1578199-75-3
Episodic Cluster Headache, Migraine
Galcanezumab represents a significant advancement in the management of debilitating headache disorders, specifically migraine and episodic cluster headache. It is a humanized immunoglobulin G4 (IgG4) monoclonal antibody, a type of biologic therapy developed to specifically target a key molecule in the pathophysiology of these conditions.[1] Developed by Eli Lilly and Company and marketed under the brand name Emgality®, galcanezumab is a cornerstone of a new therapeutic class: calcitonin gene-related peptide (CGRP) antagonists.[4]
The advent of CGRP-targeted monoclonal antibodies marks a paradigm shift in headache medicine. For decades, preventive treatment for migraine relied on repurposed oral medications from other drug classes, such as beta-blockers, antidepressants, and anticonvulsants. While beneficial for some patients, these traditional therapies are often associated with limited efficacy, slow onset of action, and burdensome side effect profiles that lead to poor adherence and high discontinuation rates.[7] Galcanezumab and its contemporaries were engineered based on a sophisticated understanding of migraine neurobiology, offering a targeted approach designed to interrupt a core mechanism of the disease process, thereby improving efficacy and tolerability.[9]
For precise identification in clinical, regulatory, and research contexts, galcanezumab is referenced by several names and codes. Its non-proprietary or generic name is Galcanezumab. In the United States, it is designated with a four-letter suffix as galcanezumab-gnlm.[5] This suffix, assigned by the U.S. Food and Drug Administration (FDA) to biologics, is devoid of intrinsic meaning but serves to distinguish it from other products and aids in pharmacovigilance. The developmental code name used during its clinical trial phase was LY2951742.[5] Its primary brand name is Emgality®.[4] Key database identifiers include DrugBank Accession Number DB14042 and Chemical Abstracts Service (CAS) Number 1578199-75-3.[4]
Galcanezumab is a complex protein-based therapeutic. It is a humanized IgG4 kappa monoclonal antibody produced using recombinant DNA technology in a mammalian cell line (Chinese Hamster Ovary, or CHO, cells).[3] The structure consists of two identical kappa light chains and two identical gamma heavy chains, with an approximate total molecular weight of 147,000 Daltons (
147 kDa).[3] A more precise calculation of the non-glycosylated, disulfide-linked protein is 144,084 Da.[13] The selection of the IgG4 isotype is a deliberate and significant aspect of its molecular engineering. Unlike the IgG1 isotype, which is often used for therapeutic antibodies that target cancer cells, the IgG4 backbone exhibits minimal effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). This "quieter" immunological profile is highly desirable for a therapy intended to neutralize a soluble ligand like CGRP, as it minimizes the potential for off-target inflammatory responses and enhances the overall safety and tolerability profile for long-term use.[1]
The final drug product is formulated as a sterile, preservative-free, clear, and colorless to slightly yellow or brown solution for injection.[3] It is supplied in single-dose pre-filled pens or syringes for subcutaneous administration by the patient or a caregiver.[9]
Table 1: Key Identifiers and Physicochemical Properties of Galcanezumab
Property | Value | Source Snippet(s) |
---|---|---|
Generic Name | Galcanezumab | 4 |
Brand Name | Emgality | 4 |
US Adopted Name (USAN) | galcanezumab-gnlm | 4 |
DrugBank ID | DB14042 | 4 |
CAS Number | 1578199-75-3 | 4 |
Type | Biotech, Protein-Based Therapy | 4 |
Class | CGRP Antagonist, Monoclonal Antibody | 5 |
Developer | Eli Lilly and Company | 4 |
Molecular Formula | C6392H9854N1686O2018S46 | 5 |
Approximate Molecular Weight | 147,000 Da (147 kDa) | 3 |
Isotype | Humanized Immunoglobulin G4 (IgG4) kappa | 1 |
Source | Chinese Hamster Ovary (CHO) cells | 3 |
Formulation | Sterile solution in single-dose pre-filled pen or syringe | 9 |
The therapeutic rationale for galcanezumab is rooted in the central role of Calcitonin Gene-Related Peptide (CGRP) in the pathophysiology of migraine and cluster headache. CGRP is a 37-amino acid neuropeptide that is widely expressed in the central and peripheral nervous systems, with particularly high concentrations in the trigeminal ganglion sensory nerves that innervate the cranial vasculature and meninges.[2]
