繁体中文
设为首页
加入收藏
当前位置:药品说明书与价格首页 >> 呼吸系统病 >> 其他类用药 >> 奥马佐单抗注射剂Xolair(OMALIZUMAB)

奥马佐单抗注射剂Xolair(OMALIZUMAB)

2013-04-29 22:03:57  作者:新特药房  来源:互联网  浏览次数:543  文字大小:【】【】【
简介: 部分中文群多普利处方资料(仅供参考)药品英文名 Omalizumab 药品别名 Xolair 奥马佐单抗药物剂型 奥马佐单抗注射剂:150/Vial 75mg/Vial。 2~8℃冰箱冷藏,避光保存。运输温度30℃以下。 药理作用 ...

HIGHLIGHTS OF PRESCRIBING INFORMATION
These highlights do not include all the information needed to use Xolair safely and effectively. See full prescribing information for Xolair.
Initial U.S. Approval: 2003
WARNING: ANAPHYLAXIS
See full prescribing information for complete boxed warning
Anaphylaxis, presenting as bronchospasm, hypotension, syncope, urticaria, and/or angioedema of the throat or tongue, has been reported to occur after administration of Xolair. Anaphylaxis has occurred after the first dose of Xolair but also has occurred beyond 1 year after beginning treatment. Closely observe patients for an appropriate period of time after Xolair administration and be prepared to manage anaphylaxis that can be life-threatening. Inform patients of the signs and symptoms of anaphylaxis and have them seek immediate medical care should symptoms occur.
RECENT MAJOR CHANGES
Indications and Usage, Pediatric Patients (Age 0 to <12) (1) 01/2010
Use in Specific Populations, Pediatric Use (Age 0 to <12) (8.4) 01/2010
INDICATIONS AND USAGE
Xolair is indicated for:
Moderate to severe persistent asthma in patients with a positive skin test or in vitro reactivity to a perennial aeroallergen and symptoms that are inadequately controlled with inhaled corticosteroids.
Important Limitations of use:
Not indicated for other allergic conditions. (1)
Not indicated for acute bronchospasm or status asthmaticus (1, 5.3)
Not indicated for pediatric patients less than 12 years of age (1, 8.4)
DOSAGE AND ADMINISTRATION
For subcutaneous (SC) administration only.
Administer Xolair 150 to 375 mg SC every 2 or 4 weeks. (2.1)
Determine dose (mg) and dosing frequency by serum total IgE level (IU/mL), measured before the start of treatment, and body weight (kg). See the dose determination charts (2.1)
Divide doses of more than 150 mg among more than one injection site to limit injections to not more than 150 mg per site. (2.3)
DOSAGE FORMS AND STRENGTHS
Lyophilized, sterile powder in a single-use 5mL vial, 150 mg (3)
CONTRAINDICATIONS
Severe hypersensitivity reaction to Xolair or any ingredient of Xolair. (4, 5.1)
WARNINGS AND PRECAUTIONS
Anaphylaxis—Administer only in a healthcare setting prepared to manage anaphylaxis that can be life-threatening and observe patients for an appropriate period of time after administration. (5.1)
Malignancy— Malignancies have been observed in clinical studies. (5.2)
Acute Asthma Symptoms—Do not use for the treatment of acute bronchospasm or status asthmaticus. (5.3)
Corticosteroid Reductions—Do not abruptly discontinue corticosteroids upon initiation of Xolair therapy. (5.4)
Eosinophilic Conditions—Be alert to eosinophilia, vasculitic rash, worsening pulmonary symptoms, cardiac complications, and/or neuropathy, especially upon reduction of oral corticosteroids. (5.5)
ADVERSE REACTIONS
 In the adult and adolescent patients (≥12 years of age), the most commonly observed adverse reactions in clinical studies (≥1% more frequent in Xolair-treated patients) were arthralgia, pain (general), leg pain, fatigue, dizziness, fracture, arm pain, pruritus, dermatitis, and earache. (6.1)
To report SUSPECTED ADVERSE REACTIONS, contact Genentech at 1-888-835-2555 or FDA at 1-800-FDA-1088 or www.fda.gov/medwatch
DRUG INTERACTIONS
No formal drug interaction studies have been performed. (7)
USE IN SPECIFIC POPULATIONS
Pregnancy: No adequate data in humans. Xolair Pregnancy Exposure Registry available (1-866-496-5247) (8.1)
See 17 for PATIENT COUNSELING INFORMATION and the FDA-approved Medication Guide 
Revised: 01/2010
欧盟批准诺华Xolair(Omalizumab)治疗CSU
2014年1月25日,诺华(Novartis)1月24日宣布,欧洲药品管理局(EMA)人用医药产品委员会(CHMP)建议批准单抗药Xolair(omalizumab,奥马珠单抗),作为一种附加药物,用于对H1抗组胺药物反应不足的12岁及以上青少年和成人慢性自发性荨麻疹(CSU)患者的治疗。CHMP推荐的剂量为每4周皮下注射300mg。
欧盟委员会(EC)通常会遵循CHMP的建议,并在2个月内做出最终审查决定。
CHMP的积极意见,是基于3项关键性III期注册研究(ASTERIA I、ASTERIA II、GLACIAL)的积极及一致性结果。这些研究涉及近1000名对抗组胺药物不响应的CSU患者,研究结果表明,300mg剂量omalizumab达到了所有的主要终点及预先指定的次要终点,表明omalizumab能够显著改善瘙痒和荨麻疹,包括快速缓解瘙痒,并在许多情况下完全清除症状。在整个III期项目中,300mg剂量omalizumab治疗组,生活质量显著改善。
诺华曾于2013年公布了Xolair治疗CSU的3项关键性注册研究的数据。
目前,Xolair已获4国(埃及、土耳其、危地马拉、萨尔多瓦)批准,用于慢性自发性荨麻疹(CSU)的治疗。同时,有超过20多个国家正在审查Xolair治疗CSU的监管文件,包括美国、加拿大、澳大利亚、瑞士。
CSU又名慢性特发性荨麻疹(CIU),是一种严重的皮肤疾病,特征是皮肤红肿、痒、时而出现荨麻疹。在世界范围内,CSU的患病率为0.5%至1.0%,超过50%的患者对获批剂量的抗组胺药物治疗无反应。
关于Xolair(omalizumab):
omalizumab是一种实验性单克隆抗体,靶向结合免疫球蛋白E(IgE),该药可能通过减少IgE和细胞激活机制的下游效应,来抑制组胺诱导的皮肤反应。
目前,omalizumab已获全球90多个国家批准,以品牌名索雷尔(Xolair)上市,用于治疗中度至重度持续性过敏性哮喘,该药由诺华和罗氏旗下基因泰克(Genentech)合作开发。

