US20100184082A1 - Antibody and immunoassays for determining the presence of delta9-tetrahydrocannabinol - Google Patents

Antibody and immunoassays for determining the presence of delta9-tetrahydrocannabinol Download PDF

Info

Publication number
US20100184082A1
US20100184082A1 US11/478,972 US47897206A US2010184082A1 US 20100184082 A1 US20100184082 A1 US 20100184082A1 US 47897206 A US47897206 A US 47897206A US 2010184082 A1 US2010184082 A1 US 2010184082A1
Authority
US
United States
Prior art keywords
thc
antibody
seq
amino acid
sample
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
US11/478,972
Other versions
US7749772B1 (en
Inventor
Daniel Wang
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.)
Agilent Technologies Inc
Original Assignee
Varian Inc
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 Varian Inc filed Critical Varian Inc
Priority to US11/478,972 priority Critical patent/US7749772B1/en
Assigned to VARIAN, INC. reassignment VARIAN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WANG, DANIEL
Application granted granted Critical
Publication of US7749772B1 publication Critical patent/US7749772B1/en
Publication of US20100184082A1 publication Critical patent/US20100184082A1/en
Assigned to AGILENT TECHNOLOGIES, INC. reassignment AGILENT TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VARIAN, INC.
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/16Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from plants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • Y10S436/807Apparatus included in process claim, e.g. physical support structures
    • Y10S436/81Tube, bottle, or dipstick