A substantial body of evidence implicates CGRP as a key mediator of headache pain. During acute migraine attacks, CGRP is released from trigeminal nerve endings, leading to elevated levels in the jugular venous blood.[2] This release triggers a cascade of events, including potent vasodilation of cranial blood vessels and neurogenic inflammation in the dura mater (the brain's protective outer layer), which are believed to activate and sensitize pain pathways, resulting in the characteristic throbbing pain of a migraine.[5] The critical role of CGRP is further supported by two key clinical observations: first, the administration of effective acute migraine treatments, such as triptans, correlates with the normalization of elevated CGRP levels; and second, intravenous infusion of CGRP can reliably trigger migraine-like attacks in individuals with a history of the disorder.[2]
Galcanezumab functions as a CGRP antagonist, but its mechanism is distinct from small-molecule CGRP receptor blockers (gepants). Instead of occupying the CGRP receptor, galcanezumab is a ligand-binding antibody, acting as a "CGRP scavenger" in the circulation and tissues.[17] It is engineered to bind directly and with very high affinity to the CGRP neuropeptide itself.[1] By sequestering the CGRP ligand, galcanezumab effectively neutralizes it, preventing it from binding to and activating its cognate receptor. This blockade of the CGRP pathway interrupts the downstream signaling that leads to vasodilation and pain transmission, thereby preventing the onset of a migraine or reducing the frequency of cluster headache attacks.[4]
The binding characteristics of galcanezumab are exceptionally precise. It exhibits a very high binding affinity for the CGRP ligand, with a dissociation constant (KD) of 31 pM.[2] Furthermore, it is highly specific, demonstrating over 10,000-fold greater selectivity for CGRP compared to structurally related peptides such as adrenomedullin, amylin, calcitonin, and intermedin.[2] This high degree of specificity is crucial for minimizing the risk of off-target effects and contributes to its favorable safety profile. Because of its large molecular size, galcanezumab is not expected to cross the blood-brain barrier in significant amounts, suggesting its primary site of action is in the peripheral nervous system, likely at the trigeminal ganglion and the neurovascular junctions within the meninges.[8]
The molecular mechanism of galcanezumab translates into observable physiological effects that can be measured in both in vitro and in vivo models. In cell-based assays, galcanezumab demonstrates a dose-dependent inhibition of CGRP-mediated production of cyclic adenosine monophosphate (cAMP), a key intracellular second messenger, confirming its ability to neutralize CGRP's biological activity.[2]
More importantly, this pharmacodynamic activity has been validated in human clinical studies. A key CGRP-mediated process is vasodilation, which can be provoked experimentally using a topical application of capsaicin. Capsaicin induces the release of CGRP from sensory nerve endings, causing a measurable increase in local dermal blood flow. In a double-blind study in healthy adults, single subcutaneous doses of galcanezumab produced a dose-dependent inhibition of this capsaicin-induced vasodilation.[2] This effect was rapid, significant, and sustained. Furthermore, repeated monthly dosing resulted in a sustained inhibition of this response for over 175 days.[2] This study provides a critical bridge between the drug's molecular action (binding CGRP) and its intended physiological outcome (preventing CGRP-mediated vasodilation), serving as a powerful
in vivo confirmation of its mechanism of action and supporting the rationale for its use in preventing migraine.
The pharmacokinetic profile of galcanezumab, which describes its absorption, distribution, metabolism, and elimination, is characteristic of a monoclonal antibody and fundamentally dictates its clinical utility, including its dosing frequency and low potential for drug-drug interactions.
Galcanezumab is administered exclusively via subcutaneous injection, as oral administration would result in its degradation in the gastrointestinal tract.[4] Following subcutaneous injection, it exhibits a linear pharmacokinetic profile, meaning that its peak concentration (
Cmax) and total exposure (Area Under the Curve, or AUC) increase proportionally with the dose across a wide range of 1 mg to 600 mg.[2] The absorption from the subcutaneous tissue is slow and prolonged, with the time to reach maximum serum concentration (
Tmax) being approximately 5 days after a single dose.[1] The subcutaneous bioavailability is estimated to be within the typical range for monoclonal antibodies, between 50% and 100%.[1] Studies have shown that the specific site of injection (e.g., abdomen, thigh, or arm) does not have a clinically significant impact on its absorption.[2]
Once absorbed into the systemic circulation, galcanezumab has an apparent volume of distribution (Vd) of approximately 7.3 L.[1] This relatively small volume of distribution indicates that the drug is primarily confined to the vascular and interstitial fluid compartments and does not extensively penetrate into deep tissues or cross the blood-brain barrier.
