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 >> 罕见病治疗药物 >> 其他治疗药 >> Soliris(eculizumab,艾库组单抗注射剂)

Soliris(eculizumab,艾库组单抗注射剂)

2012-11-10 14:49:41  作者:新特药房  来源:互联网  浏览次数:867  文字大小:【】【】【
简介: 2011年9月26日,美国食品药品管理局(FDA)已批准Soliris(eculizumab)用于治疗儿童和成人非典型溶血尿毒综合征(aHUS)。据Alexion介绍,aHUS是一种非常罕见、可危及生命的遗传性疾病。该病进行性损害重要器 ...

2011年9月23日,美国食品与药物管理局(FDA)批准Soliris(依库珠单抗)治疗一种罕见的慢性血液病—非典型溶血性尿毒症综合征(aHUS),该病可导致肾衰,并与死亡和卒中危险增加相关。
非典型HUS占所有溶血性尿毒症综合征病例的5%~10%
Soliris是一种靶向治疗药物,通过抑制在aHUS中起作用的蛋白质发挥作用。FDA最初于2007年3月批准Soliris治疗阵发性睡眠性血红蛋白尿(PNH),该病是一种罕见的血液病,可导致患者失能和过早死亡。
Soliris被归类为孤儿药(orphan drug)
对于aHUS,目前没有FDA批准的其他治疗药物,当前标准治疗(血浆置换或新鲜冷冻血浆输注)的安全性和疗效,尚没有在良好的对照试验中进行过研究。
美国FDA药物评价与研究中心血液学和肿瘤产品办公室主任理查(Richard Pazdur)说,“这是首个获准用于治疗这种致命性疾病的药物,首次批准将Soliris用于儿童”。
Soliris的安全性和有效性,在两项纳入37例aHUS成人和青少年患者的单组试验,以及一项纳入19例儿科病人和11例成年aHUS病人的回顾性研究中得到确立。在这些研究中,接受Soliris治疗病人的肾功能获得良好改善,包括对血浆疗法无反应的严重aHUS病人可停止透析治疗。接受Soliris治疗的病人还显示出血小板计数和其他与aHUS活动相关的血液参数的改善。
在用Soliris治疗的aHUS病人中,最常见的副作用包括高血压、腹泻、头痛、贫血、呕吐、恶心,上呼吸道和泌尿道感染,以及白细胞减少。
在Soliris新适应证获准的同时,对现存危险评价与减轻策略(REMS)也进行了扩展,以告知医务人员和病人关于已知的威胁生命的脑膜炎双球菌感染的危险。
Soliris将继续仅通过限制程序获取,处方医师必须加入一个注册程序,并对接受该药的病人提供用药指导。
美国初始批准:2007
Soliris® (艾库组单抗eculizumab)为静脉注射浓缩液
适应证和用途
Soliris是一种补体抑制剂适用于:
(1)T治疗阵发性睡眠性血红蛋白尿(PNH)患者减低溶血.
(2)治疗非典型溶血尿毒综合症(aHUS)患者抑制补体-介导血栓性微血管病.
Soliris在aHUS中的有效性是根据对血栓性微血管病(TMA)和肾功能的作用。正在另外患者中进行前瞻性临床试验证实Soliris在aHUS患者中的效益。
使用的限制
Soliris不适用于治疗有志贺毒素大肠杆菌[Shiga toxin E. coli]相关溶血尿毒综合征(STEC-HUS)患者。
剂量和给药方法
仅静脉输注给药。
PNH给药方案:
aHUS给药方案:
剂型和规格
300 mg单次使用小瓶每瓶含30mL的10mg/mL无菌,无防腐剂溶液.
禁忌证
下列情况禁忌Soliris:
(1)有未解决的严重的奈瑟菌感染脑膜炎患者.
(2)当前未接种对脑膜炎奈瑟菌疫苗患者,除非延迟Soliris治疗风险胜过发生脑膜炎双球菌感染。
警告和注意事项
(1)正在对严重的脑膜炎双球菌感染治疗患者终止Soliris。
(2)当给予Soliris至有任何其它系统感染患者慎用.
不良反应
(1)在PNH随机试验中最常报道不良反应(≥10%总体和大于安慰剂)是:头痛,鼻咽炎,背痛,和恶心.
(2)最常报道不良反应在aHUS单组前瞻性试验(≥15%合计每患者发生率)是:高血压,上呼吸道感染,腹泻,头痛,贫血,呕吐,恶心,泌尿道感染,和白细胞减少。
特殊人群中使用
(1)妊娠:根据动物资料,Soliris可能致胎儿危害.
(2)哺乳母亲:当给予哺乳妇女应谨慎对待 .
(3)儿童使用:PNH:未确定安全性和有效性。aHUS:安全性和有效性与成年患者相似.


Soliris 300 mg concentrate for solution for infusion
1. Name of the medicinal product
Soliris 300 mg concentrate for solution for infusion
2. Qualitative and quantitative composition
Eculizumab is a humanised monoclonal (IgG2/4κ ) antibody produced in NS0 cell line by recombinant DNA technology.
One vial of 30 ml contains 300 mg of eculizumab (10 mg/ml).
After dilution, the final concentration of the solution to be infused is 5 mg/ml.
Excipients with known effect: Sodium (5 mmol per vial)
For the full list of excipients, see section 6.1.
3. Pharmaceutical form
Concentrate for solution for infusion.
Clear, colorless, pH 7.0 solution.
4. Clinical particulars
4.1 Therapeutic indications
Soliris is indicated in adults and children for the treatment of patients with
- Paroxysmal nocturnal haemoglobinuria (PNH).
Evidence of clinical benefit of Soliris in the treatment of patients with PNH is limited to patients with history of transfusions.
- Atypical haemolytic uremic syndrome (aHUS) (see section 5.1).
4.2 Posology and method of administration
Soliris must be administered by a healthcare professional and under the supervision of a physician experienced in the management of patients with haematological and/or renal disorders.
Posology
Adult Patients:
In Paroxysmal Nocturnal Haemoglobinuria (PNH):
The PNH dosing regimen for adult patients (≥18 years of age) consists of a 4-week initial phase followed by a maintenance phase:
• Initial phase: 600 mg of Soliris administered via a 25 – 45 minute intravenous infusion every week for the first 4 weeks.
• Maintenance phase: 900 mg of Soliris administered via a 25 – 45 minute intravenous infusion for the fifth week, followed by 900 mg of Soliris administered via a 25 – 45 minute intravenous infusion every 14 ± 2 days (see section 5.1).
In Atypical Haemolytic Uremic Syndrome (aHUS):
The aHUS dosing regimen for adult patients (≥18 years of age) consists of a 4 week initial phase followed by a maintenance phase:
• Initial phase: 900 mg of Soliris administered via a 25 – 45 minute intravenous infusion every week for the first 4 weeks.
• Maintenance phase: 1,200 mg of Soliris administered via a 25 – 45 minute intravenous infusion for the fifth week, followed by 1,200 mg of Soliris administered via a 25 – 45 minute intravenous infusion every 14 ± 2 days (see section 5.1).
Paediatric patients:
Paediatric PNH and aHUS patients with body weight ≥ 40kg are treated with the adult dosing recommendations, respectively.
In paediatric PNH and aHUS patients with body weight below 40 kg, the Soliris dosing regimen consists of:

