CA2597044A1 - 5-fluoro-uracil immunoassay - Google Patents

5-fluoro-uracil immunoassay Download PDF

Info

Publication number
CA2597044A1
CA2597044A1 CA002597044A CA2597044A CA2597044A1 CA 2597044 A1 CA2597044 A1 CA 2597044A1 CA 002597044 A CA002597044 A CA 002597044A CA 2597044 A CA2597044 A CA 2597044A CA 2597044 A1 CA2597044 A1 CA 2597044A1
Authority
CA
Canada
Prior art keywords
compound
antibody
conjugate
formula
carrier
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
CA002597044A
Other languages
French (fr)
Other versions
CA2597044C (en
Inventor
Salvatore Salamone
Jodi Blake Courtney
Dennis Stocker
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Saladax Biomedical Inc
Original Assignee
Saladax Biomedical Inc.
Salvatore Salamone
Jodi Blake Courtney
Dennis Stocker
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 Saladax Biomedical Inc., Salvatore Salamone, Jodi Blake Courtney, Dennis Stocker filed Critical Saladax Biomedical Inc.
Publication of CA2597044A1 publication Critical patent/CA2597044A1/en
Application granted granted Critical
Publication of CA2597044C publication Critical patent/CA2597044C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/961Chemistry: molecular biology and microbiology including a step of forming, releasing, or exposing the antigen or forming the hapten-immunogenic carrier complex or the antigen per se
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/975Kit
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/815Test for named compound or class of compounds
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/81Carrier - bound or immobilized peptides or proteins and the preparation thereof, e.g. biological cell or cell fragment as carrier
    • Y10S530/811Peptides or proteins is immobilized on, or in, an inorganic carrier
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/13Tracers or tags

Abstract

Novel conjugates of 5-fluoro-uracil and novel 5-fhioro-uracil immunogens and monoclonal antibodies generated by these immunogens which are useful in immunoassays for the quantification and monitoring of 5-fluoro-uracil in biological fluids.

Description

r;-FLUORO-URACIL IMMUNOASSAY

FIELD OF THE INVENTION

This invention relates to the field of immunological assays for determining the presence and/or quantifying the amount of 5-fluoro-uracil [5-FU] in human biological samples in order to rapidly determine optimal drug concentrations during chemotherapy.

BACKGROUND OF THE INVENTION

Cancer is a term used to describe a group of malignancies that all share the common trait of developing when cells in a part of the body begin to grow out of control. Most.
cancers form as tumors, but can also manifest in the blood and circulate through other 1o tissues where they grow. Cancer malignancies are most commonly treated with a combination of surgery, chemotherapy, and/or radiation therapy. The type of treatment used to treat a specific cancer depends upon several factors including the type of cancer malignancy and the stage during which it was diagnosed.

5-FU is one of the more commonly used cytotoxic agents that are used for the treatment of Breast and Colorectal cancer. This chemotherapeutic agent has the formula:

H F
N

I

I
H I
~

This compound has been associated with debilitating side effects such as bone marrow density loss, mucositis, nausea and vomiting. By monitoring the levels of 5-FU
in the body and adjusting the dose these side effects can be better controlled and limited in patients.

At the same time, there is often a highly variable relationship between the dose of 5-FU
and the resulting serum drug concentration that affects therapeutic effect.
The degree of intra- and inter- individual pharmacokinetic variability of 5-FU can be as high as lo-fold (Diasio et. al. J. Clin. Invest. 81: pp 47-51, 1988, Wei et. al. J. Clin.
Invest. 98:

pp61o-615, 1996) and is impacted by many factors, including:
o Organ function o Genetic regulation o Disease state o Age o Drug-drug interaction o Time of drug ingestion, o Mode of drug administration, and o Technique-related administration.

As a result of this variability, equal doses of the same drug in different individuals can result in dramatically different clinical outcomes (Hon et. al. Clinical Chemistry 44, pp 388-40o,1998). The effectiveness of the same 5-FU dosage varies significantly based upon individual drug clearance and the ultimate serum drug concentration in the patient. Therapeutic drug management would provide the clinician with insight on patient variation in both oral and intravenous drug administrations. With therapeutic
2 drug management, drug dosages could be individualized to the patient, and the chances of effectively treating the cancer without the unwanted side effects would be much higher (Nieto, Current Drug Metabolism 2: pp 53 -66, 2001).

In addition, therapeutic drug management of 5-FU would serve as an excellent tool to ensure compliance in administering chemotherapy with the actual prescribed dosage and achievement of the effective serum concentration levels. It has been found that variability in serum concentration is not only due to physiological factors, but can also result from variation in administration technique and ability of the body to absorb 5-FU.
As a chemotherapeutic agent, 5-FU can be administered in its pro-drug form as tegafur 1o which has the structure:

H F
~~ ~~

~
~

Tegafur, when administered to a patient, is generally absorbed and metabolized into 5-FU by the patient at different rates. Therefore, in monitoring the level of 5-FU in patients by means of an immunoassay, it is important that the immunoassay be able to distinguish between tegafur, the inactive substance, and 5-FU, the active substance, into which tegafur metabolizes. The problem with antibodies to 5-FU is that they could be cross-reactive with tegafur making these immunoassays not useful.
3 Routine therapeutic drug management of 5-FU would require the availability of simple automated tests adaptable to general laboratory equipment. Tests that best fit these criteria are immunoassays. Currently there are no immunoassays for 5-FU
available and monitoring levels of this drug is conducted by physical methods like high pressure liquid chromatography (HPLC) (Escoriaza et. al. J. of Chromatography B: Biomedical Sciences and applications, 736 (1+2): pp 97-102,1999)= In order to be most effective in monitoring drug levels the antibody should be most specific to 5-FU and display very low cross-reactivity to no cross-reactivity to related pyrimidine bases, particularly tegafur.

SUMMARY OF INVENTION

In accordance with this invention, a new class of antibodies have been produced which are substantially selectively reactive to 5-FU so as to bind to 5-FU without any substantial cross reactivity to tegafur, as well as, to other interfering pyrimidine bases, uracil and cytosine. By selectively reactive it is meant that this antibody reacts with the 5-FU molecule and does not substantially react with the other interfering pyrimidine bases such as analogues of 5-FU, the most important blocking pyrimidine base being tegafur. By providing an antibody that does not substantially cross-react with tegafur, allows one to provide an immunoassay for 5-FU which can accurately monitor levels of 5-FU for therapeutic management of patients being treated with 5-FU.

It has been found that by using immunogens which are conjugates of an immunogenic polyamine polymer with a compound of the formula:
4 X - (1')P Z F
N

H II-A
wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a polyamine polymer; and p is an integer from o to 1.

produce antibodies which are specific for 5-FU and do not substantially react with or bind to tegafur, as well as other pyrimidine bases such as uracil, and cytosine. The provision of these antibodies which substantially selectively react with 5-FU
and do not cross react with tegafur allows one to produce an immunoassay which can specifically 1o detect and monitor 5-FU in the fluid samples of patients being treated with
5-FU. Also included within this invention are reagents and kits for said immunoassay. The presence of tegafur is the major cause for false positive readings which have made immunoassays for 5-FU unsuitable.

DETAILED DESCRIPTION

In accordance with this invention, a new class of antibodies is provided which substantially selectively reacts with 5-FU and does not substantially react or cross react with tegafur, as well as other interfering pyrimidine bases such as uracil, and cytosine.
It has been discovered that through the use of the 3-substituted 5-FU
derivative of formula II-A as an immunogen, this new class of antibodies of this invention are provided. It is through the use of these antibodies that an immunoassay, including reagents and kits for such immunoassay for detecting and/or quantifying 5-FU
in blood, plasma or other body fluid samples has been developed. By use of this immunoassay, the presence and amount of 5-FU in body fluid samples, preferably a blood or plasma sample, can be detected and/or quantified. In this manner, a patient being treated with 5-FU can be monitored during therapy and his treatment adjusted in accordance with said monitoring. By means of this invention one achieves the therapeutic drug management of 5-FU in cancer patients being treated with 5-FU as a chemotherapeutic agent.

1o The reagents utilized in the immunoassay of this invention are conjugates of a carrier with the i-substituted 5-FU compound of formula II-B:

H F
\
N

( O N
I
CH,,--- (Y)P X 11-B
wherein p, X and Y are as above;

or a compound of the formula
6 N
X - (Y)p :"'' F

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a polyamine polymer;

p is an integer from o to i.;
or mixtures thereof.

In the reagents of formula II-A and II-B, the carrier can be any of the conventional reagents carriers utilized in carrying out immunoassays, preferably these carriers are labeled for detection. In the compound of formula II-A, which are utilized in forming io the reagents used in the assay, X can be any functional group capable of bonding to the carrier. The preferred carriers contain a polymeric polyamine polymer with a reactive amino group and X is a terminal functional group capable of binding to a polyamine polymer.

In the immunoassay of this invention, these conjugates are competitive binding partners with the 5-FU present in the sample for the binding with the antibodies of this invention. Therefore, the amount of conjugate reagent which binds to the antibody will be inversely proportional to the amount of 5-FU in the sample. In accordance with this invention, the assay utilizes any conventional measuring means for detecting and measuring the amount of said conjugate which is bound or unbound to the antibody.
7
8 PCT/US2006/003674 Through the use of said means, the amount of the bound or unbound conjugate can be determined. Generally, the amount of 5-FU in a sample is determined by correlating the measured amount of the bound or unbound conjugate produced by the 5-FU in the sample with values of the bound or unbound conjugate determined from standard or calibration curve samples containing known amounts of 5-FU, which known amounts are in the range expected for the sample to be tested. These studies for producing calibration curves are determined using the same immunoassay procedure as used for the sample.

The conjugates are prepared from compounds of formulae II-A and II-B whereas io immunogens are prepared from compounds of the formula II-A. In performing the immunoassay in accordance with this invention, it is important that the conjugate be formed from the compound of formulae II-A or II-B and the immunogen be formed from the compound of formula II-A. In the conjugates including the immunogens, the polyamine polymer is conjugated with the ligand portion which has the formula:

X- (1')r ~a F
N I

wherein Y and p are as above; and X' is -CH2- or a functional linking group;

on with the ligand portion of the compound of formula II-B which has the formula:

H\ F
N

I
O N
I
CHr- (Y)P X' III-B
wherein X', Y and p are as above.