As a large protein, galcanezumab follows a metabolic pathway that is fundamentally different from that of small-molecule drugs. It is not metabolized by the hepatic cytochrome P450 (CYP450) enzyme system, which is responsible for the breakdown of a vast number of common medications.[2] Instead, galcanezumab is expected to be cleared from the body through general protein catabolism. This process involves being taken up by cells via endocytosis and degraded by proteases into small peptides and constituent amino acids, which are then recycled by the body for new protein synthesis.[2] This metabolic pathway is the same one used by the body to clear endogenous immunoglobulins.[1]
The clearance of galcanezumab from the body is slow, with an apparent clearance rate of 0.008 L/h.[1] This slow clearance contributes to its very long elimination half-life, which is approximately 27 days, with a reported range of 25 to 30 days.[1] This extended half-life is the key pharmacokinetic property that enables a convenient once-monthly dosing interval. It ensures that therapeutic concentrations of the drug are maintained in the body throughout the entire month, providing sustained CGRP blockade and continuous prophylactic benefit. This represents a major advantage over traditional daily oral preventives, which require constant patient adherence to maintain efficacy.
Population pharmacokinetic analyses, which pool data from a large number of individuals across clinical trials, have shown that the pharmacokinetic profile of galcanezumab is not clinically affected by intrinsic factors such as age, sex, race, or body weight.[2] Furthermore, its pharmacokinetics do not differ between patients with episodic or chronic migraine.[2] Because its metabolism and clearance are not dependent on renal or hepatic function, dose adjustments are not expected to be necessary for patients with mild to moderate renal or hepatic impairment.[1] However, it has not been formally studied in patients with severe renal impairment (creatinine clearance
<30 mL/min).[20] The independence from CYP450 metabolism also confers a significant clinical advantage by virtually eliminating the risk of pharmacokinetic drug-drug interactions, simplifying its use in patients who may be taking multiple other medications.[2]
Table 2: Summary of Pharmacokinetic Parameters of Galcanezumab
Parameter | Value | Comment/Significance | Source Snippet(s) |
---|---|---|---|
Route of Administration | Subcutaneous injection | Bypasses gastrointestinal degradation. | 4 |
Tmax (Time to Peak Concentration) | ≈5 days | Slow and sustained absorption from subcutaneous tissue. | 2 |
Bioavailability | 50%−100% (estimated) | Typical range for subcutaneously administered monoclonal antibodies. | 1 |
Volume of Distribution (Vd) | 7.3 L | Distribution primarily limited to vascular and interstitial fluids. | 1 |
Metabolism Pathway | Proteolytic catabolism | Degraded into amino acids; not metabolized by CYP450 enzymes. | 2 |
Elimination Half-life | ≈27 days | Long half-life is the basis for the once-monthly dosing regimen. | 2 |
Clearance | 0.008 L/h | Slow clearance contributes to the long half-life. | 1 |
Impact of Renal/Hepatic Impairment | Not expected to be clinically significant | Clearance pathway is independent of kidney and liver function. | 2 |
The clinical efficacy of galcanezumab for the preventive treatment of migraine was established in a robust program of three large, Phase 3, randomized, double-blind, placebo-controlled trials: EVOLVE-1, EVOLVE-2, and REGAIN. These studies evaluated the drug's ability to reduce the frequency of migraine headache days (MHDs) in adults with both episodic and chronic migraine.