Definitions

  • This invention relates generally to immunoassays for drugs of abuse and the like.
  • Marijuana is a member of the hemp family and is known to contain significant amounts of cannabinoids.
  • the most important cannabinoid is ⁇ 9 -Tetrahydrocannabinol ( ⁇ 9 -THC), the major physiologically active constituent of marijuana.
  • ⁇ 9 -THC is a controlled substance because it has both sedative and depressant-like effects on the cardiovascular and central nervous systems, as opposed to cannabidiol, a non-psychoactive constituent of marijuana.
  • ⁇ 9 -THC is rapidly absorbed from the lungs into the blood stream and metabolized through 11-nor ⁇ 9 -THC to a series of polar metabolites with 11-nor- ⁇ 9 -THC-carboxylic acid as the primary metabolite.
  • cannabinoids in biological samples can be detected by a number of techniques such as thin layer chromatography (TLC), gas chromatography/mass spectrometry (GC/MS), radioimmunoassay, enzyme immunoassay or continuous flow displacement immunoassay (U.S. Pat. No. 6,686,208).
  • TLC thin layer chromatography
  • GC/MS gas chromatography/mass spectrometry
  • radioimmunoassay enzyme immunoassay or continuous flow displacement immunoassay
  • cannabinoid metabolites may be detected in the urine for up to 10 days in occasional smokers and 36 days in chronic smokers. See U.S. Pat. No. 5,264,373.
  • Tetrahydrocannabinol the active ingredient in the marijuana plant (cannabis sativa), is detectable in saliva shortly after use.
  • the detection of the drug in saliva is thought to be primarily due to the direct exposure of the drug to the mouth (oral and smoking administrations) and the subsequent sequestering of the drug in the buccal cavity.
  • the non-metabolized drug, ⁇ 9 -THC is the main form of THC present in the marijuana smoker's oral fluid sample.
  • ⁇ 9 -THC is an excellent indicator of recent marijuana consumption and may indicate that the person is under the drug's influence. Detection of the parent THC therefore is of great interest for road side testing by law enforcement, on-site testing in work places and many other circumstances.
  • saliva THC assays and procedures are still problematic. Due to the short detectable time window of the parent THC in the oral fluid, low quantity, the low THC solubility and “stickiness” in aqueous solution, and sensitivity limitations of currently available murine anti-THC antibodies, current lateral flow immunoassays for parent THC can not meet the expected sensitivity requirement recommended by the Substance Abuse and Mental Health Services Administration (SAMHSA).
  • SAMHSA Substance Abuse and Mental Health Services Administration
  • the THC assay contained within the Oral Fluid Drug Screen Device is purported to detect the presence of THC-COOH.
  • this assay requires the presence of the THC metabolite THC-COOH, and does not detect the parent ⁇ 9 -THC molecule with a high sensitivity.
  • the THC “One Step Marijuana Test Strip” (Biomerica, Inc. Newport Beach, Calif.) is reported to be a rapid chromatographic immunoassay for the detection of 11-nor-9-THC-9 COOH in human urine at a cut-off concentration of 50 ng/mL.
  • this assay system cannot be used to detect the parent ⁇ 9 -THC molecule in saliva and requires urine collection and testing.
  • a fluorescence immunoassay has been reported for the detection and quantitation of tetrahydrocannabinols in oral fluids for use in testing for marijuana abuse.
  • the lower detection limits were reported to be 1.5 ng/mL for (1)-, 9-THC and 5.5 ng/mL for (1)9-Carboxy-11-nor-, 9-THC and to require a total assay reaction time of less than 10 minutes.
  • fluorescence detection is required for this assay, necessitating the use of expensive equipment and limiting the availability and widespread usage of this assay.
  • ⁇ 9 -THC-based or ⁇ 9 -THC-analog based tracers is problematic because the ⁇ 9 -THC analyte does not effectively displace the ⁇ 9 -THC-based or ⁇ 9 -THC-analog based tracer. This results in a lower sensitivity of the assay.
  • the amino acid sequence for the light chain variable region comprises the sequence:
  • the amino acid sequence for the heavy chain variable region comprises the sequence: QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGKGLEWIGTIVSGTTYYAS WAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYITGDIWGPGTLVTVSLG (SEQ ID NO:8), or homologous amino acid sequences having at least 85% homology thereto.
  • the antibody is a monoclonal antibody produced by a hybridoma. In additional preferred embodiments, the antibody is a recombinant antibody produced by a host cell. In a preferred embodiment, the antibody is a rabbit antibody.
  • a host cell line that expresses an antibody having specific binding for ⁇ 9 -THC and its metabolites, wherein said antibody comprises CDR amino acid sequences selected from the group consisting of L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) or homologous amino acid sequences having at least 85% homology thereto.
  • the host cell can be any cell capable of expressing antibody.
  • the host cell is a HEK 293 cell.
  • an immunoassay for detecting the presence of THC and its metabolites in a sample.
  • the immunoassay utilizes an antibody having specific binding for THC and its metabolites, wherein said antibody comprises CDR amino acid sequences selected from the group consisting of L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) or homologous amino acid sequences having at least 85% homology thereto.
  • the antibody is labeled or conjugated to a solid support such as microparticles or a solid surface.
  • the immunoassay is a lateral flow immunoassay strip for detecting the presence of THC and/or its metabolites in a fluid sample.
  • a method for detecting the presence of THC in a sample comprising the steps of: (1) providing a lateral flow immunoassay system comprising an antibody having specific binding for ⁇ 9 -THC; (2) contacting said lateral flow immunoassay system with a sample (e.g., an extract or sample of saliva obtained from an individual) to be tested for the presence of ⁇ 9 -THC in saliva; (3) performing the lateral flow immunoassay; (4) determining whether the immunoassay is positive or negative for the presence of ⁇ 9 -THC in the sample.
  • a sample e.g., an extract or sample of saliva obtained from an individual
  • kits for testing for the presence of THC in saliva, comprising a lateral flow immunoassay strip for THC, and instructions for performing the test.
  • binding refers to a binding affinity of an antibody to THC or its metabolites and analogs of at least 10 6 M ⁇ 1 , more preferably between about 10 7 and about 10 11 M ⁇ 1 , or typically between about 10 8 and 10 10 M ⁇ 1 .
  • THC refers to tetrahydrocannabinols in general, and more particularly, to the parent molecule commonly found in marijuana, ⁇ 9 -THC.
  • antibody is used in its broadest sense and includes the antibody classes such as IgG (and its subclasses IgG 1 , IgG 2 , etc.), IgA, IgD, IgE, IgM, as well as Fab, Fab 2 , Fv, diabodies, and other forms of antibody variants, constructs and fragments known in the art.
  • the present invention provides an isolated antibody having specific binding for the parent THC ( ⁇ 9 -THC) and/or its major metabolites, which is a significant advance over the performance of other antibodies known in the art which are not sufficiently sensitive for the parent ⁇ 9 -THC. Exhaustive testing was performed on additional murine anti-THC antibodies using ELISA assays but none of the murine antibodies provided satisfactory performance or binding characteristics for ⁇ 9 -THC.
  • the antibodies described herein present a significant increase in sensitivity of immunoassays such as lateral flow immunoassays and ELISA for THC and allow the determination of the presence of ⁇ 9 -THC as well as its metabolites with higher sensitivity than previously possible.
  • the superior performance characteristics are demonstrated in Example 4.
  • the present invention also provides a hybridoma cell clone producing the antibody as a monoclonal antibody, a recombinant antibody, further molecularly engineered recombinant antibodies and host cells producing the recombinant antibodies.
  • the invention also provides methods of using the antibody in immunochemical applications and immunoassays, particularly lateral flow immunoassays, and methods for detecting THC in body fluids, particularly saliva, and kits for determining the presence of THC.
  • the present invention provides high sensitivity anti-THC antibodies having antigen binding regions for detecting the presence of THC and its analogs in body fluids. Any antibody having the requisite sensitivity and binding affinity can be utilized.
  • Preferred antibodies include rabbit monoclonal antibodies.
  • Particularly preferred antibodies include CDR sequences as follows: L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6).
  • preferred antibodies comprise the amino acid sequence for the light chain variable region:
  • Conservative amino acid substitutions of these sequences are also encompassed within the scope of the anti-THC antibodies.
  • Conservative amino acid substitutions are well known to those of skill in the art, and are generally defined as amino acid replacements that preserve the structure and functional properties of proteins.
  • the PAM250 and Blosum 45 similarity matrices can be utilized to determine which amino acids are most likely to be acceptable substitutions to an existing sequence without causing loss of native structure and function of the protein.
  • the present invention also includes amino acid sequences which are homologous to SEQ ID NOS: 1-6, and the fragments above.
  • “homologous amino acid sequence” is any polypeptide which is encoded, in whole or in part, by a nucleic acid sequence which hybridizes at 25° C. to 35° C. below critical melting temperature (T m ), to any portion of the nucleic acid sequence encoding SEQ ID NOS: 1-6, or specifically to the nucleic acid sequences encoding the six CDR amino acid sequences, as shown below:
  • CDR-L1 (SEQ ID NO: 9) CAG GCC AGT CAA AGT GTT TAT AAT AAC AAC CAA TTA TCC; CDR-L2: (SEQ ID NO: 10) GGT ACA TCC AAT CTG GCA TCT; CDR-L3: (SEQ ID NO: 11) CAA GGC GGT TAT ACT AGT GGT GAT GGT AAT GCT; CDR-H1: (SEQ ID NO: 12) GGA TTC TCC CTC AGT AAC TAT GTA TTG GCC; CDR-H2: (SEQ ID NO: 13) ACC ATT GTT AGC GGT ACC ACA TAC TAC GCG AGT TGG; CDR-H3: (SEQ ID NO: 14) GGT TTG CCT CAT TAT ATT ACT GGG GAC ATC.
  • a homologous amino acid sequence can be one that differs from an amino acid sequence shown in SEQ ID NOS: 1-6 by one or more conservative amino acid substitutions.
  • Homologous amino acid sequences include sequences that are identical or substantially identical to SEQ ID NOS: 1-6.
  • amino acid sequence substantially identical is meant a sequence that is at least 85%, preferably 90%, more preferably 95%, most preferably 97%-99% identical to an amino acid sequence of reference and that preferably differs from the sequence of reference by a majority of conservative amino acid substitutions.
  • Conservative amino acid substitutions are substitutions among amino acids of the same class.
  • amino acids having uncharged polar side chains such as asparagines (N), glutamine (Q), serine (S), threonine (T), and tyrosine (Y); amino acids having basic side chains, such as lysine (K), arginine (R), and histidine (H); amino acids having acidic side chains, such as aspartic acid (D) and glutamic acid (E); and amino acids having nonpolar side chains, such as glycine (G), alanine (A), valine (V), leucine (L), isoleucine (I), praline (P), phenylalanine (F), methionine (M), tryptophan (W), and cysteine (C).
  • amino acids having uncharged polar side chains such as asparagines (N), glutamine (Q), serine (S), threonine (T), and tyrosine (Y); amino acids having basic side chains, such as lysine (K), arginine (R), and his
  • sequence analysis software such as Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705. Amino acid sequences are aligned to maximize identity. Gaps can be artificially introduced into the sequence to attain proper alignment. Once the optimal alignment has been set up, the degree of homology is established by recording all of the positions in which the amino acids of both sequences are identical, relative to the total number of positions.
  • homologous polynucleotide sequences are defined in a similar way.
  • a homologous sequence is one that is at least 45%, more preferably 50%, 55%, 60%, 65%, 70%, 75%, 80%, and even more preferably 85%, 87%, 90%, 93%, 96% and most preferably 99% identical to a nucleotide sequence encoding amino acid sequences SEQ ID NOS:1-6.
  • antigen binding regions i.e., complementarity determining regions
  • conservative amino acid substitutions are made such that the structure, hydrophobicity, charge, etc. of a particular amino acid or amino acid sequence is unchanged or changed only minimally.
  • the objective is to maintain antigen binding affinity, but allow for maturation or adjustment of the binding affinity to identify additional amino acid sequences that provide useful binding characteristics to a given antigen.
  • Conservative amino acid substitutions thus involve replacing one or more amino acids at a time with amino acids of the same type, i.e., amino acids having charged (basic or acidic) or uncharged polar side chains, or nonpolar side chains, for example, substitution of an uncharged amino acid such as asparagine (N), with another uncharged amino acid such as glutamine (Q), serine (S), threonine (T), or tyrosine (Y; substitution of a basic amino acid such as lysine (K), with another basic amino acid such as arginine (R) or histidine (H); substitution of an acidic amino acid such as aspartic acid (D), with another acidic amino acid such as glutamic acid (E); or substitution of a nonpolar amino acid such as glycine (G) with another nonpolar amino acid such as alanine (A), valine (V), leucine (L), isoleucine (I), methionine (M), proline (P), phenylalanine (F),
  • L1 QATQSVYNNNQLS (SEQ ID NO:15), QASQSVYNNNQLT (SEQ ID NO:16), QASQTVYNNNQLS (SEQ ID NO:17), QASQSAYNNNQLS (SEQ ID NO:18), QVSQSVYNNNQLS (SEQ ID NO:19), QASQSVYNNNQIS (SEQ ID NO:20), QASQSVFNNNQLS (SEQ ID NO:21), QASQSVYNNNQLS (SEQ ID NO:22), and the like;
  • GTSNLAS SEQ ID NO:23
  • GSSNLAS SEQ ID NO:24
  • GTSNLAT SEQ ID NO:25
  • GTSNIAS SEQ ID NO:26
  • GTSNLVS SEQ ID NO:27
  • GTSNLAS SEQ ID NO:28
  • GSSNIAS SEQ ID NO:29
  • GTTNLVS SEQ ID NO:30
  • QGGYTSGDGIA SEQ ID NO:31; QGAYTSGDGIA (SEQ ID NO:32); QGGYSSGDGIA (SEQ ID NO:33); QGGYTSGEGIA (SEQ ID NO:34); QGGYTSGDGLA (SEQ ID NO:35); QGGYTSGDGIV (SEQ ID NO:36); QGGFTSGDGIA (SEQ ID NO:37); QGGYTTGEGIA (SEQ ID NO:38), and the like;
  • GFSLSNYVLA SEQ ID NO:39
  • GYSLSNYVLA SEQ ID NO:40
  • GFTLSNYVLA SEQ ID NO:41
  • GFSLTNYVLA SEQ ID NO:42
  • GFSLSNFVLA SEQ ID NO:43
  • GFSLSNYVIA SEQ ID NO:44
  • GFSLSNYVLV SEQ ID NO:45
  • H2 TIVSGTTYYASW (SEQ ID NO:46), TLVSGTTYYASW (SEQ ID NO:47), TIVSGTSYYASW (SEQ ID NO:48), TIVSGTTYYVSW (SEQ ID NO:49), TIVSGTTFYASW (SEQ ID NO:50), TIVSGSTYYATW (SEQ ID NO:51), and the like;
  • H3 GLPHFITGDI (SEQ ID NO:52), GIPHYITGDI (SEQ ID NO:53), GLPHYITGDL (SEQ ID NO:54), GLPHYLTGDI (SEQ ID NO:55), GLPHYITGEI (SEQ ID NO:56), GLPNYITGDI (SEQ ID NO:57), GLPHFITGEI (SEQ ID NO:58), and the like.
  • substitutions include no more than about 5% changes from the preferred sequences, preferably no more than about 10% changes and most preferably, no more than about 15% changes in the preferred sequences.
  • Such sequences are 95%, 90% and 85% homologous, respectively, with the preferred CDR sequences.
  • each CDR sequence can be substituted independently, such that one CDR sequence can remain unsubstituted while one or more of the other CDR sequences can have one or more amino acid substitutions, as desired.
  • One skilled in the art can easily envision acceptable substitutions and using recombinant methodologies, prepare and test the resulting antibody variants for binding to THC. In this manner, variants and improvements on the preferred CDR sequences disclosed above can be prepared, enabling the practice of the full scope of the invention.
  • an antibody that specifically binds to THC and has CDR1 in the V L region (V L /CDR1) comprising the amino acid sequence of Q X 1 X 2 Q X 3 X 4 X 5 N N N Q X 6 X 7 (SEQ ID NO:59) wherein X 1 can be G, A or V; X 2 is S or T; X 3 is S or T; X 4 is M, L, I, or V; X 5 is Y, or F; X 6 is L, I; X 7 is S or T.
  • an antibody that specifically binds to THC and has CDR2 in the V L region (V L /CDR2) comprising the amino acid sequence of G X 1 X 2 X 3 X 4 X 5 X 6 (SEQ ID NO:60), wherein X 1 , X 2 , X 3 and X 6 can be T, Y, S, Q or N; and X 4 and X 5 can be G, A, V, L, I, M, P, F, W or C.
  • an antibody that specifically binds to THC and has CDR3 in the V L region (V L /CDR3) comprising the amino acid sequence of Q X 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 X 10 , (SEQ ID NO:61), wherein X 1 , X 2 , X 6 and X 8 , X 9 and X 10 can be G, A, V, L, I, M, P, F, W or C; X 3 , X 4 and X 5 can be T, Y, S, Q or N; X 7 can be D or E.
  • an antibody that specifically binds to THC and has CDR1 in the V H region (V H /CDR1) comprising the amino acid sequence of X 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 X 10 , (SEQ ID NO:62), wherein X 1 , X 4 , X 8 , X 9 and X 10 can be G, A, V, L, M or I; X 2 and X 7 can be F, W or Y; X 3 , and X 5 can be T, Y, or S; and X 6 can be N or Q.
  • an antibody that specifically binds to THC and has CDR2 in the V H region (V H /CDR2) comprising the amino acid sequence of X 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 X 10 X 11 X 12 , (SEQ ID NO:63), wherein X 1 , X 4 , X 6 , X 7 and X 11 can be T, S or Y; X 2 , X 3 or X 10 , can be G, A, V, L, M or I; X 5 can be G, V, A, L or I, X 8 and X 9 can be F or Y, and X 12 can be W, Y or F.
  • an antibody that specifically binds to THC and has CDR3 in the V H region (V H /CDR3) comprising the amino acid sequence of X 1 X 2 P X 3 X 4 X 5 X 6 X 7 X 8 X 9 , (SEQ ID NO:64), wherein X 1 , X 2 , X 5 X 7 and X 9 can be G, L, M, I, V, or A, X 6 can be T, S or Y, X 8 can be D or E, X 3 can be H, K or R, X 4 can be Y or F.
  • the anti-THC antibodies may be synthesized or expressed in cells of any organism, including but not limited to bacteria, yeast, plant, insect, and mammal. Particular types of cells include, but are not limited to, Drosophila melanogaster cells, Saccharomyces cerevisiae and other yeasts, E. coli, Bacillus subtilis , SF9 cells, C129 cells, 293 cells, Neurospora, BHK, CHO, COS, and HeLa cells, fibroblasts, Schwanoma cell lines, immortalized mammalian myeloid and lymphoid cell lines, Jurkat cells, mast cells and other endocrine and exocrine cells, and neuronal cells.
  • Drosophila melanogaster cells Saccharomyces cerevisiae and other yeasts
  • E. coli Bacillus subtilis
  • SF9 cells SF9 cells
  • C129 cells 293 cells
  • Neurospora BHK, CHO, COS, and HeLa cells
  • fibroblasts Schwanoma cell lines
  • mammalian cells include, but are not limited to, tumor cells of all types (particularly melanoma, myeloid leukemia, carcinomas of the lung, breast, ovaries, colon, kidney, prostate, pancreas and testes), cardiomyocytes, endothelial cells, epithelial cells, lymphocytes (T-cell and B cell), mast cells, eosinophils, vascular intimal cells, hepatocytes, leukocytes including mononuclear leukocytes, stem cells such as haemopoetic, neural, skin, lung, kidney, liver and myocyte stem cells, osteoclasts, chondrocytes and other connective tissue cells, keratinocytes, melanocytes, liver cells, kidney cells, and adipocytes.
  • tumor cells of all types particularly melanoma, myeloid leukemia, carcinomas of the lung, breast, ovaries, colon, kidney, prostate, pancreas and testes
  • cardiomyocytes endot
  • the anti-THC antibodies can be purified or isolated after expression according to methods known to those skilled in the art.
  • purification methods include electrophoretic, molecular, immunological and chromatographic techniques, including ion exchange, hydrophobic, affinity, and reverse-phase HPLC chromatography, and chromatofocusing, and the like.
  • the degree of purification necessary will vary depending on the contaminants present with the antibodies. In some instances no purification will be necessary.
  • the anti-THC antibodies can be screened for a desired function, preferably a biological function such as their binding to THC or particular metabolites or analogs thereof.
  • Screening assays are well known to those skilled in the art of immunoassays and immunochemistry, and include without limitation, ELISAs, flow cytometry, immunodiffusion precipitation assays, and the like.
  • the anti-THC antibodies can be used as affinity purification agents.
  • the antibodies are immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art.
  • the immobilized antibody is contacted with a sample containing THC or metabolites or analogs thereof, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the THC, which is bound to the immobilized antibody. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the THC from the antibody.
  • Anti-THC antibodies can also be useful in diagnostic assays for THC or its metabolites, e.g., detecting its presence in specific cells, tissues, or body fluids, such as saliva, serum or urine. Such diagnostic methods are useful in detecting and managing THC abuse, or dosing, such as when performing pharmacokinetic studies.
  • the THC binding antibody can be labeled with a detectable moiety.
  • the antibody can be labeled with a radioisotope, such as 35 S, 14 C, 125 I, 3 H, and 131 I, and the like, using techniques such as those described in Current Protocols in Immunology , Volumes 1 and 2, Coligen et al., Ed., Wiley-Interscience, New York, N.Y., Pubs. (1991). for example. Radioactive labeling can be measured using scintillation counting, for example.
  • the THC binding antibody can also be conjugated with a fluorescent label such as, without limitation, rare earth chelates (e.g., europium chelates), fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red.
  • a fluorescent label such as, without limitation, rare earth chelates (e.g., europium chelates), fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red.
  • the fluorescent labels can be conjugated to the antibody using techniques disclosed in Current Protocols in Immunology , supra, for example. Fluorescence can be quantified using a fluorimeter or UV/vis spectrophotometer using the known extinction coefficient of the fluorescent label.
  • the THC binding antibody can be labeled with various enzyme-substrate labels such as those disclosed in U.S. Pat. No. 4,275,149.
  • the enzyme generally catalyzes a chemical alteration of a chromogenic substrate which can be measured using various techniques, typically optical absorption.
  • the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically.
  • the enzyme may alter the fluorescence or chemiluminescence of the substrate.
  • the labeled anti-THC antibodies can be used for immunohistochemistry as well.
  • the THC binding antibody can be labeled with various particles such as that are not limited to any particular materials, and can comprise metals such as colloidal gold, natural polymers such as latex (e.g., latex particle, latex microparticle), polymer microspheres or microbeads, quantum dots, magnetic particles, glass beads, and the like.
  • the particles are detectable and can be easily visualized by eye or using an instrument, for example, by being colored.
  • Affinity maturation experiments can be performed in silico or in vitro, to optimize the CDR amino acid sequences to increase the binding affinity for THC or a specific metabolite or analog of THC.
  • Affinity maturation techniques are known in the art and include the methods taught, for example, in U.S. Patent Application Publication Nos. 2004/0133357 and 2003/0022240.
  • the framework regions of the THC binding antibodies described herein can be subjected to mutagenesis and/or amino acid substitution, making conservative amino acid substitutions, or nonconservative amino acid substitutions, according to theoretical predictions based on protein structural databases, protein modeling and force field calculations, and the like.
  • energy scoring functions utilizing electrostatic interactions, van der Waals interactions, electrostatic solvation energy, solvent-accessible surface solvation energy, and conformational entropy can be incorporated into the affinity maturation methods.
  • Force fields such as the Amber force field, Charmm force fields, the Discover cvff force fields, the ECEPP force fields, the GROMOS force fields, the OPLS force fields, the MMFF94 force field, the Tripos force field, the MM3 force field, the Dreiding force field, and UNRES force field, and other knowledge-based statistical force field (mean field) and structure-based thermodynamic potential functions can also be helpful in providing guidance for amino acid substitutions to optimize or provide selectivity between the parent ⁇ 9 -THC molecule and THC metabolites or analogs, etc.
  • Analytes to be measured using the present immunoassays include ⁇ 9 -THC and its metabolites, analogs, precursors and derivatives, and the like, without limitation.
  • the anti-THC antibodies bind ⁇ 9 -THC with highest affinity and thereby provide a more sensitive assay for ⁇ 9 -THC, which can be present in unmetabolized form in body fluid (e.g, serum, saliva), in contrast to other THC assays which measure THC metabolites in urine or serum.
  • the anti-THC antibodies bind ⁇ 9 -THC-COOH.
  • the anti-THC antibodies also bind to protein-conjugated THC or protein conjugated THC metabolites with high affinity.
  • the assays utilizing the anti-THC antibodies described herein solve these problems in several ways.
  • ⁇ 9 -THC is an excellent indicator of recent marijuana consumption and may indicate that the person is under the drug's influence.
  • drug levels in saliva can be readily correlated with drug levels in blood, therefore providing a better indicator of current drug use.
  • Saliva samples from a subject can also be collected easily in view of a law enforcement officer without invasion of privacy and without using invasive methods.
  • the present tests are easy to use and can be quickly performed by law enforcement or other personnel on site.
  • saliva samples can be conveniently collected and transferred to the testing device or assay using one of the embodiments of the devices disclosed for fluid collection and testing in U.S. Pat. No. 6,468,474 to Bachand, which discloses a saliva testing and confirmation device that generally includes an expresser cup adapted to receive and compress a foam collection swab pressed into the cup.
  • the swab includes a sponge or foam portion capable of absorbing a fluid specimen such as saliva, and a generally rigid handle for facilitating manipulation thereof.
  • the device also includes a testing and confirmation platform integral with the expresser cup.
  • a channel or groove in the cup is provided for directing a flow of expressed fluid from the expresser into both a test well and a confirmation well.
  • a lateral flow reagent test strip, in fluid communication with the test well may be encased in the platform and partially revealed for analysis of test results.
  • the confirmation well includes a fluid tight cap having a tamper evident seal.
  • the fluid collection and testing devices include a collection vial having a bottom and an open top along with an expresser sized for insertion into the vial open end.
  • the expresser includes a ribbed base enabling fluid to pass therethrough and a support member holds the expresser within the collection vial with the ribbed base in a spaced apart relationship with the vial bottom.
  • a fluid collector includes an absorbent member capturing a fluid and the absorbent member is sized for insertion into the expresser and is compressible against the ribbed base for releasing captured fluid through the ribbed base and into the vial bottom.
  • a catch mechanism is provided for latching the fluid collector to the expresser after insertion into the expresser for enabling simultaneous removal of the expresser and the fluid collector from the collection vial.
  • THC binding antibodies described herein can be used in any diagnostic or analytic assay to determine the presence of THC or its analogs, metabolites, etc.
  • immunoassays and immunochemical applications including, but not limited to, ELISAs, western blotting, immunohistochemical assays, radioimmunoassays, fluorescence immunoassays, immunocytochemical assays, immunoaffinity chromatography, immunoprecipitation, flow cytometry, and the like. Any of these assay format and detection schemes can be performed using the anti-THC antibodies described herein.
  • Preferred assays using the anti-THC antibodies of the invention include, without limitation, ELISAs, histoimmunochemical assays, lateral flow immunoassays, continuous flow displacement assays, and the like, as discussed further below.
  • the assays can be used in competitive or noncompetitive manners and with any number of detection aids such as conjugating the anti-THC antibodies or THC or THC analogs to labels, enzymes, or particles, or the like, as will be readily understood by one skilled in the art.
  • immunoassay The most common type of immunoassay is competitive and non-competitive heterogeneous assays, such as enzyme-linked immunosorbent assays (ELISA).
  • ELISA enzyme-linked immunosorbent assays
  • unlabeled antigen is bound to a solid support or reaction vessel, such as the surface of a microtiter plate or biochip.
  • Biological sample is combined with antigens bound to the reaction vessel, and antibodies (primary antibodies) in the biological sample are allowed to bind to the antigens, thus forming the immune complexes.
  • excess biological sample is removed and the vessel is washed to remove nonspecifically bound antibodies.
  • the immune complexes are then reacted with an appropriate enzyme-labeled anti-immunoglobulin (secondary antibody).
  • the secondary antibody reacts with antibodies in the immune complexes, not with other antigens bound to the vessel.
  • Secondary antibodies specific for binding antibodies of different species, including humans, are well known in the art and are commercially available, such as from Sigma Chemical Co. (St. Louis, Mo.) and Santa Cruz Biotechnology (Santa Cruz, Calif.).
