1
|
Pompili SVB, Fanzini S, Schachner M, Chen S. In Vitro and In Vivo Studies of Melanoma Cell Migration by Antagonistic Mimetics of Adhesion Molecule L1CAM. Int J Mol Sci 2024; 25:4811. [PMID: 38732030 PMCID: PMC11084881 DOI: 10.3390/ijms25094811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Melanoma, the deadliest type of skin cancer, has a high propensity to metastasize to other organs, including the brain, lymph nodes, lungs, and bones. While progress has been made in managing melanoma with targeted and immune therapies, many patients do not benefit from these current treatment modalities. Tumor cell migration is the initial step for invasion and metastasis. A better understanding of the molecular mechanisms underlying metastasis is crucial for developing therapeutic strategies for metastatic diseases, including melanoma. The cell adhesion molecule L1CAM (CD171, in short L1) is upregulated in many human cancers, enhancing tumor cell migration. Earlier studies showed that the small-molecule antagonistic mimetics of L1 suppress glioblastoma cell migration in vitro. This study aims to evaluate if L1 mimetic antagonists can inhibit melanoma cell migration in vitro and in vivo. We showed that two antagonistic mimetics of L1, anagrelide and 2-hydroxy-5-fluoropyrimidine (2H5F), reduced melanoma cell migration in vitro. In in vivo allograft studies, only 2H5F-treated female mice showed a decrease in tumor volume.
Collapse
Affiliation(s)
- Stefano Vito Boccadamo Pompili
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Sophia Fanzini
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA;
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- VA New Jersey Health System, East Orange, NJ 07018, USA
| |
Collapse
|
2
|
smProdrugs: A repository of small molecule prodrugs. Eur J Med Chem 2023; 249:115153. [PMID: 36724634 DOI: 10.1016/j.ejmech.2023.115153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
In modern drug discovery and development, the prodrug approach has become a crucial strategy for enhancing the pharmacokinetic profiles of drugs. A prodrug is a chemical compound, which gets metabolized into a pharmacologically active form (drug) inside the body after its administration. In the current work, we report 'smProdrugs' (http://cheminfolab.in/databases/prodrug/), which is one of the first exclusive databases on small molecule prodrugs. It stores the structures, physicochemical properties and experimental ADMET data manually curated from literature. SmProdrugs lists 626 small molecule prodrugs and their active compounds with the above mentioned experimental data from 1808 research articles and 61 patents have been stored. The information page of each record gives the structures and properties of the prodrug and the active drug side by side which makes it easy for the user to instantly compare them. The structural modifications in the prodrug/active drugs are highlighted in a different colour for easy comparison. Experimental data has been curated from the downloaded PubMed and patent articles and were catalogued in a tabular form with more than 25 fields under sub-sections i) name and structures of the prodrugs and their active compounds, ii) mode of activation of the prodrug and enzyme/biocatalyst involved in the conversion, iii) indications/disease, iv) pharmacological target, v) experimental pharmacokinetic properties such as solubility, absorption, volume of distribution, half-life, clearance etc. and vi) information on the purpose/gain from the prodrug strategies. Considering the ever expanding utility of the prodrug approach smProdrugs will be of great use to the scientific community working on rational design of small molecule prodrugs.
Collapse
|
3
|
Manevski N, King L, Pitt WR, Lecomte F, Toselli F. Metabolism by Aldehyde Oxidase: Drug Design and Complementary Approaches to Challenges in Drug Discovery. J Med Chem 2019; 62:10955-10994. [PMID: 31385704 DOI: 10.1021/acs.jmedchem.9b00875] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aldehyde oxidase (AO) catalyzes oxidations of azaheterocycles and aldehydes, amide hydrolysis, and diverse reductions. AO substrates are rare among marketed drugs, and many candidates failed due to poor pharmacokinetics, interspecies differences, and adverse effects. As most issues arise from complex and poorly understood AO biology, an effective solution is to stop or decrease AO metabolism. This perspective focuses on rational drug design approaches to modulate AO-mediated metabolism in drug discovery. AO biological aspects are also covered, as they are complementary to chemical design and important when selecting the experimental system for risk assessment. The authors' recommendation is an early consideration of AO-mediated metabolism supported by computational and in vitro experimental methods but not an automatic avoidance of AO structural flags, many of which are versatile and valuable building blocks. Preferably, consideration of AO-mediated metabolism should be part of the multiparametric drug optimization process, with the goal to improve overall drug-like properties.