These ligand portions may be in one or more active sites on the carrier of the conjugate.
Definitions Throughout this description the following definitions are to be understood:

The terms "immunogen" and "immunogenic" refer to substances capable of eliciting, producing, or generating an immune response in an organism.

The term "conjugate" refers to any substance formed from the joining together of two parts. Representative conjugates in accordance with the present invention include those 1o formed by the joining together of a small molecule, such as the compound of formula II-B, and a large molecule, such as a carrier or a polyamine polymer, particularly protein.
In the conjugate the small molecule may be joined at one or more active sites on the large molecule.

"Haptens" are partial or incomplete antigens. They are protein-free substances, mostly low molecular weight substances, which are not capable of stimulating antibody formation, but which do react with antibodies. The latter are formed by coupling a hapten to a high molecular weight immunogenic carrier and then injecting this coupled
9 product, i.e., immunogen, into a human or animal subject. The hapten of this invention is 5-FU.

As used herein, a "spacing group" or "spacer" refers to a portion of a chemical structure which connects two or more substructures such as haptens, carriers, immunogens, labels, or tracers through a CH2 or functional linking group. These spacer groups will be enumerated hereinafter in this application. The atoms of a spacing group and the atoms of a chain within the spacing group are themselves connected by chemical bonds.
Among the preferred spacers are straight or branched, saturated or unsaturated, carbon chains. Theses carbon chains may also include one or more heteroatoms within the 1o chain or at termini of the chains. By "heteroatoms" is meant atoms other than carbon which are chosen from the group consisting of oxygen, nitrogen and sulfur.
Spacing groups may also include cyclic or aromatic groups as part of the chain or as a substitution on one of the atoms in the chain.

The number of atoms in the spacing group is determined by counting the atoms other than hydrogen. The number of atoms in a chain within a spacing group is determined by counting the number of atoms other than hydrogen along the shortest route between the substructures being connected. A functional linking group may be used to activate, e.g., provide an available functional site on, a hapten or spacing group for synthesizing a conjugate of a hapten with a label or carrier or polyamine polymer.

2o An "immunogenic carrier," as the terms are used herein, is an immunogenic substance, commonly a protein, that can join with a hapten, in this case 5-FU or the 5-FU
derivatives of formula II-A described, thereby enabling these hapten derivatives to induce an immune response and elicit the production of antibodies that can bind specifically with these haptens. The immunogenic carriers and the linking groups will be enumerated hereinafter in this application. Among the immunogenic carrier substances are included proteins, glycoproteins, complex polyamino-polysaccharides, particles, and nucleic acids that are recognized as foreign and thereby elicit an immunologic response from the host. The polyamino- polysaccharides may be prepared from polysaccharides using any of the conventional means known for this preparation.
Also various protein types may be employed as a poly(amino acid) immunogenic carrier.
These types include albumins, serum proteins, lipoproteins, etc. Illustrative proteins include bovine serum albumin (BSA), keyhole limpet hemocyanin (KLH), egg io ovalbumin, bovine thyroglobulin (BTG) etc. Alternatively, synthetic poly(amino acids) may be utilized.

Immunogenic carriers can also include poly amino-polysaccharides, which are a high molecular weight polymer built up by repeated condensations of monosaccharides.
Examples of polysaccharides are starches, glycogen, cellulose, carbohydrate gums such as gum arabic, agar, and so forth. The polysaccharides also contain polyamino acid residues and/or lipid residues.

The immunogenic carrier can also be a poly(nucleic acid) either alone or conjugated to one of the above mentioned poly(amino acids) or polysaccharides.

The immunogenic carrier can also include solid particles. The particles are generally at least about 0.02 microns ( m) and not more than about 1oo m, and usually about 0.05 m to 1o m in diameter. The particle can be organic or inorganic, swellable or non-swellable, porous or non-porous, optimally of a density approximating water, generally from about 0.7 to 1.5 g/mL, and composed of material that can be transparent, partially transparent, or opaque. The particles can be biological materials such as cells and microorganisms, including non-limiting examples such as erythrocytes, leukocytes, lymphocytes, hybridomas, Streptococcus, Staphylococcus aureus, E. co1i, and viruses.
The particles can also be comprised of organic and inorganic polymers, liposomes, latex, phospholipid vesicles, or lipoproteins.

"Poly(amino acid)" or "polypeptide" is a polyamide formed from amino acids.
Poly(amino acids) will generally range from about 2,000 molecular weight, having no upper molecular weight limit, normally being less than io,ooo,ooo and usually not more than about 6oo,ooo daltons. There will usually be different ranges, depending on 1o whether an immunogenic carrier or an enzyme is involved.

A "peptide" is any compound formed by the linkage of two or more amino acids by amide (peptide) bonds, usually a polymer of a-amino acids in which the a-amino group of each amino acid residue (except the NH2 terminus) is linked to the a-carboxyl group of the next residue in a linear chain. The terms peptide, polypeptide and poly(amino acid) are used synonymously herein to refer to this class of compounds without restriction as to size. The largest members of this class are referred to as proteins.

A "label," "detector molecule," or "tracer" is any molecule which produces, or can be induced to produce, a detectable signal. The label can be conjugated to an analyte, immunogen, antibody, or to another molecule such as a receptor or a molecule that can 2o bind to a receptor such as a ligand, particularly a hapten. Non-limiting examples of labels include radioactive isotopes, enzymes, enzyme fragments, enzyme substrates, enzyme inhibitors, coenzymes, catalysts, fluorophores, dyes, chemiluminescers, luminescers, or sensitizers; a non-magnetic or magnetic particle, a solid support, a liposome, a ligand, or a receptor.

The term "antibody" refers to a specific protein binding partner for an antigen and is any substance, or group of substances, which has a specific binding affinity for an antigen to the exclusion of other substances. The generic term antibody subsumes polyclonal antibodies, monoclonal antibodies and antibody fragments.

The term "derivative" refers to a chemical compound or molecule made from a parent compound by one or more chemical reactions.

The term "carrier" for forming the conjugate of formula II-B refers to solid particles io and/or polymeric polymers such as immunogenic polymers such as those mentioned above. Where the carrier is a solid particle, the solid particle may be bound, coated with or otherwise attached to a polyamine polymer to provide one or more reactive sites for bonding to the terminal functional group X in the compounds of the formula II-B.

The term "reagent kit," or "test kit," refers to an assembly of materials that are used in performing an assay. The reagents can be provided in packaged combination in the same or in separate containers, depending on their cross-reactivities and stabilities, and in liquid or in lyophilized form. The amounts and proportions of reagents provided in the kit can be selected so as to provide optimum results for a particular application. A
reagent kit embodying features of the present invention comprises antibodies specific for 5-FU. The kit may further comprise ligands of the analyte and calibration and control materials. The reagents may remain in liquid form or may be lyophilized.
The phrase "calibration and control materials" refers to any standard or reference material containing a known amount of a drug to be measured. The concentration of drug is calculated by comparing the results obtained for the unknown specimen with the results obtained for the standard. This is commonly done by constructing a calibration curve.

The term "biological sample" includes, but is not limited to, any quantity of a substance from a living thing or formerly living thing. Such living things include, but are not limited to, humans, mice, monkeys, rats, rabbits, horses, and other animals.
Such substances include, but are not limited to, blood, serum, plasma, urine, cells, organs, tissues, bone, bone marrow, lymph, lymph nodes, synovial tissue, chondrocytes, synovial macrophages, endothelial cells, and skin.

Reagents and Immunogens In constructing an immunoassay, a conjugate of 5-FU is constructed to compete with the 5-FU in the sample for binding sites on the antibodies of this invention. In the immunoassay of this invention, the immunogen for producing the antibodies of this invention is the 3-substituted 5-FU derivatives of the compounds of formula III-A and the reagent is the i-substituted 5-FU derivatives of formulae III-A or III-B.
In the compounds of formula III-A and III-B, the linker spacer constitutes the -CH2_(Y)P _X'-portion of this molecule . The linker X' and the spacer - CH2_(Y)p - are conventional in preparing conjugates and immunogens. Any of the conventional spacer-linking groups utilized to prepare conjugates and immunogens for immunoassays can be utilized in the compounds of formula III-A and III-B. Such conventional linkers and spacers are disclosed in U.S. Patent 5,501,987 and U.S. Patent 5,101,015.

Among the preferred spacer groups are included the spacer groups hereinbefore mentioned. Particularly preferred spacing groups are groups such as alkylene 14.

containing from 1 to 1o carbon atoms, -(CH2)m-i-NH-(CH2)n - ; (CH2)o-, -NH-(CH2)m' O or (CH2)o-wherein n and o are integers from o to 6, and m is an integer from 1 to 6 with alkylene being the especially preferred spacing group. With respect to the above structures represented by Y, terminal functional group X, is connected to these substituents at their terminal and on the right side of the structures, i.e., a the end designated by (CH2)0 or (CH2)m=

In the compounds of formula III-A and III-B, X' is -CH2- or a functional group linking 1o the spacer, preferably to an amine group on the polymer or the carrier. The group X' is the result of the terminal functional group X in the compounds of Formula II A
and II-B
which is capable of binding to the amino group in the polyamine polymer used as either the carrier or the immunogen. Any terminal functional group capable of reacting with an amine can be utilized as the functional group X in the compounds of formula II-A

and II-B. These terminal functional groups preferably included within X are:
-Ii-OR3 , -N =C =R4, or 11 H

wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur. The radical -N --C =R4 1 can be an isocyanate or as isothiocyanate. The active esters formed by OR3 include imidoester, such as N-hydroxysuccinamide,l-hydroxy benzotriazole and p-nitrophenyl ester.
However any active ester which can react with an amine group can be used.

The carboxylic group and the active esters are coupled to the carrier or immunogenic polymer by conventional means. The amine group on the polyamine polymer, such as a protein, produces an amide group which connects the spacer to the polymer, immunogens or carrier and/or conjugates of this invention.

