WO1997039352A1 - Assays for detection of purine metabolites - Google Patents

Assays for detection of purine metabolites Download PDF

Info

Publication number
WO1997039352A1
WO1997039352A1 PCT/US1997/002839 US9702839W WO9739352A1 WO 1997039352 A1 WO1997039352 A1 WO 1997039352A1 US 9702839 W US9702839 W US 9702839W WO 9739352 A1 WO9739352 A1 WO 9739352A1
Authority
WO
WIPO (PCT)
Prior art keywords
sample
uric acid
hydrogen peroxide
purine
reagent
Prior art date
Application number
PCT/US1997/002839
Other languages
French (fr)
Inventor
James L. Sherley
Stephen S. Ortiz
Original Assignee
Fox Chase Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fox Chase Cancer Center filed Critical Fox Chase Cancer Center
Priority to AU19708/97A priority Critical patent/AU1970897A/en
Publication of WO1997039352A1 publication Critical patent/WO1997039352A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/62Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving uric acid

Definitions

  • This invention generally relates to detection and quantitation of selected analytes within a mixed population of analytes. Specifically, simple assays are provided to detect and measure purine bases and/or purine nucleotides in a biological sample.
  • the concentration of a light-absorbing (i.e., colored) substance in solution can be estimated by comparing the intensity of its color with that of several standard solutions of known ' concentration.
  • Such methods using a spectrophotometer as a detector, are known as colorimetric methods; in various forms, they have played a major role historically in analytical chemistry.
  • Colorimetric assays have been developed to analyze levels of various components in biological or chemical solutions.
  • light-emitting (i.e., fluorescent) substances can be detected and quantified using appropriate detection devices, e.g., a fluorometer.
  • Antimetabolites that interfere with de novo purine synthesis constitute an important part of the arsenal of drugs available for the treatment of leukemia and other cancers. However, not all patients respond equally well to these drugs, and some eventually become refractory to therapy. Resistance to purine analogs has been attributed to the deficiency of enzymes necessary to convert them to their nucleotide forms. Other mechanisms have been observed in experimental tumors, such as increased degradation of drugs and inability of resistant cells to convert the ribonucleotide analogues to the deoxyribonucleotide analogue. Increased plasma concentrations of hypoxanthine during treatment with these drugs may also be a mechanism of resistance to antimetabolites acting in the purine biosynthetic pathways.
  • Extracellular hypoxanthine is important for the activity of purine antimetabolites in two aspects: it can be utilized to restore purine nucleotide pools in cells, the de novo synthesis of which has been blocked, and it potentially may compete with some of these antimetabolites for transport into the cell or for the enzymes necessary to convert the drugs to their active nucleotides.
  • the present invention provides simple assays for determining the presence or quantity of selected purine metabolites in a liquid sample.
  • a method is provided for determining the presence or quantity of a selected subpopulation of analytes in a mixed population of analytes, wherein the analyte subpopulation comprises purine metabolites capable of being further metabolized to uric acid.
  • the method comprises providing a liquid sample of the mixed analyte population, adding to that sample at least one reagent that catalyzes the conversion of the purine metabolites to uric acid (under conditions whereby the purine metabolites in the subpopulation are converted to uric acid) , adding to the sample at least one reagent that reacts with uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide (and providing conditions whereby the hydrogen peroxide is produced in the sample) and quantitatively detecting the hydrogen peroxide in the sample.
  • the quantity of hydrogen peroxide produced by the method is proportional to the quantity of purine metabolite analyte subpopulation in the sample.
  • the mixed analyte population is disposed within a biological fluid or tissue and a liquid sample is prepared from that biological fluid or tissue.
  • the analyte subpopulation preferably comprises purine bases (such as adenine, xanthine and hypoxanthine) or purine nucleosides (such as inosine, xanthosine and adenosine) .
  • Reagents that catalyze the conversion of purine bases to uric acid preferably include the enzyme xanthine oxidase; reagents for catalyzing the conversion of purine nucleosides to uric acid preferably comprise a combination of the enzymes purine nucleoside phosphorylase and xanthine oxidase.
  • the uric acid in the sample is exposed to the enzyme, uricase, producing as a byproduct a stoichiometrically equivalent amount of a hydrogen peroxide.
  • the hydrogen peroxide is thereafter detected, preferably by optical means.
  • Detection is accomplished by exposing the hydrogen peroxide in the sample substrates and other reactant which, upon reaction with hydrogen peroxide form the optically detectable product is preferably detected spectrophotometrically or spectrofluorometrically.
  • a method for determining the presence or quantity of purine bases in a biological fluid or tissue. The method comprises providing a liquid test sample of the biological fluid or tissue and a volumetrically equivalent liquid control sample. To the test sample is added an amount of xanthine oxidase effective to catalyze the conversion of the purine bases to uric acid. The sample is placed under conditions whereby the purine bases are converted to uric acid.
  • a volume of a non-reactive liquid medium such as water or buffer
  • a volume of xanthine oxidase added to the test sample is subjected to the same conditions as the test sample.
  • To each of the test sample and control sample is added at least one reagent that reacts with uric acid in the samples to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide.
  • the sample is subjected to conditions whereby the reaction to form the hydrogen peroxide occurs. Thereafter, the hydrogen peroxide is quantitatively detected in each of the test sample and the control sample, the quantity of hydrogen peroxide is proportional to the quantity of the uric acid in each sample.
  • a method for determining the presence or quantity of purine nucleosides in a biological fluid or tissue comprises providing volumetrically equivalent liquid test sample and control sample of the biological fluid or tissue.
  • an amount of nucleoside phosphorylase and xanthine oxidase effective to catalyze the conversion of the purine nucleosides to uric acid.
  • To the control sample is added a volume of a non-reactive liquid medium equivalent to the volume of nucleoside phosphorylase added to the test sample.
  • the control sample is also given an equivalent amount of the xanthine oxidase as added to the test sample.
  • the respective samples are subjected to conditions whereby, in the test sample the purine nucleoside are converted to uric acid.
  • To each of the test sample and the control sample is added at least one reagent that reacts with the uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide.
  • the hydrogen peroxide in each sample is quantitatively detected, that quantity being proportional to the quantity of uric acid present in each sample.
