Home>>Signaling Pathways>> Tyrosine Kinase>> EGFR>>Cetuximab (C225)

Cetuximab (C225) Sale

(Synonyms: 西妥昔单抗; C225) 目录号 : GC34217

西妥昔单抗是一种嵌合单克隆抗体,由鼠类抗 EGFR 单克隆抗体 M225 的可变区和人 IgG1 恒定区融合而成。

Cetuximab (C225) Chemical Structure

Cas No.:205923-56-4

规格 价格 库存 购买数量
1mg
¥1,890.00
现货
5mg
¥3,990.00
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.

101

客户使用产品发表文献 1

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

SCC1 and UM-SCC-22B cells

Preparation Method

The toxicity of cetuximab to SCC1 and UM-SCC-22B cells was determined by MTT assay. At 72 h or 120 h after treatment with different doses of cetuximab (ranging from 0.1 nM to 0.5 μM), the culture medium was replaced and 50 μl of 1.0 mg/ml sterile filtered 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT; Sigma) was added to each well.

Reaction Conditions

0.1 nM to 0.5 μM at 72 h or 120 h

Applications

Treatment with cetuximab produced only modest inhibition of cell proliferation on SCC1 cells in vitro as determined by MMT assay.

Animal experiment [2]:

Animal models

BALB/c (nu/nu) female nude mice

Preparation Method

Xenografts were established in female nude mice (BALB c[nu/nu]) by subcutaneous injection of head and neck squamous cell carcinoma cell lines a UT-SCC-14 and b UT-SCC-2. Cetuximab (1 mg/injection) was administered by intraperitoneal injection at day 10, 13 and 16. The tumour size was recorded at an interval of 2–3 days, n = 10-14.

Dosage form

1 mg, i.p.

Applications

Cetuximab treatment showed reduction in the nuclear accumulation of HIF-1α, while the overall HIF-1α expression was not significantly altered. And after cetuximab treatment a downregulation of CAIX was only found in UT-SCC-14 xenografts. Cetuximab treatment affects the tumour growth and the tumour partial oxygen pressure as measured by LiPc EPR oximetry.

References:

[1]. Niu G, et al. Cetuximab-based immunotherapy and radioimmunotherapy of head and neck squamous cell carcinoma. Clin Cancer Res. 2010 Apr 1;16(7):2095-105.

[2]. Gustafsson H, et al. EPR Oximetry of Cetuximab-Treated Head-and-Neck Tumours in a Mouse Model. Cell Biochem Biophys. 2017 Dec;75(3-4):299-309.

产品描述

Cetuximab is a chimeric monoclonal antibody generated from fusion of the variable region of the murine anti-EGFR monoclonal antibody M225 and the human IgG1 constant region. It produced antibody retains high affinity and specificity to EGFR and reduces immunogenicity.[1] Cetuximab bound with high affinity to FcγRI (EC50 = 0.13 nM) and FcγRIIIa (EC50 = 6 nM). It effectively induced ADCC across multiple tumor cell lines.[4] Treatment with 100?μg/ml cetuximab for 24h enhances the cytotoxicity effect of RSL3 treatment on KRAS mutant CRC cells.[2]

In vitro experiment indicated it that radiation enhances cetuximab (0.5 μg/ml)-mediated ADCC and activation of NK cells.[3] Treatment with 20 μg/mL cetuximab inhibited the proliferation of the parental UMSCC1 cell line (UMSCC1-P) ,while the three HNSCC cetuximab-resistant clones (C2, C5, and C11) were completely refractory to cetuximab.[6]

In vivo experiment it shown that cetuximab (13?mg/kg, s.c.) enhances the inhibitory effects of RSL3 and RSL3-induced ferroptosis.[2] In vivo, after i.v. injection of 4 doses of 10 mg/kg body-weight demonstrated that cetuximab markly inhibited tumor growth in SCC1 tumor bearing mice.[5] In vivo experiment it illustrated that cetuximab-treated (50 mg/kg, i.p.) tumors showed delayed growth, when mice were inoculated with the NSCLC H226 cell line individually with 2x106 tumor cells in the dorsal flank.[6]