Patient Body Weight

Initial Phase

Maintenance Phase

30 to <40 kg

600 mg weekly x 2

900 mg at week 3; then 900 mg every 2 weeks

20 to <30 kg

600 mg weekly x 2

600 mg at week 3; then 600 mg every 2 weeks

10 to <20 kg

600 mg weekly x 1

300 mg at week 2; then 300 mg every 2 weeks

5 to <10 kg

300 mg weekly x 1

300 mg at week 2; then 300 mg every 3 weeks

Soliris has not been studied in patients with PNH who weigh less than 40kg. The posology of Soliris for PNH patients less than 40kg weight is based on the posology used for patients with aHUS and who weigh less than 40kg.
For adults and paediatric aHUS patients supplemental dosing of Soliris is required in the setting of concomitant PE/PI (plasmapheresis or plasma exchange, or fresh frozen plasma infusion):

Type of Plasma Intervention

Most Recent Soliris Dose

Supplemental Soliris Dose With Each Plasma Intervention

Timing of Supplemental Soliris Dose

Plasmapheresis or plasma exchange

300 mg

300 mg per each plasmapheresis or plasma exchange session

 

Within 60 minutes after each plasmapheresis or plasma exchange

≥600 mg

600 mg per each plasmapheresis or plasma exchange session

Fresh frozen plasma infusion

≥300 mg

300 mg per each unit of fresh frozen plasma

60 minutes prior to each 1 unit of fresh frozen plasma infusion


Treatment monitoring
aHUS patients should be monitored for signs and symptoms of thrombotic microangiopathy (TMA) (see section 4.4 aHUS laboratory monitoring).
Soliris treatment is recommended to continue for the patient's lifetime, unless the discontinuation of Soliris is clinically indicated (see section 4.4).
Elderly population
Soliris may be administered to patients aged 65 years and over. There is no evidence to suggest that any special precautions are needed when older people are treated – although experience with Soliris in this patient population is still limited.
Renal impairment
No dose adjustment is required for patients with renal impairment (see section 5.1).
Hepatic impairment
The safety and efficacy of Soliris have not been studied in patients with hepatic impairment.
Method of administration
Do not administer as an intravenous push or bolus injection. Soliris should only be administered via intravenous infusion as described below.
For instructions on dilution of the medicinal product before administration, see section 6.6.
The diluted solution of Soliris should be administered by intravenous infusion over 25 – 45 minutes via gravity feed, a syringe-type pump, or an infusion pump. It is not necessary to protect the diluted solution of Soliris from light during administration to the patient.
Patients should be monitored for one hour following infusion. If an adverse event occurs during the administration of Soliris, the infusion may be slowed or stopped at the discretion of the physician. If the infusion is slowed, the total infusion time may not exceed two hours in adults and adolescents (aged 12 years to under 18 years) and four hours in children aged less than 12 years.
4.3 Contraindications
Hypersensitivity to eculizumab, murine proteins or to any of the excipients listed in section 6.1.
Soliris therapy must not be initiated (see section 4.4):
in PNH patients:
with unresolved Neisseria meningitidis infection.
who are not currently vaccinated against Neisseria meningitidis.
in aHUS patients:
with unresolved Neisseria meningitidis infection.
who are not currently vaccinated against Neisseria meningitidis or do not receive prophylactic treatment with appropriate antibiotics until 2 weeks after vaccination.
4.4 Special warnings and precautions for use
Soliris is not expected to affect the aplastic component of anaemia in patients with PNH.
Meningococcal Infection
Due to its mechanism of action, the use of Soliris increases the patient's susceptibility to meningococcal infection (Neisseria meningitidis). These patients might be at risk of disease by uncommon serogroups (particularly Y, W135 and X), although meningococcal disease due to any serogroup may occur. To reduce the risk of infection, all patients must be vaccinated at least 2 weeks prior to receiving Soliris. PNH patients must be vaccinated 2 weeks prior to Soliris initiation. aHUS patients who are treated with Soliris less than 2 weeks after receiving a meningococcal vaccine must receive treatment with appropriate prophylactic antibiotics until 2 weeks after vaccination. Patients must be re-vaccinated according to current medical guidelines for vaccination use. Tetravalent vaccines against serotypes A, C, Y and W135 are strongly recommended, preferably conjugated ones.
Vaccination may not be sufficient to prevent meningococcal infection. Consideration should be given to official guidance on the appropriate use of antibacterial agents. Cases of serious or fatal meningococcal infections have been reported in Soliris-treated patients. All patients should be monitored for early signs of meningococcal infection, evaluated immediately if infection is suspected, and treated with appropriate antibiotics if necessary. Patients should be informed of these signs and symptoms and steps taken to seek medical care immediately. Physicians must discuss the benefits and risks of Soliris therapy with patients and provide them with a patient information brochure and a patient safety card (see Package Leaflet for a description).
Other Systemic Infections
Due to its mechanism of action, Soliris therapy should be administered with caution to patients with active systemic infections. Patients may have increased susceptibility to infections, especially with encapsulated bacteria. Patients should be provided with information from the Package Leaflet to increase their awareness of potential serious infections and the signs and symptoms of them.
Infusion Reactions
Administration of Soliris may result in infusion reactions or immunogenicity that could cause allergic or hypersensitivity reactions (including anaphylaxis), though immune system disorders within 48 hours of Soliris administration did not differ from placebo treatment in PNH, aHUS and other studies conducted with Soliris. In clinical trials, no PNH or aHUS patients experienced an infusion reaction which required discontinuation of Soliris. Soliris administration should be interrupted in all patients experiencing severe infusion reactions and appropriate medical therapy administered.
Immunogenicity
Infrequent, low titre antibody responses have been detected in Soliris-treated patients across all studies. In placebo controlled studies low titre responses have been reported with a frequency (3.4%) similar to that of placebo (4.8%). No patients have been reported to develop neutralizing antibodies following therapy with Soliris, and there has been no observed correlation of antibody development to clinical response or adverse events.
Immunization