  • the difference in quantity of uric acid in the control sample versus the test sample is compared, that difference being proportional to the quantity of purine nucleosides in the biological fluid or tissue.
  • a test kit for determining the presence or quantity of a subpopulation of analytes comprising purine metabolites capable of being further metabolized to uric acid, in a mixed population of analytes.
  • the test kit comprises at least one reagent that catalyzes the conversion of the purine metabolites to uric acid, and at least one reagent that reacts with the uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide.
  • the test kit further comprises at least one reagent for detecting the hydrogen peroxide.
  • test kits of the present invention provide a rapid, inexpensive and sensitive system for determining the presence or amount of purine metabolite in a biological sample.
  • test kit provide an inexpensive alternative to sophisticated, instrument-dependent assays presently available, and can be performed in the clinic or in the field by unskilled technicians.
  • the present invention is directed to a simple method for determining the presence or quantity of a selected subpopulation of purine metabolites in a liquid sample, preferably obtained from a biological fluid or tissue.
  • the method is designed to measure purine bases and purine nucleosides, either together or separately.
  • the assays of the invention are based on the known enzymatic activity of xanthine oxidase and purine nucleoside phosphorylase, and relies on converting the substrates of these enzymes (e.g., the purine nucleotides, inosine, xanthosine and adenosine, and purine bases xanthine, hypoxanthine and adenine) to uric acid.
  • the uric acid may be quantitatively detected in a variety of ways.
  • the uric acid is exposed to the enzyme, uricase, under oxidative conditions, which results in the formation of a stoichiometrically equivalent amount of hydrogen peroxide (H 2 0 2 ) as a reaction byproduct.
  • the ultimate step in the method of the present invention is based upon the detection of the byproduct, hydrogen peroxide.
  • Hydrogen peroxide levels may be quantitated using several different procedures.
  • a commercially available uric acid reagent kit (Sigma Chemical Co., St. Louis, MO)may be utilized. Purine metabolites initially present in a liquid sample are acted on by xanthine oxidase or purine phosphorylase. The resulting reaction product is uric acid which is then exposed to enzymes (uricase peroxidase) and other reagents present in the uric acid kit, which results in the generation of hydrogen peroxide. Hydrogen peroxide can be detected and quantitated many ways, as described in greater detail below.
  • H 2 0 2 is detected via its reaction with two substrates to form a quinoneimine dye that absorbs light at 520 nm (see Green and Hill (1984) Meth. Enz. 105: 3-22).
  • the method of this invention exemplified by the determination of hypoxanthine and xanthosine, comprises in its most preferred form: incubating a liquid sample, i.e., plasma, tissue extract or urine, with enzymes selected from the group of xanthine oxidase and purine nucleoside phosphorylase or a combination thereof; adding the above components to a tube containing uric acid reagent (Sigma) ; and reading the absorbance values of the resulting reaction product in a spectrophotometer set at 520 nm. The presence of xanthine, xanthosine, inosine, hypoxanthine, adenine or adenosine are then determined.
  • a liquid sample i.e., plasma, tissue extract or urine
  • enzymes selected from the group of xanthine oxidase and purine nucleoside phosphorylase or a combination thereof
  • a tube containing uric acid reagent Sigma
  • Another method for detecting H 2 ⁇ 2 is based upon the hydrolysis of a stable reagent, diacetyl- dichlorofluoroscin (LDADCF) , by sodium hydroxide to a less stable nonfluorescent compound dichlorofluoroscin (LDCF) , which is oxidized by hydrogen peroxide to the fluorescent dichlorofluorescin (DCF) .
  • LDADCF diacetyl- dichlorofluoroscin
  • DCF fluorescent dichlorofluorescin
  • Scopoletin may also be used to detect H 2 0 2 .
  • the method is based on quantitation of decreases in scopoletin fluorescence following its oxidation by H 2 0 2 and horseradish peroxidase (Clifford et al., (1984), Meth. Enz. JL0_5:393-398) . More specifically, when activated by light at 350 nm, scopoletin fluoresces with a peak at 460 nm. When oxidized by H 2 0 2 and horseradish peroxidase, scopoletin loses its fluorescence in direct proportion to H 2 0 2 concentration.
  • the 605 nm absorbance of this solution immediately decreases in response to the reducing action of ascorbate and/or other reductants present in the sample.
  • the extent of reoxidation of the solution upon the addition of peroxidase can be quantitatively related to the amount of H 2 0 2 in the sample. Due to the observation that reduced dichlorophenol-indophenol spontaneously reoxidizes at a rate of 0.03 nmol min '1 ⁇ M "1 with generation of H 2 0 2 , this is not a particularly preferred method as it has the potential to overestimate the H 2 0 2 content in the sample (Garcia-Castineiras et al., Exp. Eye Res. 55:9-
  • the invention provides a kit of reagents for carrying out the assays of the invention.
  • the kit contains reagents for converting prime nucleosides and/or bases to uric acid, as well as reagents for producing H 2 0 2 from uric acid and, optionally, reagents for detecting the hydrogen peroxide.
  • a kit according to the present invention may comprise reaction vials, preferably cuvettes, lyophilized enzymes (e.g., xanthine oxidase, nucleoside phosphorylase) , calibrated reference solutions, an appropriate diluent, and the uric acid reagent (Sigma) or an appropriate substitute (e.g., one or more of the individual components of the reagent obtained from Sigma or other suppliers, or modifications of the reagent, such as described by James and Price (1984) Ann. Clin. Biochem. 21' 405-410.
  • lyophilized enzymes e.g., xanthine oxidase, nucleoside phosphorylase
  • calibrated reference solutions e.g., calibrated reference solutions
  • an appropriate diluent e.g., calibrated reference solutions
  • an appropriate diluent e.g., calibrated reference solutions
  • an appropriate diluent e.g., calibrated reference solutions
  • Examples 1 and 2 describe assays for detecting purine bases and nucleosides.
  • Example 3 shows typical results of the assays.
  • Example 4 describes use of the assays to detect altered plasma hypoxanthine levels in cancer patients.
  • the degradation of purines to the end product uric acid occurs via the action of deaminase enzymes that first convert adenine and guanine to hypoxanthine and xanthine, respectively. Deamination may also occur non- enzymatically. This is a more frequent occurrence for adenine because, unlike guanine, the amino group in adenine is attached to a 6-membered aromatic ring. Hypoxanthine and xanthine are then oxidized by xanthine oxidase to uric acid.
  • duplicate tubes that contain 125 ⁇ l of the sample to be assayed.
  • Xanthine oxidase (0.643 units, Sigma) in approximately 40 ⁇ l is added to the first tube.
  • a comparable volume of distilled water is added to the second tube, which serves as a uric acid control. This sample is set up to determine the pre ⁇ existing uric acid level in the sample. Reaction mixtures are then incubated for 30 minutes at room temperature.