In the immunogens and conjugates of the present invention, the chemical bonds between the carboxyl group-containing 5-FU hapten and the amino groups on the 1o polyamine polymer on the carrier or the immunogen can be established using a variety of methods known to one skilled in the art. It is frequently preferable to form amide bonds. Amide bonds are formed by first activating the carboxylic acid moiety of the 5-FU hapten in the compounds of formula II-A and II-B by reacting the carboxyl group with a leaving group reagent (e.g., N-hydroxysuccinimide, 1-hydroxybenzotriazole, p-nitrophenol and the like). An activating reagent such as dicyclohexylcarbodiimide, diisopropylcarbodiimide and the like can be used. The activated form of the carboxyl group in the 5-FU hapten of formula II-A or II-B is then reacted with a buffered solution containing the protein carrier.

In cases where the 5-FU derivative of formula II-A or II-B contains a primary or secondary amino group as well as the carboxyl group, it is necessary to use an amine protecting group during the activation and coupling reactions to prevent the conjugates from reacting with themselves. Typically, the amines on the conjugate are protected by forming the corresponding N-trifluoroacetamide, N-tertbutyloxycarbonyl urethane (N-t-BOC urethane), N-carbobenzyloxy urethane or similar structure. Once the coupling reaction to the immunogenic polymer or carrier has been accomplished, as described above, the amine protecting group can be removed using reagents that do not otherwise alter the structure of the immunogen or conjugate. Such reagents and methods are known to one skilled in the art and include weak or strong aqueous or anhydrous acids, weak or strong aqueous or anhydrous bases, hydride-containing reagents such as sodium borohydride or sodium cyanoborohydride and catalytic hydrogenation.
Various methods of conjugating haptens and carriers are also disclosed in U.S. Patent 3,996,344 and U.S. Patent 4,o16,146, which are herein incorporated by reference.

1o On the other hand where X is a terminal isocyanate or thioisocyanate radical in the compound of formula II-A or II-B, these radicals when reacted with the free amine of a polyamine polymer produce the conjugate of formula II-B or the immunogen where X' NH -il- ,',~,here R4 is as above, which functionally connects with the carrier or the is immunogenic polypeptide.

Where X, in the compounds of formula II-A and II-B, is an aldehyde group these 20 compounds may be connected to the amine group of the polyamine polypeptide or carrier through an amine linkage by reductive amination. Any conventional method of condensing an aldehyde with an amine such as through reductive amination can be used to form this linkage. In this case, X' in the ligand portions of formula III-A
and Ill-B is -CFi2-.

25 The 1-nitrogen atoms in the compound of formula I can be connected to form the compound of formula II-B by reacting 5-FU with a halide of the formula:

Ri-CH2-(Y)P-X V-A

where Rl is chloro or bromo and Y, p and X are as above, to produce the compound of the formula:

H F
N

I
O N

CHZ- (y'P X 11-B

The compound of formula I is reacted at its 1-ring nitrogen atom with the halide of formula V-A to form the compounds of formula II-B by any conventional means of condensing a halide with an amine group. This condensation reaction is carried out in the presence of a base. In this reaction, the ring nitrogen atom at the 1-position of the compound of formula I is more reactive than the ring nitrogen atom at the 3-position.

1o Therefore the ring nitrogen atom at the 1-position will preferably condense with the halide. If the compound of formula V-A contains any reactive amino or other functional substituents, these substituents can be reacted with conventional protecting groups prior to the reaction of 5-FU with a compound of V-A. After the compound of formula VI-A is produced, these protecting groups can be removed by procedures well known in the art for removing such protecting groups while retaining the amine in the compound of formula II-B.

The 3-substituted 5-FU of formula II-A can be prepared from 5-FU by first converting 5-FU into the dichloro compound of the formula:

CI

F
N

CI Z~N VII

This is accomplished by treating the compound of formula I with a chlorinating agent such as phosphorous oxychloride. Any of the conditions conventional in utilizing these chlorinating agents can be used in carrying out this reaction. In the next step, the compound of formula VII is converted to the compound of the formula:

OH
N F
I
VIII-A
CI N

which enolizes into the compound of formula:
O
HN F

VIII-B
CI N

This conversion is carried out by treating the compound of formula VII with sodium hydroxide in an aqueous medium at a temperature of from 35-C to 50-C. The compound of formula VIII-B can be converted to the compound of the formula:
1g \N F
I
IX

where R2 is benzyl.

In forming the compound of formula IX, the compound of formula VIII-B is reacted with benzyl alcohol in an organic solvent in the presence of solid sodium hydroxide. In the next step the compound of formula IX is converted to the compound of formula II-A
by reacting the compound of formula IX with the halide of formula V-A, in the manner described hereinbefore in connection with the condensation of compound of formula I
with the halide of formula V-A.

The compound of formulae II-A or II-B can be converted into the conjugate carrier 1o reagent of this invention by reacting these compounds with a polyamine, polypeptide or a carrier. The same polypeptide can be utilized as the carrier in the compound of formula II-B and as the immunogenic polymer in the immunogen of formula II-A
of this invention provided that polyamine or polypeptide is immunologically active.
However, to form the conjugates, these polymers need not produce an immunological response as needed for the immunogens. In accordance with this invention, the various functional groups represented by X in the compounds of formula II-A and II-B can be conjugated to the polymeric material by conventional means of attaching a functional group to an amine group contained within the polymer. In accordance with a preferred embodiment, in the compound of formula II-A and II-B, X is a carboxylic acid group or 2o active esters thereof.

ANTIBODIES
The present invention also relates to novel antibodies including monoclonal antibodies to 5-FU produced by utilizing the aforementioned immunogens. In accordance with this invention it has been found that these antibodies produced in accordance with this invention are selectively reactive with 5-FU and do not react with tegafur or other pyrimidine containing compounds which would interfere with immunoassays for 5-FU.
The present invention relates to novel antibodies and monoclonal antibodies to 5-FU.
The antisera of the invention can be conveniently produced by immunizing host animals with the immunogens this invention. Suitable host animals include rodents, such as, for 1o example, mice, rats, rabbits, guinea pigs and the like, or higher mammals such as goats, sheep, horses and the like. Initial doses, bleedings and booster shots can be given according to accepted protocols for eliciting immune responses in animals, e.g., in a preferred embodiment mice received an initial dose of 1oo ug immunogen /mouse, i.p.
and two or more subsequent booster shots of 1oo ug immunogen/mouse over a six month period. Through periodic bleeding, the blood samples of the immunized mice were observed to develop an immune response against 5-FU binding utilizing conventional immunoassays. These methods provide a convenient way to screen for hosts which are producing antisera having the desired activity.

Monoclonal antibodies are produced conveniently by immunizing Balb/c mice 2o according to the above schedule followed by injecting the mice with 1oo ug immunogen i.p. or i.v. on three successive days starting three days prior to the cell fusion. Other protocols well known in the antibody art may of course be utilized as well.
The complete immunization protocol detailed herein provided an optimum protocol for serum antibody response for the antibody to 5-FU.

B lymphocytes obtained from the spleen, peripheral blood, lymph nodes or other tissue of the host may be used as the monoclonal antibody producing cell. Most preferred are B lymphocytes obtained from the spleen. Hybridomas capable of generating the desired monoclonal antibodies of the invention are obtained by fusing such B
lymphocytes with an immortal cell line, which is a cell line that which imparts long term tissue culture stability on the hybrid cell. In the preferred embodiment of the invention the immortal cell may be a lymphoblastoid cell or a plasmacytoma cell such as a myeloma cell, itself 1o an antibody producing cell but also malignant. Murine hybridomas which produce 5-FU
monoclonal antibodies are formed by the fusion of mouse myeloma cells and spleen cells from mice immunized against 5-FU-protein conjugates. Chimeric and humanized monoclonal antibodies can be produced by cloning the antibody expressing genes from the hybridoma cells and employing recombinant DNA methods now well known in the art to either join the subsequence of the mouse variable region to human constant regions or to combine human framework regions with complementary determining regions (CDR's) from a donor mouse or rat immunoglobulin. An improved method for carrying out humanization of murine monoclonal antibodies which provides antibodies of enhanced affinities is set forth in International Patent Application WO
92/11018.

Polypeptide fragments comprising only a portion of the primary antibody structure may be produced, which fragments possess one or more immunoglobulin activities.
These polypeptide fragments may be produced by proteolytic cleavage of intact antibodies by methods well known in the art, or by inserting stop codons at the desired locations in expression vectors containing the antibody genes using site-directed mutageneses to produce Fab fragments or (Fab')2 fragments. Single chain antibodies maybe produced by joining VL and VH regions with a DNA linker (see Huston et al., Proc. Nat1.
Acad.
Sci. U.SA., 85:5879-5883 (1988) and Bird et al., Science, 242:423-426 (1988)) The antibodies of this invention are selective for 5-FU and do not have any substantial cross-reactivity with such pyrimidine bases such as uracil , cytosine, tegafur etc. By having no substantial cross-reactivity it is meant that the antibodies of this invention have a cross reactivity relative to 5-FU with these metabolites of not greater than 12%
preferably less than 5 % .

IMMUNOASSAYS
In accordance with this invention, the aforementioned conjugates and the antibodies generated from the immunogens of these compounds of formula II-A can be utilized as reagents for the determination of 5-FU in patient samples. This determination is performed by means of an immunoassay. Any immunoassay in which the reagent conjugates formed from the compounds of formula II-B compete with the 5-FU in the sample for binding sites on the antibodies generated in accordance with this invention can be utilized to determine the presence of 5-FU in a patient sample. The manner for conducting such an assay for 5-FU in a sample suspected of containing 5-FU, comprises lo combining an (a) aqueous medium sample, (b) an antibody to 5-FU generated in accordance with this invention and (c) the conjugates formed from the compounds of formulae II-A and II-B. The amount of 5-FU in the sample can be determined by measuring the inhibition of the binding to the specific antibody of a known amount of the conjugate added to the mixture of the sample and antibody. The result of the inhibition of such binding of the known amount of conjugates by the unknown sample is compared to the results obtained in the same assay by utilizing known standard solutions of 5-FU. In determining the amount of 5-FU in an unknown sample, the sample, the conjugates formed from the compounds of formula II-B and the antibody may be added in any order.