Prior to initiating Soliris therapy, it is recommended that PNH and aHUS patients should initiate immunizations according to current immunization guidelines. Additionally, all patients must be vaccinated against meningococcus at least 2 weeks prior to receiving Soliris. Patients who are treated with Soliris less than 2 weeks after receiving a meningococcal vaccine must receive treatment with appropriate prophylactic antibiotics until 2 weeks after vaccination. If available, tetravalent, conjugated vaccines are recommended (see Meningococcal Infection).
Patients less than 18 years of age must be vaccinated against Haemophilus influenzae and pneumococcal infections, and strictly need to adhere to the national vaccination recommendations for each age group.
Anticoagulant therapy
Treatment with Soliris should not alter anticoagulant management.
PNH Laboratory Monitoring
PNH patients should be monitored for signs and symptoms of intravascular haemolysis, including serum lactate dehydrogenase (LDH) levels. PNH patients receiving Soliris therapy should be similarly monitored for intravascular haemolysis by measuring LDH levels, and may require dose adjustment within the recommended 14±2 day dosing schedule during the maintenance phase (up to every 12 days).
aHUS Laboratory Monitoring
aHUS patients receiving Soliris therapy should be monitored for thrombotic microangiopathy by measuring platelet counts, serum LDH and serum creatinine, and may require dose adjustment within the recommended 14±2 day dosing schedule during the maintenance phase (up to every 12 days).
Treatment Discontinuation for PNH
If PNH patients discontinue treatment with Soliris they should be closely monitored for signs and symptoms of serious intravascular haemolysis. Serious haemolysis is identified by serum LDH levels greater than the pre-treatment level, along with any of the following: greater than 25% absolute decrease in PNH clone size (in the absence of dilution due to transfusion) in one week or less; a haemoglobin level of <5 g/dL or a decrease of >4 g/dL in one week or less; angina; change in mental status; a 50% increase in serum creatinine level; or thrombosis. Monitor any patient who discontinues Soliris for at least 8 weeks to detect serious haemolysis and other reactions.
If serious haemolysis occurs after Soliris discontinuation, consider the following procedures/treatments: blood transfusion (packed RBCs), or exchange transfusion if the PNH RBCs are >50% of the total RBCs by flow cytometry; anticoagulation; corticosteroids; or reinstitution of Soliris. In PNH clinical studies, 16 patients discontinued the Soliris treatment regimen. Serious haemolysis was not observed.
Treatment Discontinuation for aHUS
Severe thrombotic microangiopathy complications were observed after Soliris discontinuation in the aHUS clinical studies. If aHUS patients discontinue treatment with Soliris, they should be monitored closely for signs and symptoms of severe thrombotic microangiopathy complications.
Severe thrombotic microangiopathy complications post discontinuation can be identified by (i) any two, or repeated measurement of any one, of the following: a decrease in platelet count of 25% or more as compared to either baseline or to peak platelet count during Soliris treatment; an increase in serum creatinine of 25% or more as compared to baseline or to nadir during Soliris treatment; or, an increase in serum LDH of 25% or more as compared to baseline or to nadir during Soliris treatment; or (ii) any one of the following: a change in mental status or seizures; angina or dyspnoea; or thrombosis.
Monitor any patient who discontinues Soliris for at least 12 weeks to detect severe thrombotic microangiopathy complications.
If severe thrombotic microangiopathy complications occur after Soliris discontinuation, consider reinstitution of Soliris treatment, supportive care with PE/PI, or appropriate organ-specific supportive measures including renal support with dialysis, respiratory support with mechanical ventilation or anticoagulation. In aHUS clinical studies, 18 patients (5 in the prospective studies) discontinued Soliris treatment. Seven (7) severe thrombotic microangiopathy complications were observed following the missed dose in 5 patients and Soliris was re-initiated in 4 of these 5 patients.
Educational materials
All physicians who intend to prescribe Soliris must ensure they are familiar with the physician's guide to prescribing. Physicians must discuss the benefits and risks of Soliris therapy with patients and provide them with a patient information brochure and a patient safety card.
Patients should be instructed that if they develop fever > 39°C, headache accompanied with fever and/or stiff neck or sensitivity to light, they should immediately seek medical care as these signs may be indicative of meningococcal infection.
Excipients
This medicinal product contains 5 mmol sodium per vial. It should be taken into consideration by patients on a controlled sodium diet.
4.5 Interaction with other medicinal products and other forms of interaction
No interaction studies have been performed.
4.6 Fertility, pregnancy and lactation
Women of childbearing potential
Women of childbearing potential have to use effective contraception during treatment and up to 5 months after treatment.
Pregnancy
For Soliris, no clinical data on exposed pregnancies are available.
Animal reproduction studies have not been conducted with eculizumab (see section 5.3).
Human IgG are known to cross the human placental barrier, and thus eculizumab may potentially cause terminal complement inhibition in the foetal circulation. Therefore, Soliris should be given to a pregnant woman only if clearly needed.
Breast-feeding
It is unknown whether eculizumab is excreted into human milk. Since many medicinal products and immunoglobulins are secreted into human milk, and because of the potential for serious adverse reactions in nursing infants, breast-feeding should be discontinued during treatment and up to 5 months after treatment.
Fertility
No specific study on fertility has been conducted.
4.7 Effects on ability to drive and use machines
Soliris has no or negligible influence on the ability to drive and use machines.
4.8 Undesirable effects
Summary of the safety profile
The most common adverse reactions were headache (occurred mostly in the initial phase), leukopenia, and the most serious adverse reaction was meningococcal infection.
Tabulated list of adverse reactions
Table 1 gives the adverse reactions observed from spontaneous reporting and in clinical trials in PNH and aHUS. Adverse reactions reported at a very common (≥1/10), common (≥1/100 to <1/10) or uncommon (≥1/1,000 to <1/100) frequency with eculizumab are listed by system organ class and preferred term. Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness.
Table 1: Adverse Reactions reported in 239 patients included in PNH and aHUS clinical trials and in postmarketing reports

 

MedDRA System Organ Class

Very Common

(≥1/10)

Common

(≥1/100 to <1/10)

Uncommon

(≥1/1,000 to <1/100)

Infection and infestations

 

Meningococcal sepsis, Meningococcal meningitis, Sepsis, Septic shock, Pneumonia, Aspergillus infection, Arthritis bacterial, Upper respiratory tract infection, Nasopharyngitis, Bronchitis, Oral Herpes, Gastrointestinal infection, Urinary tract infection, Cystitis, Viral infection