FULL PRESCRIBING INFORMATION

WARNING: Anaphylaxis

Anaphylaxis presenting as bronchospasm, hypotension, syncope, urticaria, and/or angioedema of the throat or tongue, has been reported to occur after administration of Xolair. Anaphylaxis has occurred as early as after the first dose of Xolair, but also has occurred beyond 1 year after beginning regularly administered treatment. Because of the risk of anaphylaxis, observe patients closely for an appropriate period of time after Xolair administration. Health care providers administering Xolair should be prepared to manage anaphylaxis that can be life-threatening. Inform patients of the signs and symptoms of anaphylaxis and instruct them to seek immediate medical care should symptoms occur [see Warnings and Precautions (5.1)].

1 INDICATIONS AND USAGE

Xolair (omalizumab) is indicated for adults and adolescents (12 years of age and above) with moderate to severe persistent asthma who have a positive skin test or in vitro reactivity to a perennial aeroallergen and whose symptoms are inadequately controlled with inhaled corticosteroids.

Xolair has been shown to decrease the incidence of asthma exacerbations in these patients.

Important Limitations of Use

  • Xolair is not indicated for treatment of other allergic conditions.
  • Xolair is not indicated for the relief of acute bronchospasm or status asthmaticus.
  • Xolair is not indicated for use in pediatric patients less than 12 years of age.

2 DOSAGE AND ADMINISTRATION

2.1 Dosing

Administer Xolair (omalizumab) 150 to 375 mg by subcutaneous (SC) injection every 2 or 4 weeks. Determine doses (mg) and dosing frequency by serum total IgE level (IU/mL), measured before the start of treatment, and body weight (kg). See the dose determination charts below (Table 1 and Table 2) for appropriate dose assignment.

Periodically reassess the need for continued therapy based upon the patient’s disease severity and level of asthma control.

Table 1
Administration Every 4 Weeks
Xolair Doses (milligrams) Administered by Subcutaneous Injection Every 4 Weeks for Adults and Adolescents 12 Years of Age and Older
Pre-treatment Serum IgE (IU/mL) Body Weight (kg)
30-60 > 60-70 > 70-90 > 90-150
≥ 30-100 150 150 150 300
> 100-200 300 300 300
> 200-300 300
> 300-400 SEE TABLE 2
> 400-500
> 500-600

Table 2
Administration Every 2 Weeks
Xolair Doses (milligrams) Administered by Subcutaneous Injection Every 2 Weeks for Adults and Adolescents 12 Years of Age and Older
Pre-treatment Serum IgE (IU/mL) Body Weight (kg)
30-60 > 60-70 > 70-90 > 90-150
≥ 30-100 SEE TABLE 1
> 100-200 225
> 200-300 225 225 300
> 300-400 225 225 300
> 400-500 300 300 375
> 500-600 300 375 DO NOT DOSE
> 600-700 375

2.2 Dosing Adjustments

Adjust doses for significant changes in body weight (see Table 1 and Table 2).

Total IgE levels are elevated during treatment and remain elevated for up to one year after the discontinuation of treatment. Therefore, re-testing of IgE levels during Xolair treatment cannot be used as a guide for dose determination.

  • Interruptions lasting less than one year: Dose based on serum IgE levels obtained at the initial dose determination.
  • Interruptions lasting one year or more: Re-test total serum IgE levels for dose determination.

2.3 Preparation and Administration

Prepare Xolair for subcutaneous injection using Sterile Water for Injection (SWFI), USP, ONLY. Each vial of Xolair is for single use only and contains no preservatives.

Reconstitution

The lyophilized product takes 15-20 minutes to dissolve. The fully reconstituted product will appear clear or slightly opalescent and it is acceptable if there are a few small bubbles or foam around the edge of the vial. The reconstituted product is somewhat viscous; in order to obtain the full 1.2 mL dose, ALL OF THE PRODUCT MUST BE WITHDRAWN from the vial before expelling any air or excess solution from the syringe.

Use the solution within 8 hours following reconstitution when stored in the vial at 2-8ºC (36-46ºF), or within 4 hours of reconstitution when stored at room temperature. Reconstituted Xolair vials should be protected from sunlight.

Preparation

STEP 1: Draw 1.4 mL of SWFI, USP into a 3  mL syringe equipped with a 1 inch, 18-gauge needle.

STEP 2: Place the vial upright on a flat surface and using standard aseptic technique, insert the needle and inject the SWFI, USP directly onto the product.

STEP 3: Keeping the vial upright, gently swirl the upright vial for approximately 1 minute to evenly wet the powder. Do not shake.

STEP 4: After completing STEP 3, gently swirl the vial for 5-10 seconds approximately every 5 minutes in order to dissolve any remaining solids. There should be no visible gel like particles in the solution. Do not use if foreign particles are present.

Note: If it takes longer than 20 minutes to dissolve completely, repeat STEP 4 until there are no visible gel-like particles in the solution. Do not use if the contents of the vial do not dissolve completely by 40 minutes.

STEP 5: Invert the vial for 15 seconds in order to allow the solution to drain toward the stopper. Using a new 3 mL syringe equipped with a 1-inch, 18-gauge needle, insert the needle into the inverted vial. Position the needle tip at the very bottom of the solution in the vial stopper when drawing the solution into the syringe. Before removing the needle from the vial, pull the plunger all the way back to the end of the syringe barrel in order to remove all of the solution from the inverted vial.

STEP 6: Replace the 18-gauge needle with a 25-gauge needle for subcutaneous injection.