The EVOLVE-1 (NCT02614183) and EVOLVE-2 trials were identically designed, six-month studies that enrolled a total of 1,773 patients with episodic migraine, defined as experiencing 4 to 14 MHDs per month.[22] At baseline, patients in these trials had an average of approximately 9.1 MHDs per month.[22] Patients were randomized to receive either placebo, galcanezumab 120 mg monthly, or galcanezumab 240 mg monthly, with all active treatment arms receiving an initial 240 mg loading dose.[22]
The primary endpoint for both studies was the mean change from baseline in the number of monthly MHDs over the six-month treatment period. In both trials, the 120 mg dose of galcanezumab demonstrated a statistically significant and clinically meaningful superiority over placebo.[22]
While the mean reduction is a crucial measure, the responder rates provide a clearer picture of the clinical impact on individual patients. In this regard, galcanezumab also showed significant benefits. The percentage of patients achieving at least a 50% reduction in their monthly MHDs—a widely accepted benchmark for a successful preventive therapy—was substantially higher in the galcanezumab groups.
Furthermore, a significant portion of patients experienced even more profound relief. The rate of achieving a ≥75% reduction in MHDs was roughly double that of placebo in both studies (e.g., 39% vs. 19% in EVOLVE-1).[22] Remarkably, some patients became completely migraine-free. The 100% responder rate was 16% in EVOLVE-1 and 12% in EVOLVE-2 for the 120 mg dose, compared to just 6% for placebo in both trials.[22] This demonstrates that while not all patients respond, a substantial proportion experience a life-altering reduction in migraine burden.
The REGAIN trial (NCT02614261) evaluated galcanezumab in a more severely affected and treatment-refractory population: patients with chronic migraine.[26] This three-month study enrolled 1,117 patients who experienced at least 15 headache days per month, of which at least 8 were MHDs. The baseline average in this cohort was a debilitating 19.4 MHDs per month.[5]
The primary endpoint was the mean change from baseline in monthly MHDs over the three-month treatment period. Galcanezumab 120 mg (following a 240 mg loading dose) was found to be significantly more effective than placebo, reducing the mean number of monthly MHDs by 4.8 days, compared to a reduction of 2.7 days in the placebo group (p<0.001).[9]
Responder rates in this harder-to-treat population were also significant. The percentage of patients achieving a ≥50% reduction in MHDs was nearly double with galcanezumab compared to placebo (28% vs. 15%, p<0.001).[21] While the 120 mg dose did not achieve statistical significance for the higher thresholds of
≥75% or 100% response rates in the three-month period, the clear benefit seen in the primary endpoint and 50% responder rate confirmed its efficacy in this highly disabled patient group.[24]
To assess the durability of the treatment effect, open-label extension studies were conducted for up to 12 months. Data from these extensions confirmed that the benefits of galcanezumab were sustained over long-term treatment in both episodic and chronic migraine populations.[26] In the open-label extension of the REGAIN study, patients who had initially been on placebo and were switched to galcanezumab experienced a robust reduction in MHDs, reaching a level of improvement similar to that seen in patients who had been on active treatment from the beginning. By the 12-month mark, the mean reduction from the original baseline was substantial, ranging from 8.0 to 9.0 MHDs across the treatment groups, demonstrating a consistent and durable effect over time.[26]
Table 3: Summary of Pivotal Phase 3 Efficacy Data for Migraine Prevention
Endpoint | EVOLVE-1 (Episodic) | EVOLVE-2 (Episodic) | REGAIN (Chronic) |
---|---|---|---|
Galcanezumab 120 mg | Placebo | Galcanezumab 120 mg | |
Baseline Mean MHDs | 9.2 | 9.1 | 9.1 |
Mean Change from Baseline in MHDs | -4.7 | -2.8 | -4.3 |
P-value vs. Placebo | p<0.001 | - | p<0.001 |
% Patients with ≥50% Response | 62% | 39% | 59% |
% Patients with ≥75% Response | 39% | 19% | 34% |
% Patients with 100% Response | 16% | 6% | 12% |
Data for 120 mg dose after 240 mg loading dose. MHDs = Migraine Headache Days. N/S = Not Statistically Significant. Sources:.22 |
Galcanezumab holds a unique position as the first therapy in its class, and one of very few medications overall, to receive FDA approval for the treatment of episodic cluster headache.[30] Cluster headache is a distinct trigeminal autonomic cephalalgia, characterized by attacks of excruciatingly severe, unilateral pain, often described as one of the most painful conditions known to medicine. The approval of galcanezumab addressed a profound unmet medical need for a safe and effective treatment to reduce the frequency of these debilitating attacks.[30]
The efficacy of galcanezumab for this indication was established in a randomized, 8-week, double-blind, placebo-controlled study involving 106 adult patients diagnosed with episodic cluster headache.[5] At the start of the study, patients were experiencing a high burden of disease, with an average of 17 to 18 cluster headache attacks per week.[21] Patients in the active treatment arm received a monthly subcutaneous dose of 300 mg of galcanezumab.[30]
The primary endpoint was the mean reduction from baseline in the weekly frequency of cluster headache attacks, averaged across the first three weeks of treatment. The results demonstrated a statistically significant benefit for galcanezumab.
