USRE45763E1 - Antibodies to troponin I and methods of use thereof - Google Patents

Antibodies to troponin I and methods of use thereof Download PDF

Info

Publication number
USRE45763E1
USRE45763E1 US14/035,420 US201314035420A USRE45763E US RE45763 E1 USRE45763 E1 US RE45763E1 US 201314035420 A US201314035420 A US 201314035420A US RE45763 E USRE45763 E US RE45763E
Authority
US
United States
Prior art keywords
antibody
binding protein
troponin
seq
binding
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.)
Active
Application number
US14/035,420
Inventor
Susan E. Brophy
Bailin Tu
Dagang Huang
Joan D. Tyner
Robert N. Ziemann
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.)
Abbott Laboratories
Original Assignee
Abbott Laboratories
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 Abbott Laboratories filed Critical Abbott Laboratories
Priority to US14/035,420 priority Critical patent/USRE45763E1/en
Priority to US14/461,082 priority patent/US20150024457A1/en
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TYNER, JOAN D., BROPHY, SUSAN E., HUANG, DAGANG, TU, BAILIN, ZIEMANN, ROBERT N.
Application granted granted Critical
Publication of USRE45763E1 publication Critical patent/USRE45763E1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4716Muscle proteins, e.g. myosin, actin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the subject invention relates to antibodies to troponin I as well as methods of use thereof.
  • Troponin I is a muscle protein which may be used in the determination of myocardial damage subsequent to or during, for example, a myocardial infarction.
  • troponin I is one of three subunits of the troponin complex which is located on the thin filament of the muscle contractile apparatus. This complex has a primary role in controlling the process of muscle contraction.
  • the other two subunits i.e., T and C are also immobilized on the thin myofilaments with troponin I in cardiac as well as skeletal muscle tissue.
  • immunoassays must also utilize antibodies which do not possess cross-reactivity to troponin I found in skeletal tissue. In particular, such immunoassays are needed so that appropriate therapy can be utilized by the treating physician thereby giving the affected patient the best possible prognosis.
  • the present invention pertains to binding proteins, particularly antibodies, capable of binding to cardiac troponin I.
  • these antibodies bind to one or more epitopes of troponin I.
  • the present invention also provides methods of producing and using these binding proteins or portions thereof, for example, in diagnostic assays.
  • the present invention encompasses a Chinese Hamster Ovary (CHO) cell line, referred to as TnI 19C7 AM1 hG1 CHO 204, designated by American Type Culture Collection (ATCC) deposit number PTA-9816 as well as the recombinant antibody produced by this cell line.
  • CHO Chinese Hamster Ovary
  • ATCC American Type Culture Collection
  • the present invention includes an isolated binding protein comprising an antigen-binding domain which binds to Troponin I, said antigen-binding domain comprising at least one complementarity determining region (CDR) comprising an amino acid sequence selected from the group consisting of: GYTFTDYNLH (SEQ ID NO:52), YIYPYNGITGYNQKFKS (SEQ ID NO:53), DAYDYDLTD (SEQ ID NO:54), RTSKNVGTNIH (SEQ ID NO:55), YASERLP (SEQ ID NO:56) and QQSNNWPYT (SEQ ID NO:57).
  • CDR complementarity determining region
  • the binding protein of the present invention may include, for example, at least 3 of these CDRs.
  • this binding protein may also comprise a human acceptor framework or scaffold.
  • This binding protein may be selected from the group consisting of, for example, an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a humanized antibody, a Fab, a Fab′, a F(ab′)2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, or a functionally active epitope-binding fragment of any one of these entities.
  • the present invention also encompasses an isolated nucleic acid molecule encoding a binding protein, wherein the amino acid sequence of the variable heavy chain of the binding protein has at least 70% identity to SEQ ID NO.: 25 (see FIG. 12 ).
  • This molecule may also comprise a variable light chain having at least 70% identity to SEQ ID NO.: 28 (see FIG. 12 ).
  • the present invention includes an isolated nucleic acid molecule encoding a binding protein, wherein the amino acid sequence of the variable heavy chain of said binding protein is SEQ ID NO.:25.
  • the present invention includes an isolated nucleic acid molecule encoding a binding protein, wherein the amino acid sequence of the variable light chain of said binding protein is SEQ ID NO.: 28.
  • the molecule may further comprise an isolated nucleic acid molecule encoding a variable heavy chain, wherein the amino acid sequence of the heavy chain is SEQ ID NO.: 25.
  • the present invention also includes a vector comprising one or more of the nucleic acid molecules described above, attached to a regulatory element (e.g., a promoter) as well as a host cell comprising this vector.
  • a regulatory element e.g., a promoter
  • the present invention includes a method of producing any of the binding proteins described above, capable of binding to Troponin I, which method comprises culturing the host cell, described above, for a time and under conditions sufficient to produce the binding protein of interest.
  • the invention also includes the binding protein produced by this method.
  • the present invention encompasses a pharmaceutical composition comprising any one or more of the binding proteins described above and a pharmaceutically acceptable carrier.
  • the present invention includes a method of detecting Troponin I antigen in a test sample.
  • This method comprises the steps of: contacting the test sample with an antibody which binds to Troponin I and comprises SEQ ID NO:25 for a time and under conditions sufficient for the formation of antibody/antigen complexes; and detecting presence of the complexes, presence of the complexes indicating presence of Troponin I antigen in said test sample.
  • the antibody may further comprise SEQ ID NO:28.
  • the antibody may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
  • the present invention also includes a method of detecting Troponin I antigen in a test sample comprising the steps of: contacting the test sample with a first antibody which binds to Troponin I and comprises SEQ ID NO:25 for a time and under conditions sufficient for the formation of first antibody/antigen complexes; adding a conjugate to the first antibody/antigen complexes, wherein said conjugate comprises a second antibody attached to a signal generating compound capable of generating a detectable signal, for a time and under conditions sufficient to form first antibody/antigen/second antibody complexes; and detecting presence of a signal generating by the signal generating compound, presence of the signal indicating presence of Troponin I antigen in said test sample.
  • the first antibody may further comprise SEQ ID NO:28 and may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
  • the present invention includes a method of detecting Troponin I antigen in a test sample comprising the steps of contacting Troponin I antigen with an antibody to Troponin I for a time and under conditions sufficient to form Troponin I antigen/antibody complexes, wherein the antibody comprises SEQ ID NO:25 and is labeled with a signal-generating compound capable of generating a detectable signal; adding the test sample to said Troponin I antigen/antibody complexes for a time and under conditions sufficient to form Troponin I antigen/antibody/Troponin I test sample antigen complexes; and detecting presence of a signal generating by the signal generating compound, presence of the signal indicating presence of Troponin I antigen in the test sample.
  • the antibody may further comprise SEQ ID NO:28 and may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
  • the present invention also encompasses another method of detecting Troponin I antigen in a test sample.
  • This method comprises the steps of: contacting the test sample with 1) a Troponin I reference antigen, wherein the antigen is attached to a signal generating compound capable of generating a detectable signal and 2) an antibody to Troponin I antigen wherein the antibody comprises SEQ ID NO:25, for a time and under conditions sufficient to form Troponin I reference antigen/antibody complexes; and detecting a signal generated by the signal generating compound, wherein the amount of Troponin I antigen detected in the test sample is inversely proportional to the amount of Troponin I reference antigen bound to the antibody.
  • the antibody may further comprise SEQ ID NO:28 and may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
  • the present invention includes pharmaceutical composition comprising any one or more of the binding proteins described above and a pharmaceutically acceptable carrier.
  • the present invention also encompasses a method of diagnosing acute coronary syndrome or myocardial infarction in a patient suspected of having one of these conditions.
  • This method comprises the steps of: isolating a biological sample from the patient; contacting the biological sample with an antibody which binds to Troponin I and comprises SEQ ID NO:25, for a time and under conditions sufficient for formation of Troponin I antigen/antibody complexes; detecting presence of the Troponin I antigen/antibody complexes; dissociating the Troponin I antigen present in the complexes from the antibody present in said complexes; and measuring the amount of dissociated Troponin I antigen, wherein an amount of Troponin I antigen greater than approximately 1-5 times the Troponin I value of the 99 th percentile of a normal population indicates a diagnosis of acute coronary syndrome or myocardial infarction in the patient.
  • the present invention includes an additional method method of diagnosing acute coronary syndrome or myocardial infarction in a patient suspected of having one of these conditions.
  • This method comprises the steps of: isolating a biological sample from the patient; contacting the biological sample with a first antibody which binds to Troponin I and comprises SEQ ID NO:25, for a time and under conditions sufficient for the formation of Troponin I antigen/antibody complexes; adding a conjugate to the resulting Troponin I antigen/antibody complexes for a time and under conditions sufficient to allow the conjugate to bind to the bound Troponin I antigen, wherein the conjugate comprises a second antibody attached to a signal generating compound capable of generating a detectable signal; detecting the presence of Troponin I antigen which may be present in said biological sample by detecting a signal generated by said signal generating compound; and measuring the amount of Troponin I antigen present in the test sample by measuring the intensity of the signal, an amount of Troponin I antigen greater than approximately 1
  • the present invention also includes a kit comprising any one or more of the monoclonal antibodies or binding proteins described above and, if needed, instructions describing the manner in which to use this kit.
  • the present invention includes an isolated binding protein which comprises an antigen-binding domain, wherein the antigen-binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of:
  • FIG. 1 is a flow chart showing the steps used to identify and create antibodies that have improved affinity for troponin I.
  • FIG. 2 is the nucleotide description (SEQ ID NO: 109 and SEQ ID NO: 110) of the wild-type TnI 19C7 single-chain variable fragment (“scFv”).
  • FIG. 3 shows that yeast expressing full-length TnI 19C7 single-chain variable fragment (scFv) bind to single chain troponin I (28-110aa)-linker-troponin C known as scTnI-C-2 (Spectral diagnostics, RP-3700). More specifically, this figure shows that TnI 19C7 scFv expressing yeast were incubated with scTnI-C-2 or anti-V5, followed by either anti-troponin mAb and goat anti mouse-phycoerythrin (GAM:PE) ( FIG. 3B ) or GAM:PE respectively ( FIG. 3A ).
  • GAM:PE goat anti mouse-phycoerythrin
  • the flow cytometry histograms illustrate the full-length expression of TnI 19C7 scFv as detected by anti-V5 and the ability of TnI 19C7 scFv to bind to scTnI-C-2.
  • PE-A units abcissa
  • 10 2 , 10 3 , 10 4 , and 10 5 Count units (ordinate): 10 2 , 10 3 , 10 4 , and 10 5 .
  • FIG. 4 shows the TnI 19C7 scFv off-rate measurement. More specifically, yeast expressing TnI 19C7 scFv were incubated with a saturating concentration of scTnI-C-2. Cells were washed twice and at each time point, cells were transferred to ice, washed and incubated with anti-TnI mAb. After 30 minutes, cells were washed again and incubated with goat anti-mouse phycoerythrin. Again after 30 minutes, cells were washed and analyzed on the flow cytometer.
  • a first order decay equation was used to fit the individual time points where ml was the theoretical maximum mean fluorescence units (“MFU”) at time 0, m2 was the off-rate (“koff”), m3 was the background MFU due to autofluorescence and M0, which is the time x (the x being the time that is being measured) was the time x that measurements are taken.
  • MFU theoretical maximum mean fluorescence units
  • m2 was the off-rate
  • m3 was the background MFU due to autofluorescence
  • M0 which is the time x (the x being the time that is being measured) was the time x that measurements are taken.
  • FIG. 5 shows the TnI 19C7 scFv equilibrium dissociation constant (KD) measurement. More specifically, yeast expressing TnI 19C7 scFv were incubated with varying concentrations of scTnI-C-2. Cells were washed twice with PBS pH6.8/2% BSA/0.02% Standapol ES-1 and incubated with anti-TnI mAb for 30 min Cells were washed again and incubated with goat anti-mouse phycoerythrin for 30 min. Finally, cells were washed and analyzed on the flow cytometer.
  • KD equilibrium dissociation constant
  • FIG. 6 is a schematic depiction that shows how degenerate oligonucleotides were designed so that primers are made such that for each CDR nucleotide residue 70% remains the wild-type residue and 30% a mix of the other three residues.
  • Two PCR products are generated for each library a spiked (sp) PCR product and a non-spiked PCR product. The spiked and non-spiked PCR products are combined to generate an intact CDR mutagenized scFv library.
  • FIG. 7 is a schematic depiction that shows how the TnI 19C7 scFv library was constructed using yeast homologous recombination. More specifically, the spiked CDR PCR product and the excised yeast display vector were transformed into S. cerevisiae strain EBY100. Transformed clones were selected in tryptophan deficient glucose media.
  • FIG. 8 is a summary showing the PCR primers that were used to generate the scFv construct (tpVHfor through tpVL-rev), those used to generate the CDR spiked libraries (19H1spfor through pYD41rev2) and those used to generate the combination library (19FRH2for to 19FRL3) (see SEQ ID Nos:1-22).
  • the bold and enlarged areas of the primers represent those regions in which a “70% wild-type, 30% other nucleotide mixture” was incorporated while the primers were being made. Such a “spiked” primer generated the diversity within the library.
  • FIG. 9 shows equilibrium dissociation constant (KD) measurements of selected TnI 19C7 scFv determined as described above in FIG. 5 .
  • FIG. 10 shows the results of relative antibody affinity as measured as an antigen 50% (Ag50).
  • Ag50 an antigen 50%
  • TnI 19C7 clones were converted into mouse IgG2ak antibodies by cloning the variable domains onto the immunoglobulin constant domains. Antibodies were expressed in a transient HEK 294 cell system.
  • the Ag50 is the concentration of scTnI-C at which is 50% of the maximum signal and represents the relative affinity ranking of the selected TnI 19C7 AM candidates.
  • TnI 19C7 AM1 exemplifies the tightest relative affinity compared to the TnI 19C7 wild-type antibody.
  • FIG. 11 illustrates TnI 19C7 AM1's ability to bind to scTnI-C in an ARCHITECT® assay format (Abbott Laboratories, Abbott Park, Ill.).
  • TnI 19C7 was labeled with acridinium and assayed for binding to scTnI-C using anti-TnI capture beads.
  • TnI 19C7 AM1 exhibited better binding in this assay format for the range of calibrators compared to the wild-type TnI 19C7 antibody.
  • FIG. 12 illustrates the nucleotide (SEQ ID NO:23, SEQ ID NO:24 (complement), SEQ ID NO:26 and SEQ ID NO:27 (complement)) and encoded amino acid sequences of the heavy (SEQ ID NO:25) and light (SEQ ID NO:28) chains of monoclonal antibody TnI 19C7 AM1 and, in particular, of the complementarity determining regions (CDRs).
  • FIG. 13 illustrates the positions within the heavy and light chains of the TnI 19C7 CDRs that may be substituted with amino acids other than those shown in FIG. 12 (SEQ ID Nos: 30-49).
  • polypeptide refers to any polymeric chain of amino acids.
  • peptide and protein are used interchangeably with the term polypeptide and also refer to a polymeric chain of amino acids.
  • polypeptide encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence.
  • a polypeptide may be monomeric or polymeric.
  • isolated protein or “isolated polypeptide” is a protein or polypeptide that by virtue of its origin or source of derivation is not associated with naturally associated components that accompany it in its native state; is substantially free of other proteins from the same species; is expressed by a cell from a different species; or does not occur in nature.
  • a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components.
  • a protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
  • recovering refers to the process of rendering a chemical species such as a polypeptide substantially free of naturally associated components by isolation, e.g., using protein purification techniques well known in the art.
