Home>>Signaling Pathways>> Others>> Inhibitory Antibodies>>Naxitamab

Naxitamab

(Synonyms: Naxitamab-gqgk; Humanized 3F8; Hu3F8) 目录号 : GC68338

Naxitamab (Hu3F8) 是一种人源化单克隆抗体,靶向二氮冈糖苷 GD2 (hu3F8)。Naxitamab 可用于神经母细胞瘤、骨肉瘤和其他 GD2 阳性癌症的研究。

Naxitamab Chemical Structure

Cas No.:1879925-92-4

规格 价格 库存 购买数量
5mg
¥5,175.00
现货
10mg
¥8,550.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

产品描述

Naxitamab (Hu3F8) is a humanized monoclonal antibody targeting the disialoganglioside GD2. Naxitamab can be used in research of neuroblastoma, osteosarcoma and other GD2-positive cancers[1].

Naxitamab (Hu3F8; 72 h) has cytotoxicity against neuroblastoma cell line LAN-1 with an EC50 value of 5.1 μg/mL[1].
Naxitamab (0.1-1 μg/mL; 4 h; peripheral blood mononuclear cells (PBMC) and polymorphonuclear leukocytes (PMN)) has antibody-dependent cell-mediated cytotoxic effects (ADCC)[1].

Naxitamab (Hu3F8; 100 mg/kg; i.v.; twice a week, for 4 weeks; athymic nude mice with LAN-1 xenografts) inhibits tumor growth in neuroblastoma xenografts[1].

Animal Model: Female athymic nude mice with LAN-1 xenografts[1]
Dosage: 100 mg/kg
Administration: Intravenous injection; twice a week, for 4 weeks
Result: Inhibited tumor growth and prolonged the survival time.

[1]. Cheung NK, et, al. Humanizing murine IgG3 anti-GD2 antibody m3F8 substantially improves antibody-dependent cell-mediated cytotoxicity while retaining targeting in vivo. Oncoimmunology. 2012 Jul 1;1(4):477-486.
[2]. Markham A. Naxitamab: First Approval. Drugs. 2021 Feb;81(2):291-296.

Chemical Properties

Cas No. 1879925-92-4 SDF Download SDF
别名 Naxitamab-gqgk; Humanized 3F8; Hu3F8
分子式 分子量
溶解度 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

Naxitamab: First Approval

Drugs 2021 Feb;81(2):291-296.PMID:33616889DOI:10.1007/s40265-021-01467-4.

Naxitamab (DANYELZA®, naxitamab-gqgk) is a humanised (IgG1) anti-GD2 (hu3F8) monoclonal antibody was developed by the Memorial Sloan Kettering Cancer Center (with commercial rights licenced to Y-mAbs therapeutics Inc.) for the treatment of neuroblastoma, osteosarcoma and other GD2-positive cancers. Naxitamab was recently granted accelerated approval by the US FDA for marketing as treatment (in combination with granulocyte-macrophage colony-stimulating factor) for paediatric patients at least one year of age and adult patients with relapsed or refractory high-risk neuroblastoma in the bone or bone marrow demonstrating a partial response, minor response, or stable disease to prior therapy. This article summarizes the milestones in the development of Naxitamab leading to this first approval.

Naxitamab combined with granulocyte-macrophage colony-stimulating factor as consolidation for high-risk neuroblastoma patients in complete remission

Pediatr Blood Cancer 2021 Oct;68(10):e29121.PMID:34022112DOI:10.1002/pbc.29121.

Background: Naxitamab is a humanized anti-disialoganglioside (GD2) monoclonal antibody approved for treatment of bone/bone marrow refractory high-risk neuroblastoma (HR-NB). Compassionate use (CU) expanded access program at Hospital Sant Joan de Deu permitted treatment of patients in complete remission (CR). We here report the survival, toxicity, and relapse pattern of patients in first or second CR treated with Naxitamab and sargramostim (GM-CSF). Procedure: Seventy-three consecutive patients with HR-NB (stage M at age >18 months or MYCN-amplified stages L1/L2 at any age) were treated in first or second CR. Treatment comprised five cycles of subcutaneous (SC) GM-CSF for 5 days at 250 μg/m2 /day (days -4 to 0), followed by Naxitamab + SC GM-CSF for 5 days at 500 μg/m2 /day (days 1-5). Naxitamab was infused over 30 minutes at 3 mg/kg/day, days 1, 3, and 5, outpatient. Results: Fifty-five patients were in first CR and 18 in second CR. Seventeen patients had MYCN-amplified NB and 11 detectable minimal residual disease in the bone marrow. Fifty-eight (79.5%) patients completed therapy. Four (5%) experienced grade 4 toxicities and 10 (14%) early relapse. Three-year event-free survival (EFS) 58.4%, 95% CI = (43.5%, 78.4%) and overall survival (OS) 82.4%, 95% CI = (66.8%, 100%). First CR patients 3-year EFS 74.3%, 95% CI = (62.7%, 88.1%), and OS 91.6%, 95% CI = (82.4%, 100%). EFS is significantly different between first and second CR (p = .0029). The pattern of relapse is predominantly (75%) of an isolated organ, mainly bone (54%). Univariate Cox models show prior history of relapse as the only statistically significant predictor of EFS but not OS. Conclusions: Consolidation with Naxitamab and GM-CSF resulted in excellent survival rates for HR-NB patients in CR.