STEP 7: Expel air, large bubbles, and any excess solution in order to obtain the required 1.2 mL dose. A thin layer of small bubbles may remain at the top of the solution in the syringe.

Administration

Administer Xolair by subcutaneous injection. The injection may take 5-10 seconds to administer because the solution is slightly viscous. Each vial delivers 1.2 mL (150 mg) of Xolair. Do not administer more than 150 mg per injection site. Divide doses of more than 150 mg among two or more injection sites. (Table 3).

Table 3 Number of Injections and Total Injection Volumes
Xolair Dose
(mg)
Number of Injections Total Volume Injected
(mL)
150 1 1.2
225 2 1.8
300 2 2.4
375 3 3.0

3 DOSAGE FORMS AND STRENGTHS

150 mg of omalizumab as lyophilized, sterile powder in a single-use 5 mLvial.

4 CONTRAINDICATIONS

The use of Xolair is contraindicated in the following:

Severe hypersensitivity reaction to Xolair or any ingredient of Xolair [see Warnings and Precautions (5.1)].

5 WARNINGS AND PRECAUTIONS

5.1 Anaphylaxis

Anaphylaxis has been reported to occur after administration of Xolair in premarketing clinical trials and in postmarketing spontaneous reports. Signs and symptoms in these reported cases have included bronchospasm, hypotension, syncope, urticaria, and/or angioedema of the throat or tongue. Some of these events have been life-threatening. In premarketing clinical trials the frequency of anaphylaxis attributed to Xolair use was estimated to be 0.1%. In postmarketing spontaneous reports, the frequency of anaphylaxis attributed to Xolair use was estimated to be at least 0.2% of patients based on an estimated exposure of about 57,300 patients from June 2003 through December 2006. Anaphylaxis has occurred as early as after the first dose of Xolair, but also has occurred beyond one year after beginning regularly scheduled treatment.

Administer Xolair only in a healthcare setting by healthcare providers prepared to manage anaphylaxis that can be life-threatening. Observe patients closely for an appropriate period of time after administration of Xolair, taking into account the time to onset of anaphylaxis seen in premarketing clinical trials and postmarketing spontaneous reports [see Adverse Reactions (6)]. Inform patients of the signs and symptoms of anaphylaxis, and instruct them to seek immediate medical care should signs or symptoms occur.

Discontinue Xolair in patients who experience a severe hypersensitivity reaction [see Contraindications (4)].

5.2 Malignancy

Malignant neoplasms were observed in 20 of 4127 (0.5%) Xolair-treated patients compared with 5 of 2236 (0.2%) control patients in clinical studies of adults and adolescents (≥ 12 years of age) with asthma and other allergic disorders. The observed malignancies in Xolair-treated patients were a variety of types, with breast, non-melanoma skin, prostate, melanoma, and parotid occurring more than once, and five other types occurring once each. The majority of patients were observed for less than 1 year. The impact of longer exposure to Xolair or use in patients at higher risk for malignancy (e.g., elderly, current smokers) is not known [see Adverse Reactions (6)].

5.3 Acute Asthma Symptoms

Xolair has not been shown to alleviate asthma exacerbations acutely. Do not use Xolair to treat acute bronchospasm or status asthmaticus.

5.4 Corticosteroid Reduction

Do not discontinue systemic or inhaled corticosteroids abruptly upon initiation of Xolair therapy. Decrease corticosteroids gradually under the direct supervision of a physician.

5.5 Eosinophilic Conditions

In rare cases, patients with asthma on therapy with Xolair may present with serious systemic eosinophilia sometimes presenting with clinical features of vasculitis consistent with Churg-Strauss syndrome, a condition which is often treated with systemic corticosteroid therapy. These events usually, but not always, have been associated with the reduction of oral corticosteroid therapy. Physicians should be alert to eosinophilia, vasculitic rash, worsening pulmonary symptoms, cardiac complications, and/or neuropathy presenting in their patients. A causal association between Xolair and these underlying conditions has not been established.

5.6 Parasitic (Helminth) Infection

Monitor patients at high risk of geohelminth infection while on Xolair therapy. Insufficient data are available to determine the length of monitoring required for geohelminth infections after stopping Xolair treatment.

In a one-year clinical trial conducted in Brazil in patients at high risk for geohelminthic infections (roundworm, hookworm, whipworm, threadworm), 53% (36/68) of Xolair-treated patients experienced an infection, as diagnosed by standard stool examination, compared to 42% (29/69) of placebo controls. The point estimate of the odds ratio for infection was 1.96, with a 95% confidence interval (0.88, 4.36) indicating that in this study a patient who had an infection was anywhere from 0.88 to 4.36 times as likely to have received Xolair than a patient who did not have an infection. Response to appropriate anti-geohelminth treatment of infection as measured by stool egg counts was not different between treatment groups.

5.7 Laboratory Tests

Serum total IgE levels increase following administration of Xolair due to formation of Xolair:IgE complexes [see Clinical Pharmacology (12.2)]. Elevated serum total IgE levels may persist for up to 1 year following discontinuation of Xolair. Do not use serum total IgE levels obtained less than 1 year following discontinuation to reassess the dosing regimen because these levels may not reflect steady state free IgE levels.

6 ADVERSE REACTIONS

Use of Xolair has been associated with:

  • Anaphylaxis [see Boxed Warning and Warning and Precautions (5.1)]
  • Malignancies [see Warnings and Precautions (5.2)]

Anaphylaxis was reported in 3 of 3507 (0.1%) patients in clinical trials. Anaphylaxis occurred with the first dose of Xolair in two patients and with the fourth dose in one patient. The time to onset of anaphylaxis was 90 minutes after administration in two patients and 2 hours after administration in one patient. In clinical trials the observed incidence of malignancy among Xolair-treated patients (0.5%) was numerically higher than among patients in control groups (0.2%).

