics of Eculizumab, necessitating supplemental dosing to maintain therapeutic efficacy.


Table 7.2: Supplemental Dosing Guidelines for Eculizumab with Concomitant Procedures

ProcedureMost Recent Soliris DoseRequired Supplemental DoseTiming of Administration
Plasmapheresis or Plasma Exchange (PE/PI)300 mg300 mg per sessionWithin 60 minutes after each session
≥600 mg600 mg per sessionWithin 60 minutes after each session
Fresh Frozen Plasma Infusion≥300 mg300 mg per unit infused60 minutes prior to each infusion
Intravenous Immunoglobulin (IVIg) for gMG≥600-900 mg600 mg per IVIg cycleVaries based on IVIg frequency; consult prescribing information
≤600 mg300 mg per IVIg cycleVaries based on IVIg frequency; consult prescribing information
Source: Data compiled from FDA prescribing information.19

7.2 Protocols for Preparation, Infusion, and Monitoring

The preparation and administration of Eculizumab must follow a precise protocol to ensure stability, sterility, and patient safety.

  • Preparation: The drug must be diluted prior to administration by a healthcare professional using aseptic technique. The required volume of Eculizumab is withdrawn from the vial and transferred to an infusion bag. It must be diluted to a final concentration of 5 mg/mL using one of the approved diluents: 0.9% Sodium Chloride Injection, 0.45% Sodium Chloride Injection, 5% Dextrose in Water (D5W) Injection, or Ringer's Injection. The infusion bag should be gently inverted to mix the solution; it must not be shaken. Any unused portion in the vial must be discarded as the product contains no preservatives. The final admixture must be allowed to adjust to room temperature (18-25°C) naturally before administration and must not be heated by any means.[6]
  • Administration: Eculizumab must only be administered as an intravenous infusion; it should never be given as an IV push or bolus injection. The standard infusion time is 35 minutes in adults. For pediatric patients, the infusion time is longer, ranging from 1 to 4 hours. The infusion can be delivered via gravity feed, a syringe-type pump, or an infusion pump.[6]
  • Post-Infusion Monitoring: Following the completion of the infusion, all patients must be monitored by healthcare personnel for at least one hour for any signs or symptoms of an infusion-related reaction.[6]

8.0 Clinically Significant Drug Interactions

The drug interactions associated with Eculizumab are primarily pharmacodynamic or pharmacokinetic in nature, stemming from its effects on the immune system and its properties as a large protein therapeutic.

8.1 Pharmacodynamic Interactions

These interactions involve the additive or synergistic effects of Eculizumab with other drugs that modulate the immune system.

  • Other Immunosuppressants: Co-administration of Eculizumab with other immunosuppressive or immunomodulating agents can increase the overall level of immunosuppression, potentially heightening the risk of infection. This is a broad category that includes, but is not limited to, drugs like aldesleukin, alefacept, baricitinib, cyclosporine, fingolimod, and tofacitinib. Clinical vigilance is required when combining Eculizumab with any agent that suppresses host defenses.[5]
  • Vaccines: The immunosuppressive effect of Eculizumab can diminish the therapeutic efficacy of vaccines, as an adequate immune response is required to generate protective immunity. This is particularly concerning for live attenuated vaccines (e.g., measles, mumps, rubella, varicella), which could theoretically pose an increased risk of causing disseminated infection in a patient on Eculizumab. Therefore, vaccination schedules should be completed according to guidelines, ideally at least two weeks before initiating Eculizumab therapy.[5]

8.2 Pharmacokinetic Interactions

These interactions involve other agents or procedures that directly affect the concentration of Eculizumab in the body.

  • Plasmapheresis and Plasma Exchange (PE/PI): As detailed previously, these extracorporeal procedures physically remove Eculizumab from the circulation along with other plasma components. This constitutes a significant pharmacokinetic interaction that necessitates supplemental dosing to prevent sub-therapeutic drug levels and potential loss of efficacy.[6]
  • Neonatal Fc Receptor (FcRn) Blockers: This interaction represents a new frontier in clinical pharmacology, driven by the proliferation of biologic therapies. The neonatal Fc receptor (FcRn) is a cellular receptor responsible for protecting IgG antibodies (including therapeutic ones like Eculizumab) from lysosomal degradation, thereby extending their half-life. A new class of drugs, known as FcRn blockers (e.g., efgartigimod, rozanolixizumab), has been developed to treat certain autoimmune diseases by intentionally blocking this receptor. This action accelerates the clearance of pathogenic autoantibodies. However, a direct and predictable consequence is that these drugs will also accelerate the clearance of any co-administered IgG-based therapeutic, including Eculizumab. This mechanism-based, protein-protein interaction can lower systemic exposure and reduce the effectiveness of Eculizumab. Co-administration requires close monitoring for reduced efficacy, and alternative therapeutic strategies may need to be considered.[20] This stands in stark contrast to traditional drug interactions mediated by small-molecule metabolic enzymes like the CYP450 system and highlights the increasing complexity of managing patients on multiple advanced therapies.