Collapse
Affiliation(s)
- Nenad Manevski
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Lloyd King
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - William R Pitt
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Fabien Lecomte
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Francesca Toselli
- UCB BioPharma , Chemin du Foriest 1 , 1420 Braine-l'Alleud , Belgium
| |
Collapse
|
4
|
Inhibition of vertebrate aldehyde oxidase as a therapeutic treatment for cancer, obesity, aging and amyotrophic lateral sclerosis. Eur J Med Chem 2019; 187:111948. [PMID: 31877540 DOI: 10.1016/j.ejmech.2019.111948] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 10/25/2022]
Abstract
The aldehyde oxidases (AOXs) are a small sub-family of cytosolic molybdo-flavoenzymes, which are structurally conserved proteins and broadly distributed from plants to animals. AOXs play multiple roles in both physiological and pathological processes and AOX inhibition is of increasing significance in the development of novel drugs and therapeutic strategies. This review provides an overview of the evolution and the action mechanism of AOX and the role of each domain. The review provides an update of the polymorphisms in the human AOX. This review also summarises the physiology of AOX in different organs and its role in drug metabolism. The inhibition of AOX is a promising therapeutic treatment for cancer, obesity, aging and amyotrophic lateral sclerosis.
Collapse
|
5
|
Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180. [PMID: 31128989 DOI: 10.1016/j.pharmthera.2019.05.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/27/2019] [Indexed: 11/29/2022]
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme that belongs to the family of structurally related molybdoflavoproteins like xanthine oxidase (XO). The enzyme is characterized by broad substrate specificity and marked species differences. It catalyzes the oxidation of aromatic and aliphatic aldehydes and various heteroaromatic rings as well as reduction of several functional groups. The references to AO and its role in metabolism date back to the 1950s, but the importance of this enzyme in the metabolism of drugs has emerged in the past fifteen years. Several reviews on the role of AO in drug metabolism have been published in the past decade indicative of the growing interest in the enzyme and its influence in drug metabolism. Here, we present a comprehensive monograph of AO as a drug metabolizing enzyme with emphasis on marketed drugs as well as other xenobiotics, as substrates and inhibitors. Although the number of drugs that are primarily metabolized by AO are few, the impact of AO on drug development has been extensive. We also discuss the effect of AO on the systemic exposure and clearance these clinical candidates. The review provides a comprehensive analysis of drug discovery compounds involving AO with the focus on developmental candidates that were reported in the past five years with regards to pharmacokinetics and toxicity. While there is only one known report of AO-mediated clinically relevant drug-drug interaction (DDI), a detailed description of inhibitors and inducers of AO known to date has been presented here and the potential risks associated with DDI. The increasing recognition of the importance of AO has led to significant progress in predicting the site of AO-mediated metabolism using computational methods. Additionally, marked species difference in expression of AO makes it is difficult to predict human clearance with high confidence. The progress made towards developing in vivo, in vitro and in silico approaches for predicting AO metabolism and estimating human clearance of compounds that are metabolized by AO have also been discussed.
Collapse
Affiliation(s)
- Deepak Dalvie
- Drug Metabolism and Pharmacokinetics, Celgene Corporation, 10300, Campus Point Drive, San Diego, CA 92121, USA.
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, UK
| |
Collapse
|
6
|
Nenajdenko V. Fluorine-Containing Diazines in Medicinal Chemistry and Agrochemistry. FLUORINE IN HETEROCYCLIC CHEMISTRY VOLUME 2 2014. [PMCID: PMC7121506 DOI: 10.1007/978-3-319-04435-4_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The combination of a fluorine atom and a diazine ring, which both possess unique structural and chemical features, can generate new relevant building blocks for the discovery of efficient fluorinated biologically active agents. Herein we give a comprehensive review on the biological activity and synthesis of fluorine containing, pyrimidine, pyrazine and pyridazine derivatives with relevance to medicinal and agrochemistry.