  • the subject invention also includes isolated nucleotide sequences (or fragments thereof) encoding the variable light and heavy chains of the antibodies described herein as well as those nucleotide sequences (or fragments thereof) having sequences comprising, corresponding to, identical to, hybridizable to, or complementary to, at least about 70% (e.g., 70% 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78% or 79%), preferably at least about 80% (e.g., 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88% or 89%), and more preferably at least about 90% (e.g, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) identity to these encoding nucleotide sequences.
  • 70% e.g., 70% 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78% or 79%
  • 80%
  • sequences may be derived from any source (e.g., either isolated from a natural source, produced via a semi-synthetic route, or synthesized de novo).
  • source e.g., either isolated from a natural source, produced via a semi-synthetic route, or synthesized de novo.
  • sequences may be isolated or derived from sources other than described in the examples (e.g., bacteria, fungus, algae, mouse or human).
  • the present invention also includes amino acid sequences of the variable light and heavy chains of the antibodies described herein (or fragments of these amino acid sequences). Further, the present invention also includes amino acid sequences (or fragments thereof) comprising, corresponding to, identical to, or complementary to at least about 70% (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78% or 79%), preferably at least about 80% (e.g., 80% 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88% or 89%), and more preferably at least about 90% identity (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%), to the amino acid sequences of the proteins of the present invention. (Again, all integers (and portions thereof) between and including 70% and 100% (as recited in connection with the nucleotide sequence identities
  • a “fragment” of a nucleotide sequence is defined as a contiguous sequence of approximately at least 6, preferably at least about 8, more preferably at least about 10 nucleotides, and even more preferably at least about 15 nucleotides corresponding to a region of the specified nucleotide sequence.
  • identity refers to the relatedness of two sequences on a nucleotide-by-nucleotide basis over a particular comparison window or segment. Thus, identity is defined as the degree of sameness, correspondence or equivalence between the same strands (either sense or antisense) of two DNA segments (or two amino acid sequences). “Percentage of sequence identity” is calculated by comparing two optimally aligned sequences over a particular region, determining the number of positions at which the identical base or amino acid occurs in both sequences in order to yield the number of matched positions, dividing the number of such positions by the total number of positions in the segment being compared and multiplying the result by 100.
  • Optimal alignment of sequences may be conducted by the algorithm of Smith & Waterman, Appl. Math. 2:482 (1981), by the algorithm of Needleman & Winch, J. Mol. Biol. 48:443 (1970), by the method of Pearson & Lipmann, Proc. Natl. Acad. Sci. (USA) 85:2444 (1988) and by computer programs which implement the relevant algorithms (e.g., Crustal Macaw Pileup (Higgins et al., CABIOS.
  • complementarity is defined as the degree of relatedness between two DNA segments. It is determined by measuring the ability of the sense strand of one DNA segment to hybridize with the antisense strand of the other DNA segment, under appropriate conditions, to form a double helix.
  • a “complement” is defined as a sequence which pairs to a given sequence based upon the canonic base-pairing rules. For example, a sequence A-G-T in one nucleotide strand is “complementary” to T-C-A in the other strand.
  • Similarity between two amino acid sequences is defined as the presence of a series of identical as well as conserved amino acid residues in both sequences. The higher the degree of similarity between two amino acid sequences, the higher the correspondence, sameness or equivalence of the two sequences. (“Identity between two amino acid sequences is defined as the presence of a series of exactly alike or invariant amino acid residues in both sequences.) The definitions of “complementarity”, “identity” and “similarity” are well known to those of ordinary skill in the art.
  • Encoded by refers to a nucleic acid sequence which codes for a polypeptide sequence, wherein the polypeptide sequence or a portion thereof contains an amino acid sequence of at least 3 amino acids, more preferably at least 8 amino acids, and even more preferably at least 15 amino acids from a polypeptide encoded by the nucleic acid sequence.
  • Bio activity refers to all inherent biological properties of an antibody against troponin I or troponin I. Such properties include, for example, the ability of the antibody to bind to troponin I and functionally-related antibodies described herein.
  • telomere binding in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope “A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled “A” and the antibody, will reduce the amount of labeled A bound to the antibody.
  • a particular structure e.g., an antigenic determinant or epitope
  • antibody broadly refers to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any functional fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule.
  • Ig immunoglobulin
  • Such mutant, variant, or derivative antibody entities are known in the art, non-limiting embodiments of which are discussed below.
  • each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG 1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass and may be from any species (e.g., mouse, human, chicken, rat, rabbit, sheep, shark and camelid).
  • the CDRs of the antibodies of the present invention are shown in Tables 1 and 2 below:
  • antibody portion refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by one or more fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific, specifically binding to two or more different antigens.
  • binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′) 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546, Winter et al., Intern. Appln. Public.
  • WO 90/05144 A1 herein incorporated by reference
  • VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • scFv single chain Fv
  • single chain antibodies are also encompassed herein within the term “antigen-binding portion” of an antibody.
  • Other forms of single chain antibodies, such as diabodies, are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123).
  • Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5).
  • antibody construct refers to a polypeptide comprising one or more the antigen binding portions of the invention linked to a linker polypeptide or an immunoglobulin constant domain.
  • Linker polypeptides comprise two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions.
  • Such linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123).
  • An immunoglobulin constant domain refers to a heavy or light chain constant domain. Human IgG heavy chain and light chain constant domain amino acid sequences are known in the art, and examples are presented in Table 3.
  • an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides.
  • immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol.
  • Antibody portions such as Fab and F(ab′) 2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies.
  • antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.
  • an “isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds troponin I is substantially free of antibodies that specifically bind antigens other than troponin I).
  • An isolated antibody that specifically binds troponin I may, however, have cross-reactivity to other antigens, such as troponin I molecules from other species.
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described below), antibodies isolated from a recombinant, combinatorial human antibody library (Hoogenboom H. R., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W. E., (2002) Clin. Biochem. 35:425-445; Gavilondo J. V., and Larrick J. W. (2002) BioTechniques 29:128-145; Hoogenboom H., and Chames P.
  • such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • chimeric antibody refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species.
  • the present invention encompasses chimeric antibodies having, for example, murine heavy and light chain variable regions linked to human constant regions.
  • CDR-grafted antibody refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.
  • humanized antibody refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more “human-like”, i.e., more similar to human germline variable sequences.
  • a non-human species e.g., a mouse
  • human CDR-grafted antibody in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences.
  • Kabat numbering “Kabat definitions” and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e. hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391 and Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3.
  • the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
  • the AbM definition as defined by Oxford Molecular's ABM antibody modeling software was used to define the CDR-H1 region from amino acids 26-35 for the heavy chain.
  • the terms “acceptor” and “acceptor antibody” refer to the antibody or nucleic acid sequence providing or encoding at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or 100% of the amino acid sequences of one or more of the framework regions.
  • the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding the constant region(s).
  • the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding one or more of the framework regions and the constant region(s).
  • the term “acceptor” refers to a human antibody amino acid or nucleic acid sequence that provides or encodes at least 80%, preferably, at least 85%, at least 90%, at least 95%, at least 98%, or 100% of the amino acid sequences of one or more of the framework regions.
  • an acceptor may contain at least 1, at least 2, at least 3, least 4, at least 5, or at least 10 amino acid residues that does (do) not occur at one or more specific positions of a human antibody.
  • acceptor framework region and/or acceptor constant region(s) may be, e.g., derived or obtained from a germline antibody gene, a mature antibody gene, a functional antibody (e.g., antibodies well-known in the art, antibodies in development, or antibodies commercially available).
  • CDR refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3, for each of the variable regions.
  • CDR set refers to a group of three CDRs that occur in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md.
  • CDR boundary definitions may not strictly follow one of the above systems, such as AbM definitions, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding.
  • the methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat, AbM or Chothia defined CDRs.
  • canonical residue refers to a residue in a CDR or framework that defines a particular canonical CDR structure as defined by Chothia et al. (J. Mol. Biol. 196:901-907 (1987); Chothia et al., J. Mol. Biol. 227: 799 (1992), both are incorporated herein by reference). According to Chothia et al., critical portions of the CDRs of many antibodies have nearly identical peptide backbone confirmations despite great diversity at the level of amino acid sequence. Each canonical structure specifies primarily a set of peptide backbone torsion angles for a contiguous segment of amino acid residues forming a loop.
  • the terms “donor” and “donor antibody” refer to an antibody providing one or more CDRs.
  • the donor antibody is an antibody from a species different from the antibody from which the framework regions are obtained or derived.
  • the term “donor antibody” refers to a non-human antibody providing one or more CDRs.
  • the term “framework” or “framework sequence” refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to correspondingly different interpretations.
  • the six CDRs (CDR-L1, -L2, and -L3 of light chain and CDR-H1, -H2, and -H3 of heavy chain) also divide the framework regions on the light chain and the heavy chain into four sub-regions (FR1, FR2, FR3 and FR4) on each chain, in which CDR1 is positioned between FR1 and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4.
  • a framework region represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain.
  • a FR represents one of the four sub-regions, and FRs represents two or more of the four sub-regions constituting a framework region.
  • the murine heavy chain and light chain donor sequences are selected from the sequences described below:
  • the term “germline antibody gene” or “gene fragment” refers to an immunoglobulin sequence encoded by non-lymphoid cells that have not undergone the maturation process that leads to genetic rearrangement and mutation for expression of a particular immunoglobulin. (See, e.g., Shapiro et al., Crit. Rev. Immunol. 22(3): 183-200 (2002); Marchalonis et al., Adv Exp Med Biol. 484:13-30 (2001)).
  • One of the advantages provided by various embodiments of the present invention stems from the recognition that germline antibody genes are more likely than mature antibody genes to conserve essential amino acid sequence structures characteristic of individuals in the species, hence less likely to be recognized as from a foreign source when used therapeutically in that species.
  • key residues refer to certain residues within the variable region that have more impact on the binding specificity and/or affinity of an antibody, in particular a humanized antibody.
  • a key residue includes, but is not limited to, one or more of the following: a residue that is adjacent to a CDR, a potential glycosylation site (can be either N- or O-glycosylation site), a rare residue, a residue capable of interacting with the antigen, a residue capable of interacting with a CDR, a canonical residue, a contact residue between heavy chain variable region and light chain variable region, a residue within the Vernier zone, and a residue in the region that overlaps between the Chothia definition of a variable heavy chain CDR1 and the Kabat definition of the first heavy chain framework.
  • humanized antibody is an antibody or a variant, derivative, analog or fragment thereof which immunospecifically binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody.
  • FR framework
  • CDR complementary determining region
  • substantially in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98% and most preferably at least 99% identical to the amino acid sequence of a non-human antibody CDR.
  • a humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab′, F(ab′)2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence.
  • a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain.
  • the antibody also may include the CH1, hinge, CH2, CH3, and CH4 regions of the heavy chain.
  • a humanized antibody only contains a humanized light chain. In some embodiments, a humanized antibody only contains a humanized heavy chain. In specific embodiments, a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.
  • the humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including without limitation IgG 1, IgG2, IgG3 and IgG4.
  • the humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well-known in the art.
  • the framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. In a preferred embodiment, such mutations, however, will not be extensive. Usually, at least 80%, preferably at least 85%, more preferably at least 90%, and most preferably at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences.
  • the term “consensus framework” refers to the framework region in the consensus immunoglobulin sequence.
  • the term “consensus immunoglobulin sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
  • Vernier zone refers to a subset of framework residues that may adjust CDR structure and fine-tune the fit to antigen as described by Foote and Winter (1992, J. Mol. Biol. 224:487-499, which is incorporated herein by reference). Vernier zone residues form a layer underlying the CDRs and may impact on the structure of CDRs and the affinity of the antibody.
  • activity includes activities such as the binding specificity/affinity of an antibody for an antigen, for example, an anti-troponin I antibody that binds to troponin I.
  • epitope includes any polypeptide determinant capable of specific binding to an immunoglobulin or T-cell receptor.
  • epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • An epitope is a region of an antigen that is bound by an antibody.
  • an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • BIAcore Pharmaacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.
  • K on is intended to refer to the on rate constant for association of an antibody to the antigen to form the antibody/antigen complex as is known in the art.
  • K off is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex as is known in the art.
  • K d or “K D ”, as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction as is known in the art.
  • label binding protein refers to a protein with a label incorporated that provides for the identification of the binding protein.
  • the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or calorimetric methods).
  • labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H, 14 C, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I, 177 Lu, 166 Ho, or 153 Sm); fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates.
  • radioisotopes or radionuclides e.g., 3 H, 14 C, 35 S, 90 Y, 99 Tc,
  • antibody conjugate refers to a binding protein, such as an antibody, chemically linked to a second chemical moiety, such as a therapeutic or cytotoxic agent.
  • agent is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
  • the therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • crystal refers to an antibody, or antigen-binding portion thereof, that exists in the form of a crystal.
  • Crystals are one form of the solid state of matter, which is distinct from other forms such as the amorphous solid state or the liquid crystalline state.
  • Crystals are composed of regular, repeating, three-dimensional arrays of atoms, ions, molecules (e.g., proteins such as antibodies), or molecular assemblies (e.g., antigen/antibody complexes). These three-dimensional arrays are arranged according to specific mathematical relationships that are well-understood in the field.
  • the fundamental unit, or building block, that is repeated in a crystal is called the asymmetric unit.
  • Repetition of the asymmetric unit in an arrangement that conforms to a given, well-defined crystallographic symmetry provides the “unit cell” of the crystal. Repetition of the unit cell by regular translations in all three dimensions provides the crystal. See Giege, R. and Ducruix, A. Barrett, Crystallization of Nucleic Acids and Proteins, a Practical Approach, 2nd ed., pp. 20 1-16, Oxford University Press, New York, N.Y., (1999).”
  • polynucleotide as referred to herein means a polymeric form of two or more nucleotides, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide.
  • the term includes single and double-stranded forms of DNA but preferably is double-stranded DNA.
  • isolated polynucleotide shall mean a polynucleotide (e.g., of genomic, cDNA, or synthetic origin, or some combination thereof) that, by virtue of its origin, is not associated with all or a portion of a polynucleotide with which the “isolated polynucleotide” is found in nature; is operably linked to a polynucleotide that it is not linked to in nature; or does not occur in nature as part of a larger sequence.
  • a polynucleotide e.g., of genomic, cDNA, or synthetic origin, or some combination thereof
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector may be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • operably linked refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • “Operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
  • expression control sequence refers to polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated.
  • Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • the nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence.
  • control sequences is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • Transformation refers to any process by which exogenous DNA enters a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment. Such “transformed” cells include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells that transiently express the inserted DNA or RNA for limited periods of time.
  • host cell is intended to refer to a cell into which exogenous DNA has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life. Preferred eukaryotic cells include protist, fungal, plant and animal cells.
  • host cells include but are not limited to the prokaryotic cell line E. coli; mammalian cell lines CHO, HEK 293 and COS; the insect cell line Sf9; and the fungal cell Saccharomyces cerevisiae or Picchia pastoris.
  • Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection).
  • Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein.
  • the foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.
  • Transgenic organism refers to an organism having cells that contain a transgene, wherein the transgene introduced into the organism (or an ancestor of the organism) expresses a polypeptide not naturally expressed in the organism.
  • a “transgene” is a DNA construct, which is stably and operably integrated into the genome of a cell from which a transgenic organism develops, directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic organism.
  • the term “regulate” and “modulate” are used interchangeably, and, as used herein, refers to a change or an alteration in the activity of a molecule of interest (e.g., the biological activity of troponin I). Modulation may be an increase or a decrease in the magnitude of a certain activity or function of the molecule of interest. Exemplary activities and functions of a molecule include, but are not limited to, binding characteristics, enzymatic activity, cell receptor activation, and signal transduction.