  • the enzyme substrate is added.
  • the enzyme linked to the secondary antibody catalyzes a reaction that converts the substrate into a detectable product.
  • the amount of product is directly proportional to the amount of primary antibodies present in the biological sample.
  • the product is fluorescent or luminescent, which can be measured using technology and equipment well known in the art. It is also possible to use reaction schemes that result in a colored product, which can be measured spectrophotometrically.
  • Sandwich or capture assays can also be used to identify and quantify immune complexes.
  • Sandwich assays are a mirror image of non-competitive ELISAs in that antibodies are bound to the solid phase and antigen in the biological sample is measured. These assays are particularly useful in detecting antigens, having multiple epitopes, that are present at low concentrations. This technique requires excess antibody to be attached to a solid phase, such as the biochip. The bound antibody is then incubated with the biological samples, and the antigens in the sample are allowed to form immune complexes with the bound antibody. The immune complex is incubated with an enzyme-linked secondary antibody, which recognizes the same or a different epitope on the antigen as the primary antibody. Hence, enzyme activity is directly proportional to the amount of antigen in the biological sample. D. M. Kemeny & S. J. Challacombe, ELISA and Other Solid Phase Immunoassays (1988).
  • Typical enzymes that can be linked to secondary antibodies include horseradish peroxidase, glucose oxidase, glucose-6-phosphate dehydrogenase, alkaline phosphatase, .beta.-galactosidase, and urease.
  • Secondary antigen-specific antibodies linked to various enzymes are commercially available from, for example, Sigma Chemical Co. and Amersham Life Sciences (Arlington Heights, Ill.).
  • ELISAs are similar to noncompetitive ELISAs except that enzyme linked antibodies compete with unlabeled antibodies in the biological sample for limited antigen binding sites. Briefly, a limited number of antigens are bound to the solid support. Biological sample and enzyme-labeled antibodies are added to the solid support. Antigen-specific antibodies in the biological sample compete with enzyme-labeled antibodies for the limited number of antigens bound to the solid support. After immune complexes have formed, nonspecifically bound antibodies are removed by washing, enzyme substrate is added, and the enzyme activity is measured. No secondary antibody is required. Because the assay is competitive, enzyme activity is inversely proportional to the amount of antibodies in the biological sample.
  • An alternative competitive ELISA can also be used.
  • limited amounts of antibodies from the biological sample are bound to the surface of the solid support as described herein.
  • Labeled and unlabeled antigens are then brought into contact with the solids support such that the labeled and unlabeled antigens compete with each other for binding to the antibodies on the surface of the solid support.
  • immune complexes After immune complexes have formed, nonspecifically bound antigens are removed by washing.
  • the immune complexes are detected by incubation with an enzyme-linked secondary antibody, which recognizes the same or a different epitope on the antigen as the primary antibody, as described above.
  • the activity of the enzyme is then assayed, which yields a signal that is inversely proportional to the amount of antigen present.
  • Homogeneous immunoassays can also be used when utilizing the anti-THC antibodies of the present invention. Homogeneous immunoassays may be preferred for detection of low molecular weight compounds, such as hormones, therapeutic drugs, and illegal drugs that cannot be analyzed by other methods, or compounds found in high concentration.
  • enzyme-linked antigens are enzyme-linked.
  • enzyme-linked antigens When antibodies in the biological sample bind to the enzyme-linked antigen, steric hindrances inhibit the enzyme activity.
  • Free antigens i.e., not enzyme-linked
  • enzyme activity is directly proportional to the concentration of antigen in the biological sample.
  • Enzymes useful in homogeneous immunoassays include lysozyme, neuraminidase, trypsin, papain, bromelain, glucose-6-phosphate dehydrogenase, and .beta.-galactosidase. T. Persoon, Immunochemical Assays in the Clinical Laboratory, 5 Clinical Laboratory Science 31 (1992).
  • Enzyme-linked antigens are commercially available or can be linked using various chemicals well known in the art, including glutaraldehyde and maleimide derivatives.
  • Fluorescent immunoassays can also be used when utilizing the Anti-THC antibodies of the present invention. Fluorescent immunoassays are similar to ELISAs except the enzyme is substituted for fluorescent compounds called fluorophores or fluorochromes. These compounds have the ability to absorb energy from incident light and emit the energy as light of a longer wavelength and lower energy. Fluorescein and rhodamine, usually in the form of isothiocyanates that can be readily coupled to antigens and antibodies, are most commonly used in the art. D. P. Stites et al., Basic and Clinical Immunology (1994). Fluorescein absorbs light of 490 to 495 nm in wavelength and emits light at 520 nm in wavelength.
  • Tetramethylrhodamine absorbs light of 550 nm in wavelength and emits light of 580 nm in wavelength.
  • Illustrative fluorescence-based detection methods include ELF-97 alkaline phosphatase substrate (Molecular Probes Inc., Eugene, Oreg.); PBXL1 and PBXL-3 (phycobilisomes conjugated to streptavidin) (Martek Biosciences Corp., Columbia, Md.); FITC and Texas Red labeled goat anti-human IgG (Jackson ImmunoResearch Laboratories, Inc., West Grove, Pa.); and B-Phycoerythrin and R-Phycoerythrin conjugated to streptavidin (Molecular Probes Inc.).
  • ELF-97 is a nonfluorescent chemical that is digested by alkaline phosphatase to form a fluorescent molecule. Because of turn over of the alkaline phosphatase, use of the ELF-97 substrate results in signal amplification. Fluorescent molecules attached to secondary antibodies do not exhibit this amplification.
  • Phycobiliproteins isolated from algae, porphyrins, and chlorophylls, which all fluoresce at about 600 nm, are also being used in the art. I. Hemmila, Fluoroimmunoassays and Immunofluorometric Assays, 31 Clin. Chem. 359 (1985); U.S. Pat. No. 4,542,104. Phycobiliproteins and derivatives thereof are commercially available under the names R-phycoerythrin (PE) and Quantum RedTM from, for example, Sigma Chemical Co. In addition, Cy-conjugated secondary antibodies and antigens are useful in immunoassays and are commercially available.
  • PE phytophycoerythrin
  • Quantum RedTM Quantum RedTM
  • Cy-3 for example, is maximally excited at 554 nm and emits light of between 568 and 574 nm. Cy-3 is more hydrophilic than other fluorophores and thus has less of a tendency to bind nonspecifically or aggregate. Cy-conjugated compounds are commercially available from Amersham Life Sciences.
  • Illustrative luminescence-based detection methods include CSPD and CDP star alkaline phosphatase substrates (Roche Molecular Biochemicals); and SuperSignal®. horseradish peroxidase substrate (Pierce Chemical Co., Rockford, Ill.). Chemiluminescence, electroluminescence, and electrochemiluminescenc-e (ECL) detection methods are also attractive means for quantifying antigens and antibodies in a biological sample. Luminescent compounds have the ability to absorb energy, which is released in the form of visible light upon excitation. In chemiluminescence, the excitation source is a chemical reaction; in electroluminescence the excitation source is an electric field; and in ECL an electric field induces a luminescent chemical reaction.
  • Molecules used with ECL detection methods generally comprise an organic ligand and a transition metal.
  • the organic ligand forms a chelate with one or more transition metal atoms forming an organometallic complex.
  • Various organometallic and transition metal-organic ligand complexes have been used as ECL labels for detecting and quantifying analytes in biological samples. Due to their thermal, chemical, and photochemical stability, their intense emissions and long emission lifetimes, ruthenium, osmium, rhenium, iridium, and rhodium transition metals are favored in the art.
  • the types of organic ligands are numerous and include anthracene and polypyridyl molecules and heterocyclic organic compounds.
  • bipyridyl, bipyrazyl, terpyridyl, and phenanthrolyl, and derivatives thereof are common organic ligands in the art.
  • a common organometallic complex used in the art includes tris-bipyridine ruthenium (II), commercially available from IGEN, Inc. (Rockville, Md.) and Sigma Chemical Co.
  • ECL can be performed under aqueous conditions and under physiological pH, thus minimizing biological sample handling.
  • J. K. Leland et al. Electrogenerated Chemiluminescence: An Oxidative-Reduction Type ECL Reactions Sequence Using Triprophyl Amine, 137 J. Electrochemical Soc. 3127-3131 (1990); WO 90/05296; U.S. Pat. No. 5,541,113.
  • the luminescence of these compounds may be enhanced by the addition of various cofactors, such as amines.
  • a tris-bipyridine ruthenium (II) complex may be attached to a secondary antibody using strategies well known in the art, including attachment to lysine amino groups, cysteine suithydryl groups, and histidine imidazole groups.
  • secondary antibodies would recognize ISAs bound to antigens, but not unbound antigens.
  • the tris-bipyridine ruthenium (II) complex would be excited by chemical, photochemical, and electrochemical excitation means, such as by applying current to the biochip.
  • chemical, photochemical, and electrochemical excitation means such as by applying current to the biochip.
  • the excitation would result in a double oxidation reaction of the tris-bipyridine ruthenium (II) complex, resulting in luminescence that could be detected by, for example, a photomultiplier tube.
  • Instruments for detecting luminescence are well known in the art and are commercially available, for example, from IGEN, Inc.
  • Solid state color detection circuitry can also be used to monitor the color reactions on the biochip and, on command, compare the color patterns before and after the sample application.
  • a color camera image can also be used and the pixel information analyzed to obtain the same information.
  • Still another detection method involves detection using a surface plasmon resonance (SPR) chip.
  • SPR surface plasmon resonance
  • the surface of the chip is scanned before and after sample application and a comparison is made.
  • the SPR chip relies on the refraction of light when the molecules of interest are exposed to a light source. Each molecule has its own refraction index by which it can be identified. This method requires precise positioning and control circuitry to scan the chip accurately.
  • Yet another method involves a fluid rinse of the biochip with a fluorescing reagent.
  • the locations that combine with the biological sample will fluoresce and can be detected with a charge-coupled device (CCD) array.
  • CCD charge-coupled device
  • the output of such a CCD array is analyzed to determine the unique pattern associated with each sample. This approach avoids the problems associated with scanning technologies. Speed is not a factor with any of the methods since the chemical combining of sample and reference takes only a few minutes to occur.
  • the GMS 418 Array Scanner uses laser optics to rapidly move a focused beam of light over the biochip.
  • This system uses a dual-wavelength system including high-powered, solid-state lasers that generate high excitation energy to allow for reduced excitation time.
  • the GMS 418 can scan a 22.times.75-mm slide with 10-.mu.m resolution in about 4 minutes.
  • the THC binding antibody is utilized in a lateral flow immunoassay.
  • Lateral Flow immunoassays are described, for example, in U.S. Pat. Nos. 5,770,458 to Klimov, 6,001,658 to Fredrickson and 6,368,876 to Huang. Lateral flow immunoassay is based on the principle of competitive binding. Analytes (e.g., drugs, THC) that may be present in the aqueous sample to be tested (e.g., an oral fluid specimen) compete against their respective conjugate for binding sites on an antibody having specific binding activity for the drug that is conjugated to a visible (e.g., colored) particle.
  • Analytes e.g., drugs, THC
  • aqueous sample to be tested e.g., an oral fluid specimen
  • the particles that can be used are not limited to any particular materials, and can comprise metals such as colloidal gold, natural polymers such as latex (e.g., latex particle, latex microparticle), polymer microspheres or microbeads, quantum dots, magnetic particles, glass beads, and the like.
  • metals such as colloidal gold
  • natural polymers such as latex (e.g., latex particle, latex microparticle), polymer microspheres or microbeads, quantum dots, magnetic particles, glass beads, and the like.
  • the binding sites of the antibody will be saturated and the antibody will not complex with the drug conjugate and form a visible line in the test line region.
  • a drug-positive oral fluid specimen will not generate a colored line in the specific test line region of the strip because of drug competition, while a drug-negative oral fluid specimen will generate a line in the test line region because of the absence of drug competition.
  • a colored line will always appear at the control line region, indicating that proper volume of specimen has been added and membrane wicking has occurred.
  • the lateral immunoassay test typically contains membrane strips coated with drug-protein conjugates (purified bovine albumin) on the test line, an anti-antibody (e.g., a goat polyclonal anti-rabbit or anti-mouse antibody) or anti-BSA antibody against gold-protein conjugate at the control line, and a dye pad which contains particles, such as colloidal gold particles, coated with a monoclonal antibody specific to the drug to be tested (e.g., ⁇ 9 -THC or its metabolites).
  • drug-protein conjugates purified bovine albumin
  • an anti-antibody e.g., a goat polyclonal anti-rabbit or anti-mouse antibody
  • anti-BSA antibody anti-BSA antibody against gold-protein conjugate at the control line
  • a dye pad which contains particles, such as colloidal gold particles, coated with a monoclonal antibody specific to the drug to be tested (e.g., ⁇ 9 -THC or its metabolites).
  • the anti-THC antibody is a recombinant rabbit antibody comprising CDR amino acid sequences selected from L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) and homologous amino acid sequences having at least 85% homology thereto.
  • a lateral flow immunoassay utilizing this antibody provides a much greater sensitivity and specificity for ⁇ 9 -THC than provided by murine anti-THC antibodies tested, as described in Example 4, and is particularly suitable and useful for oral fluid testing for the presence of ⁇ 9 -THC.
  • the anti-THC antibody comprises the amino acid sequence for the light chain variable region:
  • amino acid sequences having at least 85% homology thereto.
  • the amino acid sequence has about 90% or 95%, or even greater homology thereto.
  • the anti-THC antibody comprises the amino acid sequence for the heavy chain variable region: QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGKGLEWIGTIVSGTTYYAS WAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYITGDIWGPGTLVTVSLG (SEQ ID NO:8), or homologous amino acid sequences having at least 85% homology thereto.
  • the amino acid sequence has about 90% or 95%, or even greater homology thereto.
  • the anti-THC antibodies described herein can also be used in a continuous flow displacement immunoassay for ⁇ 9 -THC or its metabolites.
  • a continuous flow displacement immunoassay for cannabinoids the binding kinetics of the cannabinoid analyte (e.g., ⁇ 9 -THC or its metabolites) to the antibody play a very important role.
  • a labeled tracer compound is allowed to compete with the cannabinoid analyte for binding to the antibody.
  • the binding kinetics of the antibody to the tracer compound provide a fast dissociation rate of the bound tracer from the antibody, thereby permitting a rapid binding of the cannabinoid analyte.
  • a continuous flow displacement cannabinoid immunoassay utilizes an antibody having a high affinity for the cannabinoid analyte and a lower affinity for the tracer.
  • the affinity of the antibody for the tracer may be anywhere between 15-100% cross-reactivity.
  • a preferred cannabinoid-based tracer has about 40-80% cross-reactivity for the antibody.
  • a typical continuous flow displacement cannabinoid immunoassay involves a solid-phase immobilized antibody to ⁇ 9 -THC or its metabolites.
  • the antigen binding site of the antibody can be exposed to a synthetic labeled tracer to form a labeled synthetic tracer-antibody complex such that the antigen binding sites of the antibody are saturated with the labeled synthetic tracers.
  • a biological sample suspected of containing the analyte ⁇ 9 -THC is continuously flowed past the solid-phase immobilized antibody-labeled synthetic tracer complex, if ⁇ 9 -THC is present in the sample, the cannabinoid analyte binds to the antibody and displaces the labeled synthetic tracer. Detection of the labeled tracer downstream from the binding point indicates the presence and/or quantity of the analyte present in the biological sample. See U.S. Pat. No. 5,183,740 to Ligler, which is incorporated by reference herein.
  • the success of developing a continuous flow displacement immunoassay is based on the selection of antibody and tracer to achieve a fast dissociation rate of the bound tracer from the antibody, thereby permitting a rapid binding of the analyte.
  • the ideal continuous flow displacement immunoassay utilizes a system where the antibody has a high affinity for the analyte, and a lower affinity for the tracer.
  • the affinity of the antibody for the tracer may be anywhere between 15-100% cross-reactivity.
  • a preferred THC-based tracer has about 40-80% cross-reactivity for the antibody.
  • condition for displacing a tracer from the recognition molecule is preferably carried out in non-equilibrium condition or kinetic flow reaction, such that the sample flows past the recognition molecule-tracer complex at a rate where a stable equilibrium state between the recognition molecule, tracer, and analyte has not been achieved.
  • anti-THC antibodies described herein can also be utilized in additional useful commercially marketed immunoassay formats, for example, the OnTrak TesTcup, On•Site CupKitTM, On•Site OraLab®, OnTrak TesTstik assay systems marketed by Varian, Inc.
  • the anti-THC antibodies described herein can also be utilized in pharmaceutical and therapeutic applications, for example, to treat overdose of THC or reduce serum concentrations of THC.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins;
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active compound preferably those with complementary activities that do not adversely affect each other.
  • it may be desirable to further provide an antipsychotic agent.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients can also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • compositions to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • Therapeutic formulations of the antibodies can be prepared for storage by mixing the antibodies having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Sustained-release preparations comprising the antibodies can be prepared.
  • suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibodies, which matrices are in the form of shaped articles, e.g., films, or microcapsule.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-1S methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)
  • poly-D-( ⁇ )-3-hydroxybutyric acid While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated antibodies When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • the antibodies can be administered to the patient by a variety of different means and will vary depending upon the intended application. As one skilled in the art would recognize, administration of the therapeutic compositions can be carried out in various fashions, and more typically by parenteral injection into body cavity or vessel, e.g., intraperitoneal, intravenous, intralymphatic, intratumoral, intramuscular, interstitial, intraarterial, subcutaneous, intralesional, intraocular, intrasynovial, intraarticular.
  • parenteral injection into body cavity or vessel e.g., intraperitoneal, intravenous, intralymphatic, intratumoral, intramuscular, interstitial, intraarterial, subcutaneous, intralesional, intraocular, intrasynovial, intraarticular.
  • topical administration including, but not limited to, dermal, ocular and rectal; transdermal, via passive or active means, e.g., using a patch, a carrier, or iontophoresis; transmucosal, e.g., sublingual, buccal, rectal, vaginal, or transurethral; oral, e.g., gastric or duodenal; via inhalation, e.g., pulmonary or nasal inhalation, using e.g., a nebulizer.
  • topical administration including, but not limited to, dermal, ocular and rectal; transdermal, via passive or active means, e.g., using a patch, a carrier, or iontophoresis; transmucosal, e.g., sublingual, buccal, rectal, vaginal, or transurethral; oral, e.g., gastric or duodenal; via inhalation, e.g., pulmonary or nasal in
  • about 1 ⁇ g/kg to about 50 mg/kg (e.g., 0.1-20 mg/kg, 0.5-15 mg/Kg, and 1-10 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily or weekly dosage might range from about 1 ⁇ g/kg to about 20 mg/kg or more, depending on the factors mentioned above.
  • the treatment is repeated until the THC levels have been reduced to a desired level.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays, including, for example, immunoassays of THC and/or its metabolites in urine or saliva.
  • An antibody having specific binding for THC was prepared by the following procedures:
  • New Zealand White rabbits were immunized. Rabbits were injected subcutaneously with 0.4 mg of THC-KLH in complete Freund's adjuvant. After the initial immunization, animals were boosted 5 more times every 21 days in the same manner with incomplete Freund's adjuvant and the sera tested by immunoassay and immunohistochemical staining. The rabbit with the best titer in the immunoassay and IHC (immunohistochemistry) was selected for a final intravenous boost of 0.4 mg of THC-KLH in saline, four days before removal of the spleen.
  • Fusions were performed using conventional methodology: 1.5-3 ⁇ 10 8 lymphocytes from an immunized rabbit and the fusion partner (240E-w) were fused at a ratio of 2:1 with PEG 4000 at 37° C. in serum-free medium. The cells were distributed in 96-well cell culture plates at approximately 1 ⁇ 10 5 lymphocytes per well in medium with 15% FBS (or FCS), and after 48 hr HAT medium was added. Medium was changed 2-3 times before screening. Hybridoma colonies were ready for screening in 3-5 weeks. Supernatants were tested for the presence of antibody, specific for the immunogen, by ELISA. Immunohistochemistry was used as a secondary screening assay. The hybridomas were sub-cloned by limit dilution. For feeder cells, the fusion partner 240E-w at 2 ⁇ 10 4 cells per well was used.
  • ELISA was performed in 96-well micro-titer plates that had been coated overnight with immunogen at 1.0 ⁇ g/ml, plates were then saturated with 2% BSA, followed by incubation with the antibody-containing supernatant for 1 hr at room temperature. After washing with PBS-Tween, alkaline phosphatase conjugated goat anti-rabbit immunoglobulin (IgG) was added and incubation continued for another hour; plates were washed again and developed in the presence of P-NPP (para-nitrophenyl phosphate). Color was read at 405 nm with a plate reader (Multiskan MCC/340 from Fisher Scientific).
  • Tissue culture supernatants containing rabbit monoclonal antibody against ER were incubated with Protein A immobilized on a column.
  • the column was washed extensively with PBS to remove nonspecific binding proteins until the OD 280 was less than 0.01.
  • the rabbit IgGs were eluted with 0.2 M, pH 2.5 citrate. The eluates was dialyzed against PBS overnight at 4° C. (28).
  • ELISA was performed to determine antibody affinities. 96-well micro-titer plates was coated overnight with the THC conjugated BSA or a human recombinant protein (1-300aa) at 1.0 ⁇ g/ml, respectively. The plates were saturated with 2% BSA, followed by incubation with the antibody serially diluted (200 nM, 100 nM, 50 nM, 25 nM, 12.5 nM, 6.25 nM, 3.125 nM, 1.56 nM, 0.78 nM, 0.39 nM, and 0.2 nM).
  • ER clone SP1 was incubated with the wells coated with the BSA conjugated ER peptide and clone 1D5 was incubated with wells coated with ER alpha recombinant protein. Samples were in duplicate. After washing with PBS-Tween, HRP-conjugated goat anti-rabbit and mouse IgGs (Spring Bioscience Corporation, CA) were added. The plates were washed again, and were developed with TMB solution for 15 minutes. The reaction was stopped by adding 0.5N H 2 SO 4 . Optical density (OD) was measured at 450 nm.
  • mRNA was isolated from rabbit hybridomas by using Qiagen TurboCapture mRNA kit.
  • cDNA was made directly in the TurboCapture tube by solid phase synthesis.
  • Rabbit IgG cDNAs were amplified with heavy or light chain specific primers by PCR.
  • L chain the PCR product was sequenced and a consensus sequence was found from three independent mRNA and PCR reactions.
  • the PCR product was digested with restriction enzymes and cloned into an expression vector. A cDNA clone with the consensus PCR sequence was selected for expression.
  • V H variable region
  • V H cDNA was purified and recovered from an agarose gel.
  • the purified V H product was cloned into pCR 2.1-TOPO vector (Invitrogen) and cDNA clones were screened.
  • the V H cDNA in the desired clone was subcloned into an expression vector with rabbit IgG heavy chain constant region (CH).
  • CH rabbit IgG heavy chain constant region
  • both light and heavy chain expression constructs were transfected into HEK 293 by using 293 fectin (Invitrogen) as a vehicle. Supernatant containing recombinant antibody transiently expressed from the cells were harvested after 5-day transfection.
  • V L The nucleotide and amino acid sequences for the THC binding antibody variable regions of the light chain (V L ) and the variable region of the heavy chain (V H ) are depicted below:
  • Antibodies to be utilized in lateral flow immunoassay systems were conjugated to particles that can be visualized. Conjugation conditions were tested for each antibody lot, and monobasic/dibasic phosphate buffer pH was adjusted to optimize the conjugation, which was typically between pH 6.5 and 8.5. Antibody was diluted to 0.1 mg/ml using monobasic/dibasic phosphate buffer at room temperature and mixed with gold particles (30 nm) in the proportion 20 ⁇ l antibody per 1 mL gold for 5 minutes. 10% BSA was added to create a 1:10 dilution BSA: total volume Gold solution, and mixing was continued for five (5) minutes. The sample was centrifuged at 10,000 rpm at 4° C. for forty (40) minutes, and the supernatant removed to achieve a final suspension at 10% (w/w) gold. The gold conjugated antibodies are then utilized in the lateral flow immunoassay system, such as the system sold under the tradename OraLab®.
  • a murine monoclonal anti-THC antibody used in a lateral flow immunoassay was used as a comparator to assess the sensitivity of a recombinant rabbit anti-THC antibody in the same immunoassay system.
  • the murine anti-THC antibody chosen for comparison exhibited a sensitivity higher or comparable to other available anti-THC antibodies used in lateral flow immunoassays.
  • both the mouse monoclonal anti-THC antibody and the recombinant rabbit anti-THC antibody were conjugated to gold particles using the same procedures.
  • the conjugation buffer pH was adjusted to assure the best conjugation. All other materials and procedure were identical.
  • Both a murine anti-THC antibody strip and a rabbit anti-THC antibody strip were prepared on the same card with the murine antibody strip on the left and rabbit antibody strip on the right.
  • the recombinant rabbit anti-THC antibody is more sensitive than the reference mouse anti-THC antibody.
  • the reading with the murine anti-THC antibody is only slightly decreased, and only at THC concentrations of 200 ng/mL does the murine anti-THC antibody clearly indicate that THC was present in the sample solution tested.
  • the reading for the recombinant rabbit anti-THC antibody was almost completely abolished at a sample concentration of 50 ng/mL THC, indicating the presence of THC in the sample at the lowest concentration of THC tested.
  • a sensitivity of 35 ng/mL or lower is achievable.
  • a lateral flow immunoassay system for the detection of THC is provided to an individual.
  • the individual is instructed to open the collector bag, place the collector foam inside his or her mouth for three minutes, and completely saturate the collector foam with saliva.
  • the individual or other user is instructed to place the OraLab® cassette (Varian, Inc. 25200 Commercentre Drive, Lake Forest, Calif.) on a flat surface and lift the confirmation well cap to assure unobstructed saliva flow into the well.
  • the individual then removes the collector foam from his or her mouth, places it vertically into the sample well on top of the device, and pushes the collector downward while counting slowly to five until it stops.
  • the user is instructed to push the cap down firmly to seal the confirmation well after ten minutes have passed, and to remove the label covering the results windows and interpret the results.
  • the saliva sample is negative for the presence of THC
  • the antibody-gold particles migrate with the fluid flow by capillary action until they reach the THC loaded line, where they stop and line up to indicate a negative result.
  • the saliva sample is positive for the presence of THC
  • the antibody-gold particles migrate with the fluid flow but do not stop at the THC loaded line and instead continue upwards to the positive zone, indicating a positive result.