6.1 Clinical Trials Experience

Adult and Adolescent Patients 12 years of Age and Older

The data described below reflect Xolair exposure for 2076 adult and adolescent patients ages 12 and older, including 1687 patients exposed for six months and 555 exposed for one year or more, in either placebo-controlled or other controlled asthma studies. The mean age of patients receiving Xolair was 42 years, with 134 patients 65 years of age or older; 60% were women, and 85% Caucasian. Patients received Xolair 150 to 375 mg every 2 or 4 weeks or, for patients assigned to control groups, standard therapy with or without a placebo. Because clinical studies are conducted under widely varying conditions, adverse reaction rates observed in the clinical studies of one drug cannot be directly compared with rates in the clinical studies of another drug and may not reflect the rates observed in medical practice.

The adverse events most frequently resulting in clinical intervention (e.g., discontinuation of Xolair, or the need for concomitant medication to treat an adverse event) were injection site reaction (45%), viral infections (23%), upper respiratory tract infection (20%), sinusitis (16%), headache (15%), and pharyngitis (11%). These events were observed at similar rates in Xolair-treated patients and control patients.

Table 4 shows adverse reactions from four placebo-controlled asthma studies that occurred  ≥ 1% and more frequently in patients receiving Xolair than in those receiving placebo. Adverse events were classified using preferred terms from the International Medical Nomenclature (IMN) dictionary. Injection site reactions were recorded separately from the reporting of other adverse events and are described following Table 4 .

Table 4
Adverse Reactions ≥ 1% More Frequent in Xolair-Treated Adult or Adolescent Patients 12 years of age and older

Four placebo-controlled asthma studies
Adverse reaction Xolair
n = 738
(%)
Placebo
n = 717
(%)
Body as a whole
    Pain 7 5
    Fatigue 3 2
Musculoskeletal system
    Arthralgia 8 6
    Fracture 2 1
    Leg pain 4 2
    Arm pain 2 1
Nervous system
    Dizziness 3 2
Skin and appendages
    Pruritus 2 1
    Dermatitis 2 1
Special senses
    Earache 2 1

There were no differences in the incidence of adverse reactions based on age (among patients under 65), gender or race.

Injection Site Reactions

Injection site reactions of any severity occurred at a rate of 45% in Xolair-treated patients compared with 43% in placebo-treated patients. The types of injection site reactions included: bruising, redness, warmth, burning, stinging, itching, hive formation, pain, indurations, mass, and inflammation.

Severe injection site reactions occurred more frequently in Xolair-treated patients compared with patients in the placebo group (12% versus 9%).

The majority of injection site reactions occurred within 1 hour-post injection, lasted less than 8 days, and generally decreased in frequency at subsequent dosing visits.

Immunogenicity

Antibodies to Xolair were detected in approximately 1/1723 (< 0.1%) of patients treated with Xolair. The data reflect the percentage of patients whose test results were considered positive for antibodies to Xolair in an ELISA assay and are highly dependent on the sensitivity and specificity of the assay. Additionally, the observed incidence of antibody positivity in the assay may be influenced by several factors including sample handling, timing of sample collection, concomitant medications, and underlying disease. Therefore, comparison of the incidence of antibodies to Xolair with the incidence of antibodies to other products may be misleading.

6.2 Postmarketing Experience

The following adverse reactions have been identified during postapproval use of Xolair in adult and adolescent patients 12 years of age and older. Because these reactions are reported voluntarily from a population of uncertain size, it is not always possible to reliably estimate their frequency or establish a causal relationship to drug exposure.

Anaphylaxis: Based on spontaneous reports and an estimated exposure of about 57,300 patients from June 2003 through December 2006, the frequency of anaphylaxis attributed to Xolair use was estimated to be at least 0.2% of patients. Diagnostic criteria of anaphylaxis were skin or mucosal tissue involvement, and, either airway compromise, and/or reduced blood pressure with or without associated symptoms, and a temporal relationship to Xolair administration with no other identifiable cause. Signs and symptoms in these reported cases included bronchospasm, hypotension, syncope, urticaria, angioedema of the throat or tongue, dyspnea, cough, chest tightness, and/or cutaneous angioedema. Pulmonary involvement was reported in 89% of the cases. Hypotension or syncope was reported in 14% of cases. Fifteen percent of the reported cases resulted in hospitalization. A previous history of anaphylaxis unrelated to Xolair was reported in 24% of the cases.

Of the reported cases of anaphylaxis attributed to Xolair, 39% occurred with the first dose, 19% occurred with the second dose, 10% occurred with the third dose, and the rest after subsequent doses. One case occurred after 39 doses (after 19 months of continuous therapy, anaphylaxis occurred when treatment was restarted following a 3 month gap). The time to onset of anaphylaxis in these cases was up to 30 minutes in 35%, greater than 30 and up to 60 minutes in 16%, greater than 60 and up to 90 minutes in 2%, greater than 90 and up to 120 minutes in 6%, greater than 2 hours and up to 6 hours in 5%, greater than 6 hours and up to 12 hours in 14%, greater than 12 hours and up to 24 hours in 8%, and greater than 24 hours and up to 4 days in 5%. In 9% of cases the times to onset were unknown.

Twenty-three patients who experienced anaphylaxis were rechallenged with Xolair and 18 patients had a recurrence of similar symptoms of anaphylaxis. In addition, anaphylaxis occurred upon rechallenge with Xolair in 4 patients who previously experienced urticaria only.

Eosinophilic Conditions: Eosinophilic conditions have been reported [see Warnings and Precautions (5.5)].

Hematologic: Severe thrombocytopenia has been reported.

Skin: Hair loss has been reported.

7 DRUG INTERACTIONS

No formal drug interaction studies have been performed with Xolair. The concomitant use of Xolair and allergen immunotherapy has not been evaluated.

8 USE IN SPECIFIC POPULATIONS

8.1 Pregnancy

Teratogenic Effects: Pregnancy Category B

There are no adequate and well-controlled studies of Xolair in pregnant women. Reproduction studies have been performed in Cynomolgus monkeys at subcutaneous doses up to 10 times the maximum recommended human dose on a mg/kg basis and have revealed no evidence of impaired fertility or harm to the fetus due to Xolair. Because animal reproduction studies are not always predictive of human response, administer Xolair during pregnancy only if clearly needed [see Nonclinical Toxicology (13.2)].