  • a modulator is a compound capable of changing or altering an activity or function of a molecule of interest (e.g., the biological activity of troponin I).
  • a modulator may cause an increase or decrease in the magnitude of a certain activity or function of a molecule compared to the magnitude of the activity or function observed in the absence of the modulator.
  • a modulator is an inhibitor, which decreases the magnitude of at least one activity or function of a molecule.
  • Exemplary inhibitors include, but are not limited to, proteins, peptides, antibodies, peptibodies, carbohydrates or small organic molecules. Peptibodies are described, e.g., in International Application Publication No. WO 01/83525.
  • agonist refers to a modulator that, when contacted with a molecule of interest, causes an increase in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the agonist.
  • agonists of interest may include, but are not limited to, troponin I polypeptides, nucleic acids, carbohydrates, or any other molecules that bind to troponin I.
  • antagonist refers to a modulator that, when contacted with a molecule of interest causes a decrease in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the antagonist.
  • the term “effective amount” refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity and/or duration of a disorder or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g., prophylactic or therapeutic agent).
  • sample includes, but is not limited to, any quantity of a substance from a living thing or formerly living thing.
  • living things include, but are not limited to, humans, mice, rats, monkeys, dogs, rabbits and other mammalian or non-mammalian animals.
  • substances include, but are not limited to, blood, serum, urine, synovial fluid, cells, organs, tissues (e.g., brain), bone marrow, lymph nodes, cerebrospinal fluid, and spleen.
  • Antibodies of the present invention may be made by any of a number of techniques known in the art. For example, antibodies can be prepared using a wide variety of techniques including the use of recombinant or phage display technologies, or a combination thereof.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • the present invention provides a method of generating recombinant antibodies (as well as antibodies produced by the method) comprising culturing a Chinese Hamster Ovary cell line secreting an antibody of the invention.
  • fragments of the antibody of the present invention which recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab′)2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′)2 fragments).
  • F(ab′)2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
  • In vitro methods also can be used to make the antibodies of the invention, wherein an antibody library is screened to identify an antibody having the desired binding specificity.
  • Methods for such screening of recombinant antibody libraries are well known in the art and include methods described in, for example, Ladner et al., U.S. Pat. No. 5,223,409; Kang et al., International Appln. Publication No. WO 92/18619; Dower et al., International Appln. Publication No. WO 91/17271; Winter et al., International Appln. Publication No. WO 92/20791; Markland et al., International Appln. Publication No.
  • the recombinant antibody library may be from a subject immunized with troponin I, or a portion thereof Alternatively, the recombinant antibody library may be from a naive subject, i.e., one who has not been immunized with troponin I, such as a human antibody library from a human subject who has not been immunized with human troponin I.
  • Antibodies of the invention are selected by screening the recombinant antibody library with the peptide comprising human troponin I to thereby select those antibodies that recognize troponin I. Methods for conducting such screening and selection are well known in the art, such as described in the references in the preceding paragraph.
  • the invention pertains to an isolated antibody, or an antigen-binding portion thereof, that binds human troponin I.
  • the antibody is a recombinant antibody.
  • the antibodies of the present invention can also be generated using yeast display methods known in the art.
  • yeast display methods genetic methods are used to tether antibody domains to the yeast cell wall and display them on the surface of yeast.
  • yeast can be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • yeast display methods that can be used to make the antibodies of the present invention include those disclosed Wittrup et al., U.S. Pat. No. 6,699,658 incorporated herein by reference.
  • antibodies of the present invention may be produced by any number of techniques known in the art. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques is the preferred method of producing the antibodies of the present invention.
  • transfection is intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • prokaryotic or eukaryotic host cells expression of antibodies in eukaryotic cells is preferable, and most preferable in mammalian host cells, because such eukaryotic cells (and in particular mammalian cells) are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr—CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R. J. Kaufman and P. A. Sharp (1982) Mol. Biol. 159:601-621), NS0 myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr CHO cells
  • DHFR selectable marker e.g., as described in R. J. Kaufman and P. A. Sharp (1982) Mol. Biol. 159:601-621
  • NS0 myeloma cells e.g., as described in R. J. Kaufman and P. A. Sharp (1982) Mol. Biol. 159:601-621
  • the antibodies When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
  • Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
  • bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr—CHO cells by calcium phosphate-mediated transfection.
  • the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes.
  • the recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification.
  • the selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium.
  • Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell of the invention in a suitable culture medium until a recombinant antibody of the invention is synthesized. The method can further comprise isolating the recombinant antibody from the culture medium.
  • the isolated anti-troponin I antibody CDR sequences described herein establish a novel family of troponin I binding proteins, isolated in accordance with this invention, and comprising polypeptides that include the CDR sequences listed in Tables 1 and 2 above.
  • standard methods known in the art for generating binding proteins of the present invention and assessing the binding characteristics thereof may be used, including but not limited to those specifically described herein.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies, such as those of the present invention are well known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4;214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entireties.
  • the chimeric antibodies of the invention are produced by replacing the heavy chain constant region of the antibodies described above with a human IgG1 constant region.
  • the chimeric antibody of the invention comprises a heavy chain variable region (V H ) comprising the amino acid sequence of SEQ ID NO:25 and a light chain variable region (V L ) comprising the amino acid sequence of SEQ ID NO:28.
  • CDR-grafted antibodies of the invention comprise heavy and light chain variable region sequences from a human antibody wherein one or more of the CDR regions of V H and/or V L are replaced with CDR sequences of the murine antibodies of the invention.
  • a framework sequence from any human antibody may serve as the template for CDR grafting.
  • straight chain replacement onto such a framework often leads to some loss of binding affinity to the antigen. The more homologous a human antibody is to the original murine antibody, the less likely the possibility that combining the murine CDRs with the human framework will introduce distortions in the CDRs that could reduce affinity.
  • the human variable framework that is chosen to replace the murine variable framework apart from the CDRs have at least a 65% sequence identity with the murine antibody variable region framework. It is more preferable that the human and murine variable regions apart from the CDRs have at least 70% sequence identify. It is even more preferable that the human and murine variable regions apart from the CDRs have at least 75% sequence identity. It is most preferable that the human and murine variable regions apart from the CDRs have at least 80% sequence identity.
  • Methods for producing chimeric antibodies are known in the art and discussed in detail in Example 2.2. (See also EP 239,400; Intern. Appln. Publication No. WO 91/09967; U.S. Pat. Nos.
  • Humanized antibodies are antibody molecules from non-human species antibody that bind the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule Such imported sequences can be used to reduce immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic, as known in the art.
  • CDRs complementarity determining regions
  • Framework residues in the human framework regions may be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Antibodies can be humanized using a variety of techniques known in the art, such as but not limited to those described in Jones et al., Nature 321:522 (1986); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol. 151: 2296 (1993); Chothia and Lesk, J. Mol. Biol. 196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285 (1992); Presta et al., J. Immunol.
  • antibodies of the present invention exhibit a high capacity to bind specifically to troponin I, e.g., as assessed by any one of several in vitro and in vivo assays known in the art (e.g., see examples below).
  • the antibody comprises a heavy chain constant region, such as an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region.
  • the heavy chain constant region is an IgG1 heavy chain constant region or an IgG4 heavy chain constant region.
  • the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region.
  • the antibody comprises a kappa light chain constant region.
  • the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
  • the Fc portion of an antibody mediates several important effector functions, for example, cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/clearance rate of antibody- and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives.
  • Neonatal Fc receptors are the critical components determining the circulating half-life of antibodies.
  • at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered.
  • a labeled binding protein wherein an antibody or antibody portion of the invention is derivatized or linked to another functional molecule (e.g., another peptide or protein).
  • a labeled binding protein of the invention can be derived by functionally linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • Useful detectable agents with which an antibody or antibody portion of the invention may be derivatized include fluorescent compounds.
  • Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like.
  • An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • the detectable agent horseradish peroxidase when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable.
  • An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • Another embodiment of the invention provides a crystallized binding protein.
  • the invention relates to crystals of whole anti-troponin I antibodies and fragments thereof as disclosed herein, and formulations and compositions comprising such crystals.
  • the crystallized binding protein has a greater half-life in vivo than the soluble counterpart of the binding protein.
  • the binding protein -retains biological activity after crystallization.
  • Crystallized binding protein of the invention may be produced according methods known in the art and as disclosed in International Appln. Publication No. WO 02/072636, incorporated herein by reference.
  • Another embodiment of the invention provides a glycosylated binding protein wherein the antibody or antigen-binding portion thereof comprises one or more carbohydrate residues.
  • Nascent in vivo protein production may undergo further processing, known as post-translational modification.
  • sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation.
  • glycosylation The resulting proteins bearing covalently linked oligosaccharide side chains are known as glycosylated proteins or glycoproteins.
  • Antibodies are glycoproteins with one or more carbohydrate residues in the Fc domain, as well as the variable domain.
  • Carbohydrate residues in the Fc domain have important effect on the effector function of the Fc domain, with minimal effect on antigen binding or half-life of the antibody (R. Jefferis, Biotechnol. Prog. 21 (2005), pp. 11-16).
  • glycosylation of the variable domain may have an effect on the antigen binding activity of the antibody.
  • Glycosylation in the variable domain may have a negative effect on antibody binding affinity, likely due to steric hindrance (Co, M. S., et al., Mol. Immunol. (1993) 30:1361-1367), or result in increased affinity for the antigen (Wallick, S. C., et al., Exp. Med. (1988) 168:1099-1109; Wright, A., et al., EMBO J. (1991) 10:2717 2723).
  • One aspect of the present invention is directed to generating glycosylation site mutants in which the O- or N-linked glycosylation site of the binding protein has been mutated.
  • One skilled in the art can generate such mutants using standard well-known technologies.
  • the creation of glycosylation site mutants that retain the biological activity but have increased or decreased binding activity are another object of the present invention.
  • the glycosylation of the antibody or antigen-binding portion of the invention is modified.
  • an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen.
  • carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may increase the affinity of the antibody for antigen.
  • Such an approach is described in further detail in International Appln. Publication No. WO 03/016466A2, and U.S. Pat. Nos. 5,714,350 and 6,350,861, each of which is incorporated herein by reference in its entirety.
  • a modified antibody of the invention can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNAc structures.
  • altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol. Chem.
  • Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms may produce different glycosylation enzymes (e.g., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid.
  • the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human.
  • a therapeutic protein produced in a microorganism host such as yeast
  • glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line.
  • Such glycoproteins may also be immunogenic in humans and show reduced half-life in vivo after administration.
  • Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream.
  • a practitioner may prefer a therapeutic protein with a specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal.
  • glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. Using techniques known in the art a practitioner may generate antibodies or antigen-binding portions thereof exhibiting human protein glycosylation. For example, yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins (glycoproteins) produced in these yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S. Patent Application Publication Nos. 20040018590 and 20020137134 and International Appln. Publication No. WO 05/100584 A2).
  • multivalent binding protein is used in this specification to denote a binding protein comprising two or more antigen binding sites.
  • the multivalent binding protein is preferably engineered to have the three or more antigen binding sites, and is generally not a naturally occurring antibody.
  • multispecific binding protein refers to a binding protein capable of binding two or more related or unrelated targets.
  • Dual variable domain (DVD) binding proteins as used herein, are binding proteins that comprise two or more antigen binding sites and are tetravalent or multivalent binding proteins. Such DVDs may be monospecific, i.e., capable of binding one antigen or multispecific, i.e., capable of binding two or more antigens.
  • DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to a DVD Ig.
  • Each half of a DVD Ig comprises a heavy chain DVD polypeptide, and a light chain DVD polypeptide, and two antigen binding sites.
  • Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site.
  • DVD binding proteins and methods of making DVD binding proteins are disclosed in U.S. patent application Ser. No. 11/507,050 and incorporated herein by reference.
  • DVD binding protein comprising binding proteins capable of binding to troponin I.
  • the DVD binding protein is capable of binding troponin I and a second target.
  • the present invention also encompasses triple-variable domain (TVD) binding proteins in which the antibody is capable of binding troponin I as well as two additional targets (i.e., a second and third target).
  • TVD triple-variable domain
  • an anti-idiotypic (anti-Id) antibody specific for such binding proteins of the invention.
  • An anti-Id antibody is an antibody, which recognizes unique determinants generally associated with the antigen-binding region of another antibody.
  • the anti-Id can be prepared by immunizing an animal with the binding protein (e.g., antibody of interest) or a CDR containing region thereof. The immunized animal will recognize, and respond to the idiotypic determinants of the immunizing antibody and produce an anti-Id antibody.
  • the anti-Id antibody may also be used as an “immunogen” to induce an immune response in yet another animal, producing a so-called anti-anti-Id antibody.
  • a protein of interest may be expressed using a library of host cells genetically engineered to express various glycosylation enzymes, such that member host cells of the library produce the protein of interest with variant glycosylation patterns.
  • a practitioner may then select and isolate the protein of interest with particular novel glycosylation patterns.
  • the protein having a particularly selected novel glycosylation pattern exhibits improved or altered biological properties.
  • the anti-troponin I antibodies, or portions thereof, of the invention can be used to detect troponin I (e.g., in a biological sample such as serum, whole blood, CSF, brain tissue or plasma), using a conventional immunoassay competitive or non-competitive assay such as, for example, an enzyme linked immunosorbent assay (ELISA), a radioimmunoassay (RIA), an immunometric assay or tissue immunohistochemistry.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • tissue immunohistochemistry e.g., tissue immunohistochemistry.
  • the invention therefore provides a method for detecting troponin I in a biological sample comprising contacting a biological sample with an antibody, or antibody portion, of the invention and detecting either the antibody (or antibody portion) bound to troponin I or unbound antibody (or antibody portion), to thereby detect troponin I in the biological sample.
  • the antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody.
  • Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetyl-cholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3 H, 14 C, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I, 177 Lu, 166 Ho, or 153 Sm.
  • troponin I can be assayed in biological fluids by a competition immunoassay utilizing recombinant troponin I standards labeled with a detectable substance and an unlabeled anti-troponin I antibody.
  • the biological sample, the labeled recombinant troponin I standards and the anti-troponin I antibody are combined, and the amount of labeled recombinant troponin I standard bound to the unlabeled antibody is determined.
  • the amount of troponin I in the biological sample is inversely proportional to the amount of labeled recombinant troponin I standard bound to the anti-troponin I antibody.
  • one or more of the antibodies of the present invention are coated on a solid phase (or are in a liquid phase).
  • the test or biological sample e.g., serum, plasma, urine, etc.
  • the direct method comprises simply detecting presence of the complex itself and thus presence of the antigens.
  • a “fragment” or “portion” of an antibody is defined as a subunit of the antibody which reacts in the same manner, functionally, as the full antibody with respect to binding properties.
  • a conjugate is added to the bound antigen.
  • the conjugate comprises a second antibody, which binds to the bound antigen, attached to a signal-generating compound or label. Should the second antibody bind to the bound antigen, the signal-generating compound generates a measurable signal. Such signal then indicates presence of the antigen (i.e., troponin I) in the test sample.
  • solid phases used in diagnostic immunoassays are porous and non-porous materials, latex particles, magnetic particles, microparticles (see U.S. Pat. No. 5,705,330), beads, membranes, microtiter wells and plastic tubes.
  • the choice of solid phase material and method of labeling the antigen or antibody present in the conjugate, if desired, are determined based upon desired assay format performance characteristics.
  • the conjugate (or indicator reagent) will comprise an antibody (or perhaps anti-antibody, depending upon the assay), attached to a signal-generating compound or label.
  • This signal-generating compound or “label” is itself detectable or may be reacted with one or more additional compounds to generate a detectable product.