Neisseria infection, Lower respiratory tract infection, Fungal infection, Haemophilus influenzae infection, Abscess, Cellulitis, Influenza, Gingival infection, Infection, Sinusitis, Tooth infection, Impetigo

Neoplasms benign, malignant and unspecified (including cysts and polyps)

   

Malignant melanoma, Myelodysplastic syndrome

Blood and lymphatic system disorders

Leukopenia

Thrombocytopenia, Haemolysis*

Coagulopathy, Red blood cell agglutination, Abnormal clotting factor, Anaemia, Lymphopenia

Immune system disorders

 

Anaphylactic reaction

Hypersensitivity

Endocrine disorders

   

Basedow's disease

Metabolism and nutrition disorders

   

Anorexia, Decreased appetite

Psychiatric disorders

   

Depression, Anxiety, Insomnia, Sleep disorder, Abnormal dreams, Mood swings

Nervous system disorders

Headache

Dizziness, Paraesthesia, Dysgeusia

Syncope, Tremor

Eye disorders

   

Vision blurred, Conjunctival irritation

Ear and labyrinth disorders

 

Vertigo

Tinnitus

Cardiac disorders

   

Palpitation

Vascular disorders

 

Accelerated hypertension

Hypertension, Hypotension, Haematoma, Hot flush, Vein disorder

Respiratory, thoracic and mediastinal disorders

 

Cough, Nasal congestion, Pharyngolaryngeal pain, Throat irritation

Epistaxis, Rhinorrhoea

Gastrointestinal disorders

 

Diarrhoea, Vomiting, Nausea, Abdominal pain, Constipation, Dyspepsia

Peritonitis, Gastrooesophagal reflux disease, Abdominal distension, Gingival pain

Hepatobiliary disorders

   

Jaundice

Skin and subcutaneous tissue disorders

 

Rash, Alopecia, Dry skin, Pruritus

Urticaria, Dermatitis, Erythema, Petechiae, Skin depigmentation, Hyperhidrosis,

Musculoskeletal and connective tissue disorders

 

Arthralgia, Myalgia, Back pain, Neck pain, Pain in extremity

Trismus, Joint swelling, Muscle spasms, Bone pain

Renal and urinary disorders

 

Dysuria

Renal impairment, Haematuria

Reproductive system and breast disorders

 

Spontaneous penile erection

Menstrual disorder

General disorders and administration site conditions

 

Oedema, Infusion related reaction, Chest discomfort, Pyrexia, Chills, Fatigue, Asthenia

Chest pain, Infusion site paraesthesia, Infusion site pain, Extravasation, Influenza like illness, Feeling hot

Investigations

 

Coombs test positive*

Alanine aminotransferase increased, Aspartate aminotransferase increased, Gamma-glutamyltransferase increased, Haematocrit decreased, Haemoglobin decreased


*See paragraph Description of selected adverse reactions
Description of selected adverse reactions
In all PNH clinical studies the most serious adverse reaction was meningococcal septicaemia in two vaccinated PNH patients (see section 4.4). There were no meningococcal infections or deaths in the aHUS clinical studies.
Low titres of antibodies were detected in 2% of patients with PNH treated with Soliris. As with all proteins there is a potential for immunogenicity.
Cases of haemolysis have been reported in the setting of missed or delayed Soliris dose in PNH clinical trials (see also Section 4.4).
Cases of thrombotic microangiopathy have been reported in the setting of missed or delayed Soliris dose in aHUS clinical trials (see also Section 4.4).
Paediatric population
In children and adolescent PNH patients (aged 11 years to less than 18 years) included in the paediatric PNH Study M07-005, the safety profile appeared similar to that observed in adult PNH patients. The most common adverse reaction reported in paediatric patients was headache.
In aHUS patients, the safety profile in adolescents (patients aged 12 years to less than 18 years) is consistent with that observed in adults. In infant and children aHUS patients (aged 2 months to less than 12 years) included in the retrospective study C09-001r, the safety profile is similar to that observed in adult/adolescent aHUS patients. The most common (>10%) adverse reactions reported in paediatric patients were diarrhoea, vomiting, pyrexia, upper respiratory tract infection and headache.
Patients with other diseases
Safety Data from Other Clinical Studies
Supportive safety data were obtained in 11 clinical studies that included 716 patients exposed to eculizumab in six disease populations other than PNH and aHUS. There was an un-vaccinated patient diagnosed with idiopathic membranous glomerulonephropathy who experienced meningococcal meningitis. With regard to other AEs and considering all double-blind, placebo-controlled studies in patients diagnosed with diseases other than PNH (N=526 patients with Soliris; N=221 patients with placebo), AEs reported with Soliris at a frequency of 2% or greater than the frequency reported with placebo were: upper respiratory tract infection, rash, and injury.
4.9 Overdose

