Exdensur by is a Prescription medication manufactured, distributed, or labeled by GlaxoSmithKline LLC. Drug facts, warnings, and ingredients follow.
EXDENSUR is an interleukin‑5 (IL-5) antagonist, a monoclonal antibody (humanized immunoglobulin G [IgG]1 kappa) indicated for add-on maintenance treatment of severe asthma characterized by an eosinophilic phenotype in adult and pediatric patients aged 12 years and older. (1)
Limitations of Use: Not for relief of acute bronchospasm or status asthmaticus. (1)
None. (4)
The most common adverse reactions (incidence ≥4%) are upper respiratory tract infection, allergic rhinitis, influenza, arthralgia, and pharyngitis. (6.1)
To report SUSPECTED ADVERSE REACTIONS, contact GlaxoSmithKline at 1-888-825-5249 or FDA at 1-800-FDA-1088 or www.fda.gov/medwatch.
Pregnancy: EXDENSUR can cross the placenta during pregnancy and the presence of the YTE modification may prolong and increase exposure to the infant exposed in utero. The impact of EXDENSUR transmission to the fetus should be considered. (8.1)
See 17 for PATIENT COUNSELING INFORMATION and FDA-approved patient labeling.
Revised: 12/2025
EXDENSUR is indicated for the add‑on maintenance treatment of severe asthma characterized by an eosinophilic phenotype in adult and pediatric patients aged 12 years and older.
Limitations of Use
EXDENSUR is not indicated for the relief of acute bronchospasm or status asthmaticus [see Warnings and Precautions (5.2)].
The recommended dosage is 100 mg once every 6 months administered by subcutaneous (SC) injection into the upper arm, thigh, or abdomen avoiding 2 inches (5 cm) around the navel [see Dosage and Administration (2.2)].
Missed Dose(s)
If a dose is missed, administer the missed dose as soon as possible and resume the once every 6‑month injection schedule from the date of when the missed dose was given.
Preparation Instructions
Administration Instructions for Single‑Dose Prefilled Pen
Figure 1. EXDENSUR Prefilled Pen Components
Dispose of used pen and clear cap according to local health and safety laws.
Administration Instructions for Single‑Dose Prefilled Syringe
Figure 2. EXDENSUR Prefilled Syringe Components
Dispose of used syringe according to local health and safety laws.
Hypersensitivity reactions, including anaphylaxis, can occur following administration of EXDENSUR. If a hypersensitivity reaction occurs, discontinue EXDENSUR and initiate appropriate therapy.
EXDENSUR should not be used to treat acute asthma symptoms or acute exacerbations. Do not use EXDENSUR to treat acute bronchospasm or status asthmaticus. Patients should seek medical advice if their asthma remains uncontrolled or worsens after initiation of treatment with EXDENSUR.
Reduction in corticosteroid dose may be associated with systemic withdrawal symptoms and/or unmask conditions previously suppressed by systemic corticosteroid therapy. Do not abruptly discontinue systemic or inhaled corticosteroids upon initiation of EXDENSUR therapy. Reductions in corticosteroid dose, if appropriate, should be gradual and performed under the supervision of a healthcare provider.
Eosinophils may be involved in the immunological response to some helminth infections. Patients with pre‑existing helminth infections were excluded from participation in the clinical trials. It is unknown if EXDENSUR will influence a patient’s response against parasitic infections. Patients with pre‑existing helminth infections should be treated for their infection prior to initiation of EXDENSUR therapy. If patients become infected while receiving treatment with EXDENSUR and do not respond to anti‑helminth treatment, discontinue treatment with EXDENSUR until the infection resolves.
The following adverse reactions are discussed in greater detail in other sections of the labeling:
Because clinical trials are conducted under widely varying conditions, adverse reaction rates observed in the clinical trials of a drug cannot be directly compared with rates in the clinical trials of another drug and may not reflect the rates observed in practice.