  • signal-generating compounds include chromogens, radioisotopes (e.g., 125I, 131I, 32P, 3H, 35S and 14C), chemiluminescent compounds (e.g., acridinium), particles (visible or fluorescent), nucleic acids, complexing agents, or catalysts such as enzymes (e.g., alkaline phosphatase, acid phosphatase, horseradish peroxidase, beta-galactosidase and ribonuclease).
  • radioisotopes e.g., 125I, 131I, 32P, 3H, 35S and 14C
  • chemiluminescent compounds e.g., acridinium
  • particles visible or fluorescent
  • nucleic acids e.g., alkaline phosphatase, acid phosphatase, horseradish peroxidase, beta-galactosidase and ribonuclease.
  • enzymes
  • chromo-, fluro-, or lumo-genic substrate results in generation of a detectable signal.
  • detection systems such as time-resolved fluorescence, internal-reflection fluorescence, amplification (e.g., polymerase chain reaction) and Raman spectroscopy are also useful.
  • biological fluids which may be tested by the above immunoassays include plasma, urine, whole blood, dried whole blood, serum, cerebrospinal fluid, saliva, tears, nasal washes or aqueous extracts of tissues and cells.
  • the initial capture antibody (for detecting troponin I antigens) used in the immunoassay may be covalently or non-covalently (e.g., ionic, hydrophobic, etc.) attached to the solid phase.
  • Linking agents for covalent attachment are known in the art and may be part of the solid phase or derivatized to it prior to coating.
  • the assays and kits of the present invention optionally can be adapted or optimized for point of care assay systems, including Abbott's Point of Care (i-STATTM) electrochemical immunoassay system.
  • i-STATTM Abbott's Point of Care
  • Immunosensors and methods of manufacturing and operating them in single-use test devices are described, for example in U.S. Pat. No. 5,063,081 and published U.S. Patent Application Nos. 20030170881, 20040018577, 20050054078, and 20060160164 (incorporated by reference herein for their teachings regarding same).
  • any of the exemplary formats herein and any assay or kit according to the invention can be adapted or optimized for use in automated and semi-automated systems (including those in which there is a solid phase comprising a microparticle), as described, e.g., in U.S. Pat. Nos. 5,089,424 and 5,006,309, and as, e.g., commercially marketed by Abbott Laboratories (Abbott Park, Ill.) including but not limited to Abbott's ARCHITECT®, AXSYM, IMX, PRISM, and Quantum II platforms, as well as other platforms.
  • assay formats which may be used for purposes of the present invention include, for example, a rapid test, a Western blot, as well as the use of paramagnetic particles in, for example, an ARCHITECT® assay (see Frank Quinn, The Immunoassay Handbook, Second edition, edited by David Wild, pages 363-367, 2001, herein incorporated in its entirety by reference). Such formats are known to those of ordinary skill in the art.
  • kits may also comprise one container such as a vial, bottle or strip. These kits may also contain vials or containers of other reagents needed for performing the assay, such as washing, processing and indicator reagents.
  • the invention also provides pharmaceutical compositions comprising an antibody, or antigen-binding portion thereof, of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions comprising antibodies of the invention are for use in, but not limited to, diagnosing, detecting, or monitoring a disorder, in preventing, treating, managing, or ameliorating of a disorder or one or more symptoms thereof, and/or in research.
  • a composition comprises one or more antibodies of the invention.
  • the pharmaceutical composition comprises one or more antibodies of the invention and one or more prophylactic or therapeutic agents other than antibodies of the invention for treating a disorder in which troponin I activity is detrimental.
  • the composition may further comprise of a carrier, diluent or excipient.
  • the antibodies and antibody-portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject.
  • the pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion.
  • Various delivery systems are known and can be used to administer one or more antibodies of the invention or the combination of one or more antibodies of the invention and a prophylactic agent or therapeutic agent useful for preventing, managing treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • a prophylactic agent or therapeutic agent useful for preventing, managing treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor-mediated endocytosis (see, e.
  • Methods of administering a prophylactic or therapeutic agent of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural administration, intratumoral administration, and mucosal administration (e.g., intranasal and oral routes).
  • parenteral administration e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous
  • epidural administration e.g., intratumoral administration
  • mucosal administration e.g., intranasal and oral routes
  • pulmonary administration can be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Pat. Nos. 6,019,968, 5,985,320, 5,985,309, 5,934,272, 5,874,064, 5,855,913, 5,290,540, and 4,880,078; and
  • an antibody of the invention, combination therapy, or a composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.).
  • prophylactic or therapeutic agents of the invention are administered intramuscularly, intravenously, intratumorally, orally, intranasally, pulmonary, or subcutaneously.
  • the prophylactic or therapeutic agents may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the prophylactic or therapeutic agents of the invention may be desirable to administer the prophylactic or therapeutic agents of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous or non-porous material, including membranes and matrices, such as sialastic membranes, polymers, fibrous matrices (e.g., Tissuel®), or collagen matrices.
  • an effective amount of one or more antibodies of the invention antagonists is administered locally to the affected area to a subject to prevent, treat, manage, and/or ameliorate a disorder or a symptom thereof.
  • an effective amount of one or more antibodies of the invention is administered locally to the affected area in combination with an effective amount of one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than an antibody of the invention of a subject to prevent, treat, manage, and/or ameliorate a disorder or one or more symptoms thereof.
  • therapies e.g., one or more prophylactic or therapeutic agents
  • the prophylactic or therapeutic agent can be delivered in a controlled release or sustained release system.
  • a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574).
  • polymeric materials can be used to achieve controlled or sustained release of the therapies of the invention (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla.
  • polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly (ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters.
  • the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable.
  • a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more therapeutic agents of the invention. See, e.g., U.S. Pat. No. 4,526,938, International Appln. Publication No. WO 91/05548, International Appln. Publication No.
  • the antibodies of the invention or antigen binding portion thereof can be used alone or in combination with one or more additional agents, e.g., a therapeutic agent (for example, a small molecule or biologic), said additional agent being selected by the skilled artisan for its intended purpose.
  • additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent that affects the viscosity of the composition.
  • the combinations which are to be included within this invention are those combinations useful for their intended purpose.
  • the agents set forth below are illustrative for purposes and not intended to be limiting.
  • the combinations, which are part of this invention can be the antibodies of the present invention and at least one additional agent selected from the lists below.
  • the combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
  • compositions of the invention may include a “therapeutically effective amount” or a “prophylactically effective amount” of an antibody or antibody portion of the invention.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the antibody or antibody portion may be determined by a person skilled in the art and may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody, or antibody portion, are outweighed by the therapeutically beneficial effects.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • TnI 19C7 messenger RNA was isolated from subcloned anti-TnI 19C7-144 hybridoma cells.
  • Hybridoma cell line TnI 19C7 is described in U.S. Patent Application Publication No. US2006/0018897.
  • TnI 19C7 mRNA was utilized in a reverse transcriptase-polymerase chain reaction using a mouse Ig primer set kit purchased from Novagen (Novagen (which is an affiliate of Merck KGaA, Darmstadt, Germany), Cat No. 69831-3) with immunoglobulin gene specific primers contained in the kit.
  • the resulting PCR products were sequenced and thus the immunoglobulin variable heavy and variable light chain genes were identified (see FIG. 2 ).
  • a yeast display system was used to express unmutated or wild-type anti-TnI proteins (described herein infra) and a library of anti-TnI proteins on the yeast surface as a fusion to the yeast protein AGA2.
  • a yeast display vector called pYD (Invitrogen, Carlsbad, Calif.) was used as it allows for cloning of the anti-TnI gene at the C-terminus of the AGA2 gene, a yeast mating factor (See, Boder and Wittrup, Nature Biotechnology, 15:553-557 (June 1997)).
  • Other critical features of the pYD vector include a galactose inducible promoter and an epitope tag, V5, on the C-terminus of the inserted anti-TnI gene (see FIG. 12 ).
  • the yeast display platform utilizes an antibody format known as the single-chain variable fragment.
  • the variable heavy domain is connected to the variable light domain through a flexible linker (variable heavy domain—Linker GPAKELTPLKEAKVS (SEQ ID NO:58)—variable light domain).
  • PCR single overlap extension was used to combine the variable heavy (VH) and the variable light genes (VL) for the TnI 19C7 scFv construct ( FIG. 2 , and SEQ ID NOs:54 and 55).
  • the TnI 19C7 scFv DNA was cloned into the yeast display vector pYD41 using vector restriction sites SfiI and XhoI.
  • the pYD41-TnI 19C7scFv vector was transformed into DH5 ⁇ E. coli. Plasmid DNA was then isolated from the E. coli and the TnI 19C7 scFv insert was sequenced to ensure the scFv was cloned in frame with the AGA2 protein.
  • the cloning site for the scFv into the yeast display vector pYD41 is in an ORF that includes the following genes: AGA2-tether linker 41-X press epitope tag-TnI 19C7 variable heavy chain-Linker 40-TnI 19C7 variable light chain-V5 epitope tag—Six His tag (SEQ ID NO: 29).
  • the yeast strain EBY100 is a tryptophan auxotroph and the pYD41 vector encodes for tryptophan as the system's selectable marker.
  • Yeast display plasmid pYD41-TnI 19C7 scFv
  • Gietz and Schiestl Method See Schiestl and Gietz, Current Genetics, 16(5-6):339-46 (December 1989)
  • Dilutions of the transformation reaction were plated on selective glucose plates (2% glucose (0.67% yeast nitrogen base, 0.105% HSM-trp-ura, 1.8% bacterial agar, 18.2% sorbitol, 0.86% NaH 2 PO 4 H 2 O, 1.02% Na 2 HPO 4 7H 2 O)) and incubated at 30° C. for 48-72 hours.
  • Selective glucose media was inoculated with individual colonies and grown shaking at 30° C.
  • scTnI-C-2 is a linked, single-chain TnI (28-110aa)-linker-TnC (1-160aa) from Spectral Diagnostics, Toronto, Canada.
  • ScTnI-C-2 is abbreviated as “scTnI-C” for purposes of the present discussion.
  • GAM:PE goat anti mouse-phycoerythrin
  • FIG. 3A The flow cytometry histograms illustrate the full-length expression of TnI 19C7 scFv as detected by anti-V5 and the ability of TnI 19C7 scFv to bind to scTnI-C-2.
  • TnI 19C7 scFv and TnI 19C7 variants on yeast were measured by incubating 0.05OD yeast (1 ⁇ 10 6 cells) with 50 nM scTnI-C-2 for 30-60 minutes at room temperature. Cells were then washed twice with blocking buffer containing phosphate buffered saline pH 6.8 with 2% bovine serum albumin and 0.2% Standapol ES-1 (PBS/BSA/Standapol) and incubated at room temperature for varying amounts of time (0, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4.25 hr, 25.5 hr, 50 hr 75 hr and 144 hr (see FIG. 4 ).
  • TnI 19C7 scFv unmutated or wildtype (“wt”)
  • wt wildtype
  • TnI 19C7 scFv and TnI 19C7 variants on yeast were measured by incubating 0.05 OD yeast (1 ⁇ 10 6 cells) with varying concentrations of scTnI-C-2 for 45-60 minutes at room temperature.
  • Blocking buffer containing phosphate buffered saline pH 6.8 with 2% bovine serum albumin and 0.2% Standapol ES-1 (PBS/BSA/Standapol) was used for washes and reagent dilutions. Cells were then washed twice and incubated for 30 min with anti-TnI mAb, 8E10. Cells were washed again and incubated with goat anti-mouse phycoerythrin for 30 min.
  • FIG. 5 shows the KD data plotted as normalized mean fluorescence units (“MFU”) versus concentration scTn-I-C-2 (in Molarity).
  • MFU normalized mean fluorescence units
  • concentration scTn-I-C-2 in Molarity
  • CDR loop lengths and numbering were defined using Kabat and Oxford's Molecular AbM modeling nomenclature. Individual libraries were composed such that random mutations are incorporated at each amino acid position of the CDR for a single library. A total of six libraries were generated corresponding to one library per each CDR.
  • Libraries were generated by combining SfiI/XhoI digested. pYD41 vector and PCR products with chemically competent EBY100 yeast (see FIG. 7 ).
  • Two PCR products were generated for each CDR library to allow for PCR sorting and homologous recombination into yeast.
  • One PCR product used a primer that was designed, such that for the entire length of the CDR, a 70% wild type to 30% other nucleotide ratio was used in the primer synthesis. This product was called the spiked (sp) product (see FIG. 7 ).
  • the second PCR product was designed to include the remaining portion of the scFv gene. The two PCR products were combined and used to generate a single-chain variable fragment or a scFv product.
  • Digested vector (1 ug) and the scFv PCR products (5 ug) were combined with EBY100 yeast (5.2e8-6.4e8 cells) and transformed using electroporation.
  • the scFv PCR product and the pYD41 digested vector cyclize during transformation due to homologous recombination facilitated by the nucleotide overlap and the mechanism of yeast endogenous gap repair.
  • Libraries were grown at 30° C. for 48-72 hours in selective glucose media and passed again in selective glucose media prior to induction of protein expression for library sorting.
  • TnI 19C7 libraries were sorted based on an off-rate sorting strategy.
  • TnI 19C7 CDR mutagenic libraries were induced in galactose expression media at 20° C. for 18-24 hours.
  • TnI 19C7 scFv and TnI 19C7 libraries on yeast were incubated with 25-50 nM scTnI-C-2 for 10-15 minutes at room temperature. Cells were then washed twice with blocking buffer containing phosphate buffered saline pH 6.8 with 2% bovine serum albumin and 0.2% Standapol ES-1 (PBS/BSA/Standapol) and incubated at room temperature for 8 min. Yeast cells were transferred to ice to halt the reaction.
  • Sort gates were set based on unmutated TnI 19C7 binding at 8-9 min with a gate set to sort full-length TnI binding clones. Each sort collected the top 0.1-0.5% of the TnI binding population. Sorted cells were grown in selective glucose media and grown 18-24 hours at 30° C. Sort 1 cells were induced and sorting was repeated for two or three additional rounds.
  • sorted cells were plated onto selective glucose plates and placed at 30° C. for 72 hours. Individual yeast colonies from these libraries were inoculated in selective glucose media, cryopreserved and induced in selective galactose media. Individual colonies were then characterized and ranked in an off-rate assay. TnI 19C7 AM4 was isolated and identified from this sorting output.
  • Clones that were characterized for off-rate from each master CDR library or the total master CDR library output were used to construct scFv genes containing different pairings of the individual mutations. This approach enabled determination of whether the binding properties were further enhanced upon combining individual mutations.
  • Combinatorial clones containing various mutations in each CDR region were constructed by PCR amplification and combined using routine techniques known to those skilled in the art.
  • Combinatorial mutant libraries were transformed into yeast as described above and sorted two times using off-rate and KD selection pressures. For KD selection, 100 pM (round 1) and 50 pM (round 2) scTnI-C were used in the KD experiment as described above ( FIG. 5 ).
  • sorting was conducted as described above with incubation times following washing away of antigen of 3 hr 40 min (round 1) and 4 hrs 25 min (round 2).
  • Sort gates were set based on unmutated TnI 19C7 binding for each condition with a gate set to sort full-length TnI binding clones. Each sort collected the top 0.1% of the TnI binding population. Sorted cells were grown in selective glucose media for 18-24 hours at 30° C. Sort 1 cells were induced and sorting was repeated for one additional round.
  • sorted cells were plated onto selective glucose plates and placed at 30° C. for 72 hours. Individual yeast colonies from these libraries were inoculated in selective glucose media, cryopreserved and induced in selective galactose media. Individual colonies were then characterized and ranked in an off-rate assay. TnI 19C7 AM1, AM2, and AM3 were isolated and identified from this combinatorial library.