Various means can be utilized to measure the amount of conjugate formed from the compounds of formulae II-A and II-B bound to the antibody. One method is where binding of the conjugates to the antibody causes a decrease in the rate of rotation of a fluorophore conjugate. The amount of decrease in the rate of rotation of a fluorophore conjugate in the liquid mixture can be detected by the fluorescent polarization technique such as disclosed in U.S. Patent 4,269,511 and U.S. Patent 4,420,568.

On the other hand, the antibody can be coated or absorbed on nanoparticles so that when these particles react with the 5-FU conjugates formed from the compounds of formulae II-A and II-B, these nanoparticles form an aggregate. However, when the antibody coated or absorbed nanoparticles react with the 5-FU in the sample, the 5-FU
from the sample bound to these nanoparticles does not cause aggregation of the antibody nanoparticles. The amount of aggregation or agglutination can be measured in the assay mixture by absorbance.

1o On the other hand, these assays can be carried out by having either the antibody or the 5-FU conjugates attached to a solid support such as a microtiter plate or any other conventional solid support including solid particles. Attaching antibodies and proteins to such solid particles is well known in the art. Any conventional method can be utilized for carrying out such attachments. In many cases, in order to aid measurement, labels may be placed upon the antibodies, conjugates or solid particles , such as radioactive labels or enzyme labels, as aids in detecting the amount of the conjugates formed from the compounds of formula II-B which is bound or unbound with the antibody.
Other suitable labels include chromophores, fluorophores, etc.

As a matter of convenience, assay components of the present invention can be provided in a kit, a packaged combination with predetermined amounts of new reagents employed in assaying for 5-FU. These reagents include the antibody of this invention, as well as, the conjugates formed from the compounds of formulae II-A and II-B.

In addition to these necessary reagents, additives such as ancillary reagents may be included, for example, stabilizers, buffers and the like. The relative amounts of the various reagents may vary widely to provide for concentrations in solution of the reagents which substantially optimize the sensitivity of the assay. Reagents can be provided in solution or as a dry powder, usually lyophilized, including excipients which on dissolution will provide for a reagent solution having the appropriate concentrations for performing the assay.

EXAMPLES
In the examples, the following abbreviations are used for designating the following:
THF Tetrahydrofuran EA Ethyl alcohol DCM Dichloromethane DMAP Dimethylaminopyridine NHS N-hydroxy-succinimide EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride TLC Thin Layer Chromatrography ANS 8-Anilino-i-naphthalenesulfonic acid i.p. Intraperitoneal HRP Horse radish-peroxidase TMB 3,3',5,5'-Tetramethylbenzidine TRIS Tris(hydroxymethyl)aminomethane hydrochloride BSA Bovine serum albumin BTG Bovine thyroglobulin PBS Phosphate buffered saline di deionized water In the examples, Scheme 1, Scheme 2, Schemes 3a, 3b and Scheme 4, below set forth the specific compounds prepared and referred to by numbers in the Examples. The schemes are as follows:

Scheme 1 F
H\ F H I
N I

O~N O OIN
H Br~OEt OEt H~ F
N I HI F
O~N ON
O
4 o N 3 3 oH

Scheme 2 O
H~N I F _ H\ F
HOOC N
~ ~ I
N NH2 ~
O N COOH
NHS N
O

Scheme 3a I F Ci H\N ~ N~ F ~
O N ~
H CI N

N F H, N F
~ i O
CI N BnO N Br~OEt EtO~N F EtO~N I F
O ~ I
BnO N O N
H

0 0 p ~N-o -tr N---~N F Hp~N F
O p O
O N O N

_ 12 1 HOOC ~ ~

HOOC (:) N~N F
~ ~

H

Scheme 3b HiN F F
~
/ I
Bn0I H COOC ~Br 3 H3COOC \ NBn0" N

y F N F
lob ON ~N HOOC O~~N
I I
H H

Scheme 4 _ HOOC / 0 F HOOC \~ \ I N
SHN
N NH2 -n-"~~N F
0 0 O~N I Na2CO3/THF 2b 0 N
I I
2a H Step 9 H
EDC/NHS
Step 10 \ N\~'"~N F
~O I
2c O N
H

Example i Scheme i., Preparation of i-substituted 5-FU activated ester [4]
To a solution of Fluorouracil (50 g) [i.] in DMF (1oo mL), triethylamine (78 g) was added at 300C while stirring. Then ethy-4-bromobutyrate (88.5 g) was added drop wise.

After the addition was completed, the resulting reaction mixture was stirred for 48 hours at room temperature. The reaction mixture was filtered and the solvent was removed under reduced pressure. The residue was crystallized in ethyl acetate to afford 26 g (29%) of compound [2].

To a solution of [2] (20 g) in methanol (loo mL), 20% of potassium hydroxide aqueous solution (27 mL) was added. The resulting solution was stirred at room temperature for 3 hours, and then the mixture was concentrated under reduced pressure. The residue was dissolved in acetone (50-100 mL) and adjusted to pH 2-3 with concentrated HCl. It was then filtered and washed with acetone. The solid product was dissolved in acetone (50 mL) by heating. After cooling to room temperature, the solid was precipitated out by adding ethyl acetate (ioo mL). The solid product was collected by filtration, followed by drying to afford about 10 g of [3]. The TLC condition for ester was ethyl acetate: ether (3:1). The TLC condition for acid is chloroform : methanol (15 : 1) with 2 drops of acetic acid.

To 6.3 g of compound 131 in 6oo mL of dichloromethane at ooC, NHS was added.
To this a solution of DCC 48 g) in dichloromethane was added drop wise. After stirring for 2 hours at ooC, the resulting reaction mixture was stirred at room temperature for 15 hours. The reaction mixture was concentrated. The residue was crystallized in acetone to give crude product. The crude product was purified on a silica gel column (eluted with ethyl acetate:ether, 3: i) to provide 4 g of compound [41.

Example 2 Scheme 2, Preparation of i-substituted 5-FU acid [5]

To a solution of compound [4] (3.2 g) in acetonitrile (300 mL), water (9oo mL) was added, followed by addition of 1.2 eq. of p-methylamino-benzoic acid. The resulting reaction mixture was stirred at room temperature for 2o hours. The mixture was concentrated under reduced pressure to remove acetonitrile. A precipitate formed and was collected by filtration. It was then crystallized in acetone to give 2.8 g of crude 1o product. The crude product was purified on a silica gel column (eluted with chloroform :
methano1,15 : 1 with 1-2 drops of acetic acid) to afford 2 g of compound [5].

Example 3a Scheme 3a, Preparation of 3-substituted 5-FU acid derivatives [12], [14]
A mixture of 15.6 g of 5-FU [i] in 8o mL of POC13 was stirred in a three neck-flask equipped with condenser, thermometer and dropping funnel at 4ooC. After addition of mL of N,N dimethylaniline drop wise, the resulting mixture was heated to reflux for 3 hours. The excess of POC13 was evaporated under reduced pressure. The mixture was cooled to room temperature and poured into 75 g of crushed ice. It was then extracted with chloroform (50 mL three times). The combined extracts were washed with water, 2o dried with MgSO4, and concentrated to give a yellowish solid of compound [7] in about 50% yield.

A mixture of 16 g of oxychloride [7] in 48 mL solution of 2 N NaOH was stirred at 450C
for one hour. The pH of the reaction mixture was reduced to 7. Another 48 mL
solution of 2 N NaOH was added and continued to stir until no more oily materials were observed in the reaction mixture. After the mixture was cooled to room temperature, the pH was adjusted to pH 3 with concentrated HCl. It was cooled and the product [8]
precipitated out. Compound oxychloride [8] was collected and washed with water until the washing solution became neutral. The yield was about 55%.

To a three-neck flask equipped with condenser, thermometer and dean stark apparatus was added 20 mL of toluene, 52 mL of benzyl alcohol and 2.44 g of solid NaOH.
The resulting mixture was refluxed until dry. Then, 3 g of Compound [8] was added and continued to be refluxed for 3 hours. After the reaction mixture was cooled to room 1o temperature, 50 mL of water was added. The organic phase was washed with water twice (50 mL each). The aqueous phases were combined and the residues of toluene and benzyl alcohol were removed under reduced pressure. The solution was adjusted to pH 3 with concentrated HCl, cooled down, and a precipitate formed, which was collected. It was re-crystallized in ethanol to afford compound [9] in about 6o% yield.

After a mixture of 20 mL of benzene, 20 mL of water and 0.5 g of tetrabutylammonium bromide was heated to 550C, solution A (2g of [91 in 20 mL of 1 N NaOH aqueous solution) and solution B (1.9 g of ethyl 4-bromobutyrate in 20 mL of benzene) were added into the mixture alternatively in drop wise fashion. The pH of the reaction mixture was controlled between pH 8 - lo. After the addition was completed, the 2o reaction mixture was refluxed for two and a half hours. The organic phase was separated, washed with a 5% NaOH and water and dried with MgSO4. The organic solvent was removed under reduced pressure. The residue was purified on a silica gel column (eluted with ether and ethyl acetate, io:1) to give compound [so] as an oily product in about 40% yield.

A mixture of 3 g of compound [:to], 0.3 g of 1o% Pd/C in 50 mL of methanol was stirred under hydrogen gas (15 psi) for about 24 hours. The catalyst was removed by filtration.
To the filtrate containing [io], 2 g of NaOH and 50 mL of water were added.
The resulting mixture was stirred for 8 hours at room temperature. The methanol was removed under reduced pressure. The mixture was adjusted to pH 3 with concentrated HCl. After cooling, a precipitate was formed and collected by filtration. The precipitate was re-crystallized from ethanol to afford [12] in about 50% yield.

1o A mixture of i. g of dried [12], o.74 g of NHS, 1.47 g of DCC in 50 mL of chloroform was stirred for overnight (about 24 hours) at room temperature. The solvent was removed under reduced pressure and the residue was purified on a silica gel column (eluted with ethyl acetate and methanol,lo:1) to afford compound [13] in about 40% yield.