The safety of EXDENSUR was based on a pooled safety population from 2 replicate, randomized, double‑blind, parallel‑group, placebo‑controlled, multicenter clinical trials (SWIFT‑1 and SWIFT‑2) of 52 weeks duration. The 2 trials included 762 adult and pediatric patients 12 years of age and older with asthma, who received either EXDENSUR 100 mg or placebo administered subcutaneously once every 6 months in addition to their existing background medications for asthma [see Clinical Studies (14)]. A total of 475 patients received 2 doses of EXDENSUR 100 mg in these trials.
Adverse reactions with EXDENSUR with incidence of ≥4% are shown in Table 1.
|
Adverse Reaction |
EXDENSUR (N = 501) n (%) |
Placebo (N = 261) n (%) |
|
Upper respiratory tract infection |
46 (9) |
20 (8) |
|
Allergic rhinitis |
29 (6) |
7 (3) |
|
Influenza |
24 (5) |
11 (4) |
|
Arthralgia |
19 (4) |
8 (3) |
|
Pharyngitis |
18 (4) |
3 (1) |
Specific Adverse Reactions
Injection Site Reactions: In the pooled safety population (SWIFT‑1 and SWIFT‑2), in which EXDENSUR was administered by a healthcare provider, injection site reactions (e.g., erythema, swelling, and itching) occurred in 7 (1%) and 2 (<1%) patients receiving EXDENSUR and placebo, respectively.
Risk Summary
Available data from clinical trials with EXDENSUR use in pregnant women are insufficient to identify a drug‑associated risk of major birth defects, miscarriage, or other adverse maternal or fetal outcomes. There are risks to the mother and fetus associated with asthma in pregnancy (see Clinical Considerations).
Transport of endogenous IgG antibodies and monoclonal antibodies, such as depemokimab-ulaa, across the placenta increases as pregnancy progresses and peaks during the third trimester. The impact of the YTE modification on placental transfer is uncertain [see Clinical Pharmacology (12.1)]; however, the presence of the YTE modification may lead to prolonged and increased exposure of the infant exposed in utero, and the potential of clinical impact is unknown and should be considered. No treatment-related effects on embryofetal or postnatal development have been shown in animal studies targeting IL‑5 signaling pathways (see Data).
The background risk of major birth defects and miscarriage for the indicated population(s) is unknown. All pregnancies have a background risk of birth defect, loss, or other adverse outcomes. In the U.S. general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2% to 4% and 15% to 20%, respectively.
Pregnant women exposed to EXDENSUR, or their healthcare providers, should report EXDENSUR exposure by calling 1‑888‑825‑5249.
Clinical Considerations
Disease‑Associated Maternal and/or Embryofetal Risk: In women with poorly or moderately controlled asthma, evidence demonstrates that there is an increased risk of preeclampsia in the mother, and prematurity, low birth weight, and small for gestational age in the neonate. The level of asthma control should be closely monitored in pregnant women and treatment adjusted as necessary to maintain optimal control.
Data
Animal Data: Reproductive toxicology studies have not been conducted with depemokimab‑ulaa. In animal studies targeting the IL‑5 signaling pathway with a related biologic product without the YTE modification, there were no developmental effects observed [see Nonclinical Toxicology (13)]. Embryofetal development of IL‑5 deficient mice has been reported to be generally unaffected relative to wild‑type mice.
Risk Summary
There are no data on the presence of depemokimab‑ulaa in either human or animal milk, the effects on the breastfed infant, or the effects on milk production. However, depemokimab‑ulaa is a humanized monoclonal antibody (immunoglobulin G1 [IgG1] kappa), and maternal IgG is present in human milk in small amounts. The effects of local gastrointestinal exposure and the extent of systemic exposure in the breastfed infant to EXDENSUR are unknown.
The developmental and health benefits of breastfeeding should be considered along with the mother’s clinical need for EXDENSUR, and any potential adverse effects on the breastfed child from EXDENSUR or from the underlying maternal condition.
Asthma
The safety and effectiveness of EXDENSUR for add‑on maintenance treatment of severe asthma characterized by an eosinophilic phenotype have been established in pediatric patients aged 12 years and older.