Abstract

Antibodies having specific binding for the parent THC (Δ9-THC) and its major metabolites are provided which present a significant increase in sensitivity of immunoassays such as lateral flow immunoassays and ELISA for THC. The present invention also provides a rabbit hybridoma producing the antibody as a monoclonal antibody, a recombinant antibody, further molecularly engineered recombinant antibodies against parent Δ9-THC and its metabolites and cell lines producing the recombinant antibodies. The invention also provides applications of the antibody in immunoassays, particularly lateral flow immunoassays, specifically applications in detecting THC in body fluids, particularly saliva, and kits for determining the presence of THC.

Description

    FIELD OF THE INVENTION
  • This invention relates generally to immunoassays for drugs of abuse and the like.
  • BACKGROUND OF THE INVENTION
  • Marijuana is a member of the hemp family and is known to contain significant amounts of cannabinoids. In particular, the most important cannabinoid is Δ9-Tetrahydrocannabinol (Δ9-THC), the major physiologically active constituent of marijuana. Δ9-THC is a controlled substance because it has both sedative and depressant-like effects on the cardiovascular and central nervous systems, as opposed to cannabidiol, a non-psychoactive constituent of marijuana. Through smoking marijuana, Δ9-THC is rapidly absorbed from the lungs into the blood stream and metabolized through 11-norΔ9-THC to a series of polar metabolites with 11-nor-Δ9-THC-carboxylic acid as the primary metabolite.
  • Due to the common abuse of cannabinoids, there is a need for non-invasive and rapid tests to detect the presence of these controlled drugs in biological specimens. Currently, cannabinoids in biological samples can be detected by a number of techniques such as thin layer chromatography (TLC), gas chromatography/mass spectrometry (GC/MS), radioimmunoassay, enzyme immunoassay or continuous flow displacement immunoassay (U.S. Pat. No. 6,686,208). Depending upon assay sensitivity, cannabinoid metabolites may be detected in the urine for up to 10 days in occasional smokers and 36 days in chronic smokers. See U.S. Pat. No. 5,264,373.
  • Tetrahydrocannabinol, the active ingredient in the marijuana plant (cannabis sativa), is detectable in saliva shortly after use. The detection of the drug in saliva is thought to be primarily due to the direct exposure of the drug to the mouth (oral and smoking administrations) and the subsequent sequestering of the drug in the buccal cavity. The non-metabolized drug, Δ9-THC is the main form of THC present in the marijuana smoker's oral fluid sample. Thus the presence of Δ9-THC in oral fluids is an excellent indicator of recent marijuana consumption and may indicate that the person is under the drug's influence. Detection of the parent THC therefore is of great interest for road side testing by law enforcement, on-site testing in work places and many other circumstances.
  • Previous studies have shown a time window of detection for THC in saliva of up to 14 hours after drug use. In recent years, there have been efforts to utilize saliva for drug monitoring for the presence of Δ9-THC, for pharmacokinetic studies or for the management of patients in chronic drug therapy. Although saliva testing does not quantify the blood concentration of the drug, saliva testing is of particular interest since the presence of Δ9-THC in the saliva is indicative of recent smoking of cannabinoids. The concentration of Δ9-THC in the blood is usually much higher than the drug concentration in saliva.
  • The use of saliva THC assays and procedures however are still problematic. Due to the short detectable time window of the parent THC in the oral fluid, low quantity, the low THC solubility and “stickiness” in aqueous solution, and sensitivity limitations of currently available murine anti-THC antibodies, current lateral flow immunoassays for parent THC can not meet the expected sensitivity requirement recommended by the Substance Abuse and Mental Health Services Administration (SAMHSA).
  • For example, the THC assay contained within the Oral Fluid Drug Screen Device (ORATECT® II, Branan Medical Corp., Irvine, Calif.) is purported to detect the presence of THC-COOH. However, this assay requires the presence of the THC metabolite THC-COOH, and does not detect the parent Δ9-THC molecule with a high sensitivity.
  • The THC “One Step Marijuana Test Strip” (Biomerica, Inc. Newport Beach, Calif.) is reported to be a rapid chromatographic immunoassay for the detection of 11-nor-9-THC-9 COOH in human urine at a cut-off concentration of 50 ng/mL. However, this assay system cannot be used to detect the parent Δ9-THC molecule in saliva and requires urine collection and testing.
  • A fluorescence immunoassay has been reported for the detection and quantitation of tetrahydrocannabinols in oral fluids for use in testing for marijuana abuse. The lower detection limits were reported to be 1.5 ng/mL for (1)-, 9-THC and 5.5 ng/mL for (1)9-Carboxy-11-nor-, 9-THC and to require a total assay reaction time of less than 10 minutes. However, fluorescence detection is required for this assay, necessitating the use of expensive equipment and limiting the availability and widespread usage of this assay.
  • The use of a continuous flow displacement immunoassay technology (U.S. Pat. No. 5,183,740 to Ligler) has been demonstrated for detection of controlled drugs in saliva and urine. See also U.S. Pat. No. 6,686,208 to Wang. However, this assay procedure requires the use of a tracer molecule capable of binding to the same antigen or ligand binding site(s) of an antibody or receptor to Δ9-THC or its metabolites, to compete with Δ9-THC or its metabolites in an immunoassay, and the binding affinity of the antibody to the tracer molecules must be lower than the binding affinity of the antibody to the analyte to facilitate effective displacement. Using a Δ9-THC-based or Δ9-THC-analog based tracers is problematic because the Δ9-THC analyte does not effectively displace the Δ9-THC-based or Δ9-THC-analog based tracer. This results in a lower sensitivity of the assay.
  • There exist more sensitive immunoassay methods such as ELISA, however, there is a great need in the art for more sensitive assays and assay systems for ascertaining the presence of Δ9-THC that are inexpensive to prepare, purchase and operate, that operate in a noninvasive manner and that are easy to use for persons unskilled in laboratory assay procedures. To satisfy this need, more sensitive and specific antibodies for use in detecting Δ9-THC are required for use in lateral flow immunoassays. In particular, there is a need for a test comprising a sensitive and specific antibody having binding affinity for the parent Δ9-THC molecule, in order to detect the presence of the parent Δ9-THC molecule in saliva.
  • These and other deficiencies in the art are addressed by the invention described below.
  • SUMMARY OF THE INVENTION
  • Accordingly, it is a primary object of the invention to address the aforementioned need in the art by providing an antibody having specific binding for THC and its metabolites, wherein said antibody comprises CDR amino acid sequences selected from the group consisting of L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) or homologous amino acid sequences having at least 85% homology thereto. In certain embodiments, the amino acid sequence for the light chain variable region comprises the sequence:
  • QVLTQTPSPVSAAVGGTVTINCQASQSVYNNNQLSWYQQKPGQPPKLLIYGTSNLASG VPSRFKGSGSGTQFTLTISSVQCDDAATYYCQGGYTSGDGIAFGGGTEVVVK (SEQ ID NO:7), or homologous amino acid sequences having at least 85% homology thereto. In certain other embodiments, the amino acid sequence for the heavy chain variable region comprises the sequence:
    QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGKGLEWIGTIVSGTTYYAS WAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYITGDIWGPGTLVTVSLG (SEQ ID NO:8), or homologous amino acid sequences having at least 85% homology thereto.
  • In certain preferred embodiments, the antibody is a monoclonal antibody produced by a hybridoma. In additional preferred embodiments, the antibody is a recombinant antibody produced by a host cell. In a preferred embodiment, the antibody is a rabbit antibody.
  • In additional aspects, a host cell line is provided that expresses an antibody having specific binding for Δ9-THC and its metabolites, wherein said antibody comprises CDR amino acid sequences selected from the group consisting of L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) or homologous amino acid sequences having at least 85% homology thereto. The host cell can be any cell capable of expressing antibody. In a preferred embodiment, the host cell is a HEK 293 cell.
  • In an additional aspect, an immunoassay for detecting the presence of THC and its metabolites in a sample is provided. Preferably the immunoassay utilizes an antibody having specific binding for THC and its metabolites, wherein said antibody comprises CDR amino acid sequences selected from the group consisting of L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) or homologous amino acid sequences having at least 85% homology thereto. In a preferred embodiment, the antibody is labeled or conjugated to a solid support such as microparticles or a solid surface. In a particularly preferred embodiment, the immunoassay is a lateral flow immunoassay strip for detecting the presence of THC and/or its metabolites in a fluid sample.
  • In yet another aspect, a method is provided for detecting the presence of THC in a sample, comprising the steps of: (1) providing a lateral flow immunoassay system comprising an antibody having specific binding for Δ9-THC; (2) contacting said lateral flow immunoassay system with a sample (e.g., an extract or sample of saliva obtained from an individual) to be tested for the presence of Δ9-THC in saliva; (3) performing the lateral flow immunoassay; (4) determining whether the immunoassay is positive or negative for the presence of Δ9-THC in the sample.
  • In an additional aspect, a kit is provided for testing for the presence of THC in saliva, comprising a lateral flow immunoassay strip for THC, and instructions for performing the test.
  • Additional objects, advantages and novel features of the invention will be set forth in part in the description which follows, and in part will become apparent to those skilled in the art upon examination of the following, or may be learned by practice of the invention.
  • DETAILED DESCRIPTION OF THE INVENTION I. Definitions and Overview
  • Before the present invention is described in detail, it is to be understood that unless otherwise indicated this invention is not limited to specific amino acid sequences, conjugation methods, materials and test systems, or the like, as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to limit the scope of the present invention.
  • It must be noted that as used herein and in the claims, the singular forms “a,” “and” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a particle” includes two or more particles; reference to “an antibody” includes two or more antibodies, and so forth.
  • Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range, and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
  • The term “specific binding” refers to a binding affinity of an antibody to THC or its metabolites and analogs of at least 106 M−1, more preferably between about 107 and about 1011 M−1, or typically between about 108 and 1010 M−1.
  • The term “THC” refers to tetrahydrocannabinols in general, and more particularly, to the parent molecule commonly found in marijuana, Δ9-THC.
  • The term “antibody” is used in its broadest sense and includes the antibody classes such as IgG (and its subclasses IgG1, IgG2, etc.), IgA, IgD, IgE, IgM, as well as Fab, Fab2, Fv, diabodies, and other forms of antibody variants, constructs and fragments known in the art.
  • The present invention provides an isolated antibody having specific binding for the parent THC (Δ9-THC) and/or its major metabolites, which is a significant advance over the performance of other antibodies known in the art which are not sufficiently sensitive for the parent Δ9-THC. Exhaustive testing was performed on additional murine anti-THC antibodies using ELISA assays but none of the murine antibodies provided satisfactory performance or binding characteristics for Δ9-THC.
  • The antibodies described herein present a significant increase in sensitivity of immunoassays such as lateral flow immunoassays and ELISA for THC and allow the determination of the presence of Δ9-THC as well as its metabolites with higher sensitivity than previously possible. The superior performance characteristics are demonstrated in Example 4.
  • The present invention also provides a hybridoma cell clone producing the antibody as a monoclonal antibody, a recombinant antibody, further molecularly engineered recombinant antibodies and host cells producing the recombinant antibodies. The invention also provides methods of using the antibody in immunochemical applications and immunoassays, particularly lateral flow immunoassays, and methods for detecting THC in body fluids, particularly saliva, and kits for determining the presence of THC.
  • The invention is described in more detail below with respect to each of these embodiments.
  • II. Antibodies
  • The present invention provides high sensitivity anti-THC antibodies having antigen binding regions for detecting the presence of THC and its analogs in body fluids. Any antibody having the requisite sensitivity and binding affinity can be utilized. Preferred antibodies include rabbit monoclonal antibodies. Particularly preferred antibodies include CDR sequences as follows: L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6). In addition, preferred antibodies comprise the amino acid sequence for the light chain variable region:
  • QVLTQTPSPVSAAVGGTVTINCQASQSVYNNNNQLSWYQQKPGQPPKLLIYGTSNLASG VPSRFKGSGSGTQFTLTISSVQCDDAATYYCQGGYTSGDGIAFGGGTEVVVK (SEQ ID NO:7), and the heavy chain variable region:
    QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGKGLEWIGTIVSGTTYYAS WAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYITGDIWGPGTLVTVSLG (SEQ ID NO:8), with the CDR sequences (SEQ ID NOS: 1-6) indicated by underlining.
  • Conservative amino acid substitutions of these sequences are also encompassed within the scope of the anti-THC antibodies. Conservative amino acid substitutions are well known to those of skill in the art, and are generally defined as amino acid replacements that preserve the structure and functional properties of proteins. For example, the PAM250 and Blosum 45 similarity matrices can be utilized to determine which amino acids are most likely to be acceptable substitutions to an existing sequence without causing loss of native structure and function of the protein.
  • PAM 250 Amino Acid Similarity Matrix
    C 12
    G −3 5
    P −3 −1 6
    S 0 1 1 1
    A −2 1 1 1 2
    T −2 0 0 1 1 3
    D −5 1 −1 0 0 0 4
    E −5 0 −1 0 0 0 3 4
    N −4 0 −1 1 0 0 2 1 2
    Q −5 −1 0 −1 0 −1 2 2 1 4
    H −3 −2 0 −1 −1 −1 1 1 2 3 6
    K −5 −2 −1 0 −1 0 0 0 1 1 0 5
    R −4 −3 0 0 −2 −1 −1 −1 0 1 2 3 6
    V −2 −1 −1 −1 0 0 −2 −2 −2 −2 −2 −2 −2 4
    M −5 −3 −2 −2 −1 −1 −3 −2 0 −1 −2 0 0 2 6
    I −2 −3 −2 −1 −1 0 −2 −2 −2 −2 −2 −2 −2 4 2 5
    L −6 −4 −3 −3 −2 −2 −4 −3 −3 −2 −2 −3 −3 2 4 2 6
    F −4 −5 −5 −3 −4 −3 −6 −5 −4 −5 −2 −5 −4 −1 0 1 2 9
    Y 0 −5 −5 −3 −3 −3 −4 −4 −2 −4 0 −4 −5 −2 −2 −1 −1 7 10
    W −8 −7 −6 −2 −6 −5 −7 −7 −4 −5 −3 −3 2 −6 −4 −5 −2 0 0 17
    C G P S A T D E N Q H K R V M I L F Y W