Collapse
|
7
|
Garattini E, Terao M. The role of aldehyde oxidase in drug metabolism. Expert Opin Drug Metab Toxicol 2012; 8:487-503. [DOI: 10.1517/17425255.2012.663352] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
8
|
Sanoh S, Nozaki K, Murai H, Terashita S, Teramura T, Ohta S. Prediction of Human Metabolism of FK3453 by Aldehyde Oxidase Using Chimeric Mice Transplanted with Human or Rat Hepatocytes. Drug Metab Dispos 2011; 40:76-82. [DOI: 10.1124/dmd.111.041954] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
9
|
Garattini E, Terao M. Increasing recognition of the importance of aldehyde oxidase in drug development and discovery. Drug Metab Rev 2011; 43:374-86. [DOI: 10.3109/03602532.2011.560606] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
10
|
Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde Oxidase: An Enzyme of Emerging Importance in Drug Discovery. J Med Chem 2010; 53:8441-60. [DOI: 10.1021/jm100888d] [Citation(s) in RCA: 282] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- David C. Pryde
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ, England
| | - Deepak Dalvie
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, 10628 Science Center Drive, La Jolla, California 92121
| | - Qiyue Hu
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, 10628 Science Center Drive, La Jolla, California 92121
| | - Peter Jones
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ, England
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340
| | - Thien-Duc Tran
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ, England
| |
Collapse
|
11
|
Tayama Y, Miyake K, Sugihara K, Kitamura S, Kobayashi M, Morita S, Ohta S, Kihira K. Developmental Changes of Aldehyde Oxidase Activity in Young Japanese Children. Clin Pharmacol Ther 2007; 81:567-72. [PMID: 17375106 DOI: 10.1038/sj.clpt.6100078] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aldehyde oxidase (AO) plays an important role in metabolizing many drugs, so AO activity in individual patients may be a useful parameter for dose adjustment to avoid severe toxicity. In this study, we investigated the developmental changes of AO activity in 101 children. Urine was collected in the morning, and AO activity was assessed in terms of the ratio of pyridone formation from N(1)-methylnicotinamide, an AO substrate. Significant correlations were found between AO activity and various growth indices (age, body weight, body surface area, and liver volume). Age showed the moderate correlation (r(2)=0.506). AO activity rapidly increased with increase of the subjects' age up to about 1 year. These findings suggest that the AO activity begins to increase soon after birth. Because AO activity is immature in children below 1 year of age, dose adjustment based on individual AO activity should be made for such patients.
Collapse
Affiliation(s)
- Y Tayama
- Graduate School of Biomedical Sciences, Faculty of Pharmaceutical Science, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Beumer JH, Joseph E, Egorin MJ, Parker RS, D'argenio DZ, Covey JM, Eiseman JL. A mass balance and disposition study of the DNA methyltransferase inhibitor zebularine (NSC 309132) and three of its metabolites in mice. Clin Cancer Res 2006; 12:5826-33. [PMID: 17020990 DOI: 10.1158/1078-0432.ccr-06-1234] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To elucidate the in vivo metabolic fate of zebularine (NSC 309132), a DNA methyltransferase inhibitor proposed for clinical evaluation in the treatment of cancer. EXPERIMENTAL DESIGN Male, CD(2)F(1) mice were dosed i.v. with 100 mg/kg 2-[(14)C]zebularine. At specified times between 5 and 1,440 minutes, mice were euthanized. Plasma, organs, carcass, urine, and feces were collected and assayed for total radioactivity. Plasma and urine were also analyzed for zebularine and its metabolites with a previously validated high-pressure liquid chromatography assay. A similar experiment was done with 2-[(14)C]uridine, the proposed primary metabolite of zebularine. RESULTS Maximum plasma concentrations were 462, 306, 33.6, 21.7, and 11.5 mumol/L for total radioactivity, zebularine, uridine, uracil (each at 5 minutes), and dihydrouracil (at 15 minutes), respectively. Total radioactivity, zebularine, uridine, uracil, and dihydrouracil were rapidly eliminated from plasma, and after 45 minutes, none of the individual compounds could be quantitated by high-pressure liquid chromatography. Plasma data were consistent with sequential conversion of zebularine to uridine, uracil, and dihydrouracil. 2-Pyrimidinone was not observed. Prolonged retention of radioactivity, at concentrations higher than in plasma, was observed in tissues. Recovery of given radioactivity in urine (30.3% of dose), feces (0.4% of dose), cage wash (7.9% of dose), and tissues and carcass (6.1% of dose) after 24 hours implied that up to 55% of radioactivity was expired as (14)CO(2). Comparison of zebularine and uridine pharmacokinetic data indicated that approximately 40% of the zebularine dose was converted to uridine. CONCLUSIONS Zebularine is extensively and rapidly metabolized into endogenous compounds that are unlikely to have effects at the concentrations observed.