No case of overdose has been reported.
5. Pharmacological properties
5.1 Pharmacodynamic properties
 Pharmacotherapeutic group: Selective immunosuppressants, ATC code: L04AA25
Soliris is a recombinant humanised monoclonal IgG2/4k antibody that binds to the human C5 complement protein and inhibits the activation of terminal complement. The Soliris antibody contains human constant regions and murine complementarity-determining regions grafted onto the human framework light- and heavy-chain variable regions. Soliris is composed of two 448 amino acid heavy chains and two 214 amino acid light chains and has a molecular weight of approximately 148 kDa.
Soliris is produced in a murine myeloma (NS0 cell line) expression system and purified by affinity and ion exchange chromatography. The bulk drug substance manufacturing process also includes specific viral inactivation and removal steps.
Mechanism of action
Eculizumab, the active ingredient in Soliris, is a terminal complement inhibitor that specifically binds to the complement protein C5 with high affinity, thereby inhibiting its cleavage to C5a and C5b and preventing the generation of the terminal complement complex C5b-9. Eculizumab preserves the early components of complement activation that are essential for opsonization of microorganisms and clearance of immune complexes.
In PNH patients, uncontrolled terminal complement activation and the resulting complement-mediated intravascular haemolysis are blocked with Soliris treatment.
In most PNH patients, eculizumab serum concentrations of approximately 35 microgram/ml are sufficient for essentially complete inhibition of terminal complement-mediated intravascular haemolysis.
In PNH, chronic administration of Soliris resulted in a rapid and sustained reduction in complement-mediated haemolytic activity.
In aHUS patients, uncontrolled terminal complement activation and the resulting complement-mediated thrombotic microangiopathy are blocked with Soliris treatment.
All patients treated with Soliris when administered as recommended demonstrated rapid and sustained reduction in terminal complement activity. In all aHUS patients, eculizumab serum concentrations of approximately 50 - 100 microgram/ml are sufficient for essentially complete inhibition of terminal complement activity.
In aHUS, chronic administration of Soliris resulted in a rapid and sustained reduction in complement-mediated thrombotic microangiopathy.
Clinical efficacy and safety
Paroxysmal Nocturnal Haemoglobinuria
The safety and efficacy of Soliris in PNH patients with haemolysis were assessed in a randomized, double-blind, placebo-controlled 26 week study (C04-001). PNH patients were also treated with Soliris in a single arm 52 week study (C04-002), and in a long term extension study (E05-001). Patients received meningococcal vaccination prior to receipt of Soliris. In all studies, the dose of eculizumab was 600 mg every 7 ± 2 days for 4 weeks, followed by 900 mg 7 ± 2 days later, then 900 mg every 14 ± 2 days for the study duration. Eculizumab was administered as an intravenous infusion over 25 – 45 minutes.
In study C04-001 (TRIUMPH) PNH patients with at least 4 transfusions in the prior 12 months, flow cytometric confirmation of at least 10% PNH cells and platelet counts of at least 100,000/microliter were randomized to either Soliris (n = 43) or placebo (n = 44). Prior to randomization, all patients underwent an initial observation period to confirm the need for RBC transfusion and to identify the haemoglobin concentration (the "set-point") which would define each patient's haemoglobin stabilization and transfusion outcomes. The haemoglobin set-point was less than or equal to 9 g/dL in patients with symptoms and was less than or equal to 7 g/dL in patients without symptoms. Primary efficacy endpoints were haemoglobin stabilization (patients who maintained a haemoglobin concentration above the haemoglobin set-point and avoid any RBC transfusion for the entire 26 week period) and blood transfusion requirement. Fatigue and health-related quality of life were relevant secondary endpoints. Haemolysis was monitored mainly by the measurement of serum LDH levels, and the proportion of PNH RBCs was monitored by flow cytometry. Patients receiving anticoagulants and systemic corticosteroids at baseline continued these medications. Major baseline characteristics were balanced (see Table 2).
In the non-controlled study C04-002 (SHEPHERD), PNH patients with at least one transfusion in the prior 24 months and at least 30,000 platelets/microliter received Soliris over a 52-week period. Concomitant medications included anti-thrombotic agents in 63% of the patients and systemic corticosteroids in 40% of the patients. Baseline characteristics are shown in Table 2.
Table 2: Patient Demographics and Characteristics in C04-001 and C04-002

 

C04-001

C04-002

Parameter

Placebo

N = 44

Soliris

N = 43

Soliris

N = 97

Mean Age (SD)

38.4 (13.4)

42.1 (15.5)

41.1 (14.4)

Gender - Female (%)

29 (65.9)

23 (53.5)

49 (50.5)

History of Aplastic Anaemia or MDS (%)

12 (27.3)

8 (18.7)

29 (29.9)

Concomitant Anticoagulants (%)

20 (45.5)

24 (55.8)

59 (61)

Concomitant Steroids/Immunosuppressant Treatments (%)

16 (36.4)

14 (32.6)

46 (47.4)

Discontinued treatment

10

2

1

PRBC in previous 12 months (median (Q1,Q3))

17.0 (13.5, 25.0)

18.0 (12.0, 24.0)

8.0 (4.0, 24.0)

Mean Hgb level (g/dL) at setpoint (SD)

7.7 (0.75)

7.8 (0.79)

N/A

Pre-treatment LDH levels (median, U/L)

2,234.5

2,032.0

2,051.0

Free Haemoglobin at baseline (median, mg/dL)

46.2

40.5

34.9


In TRIUMPH, study patients treated with Soliris had significantly reduced (p< 0.001) haemolysis resulting in improvements in anaemia as indicated by increased haemoglobin stabilization and reduced need for RBC transfusions compared to placebo treated patients (see Table 3). These effects were seen among patients within each of the three pre-study RBC transfusion strata (4 - 14 units; 15 - 25 units; > 25 units). After 3 weeks of Soliris treatment, patients reported less fatigue and improved health-related quality of life. Because of the study sample size and duration, the effects of Soliris on thrombotic events could not be determined. In SHEPHERD study, 96 of the 97 enrolled patients completed the study (one patient died following a thrombotic event). A reduction in intravascular haemolysis as measured by serum LDH levels was sustained for the treatment period and resulted in increased transfusion avoidance, a reduced need for RBC transfusion and less fatigue. See Table 3.
Table 3: Efficacy Outcomes in C04-001 and C04-002

C04-001

C04-002*

 

Placebo

N = 44

Soliris

N = 43

P – Value

Soliris

N = 97

P – Value

Percentage of patients with stabilized Haemoglobin levels at end of study

0

49

< 0.001

N/A

PRBC transfused during treatment (median)

10

0

< 0.001

0

< 0.001

Transfusion Avoidance during treatment (%)

0

51

< 0.001

51

< 0.001

LDH levels at end of study (median, U/L)

2,167

239

< 0.001

269

< 0.001

LDH AUC at end of study (median, U/L x Day)

411,822

58,587

< 0.001

-632,264

< 0.001

Free Haemoglobin at end of study (median, mg/dL)

62

5

< 0.001

5

< 0.001

FACIT-Fatigue (effect size)

 