    The PAM 250 matrix above is arranged such that similar amino acids are close to each other, therefore regions along the diagonal of the matrix contain only positive scores. These regions provide an objective basis for defining conservative substitutions, i.e., amino acids that replace each other more frequently than would be expected from random replacements are shown on the diagonal. The variation in the diagonal terms of the matrix reflects both how often an amino acid is found in protein sequences and how often it is observed to be replaced by other amino acids. Thus rare amino acids which are replaced infrequently have the highest scores.
  • Blosum 45 Amino Acid Similarity Matrix
    G 7
    P −2 9
    D −1 −1 7
    E −2 0 2 6
    N 0 −2 2 0 6
    H −2 −2 0 0 1 10
    Q −2 −1 0 2 0 1 6
    K −2 −1 0 1 0 −1 1 5
    R −2 −2 −1 0 0 0 1 3 7
    S 0 −1 0 0 1 −1 0 −1 −1 4
    T −2 −1 −1 −1 0 −2 −1 −1 −1 2 5
    A 0 −1 −2 −1 −1 −2 −1 −1 −2 1 0 5
    M −2 −2 −3 −2 −2 0 0 −1 −1 −2 −1 −1 6
    V −3 −3 −3 −3 −3 −3 −3 −2 −2 −1 0 0 1 5
    I −4 −2 −4 −3 −2 −3 −2 −3 −3 −2 −1 −1 2 3 5
    L −3 −3 −3 −2 −3 −2 −2 −3 −2 −3 −1 −1 2 1 2 5
    F −3 −3 −4 −3 −2 −2 −4 −3 −2 −2 −1 −2 0 0 0 1 8
    Y −3 −3 −2 −2 −2 2 −1 −1 −1 −2 −1 −2 0 −1 0 0 3 8
    W −2 −3 −4 −3 −4 −3 −2 −2 −2 −4 −3 −2 −2 −3 −2 −2 1 3 15
    C −3 −4 −3 −3 −2 −3 −3 −3 −3 −1 −1 −1 −2 −1 −3 −2 −2 −3 −5 12
    G P D E N H Q K R S T A M V I L F Y W C
  • For a discussion of widely used matrices (including both PAM and Blosum matrices), see the following references: generally: “Amino acid substitution matrices from an information theoretic perspective,” Altschul, S. F. (1991) Journal of Molecular Biology 219, 555-665; PAM matrices for proteins: “A model of evolutionary change in proteins,” Dayhoff, M. O., Schwartz, R. M., Orcutt, B. C. (1978) In “Atlas of Protein Sequence and Structure” 5(3) M. O. Dayhoff (ed.), pp. 345-352, National Biomedical Research Foundation, Washington; PAM matrices for nucleic acids: “Improved Sensitivity of Nucleic Acid Database Search Using Application-Specific Scoring Matrices,” States, D. J., Gish, W., Altschul, S. F. (1991) Methods: A companion to Methods in Enzymology 3(1), 66-77; Blosum (Block sums) matrices: “Amino acid substitution matrices from protein blocks,” Steven Henikoff and Jorja G. Henikoff, (1992) Proc. Natl. Acad. Sci. USA 89 (Biochemistry), 10915-10919; Comparison of Amino Acid substitution matrices with visual representation of the differences:
  • “A Structural Basis of Sequence Comparisons: An evaluation of scoring methodologies,” (1993) M. S. Johnson and J. P. Overington, Journal of Molecular Biology 233, 716-738; and “Performance Evaluation of Amino Acid Substitution Matrices” (1993) Steven Henikoff and Jorja G. Henikoff, Proteins: Structure, Function, and Genetics 17, 49-61.
  • The present invention also includes amino acid sequences which are homologous to SEQ ID NOS: 1-6, and the fragments above. As used herein, “homologous amino acid sequence” is any polypeptide which is encoded, in whole or in part, by a nucleic acid sequence which hybridizes at 25° C. to 35° C. below critical melting temperature (Tm), to any portion of the nucleic acid sequence encoding SEQ ID NOS: 1-6, or specifically to the nucleic acid sequences encoding the six CDR amino acid sequences, as shown below:
  • CDR-L1:
    (SEQ ID NO: 9)
    CAG GCC AGT CAA AGT GTT TAT AAT AAC AAC CAA TTA
    TCC;
    CDR-L2:
    (SEQ ID NO: 10)
    GGT ACA TCC AAT CTG GCA TCT;
    CDR-L3:
    (SEQ ID NO: 11)
    CAA GGC GGT TAT ACT AGT GGT GAT GGT AAT GCT;
    CDR-H1:
    (SEQ ID NO: 12)
    GGA TTC TCC CTC AGT AAC TAT GTA TTG GCC;
    CDR-H2:
    (SEQ ID NO: 13)
    ACC ATT GTT AGC GGT ACC ACA TAC TAC GCG AGT TGG;
    CDR-H3:
    (SEQ ID NO: 14)
    GGT TTG CCT CAT TAT ATT ACT GGG GAC ATC.
  • A homologous amino acid sequence can be one that differs from an amino acid sequence shown in SEQ ID NOS: 1-6 by one or more conservative amino acid substitutions. Homologous amino acid sequences include sequences that are identical or substantially identical to SEQ ID NOS: 1-6. By “amino acid sequence substantially identical” is meant a sequence that is at least 85%, preferably 90%, more preferably 95%, most preferably 97%-99% identical to an amino acid sequence of reference and that preferably differs from the sequence of reference by a majority of conservative amino acid substitutions. Conservative amino acid substitutions are substitutions among amino acids of the same class. These classes include, for example, amino acids having uncharged polar side chains, such as asparagines (N), glutamine (Q), serine (S), threonine (T), and tyrosine (Y); amino acids having basic side chains, such as lysine (K), arginine (R), and histidine (H); amino acids having acidic side chains, such as aspartic acid (D) and glutamic acid (E); and amino acids having nonpolar side chains, such as glycine (G), alanine (A), valine (V), leucine (L), isoleucine (I), praline (P), phenylalanine (F), methionine (M), tryptophan (W), and cysteine (C). Homology can be measured using sequence analysis software such as Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705. Amino acid sequences are aligned to maximize identity. Gaps can be artificially introduced into the sequence to attain proper alignment. Once the optimal alignment has been set up, the degree of homology is established by recording all of the positions in which the amino acids of both sequences are identical, relative to the total number of positions.
  • Homologous polynucleotide sequences are defined in a similar way. Preferably, a homologous sequence is one that is at least 45%, more preferably 50%, 55%, 60%, 65%, 70%, 75%, 80%, and even more preferably 85%, 87%, 90%, 93%, 96% and most preferably 99% identical to a nucleotide sequence encoding amino acid sequences SEQ ID NOS:1-6.
  • In designing antigen binding regions (i.e., complementarity determining regions), typically, conservative amino acid substitutions are made such that the structure, hydrophobicity, charge, etc. of a particular amino acid or amino acid sequence is unchanged or changed only minimally. The objective is to maintain antigen binding affinity, but allow for maturation or adjustment of the binding affinity to identify additional amino acid sequences that provide useful binding characteristics to a given antigen. Conservative amino acid substitutions thus involve replacing one or more amino acids at a time with amino acids of the same type, i.e., amino acids having charged (basic or acidic) or uncharged polar side chains, or nonpolar side chains, for example, substitution of an uncharged amino acid such as asparagine (N), with another uncharged amino acid such as glutamine (Q), serine (S), threonine (T), or tyrosine (Y; substitution of a basic amino acid such as lysine (K), with another basic amino acid such as arginine (R) or histidine (H); substitution of an acidic amino acid such as aspartic acid (D), with another acidic amino acid such as glutamic acid (E); or substitution of a nonpolar amino acid such as glycine (G) with another nonpolar amino acid such as alanine (A), valine (V), leucine (L), isoleucine (I), methionine (M), proline (P), phenylalanine (F), tryptophan (W), or cysteine (C). Thus, conservative amino acid sequence substitutions are within the scope of the appended claims, so long as the overall sequence remains at least 85% homologous to each of SEQ ID NOS: 1-6. For example, such substitutions give rise to additional preferred CDR sequences, such as the following variations in L1, L2, L3, H1, H2, and H3:
  • L1: QATQSVYNNNQLS (SEQ ID NO:15), QASQSVYNNNQLT (SEQ ID NO:16), QASQTVYNNNQLS (SEQ ID NO:17), QASQSAYNNNQLS (SEQ ID NO:18), QVSQSVYNNNQLS (SEQ ID NO:19), QASQSVYNNNQIS (SEQ ID NO:20), QASQSVFNNNQLS (SEQ ID NO:21), QASQSVYNNNQLS (SEQ ID NO:22), and the like;
  • L2: GTSNLAS (SEQ ID NO:23), GSSNLAS (SEQ ID NO:24), GTSNLAT (SEQ ID NO:25), GTSNIAS (SEQ ID NO:26), GTSNLVS (SEQ ID NO:27), GTSNLAS (SEQ ID NO:28), GSSNIAS (SEQ ID NO:29), GTTNLVS (SEQ ID NO:30), and the like;
  • L3: QGGYTSGDGIA (SEQ ID NO:31); QGAYTSGDGIA (SEQ ID NO:32); QGGYSSGDGIA (SEQ ID NO:33); QGGYTSGEGIA (SEQ ID NO:34); QGGYTSGDGLA (SEQ ID NO:35); QGGYTSGDGIV (SEQ ID NO:36); QGGFTSGDGIA (SEQ ID NO:37); QGGYTTGEGIA (SEQ ID NO:38), and the like;
  • H1: GFSLSNYVLA (SEQ ID NO:39), GYSLSNYVLA (SEQ ID NO:40), GFTLSNYVLA (SEQ ID NO:41), GFSLTNYVLA (SEQ ID NO:42), GFSLSNFVLA (SEQ ID NO:43), GFSLSNYVIA (SEQ ID NO:44), GFSLSNYVLV (SEQ ID NO:45), and the like;
  • H2: TIVSGTTYYASW (SEQ ID NO:46), TLVSGTTYYASW (SEQ ID NO:47), TIVSGTSYYASW (SEQ ID NO:48), TIVSGTTYYVSW (SEQ ID NO:49), TIVSGTTFYASW (SEQ ID NO:50), TIVSGSTYYATW (SEQ ID NO:51), and the like;
  • H3: GLPHFITGDI (SEQ ID NO:52), GIPHYITGDI (SEQ ID NO:53), GLPHYITGDL (SEQ ID NO:54), GLPHYLTGDI (SEQ ID NO:55), GLPHYITGEI (SEQ ID NO:56), GLPNYITGDI (SEQ ID NO:57), GLPHFITGEI (SEQ ID NO:58), and the like.
  • These and other conservative amino acid substitutions are contemplated, as well as combinations thereof. In general such substitutions include no more than about 5% changes from the preferred sequences, preferably no more than about 10% changes and most preferably, no more than about 15% changes in the preferred sequences. Such sequences are 95%, 90% and 85% homologous, respectively, with the preferred CDR sequences.
  • In addition, each CDR sequence can be substituted independently, such that one CDR sequence can remain unsubstituted while one or more of the other CDR sequences can have one or more amino acid substitutions, as desired. One skilled in the art can easily envision acceptable substitutions and using recombinant methodologies, prepare and test the resulting antibody variants for binding to THC. In this manner, variants and improvements on the preferred CDR sequences disclosed above can be prepared, enabling the practice of the full scope of the invention.
  • In an additional embodiment, there is provided an antibody that specifically binds to THC and has CDR1 in the VL region (VL/CDR1) comprising the amino acid sequence of Q X1 X2 Q X3 X4 X5 N N N Q X6 X7 (SEQ ID NO:59) wherein X1 can be G, A or V; X2 is S or T; X3 is S or T; X4 is M, L, I, or V; X5 is Y, or F; X6 is L, I; X7 is S or T.
  • In an additional embodiment, there is provided an antibody that specifically binds to THC and has CDR2 in the VL region (VL/CDR2) comprising the amino acid sequence of G X1 X2 X3 X4 X5 X6 (SEQ ID NO:60), wherein X1, X2, X3 and X6 can be T, Y, S, Q or N; and X4 and X5 can be G, A, V, L, I, M, P, F, W or C.
  • In an additional embodiment, there is provided an antibody that specifically binds to THC and has CDR3 in the VL region (VL/CDR3) comprising the amino acid sequence of Q X1 X2 X3 X4 X5 X6 X7 X8 X9 X10, (SEQ ID NO:61), wherein X1, X2, X6 and X8, X9 and X10 can be G, A, V, L, I, M, P, F, W or C; X3, X4 and X5 can be T, Y, S, Q or N; X7 can be D or E.
  • In an additional embodiment, there is provided an antibody that specifically binds to THC and has CDR1 in the VH region (VH/CDR1) comprising the amino acid sequence of X1 X2 X3 X4 X5 X6 X7 X8 X9 X10, (SEQ ID NO:62), wherein X1, X4, X8, X9 and X10 can be G, A, V, L, M or I; X2 and X7 can be F, W or Y; X3, and X5 can be T, Y, or S; and X6 can be N or Q.
  • In an additional embodiment, there is provided an antibody that specifically binds to THC and has CDR2 in the VH region (VH/CDR2) comprising the amino acid sequence of X1 X2 X3 X4 X5 X6 X7 X8 X9 X10 X11 X12, (SEQ ID NO:63), wherein X1, X4, X6, X7 and X11 can be T, S or Y; X2, X3 or X10, can be G, A, V, L, M or I; X5 can be G, V, A, L or I, X8 and X9 can be F or Y, and X12 can be W, Y or F.
  • In an additional embodiment, there is provided an antibody that specifically binds to THC and has CDR3 in the VH region (VH/CDR3) comprising the amino acid sequence of X1 X2 P X3 X4 X5 X6 X7 X8 X9, (SEQ ID NO:64), wherein X1, X2, X5 X7 and X9 can be G, L, M, I, V, or A, X6 can be T, S or Y, X8 can be D or E, X3 can be H, K or R, X4 can be Y or F.
  • The anti-THC antibodies may be synthesized or expressed in cells of any organism, including but not limited to bacteria, yeast, plant, insect, and mammal. Particular types of cells include, but are not limited to, Drosophila melanogaster cells, Saccharomyces cerevisiae and other yeasts, E. coli, Bacillus subtilis, SF9 cells, C129 cells, 293 cells, Neurospora, BHK, CHO, COS, and HeLa cells, fibroblasts, Schwanoma cell lines, immortalized mammalian myeloid and lymphoid cell lines, Jurkat cells, mast cells and other endocrine and exocrine cells, and neuronal cells. Examples of mammalian cells include, but are not limited to, tumor cells of all types (particularly melanoma, myeloid leukemia, carcinomas of the lung, breast, ovaries, colon, kidney, prostate, pancreas and testes), cardiomyocytes, endothelial cells, epithelial cells, lymphocytes (T-cell and B cell), mast cells, eosinophils, vascular intimal cells, hepatocytes, leukocytes including mononuclear leukocytes, stem cells such as haemopoetic, neural, skin, lung, kidney, liver and myocyte stem cells, osteoclasts, chondrocytes and other connective tissue cells, keratinocytes, melanocytes, liver cells, kidney cells, and adipocytes.
  • The anti-THC antibodies can be purified or isolated after expression according to methods known to those skilled in the art. Examples of purification methods include electrophoretic, molecular, immunological and chromatographic techniques, including ion exchange, hydrophobic, affinity, and reverse-phase HPLC chromatography, and chromatofocusing, and the like. The degree of purification necessary will vary depending on the contaminants present with the antibodies. In some instances no purification will be necessary.
  • Also according to any of the embodiments described above, the anti-THC antibodies can be screened for a desired function, preferably a biological function such as their binding to THC or particular metabolites or analogs thereof. Screening assays are well known to those skilled in the art of immunoassays and immunochemistry, and include without limitation, ELISAs, flow cytometry, immunodiffusion precipitation assays, and the like.
  • The anti-THC antibodies can be used as affinity purification agents. In this process, the antibodies are immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art. The immobilized antibody is contacted with a sample containing THC or metabolites or analogs thereof, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the THC, which is bound to the immobilized antibody. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the THC from the antibody.
  • Anti-THC antibodies can also be useful in diagnostic assays for THC or its metabolites, e.g., detecting its presence in specific cells, tissues, or body fluids, such as saliva, serum or urine. Such diagnostic methods are useful in detecting and managing THC abuse, or dosing, such as when performing pharmacokinetic studies.
  • For diagnostic applications, the THC binding antibody can be labeled with a detectable moiety. For example, the antibody can be labeled with a radioisotope, such as 35S, 14C, 125I, 3H, and 131I, and the like, using techniques such as those described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al., Ed., Wiley-Interscience, New York, N.Y., Pubs. (1991). for example. Radioactive labeling can be measured using scintillation counting, for example.
  • The THC binding antibody can also be conjugated with a fluorescent label such as, without limitation, rare earth chelates (e.g., europium chelates), fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red. The fluorescent labels can be conjugated to the antibody using techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter or UV/vis spectrophotometer using the known extinction coefficient of the fluorescent label.
  • The THC binding antibody can be labeled with various enzyme-substrate labels such as those disclosed in U.S. Pat. No. 4,275,149. The enzyme generally catalyzes a chemical alteration of a chromogenic substrate which can be measured using various techniques, typically optical absorption. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. The labeled anti-THC antibodies can be used for immunohistochemistry as well.
  • The THC binding antibody can be labeled with various particles such as that are not limited to any particular materials, and can comprise metals such as colloidal gold, natural polymers such as latex (e.g., latex particle, latex microparticle), polymer microspheres or microbeads, quantum dots, magnetic particles, glass beads, and the like. Preferably, the particles are detectable and can be easily visualized by eye or using an instrument, for example, by being colored.
  • Affinity maturation experiments can be performed in silico or in vitro, to optimize the CDR amino acid sequences to increase the binding affinity for THC or a specific metabolite or analog of THC. Affinity maturation techniques are known in the art and include the methods taught, for example, in U.S. Patent Application Publication Nos. 2004/0133357 and 2003/0022240. In addition to, or as an alternative to affinity maturation of the CDR sequences, the framework regions of the THC binding antibodies described herein can be subjected to mutagenesis and/or amino acid substitution, making conservative amino acid substitutions, or nonconservative amino acid substitutions, according to theoretical predictions based on protein structural databases, protein modeling and force field calculations, and the like. For example, energy scoring functions utilizing electrostatic interactions, van der Waals interactions, electrostatic solvation energy, solvent-accessible surface solvation energy, and conformational entropy can be incorporated into the affinity maturation methods. Force fields such as the Amber force field, Charmm force fields, the Discover cvff force fields, the ECEPP force fields, the GROMOS force fields, the OPLS force fields, the MMFF94 force field, the Tripos force field, the MM3 force field, the Dreiding force field, and UNRES force field, and other knowledge-based statistical force field (mean field) and structure-based thermodynamic potential functions can also be helpful in providing guidance for amino acid substitutions to optimize or provide selectivity between the parent Δ9-THC molecule and THC metabolites or analogs, etc.
  • III. Analytes
  • Analytes to be measured using the present immunoassays include Δ9-THC and its metabolites, analogs, precursors and derivatives, and the like, without limitation. Preferably, the anti-THC antibodies bind Δ9-THC with highest affinity and thereby provide a more sensitive assay for Δ9-THC, which can be present in unmetabolized form in body fluid (e.g, serum, saliva), in contrast to other THC assays which measure THC metabolites in urine or serum. In another preferred embodiment, the anti-THC antibodies bind Δ9-THC-COOH. Preferably, the anti-THC antibodies also bind to protein-conjugated THC or protein conjugated THC metabolites with high affinity.
  • IV. Obtaining Saliva Samples for Testing
  • There are several problems associated with obtaining samples for drug testing from subjects, including privacy issues associated with sample collection, maintenance of sample chain of custody, prevention of sample adulteration by the subject, and facilitating more rapid turn around time on sample analyses. Drug testing protocols utilizing urine samples present invasion of privacy issues. For example, with urine samples, it is necessary to observe the individual providing the sample to maintain the chain of custody and eliminate the possibility of sample switching or adulteration. Urine samples are also not a good indicator of the current level of intoxication since many drug metabolites continue to be excreted into urine for days or weeks after the drugs are initially taken. While blood samples do not suffer from these problems, collecting blood is an invasive procedure requiring special facilities and trained personnel that may not always be available when the need arises. It is necessary for law enforcement personnel to maintain strict chain of custody for all samples collected to ensure that mishandling or deliberate tampering do not occur. A break or even a perceived break in the chain of custody can result in evidence being dismissed outright or given little weight.
  • The assays utilizing the anti-THC antibodies described herein solve these problems in several ways. First, integration of the antibody into the drug test makes identification of the sample donor integral to the test and eliminates the need for complex chain of custody procedures. Second, a saliva-based drug test is better than a urine test for determining recent THC use because the non-metabolized drug, Δ9-THC, is the main form of THC present in the marijuana smoker's oral fluid sample. Thus the presence of Δ9-THC in oral fluids is an excellent indicator of recent marijuana consumption and may indicate that the person is under the drug's influence. In addition, drug levels in saliva can be readily correlated with drug levels in blood, therefore providing a better indicator of current drug use. Saliva samples from a subject can also be collected easily in view of a law enforcement officer without invasion of privacy and without using invasive methods. Finally, the present tests are easy to use and can be quickly performed by law enforcement or other personnel on site.
  • Accordingly, saliva samples can be conveniently collected and transferred to the testing device or assay using one of the embodiments of the devices disclosed for fluid collection and testing in U.S. Pat. No. 6,468,474 to Bachand, which discloses a saliva testing and confirmation device that generally includes an expresser cup adapted to receive and compress a foam collection swab pressed into the cup. The swab includes a sponge or foam portion capable of absorbing a fluid specimen such as saliva, and a generally rigid handle for facilitating manipulation thereof. The device also includes a testing and confirmation platform integral with the expresser cup. A channel or groove in the cup is provided for directing a flow of expressed fluid from the expresser into both a test well and a confirmation well. A lateral flow reagent test strip, in fluid communication with the test well may be encased in the platform and partially revealed for analysis of test results. The confirmation well includes a fluid tight cap having a tamper evident seal.
  • Additional fluid collection devices are disclosed in U.S. Ser. No. 10/942,493, published as U.S. Patent Application Publication No. 2006/0057027 by Hudak, et al. Briefly, the fluid collection and testing devices include a collection vial having a bottom and an open top along with an expresser sized for insertion into the vial open end. The expresser includes a ribbed base enabling fluid to pass therethrough and a support member holds the expresser within the collection vial with the ribbed base in a spaced apart relationship with the vial bottom. A fluid collector includes an absorbent member capturing a fluid and the absorbent member is sized for insertion into the expresser and is compressible against the ribbed base for releasing captured fluid through the ribbed base and into the vial bottom. A catch mechanism is provided for latching the fluid collector to the expresser after insertion into the expresser for enabling simultaneous removal of the expresser and the fluid collector from the collection vial.
  • V. Assays
  • The THC binding antibodies described herein can be used in any diagnostic or analytic assay to determine the presence of THC or its analogs, metabolites, etc. There are many types of immunoassays and immunochemical applications known in the art, including, but not limited to, ELISAs, western blotting, immunohistochemical assays, radioimmunoassays, fluorescence immunoassays, immunocytochemical assays, immunoaffinity chromatography, immunoprecipitation, flow cytometry, and the like. Any of these assay format and detection schemes can be performed using the anti-THC antibodies described herein. Preferred assays using the anti-THC antibodies of the invention include, without limitation, ELISAs, histoimmunochemical assays, lateral flow immunoassays, continuous flow displacement assays, and the like, as discussed further below. The assays can be used in competitive or noncompetitive manners and with any number of detection aids such as conjugating the anti-THC antibodies or THC or THC analogs to labels, enzymes, or particles, or the like, as will be readily understood by one skilled in the art.
  • A. General Considerations of Immunoassays
  • The most common type of immunoassay is competitive and non-competitive heterogeneous assays, such as enzyme-linked immunosorbent assays (ELISA). In a non-competitive ELISA, unlabeled antigen is bound to a solid support or reaction vessel, such as the surface of a microtiter plate or biochip. Biological sample is combined with antigens bound to the reaction vessel, and antibodies (primary antibodies) in the biological sample are allowed to bind to the antigens, thus forming the immune complexes. After the immune complexes have formed, excess biological sample is removed and the vessel is washed to remove nonspecifically bound antibodies. The immune complexes are then reacted with an appropriate enzyme-labeled anti-immunoglobulin (secondary antibody). The secondary antibody reacts with antibodies in the immune complexes, not with other antigens bound to the vessel. Secondary antibodies specific for binding antibodies of different species, including humans, are well known in the art and are commercially available, such as from Sigma Chemical Co. (St. Louis, Mo.) and Santa Cruz Biotechnology (Santa Cruz, Calif.). After a second wash step, the enzyme substrate is added. The enzyme linked to the secondary antibody catalyzes a reaction that converts the substrate into a detectable product. When excess antigen is present, the amount of product is directly proportional to the amount of primary antibodies present in the biological sample. Preferably, the product is fluorescent or luminescent, which can be measured using technology and equipment well known in the art. It is also possible to use reaction schemes that result in a colored product, which can be measured spectrophotometrically.
  • Sandwich or capture assays can also be used to identify and quantify immune complexes. Sandwich assays are a mirror image of non-competitive ELISAs in that antibodies are bound to the solid phase and antigen in the biological sample is measured. These assays are particularly useful in detecting antigens, having multiple epitopes, that are present at low concentrations. This technique requires excess antibody to be attached to a solid phase, such as the biochip. The bound antibody is then incubated with the biological samples, and the antigens in the sample are allowed to form immune complexes with the bound antibody. The immune complex is incubated with an enzyme-linked secondary antibody, which recognizes the same or a different epitope on the antigen as the primary antibody. Hence, enzyme activity is directly proportional to the amount of antigen in the biological sample. D. M. Kemeny & S. J. Challacombe, ELISA and Other Solid Phase Immunoassays (1988).
  • Typical enzymes that can be linked to secondary antibodies include horseradish peroxidase, glucose oxidase, glucose-6-phosphate dehydrogenase, alkaline phosphatase, .beta.-galactosidase, and urease. Secondary antigen-specific antibodies linked to various enzymes are commercially available from, for example, Sigma Chemical Co. and Amersham Life Sciences (Arlington Heights, Ill.).
  • Competitive ELISAs are similar to noncompetitive ELISAs except that enzyme linked antibodies compete with unlabeled antibodies in the biological sample for limited antigen binding sites. Briefly, a limited number of antigens are bound to the solid support. Biological sample and enzyme-labeled antibodies are added to the solid support. Antigen-specific antibodies in the biological sample compete with enzyme-labeled antibodies for the limited number of antigens bound to the solid support. After immune complexes have formed, nonspecifically bound antibodies are removed by washing, enzyme substrate is added, and the enzyme activity is measured. No secondary antibody is required. Because the assay is competitive, enzyme activity is inversely proportional to the amount of antibodies in the biological sample.
  • An alternative competitive ELISA can also be used. In this alternative embodiment, limited amounts of antibodies from the biological sample are bound to the surface of the solid support as described herein. Labeled and unlabeled antigens are then brought into contact with the solids support such that the labeled and unlabeled antigens compete with each other for binding to the antibodies on the surface of the solid support. After immune complexes have formed, nonspecifically bound antigens are removed by washing. The immune complexes are detected by incubation with an enzyme-linked secondary antibody, which recognizes the same or a different epitope on the antigen as the primary antibody, as described above. The activity of the enzyme is then assayed, which yields a signal that is inversely proportional to the amount of antigen present.
  • Homogeneous immunoassays can also be used when utilizing the anti-THC antibodies of the present invention. Homogeneous immunoassays may be preferred for detection of low molecular weight compounds, such as hormones, therapeutic drugs, and illegal drugs that cannot be analyzed by other methods, or compounds found in high concentration.
  • In homogeneous techniques, bound or unbound antigens are enzyme-linked. When antibodies in the biological sample bind to the enzyme-linked antigen, steric hindrances inhibit the enzyme activity. Free antigens (i.e., not enzyme-linked) compete with the enzyme-linked antigen for limited antibody binding sites. Thus, enzyme activity is directly proportional to the concentration of antigen in the biological sample. Enzymes useful in homogeneous immunoassays include lysozyme, neuraminidase, trypsin, papain, bromelain, glucose-6-phosphate dehydrogenase, and .beta.-galactosidase. T. Persoon, Immunochemical Assays in the Clinical Laboratory, 5 Clinical Laboratory Science 31 (1992). Enzyme-linked antigens are commercially available or can be linked using various chemicals well known in the art, including glutaraldehyde and maleimide derivatives.