Use of EXDENSUR for this indication is supported by evidence from adequate and well‑controlled trials (SWIFT‑1 and SWIFT‑2) in adults and pediatric patients aged 12 years and older, and pharmacokinetic data in pediatric patients aged 12 years and older. A total of 30 pediatric patients aged 12 to 17 years with asthma were enrolled in the SWIFT‑1 and SWIFT‑2 trials, of whom 15 received EXDENSUR 100 mg. Pharmacokinetic and pharmacodynamic data have demonstrated no clinically significant differences in systemic exposure of depemokimab‑ulaa and reduction in blood eosinophil counts in pediatric patients aged 12 years and older compared to that observed in adults following administration of the recommended dosage of EXDENSUR. The safety of EXDENSUR in pediatric patients aged 12 years and older was generally similar to that of the adult population in SWIFT‑1 and SWIFT‑2 [see Adverse Reactions (6.1), Clinical Pharmacology (12.3), Clinical Studies (14)].
The safety and effectiveness of EXDENSUR have not been established in pediatric patients younger than 12 years of age.
Depemokimab‑ulaa is an interleukin‑5 (IL‑5) antagonist monoclonal antibody (humanized Immunoglobulin G1 [IgG1] kappa). Depemokimab‑ulaa is produced by recombinant DNA technology in Chinese hamster ovary cells. The estimated molecular weight of depemokimab‑ulaa is 149 kDa.
EXDENSUR (depemokimab‑ulaa) injection is a sterile, preservative‑free, colorless, yellow to brown, clear to opalescent solution for subcutaneous (SC) use.
EXDENSUR injection is supplied in a single‑dose, 1‑mL, prefilled pen with a fixed 29‑gauge, half‑inch needle or in a single‑dose, 1‑mL, prefilled syringe with a fixed 29‑gauge, half‑inch needle with a needle guard. Each 1 mL delivers 100 mg depemokimab‑ulaa, (8.43 mg) arginine HCl, (0.017 mg) edetate disodium, (1.41 mg) histidine, (2.29 mg) L‑histidine HCl monohydrate, (0.20 mg) polysorbate 80, (61.6 mg) trehalose, and Water for Injection with a pH of 6.0.
Depemokimab‑ulaa is an IL‑5 antagonist (humanized IgG1 kappa monoclonal antibody), which binds to IL‑5 with a dissociation constant of 10.5 pM, inhibiting the bioactivity of IL‑5 with in vitro IC50 value of 4 pM by blocking its binding to the alpha chain of the IL‑5 receptor complex expressed on the cell surface. Depemokimab‑ulaa contains a triple amino acid substitution (YTE) in the fragment crystallizable (Fc) region which increases binding to the neonatal Fc receptor and thereby extends the elimination half‑life. These properties support the dosing interval of every 6 months.
IL‑5 is the major cytokine responsible for the growth and differentiation, recruitment, activation, and survival of eosinophils. Inflammation is an important component in the pathogenesis of asthma. Multiple cell types (e.g., mast cells, eosinophils, neutrophils, macrophages, lymphocytes) and mediators (e.g., histamine, eicosanoids, leukotrienes, cytokines) are involved in inflammation. Depemokimab‑ulaa, by inhibiting IL‑5 signaling, reduced the production and survival of eosinophils; however, the mechanism of depemokimab‑ulaa action in asthma has not been definitively established.
In placebo‑controlled studies involving adult and pediatric patients aged 12 years and older with asthma, a 100 mg dose of EXDENSUR administered SC every 6 months for 52 weeks reduced blood eosinophils to a geometric mean count of 57 cells/mcL (95% CI: 51, 63) at Week 52. This corresponded to a geometric mean reduction of 79% (95% CI: 75.8, 81.8) compared to placebo. This magnitude of blood eosinophil reduction was observed within 2 weeks of treatment (at the first assessment) and was maintained throughout the treatment period.