A mixture of 1 g of compound [13], 0.5 g4-(aminomethyl)benzoic acid in 30 mL
of DMF was stirred at room temperature for 8 hours. A portion of 150 mL water was added to the reaction mixture. The resulting mixture was washed with 1oo mL of ethyl acetate.
The aqueous phase was allowed to stand at 40C and the product [141 slowly precipitated out of the solution, was collected by filtration and dried under vacuum at room temperature in the presence of Pz05 to yield about 65% of [141.
Example 3b Scheme 3b, Preparation of 3-substituted 5-FU acid derivative [6]
After a mixture of 20 mL of benzene, 20 mL of water and 0.5 g of tetrabutylammonium bromide was heated to 550C, solution A (2g of L91 in 20 mL of 1 N NaOH aqueous solution) and solution B(2.05 g of ethyl 4-bromobutyrate in 20 mL of benzene) were added into the mixture simultaneously. The pH of the reaction mixture was controlled between pH 8-1o. After the addition was completed, the reaction mixture was refluxed for two and a half hours. The organic phase was separated, washed with a 5%
NaOH and water and dried with MgSO4. The organic solvent was removed under reduced pressure.
The residue was purified on a silica gel column (eluted with ether and ethyl acetate, 1o:1) to give compound [15] as an oily product in about 38% yield.

A mixture of 2 g of compound [15], 0.2 g of 10% Pd/C in 6o mL of methanol was stirred under hydrogen gas (i5 psi) for about 24 hours. The catalyst was removed by filtration.
1o The filtrate containing compound [16] was concentrated to approximately 20 mL to which was added 1 g of NaOH and 20 mL of water. The resulting mixture was stirred for 8 hours at room temperature. The methanol was removed under reduced pressure and the mixture adjusted to pH 3 with concentrated HCI. After cooling, a precipitate was formed and collected by filtration. The precipitate was re-crystallized from ethanol to afford [6] in about 6o% yield.

Example 4 General method for preparing NHS activated esters from the corresponding acids [3, 5,12,14, 6]

To a stirred solution of NHS (1.39 mmol) in 20 mL of dry CH2C12 the acid (o.695 mmol) [3~ 5~ 12, 14 or 6] and EDC (2.o85 mmol) were added. The solution was stirred for 18 hours at room temperature under a nitrogen atmosphere. The reaction was quenched by the addition of 3 mL of hydrochloric acid (0.3 N) and stirred for an additional 5 minutes. The organic layer was separated, dried (Na2SO4), filtered and evaporated (under vacuum) to yield a white solid.

Example 5 Preparation of i-substituted 5-FU KLH Immunogen To 5.86 mL of KLH (31.2 mg/mL) in 50 mM phosphate buffer (50 mM, pH 7.5) o.692 mL of compound 141 (12.8 mg/mL in DMSO), that was prepared in Example 1, was added drop wise and the pH was adjusted to 8.5. The mixture was allowed to stir 18 hours at room temperature. This immunogenic conjugate was then purified by dialysis 1o and characterized according to procedures described previously (Wu et. al., Bioconj.
Chem., 8: pp 385-390,1997a Li et al., Bioconj. Chem., 8: pp 896-9o5a 1997a Salamone et al., J. Forensic Sci. pp 821-826,1998)=

Example 6a Preparation of 3-substituted 5-FU BTG Immunogen To 11.4 mL of BTG (16.9 mg/mL) in 50 mM phosphate buffer (5o mM, pH 7.5) 1.2 mL of DMSO was added drop wise and the pH was checked to be at 7.5. To this 0.277 mL
of compound [131 (52.5 mg/mL in DMSO), that was prepared in Example 3a, was added drop wise and the pH was again checked to be 7.5. The mixture was allowed to stir 18 hours at room temperature. This immunogenic conjugate was then purified by dialysis 2o and characterized according to procedures described previously (Wu et. al., Bioconj.
Chem., 8: pp 385-39oa 1997, Li et al., Bioconj. Chem., 8: pp 896-905,1997a Salamone et al., J. Forensic Sci. pp 821-826,1998)=

Example 6b Preparation of 3-substituted 5-FU KLH Immunogen To 8.3 mL of KLH (24.9 mg/mL) in 50 mM phosphate buffer (50 mM, pH 7.5) 0.922 mL of DMSO was added drop wise and the pH was checked to be at 7.5. To this 0.277 mL of compound [13] (52.6 mg/mL in DMSO), that was prepared in Example 3a, was added drop wise and the pH was again checked to be 7.5. The mixture was allowed to stir i8 hours at room temperature. This immunogenic conjugate was then purified by dialysis and characterized according to procedures described previously (Wu et. al., Bioconj. Chem., 8: pp 385-39oa 1997, Li et al., Bioconj. Chem., 8: pp 896-9o5a 1997a 1o Salamone et al., J. Forensic Sci. pp 821-826, 1998).
Example 7a Preparation of 3-substituted 5-FU BSA conjugate (i.o:i ratio) with Derivative 12 To a 1 mL solution of BSA (50 mg/mL) in 50 mM phosphate buffer (50 mM, pH 7.5) o.111 mL of DMSO was added drop wise. The activated N-Hydroxysuccinimide ester of compound [12] prepared as in example 4(0.045 mL of a 52.5 mg/mL in DMSO
solution) was added drop wise. The mixture was allowed to stir overnight at room temperature to produce the conjugate of the 3-substituted 5-FU and BSA. This conjugate was then purified by dialysis and characterized according to procedures 2o described previously (Wu et al., Bioconj. Chem., 8: pp 385-390, 1997, Li et al., Bioconj.
Chem., 8: pp 896-905, 1997, Salamone et al., J. Forensic Sci. pp 821-826,1998)=

Example 7b Preparation of 3-substituted 5-FU BSA conjugate (I:iL ratio) with Derivative To 20.0 mL of BSA (50.0 mg/mL) in 50 mM phosphate buffer (50 mM, pH 7.5) 2.222 mL of DMSO was added drop wise and the pH was checked to be at 7.5. To this 0.272 mL of the activated N-Hydroxysuccinimide ester of compound [6] (20.0 mg/mL in DMSO), that was prepared in Example 4, was added drop wise and the pH was again checked to be 7.5. The mixture was allowed to stir 18 hours at room temperature. This immunogenic conjugate was then purified by dialysis and characterized according to 1o procedures described previously (Wu et. al., Bioconj. Chem., 8: pp 385-39oa 1997, Li et al., Bioconj. Chem., 8: pp 896-905,1997a Salamone et al., J. Forensic Sci. pp 821-826, 1998).

Example 8 Preparation of i-substituted 5-FU BSA conjugate (20:1 ratio) with Derivative 5 To a i4 mL solution of BSA (5o mg/mL) in 50 mM phosphate buffer (50 mM, pH
7.5) in an ice bath 14 mL of DMSO was added drop wise. The activated N-Hydroxysuccinimide ester of compound [5] prepared as in example 4 (1.65 mL of a 57 mg/mL in DMSO
solution) was added drop wise. The mixture was allowed to stir overnight at room temperature to produce the conjugate of the i-substituted 5-FU and BSA. This conjugate was then purified by dialysis and characterized according to procedures described previously (Wu et. al., Bioconj. Chem., 8: pp 385-390,1997, Li et al., Bioconj. Chem., 8:
pp 896-905a 1997a Salamone et al., J. Forensic Sci. pp 821-826,1998)=

Example 9 Preparation of 5-FU Antibodies Ten Female BALB/c mice were immunized i.p. with 1oo g/mouse of 5-FU-KLH
prepared in example 5 or with 5-FU-BTG prepared in example 6a, emulsified in Complete Freund's Adjuvant. Mice were boosted once four weeks after the initial injection with 1oo g /mouse of the same immunogens emulsified in Incomplete Freund's Adjuvant. Ten days after the boost test bleeds from each mouse were obtained by orbital bleed. The anti-serum from these test bleeds contained 5-FU
antibodies evaluated in Examples 12a,13, and 14. For monoclonal antibodies ten Female BALB/c 1o mice were immunized i.p. with ioo g/mouse of 3-substituted 5-FU-KLH
prepared in example 6b, emulsified in Complete Freund's Adjuvant. Mice were boosted once four weeks after the initial injection with ioo gg /mouse of the same immunogens emulsified in Incomplete Freund's Adjuvant. Ten days after the boost test bleeds from each mouse were obtained by orbital bleed and these were screened as in examples 12a and 15. To produce monoclonal antibodies starting four days before the fusion (day o), the mice were injected i.p. with 400 g (day 3), 200 g (day 2) and 200 g (day 1) 3-substituted 5-FU KLH immunogen in PBS on three successive days. Spleen cells were isolated from the selected mice and fused with 2 x 107 SP2/o cells with 50% polyethylene glycol 1500 according to the method of Coligan, J.E. et al., eds., Current Protocols in Immunology, 2.5.1 - 2.5.8, (1992), Wiley & Sons, NY. The fused cells were plated on 1o 96-well plates in DMEM/F12 supplemented with 2o Jo FetalClone I, 2% L-glutamine (1oo mM) and 2%
5oX HAT. Two weeks later, the hybridoma supernatant was assayed for the presence of anti-5-FU by ELISA (example 12b). Positive wells were expanded and again screened by the same method. The positive clones were confirmed for 5-FU binding by a competitive ELISA (examples 12a and 15). Clones positive by ELISA were subcloned once or twice by limiting dilution according to the method disclosed in Coligan, J.E. et al., eds., Current Protocols in Immunelogy, 2.5.8 - 2=5=17, (1992), Wiley &
Sons, NY.

Example io Microtiter Plate Sensitization Procedure with 5-FU Derivative 5 - BSA
Conjugate The ELISA method for measuring 5-FU concentrations was performed in polystyrene microtiter plates (Nunc MaxiSorp C8 or F8 Immunomodules) optimized for protein lo binding and containing 96 wells per plate. Each well was coated with 5-FU-BSA
conjugate (prepared as in example 8) by adding 300 gL of 5-FU-BSA conjugate at io g/mL in o.o5M sodium bicarbonate, pH=9.6, and incubating for three hours at room temperature. The wells were washed with 0.05M sodium bicarbonate, pH 9.6 and then were blocked with 400 L of 5% sucrose, 0.2% sodium caseinate solution for 30 minutes at room temperature. After removal of the post-coat solution the plates were dried at 37 C overnight.