  • Fluorescent immunoassays can also be used when utilizing the Anti-THC antibodies of the present invention. Fluorescent immunoassays are similar to ELISAs except the enzyme is substituted for fluorescent compounds called fluorophores or fluorochromes. These compounds have the ability to absorb energy from incident light and emit the energy as light of a longer wavelength and lower energy. Fluorescein and rhodamine, usually in the form of isothiocyanates that can be readily coupled to antigens and antibodies, are most commonly used in the art. D. P. Stites et al., Basic and Clinical Immunology (1994). Fluorescein absorbs light of 490 to 495 nm in wavelength and emits light at 520 nm in wavelength. Tetramethylrhodamine absorbs light of 550 nm in wavelength and emits light of 580 nm in wavelength. Illustrative fluorescence-based detection methods include ELF-97 alkaline phosphatase substrate (Molecular Probes Inc., Eugene, Oreg.); PBXL1 and PBXL-3 (phycobilisomes conjugated to streptavidin) (Martek Biosciences Corp., Columbia, Md.); FITC and Texas Red labeled goat anti-human IgG (Jackson ImmunoResearch Laboratories, Inc., West Grove, Pa.); and B-Phycoerythrin and R-Phycoerythrin conjugated to streptavidin (Molecular Probes Inc.). ELF-97 is a nonfluorescent chemical that is digested by alkaline phosphatase to form a fluorescent molecule. Because of turn over of the alkaline phosphatase, use of the ELF-97 substrate results in signal amplification. Fluorescent molecules attached to secondary antibodies do not exhibit this amplification.
  • Phycobiliproteins isolated from algae, porphyrins, and chlorophylls, which all fluoresce at about 600 nm, are also being used in the art. I. Hemmila, Fluoroimmunoassays and Immunofluorometric Assays, 31 Clin. Chem. 359 (1985); U.S. Pat. No. 4,542,104. Phycobiliproteins and derivatives thereof are commercially available under the names R-phycoerythrin (PE) and Quantum Red™ from, for example, Sigma Chemical Co. In addition, Cy-conjugated secondary antibodies and antigens are useful in immunoassays and are commercially available. Cy-3, for example, is maximally excited at 554 nm and emits light of between 568 and 574 nm. Cy-3 is more hydrophilic than other fluorophores and thus has less of a tendency to bind nonspecifically or aggregate. Cy-conjugated compounds are commercially available from Amersham Life Sciences.
  • Illustrative luminescence-based detection methods include CSPD and CDP star alkaline phosphatase substrates (Roche Molecular Biochemicals); and SuperSignal®. horseradish peroxidase substrate (Pierce Chemical Co., Rockford, Ill.). Chemiluminescence, electroluminescence, and electrochemiluminescenc-e (ECL) detection methods are also attractive means for quantifying antigens and antibodies in a biological sample. Luminescent compounds have the ability to absorb energy, which is released in the form of visible light upon excitation. In chemiluminescence, the excitation source is a chemical reaction; in electroluminescence the excitation source is an electric field; and in ECL an electric field induces a luminescent chemical reaction. Molecules used with ECL detection methods generally comprise an organic ligand and a transition metal. The organic ligand forms a chelate with one or more transition metal atoms forming an organometallic complex. Various organometallic and transition metal-organic ligand complexes have been used as ECL labels for detecting and quantifying analytes in biological samples. Due to their thermal, chemical, and photochemical stability, their intense emissions and long emission lifetimes, ruthenium, osmium, rhenium, iridium, and rhodium transition metals are favored in the art. The types of organic ligands are numerous and include anthracene and polypyridyl molecules and heterocyclic organic compounds. For example, bipyridyl, bipyrazyl, terpyridyl, and phenanthrolyl, and derivatives thereof, are common organic ligands in the art. A common organometallic complex used in the art includes tris-bipyridine ruthenium (II), commercially available from IGEN, Inc. (Rockville, Md.) and Sigma Chemical Co.
  • Advantageously, ECL can be performed under aqueous conditions and under physiological pH, thus minimizing biological sample handling. J. K. Leland et al., Electrogenerated Chemiluminescence: An Oxidative-Reduction Type ECL Reactions Sequence Using Triprophyl Amine, 137 J. Electrochemical Soc. 3127-3131 (1990); WO 90/05296; U.S. Pat. No. 5,541,113. Moreover, the luminescence of these compounds may be enhanced by the addition of various cofactors, such as amines.
  • In practice, a tris-bipyridine ruthenium (II) complex, for example, may be attached to a secondary antibody using strategies well known in the art, including attachment to lysine amino groups, cysteine suithydryl groups, and histidine imidazole groups. In a typical ELISA immunoassay, secondary antibodies would recognize ISAs bound to antigens, but not unbound antigens. After washing nonspecific binding complexes, the tris-bipyridine ruthenium (II) complex would be excited by chemical, photochemical, and electrochemical excitation means, such as by applying current to the biochip. E.g., WO 86/02734. The excitation would result in a double oxidation reaction of the tris-bipyridine ruthenium (II) complex, resulting in luminescence that could be detected by, for example, a photomultiplier tube. Instruments for detecting luminescence are well known in the art and are commercially available, for example, from IGEN, Inc. Solid state color detection circuitry can also be used to monitor the color reactions on the biochip and, on command, compare the color patterns before and after the sample application. A color camera image can also be used and the pixel information analyzed to obtain the same information.
  • Still another detection method involves detection using a surface plasmon resonance (SPR) chip. The surface of the chip is scanned before and after sample application and a comparison is made. The SPR chip relies on the refraction of light when the molecules of interest are exposed to a light source. Each molecule has its own refraction index by which it can be identified. This method requires precise positioning and control circuitry to scan the chip accurately.
  • Yet another method involves a fluid rinse of the biochip with a fluorescing reagent. The locations that combine with the biological sample will fluoresce and can be detected with a charge-coupled device (CCD) array. The output of such a CCD array is analyzed to determine the unique pattern associated with each sample. This approach avoids the problems associated with scanning technologies. Speed is not a factor with any of the methods since the chemical combining of sample and reference takes only a few minutes to occur.
  • Moreover, array scanners are commercially available, such as from Genetic MicroSystems. The GMS 418 Array Scanner uses laser optics to rapidly move a focused beam of light over the biochip. This system uses a dual-wavelength system including high-powered, solid-state lasers that generate high excitation energy to allow for reduced excitation time. At a scanning speed of 30 Hz, the GMS 418 can scan a 22.times.75-mm slide with 10-.mu.m resolution in about 4 minutes.
  • Software for image analysis obtained with an array scanner is readily available. Available software packages include ImaGene (BioDiscovery, Los Angeles, Calif.); ScanAlyze (available at no charge; developed by Mike Eisen, Stanford University); DeArray (developed by Yidong Chen and Jeff Trent of the National Institutes of Health; used with IP Lab from Scanalytics, Fairfax, Va.); Pathways (Research Genetics, Huntsville, Ala.); GEM tools (Incyte Pharmaceuticals, Inc., Palo Alto, Calif.); and Imaging Research (Amersham Pharmacia Biotech, Inc., Piscataway, N.J.).
  • B. Lateral Flow Immunoassays
  • In a preferred embodiment, the THC binding antibody is utilized in a lateral flow immunoassay. Lateral Flow immunoassays are described, for example, in U.S. Pat. Nos. 5,770,458 to Klimov, 6,001,658 to Fredrickson and 6,368,876 to Huang. Lateral flow immunoassay is based on the principle of competitive binding. Analytes (e.g., drugs, THC) that may be present in the aqueous sample to be tested (e.g., an oral fluid specimen) compete against their respective conjugate for binding sites on an antibody having specific binding activity for the drug that is conjugated to a visible (e.g., colored) particle. The particles that can be used are not limited to any particular materials, and can comprise metals such as colloidal gold, natural polymers such as latex (e.g., latex particle, latex microparticle), polymer microspheres or microbeads, quantum dots, magnetic particles, glass beads, and the like. During testing, a portion of the oral fluid specimen migrates upward by capillary action. A drug, if present in the oral fluid specimen below its cut-off concentration, will not saturate the binding sites of an antibody having specific binding activity for the drug. The antibody without bound antigen will then react with the drug-protein conjugate, and a visible colored line will show up in the test line region of the specific drug strip due to the colored particle conjugated to the antibody. However, in the presence of drug above the cut-off concentration in the oral fluid specimen, the binding sites of the antibody will be saturated and the antibody will not complex with the drug conjugate and form a visible line in the test line region. A drug-positive oral fluid specimen will not generate a colored line in the specific test line region of the strip because of drug competition, while a drug-negative oral fluid specimen will generate a line in the test line region because of the absence of drug competition. To serve as a procedural control, a colored line will always appear at the control line region, indicating that proper volume of specimen has been added and membrane wicking has occurred.
  • The lateral immunoassay test typically contains membrane strips coated with drug-protein conjugates (purified bovine albumin) on the test line, an anti-antibody (e.g., a goat polyclonal anti-rabbit or anti-mouse antibody) or anti-BSA antibody against gold-protein conjugate at the control line, and a dye pad which contains particles, such as colloidal gold particles, coated with a monoclonal antibody specific to the drug to be tested (e.g., Δ9-THC or its metabolites).
  • In a particularly preferred embodiment, the anti-THC antibody is a recombinant rabbit antibody comprising CDR amino acid sequences selected from L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6) and homologous amino acid sequences having at least 85% homology thereto. A lateral flow immunoassay utilizing this antibody provides a much greater sensitivity and specificity for Δ9-THC than provided by murine anti-THC antibodies tested, as described in Example 4, and is particularly suitable and useful for oral fluid testing for the presence of Δ9-THC.
  • In an additional preferred embodiment, the anti-THC antibody comprises the amino acid sequence for the light chain variable region:
  • QVLTQTPSPVSAAVGGTVTINCQASQSVYNNNQLSWYQQKPGQPPKLLIYGTSNLASG VPSRFKGSGSGTQFTLTISSVQCDDAATYYCQGGYTSGDGIAFGGGTEVVVK (SEQ ID NO:7), or homologous amino acid sequences having at least 85% homology thereto. In certain embodiments, the amino acid sequence has about 90% or 95%, or even greater homology thereto. In additional preferred embodiments, the anti-THC antibody comprises the amino acid sequence for the heavy chain variable region:
    QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGKGLEWIGTIVSGTTYYAS WAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYITGDIWGPGTLVTVSLG (SEQ ID NO:8), or homologous amino acid sequences having at least 85% homology thereto. In certain embodiments, the amino acid sequence has about 90% or 95%, or even greater homology thereto. These sequences depict both the framework region sequences and the CDR sequences (indicated with underlining).
  • C. Continuous Flow Displacement Cannabinoid Immunoassay
  • The anti-THC antibodies described herein can also be used in a continuous flow displacement immunoassay for Δ9-THC or its metabolites. In a continuous flow displacement immunoassay for cannabinoids, the binding kinetics of the cannabinoid analyte (e.g., Δ9-THC or its metabolites) to the antibody play a very important role. To allow for determination of the presence of the cannabinoid analyte, a labeled tracer compound is allowed to compete with the cannabinoid analyte for binding to the antibody. Preferably, the binding kinetics of the antibody to the tracer compound provide a fast dissociation rate of the bound tracer from the antibody, thereby permitting a rapid binding of the cannabinoid analyte. In general, a continuous flow displacement cannabinoid immunoassay utilizes an antibody having a high affinity for the cannabinoid analyte and a lower affinity for the tracer. The affinity of the antibody for the tracer may be anywhere between 15-100% cross-reactivity. A preferred cannabinoid-based tracer has about 40-80% cross-reactivity for the antibody.
  • A typical continuous flow displacement cannabinoid immunoassay involves a solid-phase immobilized antibody to Δ9-THC or its metabolites. The antigen binding site of the antibody can be exposed to a synthetic labeled tracer to form a labeled synthetic tracer-antibody complex such that the antigen binding sites of the antibody are saturated with the labeled synthetic tracers. When a biological sample suspected of containing the analyte Δ9-THC is continuously flowed past the solid-phase immobilized antibody-labeled synthetic tracer complex, if Δ9-THC is present in the sample, the cannabinoid analyte binds to the antibody and displaces the labeled synthetic tracer. Detection of the labeled tracer downstream from the binding point indicates the presence and/or quantity of the analyte present in the biological sample. See U.S. Pat. No. 5,183,740 to Ligler, which is incorporated by reference herein.
  • The success of developing a continuous flow displacement immunoassay is based on the selection of antibody and tracer to achieve a fast dissociation rate of the bound tracer from the antibody, thereby permitting a rapid binding of the analyte. In general, the ideal continuous flow displacement immunoassay utilizes a system where the antibody has a high affinity for the analyte, and a lower affinity for the tracer. The affinity of the antibody for the tracer may be anywhere between 15-100% cross-reactivity. A preferred THC-based tracer has about 40-80% cross-reactivity for the antibody. In addition, the condition for displacing a tracer from the recognition molecule is preferably carried out in non-equilibrium condition or kinetic flow reaction, such that the sample flows past the recognition molecule-tracer complex at a rate where a stable equilibrium state between the recognition molecule, tracer, and analyte has not been achieved.
  • D. Additional Assay Formats
  • The anti-THC antibodies described herein can also be utilized in additional useful commercially marketed immunoassay formats, for example, the OnTrak TesTcup, On•Site CupKit™, On•Site OraLab®, OnTrak TesTstik assay systems marketed by Varian, Inc.
  • VI. Pharmaceutical Applications and Formulations
  • The anti-THC antibodies described herein can also be utilized in pharmaceutical and therapeutic applications, for example, to treat overdose of THC or reduce serum concentrations of THC. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).
  • The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, it may be desirable to further provide an antipsychotic agent. Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • The active ingredients can also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes. Therapeutic formulations of the antibodies can be prepared for storage by mixing the antibodies having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Sustained-release preparations comprising the antibodies can be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibodies, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-1S methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(−)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • For use in humans, humanization of the rabbit anti-THC antibody is required, and can be accomplished using methods such as those described in U.S. Pat. Nos. 6,800,738 to Carter, and 6,719,971. CDR repair can be effected if binding affinity and/or specificity is compromised utilizing, for example, the methods described in U.S. Patent Application Publication No. 20060122377, all of which are incorporated by reference herein in their entireties.
  • The antibodies can be administered to the patient by a variety of different means and will vary depending upon the intended application. As one skilled in the art would recognize, administration of the therapeutic compositions can be carried out in various fashions, and more typically by parenteral injection into body cavity or vessel, e.g., intraperitoneal, intravenous, intralymphatic, intratumoral, intramuscular, interstitial, intraarterial, subcutaneous, intralesional, intraocular, intrasynovial, intraarticular. However, other methods of administration can be utilized for particular purposes, for example, via topical administration, including, but not limited to, dermal, ocular and rectal; transdermal, via passive or active means, e.g., using a patch, a carrier, or iontophoresis; transmucosal, e.g., sublingual, buccal, rectal, vaginal, or transurethral; oral, e.g., gastric or duodenal; via inhalation, e.g., pulmonary or nasal inhalation, using e.g., a nebulizer.
  • Depending on the THC concentration in serum or other body fluids, about 1 μg/kg to about 50 mg/kg (e.g., 0.1-20 mg/kg, 0.5-15 mg/Kg, and 1-10 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily or weekly dosage might range from about 1 μg/kg to about 20 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until the THC levels have been reduced to a desired level. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays, including, for example, immunoassays of THC and/or its metabolites in urine or saliva.
  • It is to be understood that while the invention has been described in conjunction with the preferred specific embodiments thereof, that the description above as well as the examples that follow are intended to illustrate and not limit the scope of the invention. The practice of the present invention will employ, unless otherwise indicated, conventional techniques of immunochemistry, organic chemistry, polymer chemistry, biochemistry and the like, which are within the skill of the art. Other aspects, advantages and modifications within the scope of the invention will be apparent to those skilled in the art to which the invention pertains. Such techniques are explained fully in the literature.
  • In the following examples, efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental error and deviation should be accounted for. Unless indicated otherwise, temperature is in degrees ° C. and pressure is at or near atmospheric.
  • Example 1 Preparation of a Rabbit Monoclonal Antibody Having Specific Binding for THC
  • An antibody having specific binding for THC was prepared by the following procedures:
  • Immunization
  • New Zealand White rabbits were immunized. Rabbits were injected subcutaneously with 0.4 mg of THC-KLH in complete Freund's adjuvant. After the initial immunization, animals were boosted 5 more times every 21 days in the same manner with incomplete Freund's adjuvant and the sera tested by immunoassay and immunohistochemical staining. The rabbit with the best titer in the immunoassay and IHC (immunohistochemistry) was selected for a final intravenous boost of 0.4 mg of THC-KLH in saline, four days before removal of the spleen.
  • Hybridization, Fusion
  • Fusions were performed using conventional methodology: 1.5-3×108 lymphocytes from an immunized rabbit and the fusion partner (240E-w) were fused at a ratio of 2:1 with PEG 4000 at 37° C. in serum-free medium. The cells were distributed in 96-well cell culture plates at approximately 1×105 lymphocytes per well in medium with 15% FBS (or FCS), and after 48 hr HAT medium was added. Medium was changed 2-3 times before screening. Hybridoma colonies were ready for screening in 3-5 weeks. Supernatants were tested for the presence of antibody, specific for the immunogen, by ELISA. Immunohistochemistry was used as a secondary screening assay. The hybridomas were sub-cloned by limit dilution. For feeder cells, the fusion partner 240E-w at 2×104 cells per well was used.
  • ELISA
  • ELISA was performed in 96-well micro-titer plates that had been coated overnight with immunogen at 1.0 μg/ml, plates were then saturated with 2% BSA, followed by incubation with the antibody-containing supernatant for 1 hr at room temperature. After washing with PBS-Tween, alkaline phosphatase conjugated goat anti-rabbit immunoglobulin (IgG) was added and incubation continued for another hour; plates were washed again and developed in the presence of P-NPP (para-nitrophenyl phosphate). Color was read at 405 nm with a plate reader (Multiskan MCC/340 from Fisher Scientific).
  • Antibody Purification
  • Tissue culture supernatants containing rabbit monoclonal antibody against ER were incubated with Protein A immobilized on a column. The column was washed extensively with PBS to remove nonspecific binding proteins until the OD 280 was less than 0.01. The rabbit IgGs were eluted with 0.2 M, pH 2.5 citrate. The eluates was dialyzed against PBS overnight at 4° C. (28).
  • Antibody Affinity Assay
  • ELISA was performed to determine antibody affinities. 96-well micro-titer plates was coated overnight with the THC conjugated BSA or a human recombinant protein (1-300aa) at 1.0 ∝g/ml, respectively. The plates were saturated with 2% BSA, followed by incubation with the antibody serially diluted (200 nM, 100 nM, 50 nM, 25 nM, 12.5 nM, 6.25 nM, 3.125 nM, 1.56 nM, 0.78 nM, 0.39 nM, and 0.2 nM). ER clone SP1 was incubated with the wells coated with the BSA conjugated ER peptide and clone 1D5 was incubated with wells coated with ER alpha recombinant protein. Samples were in duplicate. After washing with PBS-Tween, HRP-conjugated goat anti-rabbit and mouse IgGs (Spring Bioscience Corporation, CA) were added. The plates were washed again, and were developed with TMB solution for 15 minutes. The reaction was stopped by adding 0.5N H2SO4. Optical density (OD) was measured at 450 nm.
  • Example 2 cDNA Cloning from Rabbit Hybridoma and Recombinant Antibody Expression
  • mRNA was isolated from rabbit hybridomas by using Qiagen TurboCapture mRNA kit. cDNA was made directly in the TurboCapture tube by solid phase synthesis. Rabbit IgG cDNAs were amplified with heavy or light chain specific primers by PCR. For L chain, the PCR product was sequenced and a consensus sequence was found from three independent mRNA and PCR reactions. To make an expression construct, the PCR product was digested with restriction enzymes and cloned into an expression vector. A cDNA clone with the consensus PCR sequence was selected for expression. For H chain, only the variable region (VH) was amplified and the PCR product was digested with the fusion partner VH specific restriction enzyme. Rabbit B-cell VH cDNA was purified and recovered from an agarose gel. In order to identify a desired cDNA clone, the purified VH product was cloned into pCR 2.1-TOPO vector (Invitrogen) and cDNA clones were screened. Then, the VH cDNA in the desired clone was subcloned into an expression vector with rabbit IgG heavy chain constant region (CH). To express recombinant antibody, both light and heavy chain expression constructs were transfected into HEK 293 by using 293 fectin (Invitrogen) as a vehicle. Supernatant containing recombinant antibody transiently expressed from the cells were harvested after 5-day transfection.
  • The nucleotide and amino acid sequences for the THC binding antibody variable regions of the light chain (VL) and the variable region of the heavy chain (VH) are depicted below:
  • VL Nucleotide Sequence:
  • (SEQ ID NO: 65)
    CAA GTG CTG ACC CAG ACT CCA TCC CCC GTG TCT GCA
    GCT GTG GGA GGC ACA GTC ACC ATC AAT TGC CAG GCC
    AGT CAA AGT GTT TAT AAT AAC AAC CAA TTA TCC TGG
    TAT CAG CAG AAA CCA GGG CAG CCT CCC AAG CTC CTG
    ATC TAT GGT ACA TCC AAT CTG GCA TCT GGG GTC CCA
    TCG CGG TTC AAA GGC AGT GGA TCT GGG ACA CAG TTC
    ACT CTC ACC ATC AGC AGC GTG CAG TGT GAC GAT GCT
    GCC ACT TAC TAC TGT CAA GGC GGT TAT ACT AGT GGT
    GAT GGT ATT GCT TTC GGC GGA GGG ACC GAG GTG GTC
    GTC AAA
  • VL Amino Acid Sequence:
  • (SEQ ID NO: 7)
    QVLTQTPSPVSAAVGGTVTINCQASQSVYNNNQLSWYQQKPGQPPKLLIY
    GTSNLASGVPSRFKGSGSGTQFTLTISSVQCDDAATYYCQGGTYSGDGIA
    FGGGTEVVVK
  • VH Nucleotide Sequence:
  • (SEQ ID NO: 66)
    CAG TCG GTG GAG GAG TCC GGG GGT CGC CTG GTC ACG
    CCT GGG ACA CCC CTG ACA CTC ACC TGC ACA GTC TCT
    GGA TTC TCC CTC AGT AAC TAT GTA TTG GCC TGG GTC
    CGC CAG GCT CCA GGG AAG GGG CTG GAG TGG ATC GGA
    ACC ATT GTT AGC GGT ACC ACA TAC TAC GCG AGT TGG
    GCG AAA GGC CGA TTC ACC ATC TCC AAA ACC TCG ACC
    ACG GTG CAT CTG AAA ATC ACC AGT CCG ACA ACC GAG
    GAC ACG GCC ACC TAT TTC TGT GTC AGA GGT TTG CCT
    CAT TAT ATT ACT GGG GAC ATC TGG GGC CCA GGC ACC
    CTG GTC ACC GTC TCC TTA GGG
  • VH Amino Acid Sequence:
  • (SEQ ID NO: 8)
    QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGLGLEWIGTI
    VSGTTYYASWAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYIT
    GDIWGPGTLVTVSLG.
  • Example 3 Design of a Lateral Flow Immunoassay System Using a Rabbit Monoclonal Antibody for the Detection of THC in Saliva
  • Antibodies to be utilized in lateral flow immunoassay systems were conjugated to particles that can be visualized. Conjugation conditions were tested for each antibody lot, and monobasic/dibasic phosphate buffer pH was adjusted to optimize the conjugation, which was typically between pH 6.5 and 8.5. Antibody was diluted to 0.1 mg/ml using monobasic/dibasic phosphate buffer at room temperature and mixed with gold particles (30 nm) in the proportion 20 μl antibody per 1 mL gold for 5 minutes. 10% BSA was added to create a 1:10 dilution BSA: total volume Gold solution, and mixing was continued for five (5) minutes. The sample was centrifuged at 10,000 rpm at 4° C. for forty (40) minutes, and the supernatant removed to achieve a final suspension at 10% (w/w) gold. The gold conjugated antibodies are then utilized in the lateral flow immunoassay system, such as the system sold under the tradename OraLab®.
  • Example 4 Comparison of a Lateral Flow Immunoassay System Using a Recombinant Rabbit Monoclonal Antibody with a Murine Monoclonal Antibody for the Detection of THC
  • A murine monoclonal anti-THC antibody used in a lateral flow immunoassay was used as a comparator to assess the sensitivity of a recombinant rabbit anti-THC antibody in the same immunoassay system. The murine anti-THC antibody chosen for comparison exhibited a sensitivity higher or comparable to other available anti-THC antibodies used in lateral flow immunoassays.
  • To be directly comparable, both the mouse monoclonal anti-THC antibody and the recombinant rabbit anti-THC antibody were conjugated to gold particles using the same procedures. The conjugation buffer pH was adjusted to assure the best conjugation. All other materials and procedure were identical. Both a murine anti-THC antibody strip and a rabbit anti-THC antibody strip were prepared on the same card with the murine antibody strip on the left and rabbit antibody strip on the right.
  • Samples were prepared at the indicated parent THC concentrations. Two (2) mLs of each sample was placed in the collector, and squeezed into the tube. The cards were tested and read at 10 and 15 minutes respectively (n=3).
  • Results Ten (10) Minute Color Reading
  • Parent THC Concentrations (ng/mL)
    0 50 100 200
    Ave. Ave. Ave. Ave.
    Recombinant 2.2 2.5 1.8 2.17 0 0 0.2 0.07 0 0 0 0 0 0 0 0
    rabbit anti-
    THC
    Mouse anti- 2.2 2.2 2.0 2.13 1.2 1.7 1.0 1.30 1.0 1.5 1.0 1.17 0.7 1.2 0.5 0.8
    THC
  • Fifteen (15) Minute Color Reading
  • Parent THC Concentrations (ng/mL)
    0 50 100 200
    Ave. Ave. Ave. Ave.
    Recombinant 2.5 2.7 2.3 2.50 0 0.2 0.2 0.13 0 0 0 0 0 0 0 0
    rabbit anti-
    THC
    Mouse anti- 2.5 2.4 2.5 2.47 1.5 2.2 2.0 1.90 1.2 2.0 1.5 1.57 0.7 1.5 0.5 0.9
    THC
  • Conclusion
  • The recombinant rabbit anti-THC antibody is more sensitive than the reference mouse anti-THC antibody. At 50 ng/mL THC, the reading with the murine anti-THC antibody is only slightly decreased, and only at THC concentrations of 200 ng/mL does the murine anti-THC antibody clearly indicate that THC was present in the sample solution tested. In contrast, the reading for the recombinant rabbit anti-THC antibody was almost completely abolished at a sample concentration of 50 ng/mL THC, indicating the presence of THC in the sample at the lowest concentration of THC tested. Using the recombinant rabbit anti-THC antibody in this test system, a sensitivity of 35 ng/mL or lower is achievable.
  • Example 5 Use of a Lateral Flow Immunoassay System for the Detection of THC
  • A lateral flow immunoassay system for the detection of THC is provided to an individual. The individual is instructed to open the collector bag, place the collector foam inside his or her mouth for three minutes, and completely saturate the collector foam with saliva. The individual or other user is instructed to place the OraLab® cassette (Varian, Inc. 25200 Commercentre Drive, Lake Forest, Calif.) on a flat surface and lift the confirmation well cap to assure unobstructed saliva flow into the well. The individual then removes the collector foam from his or her mouth, places it vertically into the sample well on top of the device, and pushes the collector downward while counting slowly to five until it stops. The user is instructed to push the cap down firmly to seal the confirmation well after ten minutes have passed, and to remove the label covering the results windows and interpret the results. When the saliva sample is negative for the presence of THC, the antibody-gold particles migrate with the fluid flow by capillary action until they reach the THC loaded line, where they stop and line up to indicate a negative result. When the saliva sample is positive for the presence of THC, the antibody-gold particles migrate with the fluid flow but do not stop at the THC loaded line and instead continue upwards to the positive zone, indicating a positive result.