References:
[1]. Xiong HQ, et al. Cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor, in combination with gemcitabine for advanced pancreatic cancer: a multicenter phase II Trial. J Clin Oncol. 2004 Jul 1;22(13):2610-6.
[2]. Yang J, et al. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021 Nov 13;12(11):1079.
[3]. Jin WJ, et al. Tumor-Specific Antibody, Cetuximab, Enhances the In Situ Vaccine Effect of Radiation in Immunologically Cold Head and Neck Squamous Cell Carcinoma. Front Immunol. 2020 Nov 12;11:591139.
[4]. Patel D, et al. IgG isotype, glycosylation, and EGFR expression determine the induction of antibody-dependent cellular cytotoxicity in vitro by cetuximab. Hum Antibodies. 2010;19(4):89-99.
[5]. Niu G, et al. Cetuximab-based immunotherapy and radioimmunotherapy of head and neck squamous cell carcinoma. Clin Cancer Res. 2010 Apr 1;16(7):2095-105.
[6]. Iida M, et al. Targeting the HER Family with Pan-HER Effectively Overcomes Resistance to Cetuximab. Mol Cancer Ther. 2016 Sep;15(9):2175-86.

西妥昔单抗是一种嵌合单克隆抗体,由鼠类抗 EGFR 单克隆抗体 M225 的可变区和人 IgG1 恒定区融合而成。它产生的抗体对 EGFR 保持高亲和力和特异性并降低免疫原性。[1] Cetuximab 以高亲和力结合 FcγRI (EC50 = 0.13 nM) 和 FcγRIIIa (EC50 = 6 nM)。它有效地诱导多种肿瘤细胞系的ADCC。[4]用100μg/ml西妥昔单抗处理24h增强了RSL3处理对KRAS突变CRC细胞的细胞毒性作用。[2]

体外实验表明,辐射增强了西妥昔单抗(0.5 μg/ml)介导的ADCC和NK细胞的活化。[3]用20 μg/mL西妥昔单抗治疗抑制了亲本细胞的增殖UMSCC1细胞系(UMSCC1-P),而三个HNSCC西妥昔单抗耐药克隆(C2、C5和C11)对西妥昔单抗完全耐药。[6]

体内实验表明,西妥昔单抗 (13mg/kg, s.c.) 增强了 RSL3 和 RSL3 诱导的铁死亡的抑制作用。[2] 在体内,静脉注射后。注射4次10 mg/kg体重的西妥昔单抗表明西妥昔单抗显着抑制SCC1荷瘤小鼠的肿瘤生长。[5] 体内实验表明西妥昔单抗处理(50 mg/kg, i.p.) 肿瘤显示延迟生长,当小鼠单独接种 NSCLC H226 细胞系时,在背侧有 2x106 个肿瘤细胞。[6]

Chemical Properties

Cas No. 205923-56-4 SDF
别名 西妥昔单抗; C225
Canonical SMILES [Cetuximab]
分子式 C6484H10042N1732O2023S36 分子量 145543.35
溶解度 储存条件 Store at 4°C, do not freeze
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。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 0.0069 mL 0.0344 mL 0.0687 mL
5 mM 0.0014 mL 0.0069 mL 0.0137 mL
10 mM 0.0007 mL 0.0034 mL 0.0069 mL
  • 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

Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer

Cell Death Dis 2021 Nov 13;12(11):1079.PMID:34775496DOI:10.1038/s41419-021-04367-3.

Cetuximab is approved for the treatment of metastatic colorectal cancer (mCRC) with RAS wild-type. Nevertheless, the prognosis remains poor and the effectiveness of Cetuximab is limited in KRAS mutant mCRC. Recently, emerging evidence has shown that ferroptosis, a newly discovered form of nonapoptotic cell death, is closely related to KRAS mutant cells. Here, we further investigated whether cetuximab-mediated regulation of p38/Nrf2/HO-1 promotes RSL3-induced ferroptosis and plays a pivotal role in overcoming drug resistance in KRAS mutant colorectal cancer (CRC). In our research, we used two KRAS mutant CRC cell lines, HCT116 and DLD-1, as models of intrinsic resistance to Cetuximab. The viability of cells treated with the combination of RSL3 and Cetuximab was assessed by the CCK-8 and colony formation assays. The effective of Cetuximab to promote RSL3-induced ferroptosis was investigated by evaluating lipid reactive oxygen species accumulation and the expression of the malondialdehyde and the intracellular iron assay. Cetuximab therapy contributed to regulating the p38/Nrf2/HO-1 axis, as determined by western blotting and transfection with small interfering RNAs. Cetuximab promoted RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 in KRAS mutant CRC cells, and this was further demonstrated in a xenograft nude mouse model. Our work reveals that Cetuximab enhances the cytotoxic effect of RSL3 on KRAS mutant CRC cells and that Cetuximab enhances RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 axis through the activation of p38 MAPK.

Cetuximab for treating non-small cell lung cancer

Expert Opin Biol Ther 2018 Apr;18(4):483-493.PMID:29534625DOI:10.1080/14712598.2018.1452906.