  • Uric Acid Assay reagent is comprised of Uricase (Candida, 125 U/L) ; Peroxidase (horseradish, 5000 U/L); 4-Aminoantipyrine (4-AAP) (0.3 mmol/L); 3,5Dichloro-2-hydroxybenzensulfonate (DHBS) (2 mmol/L) in a physiological buffer (pH 7.2). Reaction mixtures are incubated for 10-30 minutes.
  • the samples are then transferred to cuvettes and the absorbance measured by a spectrophotometer set at 520 nm.
  • the enzymatic reactions involved in the assay are as follows: deamination xanthine oxidase 1. adenine > hypoxanthine >xanthine
  • Uricase catalyzes the oxidation of uric acid to allantoin, carbon dioxide and hydrogen peroxide.
  • the hydrogen peroxide formed at each step reacts in the presence of peroxidase with 4-aminoantipyrine (4-AAP) and 3,5-dichloro-2-hydroxybenzenesulfonate (DHBS) to form a quinoneimine dye with an absorbance maximum at 520 nm.
  • 4-AAP 4-aminoantipyrine
  • DHBS 3,5-dichloro-2-hydroxybenzenesulfonate
  • the absorbance value of the uric acid control sample is subtracted from the absorbance value of the sample exposed to xanthine oxidase. The resulting value is absorbance due to the presence of xanthine, hypoxanthine or adenine or a combination thereof.
  • Example 2 Method for Detection of Purine Nucleosides (e.g., inosine, xanthosine, adenosine) Interconversion between purine bases and nucleosides are catalyzed by purine nucleoside phosphorylase. The reaction is as follows:
  • the procedure for the detection of purine nucleosides is the same as above with the following modifications.
  • the experimental tube contains the sample to be assayed, 0.643 units of xanthine oxidase and 2.5 units of nucleoside phosphorylase (Sigma Chemical
  • a duplicate tube is set up that contains the sample to be assayed, 0.643 units of xanthine oxidase and a comparable volume of distilled water instead of purine nucleoside phosphorylase. After a 30 minute incubation at room temperature, 150 ⁇ l of each sample is added to 850 ⁇ l of uric acid reagent and the samples are processed as in Example l.
  • the concentration of the purine nucleoside is calculated by subtracting the absorbance value of the xanthine oxidase sample from the absorbance value from the sample containing both xanthine oxidase and nucleoside phosphorylase.
  • the remaining absorbance value is due to the presence of inosine, xanthosine or adenosine, or a combination thereof.
  • hypoxanthine is by far the predominant purine base in human plasma samples. It is about 100-fold more abundant than xanthine and 5-10-fold more abundant than adenine.
  • the absorbance values of 1 mM solutions of the purine bases and nucleosides is shown in Table 1 below:
  • adenine and adenosine by the assay is thought to be due to contaminating deaminases in the xanthine oxidase preparation and/or spontaneous deamination. These reactions can convert adenosine to inosine and adenine to hypoxanthine, a substrate for xanthine oxidase.
  • 2'-deoxycorformycin can be included in the reaction mixture to inhibit such activities and provide a more specific assay for xanthine and hypoxanthine.
  • the use of enzyme preparations with greater purity would also solve this problem. If contaminating deaminases are not responsible, it should be possible to alter the reaction conditions to eliminate spontaneous chemical deamination.
  • hypoxanthine is produced by de novo biosynthesis in the liver, its primary site of production, and then transported to other tissues for utilization in the synthesis of adenine and guanine nucleotides via salvage pathways.
  • the amount of dietary contribution to this primary de novo store is uncertain. However it is likely to be a minor contribution that depends on gut flora enzymes; as purines are not an essential dietary component and nucleic acids, and nucleotides do not freely cross the plasma membrane of cells (Murray, A.W. (1971) , supra ) .
  • hypoxanthine is transported from the liver primarily in "loaded" erythrocytes to other tissues, some of which do not engage in de novo purine biosynthesis at all. There is limited information available on the relationship between plasma and tissue cell levels of hypoxanthine. However, it is clear that hypoxanthine diffuses freely across the erythrocyte plasma membrane, and as a result, erythrocyte hypoxanthine content is determined by tissue-dependent metabolic gradients. Erythrocytes load with hypoxanthine in the high-hypoxanthine-content capillary beds of the liver and release hypoxanthine in its nucleoside form, inosine, in low-hypoxanthine-content tissue capillary beds (Murray, A.W. , (1971) Ann. Rev. Biochem. 40:811- 826) .
  • hypoxanthine is catabolized sequentially by the enzyme xanthine oxidase/dehydrogenase, first to xanthine and then to uric acid. In humans, these two catabolites are excreted in the urine without further utilization (Murray, A.W. , (1971), supra ; Murray et al. , (1970)
  • hypoxanthine is arguably a much better candidate guanine nucleotide-related cancer risk factor because of its essential role in guanine nucleotide metabolism. Hypoxanthine has not been previously assessed for this purpose.
  • plasma samples from 677 individuals were collected from patients enrolled in the Family Risk Assessment Program (FRAP) at the Fox Chase Cancer Center (Philadelphia, PA) . In a number of cases, multiple samples were collected during different stages of disease and/or treatment for the same individual.
  • FRAP Family Risk Assessment Program
  • PA Fox Chase Cancer Center
  • the subject population was composed primarily of women ( ⁇ 50 male samples) , who had either sporadic or inherited breast and/or ovarian cancer, and their unaffected family members (who were for the most part, sisters and mothers) .
  • the plasma collection included the following numbers of cancer types: breast, 107; ovary, 112; breast and ovary, l; endometrium, 27; cervix, 18; miscellaneous cancers, 34; no known cancer but with a family history of cancer, 318; no known cancer and no family history of cancer, 33.
  • the two most represented cancers in the collection, breast and ovarian are known to be associated with very frequent p53 mutations (about 50% and 85% respectively) . As noted earlier, this feature is deemed an essential prerequisite to increase the likelihood for success in establishing a significant association with cancer risk.
  • the colorimetric method of the instant invention was used to quantify hypoxanthine levels in human plasma.
  • the assay was used to analyze plasma samples from 118 women with no known cancer and 49 ovarian cancer patients.
  • the results obtained demonstrate that the cancer patients had a mean plasma hypoxanthine level that was 33% lower than that of the non-cancer group (p ⁇ 0.01). This difference was not explained by differences in the age distributions of the two groups. While there are additional factors that must be analyzed in relation to this result, the data are consistent with the hypothesis of increased guanine nucleotide utilization in the tissues of cancer patients.
  • Certain preferred embodiments of the invention have been described and exemplified herein. However, other embodiments will be apparent to persons skilled in the art. For example, the incorporation of other enzymes or particular enzyme inhibitors may allow variation in the purine specificity of the assay. Thus, the invention is not limited to the embodiments specifically described, but may be varied and modified within the scope of the appended claims.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