1.12

< 0.001

1.14

< 0.001


* Results from study C04-002 refer to pre- versus post-treatment comparisons.
From the 195 patients that originated in C04-001, C04-002 and other initial studies, Soliris-treated PNH patients were enrolled in a long term extension study (E05-001). All patients sustained a reduction in intravascular haemolysis over a total Soliris exposure time ranging from 10 to 54 months. There were fewer thrombotic events with Soliris treatment than during the same period of time prior to treatment. However, this finding was shown in non-controlled clinical trials.
Atypical Haemolytic Uremic Syndrome
Data from 37 patients in two prospective controlled studies (C08-002A/B and C08-003A/B) and one retrospective study with 30 patients (C09-001r) were used to evaluate the efficacy of Soliris in the treatment of aHUS.
Study C08-002A/B was a prospective, controlled, open-label study which accrued patients in the early phase of aHUS with evidence of clinical thrombotic microangiopathy manifestations with platelet count ≤ 150 x 109/L despite PE/PI, and LDH and serum creatinine above upper limits of normal. Study C08-003A/B was a prospective, controlled, open-label study which accrued patients with longer term aHUS without apparent evidence of clinical thrombotic microangiopathy manifestations and receiving chronic PE/PI (≥1 PE/PI treatment every two weeks and no more than 3 PE/PI treatments/week for at least 8 weeks before the first dose). Patients in both prospective studies were treated with Soliris for 26 weeks and most patients enrolled into a long-term, open-label extension study. All patients enrolled in both prospective studies had an ADAMTS-13 level above 5%.
Patients received meningococcal vaccination prior to receipt of Soliris or received prophylactic treatment with appropriate antibiotics until 2 weeks after vaccination. In all studies, the dose of Soliris in adult and adolescent aHUS patients was 900 mg every 7 ± 2 days for 4 weeks, followed by 1,200 mg 7 ± 2 days later, then 1,200 mg every 14 ± 2 days for the study duration. Soliris was administered as an intravenous infusion over 35 minutes. The dosing regimen in paediatric patients and adolescents weighing less than 40 kg was defined based on a pharmacokinetic (PK) simulation that identified the recommended dose and schedule based on body weight (see section 4.2).
Primary endpoints included platelet count change from baseline in study C08-002A/B and thrombotic microangiopathy (TMA) event-free status in study C08-003A/B. Additional endpoints included TMA intervention rate, haematologic normalization, complete TMA response, changes in LDH, renal function and quality of life. TMA-event free status was defined as the absence for at least 12 weeks of the following: decrease in platelet count of > 25% from baseline, PE/PI, and new dialysis. TMA interventions were defined as PE/PI or new dialysis. Haematologic normalization was defined as normalization of platelet counts and LDH levels sustained for ≥2 consecutive measurements for ≥4 weeks. Complete TMA response was defined as haematologic normalization and a ≥25% reduction in serum creatinine sustained in ≥ 2 consecutive measurements for ≥ 4 weeks.
Baseline characteristics are shown in Table 4.
Table 4: Patient Demographics and Characteristics in C08-002A/B and C08-003A/B

Parameter

C08-002A/B

C08-003A/B

Soliris

N = 17

Soliris

N = 20

Time from first diagnosis until screening in months, median (min, max)

10 (0.26, 236)

48 (0.66, 286)

Time from current clinical TMA manifestation until screening in months, median (min, max)

< 1 (<1, 4)

9 (1, 45)

Number of PE/PI sessions for current clinical TMA manifestation, median (min, max)

17 (2, 37)

62 (20, 230)

Number of PE/PI sessions in 7 days prior to first dose of eculizumab, median (min, max)

6 (0, 7)

2 (1, 3)

Baseline platelet count (× 109/L), mean (SD)

109 (32)

228 (78)

Baseline LDH (U/L), mean (SD)

323 (138)

223 (70)

Patients without identified mutation, n (%)

4 (24)

6 (30)

Patients in aHUS Study C08-002 A/B received Soliris for a minimum of 26 weeks. After completion of the initial 26-week treatment period, most patients continued to receive Soliris by enrolling into an extension study. In aHUS Study C08-002A/B, the median duration of Soliris therapy was approximately 64 weeks (range: 2 weeks to 90 weeks).
A reduction in terminal complement activity and an increase in platelet count relative to baseline were observed after commencement of Soliris. Reduction in terminal complement activity was observed in all patients after commencement of Soliris. Table 5 summarizes the efficacy results for aHUS Study C08-002A/B.
Renal function, as measured by eGFR, was improved during Soliris therapy. Four of the five patients who required dialysis at study entry were able to discontinue dialysis for the duration of Soliris treatment, and one patient developed a new dialysis requirement. Patients reported improved health-related quality of life (QoL).
In aHUS Study C08-002A/B, responses to Soliris were similar in patients with and without identified mutations in genes encoding complement regulatory factor proteins.
Patients in aHUS study C08-003A/B received Soliris for a minimum of 26 weeks. After completion of the initial 26-week treatment period, most patients continued to receive Soliris by enrolling into an extension study. In aHUS Study C08-003A/B, the median duration of Soliris therapy was approximately 62 weeks (range: 26 to 74 weeks). Table 5 summarizes the efficacy results for aHUS Study C08-003A/B.
In aHUS Study C08-003A/B, responses to Soliris were similar in patients with and without identified mutations in genes encoding complement regulatory factor proteins. Reduction in terminal complement activity was observed in all patients after commencement of Soliris. No patient required new dialysis with Soliris. Renal function, as measured by median eGFR, increased during Soliris therapy.
Table 5: Efficacy Outcomes in Prospective aHUS Studies C08-002A/B and C08-003A/B

C08-002A/B

N=17

C08-003A/B

N=20

Change in platelet count from baseline through week 26 (× 109/L), Point Estimate (95% CI)

73 (40-105)

P =0.0001

5 (-17.5-28)

P =0.64

Normalization of platelet count

All patients, n (%) (95% CI)

Patients with abnormal baseline, n/n (%)

14 (82) (57-96)

13/15 (87)

18 (90) (68-99)

3/20 (15)

TMA event-free status, n (%) (95% CI)

15 (88) (64-99)

16 (80) (56-94)

TMA intervention rate

Daily pre-eculizumab rate, median (min, max)

Daily post-eculizumab rate, median (min, max)

P-value

 

0.88 (0.04, 1.59)







0 (0, 0.31)







P<0.0001

 

0.23 (0.05, 1.09)







0







P <0.0001

CKD improvement by ≥1 stage, n (%) (95% CI)

10 (59) (33-82)

7 (35) (15-59)

eGFR change mL/min/1.73 m2: median (range) at 26 weeks

20 (-1, 98)

5 (-1, 20)

eGFR improvement ≥15 mL/min/1.73 m2, n (%) (95% CI)

9 (53) (28-77)1

1 (5) (0-25)

Change in Hgb > 20g/L, n (%) (95% CI)

11 (65) (38-86) 2

9 (45) (23-68) 3

Haematologic normalization, n (%) (95% CI)

13 (76) (50-93)

18 (90) (68-99)

Complete TMA response, n (%) (95% CI)

11(65) (38-86)

5 (25) (9-49)