Example iia Microtiter Plate Sensitization Procedure with 5-FU Derivative 12 - BSA
Conjugate The ELISA method for measuring 5-FU concentrations was performed in polystyrene microtiter plates (Nunc MaxiSorp C8 or F8 Immunomodules) optimized for protein binding and containing 96 wells per plate. Each well was coated with 5-FU-BSA
conjugate (prepared as in example 7a) by adding 300 gL of 5-FU-BSA conjugate at lo g/mL in o.o5M sodium bicarbonate, pH=9.6, and incubating for three hours at room temperature. The wells were washed with o.o5M sodium bicarbonate, pH 9.6 and then were blocked with 400 L of 5% sucrose, 0.2% sodium caseinate solution for 30 minutes at room temperature. After removal of the post-coat solution the plates were dried at 37 C overnight.

Example iib Microtiter Plate Sensitization Procedure with 5-FU Derivative 6- BSA
Conjugate 1o The ELISA method for measuring 5-FU concentrations was performed in polystyrene microtiter plates (Nunc MaxiSorp C8 or F8 Immunomodules) optimized for protein binding and containing 96 wells per plate. Each well was coated with 5-FU-BSA
conjugate (prepared as in example 7b) by adding 300 L of 5-FU-BSA conjugate at 1o gg/mL in o.05M sodium bicarbonate, pH=9.6, and incubating for three hours at room temperature. The wells were washed with o.05M sodium bicarbonate, pH 9.6 and then were blocked with 375 L of 5% sucrose, 0.2% sodium caseinate solution for 30 minutes at room temperature. After removal of the post-coat solution the plates were dried at 37 C overnight.

Example 12a Antibody Screening Procedure - Titer The ELISA method for screening 5-FU antibodies (produced in example 9) was performed with the microtiter plates that.were sensitized with 5-FU-BSA as described in examples 7a, 7b and 8. The antibody screening assay was performed by diluting the antisera containing 5-FU antibodies to 1:1oo,l:l,ooo,l:lo,ooo and 1:1oo,ooo in phosphate buffered saline containing o.i% BSA and o.ol% thimerosal. To each well of 5-FU-BSA sensitized wells (prepared in examples 1.1a, i1b and lo)1oo L of diluted antibody was added and incubated for 1o minutes at room temperature with shaking.
During this incubation antibody binds to the 5-FU-conjugate in the well. The wells of the plates were washed three times with 0.02 M TRIS, o.go NaCl, 0.5% Tween-8o and o.oo1% Thimerosal, pH 7.8 to remove any unbound antibody. To detect the amount of 5-FU antibody bound to the 5-FU-BSA conjugate in the wells, loo L of a goat anti-mouse antibody - HRP enzyme conjugate (Jackson Immunoresearch) diluted 1/2000 in PBS with o.i! BSA, 0.05% ANS, o.o1% thimerosal, capable of binding specifically with 1o murine immunoglobulins and producing a colored product when incubated with a substrate, were added to each well. After an incubation of 1o minutes at room temperature with shaking, during which the goat anti-mouse antibody - HRP
enzyme conjugate binds to 5-FU antibodies in the wells, the plates were again washed three times to remove unbound goat anti-mouse antibody - HRP enzyme conjugate. To develop a measurable color in the wells washing was followed by the addition of 1oo L
of TMB (TMB Liquid Substrate, Sigma or BioFx), a substrate for HRP, to develop color during a io minute incubation with shaking at room temperature. Following the incubation for color development, 50 L of stop solution (1.5% sodium fluoride in di H20) was added to each well to stop the color development and after 1o seconds of shaking the absorbance was determined at 650 nm (Molecular Devices Plate Reader).
The amount of antibody in a well was proportional to the absorbance measured and was expressed as the dilution (titer) resulting in an absorbance of 1.5. Titers were determined by graphing log antibody dilution of the antibody measured (x-axis) vs.
absorbance 65o nm (y-axis) and extrapolating the titer at an absorbance of 1.5. The titer determined the concentration (dilution) of antibody used in the indirect competitive Microtiter plate assay described in examples 13, 14 and 15.

Example 12b Antibody Screening Procedure - Monoclonal Screening The ELISA method for screening 5-FU monoclonal antibodies (produced in example 9) was performed with the microtiter plates that were sensitized with 5-FU-BSA as described in example 7b. To each well of 5-FU-BSA sensitized wells (prepared in example i1b) 50 uL phosphate buffered saline containing o.1% BSA and o.o1 10 thimerosal and then 50 gL of monoclonal culture supernatant were added and 1o incubated for lo minutes at room temperature with shaking. During this incubation antibody binds to the 5-FU-conjugate in the well. The wells of the plates were washed three times with 0.02 M TRIS, o.g% NaCl, 0.5% Tween-8o and o.ool% Thimerosal, pH
7.8 to remove any unbound antibody. To detect the amount of 5-FU antibody bound to the 5-FU-BSA conjugate in the wells, ioo gL of a goat anti-mouse antibody -HRP

enzyme conjugate (Jackson Immunoresearch) diluted to a predetermined specific activity (approximately 1/2000) in PBS with o.i% BSA, 0.05% ANS, o.o10 thimerosal, capable of binding specifically with murine immunoglobulins and producing a colored product when incubated with a substrate, were added to each well. After an incubation of 10 minutes at room temperature with shaking, during which the goat anti-mouse 2o antibody - HRP enzyme conjugate binds to 5-FU antibodies in the wells, the plates were again washed three times to remove unbound goat anti-mouse antibody - HRP
enzyme conjugate. To develop a measurable color in the wells washing was followed by the addition of 1oo L of TMB (TMB Liquid Substrate, Sigma or BioFx), a substrate for HRP, to develop color during a 1o minute incubation with shaking at room temperature.

Following the incubation for color development, 50 L of stop solution (1.5%
sodium fluoride in di H20) was added to each well to stop the color development and after 1o seconds of shaking the absorbance was determined at 65o nm (Molecular Devices Plate Reader). The amount of antibody in a well was proportional to the absorbance measured. Samples with an absorbance of greater than twice background were designated as positive.

Example 13 Indirect Competitive Microtiter Plate Immunoassay Procedure Determining IC5o and Cross-Reactivity for Antibodies to 5-fU Derivative 4 Conjugate The ELISA method for measuring 5-FU concentrations was performed with the microtiter plates that were sensitized with 5-FU-BSA described in example 7a.
5-FU, uracil, thymine, cytosine and Tegafur were diluted 1o fold in PBS over a concentration range of o.o1 to 1o,ooo ng/mL. The assay was performed by incubating 5o L of the analytes to be measured with 50 L of antibody (produced in example 9 with immunogen of example 5) diluted to a titer determined in example i2a. During the 1o minute incubation (R.T., with shaking) there is a competition of antibody binding for the 5-FU conjugate in the well and the analyte in solution. Following this incubation the wells of the plate were washed three times with 0.02 M TRIS, o.gf NaCl, 0.5%
Tween-2o 8o and o.ool0 Thimerosal, pH 7.8 to remove any material that was not bound.
To detect the amount of 5-FU antibody bound to the 5-FU-BSA conjugate in the wells, 1oo L of a goat anti-mouse antibody - HRP enzyme conjugate (Jackson Immunoresearch) diluted 1/2000 in PBS with o.1J BSA, 0.05% ANS, o.o1% thimerosal, capable of binding specifically with murine immunoglobulins and producing a colored product when incubated with a substrate, were added to each well. After an incubation of 10 minutes at room temperature with shaking, during which the goat anti-mouse antibody - HRP enzyme conjugate binds to 5-FU antibodies in the wells, the plates were again washed three times to remove unbound secondary conjugate. To develop a measurable color in the wells washing was followed by the addition of 1oo L of TMB (TMB
Liquid Substrate, Sigma or BioFx), a substrate for HRP, to develop color in a lo minute incubation with shaking at room temperature. Following the incubation for color development, 50 L of stop solution (1.5% sodium fluoride in di H2 ) was added to each well to stop the color development and after 1o seconds of shaking the absorbance was 1o determined at 65o nm (Molecular Devices Plate Reader). The amount of antibody in a well was proportional to the absorbance measured and inversely proportional to the amount of 5-FU in the sample. The absorbance of the color in the wells containing analyte was compared to that with no analyte and a standard curve was generated. The IC5o value for a given analyte was defined as the concentration of analyte that is required to inhibit 50f of the absorbance for the wells containing no analyte.
The cross-reactivity of a given analyte was calculated as the ratio of the IC5o for 5-FU to the IC5o for uracil, thymine, cytosine and Tegafur expressed as a percent. When measured with an antibody as produced in example 9 with immunogen of example 5 the percent cross-reactivities relative to 5-FU for uracil, thymine, and cytosine were less than 7%

2o and 200% for tegafur. The results are in table I.