Epidermal Growth Factor Receptor (EGFR)-dependent signaling plays a crucial role in epithelial cancer biology, and dictated the development of several targeting agents. The mouse-human chimeric antibody Cetuximab was among the first to be developed. After about two decades of clinical research it has gained a significant place in the management of advanced colorectal and head and neck cancers, whereas its development in non small cell lung cancer (NSCLC) has not led to a place in routine clinical practice, because of marginal clinical benefit despite statistically significant Phase III trials. Recent data from ongoing trials suggest that more careful selection based on molecular markers may identify good responders. Areas covered: In this article, the authors review the literature concerning basic science studies identifying EGFR as a therapeutic target, pharmacological development of Cetuximab, its pharmacodynamics and pharmacokinetics, and clinical trials on Cetuximab in NSCLC, focusing on recent findings on putative predictive biomarkers. Expert opinion: Cetuximab currently has no role in NSCLC treatment outside of research settings. We argue that failure to identify a predictive biomarker early on has hampered its chances to enter routine practice. Although recent research suggests benefit in highly selected patient subsets, its potential impact is severely dampened by lack of regulatory body approval and the emergence of competitors for the same niches.

Radiotherapy plus Cetuximab or cisplatin in human papillomavirus-positive oropharyngeal cancer (NRG Oncology RTOG 1016): a randomised, multicentre, non-inferiority trial

Lancet 2019 Jan 5;393(10166):40-50.PMID:30449625DOI:10.1016/S0140-6736(18)32779-X.

Background: Patients with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma have high survival when treated with radiotherapy plus cisplatin. Whether replacement of cisplatin with cetuximab-an antibody against the epidermal growth factor receptor-can preserve high survival and reduce treatment toxicity is unknown. We investigated whether Cetuximab would maintain a high proportion of patient survival and reduce acute and late toxicity. Methods: RTOG 1016 was a randomised, multicentre, non-inferiority trial at 182 health-care centres in the USA and Canada. Eligibility criteria included histologically confirmed HPV-positive oropharyngeal carcinoma; American Joint Committee on Cancer 7th edition clinical categories T1-T2, N2a-N3 M0 or T3-T4, N0-N3 M0; Zubrod performance status 0 or 1; age at least 18 years; and adequate bone marrow, hepatic, and renal function. We randomly assigned patients (1:1) to receive either radiotherapy plus Cetuximab or radiotherapy plus cisplatin. Randomisation was balanced by using randomly permuted blocks, and patients were stratified by T category (T1-T2 vs T3-T4), N category (N0-N2a vs N2b-N3), Zubrod performance status (0 vs 1), and tobacco smoking history (≤10 pack-years vs >10 pack-years). Patients were assigned to receive either intravenous Cetuximab at a loading dose of 400 mg/m2 5-7 days before radiotherapy initiation, followed by Cetuximab 250 mg/m2 weekly for seven doses (total 2150 mg/m2), or cisplatin 100 mg/m2 on days 1 and 22 of radiotherapy (total 200 mg/m2). All patients received accelerated intensity-modulated radiotherapy delivered at 70 Gy in 35 fractions over 6 weeks at six fractions per week (with two fractions given on one day, at least 6 h apart). The primary endpoint was overall survival, defined as time from randomisation to death from any cause, with non-inferiority margin 1·45. Primary analysis was based on the modified intention-to-treat approach, whereby all patients meeting eligibility criteria are included. This study is registered with ClinicalTrials.gov, number NCT01302834. Findings: Between June 9, 2011, and July 31, 2014, 987 patients were enrolled, of whom 849 were randomly assigned to receive radiotherapy plus Cetuximab (n=425) or radiotherapy plus cisplatin (n=424). 399 patients assigned to receive Cetuximab and 406 patients assigned to receive cisplatin were subsequently eligible. After median follow-up duration of 4·5 years, radiotherapy plus Cetuximab did not meet the non-inferiority criteria for overall survival (hazard ratio [HR] 1·45, one-sided 95% upper CI 1·94; p=0·5056 for non-inferiority; one-sided log-rank p=0·0163). Estimated 5-year overall survival was 77·9% (95% CI 73·4-82·5) in the Cetuximab group versus 84·6% (80·6-88·6) in the cisplatin group. Progression-free survival was significantly lower in the Cetuximab group compared with the cisplatin group (HR 1·72, 95% CI 1·29-2·29; p=0·0002; 5-year progression-free survival 67·3%, 95% CI 62·4-72·2 vs 78·4%, 73·8-83·0), and locoregional failure was significantly higher in the Cetuximab group compared with the cisplatin group (HR 2·05, 95% CI 1·35-3·10; 5-year proportions 17·3%, 95% CI 13·7-21·4 vs 9·9%, 6·9-13·6). Proportions of acute moderate to severe toxicity (77·4%, 95% CI 73·0-81·5 vs 81·7%, 77·5-85·3; p=0·1586) and late moderate to severe toxicity (16·5%, 95% CI 12·9-20·7 vs 20·4%, 16·4-24·8; p=0·1904) were similar between the Cetuximab and cisplatin groups. Interpretation: For patients with HPV-positive oropharyngeal carcinoma, radiotherapy plus Cetuximab showed inferior overall survival and progression-free survival compared with radiotherapy plus cisplatin. Radiotherapy plus cisplatin is the standard of care for eligible patients with HPV-positive oropharyngeal carcinoma. Funding: National Cancer Institute USA, Eli Lilly, and The Oral Cancer Foundation.

