Home>>Signaling Pathways>> Antibody-drug Conjugate/ADC Related>> ADC Cytotoxin>>Monomethyl auristatin E

Monomethyl auristatin E Sale

(Synonyms: 一甲基澳瑞他汀E,Vedotin; MMAE) 目录号 : GC17276

A potent antimitotic compound

Monomethyl auristatin E Chemical Structure

Cas No.:474645-27-7

规格 价格 库存 购买数量
5mg
¥315.00
现货
10mg
¥504.00
现货
50mg
¥945.00
现货
100mg
¥1,395.00
现货
500mg
¥4,140.00
现货
1g
¥6,120.00
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:

实验参考方法

Cell experiment [1]:

Cell lines

MCF7-R1 cells

Preparation Method

100,000 MCF7-R1 cells were seeded into each well of a 12-well culture plate, allowed to adhere overnight, and treated with 10 nM Monomethyl Auristatin E (MMAE) or conjugates in the presence of 10 U/mL heparin for 72 h. Then the cells were harvested with a TrypLE Express solution, stained by Annexin V-FITC and propidium iodide, and analyzed by flow cytometry using a NovoCyte 2060R flow cytometer.

Reaction Conditions

10 nM; for 72 h

Applications

After 72 h of treatment of MCF7-R1 cells with 10 nM conjugates, the cells were mainly in the early stage of apoptosis.

Animal experiment [2]:

Animal models

Mice

Preparation Method

The breast tumor models were prepared via subcutaneous injection of 1 × 106 4T1 cells. When the tumor volumes were approximately 100 mm3, mice were divided into three groups: (i) saline; (ii) Monomethyl Auristatin E (MMAE) (0.2 mg/kg); and (iii) FRRG-MMAE nanoparticles (equivalent dose of 0.2 mg/kg based on MMAE contents). The mice were treated once every three days, and tumor volumes were calculated as the smallest diameter 2 × largest diameter × 0.53. The mice with a tumor volume of 2000 mm3 or larger were counted as dead. The toxicity study of FRRG-MMAE nanoparticles was assessed via histology.

Dosage form

0.2 mg/kg

Applications

The results demonstrated that free MMAE-treated mice showed rapid tumor growth compared to FRRG-MMAE-treated mice during monitoring for 15 days.Finally, mice treated with free MMAE showed significant body weight loss owing to the severe toxicity; eventually, all the mice were dead within five days of treatment.These results show the significantly reduced MMAE-related toxicity in the FRRG-MMAE nanoparticle group.

References:

[1] Krzyscik MA, et al. Fibroblast Growth Factor 2 Conjugated with Monomethyl Auristatin E Inhibits Tumor Growth in a Mouse Model. Biomacromolecules. 2021 Oct 11;22(10):4169-4180.

[2] Cho H, et al. Tumor-Specific Monomethyl Auristatin E (MMAE) Prodrug Nanoparticles for Safe and Effective Chemotherapy. Pharmaceutics. 2022 Oct 7;14(10):2131.

产品描述

Monomethyl Auristatin E (MMAE), as a synthetic derivative of dolastatin 10, a linear pentapeptide originally isolated from the extracts of the sea hare Dolabella auriculari. Monomethyl Auristatin E, as a synthetic derivative of dolastatin 10, a linear pentapeptide originally isolated from the extracts of the sea hare Dolabella auriculari. Monomethyl Auristatin E can inhibit tubulin polymerization, thus blocking mitosis.[1]

In vitro, Monomethyl Auristatin E and Monomethyl Auristatin E-phosphate shown the inhibition with IC50 of 2 and 48 nM, respectively, in PC-3 and C4-2B cell lines.[1] In vitro experiment it shown that 5 nM MMAE resulted in 50% of HCT-116 cells blocked in G2/M and 2 nM in PANC-1 cells.[2] MMAE shown the inhibition of cell growth with IC50 of 1.7 nM in HCT-116, 0.6 nM in PANC-1, and 5.6 nM in 779E cells, respectively.[2] In addition, MMAE also showed markedly polarized transport in both MDCK-WT and MDCK-MDR1 cells with ERs at 13.6 and 44.5, respectively, resulting ratio of ratios of 3.3. MMAE shown dose-dependent cytotoxicity in HepG2, Hep3B2, H226, N87 or OVCAR3 cells.[3]