A simple assay for purine bases or nucleosides in a liquid sample is provided wherein the purine base or nucleoside is converted to uric acid by the action of xanthine oxidase and purine nucleoside phosphorylase. In an exemplary embodiment, a commercially available uric acid reagent in combination with standards of known concentrations are used to determine the level of purine base or nucleoside in a sample. According to another aspect of the invention, test kits are provided for performing the above-described methods.

Description

ASSAYS FOR DETECTION OF PURINE METABOLITES
Pursuant to 35 U.S.C. §202(c) , it is hereby acknowledged that the U.S. Government has certain rights in the invention described herein, which was made in part with funds from the National Institutes of Health.
FIELD OF THE INVENTION
This invention generally relates to detection and quantitation of selected analytes within a mixed population of analytes. Specifically, simple assays are provided to detect and measure purine bases and/or purine nucleotides in a biological sample.
BACKGROUND OF THE INVENTION
The concentration of a light-absorbing (i.e., colored) substance in solution can be estimated by comparing the intensity of its color with that of several standard solutions of known' concentration. Such methods, using a spectrophotometer as a detector, are known as colorimetric methods; in various forms, they have played a major role historically in analytical chemistry.
Colorimetric assays have been developed to analyze levels of various components in biological or chemical solutions. Similarly, light-emitting (i.e., fluorescent) substances can be detected and quantified using appropriate detection devices, e.g., a fluorometer.
Antimetabolites that interfere with de novo purine synthesis constitute an important part of the arsenal of drugs available for the treatment of leukemia and other cancers. However, not all patients respond equally well to these drugs, and some eventually become refractory to therapy. Resistance to purine analogs has been attributed to the deficiency of enzymes necessary to convert them to their nucleotide forms. Other mechanisms have been observed in experimental tumors, such as increased degradation of drugs and inability of resistant cells to convert the ribonucleotide analogues to the deoxyribonucleotide analogue. Increased plasma concentrations of hypoxanthine during treatment with these drugs may also be a mechanism of resistance to antimetabolites acting in the purine biosynthetic pathways. Extracellular hypoxanthine is important for the activity of purine antimetabolites in two aspects: it can be utilized to restore purine nucleotide pools in cells, the de novo synthesis of which has been blocked, and it potentially may compete with some of these antimetabolites for transport into the cell or for the enzymes necessary to convert the drugs to their active nucleotides.
Thus, the levels of purine metabolites (inosine, hypoxanthine, xanthine, xanthosine, adenine and adenosine) have been extensively investigated in biological fluids in many diseases, including cancer, gout and Lesch-Nyhan syndrome. However, their determination, until a few years ago, presented several problems, as the analytical methods used were of dubious sensitivity and specificity. Methods exist for the detection of these compounds by HPLC, but these methods are expensive, requiring sophisticated instrumentation and technicians with specialized training. Because they require separation of purines from other plasma constituents, they are labor-intensive and poorly quantitative due to difficulties in ensuring uniform sample recovery.
It would therefore be beneficial to have available a rapid, inexpensive, and sensitive method for the determination of purine metabolites which can be performed by unskilled technicians. It is the object of the present invention to provide such a method. It is a further object of the invention to provide a kit for the determination of purine metabolite levels in a given biological sample i.e., blood, urine, seminal fluid, plasma or tissue extracts. SUMMARY OF THE INVENTION
The present invention provides simple assays for determining the presence or quantity of selected purine metabolites in a liquid sample. According to one aspect of the invention, a method is provided for determining the presence or quantity of a selected subpopulation of analytes in a mixed population of analytes, wherein the analyte subpopulation comprises purine metabolites capable of being further metabolized to uric acid. The method comprises providing a liquid sample of the mixed analyte population, adding to that sample at least one reagent that catalyzes the conversion of the purine metabolites to uric acid (under conditions whereby the purine metabolites in the subpopulation are converted to uric acid) , adding to the sample at least one reagent that reacts with uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide (and providing conditions whereby the hydrogen peroxide is produced in the sample) and quantitatively detecting the hydrogen peroxide in the sample. The quantity of hydrogen peroxide produced by the method is proportional to the quantity of purine metabolite analyte subpopulation in the sample.
In preferred embodiments of the aforementioned assay, the mixed analyte population is disposed within a biological fluid or tissue and a liquid sample is prepared from that biological fluid or tissue. The analyte subpopulation preferably comprises purine bases (such as adenine, xanthine and hypoxanthine) or purine nucleosides (such as inosine, xanthosine and adenosine) . Reagents that catalyze the conversion of purine bases to uric acid preferably include the enzyme xanthine oxidase; reagents for catalyzing the conversion of purine nucleosides to uric acid preferably comprise a combination of the enzymes purine nucleoside phosphorylase and xanthine oxidase. In preferred embodiments of the aforementioned method, the uric acid in the sample is exposed to the enzyme, uricase, producing as a byproduct a stoichiometrically equivalent amount of a hydrogen peroxide. The hydrogen peroxide is thereafter detected, preferably by optical means. Detection is accomplished by exposing the hydrogen peroxide in the sample substrates and other reactant which, upon reaction with hydrogen peroxide form the optically detectable product is preferably detected spectrophotometrically or spectrofluorometrically. According to another aspect of the invention, a method is provided for determining the presence or quantity of purine bases in a biological fluid or tissue. The method comprises providing a liquid test sample of the biological fluid or tissue and a volumetrically equivalent liquid control sample. To the test sample is added an amount of xanthine oxidase effective to catalyze the conversion of the purine bases to uric acid. The sample is placed under conditions whereby the purine bases are converted to uric acid. To the control sample is added a volume of a non-reactive liquid medium (such as water or buffer) equivalent to the volume of xanthine oxidase added to the test sample. The control sample is subjected to the same conditions as the test sample. To each of the test sample and control sample is added at least one reagent that reacts with uric acid in the samples to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide. The sample is subjected to conditions whereby the reaction to form the hydrogen peroxide occurs. Thereafter, the hydrogen peroxide is quantitatively detected in each of the test sample and the control sample, the quantity of hydrogen peroxide is proportional to the quantity of the uric acid in each sample. By comparing the difference in quantity of uric acid in the control sample versus the test sample the quantity of purine bases in the biological fluid or the tissue may be determined. According to another aspect of the present invention, a method for determining the presence or quantity of purine nucleosides in a biological fluid or tissue. The method comprises providing volumetrically equivalent liquid test sample and control sample of the biological fluid or tissue. To the test sample is added an amount of nucleoside phosphorylase and xanthine oxidase effective to catalyze the conversion of the purine nucleosides to uric acid. To the control sample is added a volume of a non-reactive liquid medium equivalent to the volume of nucleoside phosphorylase added to the test sample. The control sample is also given an equivalent amount of the xanthine oxidase as added to the test sample. The respective samples are subjected to conditions whereby, in the test sample the purine nucleoside are converted to uric acid. To each of the test sample and the control sample is added at least one reagent that reacts with the uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide. The hydrogen peroxide in each sample is quantitatively detected, that quantity being proportional to the quantity of uric acid present in each sample. The difference in quantity of uric acid in the control sample versus the test sample is compared, that difference being proportional to the quantity of purine nucleosides in the biological fluid or tissue.
