Home>>Signaling Pathways>> Immunology/Inflammation>> Autoimmunity>>Carboxyphosphamide

Carboxyphosphamide Sale

(Synonyms: CPCOOH, NSC 145124) 目录号 : GC49147

An inactive metabolite of cyclophosphamide

Carboxyphosphamide Chemical Structure

Cas No.:22788-18-7

规格 价格 库存 购买数量
1 mg
¥599.00
现货
5 mg
¥2,552.00
现货
10 mg
¥4,797.00
现货
25 mg
¥11,238.00
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

Carboxyphosphamide is an inactive metabolite of the alkylating agent cyclophosphamide .1 It is formed from cyclophosphamide via oxidation of the intermediate metabolite aldophosphamide by aldehyde dehydrogenase.

1.Manthey, C.L., and Sladek, N.E.Kinetic characterization of the catalysis of "activated" cyclophosphamide (4-hydroxycyclophosphamide/aldophosphamide) oxidation to carboxyphosphamide by mouse hepatic aldehyde dehydrogenasesBiochem. Pharmacol.37(14)2781-2790(1988)

Chemical Properties

Cas No. 22788-18-7 SDF
别名 CPCOOH, NSC 145124
Canonical SMILES O=C(CCOP(N)(N(CCCl)CCCl)=O)O
分子式 C7H15Cl2N2O4P 分子量 293.1
溶解度 Acetonitrile: soluble,DMSO: soluble,Methanol: soluble 储存条件 -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。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 3.4118 mL 17.059 mL 34.118 mL
5 mM 0.6824 mL 3.4118 mL 6.8236 mL
10 mM 0.3412 mL 1.7059 mL 3.4118 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

Individual variation in the activation and inactivation of metabolic pathways of cyclophosphamide

J Natl Cancer Inst 1992 Nov 18;84(22):1744-8.PMID:1433359DOI:10.1093/jnci/84.22.1744.

Background: Carboxyphosphamide is an inactive metabolite of cyclophosphamide, which is a widely used antineoplastic drug. Deficiencies in the production of this metabolite have been reported. Such deficiencies would have important consequences for therapeutic and toxic effects of oxazaphosphorines like cyclophosphamide. Purpose: This study further investigates the variability in cyclophosphamide metabolism and Carboxyphosphamide recovery in urine. Methods: The 24-hour urinary metabolic profile of cyclophosphamide was investigated in 17 Turkish patients receiving doses of 100-1080 mg orally or by short intravenous infusion. Urine samples were assayed quantitatively for cyclophosphamide and its principal metabolites (phosphoramide mustard, 4-ketocyclophosphamide, Carboxyphosphamide, and dechloroethylcyclophosphamide) with combined thin-layer chromatography-photography-densitometry. The amount of each metabolite excreted in 24 hours was expressed as a percentage of the dose. Results: Recovery of drug and metabolites varied greatly among individuals (range, 0.01%-13.56% of dose). In particular, the amount of Carboxyphosphamide varied over a thousandfold range and was undetectable in urine from four patients. The patients were classified by phenotype as demonstrating low or high carboxylation. Those with low carboxylation excreted less than 0.2% of the cyclophosphamide dose as Carboxyphosphamide, while those with high carboxylation excreted 0.8%-13.6% (median, 1.81%). No association was observed between carboxylation phenotype and patient age, sex, disease, or concomitant therapy, although the three lifetime nonsmokers all showed poor carboxylation. No correlation was observed between the percent of dose excreted as any of the other metabolites and that excreted as Carboxyphosphamide. There was a statistically significant inverse correlation between the combined recovery of Carboxyphosphamide and phosphoramide mustard and the dose of prednisolone administered. Conclusions: These data confirm an earlier observation of a phenotypic deficiency of Carboxyphosphamide excretion in British patients treated with cyclophosphamide. This deficiency may arise from a polymorphism in the enzyme aldehyde dehydrogenase. Carboxylation phenotype may have important implications for both the therapeutic effect and toxicity of cyclophosphamide.

Kinetic characterization of the catalysis of "activated" cyclophosphamide (4-hydroxycyclophosphamide/aldophosphamide) oxidation to Carboxyphosphamide by mouse hepatic aldehyde dehydrogenases

Biochem Pharmacol 1988 Jul 15;37(14):2781-90.PMID:3395357DOI:10.1016/0006-2952(88)90041-x.

A spectrophotometric assay was developed and utilized to directly characterize aldehyde dehydrogenase-catalyzed oxidation of aldophosphamide to Carboxyphosphamide by soluble and solubilized particulate fractions prepared from mouse liver homogenates. Vmax values of 3310 and 1170 nmol/min/g liver were obtained for the soluble and solubilized particulate fractions respectively. Km values were 22 and 84 microM respectively. Alkaline pH optimums were observed in each case. Aldehyde dehydrogenase-catalyzed oxidation of aldophosphamide by the soluble fraction was markedly more temperature responsive. Catalysis of aldophosphamide and acetaldehyde or benzaldehyde oxidation was apparently by the same isozyme(s) in the soluble fraction. Similarly, low Km (acetaldehyde/benzaldehyde) and high Km (acetaldehyde/benzaldehyde) isozymes each apparently catalyzed the oxidation of aldophosphamide in the solubilized particulate fraction. Our findings suggest that (1) oxidation of aldophosphamide to Carboxyphosphamide by mouse liver is catalyzed largely by the predominant aldehyde dehydrogenase isozyme present in the soluble fraction (cytosol) of this tissue, and (2) isozymes that catalyze aldophosphamide oxidation are not different from those that catalyze the oxidation of acetaldehyde and benzaldehyde, though the relative contribution of each isozyme within the solubilized particulate fraction to the catalysis of aldophosphamide oxidation remains to be determined.