In vivo, treatment with 30 mg/kg cAC10-vcMMAE in mice had no signs of toxicity.[4] In vivo efficacy test it shown that treatment with 6.5 mg/kg DD1-MMAE (the bivalent DARPin dimer) or DFc-MMAE (a DARPin-Fc) twice weekly did not cause any sequela in mice.[5] Moreover, nude mice were injected intravenously 2.5, 5, and 10 mg/kg hertuzumab-vcMMAE on day 0 resulted in obvious and sustained antitumor effects. [6]

References:
[1]Abawi A, et al. Monomethyl Auristatin E Grafted-Liposomes to Target Prostate Tumor Cell Lines. Int J Mol Sci. 2021 Apr 15;22(8):4103.
[2]Buckel L, et al. Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery. Cancer Res. 2015 Apr 1;75(7):1376-1387.
[3]Liu-Kreyche P, et al. Lysosomal P-gp-MDR1 Confers Drug Resistance of Brentuximab Vedotin and Its Cytotoxic Payload Monomethyl Auristatin E in Tumor Cells. Front Pharmacol. 2019 Jul 17;10:749.
[4]Francisco JA, et al. cAC10-vcMMAE, an anti-CD30-monomethyl auristatin E conjugate with potent and selective antitumor activity. Blood. 2003 Aug 15;102(4):1458-65.
[5]Karsten L, et al. Bivalent EGFR-Targeting DARPin-MMAE Conjugates. Int J Mol Sci. 2022 Feb 23;23(5):2468.
[6]Li H, et al. An anti-HER2 antibody conjugated with monomethyl auristatin E is highly effective in HER2-positive human gastric cancer. Cancer Biol Ther. 2016 Apr 2;17(4):346-54.

Monomethyl Auristatin E (MMAE),作为海兔毒素 10 的合成衍生物,海兔毒素 10 是一种线性五肽,最初是从海兔 Dolabella auriculari 的提取物中分离出来的。 Monomethyl Auristatin E,作为海兔毒素 10 的合成衍生物,海兔毒素 10 是一种线性五肽,最初是从海兔 Dolabella auriculari 的提取物中分离出来的。单甲基 Auristatin E 可以抑制微管蛋白聚合,从而阻断有丝分裂。[1]

在体外,Monomethyl Auristatin E 和 Monomethyl Auristatin E-phosphate 在 PC-3 和 C4-2B 细胞系中的 IC50 分别为 2 和 48 nM。[1]体外实验表明,5 nM MMAE 导致 50% 的 HCT-116 细胞在 G2/M 期受阻,2 nM 在 PANC-1 细胞中受阻。[2] MMAE 显示出抑制细胞生长的 IC50在 HCT-116 中为 1.7 nM,在 PANC-1 中为 0.6 nM,在 779E 细胞中为 5.6 nM。[2] 此外,MMAE 在 MDCK-WT 和 MDCK 中也表现出明显的极化转运-ER 分别为 13.6 和 44.5 的 MDR1 细胞,所得比率为 3.3。 MMAE 在 HepG2、Hep3B2、H226、N87 或 OVCAR3 细胞中表现出剂量依赖性细胞毒性。[3]

在体内,用 30 mg/kg cAC10-vcMMAE 处理小鼠没有毒性迹象。[4] 体内药效试验表明,用 6.5 mg/kg DD1-MMAE 处理(二价 DARPin 二聚体)或 DFc-MMAE(一种 DARPin-Fc)每周两次在小鼠体内未引起任何后遗症。[5] 此外,裸鼠静脉注射 2.5、5 和 10 mg/ kg hertuzumab-vcMMAE 在第 0 天产生明显和持续的抗肿瘤作用。 [6]