According to other aspects of the invention, a test kit for determining the presence or quantity of a subpopulation of analytes comprising purine metabolites capable of being further metabolized to uric acid, in a mixed population of analytes, is provided. The test kit comprises at least one reagent that catalyzes the conversion of the purine metabolites to uric acid, and at least one reagent that reacts with the uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide. In a preferred embodiment of the invention, the test kit further comprises at least one reagent for detecting the hydrogen peroxide.
The assays in the test kits of the present invention provide a rapid, inexpensive and sensitive system for determining the presence or amount of purine metabolite in a biological sample. These methods and test kit provide an inexpensive alternative to sophisticated, instrument-dependent assays presently available, and can be performed in the clinic or in the field by unskilled technicians.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to a simple method for determining the presence or quantity of a selected subpopulation of purine metabolites in a liquid sample, preferably obtained from a biological fluid or tissue. The method is designed to measure purine bases and purine nucleosides, either together or separately. The assays of the invention are based on the known enzymatic activity of xanthine oxidase and purine nucleoside phosphorylase, and relies on converting the substrates of these enzymes (e.g., the purine nucleotides, inosine, xanthosine and adenosine, and purine bases xanthine, hypoxanthine and adenine) to uric acid. The uric acid may be quantitatively detected in a variety of ways. In a most preferred embodiments of the present invention, the uric acid is exposed to the enzyme, uricase, under oxidative conditions, which results in the formation of a stoichiometrically equivalent amount of hydrogen peroxide (H202) as a reaction byproduct.
The ultimate step in the method of the present invention is based upon the detection of the byproduct, hydrogen peroxide. Hydrogen peroxide levels may be quantitated using several different procedures. In a preferred embodiment of the invention, a commercially available uric acid reagent kit (Sigma Chemical Co., St. Louis, MO)may be utilized. Purine metabolites initially present in a liquid sample are acted on by xanthine oxidase or purine phosphorylase. The resulting reaction product is uric acid which is then exposed to enzymes (uricase peroxidase) and other reagents present in the uric acid kit, which results in the generation of hydrogen peroxide. Hydrogen peroxide can be detected and quantitated many ways, as described in greater detail below. In the uric acid reagent kit, H202 is detected via its reaction with two substrates to form a quinoneimine dye that absorbs light at 520 nm (see Green and Hill (1984) Meth. Enz. 105: 3-22).
The method of this invention, exemplified by the determination of hypoxanthine and xanthosine, comprises in its most preferred form: incubating a liquid sample, i.e., plasma, tissue extract or urine, with enzymes selected from the group of xanthine oxidase and purine nucleoside phosphorylase or a combination thereof; adding the above components to a tube containing uric acid reagent (Sigma) ; and reading the absorbance values of the resulting reaction product in a spectrophotometer set at 520 nm. The presence of xanthine, xanthosine, inosine, hypoxanthine, adenine or adenosine are then determined. Another method for detecting H2θ2 is based upon the hydrolysis of a stable reagent, diacetyl- dichlorofluoroscin (LDADCF) , by sodium hydroxide to a less stable nonfluorescent compound dichlorofluoroscin (LDCF) , which is oxidized by hydrogen peroxide to the fluorescent dichlorofluorescin (DCF) . This fluorometric analysis is used successfully to detect ultramicroquantities of hydrogen peroxide (Sanchez-Ferrer et al., (1990) Analytical Biochem. 187:129-132).
Scopoletin may also be used to detect H202. The method is based on quantitation of decreases in scopoletin fluorescence following its oxidation by H202 and horseradish peroxidase (Clifford et al., (1984), Meth. Enz. JL0_5:393-398) . More specifically, when activated by light at 350 nm, scopoletin fluoresces with a peak at 460 nm. When oxidized by H202 and horseradish peroxidase, scopoletin loses its fluorescence in direct proportion to H202 concentration.
Another method for assessing hydrogen peroxide levels in a sample described by Pick and Mizel, (1981) J. Immunol. Meth. 4.6211-266. This is based on the horseradish peroxidase dependent conversion of phenol red by H202 into a compound with increased absorbance at 600- 610 nm. The spectrophotometric measurement is made after adjusting the pH of the reaction mixture to 12.5, in order to eliminate changes in the absorbance of phenol red due to its behavior as a pH indicator. Hydrogen peroxide levels may also be quantitated with dichlorophenol-indophenol. In this procedure, a sample is added to a blue, oxidized dichlorophenol-indophenol solution. The 605 nm absorbance of this solution immediately decreases in response to the reducing action of ascorbate and/or other reductants present in the sample. The extent of reoxidation of the solution upon the addition of peroxidase, as measured by the increase in its 605 nm absorbance, can be quantitatively related to the amount of H202 in the sample. Due to the observation that reduced dichlorophenol-indophenol spontaneously reoxidizes at a rate of 0.03 nmol min'1 μM"1 with generation of H202, this is not a particularly preferred method as it has the potential to overestimate the H202 content in the sample (Garcia-Castineiras et al., Exp. Eye Res. 55:9-
19).
It is apparent from the methods described above (which are exemplary, rather than all-inclusive) that various procedures may be utilized to detect the final reaction end product, H202, in the present assays. These methods are also contemplated for use in the instant invention. In a further aspect, the invention provides a kit of reagents for carrying out the assays of the invention. The kit contains reagents for converting prime nucleosides and/or bases to uric acid, as well as reagents for producing H202 from uric acid and, optionally, reagents for detecting the hydrogen peroxide. Thus for example, a kit according to the present invention may comprise reaction vials, preferably cuvettes, lyophilized enzymes (e.g., xanthine oxidase, nucleoside phosphorylase) , calibrated reference solutions, an appropriate diluent, and the uric acid reagent (Sigma) or an appropriate substitute (e.g., one or more of the individual components of the reagent obtained from Sigma or other suppliers, or modifications of the reagent, such as described by James and Price (1984) Ann. Clin. Biochem. 21' 405-410.
The examples set forth below are provided to describe the invention in further detail. They are not intended to limit the invention. Examples 1 and 2 describe assays for detecting purine bases and nucleosides. Example 3 shows typical results of the assays. Example 4 describes use of the assays to detect altered plasma hypoxanthine levels in cancer patients.
Example 1
Method for Detection of Purine Bases (e.g., adenine, xanthine, hypoxanthine)
The degradation of purines to the end product uric acid occurs via the action of deaminase enzymes that first convert adenine and guanine to hypoxanthine and xanthine, respectively. Deamination may also occur non- enzymatically. This is a more frequent occurrence for adenine because, unlike guanine, the amino group in adenine is attached to a 6-membered aromatic ring. Hypoxanthine and xanthine are then oxidized by xanthine oxidase to uric acid. To detect the levels of purine bases (xanthine, hypoxanthine, or adenine) in a liquid sample, duplicate tubes are prepared that contain 125 μl of the sample to be assayed. Xanthine oxidase (0.643 units, Sigma) in approximately 40 μl is added to the first tube. A comparable volume of distilled water is added to the second tube, which serves as a uric acid control. This sample is set up to determine the pre¬ existing uric acid level in the sample. Reaction mixtures are then incubated for 30 minutes at room temperature. After 30 minutes, 150 μl of the sample from each tube, (the control sample and the experimental sample) are added to two separate tubes containing 850 μl of Uric Acid Assay reagent prepared at 1.176 times the concentration specified by the supplier (Sigma Chemical Co. , St. Louis, MO) . Uric acid reagent is comprised of Uricase (Candida, 125 U/L) ; Peroxidase (horseradish, 5000 U/L); 4-Aminoantipyrine (4-AAP) (0.3 mmol/L); 3,5Dichloro-2-hydroxybenzensulfonate (DHBS) (2 mmol/L) in a physiological buffer (pH 7.2). Reaction mixtures are incubated for 10-30 minutes. The samples are then transferred to cuvettes and the absorbance measured by a spectrophotometer set at 520 nm. The enzymatic reactions involved in the assay are as follows: deamination xanthine oxidase 1. adenine > hypoxanthine >xanthine
+ H20 + 02 +H202
xanthine oxidase
2. xanthine + H20 + 02 > uric acid + H202