Pregnancy Exposure Registry

To monitor outcomes of pregnant women exposed to Xolair, including women who are exposed to at least one dose of Xolair within 8 weeks prior to conception or any time during pregnancy, a pregnancy exposure registry has been established. Encourage patients to call 1-866-4XOLAIR (1-866-496-5247) to enroll in the Xolair Pregnancy Exposure Registry. Call this number to obtain further information about this registry.

8.3 Nursing Mothers

There are no data from controlled clinical trials on the use of Xolair by nursing mothers. It is not known whether Xolair is excreted in human breast milk. However, IgG is excreted in human breast milk and therefore it is expected that Xolair will be excreted in human breast milk. The potential for Xolair absorption or harm to the infant is unknown; therefore caution should be exercised when Xolair is administered to a nursing woman.

The excretion of omalizumab in milk was evaluated in female Cynomolgus monkeys at a subcutaneous dose approximately 10 times the maximum recommended human dose on a mg/kg basis. Neonatal plasma levels of omalizumab after in utero exposure and 28 days of nursing were between 11% and 94% of the maternal plasma level. Milk levels of omalizumab were 1.5% of maternal blood concentration [see Nonclinical Toxicology (13.2)].

8.4 Pediatric Use

Safety and effectiveness of Xolair were evaluated in 2 studies in 926 (Xolair 624; placebo 302) asthma patients 6 to <12 years of age. One study was a pivotal study of similar design and conduct to that of adult and adolescent studies 1 and 2 [see Clinical Trials (14)]. The other study was primarily a safety study and included evaluation of efficacy as a secondary outcome. In the pivotal study, Xolair-treated patients had a statistically significant reduction in the rate of exacerbations (exacerbation was defined as worsening of asthma that required treatment with systemic corticosteroids or a doubling of the baseline ICS dose), but other efficacy variables such as nocturnal symptom scores, beta-agonist use, and measures of airflow (FEV1) were not significantly different in Xolair-treated patients compared to placebo. Considering the risk of anaphylaxis and malignancy seen in Xolair-treated patients ≥12 years old and the modest efficacy of Xolair in the pivotal pediatric study, the risk-benefit assessment does not support the use of Xolair in patients 6 to <12 years of age. Although patients treated with Xolair in these two studies did not develop anaphylaxis or malignancy, the studies are not adequate to address these concerns because patients with a history of anaphylaxis or malignancy were excluded, and the duration of exposure and sample size were not large enough to exclude these risks in patients 6 to <12 years of age. Furthermore, there is no reason to expect that younger pediatric patients would not be at risk of anaphylaxis and malignancy seen in adult and adolescent patients with Xolair. [see Warnings and Precautions (5.1) (5.2); and Adverse Reactions (6)].

Studies in patients 0-5 years of age were not required because of the safety concerns of anaphylaxis and malignancy associated with the use of Xolair in adults and adolescents.

8.5 Geriatric Use

In clinical trials 134 patients 65 years of age or older were treated with Xolair. Although there were no apparent age-related differences observed in these studies, the number of patients aged 65 and over is not sufficient to determine whether they respond differently from younger patients.

10 OVERDOSAGE

The maximum tolerated dose of Xolair has not been determined. Single intravenous doses of up to 4000 mg have been administered to patients without evidence of dose limiting toxicities. The highest cumulative dose administered to patients was 44,000 mg over a 20 week period, which was not associated with toxicities.

11 DESCRIPTION

Xolair (omalizumab) is a recombinant DNA-derived humanized IgG1κ monoclonal antibody that selectively binds to human immunoglobulin E (IgE). The antibody has a molecular weight of approximately 149 kiloDaltons. Xolair is produced by a Chinese hamster ovary cell suspension culture in a nutrient medium containing the antibiotic gentamicin. Gentamicin is not detectable in the final product.

Xolair is a sterile, white, preservative free, lyophilized powder contained in a single use vial that is reconstituted with Sterile Water for Injection (SWFI), USP, and administered as a subcutaneous (SC) injection. Each 202.5 mg vial of omalizumab also contains L-histidine (1.8 mg), L-histidine hydrochloride monohydrate (2.8 mg), polysorbate 20 (0.5 mg) and sucrose (145.5 mg) and is designed to deliver 150 mg of omalizumab in 1.2 mL after reconstitution with 1.4 mL SWFI, USP.

12 CLINICAL PHARMACOLOGY

12.1 Mechanism of Action

Omalizumab inhibits the binding of IgE to the high-affinity IgE receptor (FcεRI) on the surface of mast cells and basophils. Reduction in surface-bound IgE on FcεRI-bearing cells limits the degree of release of mediators of the allergic response. Treatment with Xolair also reduces the number of FcεRI receptors on basophils in atopic patients.

12.2 Pharmacodynamics

In clinical studies, serum free IgE levels were reduced in a dose dependent manner within 1 hour following the first dose and maintained between doses. Mean serum free IgE decrease was greater than 96% using recommended doses. Serum total IgE levels (i.e., bound and unbound) increased after the first dose due to the formation of omalizumab:IgE complexes, which have a slower elimination rate compared with free IgE. At 16 weeks after the first dose, average serum total IgE levels were five-fold higher compared with pre-treatment when using standard assays. After discontinuation of Xolair dosing, the Xolair-induced increase in total IgE and decrease in free IgE were reversible, with no observed rebound in IgE levels after drug washout. Total IgE levels did not return to pre-treatment levels for up to one year after discontinuation of Xolair.

12.3 Pharmacokinetics

After SC administration, omalizumab is absorbed with an average absolute bioavailability of 62%. Following a single SC dose in adult and adolescent patients with asthma, omalizumab was absorbed slowly, reaching peak serum concentrations after an average of 7-8 days. The pharmacokinetics of omalizumab are linear at doses greater than 0.5 mg/kg. Following multiple doses of Xolair, areas under the serum concentration-time curve from Day 0 to Day 14 at steady state were up to 6-fold of those after the first dose.