  • FIG. 9 shows the scFv KD values determined for four selected clones.
  • the TnI 19C7 AM1 clone exhibited the most improved binding at 0.36 nM compared to the wild-type TnI 19C7 antibody 1.7 nM.
  • Selected TnI 19C7 scFv variants were sequenced to determine the amino acid mutations being expressed.
  • plasmid DNA was isolated from yeast suspension cultures using a yeast mini-prep kit (Cat No. D2001, Zymo Research Orange, Calif.).
  • yeast mini-prep kit Cat No. D2001, Zymo Research Orange, Calif.
  • plasmid from the yeast mini-prep kit was transformed into DH5 ⁇ E. coli, and then purified from culture using E. coli mini-prep kits (Qiagen).
  • the CDR H3 was the only CDR with mutations that were different than the combinatorial consensus set of sequences. Specifically the mutations were Ala96Phe, Tyr99Ser, Trp100aAla, and Tyr102Asp.
  • CDR L1 The consensus sequence data for CDR L1 indicated a preference for threonine at position 25, lysine at position 27, asparagine at position 28, valine at position 29, and histidine at position 34.
  • each clone has unique or no mutations with only TnI 19C7 AM1 and AM2 sharing only the Ser54Arg mutation.
  • TnI 19C7 variants were converted to chimeric mouse-mouse IgG 2 ⁇ /mouse kappa and/or mouse-human IgG 1 /human kappa antibodies through cloning of the TnI 19C7 variable domains into the transient expression vector system called pBOS (Abbott Bioresearch Center, Worcester, Mass.). More specifically, PCR was used to amplify the variable heavy and variable light chain genes with restriction sites for cloning into separate pBOS vectors (Mizushima and Nagata, Nucleic Acids Research, 18:5322 (1990)). The variable heavy and variable light genes were ligated in digested and dephosphorylated vector and transformed into DH5 ⁇ E. coli.
  • Plasmid DNA was purified from E. coli and transfected into 293H cells using PEI (1 mg/ml). Transient antibody was expressed for the following TnI 19C7 variants: TnI 19C7 wt, AM1, AM2, AM3 and AM4.
  • variable heavy chain and variable light genes were ligated in frame to the human constant genes in pBV and pJV plasmids (Abbott Bioresearch Center, Worcester, Mass.), respectively, using the restriction enzymes SrfI/NotI. Ligation reactions were transformed into DH5 ⁇ E. coli and plasmid DNA was subsequently isolated from individual colonies.
  • the pBV-TnI 19C7 mouse variable heavy-human IgG1 and pJV-TnI 19C7 mouse variable light-human kappa were sequenced at the cloning sites.
  • the second cloning step involved combining the heavy chain IgG 1 genes and the light chain kappa genes into a single stable cell line vector.
  • the pBV-TnI 19C7 AM 1 human IgG1 and pJV-TnI 19C7 AM1 human kappa vectors were digested with AscI/PacI.
  • the VL-human kappa constant and the VH-human IgG1 constant DNA fragments were gel purified and ligated to produce the stable cell line vector called pBJ-TnI 19C7 AM1.
  • the pBJ-TnI 19C7 AM1 human heavy/light chimeric plasmid was transformed into CHO cells using a lipofectamine (Invitrogen) protocol.
  • Stable cell lines were sub-cloned from initial transformation.
  • a stable CHO cell line has been developed for the clone AM1 (also referred to as “TnI 19C7AM1 hG1kCHO204”) and was deposited with the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209 on Feb. 11, 2009 and received deposit designation PTA-9816.
  • Troponin I clone 19C7 wild type (full mouse construct) and affinity matured (human constant region) antibodies were evaluated for relative affinity in a microtiter enzyme immunoassay.
  • 96-well assay plates (NUNC Corporation, Rochester, N.Y.) were coated by adding 100 uL/well of a 2 ug/mL solution of either sheep anti-mouse IgG Fc ⁇ specific antibody (Jackson ImmunoResearch, West Grove, Pa.) or donkey antihuman IgG Fc ⁇ fragment specific antibody (Jackson ImmunoResearch). Both antibodies were diluted in phosphate buffered saline (PBS, Abbott Laboratories, Abbott Park, Ill.). The assay plates were incubated overnight at 15-30 deg C.
  • PBS phosphate buffered saline
  • BSA solution bovine serum albumin [Abbott Laboratories] diluted in PBS
  • the BSA solution was incubated in the assay wells for 30 minutes at 15-30 deg C., removed and the assay wells washed by adding 300 uL/well distilled water (dH2O, Abbott Laboratories) and aspirating for three wash cycles. Next, 100 uL/well test samples were added. Test samples were prepared by creating an initial 2 ug/mL solution (in BSA solution) of each antibody, followed by log 3 dilutions, in BSA solution. The test samples were incubated for 2-3 hours at 15-30 deg C.
  • test antigen solutions were added to each assay well.
  • the test antigen solutions were created by first preparing a 1000 ng/mL solution of scTnI-C-2 (aa 28-100 of cardiac troponin I linked to full length cardiac troponin C, Spectral Diagnostics) in BSA solution, followed by log 2 dilutions.
  • the antigen solutions were incubated in the assay wells for 10 minutes at 15-30 deg C. and then removed by slapping out the solutions.
  • the assay plates were then washed with dH2O as previously described.
  • substrate solution was prepared by dissolving 1 OPD tablet per 10 mL OPD diluent (o-phenylenediamine, both Abbott Laboratories). 100 uL/well of the prepared substrate solution was added to the assay plates, incubated for about 4-5 minutes and then the reaction quenched by adding 100 uL/well 1N sulfuric acid (Abbott Laboratories). The resulting signal was read at 492 nm using an optical fluorometer. Results from the experiment were plotted using kaleidagraph software. The Ag 50 value (the concentration of antigen at 50% of maximal binding) was determined and used to compare the antibodies for relative affinity to the tested antigen.
  • Troponin I clone 19C7 wild type (full mouse construct) and affinity matured (human constant region) antibodies were evaluated for relative affinity on the ARCHITECT® immunoassay analyzer (Abbott Laboratories). The assay was fully automated, and the analyzer performs all steps. Magnetic microparticles coated with a mouse anti-troponin I antibody (Abbott Laboratories, Abbott Park, Ill.) were mixed with varying levels of scTnI-C-2 (aa 28-100 of cardiac troponin I linked to full length cardiac troponin C (Spectral Diagnostics) and incubated for 18 minutes at 15-30 deg C. During this time, the microparticle coated antibody bound the scTnI-C-2.
  • microparticles were then attracted to a magnet, the remaining assay solution was aspirated, and the particles washed with assay diluent (Abbott Laboratories, Abbott Park, Ill.).
  • assay diluent Abbott Laboratories, Abbott Park, Ill.
  • the wild type or affinity matured 19C7 antibodies all of which were labeled with acridinium (Abbott Laboratories, Abbott Park, Ill.) were added to the microparticles and incubated for 4 minutes at 15-30 deg C.
  • the microparticles were attracted to a magnet, the remaining assay solution was aspirated, and the particles were washed with assay diluent.
  • Signal relative light units
  • pre-trigger and trigger solutions both Abbott Laboratories.
  • Signal ratios were calculated and used to compare antibodies. As can be established based upon the results shown in FIG. 11 , AM1 antibody gave a better signal than wild-type antibody.

Abstract

The subject invention relates to antibodies to troponin I as well as methods of use thereof. In particular, such antibodies may be used to detect Troponin I in a patient and may also be used in the diagnosis of, for example, a myocardial infarction or acute coronary syndrome.

Description

CROSS-REFERENCE TO RELATED APPLICATION(S)
This is a reissue of U.S. patent application Ser. No. 12/391,937, filed on Feb. 24, 2009, now U.S. Pat. No. 8,030,026.
BACKGROUND OF THE INVENTION
1. Field of the Invention
The subject invention relates to antibodies to troponin I as well as methods of use thereof.
2. Background Information
Troponin I is a muscle protein which may be used in the determination of myocardial damage subsequent to or during, for example, a myocardial infarction. In particular, troponin I is one of three subunits of the troponin complex which is located on the thin filament of the muscle contractile apparatus. This complex has a primary role in controlling the process of muscle contraction. The other two subunits (i.e., T and C) are also immobilized on the thin myofilaments with troponin I in cardiac as well as skeletal muscle tissue.
Assays have been described which measure cardiac troponin I in human serum. For example, a radioassay has been used for this purpose (Cummins et al., Am Heart Journal 113:1333-1344 (1987). However, the assay utilized polyclonal antibodies having significant cross-reactivity to skeletal forms of troponin I. Further, a sandwich assay has been utilized which uses two different monoclonal antibodies (Bodar et al., Clinical Chemistry 38:2203-2214 (1992); see also U.S. Pat. No. 7,285,418). Unfortunately, such assays have a very high degree of imprecision. Thus, the need certainly exists for immunoassays that are highly specific for and sensitive to troponin I. These immunoassays must also utilize antibodies which do not possess cross-reactivity to troponin I found in skeletal tissue. In particular, such immunoassays are needed so that appropriate therapy can be utilized by the treating physician thereby giving the affected patient the best possible prognosis.
All patents and publications referred to herein are hereby incorporated in their entirety by reference.
SUMMARY OF THE INVENTION
The present invention pertains to binding proteins, particularly antibodies, capable of binding to cardiac troponin I. In particular, these antibodies bind to one or more epitopes of troponin I. Further, the present invention also provides methods of producing and using these binding proteins or portions thereof, for example, in diagnostic assays.
In particular, the present invention encompasses a Chinese Hamster Ovary (CHO) cell line, referred to as TnI 19C7 AM1 hG1 CHO 204, designated by American Type Culture Collection (ATCC) deposit number PTA-9816 as well as the recombinant antibody produced by this cell line.
Additionally, the present invention includes an isolated binding protein comprising an antigen-binding domain which binds to Troponin I, said antigen-binding domain comprising at least one complementarity determining region (CDR) comprising an amino acid sequence selected from the group consisting of: GYTFTDYNLH (SEQ ID NO:52), YIYPYNGITGYNQKFKS (SEQ ID NO:53), DAYDYDLTD (SEQ ID NO:54), RTSKNVGTNIH (SEQ ID NO:55), YASERLP (SEQ ID NO:56) and QQSNNWPYT (SEQ ID NO:57). The binding protein of the present invention may include, for example, at least 3 of these CDRs. Further, this binding protein may also comprise a human acceptor framework or scaffold. This binding protein may be selected from the group consisting of, for example, an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a humanized antibody, a Fab, a Fab′, a F(ab′)2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, or a functionally active epitope-binding fragment of any one of these entities.
The present invention also encompasses an isolated nucleic acid molecule encoding a binding protein, wherein the amino acid sequence of the variable heavy chain of the binding protein has at least 70% identity to SEQ ID NO.: 25 (see FIG. 12). This molecule may also comprise a variable light chain having at least 70% identity to SEQ ID NO.: 28 (see FIG. 12).
Furthermore, the present invention includes an isolated nucleic acid molecule encoding a binding protein, wherein the amino acid sequence of the variable heavy chain of said binding protein is SEQ ID NO.:25.
Additionally, the present invention includes an isolated nucleic acid molecule encoding a binding protein, wherein the amino acid sequence of the variable light chain of said binding protein is SEQ ID NO.: 28. The molecule may further comprise an isolated nucleic acid molecule encoding a variable heavy chain, wherein the amino acid sequence of the heavy chain is SEQ ID NO.: 25.
The present invention also includes a vector comprising one or more of the nucleic acid molecules described above, attached to a regulatory element (e.g., a promoter) as well as a host cell comprising this vector.
Moreover, the present invention includes a method of producing any of the binding proteins described above, capable of binding to Troponin I, which method comprises culturing the host cell, described above, for a time and under conditions sufficient to produce the binding protein of interest. The invention also includes the binding protein produced by this method.
Furthermore, the present invention encompasses a pharmaceutical composition comprising any one or more of the binding proteins described above and a pharmaceutically acceptable carrier.
Also, the present invention includes a method of detecting Troponin I antigen in a test sample. This method comprises the steps of: contacting the test sample with an antibody which binds to Troponin I and comprises SEQ ID NO:25 for a time and under conditions sufficient for the formation of antibody/antigen complexes; and detecting presence of the complexes, presence of the complexes indicating presence of Troponin I antigen in said test sample. The antibody may further comprise SEQ ID NO:28. The antibody may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
The present invention also includes a method of detecting Troponin I antigen in a test sample comprising the steps of: contacting the test sample with a first antibody which binds to Troponin I and comprises SEQ ID NO:25 for a time and under conditions sufficient for the formation of first antibody/antigen complexes; adding a conjugate to the first antibody/antigen complexes, wherein said conjugate comprises a second antibody attached to a signal generating compound capable of generating a detectable signal, for a time and under conditions sufficient to form first antibody/antigen/second antibody complexes; and detecting presence of a signal generating by the signal generating compound, presence of the signal indicating presence of Troponin I antigen in said test sample. The first antibody may further comprise SEQ ID NO:28 and may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
Also, the present invention includes a method of detecting Troponin I antigen in a test sample comprising the steps of contacting Troponin I antigen with an antibody to Troponin I for a time and under conditions sufficient to form Troponin I antigen/antibody complexes, wherein the antibody comprises SEQ ID NO:25 and is labeled with a signal-generating compound capable of generating a detectable signal; adding the test sample to said Troponin I antigen/antibody complexes for a time and under conditions sufficient to form Troponin I antigen/antibody/Troponin I test sample antigen complexes; and detecting presence of a signal generating by the signal generating compound, presence of the signal indicating presence of Troponin I antigen in the test sample. Again, the antibody may further comprise SEQ ID NO:28 and may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
The present invention also encompasses another method of detecting Troponin I antigen in a test sample. This method comprises the steps of: contacting the test sample with 1) a Troponin I reference antigen, wherein the antigen is attached to a signal generating compound capable of generating a detectable signal and 2) an antibody to Troponin I antigen wherein the antibody comprises SEQ ID NO:25, for a time and under conditions sufficient to form Troponin I reference antigen/antibody complexes; and detecting a signal generated by the signal generating compound, wherein the amount of Troponin I antigen detected in the test sample is inversely proportional to the amount of Troponin I reference antigen bound to the antibody. Again, the antibody may further comprise SEQ ID NO:28 and may be produced by a Chinese Hamster Ovary cell line having ATCC deposit designation PTA-9816.
In addition, the present invention includes pharmaceutical composition comprising any one or more of the binding proteins described above and a pharmaceutically acceptable carrier.
The present invention also encompasses a method of diagnosing acute coronary syndrome or myocardial infarction in a patient suspected of having one of these conditions. This method comprises the steps of: isolating a biological sample from the patient; contacting the biological sample with an antibody which binds to Troponin I and comprises SEQ ID NO:25, for a time and under conditions sufficient for formation of Troponin I antigen/antibody complexes; detecting presence of the Troponin I antigen/antibody complexes; dissociating the Troponin I antigen present in the complexes from the antibody present in said complexes; and measuring the amount of dissociated Troponin I antigen, wherein an amount of Troponin I antigen greater than approximately 1-5 times the Troponin I value of the 99th percentile of a normal population indicates a diagnosis of acute coronary syndrome or myocardial infarction in the patient.