Uricase
3 . uric acid + 02 + H20 >allantoin + C02 +H202 Peroxidase
4. H202 + 4-AAP + DHBS > quinoneimine dye + 3H20
Uricase catalyzes the oxidation of uric acid to allantoin, carbon dioxide and hydrogen peroxide. The hydrogen peroxide formed at each step reacts in the presence of peroxidase with 4-aminoantipyrine (4-AAP) and 3,5-dichloro-2-hydroxybenzenesulfonate (DHBS) to form a quinoneimine dye with an absorbance maximum at 520 nm. To determine the presence of a purine base, the absorbance value of the uric acid control sample is subtracted from the absorbance value of the sample exposed to xanthine oxidase. The resulting value is absorbance due to the presence of xanthine, hypoxanthine or adenine or a combination thereof.
Example 2 Method for Detection of Purine Nucleosides (e.g., inosine, xanthosine, adenosine) Interconversion between purine bases and nucleosides are catalyzed by purine nucleoside phosphorylase. The reaction is as follows:
phosphorylase
5. purine nucleoside + Pi +* purine + ribose-1-phosphate
The procedure for the detection of purine nucleosides is the same as above with the following modifications. The experimental tube contains the sample to be assayed, 0.643 units of xanthine oxidase and 2.5 units of nucleoside phosphorylase (Sigma Chemical
Co., St. Louis, MO). As a control for pre-existing uric acid and purine base levels, a duplicate tube is set up that contains the sample to be assayed, 0.643 units of xanthine oxidase and a comparable volume of distilled water instead of purine nucleoside phosphorylase. After a 30 minute incubation at room temperature, 150 μl of each sample is added to 850 μl of uric acid reagent and the samples are processed as in Example l. In this Example, the concentration of the purine nucleoside is calculated by subtracting the absorbance value of the xanthine oxidase sample from the absorbance value from the sample containing both xanthine oxidase and nucleoside phosphorylase. The remaining absorbance value is due to the presence of inosine, xanthosine or adenosine, or a combination thereof.
Example 3
Absorbances Observed for Purine Compounds Measured According to the Assays of Examples 1 and 2
While the purine base assay presently detects three different bases, we have found by HPLC analysis that hypoxanthine is by far the predominant purine base in human plasma samples. It is about 100-fold more abundant than xanthine and 5-10-fold more abundant than adenine. The absorbance values of 1 mM solutions of the purine bases and nucleosides is shown in Table 1 below:
Absorbance at 520 nm
Purine Purine Purine comnound Base Assay Nucleoside Assay
Hypoxanthine 1.18 ± 0.02 0.0
Xanthine 0.84 ± 0.01 0.0
Adenine 0.12 ± 0.01 0.0
Guanine 0.00 ± 0.0 0.0
Uric Acid 0.00 ± 0.0 0.0
Inosine (Not Done) 0.60
Xanthosine 0.0 ± 0.0 0.61
Adenosine (Not Done) 0.68
Guanosine 0.0 ± 0.0 0.00
Deoxyguanosine 0.0 ± 0.0 0.00
The detection of adenine and adenosine by the assay is thought to be due to contaminating deaminases in the xanthine oxidase preparation and/or spontaneous deamination. These reactions can convert adenosine to inosine and adenine to hypoxanthine, a substrate for xanthine oxidase. In another embodiment of the invention, 2'-deoxycorformycin can be included in the reaction mixture to inhibit such activities and provide a more specific assay for xanthine and hypoxanthine. The use of enzyme preparations with greater purity would also solve this problem. If contaminating deaminases are not responsible, it should be possible to alter the reaction conditions to eliminate spontaneous chemical deamination.
Example 4 Detection of Altered Plasma Hypoxanthine levels in Cancer Patients
It has been shown in earlier work that p53 is one of the most frequently mutated genes in human cancer. It is believed that p53 mutations result in altered guanine nucleotide metabolism (Sherley, J. (1991) J.
Biol. Chem. 266:24815-24828; Sherley et al., (1995) Proc. Natl. Acad. Sci. USA 92.:136-140) . This alteration might be an increase in plasma hypoxanthine due to increased production of guanine nucleotides. Alternatively, decreases may be due to increased tissue utilization of hypoxanthine that results in carcinogenic levels of postulated critical cellular guanine ribonucleotides. Quantitative assays for plasma hypoxanthine will provide insight into the relationship between p53 mutation and altered guanine metabolism. Hypoxanthine is the body's primary currency for intercellular purine transport (Murray, A.W. (1971) Ann. Rev. Biochem. 4.0:811-826) . Hypoxanthine is produced by de novo biosynthesis in the liver, its primary site of production, and then transported to other tissues for utilization in the synthesis of adenine and guanine nucleotides via salvage pathways. The amount of dietary contribution to this primary de novo store is uncertain. However it is likely to be a minor contribution that depends on gut flora enzymes; as purines are not an essential dietary component and nucleic acids, and nucleotides do not freely cross the plasma membrane of cells (Murray, A.W. (1971) , supra ) . Hypoxanthine is transported from the liver primarily in "loaded" erythrocytes to other tissues, some of which do not engage in de novo purine biosynthesis at all. There is limited information available on the relationship between plasma and tissue cell levels of hypoxanthine. However, it is clear that hypoxanthine diffuses freely across the erythrocyte plasma membrane, and as a result, erythrocyte hypoxanthine content is determined by tissue-dependent metabolic gradients. Erythrocytes load with hypoxanthine in the high-hypoxanthine-content capillary beds of the liver and release hypoxanthine in its nucleoside form, inosine, in low-hypoxanthine-content tissue capillary beds (Murray, A.W. , (1971) Ann. Rev. Biochem. 40:811- 826) .
Hypoxanthine is catabolized sequentially by the enzyme xanthine oxidase/dehydrogenase, first to xanthine and then to uric acid. In humans, these two catabolites are excreted in the urine without further utilization (Murray, A.W. , (1971), supra ; Murray et al. , (1970)
Progr. Nucl. Acid Res. Mol. Biol. 10:87-119) . Unlike hypoxanthine, for which there are no clinical tests in common practice, the measurement of serum uric acid concentration is a routine clinical laboratory test. It provides diagnostic information for an array of pathological conditions including renal failure, gout, in-born errors of purine metabolism (e.g., Lesch-Nyhan's Disease), and hyper-catabolic states (e.g., cancer) Serum uric acid has been evaluated as a potential cancer risk marker. However, no significant association has been found (Hiatt et al., (1988) Cancer Research 48:2916- 2918; Kolonel, et al., (1994) Can. Epidem. Biomarkers Prev. 3_:225-228) . Hypoxanthine is arguably a much better candidate guanine nucleotide-related cancer risk factor because of its essential role in guanine nucleotide metabolism. Hypoxanthine has not been previously assessed for this purpose. To assess hypoxanthine levels in cancer patients, plasma samples from 677 individuals were collected from patients enrolled in the Family Risk Assessment Program (FRAP) at the Fox Chase Cancer Center (Philadelphia, PA) . In a number of cases, multiple samples were collected during different stages of disease and/or treatment for the same individual. The subject population was composed primarily of women (<50 male samples) , who had either sporadic or inherited breast and/or ovarian cancer, and their unaffected family members (who were for the most part, sisters and mothers) . The plasma collection included the following numbers of cancer types: breast, 107; ovary, 112; breast and ovary, l; endometrium, 27; cervix, 18; miscellaneous cancers, 34; no known cancer but with a family history of cancer, 318; no known cancer and no family history of cancer, 33. The two most represented cancers in the collection, breast and ovarian, are known to be associated with very frequent p53 mutations (about 50% and 85% respectively) . As noted earlier, this feature is deemed an essential prerequisite to increase the likelihood for success in establishing a significant association with cancer risk.