Claims (14)

1. An isolated antibody having specific binding for THC and its metabolites, wherein said antibody comprises CDR amino acid sequences as follows: L1: QASQSVYNNNQLS (SEQ ID NO:1); L2: GTSNLAS (SEQ ID NO:2); L3: QGGYTSGDGIA (SEQ ID NO:3); H1: GFSLSNYVLA (SEQ ID NO:4); H2: TIVSGTTYYASW (SEQ ID NO:5); H3: GLPHYITGDI (SEQ ID NO:6); or homologous amino acid sequences having at least 85% homology thereto.
2. The antibody of claim 1, wherein the amino acid sequence for the light chain variable region comprises the sequence:
QVLTQTPSPVSAAVGGTVTINCQASQSVYNNNQLSWYQQKPGQPPKLLIYGTSNLAS GVPSRFKGSGSGTQFTLTISSVQCDDAATYYCQGGYTSGDGIAFGGGTEVVVK (SEQ ID NO:7), or homologous amino acid sequences having at least 85% homology thereto.
3. The antibody of claim 1, wherein the amino acid sequence for the heavy chain variable region comprises the sequence:
QSVEESGGRLVTPGTPLTLTCTVSGFSLSNYVLAWVRQAPGKGLEWIGTIVSGTTYY ASWAKGRFTISKTSTTVHLKITSPTTEDTATYFCVRGLPHYITGDIWGPGTLVTVSLG (SEQ ID NO:8), or homologous amino acid sequences having at least 85% homology thereto.
4. The antibody of claim 1, wherein the antibody is a monoclonal antibody.
5. The monoclonal antibody of claim 4, wherein the antibody is produced by recombinant methods or by a hybridoma.
6. The monoclonal antibody of claim 4, wherein the antibody is a rabbit antibody.
7. A host cell expressing the antibody of claim 1.
8. The host cell of claim 7, wherein the host cell is a HEK 293 cell.
9. An immunoassay for detecting the presence of THC and its metabolites in a sample, comprising
contacting the sample with the antibody of claim 1, such that immune complexes form between the antibody and THC or its metabolites;
detecting and relating the binding of the antibody to the THC or its metabolites to the presence of said THC or its metabolites in the sample.
10. The immunoassay of claim 9, wherein the antibody is labeled or conjugated to particles.
11. A lateral flow immunoassay strip for detecting the presence of THC and/or its metabolites in a fluid sample, comprising a membrane strip coated with a THC-protein conjugate on a test line, and particles coated with the antibody of claim 1.
12. A method for detecting the presence of THC in a sample, comprising the steps of
(1) providing the lateral flow immunoassay strip of claim 11;
(2) contacting said lateral flow immunoassay strip with a sample to be tested for the presence of Δ9-THC in the sample;
(3) allowing the sample to migrate along the strip;
(4) determining whether the immunoassay is positive or negative for the presence of Δ9-THC in the sample by detecting the presence or absence of the antibody-coated particles in the test line region of the lateral flow immunoassay strip.
13. A kit for testing for the presence of THC in saliva, comprising the lateral flow immunoassay strip for THC of claim 11, and instructions for performing the test.
14-19. (canceled)
US11/478,972 2006-06-29 2006-06-29 Antibody and immunoassays for determining the presence of Δ9-Tetrahydrocannabinol Expired - Fee Related US7749772B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/478,972 US7749772B1 (en) 2006-06-29 2006-06-29 Antibody and immunoassays for determining the presence of Δ9-Tetrahydrocannabinol