The present invention includes an additional method method of diagnosing acute coronary syndrome or myocardial infarction in a patient suspected of having one of these conditions. This method comprises the steps of: isolating a biological sample from the patient; contacting the biological sample with a first antibody which binds to Troponin I and comprises SEQ ID NO:25, for a time and under conditions sufficient for the formation of Troponin I antigen/antibody complexes; adding a conjugate to the resulting Troponin I antigen/antibody complexes for a time and under conditions sufficient to allow the conjugate to bind to the bound Troponin I antigen, wherein the conjugate comprises a second antibody attached to a signal generating compound capable of generating a detectable signal; detecting the presence of Troponin I antigen which may be present in said biological sample by detecting a signal generated by said signal generating compound; and measuring the amount of Troponin I antigen present in the test sample by measuring the intensity of the signal, an amount of Troponin I antigen greater than approximately 1-5 times the value of the 99th percentile of a normal population indicating a diagnosis of acute coronary syndrome or myocardial infarction in the patient.
The present invention also includes a kit comprising any one or more of the monoclonal antibodies or binding proteins described above and, if needed, instructions describing the manner in which to use this kit.
Additionally, the present invention includes an isolated binding protein which comprises an antigen-binding domain, wherein the antigen-binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of:
CDR-VH1. X1-X2-X3-X4-X5-X6-X7-X8-X9-X10 (SEQ ID NO:63), wherein:
    • X1 is G;
    • X2 is Y;
    • X3 is T or S;
    • X4 is F;
    • X5 is T;
    • X6 is D;
    • X7 is Y;
    • X8 is N;
    • X9 is I or L; and
    • X10 is H.
CDR-VH2. X1X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17 (SEQ ID NO:64), wherein:
    • X1 is Y;
    • X2 is I;
    • X3 is Y;
    • X4 is P;
    • X5 is Y;
    • X6 is N;
    • X7 is G;
    • X8 IS I;
    • X9 is T;
    • X10 is G;
    • X11 is Y;
    • X12 is N;
    • X13 is Q;
    • X14 is K;
    • X15 is F;
    • X16 is K; and
    • X17 is S.
CDR-VH3. X1-X2-X3-X4-X5-X6-X7-X8-X9-X10 (SEQ ID NO:65), wherein:
    • X1 is D;
    • X2 is A or F;
    • X3 is Y;
    • X4 is D;
    • X5 is Y or S;
    • X6 is D;
    • X7 is W, Y or A;
    • X8 is L;
    • X9 is A or T; and
    • X10 is Y or D.
CDR-VL1. X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11 (SEQ ID NO:66), wherein:
    • X1 is R;
    • X2 is A or T;
    • X3 is S;
    • X4 is Q or K;
    • X5 is S or N;
    • X6 is I or V;
    • X7 is G;
    • X8 is T;
    • X9 is N;
    • X10 is I; and
    • X11 is Y or H.
CDR-VL2. X1-X2-X3-X4-X5-X6-X7 (SEQ ID NO:67), wherein:
    • X1 is Y;
    • X2 is A or G;
    • X3 is S or T;
    • X4 is E;
    • X5 is S or R;
    • X6 is I, L or V; and
    • X7 is S, P or F, and
CDR-VL3. X1-X2-X3-X4-X5-X6-X7-X8-X9 (SEQ ID NO:68), wherein:
    • X1 is Q;
    • X2 is Q;
    • X3 is S;
    • X4 is N;
    • X5 is N;
    • X6 is W;
    • X2 is P;
    • X8 is Y; and
    • X9 is T.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 is a flow chart showing the steps used to identify and create antibodies that have improved affinity for troponin I.
FIG. 2 is the nucleotide description (SEQ ID NO: 109 and SEQ ID NO: 110) of the wild-type TnI 19C7 single-chain variable fragment (“scFv”).
FIG. 3 shows that yeast expressing full-length TnI 19C7 single-chain variable fragment (scFv) bind to single chain troponin I (28-110aa)-linker-troponin C known as scTnI-C-2 (Spectral diagnostics, RP-3700). More specifically, this figure shows that TnI 19C7 scFv expressing yeast were incubated with scTnI-C-2 or anti-V5, followed by either anti-troponin mAb and goat anti mouse-phycoerythrin (GAM:PE) (FIG. 3B) or GAM:PE respectively (FIG. 3A). The flow cytometry histograms illustrate the full-length expression of TnI 19C7 scFv as detected by anti-V5 and the ability of TnI 19C7 scFv to bind to scTnI-C-2. PE-A units (abscissa). 102, 103, 104, and 105. Count units (ordinate): 102, 103, 104, and 105.
FIG. 4 shows the TnI 19C7 scFv off-rate measurement. More specifically, yeast expressing TnI 19C7 scFv were incubated with a saturating concentration of scTnI-C-2. Cells were washed twice and at each time point, cells were transferred to ice, washed and incubated with anti-TnI mAb. After 30 minutes, cells were washed again and incubated with goat anti-mouse phycoerythrin. Again after 30 minutes, cells were washed and analyzed on the flow cytometer. A first order decay equation was used to fit the individual time points where ml was the theoretical maximum mean fluorescence units (“MFU”) at time 0, m2 was the off-rate (“koff”), m3 was the background MFU due to autofluorescence and M0, which is the time x (the x being the time that is being measured) was the time x that measurements are taken. The half-life (t1/2) of TnI 19C7 scFv binding to TnI-C-2 was calculated using: t1/2=ln 2/koff. Five times the half-life was the time used to sort the TnI 19C7 scFv CDR mutagenic libraries.
FIG. 5 shows the TnI 19C7 scFv equilibrium dissociation constant (KD) measurement. More specifically, yeast expressing TnI 19C7 scFv were incubated with varying concentrations of scTnI-C-2. Cells were washed twice with PBS pH6.8/2% BSA/0.02% Standapol ES-1 and incubated with anti-TnI mAb for 30 min Cells were washed again and incubated with goat anti-mouse phycoerythrin for 30 min. Finally, cells were washed and analyzed on the flow cytometer.
FIG. 6 is a schematic depiction that shows how degenerate oligonucleotides were designed so that primers are made such that for each CDR nucleotide residue 70% remains the wild-type residue and 30% a mix of the other three residues. Two PCR products are generated for each library a spiked (sp) PCR product and a non-spiked PCR product. The spiked and non-spiked PCR products are combined to generate an intact CDR mutagenized scFv library.
FIG. 7 is a schematic depiction that shows how the TnI 19C7 scFv library was constructed using yeast homologous recombination. More specifically, the spiked CDR PCR product and the excised yeast display vector were transformed into S. cerevisiae strain EBY100. Transformed clones were selected in tryptophan deficient glucose media.
FIG. 8 is a summary showing the PCR primers that were used to generate the scFv construct (tpVHfor through tpVL-rev), those used to generate the CDR spiked libraries (19H1spfor through pYD41rev2) and those used to generate the combination library (19FRH2for to 19FRL3) (see SEQ ID Nos:1-22). The bold and enlarged areas of the primers represent those regions in which a “70% wild-type, 30% other nucleotide mixture” was incorporated while the primers were being made. Such a “spiked” primer generated the diversity within the library.
FIG. 9 shows equilibrium dissociation constant (KD) measurements of selected TnI 19C7 scFv determined as described above in FIG. 5.
FIG. 10 shows the results of relative antibody affinity as measured as an antigen 50% (Ag50). Four TnI 19C7 clones were converted into mouse IgG2ak antibodies by cloning the variable domains onto the immunoglobulin constant domains. Antibodies were expressed in a transient HEK 294 cell system. The Ag50 is the concentration of scTnI-C at which is 50% of the maximum signal and represents the relative affinity ranking of the selected TnI 19C7 AM candidates. TnI 19C7 AM1 exemplifies the tightest relative affinity compared to the TnI 19C7 wild-type antibody.
FIG. 11 illustrates TnI 19C7 AM1's ability to bind to scTnI-C in an ARCHITECT® assay format (Abbott Laboratories, Abbott Park, Ill.). TnI 19C7 was labeled with acridinium and assayed for binding to scTnI-C using anti-TnI capture beads. (X=signal generated with given calibrator concentration of scTnI-C; X/A=ratio of calibrator X signal to calibrator A signal; RLU=Relative Light Units). TnI 19C7 AM1 exhibited better binding in this assay format for the range of calibrators compared to the wild-type TnI 19C7 antibody.
FIG. 12 illustrates the nucleotide (SEQ ID NO:23, SEQ ID NO:24 (complement), SEQ ID NO:26 and SEQ ID NO:27 (complement)) and encoded amino acid sequences of the heavy (SEQ ID NO:25) and light (SEQ ID NO:28) chains of monoclonal antibody TnI 19C7 AM1 and, in particular, of the complementarity determining regions (CDRs).
FIG. 13 illustrates the positions within the heavy and light chains of the TnI 19C7 CDRs that may be substituted with amino acids other than those shown in FIG. 12 (SEQ ID Nos: 30-49).
DETAILED DESCRIPTION OF THE INVENTION
Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. The meaning and scope of the terms should be clear; however, in the event of any latent ambiguity, definitions provided herein take precedent over any dictionary or extrinsic definition. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. In this application, the use of “or” means “and/or” unless stated otherwise. Furthermore, the use of the term “including”, as well as other forms, such as “includes” and “included”, is not limiting. Also, terms such as “element” or “component” encompass both elements and components comprising one unit and elements and components that comprise more than one subunit unless specifically stated otherwise.
Generally, nomenclatures-used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well known and commonly used in the art. The methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated. Enzymatic reactions and purification techniques are performed according to manufacturer's specifications, as commonly accomplished in the art or as described herein. The nomenclatures used in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients.
In order that the present invention may be more readily understood, select terms are defined below.
The term “polypeptide” as used herein, refers to any polymeric chain of amino acids. The terms “peptide” and “protein” are used interchangeably with the term polypeptide and also refer to a polymeric chain of amino acids. The term “polypeptide” encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence. A polypeptide may be monomeric or polymeric.
The term “isolated protein” or “isolated polypeptide” is a protein or polypeptide that by virtue of its origin or source of derivation is not associated with naturally associated components that accompany it in its native state; is substantially free of other proteins from the same species; is expressed by a cell from a different species; or does not occur in nature. Thus, a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components. A protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
The term “recovering” as used herein, refers to the process of rendering a chemical species such as a polypeptide substantially free of naturally associated components by isolation, e.g., using protein purification techniques well known in the art.
The subject invention also includes isolated nucleotide sequences (or fragments thereof) encoding the variable light and heavy chains of the antibodies described herein as well as those nucleotide sequences (or fragments thereof) having sequences comprising, corresponding to, identical to, hybridizable to, or complementary to, at least about 70% (e.g., 70% 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78% or 79%), preferably at least about 80% (e.g., 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88% or 89%), and more preferably at least about 90% (e.g, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) identity to these encoding nucleotide sequences. (All integers (and portions thereof) between and including 70% and 100% are considered to be within the scope of the present invention with respect to percent identity.) Such sequences may be derived from any source (e.g., either isolated from a natural source, produced via a semi-synthetic route, or synthesized de novo). In particular, such sequences may be isolated or derived from sources other than described in the examples (e.g., bacteria, fungus, algae, mouse or human).
In addition to the nucleotide sequences described above, the present invention also includes amino acid sequences of the variable light and heavy chains of the antibodies described herein (or fragments of these amino acid sequences). Further, the present invention also includes amino acid sequences (or fragments thereof) comprising, corresponding to, identical to, or complementary to at least about 70% (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78% or 79%), preferably at least about 80% (e.g., 80% 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88% or 89%), and more preferably at least about 90% identity (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%), to the amino acid sequences of the proteins of the present invention. (Again, all integers (and portions thereof) between and including 70% and 100% (as recited in connection with the nucleotide sequence identities noted above) are also considered to be within the scope of the present invention with respect to percent identity.)
For purposes of the present invention, a “fragment” of a nucleotide sequence is defined as a contiguous sequence of approximately at least 6, preferably at least about 8, more preferably at least about 10 nucleotides, and even more preferably at least about 15 nucleotides corresponding to a region of the specified nucleotide sequence.
The term “identity” refers to the relatedness of two sequences on a nucleotide-by-nucleotide basis over a particular comparison window or segment. Thus, identity is defined as the degree of sameness, correspondence or equivalence between the same strands (either sense or antisense) of two DNA segments (or two amino acid sequences). “Percentage of sequence identity” is calculated by comparing two optimally aligned sequences over a particular region, determining the number of positions at which the identical base or amino acid occurs in both sequences in order to yield the number of matched positions, dividing the number of such positions by the total number of positions in the segment being compared and multiplying the result by 100. Optimal alignment of sequences may be conducted by the algorithm of Smith & Waterman, Appl. Math. 2:482 (1981), by the algorithm of Needleman & Winch, J. Mol. Biol. 48:443 (1970), by the method of Pearson & Lipmann, Proc. Natl. Acad. Sci. (USA) 85:2444 (1988) and by computer programs which implement the relevant algorithms (e.g., Crustal Macaw Pileup (Higgins et al., CABIOS. 5L151-153 (1989)), FASTDB (Intelligentsias), BLAST (National Center for Biomedical Information; Latches et al., Nucleic Acids Research 25:3389-3402 (1997)), PILEUP (Genetics Computer Group, Madison, Wis.) or GAP, BESTFIT, FASTA and TFASTA (Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, Madison, Wis.). (See U.S. Pat. No. 5,912,120.)
For purposes of the present invention, “complementarity” is defined as the degree of relatedness between two DNA segments. It is determined by measuring the ability of the sense strand of one DNA segment to hybridize with the antisense strand of the other DNA segment, under appropriate conditions, to form a double helix. A “complement” is defined as a sequence which pairs to a given sequence based upon the canonic base-pairing rules. For example, a sequence A-G-T in one nucleotide strand is “complementary” to T-C-A in the other strand.
In the double helix, adenine appears in one strand, thymine appears in the other strand. Similarly, wherever guanine is found in one strand, cytosine is found in the other. The greater the relatedness between the nucleotide sequences of two DNA segments, the greater the ability to form hybrid duplexes between the strands of the two DNA segments.
“Similarity” between two amino acid sequences is defined as the presence of a series of identical as well as conserved amino acid residues in both sequences. The higher the degree of similarity between two amino acid sequences, the higher the correspondence, sameness or equivalence of the two sequences. (“Identity between two amino acid sequences is defined as the presence of a series of exactly alike or invariant amino acid residues in both sequences.) The definitions of “complementarity”, “identity” and “similarity” are well known to those of ordinary skill in the art.
“Encoded by” refers to a nucleic acid sequence which codes for a polypeptide sequence, wherein the polypeptide sequence or a portion thereof contains an amino acid sequence of at least 3 amino acids, more preferably at least 8 amino acids, and even more preferably at least 15 amino acids from a polypeptide encoded by the nucleic acid sequence.
“Biological activity” as used herein, refers to all inherent biological properties of an antibody against troponin I or troponin I. Such properties include, for example, the ability of the antibody to bind to troponin I and functionally-related antibodies described herein.
The terms “specific binding” or “specifically binding”, as used herein, in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope “A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled “A” and the antibody, will reduce the amount of labeled A bound to the antibody.
The term “antibody”, as used herein, broadly refers to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any functional fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule. Such mutant, variant, or derivative antibody entities are known in the art, non-limiting embodiments of which are discussed below.
In a full-length antibody, each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG 1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass and may be from any species (e.g., mouse, human, chicken, rat, rabbit, sheep, shark and camelid).
The CDRs of the antibodies of the present invention are shown in Tables 1 and 2 below:
TABLE 1
CDRs OF HEAVY CHAIN FOR TnI 19C7 AM1
SEQ ID NO. Protein region Sequence
52 CDR H1 GYTFTDYNLH
53 CDR H2 YIYPYNGITGYNQKFKS
54 CDR H3 DAYDYDYLTD
TABLE 2
CDRs OF LIGHT CHAIN FOR TnI 19C7 AM1
SEQ ID NO. Protein region Sequence
55 CDR Ll RTSKNVGTNIH
56 CDR L2 YASERLP
57 CDR L3 QQSNNWPYT
The term “antigen-binding portion” of an antibody (or simply “antibody portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by one or more fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific, specifically binding to two or more different antigens. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546, Winter et al., Intern. Appln. Public. No. WO 90/05144 A1 herein incorporated by reference), which comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also encompassed herein within the term “antigen-binding portion” of an antibody. Other forms of single chain antibodies, such as diabodies, are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5).