The colorimetric method of the instant invention, as set forth in Examples 1 and 2, was used to quantify hypoxanthine levels in human plasma. The assay was used to analyze plasma samples from 118 women with no known cancer and 49 ovarian cancer patients. The results obtained demonstrate that the cancer patients had a mean plasma hypoxanthine level that was 33% lower than that of the non-cancer group (p<0.01). This difference was not explained by differences in the age distributions of the two groups. While there are additional factors that must be analyzed in relation to this result, the data are consistent with the hypothesis of increased guanine nucleotide utilization in the tissues of cancer patients. Certain preferred embodiments of the invention have been described and exemplified herein. However, other embodiments will be apparent to persons skilled in the art. For example, the incorporation of other enzymes or particular enzyme inhibitors may allow variation in the purine specificity of the assay. Thus, the invention is not limited to the embodiments specifically described, but may be varied and modified within the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A method for determining the presence or quantity of a selected subpopulation of analytes in a mixed population of analytes, the analyte subpopulation comprising purine metabolites capable of being further metabolized to uric acid, the method comprising: a) providing a liquid sample of the mixed analyte population; b) adding to the sample at least one reagent that catalyzes the conversion of the purine metabolites to uric acid, under conditions whereby the purine metabolites in the subpopulation are converted to uric acid; c) adding to the sample at least one reagent that reacts with uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide, under conditions whereby the stoichiometrically equivalent amount of hydrogen peroxide is produced in the sample; and d) quantitatively detecting the hydrogen peroxide in the sample, the quantity of hydrogen peroxide being proportional to the quantity of the analyte subpopulation in the sample.
2. The method of claim 1, wherein the mixed analyte population is disposed within a biological fluid or tissue and the liquid sample is prepared from the biological fluid or tissue.
3. The method of claim 2, wherein the biological fluid is selected from the group consisting of plasma, urine, seminal fluid, saliva, tissue extract, cell extract and conditioned medium of cultured cells.
4. The method of claim 3, wherein the biological fluid or tissue is plasma.
5. The method of claim 1, wherein the analyte subpopulation comprises purine bases.
6. The method of claim 5, wherein the purine bases are selected from the group consisting of adenine, xanthine and hypoxanthine.
7. The method of claim 5, wherein said at least one reagent that catalyzes the conversion to uric acid includes xanthine oxidase.
8. The method of claim 1, wherein the analyte subpopulation comprises purine nucleosides.
9. The method of claim 8, wherein the purine nucleosides are selected from the group consisting of inosine, xanthosine and adenosine.
10. The method of claim 8, wherein said at least one reagent that catalyzes the conversion to uric acid includes phosphorylase and xanthine oxidase.
11. The method of claim 1, wherein the reagent that reacts with the uric acid is uricase.
12. The method of claim 1, wherein the hydrogen peroxide is detected by adding to the liquid sample at least one reagent that reacts with the hydrogen peroxide to form a proportional amount of an optically detectable product.
13. The method of claim 12, wherein the optically detectable product is a spectrophotometrically detectable product.
14. The method of claim 13, wherein said at least one reagent that reacts with hydrogen peroxide includes a combination of peroxidase, 4-aminoantipyrine and 3, 5- dichloro-2-hydoxybenzenesulfonate, and the spectrophotometrically detectable product is a quinoneimine dye.
15. The method of claim 12, wherein the optically detectable product is a fluorescent product.
16. A method for determining the presence or quantity of purine bases in a biological fluid or tissue, the method comprising: a) providing a liquid test sample of the biological fluid or tissue; b) providing a liquid control sample of the biological fluid or tissue that is volumetrically equivalent to the test sample; c) adding to the test sample an amount of xanthine oxidase effective to catalyze the conversion of the purine bases to uric acid, under conditions whereby the purine bases in the sample are converted to uric acid; d) adding to the control sample a volume of a non-reactive liquid medium equivalent to the volume of xanthine oxidase added to the test sample; e) adding to each of the test sample and the control sample at least one reagent that reacts with uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide, under conditions whereby the stoichiometrically equivalent amount of hydrogen peroxide is produced in each sample; f) quantitatively detecting the hydrogen peroxide in each of the test sample and the control sample, the quantity of hydrogen peroxide being proportional to the quantity of the uric acid in each sample; and g) comparing the difference in quantity of uric acid in the control sample and in the test sample, the difference being proportional to the quantity of purine bases in the biological fluid or tissue.
17. The method of claim 6, wherein the biological fluid is selected from the group consisting of plasma, urine, seminal fluid, saliva, tissue extract, cell extract and conditioned medium of cultured cells.
18. The method of claim 17, wherein the biological fluid or tissue is plasma.
19. The method of claim 16, wherein the purine bases are selected from the group consisting of adenine, xanthine and hypoxanthine.
20. The method of claim 16, wherein the reagent that reacts with the uric acid is uricase.
21. The method of claim 16, wherein the hydrogen peroxide is detected by adding to each sample at least one reagent that reacts with the hydrogen peroxide to form a proportional amount of an optically detectable product.
22. The method of claim 21, wherein the optically detectable product is a spectrophotometrically detectable product.
23. The method of claim 22, wherein said at least one reagent that reacts with hydrogen peroxide includes a combination of peroxidase, 4-aminoantipyrine and 3, 5- dichloro-2-hydoxybenzenesulfonate, and the spectrophotometrically detectable product is a quinoneimine dye.
24. The method of claim 21, wherein the optically detectable product is a fluorescent product.
25. A method for determining the presence or quantity of purine nucleosides in a biological fluid or tissue, the method comprising: a) providing a liquid test sample of the biological fluid or tissue; b) providing a liquid control sample of the biological fluid or tissue that is volumetrically equivalent to the test sample; c) adding to the test sample an amount of a combination of nucleoside phosphorylase and xanthine oxidase effective to catalyze the conversion of the purine nucleosides to uric acid, under conditions whereby the purine nucleosides in the sample are converted to uric acid; d) adding to the control sample a volume of a non-reactive liquid medium equivalent to the volume of nucleoside phosphorylase added to the test sample and an equivalent amount of the xanthine oxidase as added to the test sample; e) adding to each of the test sample and the control sample at least one reagent that reacts with uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide, under conditions whereby the stoichiometrically equivalent amount of hydrogen peroxide is produced in each sample; f) quantitatively detecting the hydrogen peroxide in each of the test sample and the control sample, the quantity of hydrogen peroxide being proportional to the quantity of the uric acid in each sample; and g) comparing the difference in quantity of uric acid in the control sample and in the test sample, the difference being proportional to the quantity of purine nucleosides in the biological fluid or tissue.
26. The method of claim 25, wherein the biological fluid is selected from the group consisting of plasma, urine, seminal fluid, saliva, tissue extract, cell extract and conditioned medium of cultured cells.
27. The method of claim 26, wherein the biological fluid or tissue is plasma.
28. The method of claim 25, wherein the purine nucleosides are selected from the group consisting of inosine, xanthosine and adenosine.
29. The method of claim 25, wherein the reagent that reacts with the uric acid is uricase.
30. The method of claim 25, wherein the hydrogen peroxide is detected by adding to each sample at least one reagent that reacts with the hydrogen peroxide to form a proportional amount of an optically detectable product.
31. The method of claim 30, wherein the optically detectable product is a spectrophotometrically detectable product.
32. The method of claim 31, wherein said at least one reagent that reacts with hydrogen peroxide includes a combination of peroxidase, 4-aminoantipyrine and 3, 5- dichloro-2-hydoxybenzenesulfonate, and the spectrophotometrically detectable product is a quinoneimine dye.
33. The method of claim 29, wherein the optically detectable product is a fluorescent product.
34. A test kit for determining the presence or quantity of a selected subpopulation of analytes in a mixed population of analytes, the analyte subpopulation comprising purine metabolites capable of being further metabolized to uric acid, the kit comprising: a) at least one reagent that catalyzes the conversion of the purine metabolites to uric acid; and b) at least one reagent that reacts with uric acid to produce, as a reaction byproduct, a stoichiometrically equivalent amount of hydrogen peroxide.
35. The test kit of claim 34, which further comprises at least one reagent for detecting the hydrogen peroxide.
PCT/US1997/002839 1996-04-15 1997-02-25 Assays for detection of purine metabolites WO1997039352A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU19708/97A AU1970897A (en) 1996-04-15 1997-02-25 Assays for detection of purine metabolites