ARID1A mutations confer intrinsic and acquired resistance to Cetuximab treatment in colorectal cancer

Nat Commun 2022 Sep 19;13(1):5478.PMID:36117191DOI:10.1038/s41467-022-33172-5.

Most colorectal (CRC) tumors are dependent on EGFR/KRAS/BRAF/MAPK signaling activation. ARID1A is an epigenetic regulator mutated in approximately 5% of non-hypermutated CRC tumors. Here we show that anti-EGFR but not anti-VEGF treatment enriches for emerging ARID1A mutations in CRC patients. In addition, we find that patients with ARID1A mutations, at baseline, are associated with worse outcome when treated with cetuximab- but not bevacizumab-containing therapies; thus, this suggests that ARID1A mutations may provide both an acquired and intrinsic mechanism of resistance to anti-EGFR therapies. We find that, ARID1A and EGFR-pathway genetic alterations are mutually exclusive across lung and colorectal cancers, further supporting a functional connection between these pathways. Our results not only suggest that ARID1A could be potentially used as a predictive biomarker for Cetuximab treatment decisions but also provide a rationale for exploring therapeutic MAPK inhibition in an unexpected but genetically defined segment of CRC patients.

Adagrasib with or without Cetuximab in Colorectal Cancer with Mutated KRAS G12C

N Engl J Med 2023 Jan 5;388(1):44-54.PMID:PMC9908297DOI:10.1056/NEJMoa2212419.

Background: Adagrasib, an oral small-molecule inhibitor of mutant KRAS G12C protein, has shown clinical activity in pretreated patients with several tumor types, including colorectal cancer. Preclinical studies suggest that combining a KRAS G12C inhibitor with an epidermal growth factor receptor antibody could be an effective clinical strategy. Methods: In this phase 1-2, open-label, nonrandomized clinical trial, we assigned heavily pretreated patients with metastatic colorectal cancer with mutant KRAS G12C to receive adagrasib monotherapy (600 mg orally twice daily) or adagrasib (at the same dose) in combination with intravenous Cetuximab once a week (with an initial loading dose of 400 mg per square meter of body-surface area, followed by a dose of 250 mg per square meter) or every 2 weeks (with a dose of 500 mg per square meter). The primary end points were objective response (complete or partial response) and safety. Results: As of June 16, 2022, a total of 44 patients had received adagrasib, and 32 had received combination therapy with adagrasib and Cetuximab, with a median follow-up of 20.1 months and 17.5 months, respectively. In the monotherapy group (43 evaluable patients), a response was reported in 19% of the patients (95% confidence interval [CI], 8 to 33). The median response duration was 4.3 months (95% CI, 2.3 to 8.3), and the median progression-free survival was 5.6 months (95% CI, 4.1 to 8.3). In the combination-therapy group (28 evaluable patients), the response was 46% (95% CI, 28 to 66). The median response duration was 7.6 months (95% CI, 5.7 to not estimable), and the median progression-free survival was 6.9 months (95% CI, 5.4 to 8.1). The percentage of grade 3 or 4 treatment-related adverse events was 34% in the monotherapy group and 16% in the combination-therapy group. No grade 5 adverse events were observed. Conclusions: Adagrasib had antitumor activity in heavily pretreated patients with metastatic colorectal cancer with mutant KRAS G12C, both as oral monotherapy and in combination with Cetuximab. The median response duration was more than 6 months in the combination-therapy group. Reversible adverse events were common in the two groups. (Funded by Mirati Therapeutics; KRYSTAL-1 ClinicalTrials.gov number, NCT03785249.).