The term “antibody construct” as used herein refers to a polypeptide comprising one or more the antigen binding portions of the invention linked to a linker polypeptide or an immunoglobulin constant domain. Linker polypeptides comprise two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions. Such linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). An immunoglobulin constant domain refers to a heavy or light chain constant domain. Human IgG heavy chain and light chain constant domain amino acid sequences are known in the art, and examples are presented in Table 3.
TABLE 3
SEQUENCE OF HUMAN IgG HEAVY CHAIN CONSTANT
DOMAIN AND LIGHT CHAIN CONSTANT DOMAIN
Se-
quence
Identi- Sequence
Protein fier 12345678901234567890123456789012
Ig SEQ ID ASTKGPSVFFLAPSSKSTSGGTAALGCLVKDYFP
gamma-1 NO.: 50 EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV
constant VTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKS
region CDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI
SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSR
EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN
NYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVF
SCSVMHEALHNHYTQKSLSLSPGK
Ig SEQ ID ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFP
gamma-1 NO.: 51 EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV
constant VTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKS
region CDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMI
mutant SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA
KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKC
KVSNKALPAPIEKTISKAKGQPREPQVYTLPPSR
EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN
NYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVF
SCSVMHEALHNHYTQKSLSLSPGK
Ig Kappa SEQ ID TVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPR
constant NO.: 61 EAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLS
region STLTLSKADYEKHKVYACEVTHQGLSSPVTKSFN
RGEC
Ig SEQ ID QPKAAPSVTLFPPSSEELQANKATLVCLISDFYP
Lambda NO.: 62 GAVTVAWKADSSPVKAGVETTTPSKQSNNKYAAS
constant SYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPT
region ECS
Still further, an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.
An “isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds troponin I is substantially free of antibodies that specifically bind antigens other than troponin I). An isolated antibody that specifically binds troponin I may, however, have cross-reactivity to other antigens, such as troponin I molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
The term “human antibody”, as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
The term “recombinant human antibody”, as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described below), antibodies isolated from a recombinant, combinatorial human antibody library (Hoogenboom H. R., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W. E., (2002) Clin. Biochem. 35:425-445; Gavilondo J. V., and Larrick J. W. (2002) BioTechniques 29:128-145; Hoogenboom H., and Chames P. (2000) Immunology Today 21:371-378), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295; Kellermann, S-A. and Green, L. L. (2002) Current Opinion in Biotechnology 13:593-597; Little M. et al (2000) Immunology Today 21:364-370) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
The term “chimeric antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species. The present invention encompasses chimeric antibodies having, for example, murine heavy and light chain variable regions linked to human constant regions.
The term “CDR-grafted antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.
The term “humanized antibody” refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more “human-like”, i.e., more similar to human germline variable sequences. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences.
The terms “Kabat numbering”, “Kabat definitions and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e. hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391 and Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3. (In addition, for purposes of the present invention, the AbM definition as defined by Oxford Molecular's ABM antibody modeling software was used to define the CDR-H1 region from amino acids 26-35 for the heavy chain.)
As used herein, the terms “acceptor” and “acceptor antibody” refer to the antibody or nucleic acid sequence providing or encoding at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or 100% of the amino acid sequences of one or more of the framework regions. In some embodiments, the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding the constant region(s). In yet another embodiment, the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding one or more of the framework regions and the constant region(s). In a specific embodiment, the term “acceptor” refers to a human antibody amino acid or nucleic acid sequence that provides or encodes at least 80%, preferably, at least 85%, at least 90%, at least 95%, at least 98%, or 100% of the amino acid sequences of one or more of the framework regions. In accordance with this embodiment, an acceptor may contain at least 1, at least 2, at least 3, least 4, at least 5, or at least 10 amino acid residues that does (do) not occur at one or more specific positions of a human antibody. An acceptor framework region and/or acceptor constant region(s) may be, e.g., derived or obtained from a germline antibody gene, a mature antibody gene, a functional antibody (e.g., antibodies well-known in the art, antibodies in development, or antibodies commercially available).
As used herein, the term “CDR” refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3, for each of the variable regions. The term “CDR set” as used herein refers to a group of three CDRs that occur in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987) and (1991)) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody, but also provides precise residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Chothia and coworkers (Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987) and Chothia et al., Nature 342:877-883 (1989)) found that certain sub-portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. These sub-portions were designated as L1, L2 and L3 or H1, H2 and H3 where the “L” and the “H” designates the light chain and the heavy chains regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs have been described by Padlan (FASEB J. 9:133-139 (1995)) and MacCallum (J Mol Biol 262(5):732-45 (1996)). Still other CDR boundary definitions may not strictly follow one of the above systems, such as AbM definitions, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. The methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat, AbM or Chothia defined CDRs.
As used herein, the term “canonical” residue refers to a residue in a CDR or framework that defines a particular canonical CDR structure as defined by Chothia et al. (J. Mol. Biol. 196:901-907 (1987); Chothia et al., J. Mol. Biol. 227: 799 (1992), both are incorporated herein by reference). According to Chothia et al., critical portions of the CDRs of many antibodies have nearly identical peptide backbone confirmations despite great diversity at the level of amino acid sequence. Each canonical structure specifies primarily a set of peptide backbone torsion angles for a contiguous segment of amino acid residues forming a loop.
As used herein, the terms “donor” and “donor antibody” refer to an antibody providing one or more CDRs. In a preferred embodiment, the donor antibody is an antibody from a species different from the antibody from which the framework regions are obtained or derived. In the context of a humanized antibody, the term “donor antibody” refers to a non-human antibody providing one or more CDRs.
As used herein, the term “framework” or “framework sequence” refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to correspondingly different interpretations. The six CDRs (CDR-L1, -L2, and -L3 of light chain and CDR-H1, -H2, and -H3 of heavy chain) also divide the framework regions on the light chain and the heavy chain into four sub-regions (FR1, FR2, FR3 and FR4) on each chain, in which CDR1 is positioned between FR1 and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4. Without specifying the particular sub-regions as FR1, FR2, FR3 or FR4, a framework region, as referred by others, represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain. As used herein, a FR represents one of the four sub-regions, and FRs represents two or more of the four sub-regions constituting a framework region.
In one embodiment of the invention, the murine heavy chain and light chain donor sequences are selected from the sequences described below:
TABLE 4
HEAVY CHAIN DONOR SEQUENCES FOR TnI 19C7 AM1
Sequence
SEQ ID No. 12345678901234567890123456789012
69 EVTLRESGPALVKPTQTLTLTCTFSGFSLS
70 WIRQPPGKALEWLA
71 RLTISKDTSKNQVVLTMTNMDPVDTATYYCAR
72 WGQGTTVTVSS
73 EVTLKESGPVLVKPTETLTLTCTVSGFSLS
74 WIRQPPGKALEWLA
75 RLTISKDTSKSQVVLTMTNMDPVDTATYYCAR
76 WGQGTTVTVSS
77 EVQLVESGGGLVQPGGSLRLSCAASGFTFS
78 WVRQAPGKGLEWVG
79 RFTISRDDSKNSLYLQMNSLKTEDTAVYYCAR
80 WGQGTTVTVSS
81 EVQLVESGGGLVKPGGSLRLSCAASGFTFS
82 WVRQAPGKGLEWVS
83 RFTISRDNAKNSLYLQMNSLRAEDTAVYYCAR
84 WGQGTTVTVSS
85 EVQLVQSGAEVKKPGSSVKVSCKASGGTFS
86 WVRQAPGQGLEWMG
87 RVTITADKSTSTAYMELSSLRSEDTAVYYCAR
88 WGQGTTVTVSS
89 EVQLVQSGAEVKKPGASVKVS CKASGYTFT
90 WVRQAPGQGLEWMG
91 RVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR
92 WGQGTTVTVSS
TABLE 5
LIGHT CHAIN DONOR SEQUENCES FOR TnI 19C7 AM1
Sequence
SEQ ID No. 12345678901234567890123456789012
93 DIVMTQSPDSLAVSLGERATINC
94 WYQQKPGQPPKLLIY
95 GVPDRFSGSGSGTDFTLTISSLQAEDVAVYYC
96 FGGGTKVEIKR
97 EIVMTQSPATLSVSPGERATLSC
98 WYQQKPGQAPRLLIY
99 GIPARFSGSGSGTEFTLTISSLQSEDFAVYYC
100 FGGGTKVEIKR
101 DIQMTQSPSSLSASVGDRVTITC
102 WYQQKPEKAPKSLIY
103 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC
104 FGGGTKVEIKR
105 DIQMTQSPSSVSASVGDRVTITC
106 WYQOKPGKAPKLLIY
107 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC
108 FGGGTKVEIKR
As used herein, the term “germline antibody gene” or “gene fragment” refers to an immunoglobulin sequence encoded by non-lymphoid cells that have not undergone the maturation process that leads to genetic rearrangement and mutation for expression of a particular immunoglobulin. (See, e.g., Shapiro et al., Crit. Rev. Immunol. 22(3): 183-200 (2002); Marchalonis et al., Adv Exp Med Biol. 484:13-30 (2001)). One of the advantages provided by various embodiments of the present invention stems from the recognition that germline antibody genes are more likely than mature antibody genes to conserve essential amino acid sequence structures characteristic of individuals in the species, hence less likely to be recognized as from a foreign source when used therapeutically in that species.
As used herein, the term “key” residues refer to certain residues within the variable region that have more impact on the binding specificity and/or affinity of an antibody, in particular a humanized antibody. A key residue includes, but is not limited to, one or more of the following: a residue that is adjacent to a CDR, a potential glycosylation site (can be either N- or O-glycosylation site), a rare residue, a residue capable of interacting with the antigen, a residue capable of interacting with a CDR, a canonical residue, a contact residue between heavy chain variable region and light chain variable region, a residue within the Vernier zone, and a residue in the region that overlaps between the Chothia definition of a variable heavy chain CDR1 and the Kabat definition of the first heavy chain framework.
As used herein, the term “humanized antibody” is an antibody or a variant, derivative, analog or fragment thereof which immunospecifically binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody. As used herein, the term “substantially” in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98% and most preferably at least 99% identical to the amino acid sequence of a non-human antibody CDR. A humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab′, F(ab′)2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. Preferably, a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In some embodiments, a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain. The antibody also may include the CH1, hinge, CH2, CH3, and CH4 regions of the heavy chain. In other embodiments, a humanized antibody only contains a humanized light chain. In some embodiments, a humanized antibody only contains a humanized heavy chain. In specific embodiments, a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.
The humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including without limitation IgG 1, IgG2, IgG3 and IgG4. The humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well-known in the art.
The framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. In a preferred embodiment, such mutations, however, will not be extensive. Usually, at least 80%, preferably at least 85%, more preferably at least 90%, and most preferably at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences. As used herein, the term “consensus framework” refers to the framework region in the consensus immunoglobulin sequence. As used herein, the term “consensus immunoglobulin sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
As used herein, “Vernier” zone refers to a subset of framework residues that may adjust CDR structure and fine-tune the fit to antigen as described by Foote and Winter (1992, J. Mol. Biol. 224:487-499, which is incorporated herein by reference). Vernier zone residues form a layer underlying the CDRs and may impact on the structure of CDRs and the affinity of the antibody.
The term “activity” includes activities such as the binding specificity/affinity of an antibody for an antigen, for example, an anti-troponin I antibody that binds to troponin I.
The term “epitope” includes any polypeptide determinant capable of specific binding to an immunoglobulin or T-cell receptor. In certain embodiments, epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics. An epitope is a region of an antigen that is bound by an antibody. In certain embodiments, an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
The term “surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jönsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26; Jönsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131; and Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277.
The term “Kon”, as used herein, is intended to refer to the on rate constant for association of an antibody to the antigen to form the antibody/antigen complex as is known in the art.
The term “Koff”, as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex as is known in the art.
The term “Kd” or “KD”, as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction as is known in the art.
The term “labeled binding protein” as used herein, refers to a protein with a label incorporated that provides for the identification of the binding protein. Preferably, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or calorimetric methods). Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 35S, 90Y, 99Tc, 111In, 125I, 131I, 177Lu, 166Ho, or 153Sm); fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates.
The term “antibody conjugate” refers to a binding protein, such as an antibody, chemically linked to a second chemical moiety, such as a therapeutic or cytotoxic agent. The term “agent” is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials. Preferably the therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. The terms “crystal”, and “crystallized” as used herein, refer to an antibody, or antigen-binding portion thereof, that exists in the form of a crystal. Crystals are one form of the solid state of matter, which is distinct from other forms such as the amorphous solid state or the liquid crystalline state. Crystals are composed of regular, repeating, three-dimensional arrays of atoms, ions, molecules (e.g., proteins such as antibodies), or molecular assemblies (e.g., antigen/antibody complexes). These three-dimensional arrays are arranged according to specific mathematical relationships that are well-understood in the field. The fundamental unit, or building block, that is repeated in a crystal is called the asymmetric unit. Repetition of the asymmetric unit in an arrangement that conforms to a given, well-defined crystallographic symmetry provides the “unit cell” of the crystal. Repetition of the unit cell by regular translations in all three dimensions provides the crystal. See Giege, R. and Ducruix, A. Barrett, Crystallization of Nucleic Acids and Proteins, a Practical Approach, 2nd ed., pp. 20 1-16, Oxford University Press, New York, N.Y., (1999).”
The term “polynucleotide” as referred to herein means a polymeric form of two or more nucleotides, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes single and double-stranded forms of DNA but preferably is double-stranded DNA.
The term “isolated polynucleotide” as used herein shall mean a polynucleotide (e.g., of genomic, cDNA, or synthetic origin, or some combination thereof) that, by virtue of its origin, is not associated with all or a portion of a polynucleotide with which the “isolated polynucleotide” is found in nature; is operably linked to a polynucleotide that it is not linked to in nature; or does not occur in nature as part of a larger sequence.
The term “vector”, as used herein, is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, “plasmid” and “vector” may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
The term “operably linked” refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences. “Operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term “expression control sequence” as used herein refers to polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence. The term “control sequences” is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
“Transformation”, as defined herein, refers to any process by which exogenous DNA enters a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment. Such “transformed” cells include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells that transiently express the inserted DNA or RNA for limited periods of time.
The term “recombinant host cell” (or simply “host cell”), as used herein, is intended to refer to a cell into which exogenous DNA has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. Preferably host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life. Preferred eukaryotic cells include protist, fungal, plant and animal cells. Most preferably host cells include but are not limited to the prokaryotic cell line E. coli; mammalian cell lines CHO, HEK 293 and COS; the insect cell line Sf9; and the fungal cell Saccharomyces cerevisiae or Picchia pastoris.
Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.
“Transgenic organism”, as known in the art and as used herein, refers to an organism having cells that contain a transgene, wherein the transgene introduced into the organism (or an ancestor of the organism) expresses a polypeptide not naturally expressed in the organism. A “transgene” is a DNA construct, which is stably and operably integrated into the genome of a cell from which a transgenic organism develops, directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic organism.
The term “regulate” and “modulate” are used interchangeably, and, as used herein, refers to a change or an alteration in the activity of a molecule of interest (e.g., the biological activity of troponin I). Modulation may be an increase or a decrease in the magnitude of a certain activity or function of the molecule of interest. Exemplary activities and functions of a molecule include, but are not limited to, binding characteristics, enzymatic activity, cell receptor activation, and signal transduction.