Example 14 Indirect Competitive Microtiter Plate Immunoassay Procedure Determining IC5o and Cross-Reactivity for Antibodies to 5-FU Derivative 13 Conjugate The ELISA method for measuring 5-FU concentrations was performed with the microtiter plates that were sensitized with 5-FU-BSA described in example 8. 5-FU, uracil, thymine, cytosine and Tegafur were diluted io fold in PBS over a concentration range of o.oi to 1o,ooo ng/mL. The assay was performed by incubating 5o L of the analytes to be measured with 50 L of antibody (produced in example 9) diluted to a io titer determined in example 12a. During the 1o minute incubation (R.T., with shaking) there is a competition of antibody binding for the 5-FU conjugate in the well and the analyte in solution. Following this incubation the wells of the plate were washed three times with 0.02 M TRIS, o.9% NaCl, 0.5% Tween-8o and o.oo1 6o Thimerosal, pH
7.8 to remove any material that was not bound. To detect the amount of 5-FU antibody bound to the 5-FU-BSA conjugate in the wells,loo L of a goat anti-mouse antibody -HRP
enzyme conjugate (Jackson Immunoresearch) diluted 1/2000 in PBS with o.1 fo BSA, o.o56 ANS, o.oi% thimerosal, capable of binding specifically with murine immunoglobulins and producing a colored product when incubated with a substrate, were added to each well. After an incubation of 1o minutes at room temperature with shaking, during which the goat anti-mouse antibody - HRP enzyme conjugate binds to 5-FU antibodies in the wells, the plates were again washed three times to remove unbound secondary conjugate. To develop a measurable color in the wells washing was followed by the addition of 1oo L of TMB (TMB Liquid Substrate, Sigma or BioFx), a substrate for HRP, to develop color in a 1o minute incubation with shaking at room temperature. Following the incubation for color development, 5o gL of stop solution (1.5J sodium fluoride in di H20) was added to each well to stop the color development and after lo seconds of shaking the absorbance was determined at 65o nm (Molecular Devices Plate Reader). The amount of antibody in a well was proportional to the absorbance measured and inversely proportional to the amount of 5-FU in the sample.
The absorbance of the color in the wells containing analyte was compared to that with no analyte and a standard curve was generated. The IC5o value for a given analyte was defined as the concentration of analyte that is required to inhibit 50% of the absorbance for the wells containing no analyte. The cross-reactivity of a given analyte was io calculated as the ratio of the IC5o for 5-FU to the IC5o for uracil, thymine, cytosine and Tegafur expressed as a percent. When measured with an antibody as produced in example 9 with immunogen of example 6a the percent cross-reactivates relative to 5-FU
for uracil was less than 8%, for cytosine less than 0.03%, less than 1% for tegafur and about 12% for thymine. The results are in table I.

Example 15 Indirect Competitive Microtiter Plate Immunoassay Procedure Determining IC5o and Cross-Reactivity for Antibodies to 5-FU Derivative 13 Conjugate The ELISA method for measuring 5-FU concentrations was performed with the microtiter plates that were sensitized with 5-FU-BSA described in example 7b.
5-FU, uracil, thymine, cytosine and Tegafur were diluted 1o fold in PBS over a concentration range of o.1 to l,ooo,ooo ng/mL depending on the sample. The assay was performed by incubating 50 L of the analytes to be measured with 5o L of antibody (produced in example 9) diluted to a titer determined in example 12a. During the 1o minute incubation (R.T., with shaking) there is a competition of antibody binding for the 5-FU
conjugate in the well and the analyte in solution. Following this incubation the wells of the plate were washed three times with 0.02 M TRIS, o.g 6o NaCl, 0.5J Tween-8o and o.oo1% Thimerosal, pH 7.8 to remove any material that was not bound. To detect the amount of 5-FU antibody bound to the 5-FU-BSA conjugate in the wells, ioo L
of a goat anti-mouse antibody - HRP enzyme conjugate (Jackson Immunoresearch) diluted to a predetermined specific activity (approximately 1/2000) in PBS with o.1%
BSA, 0.05f ANS, o.oi% thimerosal, capable of binding specifically with murine immunoglobulins and producing a colored product when incubated with a substrate, Y.o were added to each well. After an incubation of lo minutes at room temperature with shaking, during which the goat anti-mouse antibody - HRP enzyme conjugate binds to 5-FU antibodies in the wells, the plates were again washed three times to remove unbound secondary conjugate. To develop a measurable color in the wells washing was followed by the addition of 1oo L of TMB (TMB Liquid Substrate, Sigma or BioFx), a substrate for HRP, to develop color in a 1o minute incubation with shaking at room temperature. Following the incubation for color development, 5o L of stop solution (1.5% sodium fluoride in di H20) was added to each well to stop the color development and after 1o seconds of shaking the absorbance was determined at 65o nm (Molecular Devices Plate Reader). The amount of antibody in a well was proportional to the 2o absorbance measured and inversely proportional to the amount of 5-FU in the sample.
The absorbance of the color in the wells containing analyte was compared to that with no analyte and a standard curve was generated. The IC5o value for a given analyte was defined as the concentration of analyte that is required to inhibit 50f of the absorbance for the wells containing no analyte. The cross-reactivity of a given analyte was calculated as the ratio of the IC5o for 5-FU to the IC5o for uracil, thymine, cytosine and Tegafur expressed as a percent. When measured with a monoclonal antibody as produced in example 9 with immunogen of example 6b the percent cross-reactivities relative to 5-FU for uracil was less than 8%, less than o.o1 1o for cytosine, about 1% for tegafur and less than 4% for thymine. The results are in table II.

C5p O
co F N
.'., O
Vj e-I C7 cz O
r= V V
'o O

~ o p., Ln U

O O
~
=~ O O
cd L~P~
cd O
CT
O
Cn 4 U y r 4-1 N U ~
a C P' 00 y y ZO
o =a+ O ~ ~ ~co ~ >C 1=4 w~lj aU ~=-' OW O ~=~ c~W U

rA

v ~r y y Q' ~
b U 0-1 Ua) p bJD U ~ 'd ~ o s a~
~ . p ;z M
~s 4-y 1"0 U] v U ~ ~,i U
H

a a r4 ~
~ o 0 U o ...

=~' .r, o b~ a N
o ~
O ci ~ ~

co o y cz to O
=~ i~-i 0 O O
'rn cd C~/~ ~
cn pq m cz ~ o =~ ~ a~ ~

ct ((w s.r M

+r q ~o ca ~

~ ~
o ~
M "
~
t~ o O CU .o U(D rd rn y H ~ o 4 M ~ 0 U L + ~, ~ a~ A+
cc ~ y W p M

The results in these tables demonstrate the importance of forming the immunogen from the compound of formula II-A and the reagent from the compound of formula II-A
or II-B. From these results it can be seen that it is when the immunogen is formed from the compound of formula II-A, rather than II-B, an antibody is produced which does not cross-react with Tegafur. It is through the antibody provided from the immunogen of the compound of the formula II-A and the reagent carrier provided from the compound of II-A or II-B, that produces an accurate immunoassay for 5-FU to monitor patients being treated with 5-FU.

Claims (51)

What is claimed:
1. An immunoassay for detecting 5-fluoro-uracil in a sample comprising providing a mixture of a) a sample, b) an antibody selectively reactive with 5-fluoro-uracil which antibody is not substantially cross-reactive with tegafur and c) a conjugate of a carrier with a compound of the formula:

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to the carrier; and p is an integer from 0 to 1 or a compound of the formula:

wherein X, Y and p are as above;
or mixtures thereof;

causing the 5-fluoro-uracil in the sample and said conjugate to bind with said antibody and thereafter measuring the amount of said conjugate in said mixture which is bound or unbound to said antibody whereby the presence of 5-fluoro-uracil in the sample can be determined.
2. The process of claim 1, wherein the sample is a human sample.
3. The immunoassay of claim 2, wherein said antibody is generated from an immunogen comprising an immunogenic carrier containing a polyamine polymer conjugated with a compound of the formula:

wherein p, X and Y are as above.
4. The immunoassay of claim 2, wherein the antibody is attached to a solid support.
5. The immunoassay of claim 4, wherein the solid support is microtitor plates.
6. The immunoassay of claim 4, wherein the solid support is nanoparticles.
7. An antibody which binds selectively to 5-fluoro-uracil and does not substantially bind to tegafur.
8. The antibody of claim 7, wherein said antibody is a monoclonal antibody.
9. The antibody of claim 8, wherein said antibody is derived from mice, rabbits or rats.
10. The antibody of claim 9, wherein said antibody does not substantially cross-react
11. The antibody of claim 7, wherein said antibody is derived from an immunogenic carrier containing a polyamine polymer conjugated to a compound of the formula:

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a polyamine polymer; and p is an integer from 0 to 1.
12. The antibody of claim 11, wherein said antibody is a monoclonal antibody,
13. The antibody of claim 11, wherein said antibody is derived from mice, rabbits or rats.
14. The antibody of claim 13, wherein said antibody does not substantially cross-react with uracil and cytosine.
15. A compound of the formula:

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a carrier; and p is an integer from 0 to 1.
16. The compound of claim 15, wherein p is 0.
17. The compound of claim 16, wherein X is wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur.
18. The compound of claim 17, wherein X is and R3 is hydrogen.
19. The compound of claim 17, wherein X is and R3 forms a reactive ester.
20. The compound of claim 19, wherein the ester formed is a lower alkyl ester, imidoester or amidoester.
21. The compound of claim 15, wherein p is 1.
22. The compound of claim 21, wherein Y is alkylene containing from 1 to 10 carbon atoms, wherein n and 0 are integers from 0 to 6, and m is an integer from 1 to 6.
23. A compound of the formula:

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a carrier; and p is an integer from 0 to 1;

or mixtures thereof.
24. The compound of claim 23, wherein p is 0.
25. The compound of claim 24, wherein X is wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur.
26. The compound of claim 25, wherein X is and R3 is hydrogen.
27. The compound of claim 26, wherein X is and R3 forms a reactive ester.
28. The compound of claim 27, wherein the ester formed is a lower alkyl ester, imidoester or amidoester.
29. The compound of claim 23, wherein p is 1.
3o. The compound of claim 29, wherein X is wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur.
31. The compound of claim 30, wherein Y is alkylene containing from 1 to 10 carbon atoms, wherein n and 0 are integers from 0 to 6, and m is an integer from 1 to 6.
32. A conjugate of a carrier with a compound of the formula:

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a carrier; and p is an integer from 0 to 1.
33. The conjugate of claim 32, wherein p is 0.
34. The conjugate of claim 33, wherein X is wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur.
35. The conjugate of claim 32, wherein p is 1 and Y is alkylene containing from 1 to 10 carbon atoms, wherein n and 0 are integers from 0 to 6, and m is an integer from 1 to 6.
36. The conjugate of claim 35, wherein the carrier contains one or more amino groups linked by wherein R4 is oxygen or sulfur.
37. A conjugate of a carrier with a compound of the formula:
wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a carrier; and p is an integer of from o to 1.
38. The conjugate of claim 37, wherein p is o.
39. The conjugate of claim 38, wherein X is wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur.
40. The conjugate of claim 37, wherein p is 1.
41. The conjugate of claim 40, wherein Y is alkylene containing from i to 1o carbon atoms, wherein n and 0 are integers from 0 to 6, and m is an integer from 1 to 6.
42. The conjugate of claim 41, wherein X is wherein R 3 is hydrogen or taken together with its attached oxygen atom forms a reactive ester and R4 is oxygen or sulfur.
43. An immunogen containing an immunogenic carrier containing a polyamine polymer linked to a compound of the formula:

wherein Y is an organic spacing group;