The pharmacokinetics of depemokimab‑ulaa were approximately dose‑proportional in patients with asthma following SC administration over a dose range of 0.1‑times the recommended dose to 3‑times the recommended dose. After SC administration of 100 mg of EXDENSUR every 6 months in patients with severe asthma, the mean (SD) model-based estimated trough concentration of depemokimab‑ulaa at Week 26 was approximately 1.30 (0.44) mcg/mL. The mean (SD) model-based estimated average steady‑state plasma concentration over a single dosing interval was 6.16 (1.62) mcg/mL.
Absorption
Depemokimab‑ulaa median Tmax was approximately 14 days. There was no accumulation following repeat SC administration once every 6 months.
Distribution
The estimated typical apparent volume of distribution is 6.3 L.
Elimination
The estimated mean (SD) elimination half‑life was 48 (4.7) days following SC administration of EXDENSUR. The estimated typical apparent clearance was 0.092 L/day.
Metabolism: Depemokimab‑ulaa is a monoclonal antibody which is expected to be metabolized into small peptides and amino acids by catabolic pathways.
Specific Populations
No clinically significant differences in the pharmacokinetics of depemokimab‑ulaa were observed based on age (12 to 93 years), sex, race (73% White, 7% Black, 19% Asian), body weight (34.6 to 161 kg), renal impairment (eGFR ≥90 mL/min/1.73 m2 [normal, N = 548]; eGFR 60 to <90 mL/min/1.73 m2 [mild, N = 380]; eGFR 30 to <60 mL/min/1.73 m2 [moderate, N = 31], and eGFR ≤30 mL/min/1.73 m2 [severe, N = 2]), or baseline hepatic function biomarkers (alanine aminotransferase [ALT; 5 to 153 IU/L], aspartate aminotransferase [AST; 9 to 115 IU/L], and bilirubin [1.7 to 42 micromol/L]).
Pediatric Patients: There are limited pharmacokinetic data available in the pediatric population. The pharmacokinetics of depemokimab‑ulaa in pediatric patients aged 12 to 17 years were consistent with adults. The pharmacokinetics of depemokimab‑ulaa have not been studied in pediatric patients aged less than 12 years.
Drug Interaction Studies
No drug interaction studies have been conducted.
The observed incidence of anti‑drug antibodies is highly dependent on the sensitivity and specificity of the assay. Differences in assay methods preclude meaningful comparisons of the incidence of anti‑drug antibodies (ADA) in the studies described below with the incidence of ADA in other studies, including those of EXDENSUR or of other depemokimab products.
In adults and pediatric patients aged 12 years and older with asthma, the incidence of ADA to depemokimab in patients treated with EXDENSUR 100 mg once every 6 months in SWIFT‑1, SWIFT‑2, and an open‑label extension was 10% (66/691). Among ADA‑positive patients, 6% (4/66) developed neutralizing antibodies.
There was no identified clinically significant effect of ADA on the pharmacokinetics, pharmacodynamics, safety, or efficacy of EXDENSUR.
No genotoxicity or carcinogenicity studies have been conducted.
Male and female fertility were unaffected based upon no adverse histopathological findings in the reproductive organs from cynomolgus monkeys receiving SC dosages up to 100 mg/kg depemokimab‑ulaa for 6 months. Mating and reproductive performance were unaffected in male and female CD‑1 mice receiving an analogous antibody, which inhibits the activity of murine IL‑5.
The efficacy of EXDENSUR for the add‑on maintenance treatment of severe asthma characterized by an eosinophilic phenotype was evaluated in 2 replicate, randomized (2:1 to EXDENSUR or placebo), double‑blind, parallel‑group, placebo‑controlled, multicenter clinical trials (SWIFT‑1 [NCT04719832] and SWIFT‑2 [NCT04718103]) of 52 weeks duration. The trials enrolled adult and pediatric patients aged 12 years and older with asthma characterized by an eosinophilic phenotype, defined as a blood eosinophil count ≥150 cells/mcL at screening or ≥300 cells/mcL documented in the year prior to study entry. Patients were required to have 2 or more asthma exacerbations requiring treatment with systemic corticosteroids (SCS) in the prior year while on background asthma therapy consisting of a medium‑ to high‑dose ICS plus at least one additional asthma controller with or without maintenance oral corticosteroids (OCS). Patients were also required to have reduced lung function at baseline (pre‑bronchodilator forced expiratory volume in 1 second [FEV1] <80% predicted normal in adults; FEV1 <90% or FEV1 to forced vital capacity ratio <0.8 in pediatric patients aged 12 years and older). Patients were enrolled without requiring a minimum baseline Asthma Control Questionnaire‑5 (ACQ‑5) score. In these trials, EXDENSUR 100 mg was administered SC once every 6 months for a total of 2 doses in addition to background asthma therapy. The efficacy population consisted of 762 patients who received at least 1 dose of EXDENSUR 100 mg or placebo in SWIFT‑1 (N = 382) and SWIFT‑2 (N = 380).