In vitro, omalizumab forms complexes of limited size with IgE. Precipitating complexes and complexes larger than 1 million daltons in molecular weight are not observed in vitro or in vivo. Tissue distribution studies in Cynomolgus monkeys showed no specific uptake of 125I-omalizumab by any organ or tissue. The apparent volume of distribution in patients following SC administration was 78 ± 32 mL/kg.

Clearance of omalizumab involves IgG clearance processes as well as clearance via specific binding and complex formation with its target ligand, IgE. Liver elimination of IgG includes degradation in the liver reticuloendothelial system (RES) and endothelial cells. Intact IgG is also excreted in bile. In studies with mice and monkeys, omalizumab:IgE complexes were eliminated by interactions with Fcγ receptors within the RES at rates that were generally faster than IgG clearance. In asthma patients omalizumab serum elimination half-life averaged 26 days, with apparent clearance averaging 2.4 ± 1.1 mL/kg/day. In addition, doubling body weight approximately doubled apparent clearance.

Special Populations

The population pharmacokinetics of omalizumab were analyzed to evaluate the effects of demographic characteristics. Analyses of these data suggest that no dose adjustments are necessary for age (12-76 years), race, ethnicity, or gender.

13 NONCLINICAL TOXICOLOGY

13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility

No long-term studies have been performed in animals to evaluate the carcinogenic potential of Xolair.

No evidence of mutagenic activity was observed in Ames tests using six different strains of bacteria with and without metabolic activation at omalizumab concentrations up to 5000 μg/mL.

There were no effects on fertility and reproductive performance in male and female Cynomolgus monkeys that received Xolair at subcutaneous doses up to 75 mg/kg/week (approximately 5 times the maximum recommended human dose on an AUC basis).

13.2 Animal Toxicology and/or Pharmacology

Reproductive Toxicology Studies:

Reproductive studies have been performed in Cynomolgus monkeys at subcutaneous doses up to 75 mg/kg (approximately 10 times the maximum recommended human dose on a mg/kg basis) and have revealed no evidence of maternal toxicity, embryotoxicity, or teratogenicity when administered throughout organogenesis and did not elicit adverse effects on fetal or neonatal growth when administered throughout late gestation, delivery and nursing. IgG molecules are known to cross the placental barrier [see Use in Specific Populations (8.1)].

Lactation Studies:

The excretion of omalizumab in milk was evaluated in female Cynomolgus monkeys receiving a subcutaneous dose of 75 mg/kg/week (approximately 10 times the maximum recommended human dose on a mg/kg basis). Neonatal plasma levels of omalizumab after in utero exposure and 28 days of nursing were between 11% and 94% of the maternal plasma level. Milk levels of Xolair were 1.5% of maternal blood concentration. [see Use in Specific Population (8.3)].

14 CLINICAL STUDIES

Adult and Adolescent Patients 12 Years of Age and Older

The safety and efficacy of Xolair were evaluated in three randomized, double-blind, placebo-controlled, multicenter trials.

The trials enrolled patients 12 to 76 years old, with moderate to severe persistent (NHLBI criteria) asthma for at least one year, and a positive skin test reaction to a perennial aeroallergen. In all trials, Xolair dosing was based on body weight and baseline serum total IgE concentration. All patients were required to have a baseline IgE between 30 and 700 IU/mL and body weight not more than 150 kg. Patients were treated according to a dosing table to administer at least 0.016 mg/kg/IU (IgE/mL) of Xolair or a matching volume of placebo over each 4-week period. The maximum Xolair dose per 4 weeks was 750 mg.

In all three studies an exacerbation was defined as a worsening of asthma that required treatment with systemic corticosteroids or a doubling of the baseline ICS dose. Most exacerbations were managed in the out-patient setting and the majority were treated with systemic steroids. Hospitalization rates were not significantly different between Xolair and placebo-treated patients; however, the overall hospitalization rate was small. Among those patients who experienced an exacerbation, the distribution of exacerbation severity was similar between treatment groups.

Studies 1 and 2

At screening, patients in Studies 1 and 2 had a forced expiratory volume in one second (FEV1) between 40% and 80% predicted. All patients had a FEV1 improvement of at least 12% following beta2-agonist administration. All patients were symptomatic and were being treated with inhaled corticosteroids (ICS) and short acting beta2-agonists. Patients receiving other concomitant controller medications were excluded, and initiation of additional controller medications while on study was prohibited. Patients currently smoking were excluded.

Each study was comprised of a run-in period to achieve a stable conversion to a common ICS (beclomethasone dipropionate), followed by randomization to Xolair or placebo. Patients received Xolair for 16 weeks with an unchanged corticosteroid dose unless an acute exacerbation necessitated an increase. Patients then entered an ICS reduction phase of 12 weeks during which ICS dose reduction was attempted in a step-wise manner.

The distribution of the number of asthma exacerbations per patient in each group during a study was analyzed separately for the stable steroid and steroid-reduction periods.

In both Studies 1 and 2 the number of exacerbations per patient was reduced in patients treated with Xolair compared with placebo (Table 5).

Measures of airflow (FEV1) and asthma symptoms were also evaluated in these studies. The clinical relevance of the treatment-associated differences is unknown. Results from the stable steroid phase Study 1 are shown in Table 6. Results from the stable steroid phase of Study 2 and the steroid reduction phases of both Studies 1 and 2 were similar to those presented in Table 6.