1.At data cut-off
2 Study C08-002: 3 patients received ESA which was discontinued after eculizumab initiation
3 Study C08-003: 8 patients received ESA which was discontinued in 3 of them during eculizumab therapy
Paediatric population
Paroxysmal Nocturnal Haemoglobinuria
A total of 7 PNH paediatric patients, with a median weight of 57.2 kg (range of 48.6 to 69.8 kg) and aged from 11 to 17 years (median age : 15.6 years), received Soliris in study M07-005.
Treatment with eculizumab at the proposed dosing regimen in the paediatric population was associated with a reduction of intravascular haemolysis as measured by serum LDH level. It also resulted in a marked decrease or elimination of blood transfusions, and a trend towards an overall improvement in general function. The efficacy of eculizumab treatment in paediatric PNH patients appears to be consistent with that observed in adult PNH patients enrolled in PNH pivotal Studies (C04-001 and C04-002) (Table 3 and 6).
Table 6: Efficacy Outcomes in Paediatric PNH Study M07-005

P – Value

 

Mean (SD)

Wilcoxon Signed Rank

Paired t-test

Change from baseline at 12 weeks of LDH Value (U/L)

-771 (914)

0.0156

0.0336

LDH AUC

(U/L x Day)

-60,634 (72,916)

0.0156

0.0350

Change from baseline at 12 weeks in Plasma Free Haemoglobin (mg/dL)

-10.3 (21.13)

0.2188

0.1232

Change from baseline Type III RBC clone size (Percent of aberrant cells)

1.80 (358.1)

   

Change from baseline at 12 weeks of PedsQLTM4.0 Generic Core scale (patients)

10.5 (6.66)

0.1250

0.0256

Change from baseline at 12 weeks of PedsQLTM4.0 Generic Core scale (parents)

11.3 (8.5)

0.2500

0.0737

Change from baseline at 12 weeks of PedsQLTM Multidimensional Fatigue (patients)

0.8 (21.39)

0.6250

0.4687

Change from baseline at 12 weeks of PedsQLTM Multidimensional Fatigue (parents)

5.5 (0.71)

0.5000

0.0289

Atypical Haemolytic Uremic Syndrome
A total of 15 paediatric patients (aged 2 months to 12 years) received Soliris in aHUS Study C09-001r. Forty seven percent of patients had an identified complement regulatory factor mutation or auto-antibody. The median time from aHUS diagnosis to first dose of Soliris was 14 months (range <1, 110 months). The median time from current thrombotic microangiopathy manifestation to first dose of Soliris was 1 month (range <1 to 16 months). The median duration of Soliris therapy was 16 weeks (range 4 to 70 weeks) for children < 2 years of age (n=5) and 31 weeks (range 19 to 63 weeks) for children 2 to <12 years of age (n=10).
Overall, the efficacy results for these paediatric patients appeared consistent with what was observed in patients enrolled in aHUS pivotal Studies C08-002 and C08-003 (Table 5). No paediatric patient required new dialysis during treatment with Soliris.
Table 7: Efficacy Results in Paediatric Patients Enrolled in aHUS C09-001r

Efficacy Parameter

<2 years

(n=5)

2 to <12 years

(n=10)

<12 years

(n=15)

Patients with platelet count normalization, n (%)

4 (80)

10 (100)

14 (93)

Complete TMA response, n (%)

2 (40)

5 (50)

7 (50)

Daily TMA intervention rate, median (range)

Before eculizumab

On eculizumab treatment

 

1 (0, 2)

<1 (0, <1)

 

<1 (0.07, 1.46)

0 (0, <1)

 

<1 (0, 2)

0 (0, <1)

Patients with eGFR improvement ≥15 mL/min/1.73 m2, n (%)

2 (40)

6 (60)

8 (53)


In paediatric patients with shorter duration of current severe clinical thrombotic microangiopathy (TMA) manifestation prior to eculizumab, there was TMA control and improvement of renal function with eculizumab treatment (Table 8).
In paediatric patients with longer duration of current severe clinical TMA manifestation prior to eculizumab, there was TMA control with eculizumab treatment. However, renal function was not changed due to prior irreversible kidney damage (Table 8).
Table 8: Efficacy Outcomes in Paediatric Patients in Study C09-001r according to duration of current severe clinical thrombotic microangiopathy (TMA) manifestation

Duration of current severe clinical TMA manifestation

 

< 2 months

N=10 (%)

>2 months

N=5 (%)

Platelet count normalization

9 (90)

5 (100)

TMA event-free status

8 (80)

3 (60)

Complete TMA response

7 (70)

0

eGFR improvement ≥ 15 mL/min/1.73m2

7 (70)