Collapse
Affiliation(s)
- Jan H Beumer
- Molecular Therapeutics/Drug Discovery Program, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
13
|
Kitamura S, Sugihara K, Ohta S. Drug-Metabolizing Ability of Molybdenum Hydroxylases. Drug Metab Pharmacokinet 2006; 21:83-98. [PMID: 16702728 DOI: 10.2133/dmpk.21.83] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Molybdenum hydroxylases, which include aldehyde oxidase and xanthine oxidoreductase, are involved in the metabolism of some medicines in humans. They exhibit oxidase activity towards various heterocyclic compounds and aldehydes. The liver cytosol of various mammals also exhibits a significant reductase activity toward nitro, sulfoxide, N-oxide and other moieties, catalyzed by aldehyde oxidase. There is considerable variability of aldehyde oxidase activity in liver cytosol of mammals: humans show the highest activity, rats and mice show low activity, and dogs have no detectable activity. On the other hand, xanthine oxidoreductase activity is present widely among species. Interindividual variation of aldehyde oxidase activity is present in humans. Drug-drug interactions associated with aldehyde oxidase and xanthine oxidoreductase are of potential clinical significance. Drug metabolizing ability of molybdenum hydroxylases and the variation of the activity are described in this review.
Collapse
Affiliation(s)
- Shigeyuki Kitamura
- Graduate School of Biomedical Sciences, Hiroshima University, Kasumi, Japan.
| | | | | |
Collapse
|
14
|
Klecker RW, Cysyk RL, Collins JM. Zebularine metabolism by aldehyde oxidase in hepatic cytosol from humans, monkeys, dogs, rats, and mice: Influence of sex and inhibitors. Bioorg Med Chem 2006; 14:62-6. [PMID: 16143537 DOI: 10.1016/j.bmc.2005.07.053] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 07/27/2005] [Accepted: 07/27/2005] [Indexed: 11/15/2022]
Abstract
To aid in the clinical evaluation of zebularine, a potential oral antitumor agent, we initiated studies on the metabolism of zebularine in liver cytosol from humans and other mammals. Metabolism by aldehyde oxidase (AO, EC 1.2.3.1) was the major catabolic route, yielding uridine as the primary metabolite, which was metabolized further to uracil by uridine phosphorylase. The inhibition of zebularine metabolism was studied using raloxifene, a known potent inhibitor of AO, and 5-benzylacyclouridine (BAU), a previously undescribed inhibitor of AO. The Michaelis-Menten kinetics of aldehyde oxidase and its inhibition by raloxifene and BAU were highly variable between species.
Collapse
Affiliation(s)
- Raymond W Klecker
- Laboratory of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| | | | | |
Collapse
|
15
|
Rooseboom M, Commandeur JNM, Vermeulen NPE. Enzyme-catalyzed activation of anticancer prodrugs. Pharmacol Rev 2004; 56:53-102. [PMID: 15001663 DOI: 10.1124/pr.56.1.3] [Citation(s) in RCA: 370] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The rationale fo the development of prodrugs relies upon delivery of higher concentrations of a drug to target cells compared to administration of the drug itself. In the last decades, numerous prodrugs that are enzymatically activated into anti-cancer agents have been developed. This review describes the most important enzymes involved in prodrug activation notably with respect to tissue distribution, up-regulation in tumor cells and turnover rates. The following endogenous enzymes are discussed: aldehyde oxidase, amino acid oxidase, cytochrome P450 reductase, DT-diaphorase, cytochrome P450, tyrosinase, thymidylate synthase, thymidine phosphorylase, glutathione S-transferase, deoxycytidine kinase, carboxylesterase, alkaline phosphatase, beta-glucuronidase and cysteine conjugate beta-lyase. In relation to each of these enzymes, several prodrugs are discussed regarding organ- or tumor-selective activation of clinically relevant prodrugs of 5-fluorouracil, axazaphosphorines (cyclophosphamide, ifosfamide, and trofosfamide), paclitaxel, etoposide, anthracyclines (doxorubicin, daunorubicin, epirubicin), mercaptopurine, thioguanine, cisplatin, melphalan, and other important prodrugs such as menadione, mitomycin C, tirapazamine, 5-(aziridin-1-yl)-2,4-dinitrobenzamide, ganciclovir, irinotecan, dacarbazine, and amifostine. In addition to endogenous enzymes, a number of nonendogenous enzymes, used in antibody-, gene-, and virus-directed enzyme prodrug therapies, are described. It is concluded that the development of prodrugs has been relatively successful; however, all prodrugs lack a complete selectivity. Therefore, more work is needed to explore the differences between tumor and nontumor cells and to develop optimal substrates in terms of substrate affinity and enzyme turnover rates fo prodrug-activating enzymes resulting in more rapid and selective cleavage of the prodrug inside the tumor cells.