Table 5 Frequency of Asthma Exacerbations per Patient by Phase in Studies 1 and 2
Stable Steroid Phase (16 wks)
Study 1 Study 2
Exacerbations per patient Xolair
N = 268
(%)
Placebo
N = 257
(%)
Xolair
N = 274
(%)
Placebo
N = 272
(%)
0 85.8 76.7 87.6 69.9
1 11.9 16.7 11.3 25.0
≥ 2 2.2 6.6 1.1 5.1
p-Value 0.005 < 0.001
Mean number exacerbations/patient 0.2 0.3 0.1 0.4
Steroid Reduction Phase (12 wks)
Exacerbations per patient Xolair
N = 268
(%)
Placebo
N = 257
(%)
Xolair
N = 274
(%)
Placebo
N = 272
(%)
0 78.7 67.7 83.9 70.2
1 19.0 28.4 14.2 26.1
≥ 2 2.2 3.9 1.8 3.7
p-Value 0.004 < 0.001
Mean number exacerbations/patient 0.2 0.4 0.2 0.3

Table 6 Asthma Symptoms and Pulmonary Function During Stable Steroid Phase of Study 1
Asthma symptom scale: total score from 0 (least) to 9 (most); nocturnal and daytime scores from 0 (least) to 4 (most symptoms).
*
Number of patients available for analysis ranges 255-258 in the Xolair group and 238-239 in the placebo group.
Comparison of Xolair versus placebo (p < 0.05).
Xolair
N = 268*
Placebo
N = 257*
Endpoint Mean
Baseline
Median Change (Baseline to Wk 16) Mean
Baseline
Median Change (Baseline to Wk 16)
Total asthma symptom score 4.3 -1.5 † 4.2 -1.1†
    Nocturnal asthma score 1.2 -0.4† 1.1 -0.2†
    Daytime asthma score 2.3 -0.9† 2.3 -0.6†
FEV1 % predicted 68 3† 68 0†
Study 3

In Study 3, there was no restriction on screening FEV1, and unlike Studies 1 and 2, long-acting beta2-agonists were allowed. Patients were receiving at least 1000 μg/day fluticasone propionate and a subset was also receiving oral corticosteroids. Patients receiving other concomitant controller medications were excluded, and initiation of additional controller medications while on study was prohibited. Patients currently smoking were excluded.

The study was comprised of a run-in period to achieve a stable conversion to a common ICS (fluticasone propionate), followed by randomization to Xolair or placebo. Patients were stratified by use of ICS-only or ICS with concomitant use of oral steroids. Patients received Xolair for 16 weeks with an unchanged corticosteroid dose unless an acute exacerbation necessitated an increase. Patients then entered an ICS reduction phase of 16 weeks during which ICS or oral steroid dose reduction was attempted in a step-wise manner.

The number of exacerbations in patients treated with Xolair was similar to that in placebo-treated patients (Table 7). The absence of an observed treatment effect may be related to differences in the patient population compared with Studies 1 and 2, study sample size, or other factors.

Table 7 Percentage of Patients with Asthma Exacerbations by Subgroup and Phase in Study 3
Stable Steroid Phase (16 wks)
Inhaled Only Oral + Inhaled
Xolair
N = 126
Placebo
N = 120
Xolair
N = 50
Placebo
N = 45
% Patients with  ≥ 1 exacerbations 15.9 15.0 32.0 22.2
Difference
(95% CI)
0.9
(-9.7, 13.7)
9.8
(-10.5, 31.4)
Steroid Reduction Phase (16 wks)
Xolair
N = 126
Placebo
N = 120
Xolair
N = 50
Placebo
N = 45
% Patients with  ≥ 1 exacerbations 22.2 26.7 42.0 42.2
Difference
(95% CI)
-4.4
(-17.6, 7.4)
-0.2
(-22.4, 20.1)

  In all three of the studies, a reduction of asthma exacerbations was not observed in the Xolair-treated patients who had FEV1 > 80% at the time of randomization. Reductions in exacerbations were not seen in patients who required oral steroids as maintenance therapy.

Pediatric Patients 6 to < 12 Years of Age

Clinical studies with Xolair in pediatric patients 6 to 11 years of age have been conducted [see Use in Specific Populations (8.4)]

Pediatric Patients <6 Years of Age

Clinical studies have with Xolair in pediatric patients less than 6 years of age have not been conducted [see Use in Specific Populations (8.4)]

16 HOW SUPPLIED/STORAGE AND HANDLING

Xolair (omalizumab)  is supplied as a lyophilized, sterile powder in a single-use, 5 mL vial without preservatives. Each vial delivers 150 mg of Xolair upon reconstitution with 1.4 mL SWFI, USP. Each carton contains one single-use vial of Xolair® (omalizumab) NDC 50242-040-62.

Xolair should be shipped at controlled ambient temperature (≤ 30°C [ ≤ 86°F]). Store Xolair under refrigerated conditions 2-8°C (36-46°F). Do not use beyond the expiration date stamped on carton.

Use the solution for subcutaneous administration within 8 hours following reconstitution when stored in the vial at 2-8°C (36-46°F), or within 4 hours of reconstitution when stored at room temperature.

Reconstituted Xolair vials should be protected from direct sunlight.