Naxitamab: a humanized anti-glycolipid disialoganglioside (anti-GD2) monoclonal antibody for treatment of neuroblastoma

Drugs Today (Barc) 2021 Nov;57(11):677-688.PMID:34821881DOI:10.1358/dot.2021.57.11.3343691.

Therapy for high-risk neuroblastoma (HR NBL) is comprised of multimodal therapy including chemotherapy, surgery, radiation therapy, myeloablative therapy followed by autologous hematopoietic stem cell transplant, and immunotherapy. GD2 is a disialoganglioside that is highly expressed on the surface of neuroblastoma cells, with limited expression on normal tissues, which makes it an attractive target for immunologic therapy. The combination of immunotherapy with murine and chimeric anti-GD2 antibody formulations has improved outcomes compared with standard therapy in HR NBL patients. Naxitamab (Danyelza), a fully humanized anti-GD2 antibody, was developed at Memorial Sloan Kettering Cancer Center (MSKCC) to mitigate adverse reactions related to intolerance of foreign murine and chimeric antigens. Phase I and II studies demonstrating the tolerability and efficacy of Naxitamab in patients with relapsed/refractory (r/r) HR NBL prompted its approval by the U.S. Food and Drug Administration (FDA) in 2020 for HR NBL with bone or bone marrow involvement. Initial outcomes with Naxitamab are encouraging; however, future trials to maximize drug tolerance and elucidate its optimal role in neuroblastoma therapy in conjunction with other treatment strategies are needed. This review discusses the use of Naxitamab in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment of r/r HR NBL.

How we approach the treatment of patients with high-risk neuroblastoma with Naxitamab: experience from the Hospital Sant Joan de Déu in Barcelona, Spain

ESMO Open 2022 Apr;7(2):100462.PMID:35397431DOI:10.1016/j.esmoop.2022.100462.

Naxitamab [humanized 3f8 (hu3F8)] is a humanized monoclonal antibody (mAb) targeting the disialoganglioside GD2. It was approved in 2020 by the United States Food and Drug Administration (FDA) in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) for treatment of pediatric and adult patients with relapsed/refractory high-risk neuroblastoma, limited to the bone or bone marrow (BM). The team at Sant Joan de Déu Children's Hospital in Barcelona, Spain, have been using Naxitamab to treat neuroblastoma under clinical trial protocols [e.g. Trial 201, and hu3F8, irinotecan, temozolomide, and sargramostim (GM-CSF) (HITS) study] and compassionate use since 2017. The team has experience with two primary regimens: Naxitamab with GM-CSF only, or Naxitamab in combination with irinotecan, temozolomide, and GM-CSF (chemoimmunotherapy). This article aims to provide a practical overview of the team's experience with Naxitamab to date, including preparing the treatment room and selecting the team. Adverse event management, including the use of ketamine to manage pain during anti-GD2 mAb infusions, is also discussed. We hope this will provide practical information for other health care providers considering offering this treatment.

Outpatient administration of Naxitamab in combination with granulocyte-macrophage colony-stimulating factor in patients with refractory and/or relapsed high-risk neuroblastoma: Management of adverse events

Cancer Rep (Hoboken) 2023 Jan;6(1):e1627.PMID:35579862DOI:10.1002/cnr2.1627.

Background: Naxitamab is a humanized GD2-binding monoclonal antibody that received accelerated approval from the U.S. Food and Drug Administration for refractory or relapsed high-risk neuroblastoma limited to bone or bone marrow. Trial 201 (NCT03363373) is an ongoing global clinical trial to evaluate the efficacy and safety of Naxitamab in combination with granulocyte-macrophage colony-stimulating factor in this population. Aims: Here, we review the safety profile and adverse event (AE) management associated with Naxitamab administration in a pediatric population, based on Trial 201 protocol guidelines and clinical trial experience. Methods and results: At least 50% of patients experienced pain, hypotension, bronchospasm, cough, vomiting, diarrhea, nausea, and tachycardia, with the following reported at grade ≥3 AEs for at least 10% of patients: pain, hypotension, urticaria, and bronchospasm. These AEs were generally manageable in the outpatient setting using premedications, supportive therapies, and appropriate monitoring post-infusion. Algorithms were established for infusion-related AEs, including hypotension and bronchospasm, to provide guidance to investigators for early recognition and timely intervention, including medication and infusion rate modification or interruption, or treatment discontinuation, based on AE severity. Educating patients and caregivers on what to expect regarding premedication at home, experience during the infusion cycle, and post-infusion monitoring helps optimize Naxitamab treatment and supportive therapies and may reduce treatment burden. Conclusion: This article highlights the protocol-based recommendations for the management of acute AEs associated with outpatient Naxitamab treatment in Trial 201. The authors recommend close monitoring and timely implementation of measures to ensure that patients can remain on treatment and obtain maximum clinical benefit from Naxitamab therapy.