9.0 Use in Specific and Vulnerable Populations

The use of Eculizumab in specific populations, such as pediatric and geriatric patients, and particularly in pregnant or lactating women, requires careful consideration of the available evidence and a nuanced risk-benefit assessment.

9.1 Pediatric Use

Eculizumab has established indications in pediatric populations for specific diseases:

  • It is approved for the treatment of aHUS in children.[10]
  • It is approved for the treatment of gMG in children aged six years and older who are anti-AChR antibody positive.[3]
  • For these indications, dosing is not fixed but is based on body weight, with specific induction and maintenance regimens for different weight brackets to ensure appropriate drug exposure.[16]
  • The safety and efficacy of Eculizumab have not been established for other conditions, such as PNH or NMOSD, in pediatric patients.[31]

9.2 Geriatric Use

Clinical studies of Eculizumab have included geriatric patients, and to date, these studies have not identified any specific safety or efficacy concerns that would limit its usefulness in this population.[31] Population pharmacokinetic analyses have confirmed that age, up to 85 years, does not have a clinically meaningful effect on the drug's disposition.[6] Therefore, no age-based dose adjustments are required for elderly patients.

9.3 Risk-Benefit Analysis in Pregnancy and Lactation

The use of Eculizumab during pregnancy and lactation exemplifies a common and challenging scenario in medicine: a dissonance between the cautious position of regulatory bodies and the pressing needs of clinical reality.

  • Pregnancy:
  • Regulatory Stance: Formal regulatory guidance is conservative. The US FDA has not assigned a pregnancy category to Eculizumab, in line with its newer labeling rules. The official prescribing information warns that, based on animal studies, the drug may cause fetal harm.[7] As an IgG antibody, Eculizumab is expected to be actively transported across the placenta, particularly during the second and third trimesters.[32] Consequently, women of childbearing potential are advised to use effective contraception during therapy and for up to 5 months after the last dose.[32] This position is based on a lack of controlled clinical trial data in pregnant women, as such trials are ethically prohibitive.
  • Clinical Reality and Emerging Evidence: In stark contrast to the label's caution, clinicians are often faced with pregnant women suffering from life-threatening, complement-mediated diseases like PNH and aHUS. The risk of the untreated disease during pregnancy is exceptionally high, including severe maternal morbidity (e.g., thrombosis, renal failure), mortality, and adverse fetal outcomes. In this context, the clinical decision becomes a risk-benefit calculation weighing the known, severe risks of the disease against the theoretical and likely lower risks of the drug. A growing body of real-world evidence from case series and registries has emerged, consistently reporting that the use of Eculizumab during pregnancy is associated with favorable maternal and fetal outcomes and is often essential for controlling the underlying disease and allowing the pregnancy to proceed safely.[34] Some reports note that physiological changes during pregnancy, particularly in the third trimester, may increase drug clearance, necessitating an adjustment to the treatment regimen, such as shortening the dosing interval.[35] This body of evidence has empowered clinicians to use Eculizumab when clinically necessary, viewing treatment as the safer option for both mother and child.
  • Lactation:
  • Official Stance: Similar to pregnancy, the official prescribing information advises that caution should be exercised when administering Eculizumab to a nursing woman, as its safety has not been formally established in this context.[7]
  • Emerging Data: Data from multiple studies have analyzed the presence of Eculizumab in the breastmilk of mothers receiving therapy. The consistent finding is that maternal Eculizumab administration results in undetectable or very low levels of the drug in breastmilk.[32] Furthermore, as a large protein molecule, any Eculizumab that is ingested by the infant is likely to be denatured and degraded by proteases in the infant's gastrointestinal tract, with minimal to no systemic absorption. To date, no adverse effects have been reported in infants who were breastfed during maternal Eculizumab therapy.[32]

10.0 Regulatory Trajectory and Evolving Market Dynamics

The regulatory and commercial history of Eculizumab is a story of pioneering science, strategic orphan drug development, and a long period of market dominance that is now transitioning into a new, more competitive phase.