部分中奥马佐单抗处方资料(仅供参考)
药品英文名
Omalizumab
药品别名
Xolair  奥马佐单抗
药物剂型
奥马佐单抗注射剂:150/Vial  75mg/Vial。 2~8℃冰箱冷藏,避光保存。运输温度30℃以下。
药理作用
本品为从仓鼠卵巢细胞组织混悬液在含庆大霉素的培养基培养而得到的基因重组人源化IgG单克隆抗体,能选择性与人免疫球蛋白E结合,抑制IgE与高亲和力的IgE受体(FcεRI)在肥大细胞和嗜碱性粒细胞表面的结合,从而减少过敏性介质的释放。在特异反应性患者中还能减少FcεRI的数量。
本品具有以下特性。
①与游离IgE结合,不与IgG或IgA结合。
②阻断IgE与其高亲和力受体结合。
③不与结合在肥大细胞或嗜碱性粒细胞上的IgE结合。
④抑制产IgE培养细胞合成IgE。
药动学
本品静脉给药后1~2h,血浆IgE开始降低。单次或多次静脉给药,血浆IgE显著抑制的持续时间均为2~4周。静脉给予2mg/kg负荷剂量后,在第7、第14、第28、第42、第56、第70日给予6次1mg/kg,2周内均达稳态血药浓度(超过30000ng/ml)。皮下用药后7~8日达血药浓度峰值,如在第1、第7、第14日给予0.15mg/kg,每2周给予相同剂量的治疗方案,达稳态血药浓度的时间也出现在2周内(稳态血药浓度约为2000ng/ml)。皮下用药后表观分布容积为78ml/kg,生物利用度为62%。能否经乳汁排泄尚不明确。总体清除率为每日2.4ml/kg。消除半衰期为20~26天。
适应证
抗哮喘,用于成人及12岁以上青少年,对常年气源性致敏原呈皮肤阳性反应,症状不能被吸人性皮质激素控制的中度至重度持续性哮喘,本品能减少这些哮喘患者哮喘加重的发生率。对其他过敏情况的安全性和疗效尚无资料。
禁忌证
对本品严重过敏者禁用。
注意事项
1.有对其他抗体制剂(尤其是来源于小鼠的制剂)过敏史者慎用。
2.急性支气管痉挛或哮喘持续状态患者慎用。
3.肝肾功能不全者慎用。
4.生殖毒性分级为B,短尾猴研究未发现母体、胚胎毒性和致畸作用,尚无孕妇体内研究资料,孕妇慎用。
5.本品经短尾猴乳汁分泌,IgG经人乳汁分泌,推测本品也通过乳汁分泌,哺乳期妇女慎用。
6.本品不能减轻严重哮喘的加重,不能用于严重支气管哮喘或其持续状态。
7.使用激素治疗改用本品时,激素不能突然停用,要在医师指导下逐渐减量。
8.没有医师指导,使用本品不能减少其他哮喘药物的剂量或停药。
9.12岁以下儿童用药安全性和疗效尚未知。
10.配制溶液单次使用,溶液2~8℃可保存8h,室温4h。
不良反应
本品严重不良反应为恶性肿瘤和过敏反应。 常见不良反应包括:注射部位反应(45%;注射部位损伤、发红、发热、烧灼感、刺激感、痒、蜂窝形成、疼痛、硬结、发炎,多在用药后1h发生,最多持续8h),病毒感染(23%),上呼吸道感染(20%),鼻窦炎(16%),头痛(15%),咽炎(11%)。 其他不良反应有:全身痛,疲乏,关节痛,骨折,腿痛,臂痛,头晕,瘙痒,皮炎,耳痛。
用法用量
1.静脉给药 过敏性哮喘 中度或重度常年性过敏性哮喘,本品2.5μg/kg或5.8μg/kg(按血清IgE的ng/ml计),与口服和(或)吸入糖皮质激素联用。其中,第1、第4日给予半量,第7日给予全量,以后每2周给予全量1次,共20周。
2.皮下给药
(1)过敏性哮喘:皮肤试验阳性或常年气源性致敏原体外反应阳性、吸入糖皮质激素不能完全控制症状的中至重度常年性哮喘,推荐用量为每次150~375mg,每2~4周1次。
(2)过敏性鼻炎:
①常年性:每次16μg/kg(按血清IgE的U/ml计),每4周皮下注射1~2次。
②季节性:每次150~300mg,每3~4周1次,给药次数根据血清总IgE而定(IgE水平在150U/ml以上时,每3周用药1次;IgE水平为30~150U/ml时,每4周用药1次)。
药物相应作用
尚未发现药物相互作用。


----------------------------------------------------------
注:以下产品规格和价格均不同,购买以咨询为准!
----------------------------------------------------------
产地国家:美国
原产地英文商品名:
XOLAIR 150mg/Vial
原产地英文药品名:
OMALIZUMAB
中文参考商品译名:
XOLAIR 150毫克/瓶
中文参考药品译名:
奥马佐单抗
生产厂家中文参考译名:
基因技术公司
生产厂家英文名:
Genentech
----------------------------------------------------------
产地国家:西班牙
原产地英文商品名:
XOLAIR 75mg/Vial
原产地英文药品名:
OMALIZUMAB
中文参考商品译名:
XOLAIR 75毫克/瓶
中文参考药品译名:
奥马佐单抗
生产厂家中文参考译名:
诺华制药
生产厂家英文名:
NOVARTIS FARMACEUTICA
----------------------------------------------------------
产地国家:西班牙
原产地英文商品名:
XOLAIR 150mg/Vial
原产地英文药品名:
OMALIZUMAB
中文参考商品译名:
XOLAIR 150毫克/瓶
中文参考药品译名:
奥马佐单抗
生产厂家中文参考译名:
诺华制药
生产厂家英文名:
NOVARTIS FARMACEUTICA
该药品相关信息网址1:
http://www.drugs.com/xolair.html
该药品相关信息网址2:
http://www.rxlist.com/xolair-drug.htm
该药品相关信息网址3:
http://dailymed.nlm.nih.gov/dailymed/archives/fdaDrugInfo.cfm?archiveid=15582

责任编辑:admin


相关文章
XOLAIR(omalizumab Solution for Injection)奥马珠单抗
Xolair(Omalizumab for s.c. injection)
奥马珠单抗获欧盟批准用于慢性荨麻疹
注射用奥马珠单抗XOLAIR(omalizumab)
注射用XOLAIR(奥马珠单抗omalizumab)-2014年获批新适应证
奥马佐单抗注射剂|Xolair(Omalizumab)
Xolair(索雷尔注射剂, 乐无喘, 柯耐尔)
Xolair(omalizumab,索雷尔,乐无喘,柯耐尔)
欧盟批准诺华公司抗哮喘药omalizumab(Xolair)上市
抗哮喘新药Xolair(omalizumab)的临床研究结果喜人
 

最新文章

更多

· FLUIMUCIL 10% (乙酰半胱...
· OLOPATADINE HYDROCHLOR...
· Tessalon Capsules(苯佐...
· OFEV(NINTEDANIB ESYLAT...
· Flutiform(复方丙酸氟替...
· Flutiform Aerosol(复方...
· ORKAMBI(lumacaftor/iva...
· METHY-F 10% Powder(消...
· PROAIR RESPICLICK(硫酸...
· Meptin Swinghaler(盐酸...

推荐文章

更多

· FLUIMUCIL 10% (乙酰半胱...
· OLOPATADINE HYDROCHLOR...
· Tessalon Capsules(苯佐...
· OFEV(NINTEDANIB ESYLAT...
· Flutiform(复方丙酸氟替...
· Flutiform Aerosol(复方...
· ORKAMBI(lumacaftor/iva...
· METHY-F 10% Powder(消...
· PROAIR RESPICLICK(硫酸...
· Meptin Swinghaler(盐酸...

热点文章

更多