Phenotypically deficient urinary elimination of Carboxyphosphamide after cyclophosphamide administration to cancer patients

Cancer Res 1988 Sep 15;48(18):5167-71.PMID:3409242doi

The 0-24-h urinary metabolic profile of cyclophosphamide was investigated in a series of 14 patients with various malignancies receiving combination chemotherapy including i.v. cyclophosphamide. This was accomplished using combined thin-layer chromatography-photography-densitometry, which can quantitate cyclophosphamide and its four principal urinary metabolites (4-ketocyclophosphamide, nor-nitrogen mustard, Carboxyphosphamide, and phosphoramide mustard). Recovery of drug-related metabolites was 36.5 +/- 17.8% (SD) dose, the most abundant metabolites being phosphoramide mustard (18.5 +/- 16.1% dose) and unchanged cyclophosphamide (12.7 +/- 9.3% dose). The most variable metabolite was Carboxyphosphamide, with five patients excreting 0.3% dose or less. These patients were termed low carboxylators (LC) and could be distinguished from high carboxylators (HC) by a carboxylation index (relative percentage as Carboxyphosphamide multiplied by 10). Mean carboxylation indices for the LC and HC phenotypes were 3.4 +/- 2.6 and 151 +/- 115, respectively. There were no associations between patient age, sex, body weight, tumor type, or concomitant drug therapy and carboxylation phenotype. Neither 4-ketocyclophosphamide nor nor-nitrogen mustard excretion differed between LC and HC phenotypes; however, HC patients had a greater excretion of cyclophosphamide (46.4 +/- 15.5 relative percentage) than LC patients (19.4 +/- 12.6%). The DNA cross-linking cytotoxic metabolite phosphoramide mustard was elevated more than 2-fold in the LC (76.5 +/- 13.9%) compared with the HC (33.0 +/- 12.2%) phenotype. It is concluded that these data represent the first evidence of a defect in cyclophosphamide metabolism, and it is proposed that this arises from a hitherto unrecognized aldehyde dehydrogenase genotype.

Cyclophosphamide metabolism in children

Cancer Res 1995 Feb 15;55(4):803-9.PMID:7850793doi

The alkylating agent cyclophosphamide is a prodrug which is metabolized in vivo to produce both therapeutic and toxic effects. Cyclophosphamide metabolism was investigated in 36 children with various malignancies. Concentrations of cyclophosphamide and its principal metabolites were measured in plasma and urine using a quantitative high-performance TLC method. The results indicated a high degree of inter-patient variation in metabolism. In contrast to previous adult studies on urinary metabolites, plasma Carboxyphosphamide concentrations did not support the existence of polymorphic metabolism. Plasma concentrations of dechlorethylcyclophosphamide and Carboxyphosphamide were correlated in individual patients, suggesting that the activity of both aldehyde dehydrogenase and cytochrome P450 enzyme(s) determine Carboxyphosphamide production in vivo. The presence of ketocyclophosphamide in plasma was strongly associated with dexamethasone pretreatment and was also accompanied by a high clearance of the parent drug. Interpatient differences in metabolism reflect individual levels of enzyme expression and may contribute to variation in clinical effect.

The effect of cimetidine on cyclophosphamide metabolism in rabbits

Cancer Chemother Pharmacol 1990;27(2):125-30.PMID:2249327DOI:10.1007/BF00689096.

Six female rabbits were given 20 mg/kg cyclophosphamide (containing 100 microCi [3H-chloroethyl]-cyclophosphamide) alone or 1 h following 100 mg/kg cimetidine. Serial plasma and urine specimens were collected and levels of cyclophosphamide and its metabolites (4-hydroxycyclophosphamide, 4-ketocyclophosphamide, phosphoramide mustard, and Carboxyphosphamide) were measured. 4-Ketocyclophosphamide was the major metabolite present in rabbit plasma and urine, with lesser amounts of 4-hydroxycyclophosphamide, Carboxyphosphamide, and phosphoramide mustard also being identified. Cimetidine pretreatment resulted in prolongation of cyclophosphamide's half-life from 24.3 +/- 7.3 to 33.5 +/- 9.5 min (mean +/- SD; P = 0.036) but did not significantly alter the AUC0-8 h for the latter drug. Cimetidine pretreatment resulted in a significantly greater AUC0-8 h for 4-hydroxycyclophosphamide (189.4 +/- 77 vs 364.6 +/- 126.7 mumol min/l-1; P = 0.016) as compared with control values. A higher AUC0-8 h value for phosphoramide mustard (53.7 +/- 69.2 vs 95.7 +/- 34.7 mumol min/l-1) was also observed after cimetidine dosing but the difference was not significant (P = 0.21). Kinetics of 4-ketocyclophosphamide and Carboxyphosphamide were not significantly affected by cimetidine treatment. Cimetidine was added to hepatic microsomes isolated from phenobarbital-treated rabbits; it did not inhibit cyclophosphamide's metabolism in vitro, suggesting that its in vivo effect may be mediated through mechanisms other than cytochrome P-450 inhibition. Cimetidine pretreatment increases exposure to cyclophosphamide and its major activated metabolite, 4-hydroxycyclophosphamide. Potentiation rather than inhibition of cyclophosphamide's pharmacodynamic effect is to be predicted when cimetidine is given concomitantly with the former. Alterations in hepatic blood flow or mechanisms other than microsomal inhibition by cimetidine may explain this potentiation.