10.1 A History of Global Regulatory Approvals and Orphan Designations

  • Developer and Marketer: Eculizumab was discovered, developed, manufactured, and marketed globally by Alexion Pharmaceuticals, Inc. Following its acquisition by AstraZeneca, this unit is now known as Alexion AstraZeneca Rare Disease.[2]
  • Orphan Drug Strategy: A key element of Eculizumab's success was its strategic development under the orphan drug framework. It received orphan drug designation from regulatory bodies like the FDA for multiple indications, including PNH, aHUS, gMG, and NMOSD.[1] This status provides incentives such as tax credits, research grants, and, most importantly, a period of market exclusivity post-approval, which protected Soliris from competition for many years.
  • Timeline of Approvals: The regulatory journey of Soliris demonstrates a clear, stepwise expansion from its initial indication, creating a narrative of successful lifecycle management.

Table 10.1: Timeline of Key U.S. FDA Regulatory Milestones for Soliris (Eculizumab)

DateMilestoneIndication
March 16, 2007Initial FDA ApprovalParoxysmal Nocturnal Hemoglobinuria (PNH)
September 23, 2011Indication ExpansionAtypical Hemolytic Uremic Syndrome (aHUS)
October 23, 2017Indication ExpansionGeneralized Myasthenia Gravis (gMG) - Adults
June 27, 2019Indication ExpansionNeuromyelitis Optica Spectrum Disorder (NMOSD) - Adults
March 10, 2025Population ExpansionGeneralized Myasthenia Gravis (gMG) - Pediatrics (age 6+)
Source: Data compiled from FDA approval history.3

10.2 The Impact of Biosimilar Entry on the Therapeutic Landscape

For over a decade, Soliris existed as a monopoly product for C5 inhibition, allowing Alexion to command a very high price and making it one of the most expensive medicines in the world. The expiration of its key patents (U.S. exclusivity ended in 2017) has fundamentally altered this market reality.[1]

  • Emergence of Biosimilars: The end of exclusivity paved the way for other pharmaceutical companies to develop and seek approval for biosimilar versions of Eculizumab. A biosimilar is a biologic product that is highly similar to, and has no clinically meaningful differences from, an existing FDA-approved reference product.
  • Approved Biosimilars: As of mid-2024, the FDA has approved multiple Eculizumab biosimilars, including Bkemv (eculizumab-aeeb) and Epysqli (eculizumab-aagh).[10] The approval of these products was based on a totality of evidence from analytical studies, non-clinical data, and clinical trials (e.g., NCT04058158 for SB12, NCT04463056 for Elizaria®) that demonstrated their similarity to the reference product, Soliris.[23]
  • Interchangeability: Notably, Bkemv has been designated by the FDA as an interchangeable biosimilar.[10] This is a higher standard of biosimilarity that means it can be substituted for Soliris at the pharmacy level by a pharmacist without needing to consult the prescribing physician, subject to state pharmacy laws. This designation is expected to accelerate its adoption and increase competitive pressure.

The arrival of biosimilars marks a pivotal inflection point. It signals the maturation of the C5 inhibitor market from a single-product monopoly to a competitive environment. This will inevitably lead to significant price erosion, which is expected to democratize access to this life-saving class of therapy for patients globally and reduce the substantial cost burden on healthcare systems. This market shift also created a powerful strategic imperative for the originator, Alexion, to innovate beyond Soliris to defend its complement franchise, a strategy manifested in its development of next-generation therapies.

11.0 The Investigational Frontier and Future Perspectives

The potent and specific mechanism of Eculizumab has made it an attractive candidate for investigation in a wide array of other diseases where complement activation is thought to play a pathogenic role. This research, coupled with the development of successor molecules, points toward a future of broader and more sophisticated complement modulation.

11.1 Analysis of Off-Label and Investigational Applications

The success of Eculizumab in its approved indications has spurred significant interest in its potential use in other conditions. This is reflected in numerous clinical trials and published case reports.