The demographics and baseline characteristics of the efficacy population in SWIFT‑1 and SWIFT‑2 are provided in Table 2.
| FEV1 = forced expiratory volume in 1 second; ICS = inhaled corticosteroid; IgE = immunoglobulin E; LABA = long‑acting beta agonist; LAMA = long‑acting muscarinic antagonist; mcL = microliter; N = number of patients in the efficacy population; n = number of patients in the respective group; OCS = oral corticosteroid; SD = standard deviation; U = units. | ||
| a Medium ICS dose = 440 mcg fluticasone propionate (FP) daily or equivalent; High ICS dose >440 mcg FP daily or equivalent. | ||
|
SWIFT‑1 N = 382 |
SWIFT‑2 N = 380 |
|
|
Age (years), n (%) | ||
|
12‑17 |
8 (2) |
22 (6) |
|
18‑64 |
276 (72) |
262 (69) |
|
≥65 |
98 (26) |
96 (25) |
|
Mean (SD) |
54 (14.2) |
53 (16.2) |
|
Female, n (%) |
223 (58) |
241 (63) |
|
Race, n (%) | ||
|
White |
316 (83) |
272 (72) |
|
Asian |
58 (15) |
75 (20) |
|
Black or African American |
8 (2) |
28 (7) |
|
Other |
0 |
5 (1) |
|
Hispanic or Latino, n (%) |
23 (6) |
65 (17) |
|
Never smoked, n (%) |
288 (75) |
294 (77) |
|
Duration of asthma (years), mean (SD) |
22 (16.2) |
25 (18.5) |
|
Pre‑bronchodilator % predicted FEV1, mean (SD) |
62 (15.2) |
62 (15.9) |
|
% reversibility, mean (SD) |
17 (15.3) |
18 (17.4) |
|
Eosinophil count (cells/mcL), median (min; max) |
310 (20; 2,360) |
340 (10; 4,440) |
|
Exacerbations in previous year, mean (SD) |
2.2 (0.7) |
2.7 (1.9) |
|
High‑dose ICS use, n (%)a |
203 (53) |
226 (59) |
|
ICS + LABA + LAMA use, n (%) |
95 (25) |
127 (33) |
|
Maintenance OCS use, n (%) |
21 (5) |
19 (5) |
|
Total IgE (U/mcL), median (min; max) |
185 (2; 12,142) |
180 (2; 16,199) |
Exacerbations
The primary efficacy endpoint for SWIFT‑1 and SWIFT‑2 was the annualized rate of clinically significant exacerbations over the 52‑week treatment period. A clinically significant exacerbation was defined as worsening of asthma requiring use of SCS such as intravenous (IV) or oral steroids for at least 3 days or a single intramuscular (IM) corticosteroid dose and/or hospitalization and/or Emergency Department visit. For patients on maintenance SCS, at least double the existing maintenance dose for at least 3 days was required. All patients experiencing an exacerbation were treated with SCS.