A key secondary endpoint was the 50% responder rate at Week 3. This analysis also favored galcanezumab, with 71% of patients in the treatment group achieving at least a 50% reduction in their weekly attack frequency, compared to 53% of patients in the placebo group (p=0.046).[21]
An important nuance in the trial data is the observation that while the initial effect was robust, the efficacy curves for the galcanezumab and placebo groups began to converge during weeks 4 through 8 of the study.[27] This finding may suggest that galcanezumab's primary benefit is in rapidly reducing attack frequency at the onset and during the most intense phase of a cluster period. The convergence could be influenced by several factors, including the natural, self-limiting course of an episodic cluster period, where attacks may begin to remit spontaneously in both groups. Nonetheless, the ability to significantly curtail the frequency of attacks during the initial, most severe weeks of a cluster period represents a highly valuable clinical outcome for patients suffering from this condition.
The safety profile of galcanezumab has been characterized through extensive clinical trials involving over 2,500 patients and is continuously monitored through post-marketing pharmacovigilance.[13] Overall, it is considered well-tolerated, but important risks have been identified.
Across the pivotal migraine trials (EVOLVE-1, EVOLVE-2, and REGAIN), the safety profile of galcanezumab was highly favorable and broadly similar to that of placebo.[9] The most frequently reported adverse reaction that occurred more often than with placebo was injection site reactions.[19] These reactions, which are generally mild to moderate in severity, encompass a range of related events including pain, erythema (redness), pruritus (itching), and swelling at the injection site.[13] In the pooled migraine studies, 18% of patients receiving the 120 mg dose of galcanezumab reported injection site reactions, compared to 13% of those receiving placebo injections.[19]
Other adverse events reported in trials, though less common, include vertigo, constipation, and pruritus.[13] Importantly, the rate of discontinuation due to adverse events was very low, at less than 2.5%, and was not significantly different from the placebo groups, underscoring the drug's general tolerability.[13] The safety profile observed in the episodic cluster headache study, which used a higher 300 mg dose, was consistent with that seen in the migraine studies.[19]
Table 4: Adverse Reactions with Incidence ≥2% and Greater than Placebo (Migraine Trials)
Adverse Reaction | Emgality 120 mg (N=705) % | Placebo (N=1451) % |
---|---|---|
Injection site reactions | 18% | 13% |
Injection site reactions include multiple related adverse event terms, such as injection site pain, injection site reaction, injection site erythema, and injection site pruritus. Source:.19 |
While clinical trials established a benign safety profile, post-marketing surveillance in a larger and more diverse patient population has identified several rare but serious risks, which are now reflected in the drug's official prescribing information.
Galcanezumab has a very low potential for drug-drug interactions. Because it is a monoclonal antibody that is cleared via protein catabolism rather than by hepatic CYP450 enzymes, it is not expected to have pharmacokinetic interactions with drugs that are substrates, inducers, or inhibitors of the CYP450 system.[2] This is a significant advantage over many oral migraine preventives. A theoretical interaction exists with drugs that bind to the neonatal Fc receptor (FcRn), such as efgartigimod, which could potentially lower galcanezumab concentrations, but the clinical significance of this is not fully established.[20]
Data on the use of galcanezumab in certain populations are limited.
Galcanezumab is approved by the U.S. Food and Drug Administration for two distinct indications in adults [4]:
The dosing regimen for galcanezumab differs based on the indication.
If a dose is missed, it should be administered as soon as possible. The subsequent monthly dosing schedule should then be based on the date of the last administered dose.[14]
Galcanezumab is provided as a sterile solution in single-dose, disposable delivery devices, designed for patient self-administration after appropriate training from a healthcare professional.[14]
The development and approval of galcanezumab was a key event in the emergence of CGRP-targeted therapies for headache disorders.