0*

*One patient achieved eGFR improvement after renal transplant
The European Medicines Agency has deferred the obligation to submit the results of studies with Soliris in one or more subsets of the paediatric population in PNH and in aHUS (see section 4.2 for information on paediatric use).
5.2 Pharmacokinetic properties
Pharmacokinetics and Drug Metabolism
Biotransformation
Human antibodies undergo endocytotic digestion in the cells of the reticuloendothelial system. Eculizumab contains only naturally occurring amino acids and has no known active metabolites. Human antibodies are predominately catabolized by lysosomal enzymes to small peptides and amino acids.
Elimination
No specific studies have been performed to evaluate the hepatic, renal, lung, or gastrointestinal routes of excretion/elimination for Soliris. In normal kidneys, antibodies are not excreted and are excluded from filtration by their size.
Pharmacokinetic Parameters
In 40 patients with PNH, a 1-compartmental model was used to estimate pharmacokinetic parameters after multiple doses. Mean clearance was 0.31 ± 0.12 ml/hr/kg, mean volume of distribution was 110.3 ± 17.9 ml/kg, and mean elimination half-life was 11.3 ± 3.4 days. Based on these data, the onset of steady state is predicted to be approximately 49 – 56 days.
In PNH patients, pharmacodynamic activity correlates directly with eculizumab serum concentrations and maintenance of trough levels above ≥ 35 microgram/ml results in essentially complete blockade of haemolytic activity in the majority of PNH patients.
A second population PK analysis with a standard 1 compartmental model was conducted on the multiple dose PK data from 37 aHUS patients receiving the recommended Soliris regimen in studies C08-002A/B and C08-003A/B. In this model, the clearance of Soliris for a typical aHUS patient weighing 70 kg was 0.0139 L/hr and the volume of distribution was 5.6 L. The elimination half-life was 297 h (approximately 12.4 days).
The clearance and half-life of eculizumab were also evaluated during plasma exchange interventions. Plasma exchange resulted in an approximately 50% decline in eculizumab concentrations following a 1 hour intervention and the elimination half-life of eculizumab was reduced to 1.3 hours. Supplemental dosing is recommended when Soliris is administered to aHUS patients receiving plasma infusion or exchange (see section 4.2).
All aHUS patients treated with Soliris when administered as recommended demonstrated rapid and sustained reduction in terminal complement activity. In aHUS patients, pharmacodynamic activity correlates directly with eculizumab serum concentrations and maintenance of trough levels of approximately 50-100 microgram/ml result in essentially complete blockade of terminal complement activity in all aHUS patients.
Special Populations
PNH
Formal studies have not been conducted to evaluate the pharmacokinetics of Soliris administration in special PNH patient populations based on gender, race, age (geriatric), or renal or hepatic impairment.
Paediatric patients
The pharmacokinetics of eculizumab was evaluated in Study M07-005 including 7 PNH paediatric patients (aged from 11 to less than 18 years).
Weight was a significant covariate resulting in a lower eculizumab clearance 0.0105 L/h in the adolescent patients. Dosing for paediatric patients <40 kg is based on paediatric patients with aHUS.
aHUS
The pharmacokinetics of Soliris have been studied in aHUS patients with a range of renal impairment and age. There have been no observed differences in pharmacokinetic parameters noted in these subpopulations of aHUS patients.
5.3 Preclinical safety data
The specificity of eculizumab for C5 in human serum was evaluated in two in vitro studies.
The tissue cross-reactivity of eculizumab was evaluated by assessing binding to a panel of 38 human tissues. C5 expression in the human tissue panel examined in this study is consistent with published reports of C5 expression, as C5 has been reported in smooth muscle, striated muscle, and renal proximal tubular epithelium. No unexpected tissue cross-reactivity was observed.
In a 26 week toxicity study performed in mice with a surrogate antibody directed against murine C5, treatment did not affect any of the toxicity parameters examined. Haemolytic activity during the course of the study was effectively blocked in both female and male mice.
Animal reproduction studies have not been conducted with eculizumab. No clear treatment-related effects or adverse effects were observed in reproductive toxicology studies in mice with a surrogate terminal complement inhibitory antibody. When maternal exposure to the antibody occurred during organogenesis, two cases of retinal dysplasia and one case of umbilical hernia were observed among 230 offspring born to mothers exposed to the higher antibody dose (approximately 4 times the maximum recommended human Soliris dose, based on a body weight comparison); however, the exposure did not increase foetal loss or neonatal death.
No animal studies have been conducted to evaluate the genotoxic and carcinogenic potential of eculizumab or its effect on fertility.
6. Pharmaceutical particulars
6.1 List of excipients
Sodium phosphate, monobasic
Sodium phosphate, dibasic
Sodium chloride
Polysorbate 80
Water for injections
6.2 Incompatibilities
This medicinal product must not be mixed with other medicinal products except those mentioned in section 6.6.
6.3 Shelf life
30months.
After dilution, the medicinal product should be used immediately. However, chemical and physical stability has been demonstrated for 24 hours at 2°C – 8°C.
6.4 Special precautions for storage
Store in a refrigerator (2°C – 8°C).
Do not freeze.
Store in the original package in order to protect from light.
Soliris vials in the original package may be removed from refrigerated storage for only one single period of up to 3 days. At the end of this period the product can be put back in the refrigerator.
For storage conditions after dilution of the medicinal product, see section 6.3.
6.5 Nature and contents of container
30 ml of concentrate in a vial (Type I glass) with a stopper (butyl, siliconised), and a seal (aluminium) with flip-off cap (polypropylene).
Pack size of one vial.
6.6 Special precautions for disposal and other handling
 Prior to administration, the Soliris solution should be visually inspected for particulate matter and discolouration.
Instructions:
Reconstitution and dilution should be performed in accordance with good practices rules, particularly for the respect of asepsis.
Withdraw the total amount of Soliris from the vial(s) using a sterile syringe.
Transfer the recommended dose to an infusion bag.
Dilute Soliris to a final concentration of 5 mg/ml by addition to the infusion bag using sodium chloride 9 mg/ml (0.9%) solution for injection, sodium chloride 4.5 mg/ml (0.45%) solution for injection, or 5% dextrose in water, as the diluent.
The final volume of a 5 mg/ml diluted solution is 60 ml for 300 mg doses, 120 ml for 600 mg doses, 180 ml for 900 mg doses and 240 ml for 1,200 mg doses. The solution should be clear and colourless.
Gently agitate the infusion bag containing the diluted solution to ensure thorough mixing of the product and diluent.
The diluted solution should be allowed to warm to room temperature prior to administration by exposure to ambient air.
Discard any unused portion left in a vial, as the product contains no preservatives.
Any unused medicinal product or waste material should be disposed of in accordance with local requirements.
7. Marketing authorisation holder
Alexion Europe SAS
25 Boulevard de l'Amiral Bruix
75016 Paris
FRANCE
8. Marketing authorisation number(s)
EU/1/07/393/001
9. Date of first authorisation/renewal of the authorisation
Date of first authorisation : 20 June 2007
Date of latest renewal : 20 June 2012
10. Date of revision of the text
10/09/2013
Detailed information on this medicinal product is available on the website of the European Medicines Agency http://www.ema.europa.eu/

责任编辑:admin


相关文章
依库珠单抗注射剂|Soliris(eculizumab)
Eculizumab-治疗视神经脊髓炎谱系疾病初显效
常见肿瘤分子靶向治疗药物应用进展
肿瘤的靶向治疗药物选择
Soliris(艾库组单抗,eculizumab)
人源型抗C5单克隆抗体|Soliris®(eculizumab)
吉非替尼靶向药物-推动肿瘤治疗理念不断进步
吉非替尼等靶向治疗药物推动肿瘤治疗理念不断进步
吉非替尼等靶向治疗药物面世,推动肿瘤治疗新理念
人源型抗C5单克隆抗体Soliris(ECULIZUMAB)-治疗阵发性睡眠性血红蛋白尿
 

最新文章

更多

· Glybera(alipogene tip...
· Xuriden(uridine triace...
· STRENSIQ Subcutaneous ...
· Ryanodex injectable su...
· CARBAGLU(carglumic aci...
· Orphacol cap(胆酸硬胶...
· Cholbam(cholic acid ca...
· Orphacol cap(胆酸硬胶...
· Translarna(Ataluren)gr...
· REPLAGAL(α-半乳糖苷酶...

推荐文章

更多

· Glybera(alipogene tip...
· Xuriden(uridine triace...
· STRENSIQ Subcutaneous ...
· Ryanodex injectable su...
· CARBAGLU(carglumic aci...
· Orphacol cap(胆酸硬胶...
· Cholbam(cholic acid ca...
· Orphacol cap(胆酸硬胶...
· Translarna(Ataluren)gr...
· REPLAGAL(α-半乳糖苷酶...

热点文章

更多

· Xuriden(uridine triace...
· STRENSIQ Subcutaneous ...
· Glybera(alipogene tip...