Collapse
Affiliation(s)
- Martijn Rooseboom
- Leiden/Amsterdam Center for Drug Research (L.A.C.D.R.), Division of Molecular Toxicology, Department of Pharmacochemistry, Vrije Universiteit Amsterdam, De Boelelaan 1083, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
16
|
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme expressed predominantly in the liver. AO is involved in the metabolism of many xenobiotics of pharmacological and toxicological importance including antivirals (famciclovir), antimalarials (quinine) and anticancer drugs (5-fluoro-2-pyrimidine and methotrexate). The aim of this study was to characterize AO activity in different strains of mice using two different substrates. AO activity in the cytosolic fraction was characterized using the metabolism of N-[2-(dimethylamino)ethyl]acridine-4-carboxamide (DACA), a novel antitumor drug, to form DACA-9(10H)-acridone (quantified by HPLC with fluorescence detection) and benzaldehyde to form benzoic acid (quantified spectrophotometrically). Characterization of mouse AO activity with DACA showed 15-fold variation in K(m), 10-fold variation in apparent V(max) and twofold differences in intrinsic clearance. Nude mice and C129/C57 had the highest intrinsic clearance (0.66 and 0.l53 ml/min per mg protein, respectively). Nude mice cleared DACA faster than nude tumor bearing mice by a factor of 2. Male Swiss CD had higher intrinsic clearance than female Swiss CD (0.36 and 0.28 ml/min per mg protein). A similar pattern of enzyme activity was observed with benzaldehyde; however, the extent of variation was less than that found with DACA. In conclusion, our results show that there are both strain and gender differences in AO activity. These differences are better detected by DACA. Furthermore, these results suggest caution when extrapolating the data obtained from mouse AO studies to humans.
Collapse
Affiliation(s)
- H S Al-Salmy
- Department of Pharmacology and Therapeutics, College of Medicine, Sultan Qaboos University, Al-Khod, P.O. Box 35, Muscat-123, Sultanate of Oman.
| |
Collapse
|
17
|
LoRusso PM, Prakash S, Wozniak A, Flaherty L, Zalupski M, Shields A, Sands H, Parchment R, Jasti B. Phase I clinical trial of 5-fluoro-pyrimidinone (5FP), an oral prodrug of 5-fluorouracil (5FU). Invest New Drugs 2002; 20:63-71. [PMID: 12003195 DOI: 10.1023/a:1014430216434] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE 5-Fluoro-Pyrimidinone (5FP) is an oral pro-drug of 5-Fluorouracil (5FU), and is converted to 5FU by hepatic aldehyde oxidase. Preclinically, 5FP demonstrated anti-tumor activity against colon 38 and P 388 leukemia models in mice. Using an accelerated titration trial design with one patient cohorts and initial 100% escalations, a Phase I trial was conducted to determine the maximum tolerated dose (MTD) of 5FP and describe its toxicity and pharmacokinetic profile. PATIENTS AND METHODS 5FP was administered orally once daily for 5 days every 4 weeks. The initial dose level was 23 mg/m2/d. Using single patient cohorts, escalation proceeded according to accelerated titration 4B design, initially by 100% and subsequently 30-35% escalations (exact escalation determined by pill size) until dose limiting toxicity was observed. A total of 19 patients were enrolled with a median age of 56 years and median performance status of 1. Most patients were heavily pre-treated with chemotherapy, radiation therapy, or both, and patient population included a wide variety of tumor types. RESULTS Dose escalation proceeded rapidly to 1715 mg/m2/d with the only toxicities observed being nausea and vomiting. The large number of pills necessary at that point required a formulation change, which resulted in appreciable hematologic toxicity. This led to rapid de-escalation of dose in subsequent patients, with the MTD finally being determined to be 625 mg/m2/d. The DLTs observed were grade 4 neutropenia for greater than 5 days and grade 3 anemia. Other toxicities included nausea, vomiting, fatigue, constipation and mucositis. Pharmacology studies confirmed that SFP was converted to 5FU in humans at all dose levels. However, the extent of conversion decreased over the five daily treatments, but returned for the subsequent cycle. The hematologic toxicity was not related to 5FU exposure per course. CONCLUSION 5FP is a tolerable oral outpatient therapy. Accelerated titration was an efficient way of conducting this phase I trial. The recommended phase 2 dose is 625 mg/m2/d orally for 5 days every 28 days.