Chemical Properties

Cas No. 474645-27-7 SDF
别名 一甲基澳瑞他汀E,Vedotin; MMAE
化学名 (2S)-N-[(2S)-1-[[(3R,4S,5S)-1-[(2S)-2-[(1R,2R)-3-[[(1S,2R)-1-hydroxy-1-phenylpropan-2-yl]amino]-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl]-3-methoxy-5-methyl-1-oxoheptan-4-yl]-methylamino]-3-methyl-1-oxobutan-2-yl]-3-methyl-2-(methylamino)butanamide
Canonical SMILES CCC(C)C(C(CC(=O)N1CCCC1C(C(C)C(=O)NC(C)C(C2=CC=CC=C2)O)OC)OC)N(C)C(=O)C(C(C)C)NC(=O)C(C(C)C)NC
分子式 C39H67N5O7 分子量 717.98
溶解度 ≥ 35.9 mg/mL in DMSO, ≥ 48.5 mg/mL in EtOH with ultrasonic and warming 储存条件 Store at -20°C, stored under nitrogen,unstable in solution, ready to use.
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 1.3928 mL 6.964 mL 13.928 mL
5 mM 0.2786 mL 1.3928 mL 2.7856 mL
10 mM 0.1393 mL 0.6964 mL 1.3928 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

cAC10-vcMMAE, an anti-CD30-monomethyl auristatin E conjugate with potent and selective antitumor activity

Blood2003 Aug 15;102(4):1458-65.PMID: 12714494DOI: 10.1182/blood-2003-01-0039

The chimeric monoclonal antibody cAC10, directed against CD30, induces growth arrest of CD30+ cell lines in vitro and has pronounced antitumor activity in severe combined immunodeficiency (SCID) mouse xenograft models of Hodgkin disease. We have significantly enhanced these activities by conjugating to cAC10 the cytotoxic agent monomethyl auristatin E (MMAE) to create the antibody-drug conjugate cAC10-vcMMAE. MMAE, a derivative of the cytotoxic tubulin modifier auristatin E, was covalently coupled to cAC10 through a valine-citrulline peptide linker. The drug was stably attached to the antibody, showing only a 2% release of MMAE following 10-day incubation in human plasma, but it was readily cleaved by lysosomal proteases after receptor-mediated internalization. Release of MMAE into the cytosol induced G2/M-phase growth arrest and cell death through the induction of apoptosis. In vitro, cAC10-vcMMAE was highly potent and selective against CD30+ tumor lines (IC50 less than 10 ng/mL) but was more than 300-fold less active on antigen-negative cells. In SCID mouse xenograft models of anaplastic large cell lymphoma or Hodgkin disease, cAC10-vcMMAE was efficacious at doses as low as 1 mg/kg. Mice treated at 30 mg/kg cAC10-vcMMAE showed no signs of toxicity. These data indicate that cAC10-vcMMAE may be a highly effective and selective therapy for the treatment of CD30+ neoplasias.

Microtubule and tubulin binding and regulation of microtubule dynamics by the antibody drug conjugate (ADC) payload, monomethyl auristatin E (MMAE): Mechanistic insights into MMAE ADC peripheral neuropathy

Toxicol Appl Pharmacol2021 Jun 15;421:115534.PMID: 33852878DOI: 10.1016/j.taap.2021.115534

Monomethyl auristatin E (MMAE) is a potent anti-cancer microtubule-targeting agent (MTA) used as a payload in three approved MMAE-containing antibody drug conjugates (ADCs) and multiple ADCs in clinical development to treat different types of cancers. Unfortunately, MMAE-ADCs can induce peripheral neuropathy, a frequent adverse event leading to treatment dose reduction or discontinuation and subsequent clinical termination of many MMAE-ADCs. MMAE-ADC-induced peripheral neuropathy is attributed to non-specific uptake of the ADC in peripheral nerves and release of MMAE, disrupting microtubules (MTs) and causing neurodegeneration. However, molecular mechanisms underlying MMAE and MMAE-ADC effects on MTs remain unclear. Here, we characterized MMAE-tubulin/MT interactions in reconstituted in vitro soluble tubulin or MT systems and evaluated MMAE and vcMMAE-ADCs in cultured human MCF7 cells. MMAE bound to soluble tubulin heterodimers with a maximum stoichiometry of ~1:1, bound abundantly along the length of pre-assembled MTs and with high affinity at MT ends, introduced structural defects, suppressed MT dynamics, and reduced the kinetics and extent of MT assembly while promoting tubulin ring formation. In cells, MMAE and MMAE-ADC (via nonspecific uptake) suppressed proliferation, mitosis and MT dynamics, and disrupted the MT network. Comparing MMAE action to other MTAs supports the hypothesis that peripheral neuropathy severity is determined by the precise mechanism(s) of each individual drug-MT interaction (location of binding, affinity, effects on morphology and dynamics). This work demonstrates that MMAE binds extensively to tubulin and MTs and causes severe MT dysregulation, providing convincing evidence that MMAE-mediated inhibition of MT-dependent axonal transport leads to severe peripheral neuropathy.