  • Investigational Trials: The ClinicalTrials.gov database lists a multitude of studies exploring Eculizumab's efficacy in various diseases. Completed or ongoing trials have investigated its use in conditions such as:
  • Guillain-Barré Syndrome (GBS): A Phase 3 trial (NCT04752566) evaluated Eculizumab as a treatment for severe GBS.[38]
  • Membranoproliferative Glomerulonephritis (MPGN): A Phase 2 trial (NCT02093533) explored its use in this rare kidney disease.[39]
  • Antibody-Mediated Transplant Rejection: Early phase trials (e.g., NCT01327573) have studied its potential to block complement-mediated damage to transplanted organs.[14]
  • Severe COVID-19: During the pandemic, trials like CORIMUNO-19 (NCT04346797) were initiated to determine if blocking complement-mediated inflammation could improve outcomes in patients with severe respiratory distress.[40]
  • Off-Label Case Reports: The medical literature contains reports of successful off-label use. For example, a 2023 case report described the use of Eculizumab in a patient with a complex neurological syndrome involving pachymeningitis, progressive vision loss, and seizures that was linked to underlying complement system dysregulation. Treatment with Eculizumab resulted in the stabilization of neurological symptoms and vision loss, allowing for the discontinuation of long-term steroid therapy.[41]

11.2 The Next Generation: Successors and Combination Therapies

Recognizing the limitations of Eculizumab—namely its bi-weekly infusion schedule and its inability to control all aspects of PNH pathology—Alexion has proactively developed next-generation therapies to evolve the standard of care.

  • Ravulizumab (Ultomiris): This molecule is a direct successor to Eculizumab and represents a significant advancement in convenience. Ravulizumab is a C5 inhibitor that was engineered with an extended half-life through modifications that enhance its recycling by the FcRn receptor. This allows for a maintenance dosing schedule of one intravenous infusion every 8 weeks, a substantial reduction in treatment burden compared to Eculizumab's 26 annual infusions.[10] It has been approved for the same core indications as Eculizumab (PNH, aHUS, gMG, NMOSD) and is rapidly becoming the new standard-of-care C5 inhibitor.
  • Danicopan (Voydeya): The development of Danicopan reveals a deeper understanding of the limitations of C5 inhibition alone. While Eculizumab is highly effective at preventing intravascular hemolysis (the destruction of red blood cells within blood vessels) by blocking the MAC, it does not prevent the upstream process of opsonization, where red blood cells are coated with the C3b complement fragment. These C3b-coated cells are then cleared by macrophages in the liver and spleen, a process known as extravascular hemolysis (EVH). In approximately 10-20% of PNH patients treated with a C5 inhibitor, this EVH remains clinically significant, leading to continued anemia and fatigue despite effective control of intravascular hemolysis.[11] Danicopan is a first-in-class, oral inhibitor of Factor D, an enzyme essential for the activation of the alternative pathway that generates C3b. By inhibiting Factor D, Danicopan reduces C3b deposition on red blood cells, thereby decreasing EVH. It has been approved in the U.S. as an add-on therapy to be used in combination with a C5 inhibitor (Eculizumab or Ravulizumab) for PNH patients with clinically significant EVH.[11] This marks a paradigm shift from monotherapy to strategic, dual-pathway combination therapy to achieve optimal disease control.

12.0 Concluding Expert Analysis and Strategic Outlook

12.1 Eculizumab's Legacy

Eculizumab will be recorded in medical history as a transformative therapeutic agent. It was the first drug to successfully and specifically target the complement system for therapeutic benefit, validating a previously undruggable pathway and providing a life-altering, and in many cases life-saving, treatment for several devastating rare diseases. Its development journey serves as a masterclass in orphan drug strategy, demonstrating how a deep understanding of pathophysiology can lead to a highly effective therapy that can command a premium price and build a successful franchise. While its own market share will inevitably decline, its impact is indelible.

12.2 The Three-Pronged Future

The therapeutic landscape that Eculizumab pioneered is now rapidly evolving and segmenting, driven by three key forces that will define the future of complement modulation:

  1. Price Erosion and Broadened Access: The entry of Eculizumab biosimilars, particularly those with interchangeability status, will introduce robust price competition. This will significantly drive down the cost of foundational C5 inhibitor therapy, thereby broadening patient access on a global scale and alleviating the immense financial strain on healthcare systems.
  2. Enhanced Convenience and Reduced Treatment Burden: Next-generation, long-acting C5 inhibitors like Ravulizumab (Ultomiris) are poised to become the new standard of care for monotherapy. By drastically reducing the frequency of infusions, these agents address a major patient-centric need, prioritizing convenience and quality of life.
  3. Targeted Combination Therapy and Optimized Outcomes: The recognition of residual disease activity, exemplified by extravascular hemolysis in PNH, is driving the field toward more sophisticated treatment paradigms. The approval of add-on therapies like the oral Factor D inhibitor Danicopan signals a move away from a "one-size-fits-all" approach. The future will likely involve the strategic use of novel agents, often oral, in combination with a C5 inhibitor backbone to address specific, unresolved aspects of disease pathology and optimize clinical outcomes for individual patients.