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US11/478,972 US7749772B1 (en) 2006-06-29 2006-06-29 Antibody and immunoassays for determining the presence of Δ9-Tetrahydrocannabinol

Publications (2)

Publication Number Publication Date
US7749772B1 US7749772B1 (en) 2010-07-06
US20100184082A1 true US20100184082A1 (en) 2010-07-22

Family

ID=42307005

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/478,972 Expired - Fee Related US7749772B1 (en) 2006-06-29 2006-06-29 Antibody and immunoassays for determining the presence of Δ9-Tetrahydrocannabinol

Country Status (1)

Country Link
US (1) US7749772B1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107238713A (en) * 2017-06-23 2017-10-10 浙江诺迦生物科技有限公司 The detection method of trace hemp and the SPR chips used in a kind of saliva sample
WO2018184031A1 (en) * 2017-03-31 2018-10-04 Wang Amanda Cannabis gender identifier
WO2021087453A1 (en) * 2019-10-31 2021-05-06 The Regents Of The University Of California Systems and methods for the detection of phenolic cannabinoids
DE102020127871B3 (en) 2020-10-22 2022-03-24 Farmako GmbH Method for detecting the presence of cannabinoids in a test material and use of an immunoassay test object for detecting the presence of cannabinoids in a test material
US11647993B2 (en) * 2017-12-22 2023-05-16 Research Triangle Institute Oral fluid collector

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9406959B2 (en) 2005-05-10 2016-08-02 Board Of Trustees Of Michigan State University Electrobiocatalytic reactors having reversible bioelectronic interfaces and methods and devices related thereto
EP1933145B1 (en) * 2006-12-11 2011-09-28 AraGen Biotechnology Co. Ltd. Rapid immunochromatographic detection by amplification of the colloidal gold signal
DE102007029772B4 (en) * 2007-06-22 2011-12-08 Aj Innuscreen Gmbh Method and rapid test for the detection of specific nucleic acid sequences
US10408786B2 (en) 2012-06-14 2019-09-10 ChemiSensor, LLP Distributable chemical sampling and sensing system
US9151747B1 (en) 2013-03-14 2015-10-06 Mercyhurst University Sucralose antibody and immunoassay
US10254298B1 (en) 2015-03-25 2019-04-09 National Technology & Engineering Solutions Of Sandia, Llc Detection of metabolites for controlled substances
US9823259B1 (en) 2016-04-08 2017-11-21 Walter Nichols Detection device for cannabinoid use
US10981174B1 (en) 2016-08-04 2021-04-20 National Technology & Engineering Solutions Of Sandia, Llc Protein and nucleic acid detection for microfluidic devices
WO2018039047A1 (en) 2016-08-23 2018-03-01 Qoolabs, Inc. Lateral flow assay for assessing recombinant protein expression or reporter gene expression
WO2018200794A1 (en) * 2017-04-28 2018-11-01 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Tetrahydrocannabinol sensor

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4375149A (en) * 1979-11-14 1983-03-01 Zinser Textilmaschinen Gmbh Textile machine shut-off device
US4542104A (en) * 1983-04-06 1985-09-17 The Board Of Trustees Of The Leland Stanford Jr. Univ. Phycobiliprotein fluorescent conjugates
US5183740A (en) * 1990-02-23 1993-02-02 The United States Of America As Represented By The Secretary Of The Navy Flow immunosensor method and apparatus
US5264373A (en) * 1987-02-17 1993-11-23 Abbott Laboratories Fluorescence polarization immunoassay for tetrahydrocannabinoids
US5541113A (en) * 1993-09-22 1996-07-30 Beckman Instruments, Inc. Method for detecting an analyte using an electrochemical luminescent transition metal label
US5770458A (en) * 1995-02-10 1998-06-23 Roche Diagnostics Systems, Inc. Apparatus and method for conducting a binding assay on an absorbant carrier material
US6001658A (en) * 1996-09-13 1999-12-14 Diagnostic Chemicals Limited Test strip apparatus and method for determining presence of analyte in a fluid sample
US6368876B1 (en) * 1995-05-18 2002-04-09 Genzyme Diagnostics One step immunochromatographic device and method of use
US6468474B2 (en) * 2000-07-06 2002-10-22 Varian, Inc. Saliva testing and confirmation device
US20030022240A1 (en) * 2001-04-17 2003-01-30 Peizhi Luo Generation and affinity maturation of antibody library in silico
US6686208B2 (en) * 1997-03-18 2004-02-03 Institut Fur Chemo- Und Biosensorik Munster E.V. Device and method for carrying out fluoresence immunotests
US6719971B1 (en) * 1991-06-14 2004-04-13 Genentech, Inc. Method for making humanized antibodies
US20040133357A1 (en) * 2001-04-17 2004-07-08 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
US6800738B1 (en) * 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
US20060057027A1 (en) * 2004-09-16 2006-03-16 Hudak Andrew T Fluid collection and testing device
US20060122377A1 (en) * 2004-02-19 2006-06-08 Genentech, Inc. CDR-repaired antibodies

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US5238808A (en) 1984-10-31 1993-08-24 Igen, Inc. Luminescent metal chelate labels and means for detection
WO1990005296A1 (en) 1988-11-03 1990-05-17 Igen, Inc. Electrochemiluminescent reaction utilizing amine-derived reductant

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4375149A (en) * 1979-11-14 1983-03-01 Zinser Textilmaschinen Gmbh Textile machine shut-off device
US4542104A (en) * 1983-04-06 1985-09-17 The Board Of Trustees Of The Leland Stanford Jr. Univ. Phycobiliprotein fluorescent conjugates
US5264373A (en) * 1987-02-17 1993-11-23 Abbott Laboratories Fluorescence polarization immunoassay for tetrahydrocannabinoids
US5183740A (en) * 1990-02-23 1993-02-02 The United States Of America As Represented By The Secretary Of The Navy Flow immunosensor method and apparatus
US6719971B1 (en) * 1991-06-14 2004-04-13 Genentech, Inc. Method for making humanized antibodies
US6800738B1 (en) * 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
US5541113A (en) * 1993-09-22 1996-07-30 Beckman Instruments, Inc. Method for detecting an analyte using an electrochemical luminescent transition metal label
US5770458A (en) * 1995-02-10 1998-06-23 Roche Diagnostics Systems, Inc. Apparatus and method for conducting a binding assay on an absorbant carrier material
US6368876B1 (en) * 1995-05-18 2002-04-09 Genzyme Diagnostics One step immunochromatographic device and method of use
US6001658A (en) * 1996-09-13 1999-12-14 Diagnostic Chemicals Limited Test strip apparatus and method for determining presence of analyte in a fluid sample
US6686208B2 (en) * 1997-03-18 2004-02-03 Institut Fur Chemo- Und Biosensorik Munster E.V. Device and method for carrying out fluoresence immunotests
US6468474B2 (en) * 2000-07-06 2002-10-22 Varian, Inc. Saliva testing and confirmation device
US20040133357A1 (en) * 2001-04-17 2004-07-08 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
US20030022240A1 (en) * 2001-04-17 2003-01-30 Peizhi Luo Generation and affinity maturation of antibody library in silico
US20060122377A1 (en) * 2004-02-19 2006-06-08 Genentech, Inc. CDR-repaired antibodies
US20060057027A1 (en) * 2004-09-16 2006-03-16 Hudak Andrew T Fluid collection and testing device

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018184031A1 (en) * 2017-03-31 2018-10-04 Wang Amanda Cannabis gender identifier
CN107238713A (en) * 2017-06-23 2017-10-10 浙江诺迦生物科技有限公司 The detection method of trace hemp and the SPR chips used in a kind of saliva sample
US11647993B2 (en) * 2017-12-22 2023-05-16 Research Triangle Institute Oral fluid collector
WO2021087453A1 (en) * 2019-10-31 2021-05-06 The Regents Of The University Of California Systems and methods for the detection of phenolic cannabinoids
DE102020127871B3 (en) 2020-10-22 2022-03-24 Farmako GmbH Method for detecting the presence of cannabinoids in a test material and use of an immunoassay test object for detecting the presence of cannabinoids in a test material

Also Published As

Publication number Publication date
US7749772B1 (en) 2010-07-06

Similar Documents

Publication Publication Date Title
US7749772B1 (en) Antibody and immunoassays for determining the presence of Δ9-Tetrahydrocannabinol
CN104634955B (en) Detection anti-TNF medicine and the test of autoantibody
EP0487289B1 (en) Cyclosporin immunoassay
JP5031585B2 (en) 5-Fluoro-uracil immunoassay
CN102112489B (en) Anti-HEPCIDIN-25 selective antibodies and uses thereof
JP4576429B2 (en) Taxol immunoassay
US9441033B2 (en) Detection of synthetic cannabinoids
US9702869B2 (en) Assays, antibodies, immunogens and compositions related to 5-FU
TW200829601A (en) Conjugate and its use as a standard in an immunoassay
CA2685997C (en) Compositions and methods for combining report antibodies
US20130065323A1 (en) Detection of Synthetic Cannabinoids
US6946547B2 (en) Ecstasy-class analogs and use of same in detection of ecstasy-class compounds
Wring et al. Shorter development of immunoassay for drugs: application of the novel RIMMS technique enables rapid production of monoclonal antibodies to ranitidine
US20210061924A1 (en) Novel anti-thymidine kinase antibodies
Wang et al. Antibody and immunoassays for determining the presence of Δ 9-tetrahydrocannabinol
WO2022133193A2 (en) Reagents, methods, and systems for detecting therapeutic agents to monitor adherence to nucleoside reverse transcriptase inhibitor metabolites
JPH03190877A (en) Assay of barbiturate and compound for use therein
US20140273023A1 (en) Gemcitabine immunoassay
JP4486994B2 (en) Busulfan immunoassay
Salamone et al. A non-cannabinoid immunogen used to elicit antibodies with broad cross-reactivity to cannabinoid metabolites
EP3245230B1 (en) Anti april (a proliferation-inducing ligand) antibodies and their uses for the prognosis and/or diagnosis of cancer
US5466610A (en) Immunoassays for insulin sensitivity enhancers, insulin sensitivity enhancer antibodies, and non-thiazolidinedione insulin sensitivity enhancer compositions
JP2014519036A (en) Gemcitabine immunoassay
AU2004201615B2 (en) Anti-plasmodium compositions and methods of use
NZ619172A (en) Assays for detecting neutralizing autoantibodies to biologic therapy with tnf alpha

Legal Events

Date Code Title Description
AS Assignment

Owner name: VARIAN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WANG, DANIEL;REEL/FRAME:018116/0113

Effective date: 20060807

AS Assignment

Owner name: AGILENT TECHNOLOGIES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VARIAN, INC.;REEL/FRAME:025368/0230

Effective date: 20101029

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20140706