In SWIFT‑1 and SWIFT‑2, the annualized rate of asthma exacerbations was significantly lower in patients receiving EXDENSUR compared to placebo (Table 3). During the 52‑week treatment period, fewer patients experienced exacerbations in the EXDENSUR group (32% and 32%) compared to the placebo group (46% and 50%) in SWIFT‑1 and SWIFT‑2, respectively.
| N = number of patients in the efficacy population. | ||||
| Note: Results obtained from a negative binomial model with an offset term for years in study and fixed effects for treatment group, asthma exacerbation history, baseline ICS dose, geographical region, and baseline pre‑bronchodilator % predicted FEV1. | ||||
|
SWIFT‑1 |
SWIFT‑2 |
|||
|
EXDENSUR N = 250 |
Placebo N = 132 |
EXDENSUR N = 252 |
Placebo N = 128 |
|
|
Annualized rate of clinically significant asthma exacerbations |
0.46 |
1.11 |
0.56 |
1.08 |
|
Rate ratio (95% CI) |
0.42 (0.30, 0.59) |
0.52 (0.36, 0.73) |
||
|
P‑value |
<0.001 |
<0.001 |
||
The percentage of patients with exacerbations requiring hospitalization and/or Emergency Department visit was numerically lower for patients treated with EXDENSUR (1% and 4%) compared with placebo (8% and 10%) in SWIFT‑1 and SWIFT‑2, respectively.
In SWIFT‑1 and SWIFT‑2, the time to first clinically significant exacerbation was longer for EXDENSUR compared to placebo (Figures 3 and 4).
Figure 3. Kaplan Meier Curve for Time to First Clinically Significant Exacerbation (SWIFT‑1)
Shaded areas represent 95% confidence intervals.
Figure 4 Kaplan Meier Curve for Time to First Clinically Significant Exacerbation (SWIFT‑2)
Shaded areas represent 95% confidence intervals.
Lung Function
In SWIFT‑1 and SWIFT‑2, the mean change from baseline in pre‑bronchodilator FEV1 for EXDENSUR was 160 mL and 240 mL, respectively, compared to 160 mL and 184 mL for placebo. The treatment difference in SWIFT‑1 and SWIFT‑2 relative to placebo was -1 mL (95% CI: -89, 88) and 56 mL (95% CI: -43, 154), respectively.
Patient-Reported Outcome
In SWIFT‑1 and SWIFT‑2, the proportion of ACQ‑5 responders (clinically meaningful improvement defined as a decrease in score of 0.5 or more) at Week 52 was 54% for EXDENSUR in both studies compared to 55% and 53%, respectively, for placebo.
How Supplied
EXDENSUR (depemokimab‑ulaa) injection is a sterile, preservative‑free, colorless, yellow to brown, clear to opalescent solution for subcutaneous (SC) use. The pens and syringes are not made with natural rubber latex. EXDENSUR injection is supplied as described in Table 4.
| Package Configuration | Strength | NDC |
|---|---|---|
|
1 single‑dose, prefilled pen with attached 29‑gauge, half‑inch needle in a carton |
100 mg/mL |
0173‑0927‑31 |
|
1 single‑dose, prefilled syringe with attached 29‑gauge, half‑inch needle with needle guard in a carton |
100 mg/mL |
0173‑0927‑42 |
Storage and Handling
Store refrigerated at 36°F to 46°F (2°C to 8°C) in the original carton to protect from light.
Do not freeze. Do not shake. Avoid exposure to heat.
The prefilled pen and prefilled syringe can be removed from the refrigerator and kept in the unopened carton, protected from light for up to 7 days at room temperature up to 86°F (30°C). Discard if left out of the refrigerator for more than 7 days.
The prefilled pen or prefilled syringe must be administered within 8 hours once removed from the carton. Discard if not administered within 8 hours.
Advise the patient to read the FDA‑approved patient labeling (Patient Information).
Hypersensitivity Reactions
Inform patients of hypersensitivity reactions, including anaphylaxis, that can occur following administration of EXDENSUR. Instruct patients to contact their healthcare provider if they experience symptoms of a hypersensitivity reaction [see Warnings and Precautions (5.1)].
Acute Asthma Symptoms or Deteriorating Disease
Inform patients that EXDENSUR does not treat acute asthma symptoms or acute exacerbations. Advise patients to seek medical advice if their asthma remains uncontrolled or worsens after initiation of treatment with EXDENSUR [see Warnings and Precautions (5.2)].