12.3 Final Outlook

While the brand Soliris itself will face a challenging commercial future due to biosimilar competition, the therapeutic principle of Eculizumab—terminal complement inhibition—is now more important than ever. The market it single-handedly created is expanding and fragmenting, moving toward a more mature and patient-focused ecosystem. This new landscape promises a future characterized by greater affordability and access, superior convenience through long-acting formulations, and more personalized and effective treatment strategies that target multiple points in the complex complement cascade.

Works cited

  1. Eculizumab - Wikipedia, accessed July 17, 2025, https://en.wikipedia.org/wiki/Eculizumab
  2. en.wikipedia.org, accessed July 17, 2025, https://en.wikipedia.org/wiki/Eculizumab#:~:text=Eculizumab%20(Soliris)%20is%20developed%2C,and%20marketed%20by%20Alexion%20Pharmaceuticals.
  3. Soliris (eculizumab) FDA Approval History - Drugs.com, accessed July 17, 2025, https://www.drugs.com/history/soliris.html
  4. Eculizumab (Soliris) | CAS 219685-50-4 | AbMole BioScience, accessed July 17, 2025, https://www.abmole.com/products/eculizumab.html
  5. Eculizumab: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed July 17, 2025, https://go.drugbank.com/drugs/DB01257
  6. Reference ID: 5540265 - accessdata.fda.gov, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2025/125166s448,761108s038lbl.pdf
  7. Soliris® (eculizumab) injection, for intravenous use - accessdata.fda.gov, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125166s417lbl.pdf
  8. Eculizumab Monograph for Professionals - Drugs.com, accessed July 17, 2025, https://www.drugs.com/monograph/eculizumab.html
  9. Eculizumab - Alexion AstraZencea Rare Disease - AdisInsight - Springer, accessed July 17, 2025, https://adisinsight.springer.com/drugs/800006080
  10. Eculizumab: Usage, Dosage, Side Effects, Warnings - Drugs.com, accessed July 17, 2025, https://www.drugs.com/eculizumab.html
  11. Voydeya approved in the US as add-on therapy to ravulizumab or eculizumab for treatment of extravascular haemolysis in adults with the rare disease PNH - AstraZeneca, accessed July 17, 2025, https://www.astrazeneca.com/media-centre/press-releases/2024/voydeya-approved-in-us.html
  12. Eculizumab - KEGG DRUG, accessed July 17, 2025, https://www.kegg.jp/entry/D03940
  13. LGM Pharma: Eculizumab - CAS# 219685-50-4 - API Supplier, accessed July 17, 2025, https://pharmaoffer.com/api-excipient-supplier/company/lgm-pharma/product/eculizumab
  14. Eculizumab (Synonyms: Anti-Human C5, Humanized Antibody) - MedchemExpress.com, accessed July 17, 2025, https://www.medchemexpress.com/eculizumab.html
  15. Soliris (Eculizumab): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed July 17, 2025, https://www.rxlist.com/soliris-drug.htm
  16. Soliris, INN-eculizumab - EMA, accessed July 17, 2025, https://www.ema.europa.eu/en/documents/product-information/soliris-epar-product-information_en.pdf
  17. Bekemv, INN-eculizumab - EMA, accessed July 17, 2025, https://www.ema.europa.eu/en/documents/product-information/bekemv-epar-product-information_en.pdf
  18. go.drugbank.com, accessed July 17, 2025, https://go.drugbank.com/drugs/DB01257#:~:text=Eculizumab%20is%20a%20monoclonal%20antibody%20that%20targets%20complement%20protein%20C5.&text=Binding%20to%20this%20protein%20prevents,a%20number%20of%20autoimmune%20conditions.
  19. Soliris (exulizumab) label - accessdata.fda.gov, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/125166s172lbl.pdf
  20. Soliris, Bkemv, Epysqli (eculizumab) dosing, indications, interactions, adverse effects, and more - Medscape Reference, accessed July 17, 2025, https://reference.medscape.com/drug/soliris-bkemv-eculizumab-342875
  21. SOLIRIS® (eculizumab) Home | Patient Site, accessed July 17, 2025, https://solirisnmosd.com/
  22. Study Details | Eculizumab to Treat Paroxysmal Nocturnal Hemoglobinuria, accessed July 17, 2025, https://www.clinicaltrials.gov/study/NCT00130000
  23. PNH Completed Phase 3 Trials for Eculizumab (DB01257) | DrugBank Online, accessed July 17, 2025, https://go.drugbank.com/indications/DBCOND0053931/clinical_trials/DB01257?phase=3&status=completed