Galcanezumab was developed and is currently marketed globally by the pharmaceutical company Eli Lilly and Company.[4]
The regulatory pathway in the United States saw two separate approvals for distinct indications.
This rapid succession of approvals for galcanezumab and two other CGRP monoclonal antibodies in 2018 (erenumab and fremanezumab) signaled a major regulatory and clinical turning point.[9] It reflected the strength of the clinical evidence, the high unmet need for effective and tolerable migraine preventives, and a commitment from regulatory bodies to advance innovative treatments for disabling neurological diseases.
The regulatory process in the European Union proceeded on a similar timeline for the migraine indication.
Galcanezumab has fundamentally altered the therapeutic landscape for migraine and episodic cluster headache. It serves as a highly effective and well-tolerated treatment option, particularly for patients who have had an inadequate response to, or cannot tolerate, older oral preventive medications.[7] Its key clinical advantages include a rapid onset of action, often seen within the first month of treatment, a favorable safety profile in controlled trials that is comparable to placebo for most adverse events, and a convenient once-monthly dosing schedule that significantly enhances patient adherence compared to daily medications.[8]
Within the class of CGRP-targeted biologics, galcanezumab (a ligand inhibitor) is often considered alongside fremanezumab (another ligand inhibitor) and erenumab (a receptor antagonist). While all are highly effective, the subtle mechanistic differences may be relevant for future research, although clear distinctions in their clinical profiles have yet to be definitively established. Galcanezumab also now competes with the newer class of oral CGRP receptor antagonists (gepants), which offer the convenience of oral administration but have much shorter half-lives, requiring daily dosing for prevention.[10]
The success of galcanezumab and its class has done more than provide a new treatment; it has validated decades of research into the neurobiology of migraine. The profound efficacy of therapies that specifically target the CGRP pathway provides the strongest evidence to date that this pathway is not merely correlated with migraine but is a central, causal element in its pathogenesis. This represents a landmark achievement in translational medicine, where a basic science discovery has led directly to a highly effective, mechanism-based therapy.
The ideal candidate for galcanezumab therapy is an adult with frequent episodic or chronic migraine who is a candidate for preventive treatment, or an adult experiencing an episodic cluster headache period.[7] It is particularly valuable for patients who have failed multiple oral preventives.
However, patient selection requires careful consideration of the emerging post-marketing safety signals. Caution is warranted when prescribing galcanezumab to patients with pre-existing and poorly controlled hypertension, or those with a history of Raynaud's phenomenon or other significant peripheral vascular, cardiovascular, or cerebrovascular diseases.[7] The potential for CGRP blockade to affect vascular tone regulation in these individuals requires a thoughtful risk-benefit discussion. Another important clinical consideration is the drug's long half-life. While beneficial for adherence, it means that if a patient experiences an intolerable adverse effect or decides to plan a pregnancy, the drug will remain in their system for an extended period, with complete washout taking up to five months.
While currently approved only for adults, there is growing interest in the off-label use of galcanezumab for adolescents with severe, refractory migraine. Expert consensus groups have cautiously suggested that its use may be considered in post-pubertal adolescents who have failed other therapies, but this should be done with close monitoring until robust pediatric trial data and formal approvals are available.[8] Physicians may also consider it off-label for other headache disorders like vestibular migraine, though evidence to support this use is currently lacking.[36]
The foremost questions for future research revolve around personalizing therapy and understanding long-term safety. A critical area of investigation is identifying biomarkers to predict which patients will be "super-responders" (experiencing a ≥75% or 100% reduction in MHDs) and which will be non-responders.[10] This could involve genetic studies of the CGRP pathway, measurement of baseline CGRP levels, or advanced neuroimaging to classify migraine subtypes. Answering this question would allow for more precise patient selection, maximizing therapeutic benefit and avoiding unnecessary treatment. Continued long-term surveillance is also essential to fully characterize the systemic effects of chronic CGRP blockade, and dedicated studies are needed to establish safety in special populations, including pregnant, lactating, pediatric, and geriatric individuals. The ongoing study of galcanezumab in breastmilk is a vital step in this direction.[34]
Published at: August 18, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.