X is a terminal functional group capable of binding to a polyamine polymer; and p is an integer from 0 to 1.
44. The immunogen of claim 43, wherein p is 0.
45. The immunogen of claim 43, wherein p is 1.
46. The immunogen of claim 45, wherein Y is alkylene containing from 1 to 10 carbon atoms, wherein n and o are integers from 0 to 6, and m is an integer from 1 to 6.
47. The immunogen of claim 46, wherein the immunogenic polymer contains one or more amino groups linked by wherein R4 is oxygen or sulfur.
48. A kit for determining the presence of 5-fluoro-uracil in a patient sample comprising reagents in separate containers, one of the reagents being a conjugate of a carrier with a compound of the formula:

wherein Y is an organic spacing group;
X is a terminal functional group capable of binding to carrier; and p is an integer from 0 to 1;
or a compound of the formula:

wherein X, Y and p are as above;
or mixtures thereof;
and the second container containing an antibody substantially selectively reactive with 5-fluoro-uracil and not substantially cross-reactive to tegafur or uracil.
49. The kit of claim 48, wherein said conjugate is present in a predetermined amount in said first container.
50. The kit of claim 49, wherein said kit is used to determine the amount of 5-fluoro-uracil in said sample.
51. The kit of claim 48, wherein, said antibody is generated from an immunogen of an immunogenic polyamine polypeptide linked to a compound of the formula:

wherein p, X and Y are as above.
CA2597044A 2005-02-08 2006-02-02 5-fluoro-uracil immunoassay Active CA2597044C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11/053,480 US20060177883A1 (en) 2005-02-08 2005-02-08 5-Fluoro-uracil immunoassay
US11/053,480 2005-02-08
PCT/US2006/003674 WO2006086208A2 (en) 2005-02-08 2006-02-02 5-fluoro-uracil immunoassay

Publications (2)

Publication Number Publication Date
CA2597044A1 true CA2597044A1 (en) 2006-08-17
CA2597044C CA2597044C (en) 2014-05-20

Family

ID=36780436

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2597044A Active CA2597044C (en) 2005-02-08 2006-02-02 5-fluoro-uracil immunoassay

Country Status (8)

Country Link
US (4) US20060177883A1 (en)
EP (4) EP1846763B1 (en)
JP (2) JP5031585B2 (en)
CN (1) CN101147063B (en)
CA (1) CA2597044C (en)
ES (2) ES2485691T3 (en)
HK (2) HK1116864A1 (en)
WO (1) WO2006086208A2 (en)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8828665B2 (en) 2007-02-16 2014-09-09 Ark Diagnostics, Inc. Compounds and methods for use in detecting gabapentin
US8168756B2 (en) 2008-10-24 2012-05-01 Ark Diagnostics, Inc. Levetiracetam immunoassays
JP5863047B2 (en) * 2009-07-17 2016-02-16 ミリアド ジェネティクス, インコーポレイテッド Method for assaying 5-FU
WO2011022174A1 (en) * 2009-08-21 2011-02-24 Myriad Genetics, Inc. Method of processing blood samples containing 5-fu
US20130109707A1 (en) * 2010-03-01 2013-05-02 Concert Pharmaceuticals ,Inc. Fluorouracil derivatives
US8426143B2 (en) 2011-05-24 2013-04-23 Saladax Biomedical Inc. Gemcitabine immunoassay
WO2013082463A2 (en) * 2011-11-30 2013-06-06 Wellstat Diagnostics, Llc. Assays, antibodies, immunogens and compositions related to 5-fu
US9625465B2 (en) 2012-05-15 2017-04-18 Defined Diagnostics, Llc Clinical diagnostic systems
US9075042B2 (en) 2012-05-15 2015-07-07 Wellstat Diagnostics, Llc Diagnostic systems and cartridges
US9213043B2 (en) 2012-05-15 2015-12-15 Wellstat Diagnostics, Llc Clinical diagnostic system including instrument and cartridge
JP6352258B2 (en) 2012-07-16 2018-07-04 センターズ フォー ディジーズ コントロール アンド プリベンション Direct-reading detection kit for surface contamination with antitumor agents
WO2014031645A1 (en) * 2012-08-21 2014-02-27 Ortho-Clinical Diagnostics, Inc Antibodies to risperidone haptens and use thereof
US9970929B2 (en) 2013-01-18 2018-05-15 Ark Diagnostics, Inc. Voriconazole immunoassays
ES2674704T3 (en) 2013-02-13 2018-07-03 Ark Diagnostics, Inc. Posaconazole Immunoassays
CN104478813A (en) * 2014-12-20 2015-04-01 苏州博源医疗科技有限公司 5-fluorouracil derivatives, 5-fluorouracil immunogens, antibodies for immunogens and 5-fluorouracil detection kit
CN107132347A (en) * 2016-11-25 2017-09-05 南方医科大学 A kind of time-resolved fluorescence detection kit of 5 fluorouracil blood concentrations of real-time monitoring
EP3684944A4 (en) 2017-09-21 2021-05-26 Becton, Dickinson and Company Hazardous contaminant collection kit and rapid testing
EP3836971A4 (en) * 2018-08-13 2022-06-22 Flagship Pioneering Innovations V, Inc. Conjugates and methods of using the same
EP3918300A4 (en) 2019-01-28 2022-11-16 Becton, Dickinson and Company Hazardous contaminant collection device with integrated swab and test device
CN110156872B (en) * 2019-05-27 2020-09-08 山东省医学科学院药物研究所(山东省抗衰老研究中心、山东省新技术制药研究所) Tegafur derivative and preparation method and application thereof
US20200393451A1 (en) 2019-06-13 2020-12-17 Becton, Dickinson And Company Multiplexed turbidimetric immunoassays to monitor environmental contamination
CN111875587B (en) * 2020-07-23 2021-07-13 湖南苏阳医疗科技有限公司 5-fluorocytosine derivative, preparation method thereof and application thereof in 5-fluorocytosine immunodetection reagent
CN113501877A (en) * 2021-08-23 2021-10-15 苏州科铭生物技术有限公司 Preparation method of anti-5-fluorouracil monoclonal antibody and enzyme-linked immunoassay method of 5-fluorouracil
CN114605338B (en) * 2022-05-11 2022-10-28 深圳厚存纳米药业有限公司 Nanoparticle and nucleic acid nanocomposite containing uracil derivative, and preparation method and application thereof

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3996344A (en) 1972-05-15 1976-12-07 Biological Developments, Inc. Phenethylamine antigenic conjugates, their preparation, antibodies and use
US4016146A (en) 1974-12-10 1977-04-05 Biological Developments, Inc. Phenethylamine antigenic conjugates, their preparation, antibodies, and use
US3996334A (en) * 1975-06-02 1976-12-07 Southwire Company Process for recovering aluminum from alunite
US4269511A (en) 1979-02-15 1981-05-26 Abbott Laboratories Apparatus and method for measuring the magnitude of polarization of light
US4420568A (en) 1980-07-30 1983-12-13 Abbott Laboratories Fluorescent polarization immunoassay utilizing substituted triazinylaminofluoresceins
JPS63222699A (en) * 1987-03-11 1988-09-16 Sendai Biseibutsu Kenkyusho Monoclonal antibody and measurement of pseudouridine phi using said antibody
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5101015A (en) 1989-04-10 1992-03-31 Abbott Laboratories Reagents for an amphetamine-class fluorescence polarization immunoassay
CA2096495C (en) 1992-06-16 2002-07-09 Kathy Palmer Ordonez Dual analyte immunoassay
US5789192A (en) 1992-12-10 1998-08-04 Schering Corporation Mammalian receptors for interleukin-10 (IL-10)
JP3479453B2 (en) * 1998-05-28 2003-12-15 大鵬薬品工業株式会社 Method for measuring tegafur in urine
JP4070372B2 (en) * 1999-10-19 2008-04-02 有限会社バイオメディカルリサーチグループ Anti-uracil monoclonal antibody

Also Published As

Publication number Publication date
CA2597044C (en) 2014-05-20
EP2280281B1 (en) 2014-05-21
WO2006086208A3 (en) 2007-04-26
US7767794B2 (en) 2010-08-03
HK1147799A1 (en) 2011-08-19
EP2280280A1 (en) 2011-02-02
JP2008530528A (en) 2008-08-07
US7205116B2 (en) 2007-04-17
US20060177883A1 (en) 2006-08-10
JP2012197285A (en) 2012-10-18
CN101147063A (en) 2008-03-19
US20100204456A1 (en) 2010-08-12
ES2485691T3 (en) 2014-08-14
WO2006086208A2 (en) 2006-08-17
CN101147063B (en) 2014-02-12
EP2317325A1 (en) 2011-05-04
EP1846763A2 (en) 2007-10-24
US20060177884A1 (en) 2006-08-10
HK1116864A1 (en) 2009-01-02
US20070154972A1 (en) 2007-07-05
JP5031585B2 (en) 2012-09-19
EP1846763B1 (en) 2012-12-05
ES2400773T3 (en) 2013-04-12
EP1846763A4 (en) 2008-04-02
US9551724B2 (en) 2017-01-24
EP2280281A1 (en) 2011-02-02

Similar Documents

Publication Publication Date Title
EP2280281B1 (en) 5-fluoro-uracil immunoassay
EP2544540B1 (en) Lenalidomide and thalidomide immunoassays
CA2769998C (en) Imatinib immunoassay
CA2572618C (en) Taxol immunoassay
US7175993B2 (en) Taxol immunoassay
EP1774331B1 (en) Cytoxan antibodies and immunoassay
US7423131B2 (en) Busulfan immunoassay
CA2836539A1 (en) Gemcitabine immunoassay
WO2011071672A1 (en) Irinotecan immunoassay
AU2012259309A1 (en) Gemcitabine immunoassay

Legal Events

Date Code Title Description
EEER Examination request