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Karmanos Cancer Institute at Wayne State University School of Medicine, Department of Internal Medicine, Detroit, MI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Affiliation(s)
- P J O'Dwyer
- Thomas Jefferson University, Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | | |
Collapse
|
19
|
Abstract
In addition to cytochrome P450, oxidation of drugs and other xenobiotics can also be mediated by non-P450 enzymes, the most significant of which are flavin monooxygenase, monoamine oxidase, alcohol dehydrogenase, aldehyde dehydrogenase, aldehyde oxidase and xanthine oxidase. This article highlights the importance of these non-P450 enzymes in drug metabolism. A brief introduction to each of the non-P450 oxidizing enzymes is given in this review and the oxidative reactions have been illustrated with clinical examples. Drug oxidation catalyzed by enzymes such as flavin monooxygenase and monoamine oxidase may often produce the same metablolites as those generated by P450 adn thus drug interactions may be difficult to predict without a clear knowledge of the underlying enzymology. In contrast, oxidation via aldehyde oxidase and xanthine oxidase gives different metabolites to those resulting from P450 hydroxylation. Although oxidation catalyzed by non-P450 enzymes can lead to drug inactivation, oxidation may be essential for the generation of active metabolite(s). The activation of a number of prodrugs by non-P450 enzymes is thus described. It is concluded that there is still much to learn about factors affecting the non-P450 enzymes in the clinical situation.
Collapse
Affiliation(s)
- C Beedham
- Pharmaceutical Chemistry, School of Pharmacy, University of Bradford, West Yorkshire, UK
| |
Collapse
|
20
|
Yoshihara S, Tatsumi K. Involvement of growth hormone as a regulating factor in sex differences of mouse hepatic aldehyde oxidase. Biochem Pharmacol 1997; 53:1099-105. [PMID: 9175714 DOI: 10.1016/s0006-2952(97)00088-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The participation of circulating growth hormone (GH) as a regulator of sex differences in hepatic aldehyde oxidase (AO) activity in ddy mice was examined. The 2- to 3-fold higher activities in adult male mice compared with adult female mice were decreased to the female levels by neonatal pretreatment with monosodium glutamate (MSG) or monosodium aspartate (MSA), either of which is known to reduce circulating GH levels. A decline of the activities in the MSG-treated male mice was restored nearly to the male control levels by subsequent injections of human GH every 12 hr for 7 days. These changes in AO activities in male mice caused by the excitotoxic amino acids were not observed in females. Hypophysectomy markedly decreased hepatic AO activities in male mice and partially in female mice. The activities in hypophysectomized male mice were restored again to levels similar to the control males by intermittent injections of human GH. Administration of testosterone propionate (TP) significantly increased the activities of hepatic AO in intact female mice, but not in MSA-treated or hypophysectomized females. On the other hand, the AO activities in adult male mice were decreased partially by the administration of estradiol benzoate. These results indicate that the pituitary GH is involved as one of the major regulatory factors of sex differences in the activities of hepatic AO in mice and TP also contributes to maintaining the higher activity in male mice mainly through the hypothalamus-pituitary system.
Collapse
Affiliation(s)
- S Yoshihara
- Institute of Pharmaceutical Sciences, Hiroshima University School of Medicine, Minami-ku, Japan.
| | | |
Collapse
|
21
|
|