Monomethyl Auristatin E Grafted-Liposomes to Target Prostate Tumor Cell Lines

Int J Mol Sci2021 Apr 15;22(8):4103.PMID: 33921088DOI: 10.3390/ijms22084103

Novel nanomedicines have been engineered to deliver molecules with therapeutic potentials, overcoming drawbacks such as poor solubility, toxicity or short half-life. Lipid-based carriers such as liposomes represent one of the most advanced classes of drug delivery systems. A Monomethyl Auristatin E (MMAE) warhead was grafted on a lipid derivative and integrated in fusogenic liposomes, following the model of antibody drug conjugates. By modulating the liposome composition, we designed a set of particles characterized by different membrane fluidities as a key parameter to obtain selective uptake from fibroblast or prostate tumor cells. Only the fluid liposomes made of palmitoyl-oleoyl-phosphatidylcholine and dioleoyl-phosphatidylethanolamine, integrating the MMAE-lipid derivative, showed an effect on prostate tumor PC-3 and LNCaP cell viability. On the other hand, they exhibited negligible effects on the fibroblast NIH-3T3 cells, which only interacted with rigid liposomes. Therefore, fluid liposomes grafted with MMAE represent an interesting example of drug carriers, as they can be easily engineered to promote liposome fusion with the target membrane and ensure drug selectivity.

Disitamab Vedotin: First Approval

Drugs2021 Nov;81(16):1929-1935.PMID: 34661865DOI: 10.1007/s40265-021-01614-x

Disitamab vedotin (Aidixi?) is an antibody-drug conjugate comprising a monoclonal antibody against human epidermal growth factor receptor 2 (HER2) conjugated via a cleavable linker to the cytotoxic agent monomethyl auristatin E. Disitamab vedotin is being developed by RemeGen for the treatment of solid tumours, including gastric cancer; Seagen has the right to develop disitamab vedotin globally outside of RemeGen's territory. In June 2021, disitamab vedotin received its first Biologics License Application (BLA) approval in China for the treatment of patients with HER2-overexpressing (defined as IHC2+ or 3+) locally advanced or metastatic gastric cancer (including gastroesophageal junction adenocarcinoma) who have received at least two systemic chemotherapy regimens. Disitamab vedotin as monotherapy or combination therapy is also in clinical development for the treatment of other solid tumours globally, including urothelial cancer in China and the USA, and biliary tract cancer, non-small cell lung cancer and HER2-positive and HER2-low expressing breast cancer in China. This article summarizes the milestones in the development of disitamab vedotin leading to this first approval for locally advanced or metastatic gastric cancer.

Tisotumab Vedotin: First Approval

Drugs2021 Dec;81(18):2141-2147.PMID: 34748188DOI: 10.1007/s40265-021-01633-8

Tisotumab vedotin (Tivdak?) is an antibody-drug conjugate comprising a fully human monoclonal antibody specific for tissue factor (TF-011) conjugated to monomethyl auristatin E (MMAE) that has been engineered to target tissue factor expressing tumours. Based on the results of a phase II trial, tisotumab vedotin has been granted accelerated approval in the USA for the treatment of adult patients with recurrent or metastatic cervical cancer with disease progression on or after chemotherapy. This article summarizes the milestones in the development of tisotumab vedotin leading to this first approval.