Risk Associated with Abrupt Reduction of Corticosteroid Dosage
Advise patients to not abruptly discontinue systemic or inhaled corticosteroids after initiation of EXDENSUR therapy. Inform patients that reduction in corticosteroid dose may be associated with systemic withdrawal symptoms and/or unmask conditions previously suppressed by systemic corticosteroid therapy. Inform patients that reductions in corticosteroid dose, if appropriate, should be gradual and performed under the supervision of a healthcare provider [see Warnings and Precautions (5.3)].
EXDENSUR Exposure to Fetus or Infants During Pregnancy
Advise patients that the potential clinical impact of prolonged and increased EXDENSUR exposure in infants that were exposed in utero is unknown. Inform providers of infants exposed to EXDENSUR in utero of the potential for the infant to be exposed to elevated levels of EXDENSUR for a prolonged period [see Use in Specific Populations (8.1)].
Advise patients who are exposed to EXDENSUR during pregnancy to contact GlaxoSmithKline at 1‑888‑825‑5249.
Trademarks are owned by or licensed to the GSK group of companies.
Manufactured by
GlaxoSmithKline LLC
2929 Walnut Street, Suite 1700
Philadelphia, PA 19104
U.S. License No. 1727
Distributed by
GlaxoSmithKline
Durham, NC 27701
©2025 GSK group of companies or its licensor.
EXD:1PI
|
PATIENT INFORMATION EXDENSUR [Ex‑DEN‑shur] (depemokimab‑ulaa) injection, for subcutaneous use |
|
|
What is EXDENSUR?
It is not known if EXDENSUR is safe and effective in children with asthma under 12 years of age. |
|
|
Before using EXDENSUR, tell your healthcare provider about all of your medical conditions, including if you:
|
|
|
How EXDENSUR is given: A healthcare provider will inject EXDENSUR under your skin (subcutaneously) every 6 months. If you miss a dose of EXDENSUR, contact your healthcare provider to get your next dose as soon as you can. |
|
|
What are the possible side effects of EXDENSUR? EXDENSUR can cause serious side effects including:
|
|
|
|
|
The most common side effects of EXDENSUR include:
These are not all of the possible side effects of EXDENSUR. Call your doctor for medical advice about side effects. You may report side effects to FDA at 1‑800‑FDA‑1088. |
|
|
General information about the safe and effective use of EXDENSUR. Medicines are sometimes prescribed for purposes other than those listed in a Patient Information leaflet. You can ask your pharmacist or healthcare provider for information about EXDENSUR that is written for health professionals. |
|
|
What are the ingredients in EXDENSUR? Active ingredients: depemokimab‑ulaa Inactive ingredients: arginine HCl, edetate disodium, histidine, L‑histidine HCl monohydrate, polysorbate 80, trehalose, Water for Injection. For more information about EXDENSUR, call 1‑833‑EXDENSUR or visit our website at EXDENSUR.com. Trademarks are owned by or licensed to the GSK group of companies. Manufactured by: GlaxoSmithKline LLC, Philadelphia, PA 19104, U.S. License No. 1727 Distributed by: GlaxoSmithKline, Durham, NC 27701 ©2025 GSK group of companies or its licensor. EXD:1PIL |
|
This Patient Information has been approved by the U.S. Food and Drug Administration Issued: December 2025
PRINCIPAL DISPLAY PANEL
NDC: 0173-0927-42
EXDENSUR
(depemokimab-ulaa)
Injection
Rx Only
GSK
100 mg/mL
For subcutaneous use
Prefilled Syringe
Contents:
- 1 Single-Dose 1-mL Prefilled Syringe
- Prescribing Information
- Patient Information
©2025 GSK group of companies or its licensor.
Rev. 12/25
62000000099386
| EXDENSUR
depemokimab injection, solution |
||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||
| Labeler - GlaxoSmithKline LLC (167380711) |
Mark Image Registration | Serial | Company Trademark Application Date |
|---|---|
![]() EXDENSUR 97160260 not registered Live/Pending |
Glaxo Group Limited 2021-12-07 |