  24. Eculizumab Completed Phase 3 Trials for Paroxysmal Nocturnal Hemoglobinuria (PNH) Treatment | DrugBank Online, accessed July 17, 2025, https://go.drugbank.com/drugs/DB01257/clinical_trials?conditions=DBCOND0028737&phase=3&purpose=treatment&status=completed
  25. Study Results | An Open-label, Multi-center Clinical Trial of Eculizumab in Adult Patients With Atypical Hemolytic-uremic Syndrome | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT01194973?term=AREA%5BBasicSearch%5D(AREA%5BBasicSearch%5D(AREA%5BConditionSearch%5D((HEMOLYTIC%20UREMIC%20SYNDROME,%20ATYPICAL)%20OR%20(C3))))&rank=9&tab=results
  26. FDA Approves Expanded Use of Eculizumab (Soliris) for Pediatric Myasthenia Gravis, Bringing New Treatment Options for Children - Muscular Dystrophy Association (MDA), accessed July 17, 2025, https://www.mda.org/press-releases/2025/fda-approves-expanded-use-of-eculizumab-soliris-for-pediatric-myasthenia-gravis-bringing-new-treatment-options-for-children
  27. Safety and Efficacy of Eculizumab in Refractory Generalized Myasthenia Gravis (REGAIN Study) | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT01997229
  28. What is the approval history and clinical development pathway of Soliris?, accessed July 17, 2025, https://synapse.patsnap.com/article/what-is-the-approval-history-and-clinical-development-pathway-of-soliris
  29. risk of meningococcal infection - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/125166s446lbl.pdf
  30. HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights do not include all the information needed to use SOLIRIS safely and effec - Alexion.US, accessed July 17, 2025, https://alexion.us/-/media/alexion_global/documents/regulatory/north-america/usa/2024/english/soliris_uspi.pdf
  31. Eculizumab (intravenous route) - Side effects & uses - Mayo Clinic, accessed July 17, 2025, https://www.mayoclinic.org/drugs-supplements/eculizumab-intravenous-route/description/drg-20070917
  32. Eculizumab Use During Pregnancy - Drugs.com, accessed July 17, 2025, https://www.drugs.com/pregnancy/eculizumab.html
  33. Reference ID: 4171013 This label may not be the latest approved by FDA. For current labeling information, please visit https:// - accessdata.fda.gov, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125166s422lbl.pdf
  34. Eculizumab - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed July 17, 2025, https://www.ncbi.nlm.nih.gov/books/NBK501921/
  35. Eculizumab application during pregnancy in a patient with paroxysmal nocturnal hemoglobinuria: A case report with review of the literature - PMC, accessed July 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6099006/
  36. Epysqli (eculizumab-aagh) FDA Approval History - Drugs.com, accessed July 17, 2025, https://www.drugs.com/history/epysqli.html
  37. A Study to Compare SB12 (Proposed Eculizumab Biosimilar) to Soliris in Subjects With Paroxysmal Nocturnal Haemoglobinuria | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT04058158
  38. Study Details | A Study to Evaluate the Efficacy and Safety of ..., accessed July 17, 2025, https://clinicaltrials.gov/study/NCT04752566
  39. Search for: Other terms: ECULIZUMAB | Card Results | ClinicalTrials.gov, accessed July 17, 2025, https://www.clinicaltrials.gov/ct2/results?submit=Search&term=ECULIZUMAB
  40. CORIMUNO19-ECU: Trial Evaluating Efficacy and Safety of Eculizumab (Soliris) in Patients With COVID-19 Infection, Nested in the CORIMUNO-19 Cohort | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT04346797
  41. pubmed.ncbi.nlm.nih.gov, accessed July 17, 2025, https://pubmed.ncbi.nlm.nih.gov/38282487/#:~:text=Off%2Dlabel%20use%20of%20eculizumab%20for%20neurological%20symptoms%20and%20progressive%20vision%20loss
  42. Off-label use of eculizumab for neurological symptoms and ..., accessed July 17, 2025, https://pubmed.ncbi.nlm.nih.gov/38282487/
  43. Off-label use of eculizumab for neurological symptoms and progressive vision loss - Termedia, accessed July 17, 2025, https://www.termedia.pl/Journal/-20/pdf-52237-10?filename=Off-label.pdf

Published at: July 17, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.