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63205596A 1996-04-15 1996-04-15
US08/632,055 1996-04-15

Publications (1)

Publication Number Publication Date
WO1997039352A1 true WO1997039352A1 (en) 1997-10-23

Family

ID=24533894

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/002839 WO1997039352A1 (en) 1996-04-15 1997-02-25 Assays for detection of purine metabolites

Country Status (2)

Country Link
AU (1) AU1970897A (en)
WO (1) WO1997039352A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000034506A2 (en) * 1998-12-04 2000-06-15 University Of North Carolina At Chapel Hill Method of screening candidate compounds for susceptibility to oxidative metabolism
WO2001076606A2 (en) * 2000-04-07 2001-10-18 Roman Rozencwaig Use of uric acid or precursors thereof as a treatment for cancer, aids and the like
KR20190081458A (en) * 2017-12-29 2019-07-09 (주)큐브바이오 Apparatus for analysing purine metabolite concentration such as hypoxanthine and xanthine for tumor diagnosis
CN110951823A (en) * 2019-12-31 2020-04-03 扬中酵诚生物技术研究有限公司 Uric acid detection kit and manufacturing process thereof
CN112161945A (en) * 2020-10-27 2021-01-01 安徽农业大学 Ultra-micro detection method for purine content of soybean grains
CN112730284A (en) * 2020-12-22 2021-04-30 成都医学院 Method for measuring adenine content and RIP activity
CN113916809A (en) * 2021-09-06 2022-01-11 首都儿科研究所附属儿童医院 POCT (Point of Care testing) detection method for hypoxanthine content in urine and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6049800A (en) * 1983-08-30 1985-03-19 Kankyo Bunseki Center:Kk Measurement of inosine and/or hypoxanthine and test paper for measuring
JPS6240300A (en) * 1985-08-13 1987-02-21 Wako Pure Chem Ind Ltd Activity measurement of adenosine deaminase in humor
JPH0380099A (en) * 1989-08-24 1991-04-04 Iatron Lab Inc Enzymatic determination of inorganic phosphorus using chemiluminescence

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6049800A (en) * 1983-08-30 1985-03-19 Kankyo Bunseki Center:Kk Measurement of inosine and/or hypoxanthine and test paper for measuring
JPS6240300A (en) * 1985-08-13 1987-02-21 Wako Pure Chem Ind Ltd Activity measurement of adenosine deaminase in humor
JPH0380099A (en) * 1989-08-24 1991-04-04 Iatron Lab Inc Enzymatic determination of inorganic phosphorus using chemiluminescence

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
AGRICULTURAL AND BIOLOGICAL CHEMISTRY, 1982, Vol. 46, No. 3, Y. MACHIDA et al., pages 807-808. *
ANALYST, June 1968, Vol. 93, R.A. CHALMERS et al., pages 354-362. *
ANALYTICA CHIMICA ACTA, 1986, Vol. 186, Y. HAYASHI et al., pages 131-137. *
ANALYTICA CHIMICA ACTA, 1988, Vol. 208, T. IMASAKI et al., pages 325-329. *
ANALYTICAL BIOCHEMISTRY, 1989, Vol. 182, H. KAWASAKI et al., pages 366-370. *
ANALYTICAL BIOCHEMISTRY, 1994, Vol. 222, P.R. LE TISSIER et al., pages 168-175. *
IGAKU KENSA, 1991, Vol. 40, No. 1, M. OCHI et al., pages 66-70. *
JOURNAL OF CHROMATOGRAPHY, 1990, Vol. 528, Y. KITO et al., pages 91-99. *
JOURNAL OF FLOW INJECTION ANALYSIS, 1990, Vol. 7, No. 2, M. TOMODA et al., pages 113-129. *
NIPPON RINSHO KAGAKKAI BUNSEKI BUKAI SHIKOKU SHIBU KAISHI, 1989, Vol. 6, No. 1, M. OCHI et al., pages 29-36. *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000034506A2 (en) * 1998-12-04 2000-06-15 University Of North Carolina At Chapel Hill Method of screening candidate compounds for susceptibility to oxidative metabolism
WO2000034506A3 (en) * 1998-12-04 2000-11-23 Univ North Carolina Method of screening candidate compounds for susceptibility to oxidative metabolism
US6312917B1 (en) 1998-12-04 2001-11-06 The University Of North Carolina At Chapel Hill Method of screening candidate compounds for susceptibility to oxidative metabolism
AU774229B2 (en) * 1998-12-04 2004-06-17 University Of North Carolina At Chapel Hill, The Method of screening candidate compounds for susceptibility to oxidative metabolism
WO2001076606A2 (en) * 2000-04-07 2001-10-18 Roman Rozencwaig Use of uric acid or precursors thereof as a treatment for cancer, aids and the like
WO2001076606A3 (en) * 2000-04-07 2002-08-01 Roman Rozencwaig Use of uric acid or precursors thereof as a treatment for cancer, aids and the like
KR20190081458A (en) * 2017-12-29 2019-07-09 (주)큐브바이오 Apparatus for analysing purine metabolite concentration such as hypoxanthine and xanthine for tumor diagnosis
KR102094024B1 (en) * 2017-12-29 2020-03-26 (주)큐브바이오 Apparatus for analysing purine metabolite concentration such as hypoxanthine and xanthine for tumor diagnosis
CN110951823A (en) * 2019-12-31 2020-04-03 扬中酵诚生物技术研究有限公司 Uric acid detection kit and manufacturing process thereof
CN112161945A (en) * 2020-10-27 2021-01-01 安徽农业大学 Ultra-micro detection method for purine content of soybean grains
CN112730284A (en) * 2020-12-22 2021-04-30 成都医学院 Method for measuring adenine content and RIP activity
CN113916809A (en) * 2021-09-06 2022-01-11 首都儿科研究所附属儿童医院 POCT (Point of Care testing) detection method for hypoxanthine content in urine and application

Also Published As

Publication number Publication date
AU1970897A (en) 1997-11-07

Similar Documents

Publication Publication Date Title
Fossati et al. Enzymic creatinine assay: a new colorimetric method based on hydrogen peroxide measurement.
US4350762A (en) Aminopyrine improved Trinder&#39;s reagent and dosing process for hydrogen peroxide from enzymatic oxidation of metabolic substrata with the same
US5516700A (en) Automated urinalysis method
EP0763132B1 (en) Assay for enzyme activity using a microfluorometric assay
EP0200540B1 (en) Analytical element and method for determination of creatine kinase isoenzyme
US20090042279A1 (en) Method of detecting mild impaired glucose tolerance or insulin secretory defect
EP0116307B1 (en) Composition, analytical element and method for the quantification of creatine kinase
Kohlbecker et al. Direct spectrophotometric determination of serum and urinary oxalate with oxalate oxidase
WO1997039352A1 (en) Assays for detection of purine metabolites
Majkić-Singh et al. Spectrophotometric assay of xanthine oxidase with 2, 2'-azino-di (3-ethylbenzthiazoline-6-sulphonate)(ABTS) as chromogen
JP3975279B2 (en) Examination method of pre-diabetes group
EP1157128B1 (en) Homogeneous enzymatic assay for vitamin b6
Orfanos et al. A rapid screening test for Duchenne muscular dystrophy using dried blood specimens
US7198905B2 (en) Method of screening methods prediabetic state and screening reagent
EP0200541B1 (en) Immunochemical method and analytical composition and element for determination of creatine kinase-mb
Isobe et al. A rapid enzymatic assay for total blood polyamines
US7374902B2 (en) Enzyme cycling based assays for alpha-methylacyl-CoA racemase
JPWO2006030866A1 (en) Method for quantitative determination of uric acid
Bais et al. Urinary glycolate measured by use of (S)-2-hydroxy-acid oxidase.
JP3036711B2 (en) Highly sensitive lactic acid or pyruvic acid quantification method and composition for quantification
SUzUKI et al. Use of the o-phenylenediamine fluorescence system in the enzymatic assay of serum uric acid
JP3034984B2 (en) Highly sensitive method and composition for quantification of D-galactose
EP0989190B1 (en) Dry analytical element for quantitative analysis of creatine kinase MB isozyme
US5998158A (en) Glucose free, stable dry analytical element for quantitative analysis of creatine kinase MB isozyme
Panteghini et al. Evaluation of a new commercial assay kit for quantification of lactate dehydrogenase isoenzyme 1 in serum

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA IL JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97537077

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: CA

122 Ep: pct application non-entry in european phase