Correspondingly, the term “modulator,” as used herein, is a compound capable of changing or altering an activity or function of a molecule of interest (e.g., the biological activity of troponin I). For example, a modulator may cause an increase or decrease in the magnitude of a certain activity or function of a molecule compared to the magnitude of the activity or function observed in the absence of the modulator. In certain embodiments, a modulator is an inhibitor, which decreases the magnitude of at least one activity or function of a molecule. Exemplary inhibitors include, but are not limited to, proteins, peptides, antibodies, peptibodies, carbohydrates or small organic molecules. Peptibodies are described, e.g., in International Application Publication No. WO 01/83525.
The term “agonist”, as used herein, refers to a modulator that, when contacted with a molecule of interest, causes an increase in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the agonist. Particular agonists of interest may include, but are not limited to, troponin I polypeptides, nucleic acids, carbohydrates, or any other molecules that bind to troponin I.
The term “antagonist” or “inhibitor”, as used herein, refer to a modulator that, when contacted with a molecule of interest causes a decrease in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the antagonist.
As used herein, the term “effective amount” refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity and/or duration of a disorder or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g., prophylactic or therapeutic agent).
The term “sample”, as used herein, is used in its broadest sense. A “biological sample”, as used herein, includes, but is not limited to, any quantity of a substance from a living thing or formerly living thing. Such living things include, but are not limited to, humans, mice, rats, monkeys, dogs, rabbits and other mammalian or non-mammalian animals. Such substances include, but are not limited to, blood, serum, urine, synovial fluid, cells, organs, tissues (e.g., brain), bone marrow, lymph nodes, cerebrospinal fluid, and spleen.
Methods of Making Antibodies
Antibodies of the present invention may be made by any of a number of techniques known in the art. For example, antibodies can be prepared using a wide variety of techniques including the use of recombinant or phage display technologies, or a combination thereof. The term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
In one embodiment, the present invention provides a method of generating recombinant antibodies (as well as antibodies produced by the method) comprising culturing a Chinese Hamster Ovary cell line secreting an antibody of the invention.
Further, fragments of the antibody of the present invention which recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab′)2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′)2 fragments). F(ab′)2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
Production of Anti-Troponin I Antibodies Using Recombinant Antibody Libraries
In vitro methods also can be used to make the antibodies of the invention, wherein an antibody library is screened to identify an antibody having the desired binding specificity. Methods for such screening of recombinant antibody libraries are well known in the art and include methods described in, for example, Ladner et al., U.S. Pat. No. 5,223,409; Kang et al., International Appln. Publication No. WO 92/18619; Dower et al., International Appln. Publication No. WO 91/17271; Winter et al., International Appln. Publication No. WO 92/20791; Markland et al., International Appln. Publication No. WO 92/15679; Breitling et al., International Appln. Publication No. WO 93/01288; McCafferty et al., PCT Publication No. WO 92/01047; Garrard et al., International Appln. Publication No. WO 92/09690; Fuchs et al. (1991), Bio/Technology 9:1370-1372; Hay et al., (1992) Hum Antibod Hybridomas 3:81-85; Huse et al. (1989), Science 246: 1275-1281; McCafferty et al., Nature (1990) 348:552-554; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al., (1992) J Mol Biol 226:889-896; Clackson et al., (1991) Nature 352:624-628; Gram et al., (1992) PNAS 89:3576-3580; Garrad et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991), Nuc Acid Res 19:4133-4137; and Barbas et al. (1991), PNAS 88:7978-7982, U.S. Patent Application Publication No. 20030186374, and International Application Publication No. WO 97/29131, the contents of each of which are incorporated herein by reference.
The recombinant antibody library may be from a subject immunized with troponin I, or a portion thereof Alternatively, the recombinant antibody library may be from a naive subject, i.e., one who has not been immunized with troponin I, such as a human antibody library from a human subject who has not been immunized with human troponin I. Antibodies of the invention are selected by screening the recombinant antibody library with the peptide comprising human troponin I to thereby select those antibodies that recognize troponin I. Methods for conducting such screening and selection are well known in the art, such as described in the references in the preceding paragraph. To select antibodies of the invention having particular binding affinities for troponin I, such as those that dissociate from human troponin I with a particular koff rate constant, the art-known method of surface plasmon resonance can be used to select antibodies having the desired koff rate constant.
In one aspect, the invention pertains to an isolated antibody, or an antigen-binding portion thereof, that binds human troponin I. In various embodiments, the antibody is a recombinant antibody.
In another approach the antibodies of the present invention can also be generated using yeast display methods known in the art. In yeast display methods, genetic methods are used to tether antibody domains to the yeast cell wall and display them on the surface of yeast. In particular, such yeast can be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Examples of yeast display methods that can be used to make the antibodies of the present invention include those disclosed Wittrup et al., U.S. Pat. No. 6,699,658 incorporated herein by reference.
Production of Recombinant Antibodies
As noted above, antibodies of the present invention may be produced by any number of techniques known in the art. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques is the preferred method of producing the antibodies of the present invention. (The various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.) Although it is possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells is preferable, and most preferable in mammalian host cells, because such eukaryotic cells (and in particular mammalian cells) are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr—CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R. J. Kaufman and P. A. Sharp (1982) Mol. Biol. 159:601-621), NS0 myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
In a preferred system for recombinant expression of an antibody, or antigen-binding portion thereof, of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr—CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium. Still further the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell of the invention in a suitable culture medium until a recombinant antibody of the invention is synthesized. The method can further comprise isolating the recombinant antibody from the culture medium.
Anti-Troponin Antibodies
The isolated anti-troponin I antibody CDR sequences described herein (see Tables 1 and 2) establish a novel family of troponin I binding proteins, isolated in accordance with this invention, and comprising polypeptides that include the CDR sequences listed in Tables 1 and 2 above. To generate and to select CDRs of the invention having preferred troponin I binding activity, standard methods known in the art for generating binding proteins of the present invention and assessing the binding characteristics thereof may be used, including but not limited to those specifically described herein.
Anti-Troponin I Chimeric Antibodies
A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies, such as those of the present invention, are well known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4;214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entireties. In addition, techniques developed for the production of “chimeric antibodies” (Morrison et al., 1984, Proc. Natl. Acad. Sci. 81:851-855; Neuberger et al., 1984, Nature 312:604-608; Takeda et al., 1985, Nature 314: 452-454 which are incorporated herein by reference in their entireties) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used.
In one embodiment, the chimeric antibodies of the invention are produced by replacing the heavy chain constant region of the antibodies described above with a human IgG1 constant region. In a specific embodiment, the chimeric antibody of the invention comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO:25 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO:28.
Anti-Troponin I CDR Grafted Antibodies
CDR-grafted antibodies of the invention comprise heavy and light chain variable region sequences from a human antibody wherein one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of the murine antibodies of the invention. A framework sequence from any human antibody may serve as the template for CDR grafting. However, straight chain replacement onto such a framework often leads to some loss of binding affinity to the antigen. The more homologous a human antibody is to the original murine antibody, the less likely the possibility that combining the murine CDRs with the human framework will introduce distortions in the CDRs that could reduce affinity. Therefore, it is preferable that the human variable framework that is chosen to replace the murine variable framework apart from the CDRs have at least a 65% sequence identity with the murine antibody variable region framework. It is more preferable that the human and murine variable regions apart from the CDRs have at least 70% sequence identify. It is even more preferable that the human and murine variable regions apart from the CDRs have at least 75% sequence identity. It is most preferable that the human and murine variable regions apart from the CDRs have at least 80% sequence identity. Methods for producing chimeric antibodies are known in the art and discussed in detail in Example 2.2. (See also EP 239,400; Intern. Appln. Publication No. WO 91/09967; U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5): 489-498 (1991); Studnicka et al., Protein Engineering 7(6): 805-814 (1994); Roguska et al., PNAS 91:969-973 (1994)), and chain shuffling (U.S. Pat. No. 5,565,352).
Humanized Antibodies
Humanized antibodies are antibody molecules from non-human species antibody that bind the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule Such imported sequences can be used to reduce immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic, as known in the art.
Framework residues in the human framework regions may be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding. Antibodies can be humanized using a variety of techniques known in the art, such as but not limited to those described in Jones et al., Nature 321:522 (1986); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol. 151: 2296 (1993); Chothia and Lesk, J. Mol. Biol. 196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285 (1992); Presta et al., J. Immunol. 151:2623 (1993), Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska et al., PNAS 91:969-973 (1994); International Appln. Publication No. WO 91/09967, PCT/: US98/16280, US96/18978, US91/09630, US91/05939, US94/01234, GB89/01334, GB91/01134, GB92/01755; WO90/14443, WO90/14424, WO90/14430, EP 229246, EP 592,106; EP 519,596, EP 239,400, U.S. Pat. Nos. 5,565,332, 5,723,323, 5,976,862, 5,824,514, 5,817,483, 5,814,476, 5,763,192, 5,723,323, 5,766,886, 5,714,352, 6,204,023, 6,180,370, 5,693,762, 5,530,101, 5,585,089, 5,225,539; 4,816,567, each entirely incorporated herein by reference, included references cited therein.
Production of Antibodies and Antibody-Producing Cell Lines
As noted above, preferably, antibodies of the present invention exhibit a high capacity to bind specifically to troponin I, e.g., as assessed by any one of several in vitro and in vivo assays known in the art (e.g., see examples below).
In certain embodiments, the antibody comprises a heavy chain constant region, such as an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region. Preferably, the heavy chain constant region is an IgG1 heavy chain constant region or an IgG4 heavy chain constant region. Furthermore, the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region. Preferably, the antibody comprises a kappa light chain constant region. Alternatively, the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
Replacements of amino acid residues in the Fc portion to alter antibody effector function are known in the art (Winter et al., U.S. Pat. Nos. 5,648,260 and 5,624,821). The Fc portion of an antibody mediates several important effector functions, for example, cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/clearance rate of antibody- and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives. Certain human IgG isotypes, particularly IgG1 and IgG3, mediate ADCC and CDC via binding to FcγRs and complement Clq, respectively. Neonatal Fc receptors (FcRn) are the critical components determining the circulating half-life of antibodies. In still another embodiment, at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered.
One embodiment provides a labeled binding protein wherein an antibody or antibody portion of the invention is derivatized or linked to another functional molecule (e.g., another peptide or protein). For example, a labeled binding protein of the invention can be derived by functionally linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
Useful detectable agents with which an antibody or antibody portion of the invention may be derivatized include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
Another embodiment of the invention provides a crystallized binding protein. Preferably, the invention relates to crystals of whole anti-troponin I antibodies and fragments thereof as disclosed herein, and formulations and compositions comprising such crystals. In one embodiment the crystallized binding protein has a greater half-life in vivo than the soluble counterpart of the binding protein. In another embodiment, the binding protein-retains biological activity after crystallization.
Crystallized binding protein of the invention may be produced according methods known in the art and as disclosed in International Appln. Publication No. WO 02/072636, incorporated herein by reference.
Another embodiment of the invention provides a glycosylated binding protein wherein the antibody or antigen-binding portion thereof comprises one or more carbohydrate residues. Nascent in vivo protein production may undergo further processing, known as post-translational modification. In particular, sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation. The resulting proteins bearing covalently linked oligosaccharide side chains are known as glycosylated proteins or glycoproteins. Antibodies are glycoproteins with one or more carbohydrate residues in the Fc domain, as well as the variable domain. Carbohydrate residues in the Fc domain have important effect on the effector function of the Fc domain, with minimal effect on antigen binding or half-life of the antibody (R. Jefferis, Biotechnol. Prog. 21 (2005), pp. 11-16). In contrast, glycosylation of the variable domain may have an effect on the antigen binding activity of the antibody. Glycosylation in the variable domain may have a negative effect on antibody binding affinity, likely due to steric hindrance (Co, M. S., et al., Mol. Immunol. (1993) 30:1361-1367), or result in increased affinity for the antigen (Wallick, S. C., et al., Exp. Med. (1988) 168:1099-1109; Wright, A., et al., EMBO J. (1991) 10:2717 2723).
One aspect of the present invention is directed to generating glycosylation site mutants in which the O- or N-linked glycosylation site of the binding protein has been mutated. One skilled in the art can generate such mutants using standard well-known technologies. The creation of glycosylation site mutants that retain the biological activity but have increased or decreased binding activity are another object of the present invention.
In still another embodiment, the glycosylation of the antibody or antigen-binding portion of the invention is modified. For example, an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen. Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for antigen. Such an approach is described in further detail in International Appln. Publication No. WO 03/016466A2, and U.S. Pat. Nos. 5,714,350 and 6,350,861, each of which is incorporated herein by reference in its entirety.
Additionally or alternatively, a modified antibody of the invention can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNAc structures. Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol. Chem. 277:26733-26740; Umana et al. (1999) Nat. Biotech. 17:176-1, as well as, European Patent No: EP 1,176,195; International Appln. Publication Nos. WO 03/035835 and WO 99/5434280, each of which is incorporated herein by reference in its entirety.
Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms may produce different glycosylation enzymes (e.g., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid. Preferably the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human.
It is known to those skilled in the art that differing protein glycosylation may result in differing protein characteristics. For instance, the efficacy of a therapeutic protein produced in a microorganism host, such as yeast, and glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line. Such glycoproteins may also be immunogenic in humans and show reduced half-life in vivo after administration. Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream. Other adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity. Accordingly, a practitioner may prefer a therapeutic protein with a specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal.
Expressing glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. Using techniques known in the art a practitioner may generate antibodies or antigen-binding portions thereof exhibiting human protein glycosylation. For example, yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins (glycoproteins) produced in these yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S. Patent Application Publication Nos. 20040018590 and 20020137134 and International Appln. Publication No. WO 05/100584 A2).
The term “multivalent binding protein” is used in this specification to denote a binding protein comprising two or more antigen binding sites. The multivalent binding protein is preferably engineered to have the three or more antigen binding sites, and is generally not a naturally occurring antibody. The term “multispecific binding protein” refers to a binding protein capable of binding two or more related or unrelated targets. Dual variable domain (DVD) binding proteins as used herein, are binding proteins that comprise two or more antigen binding sites and are tetravalent or multivalent binding proteins. Such DVDs may be monospecific, i.e., capable of binding one antigen or multispecific, i.e., capable of binding two or more antigens. DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to a DVD Ig. Each half of a DVD Ig comprises a heavy chain DVD polypeptide, and a light chain DVD polypeptide, and two antigen binding sites. Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site. DVD binding proteins and methods of making DVD binding proteins are disclosed in U.S. patent application Ser. No. 11/507,050 and incorporated herein by reference.
One aspect of the invention pertains to a DVD binding protein comprising binding proteins capable of binding to troponin I. Preferably, the DVD binding protein is capable of binding troponin I and a second target. The present invention also encompasses triple-variable domain (TVD) binding proteins in which the antibody is capable of binding troponin I as well as two additional targets (i.e., a second and third target).
In addition to the binding proteins, the present invention is also directed to an anti-idiotypic (anti-Id) antibody specific for such binding proteins of the invention. An anti-Id antibody is an antibody, which recognizes unique determinants generally associated with the antigen-binding region of another antibody. The anti-Id can be prepared by immunizing an animal with the binding protein (e.g., antibody of interest) or a CDR containing region thereof. The immunized animal will recognize, and respond to the idiotypic determinants of the immunizing antibody and produce an anti-Id antibody. The anti-Id antibody may also be used as an “immunogen” to induce an immune response in yet another animal, producing a so-called anti-anti-Id antibody.
Further, it will be appreciated by one skilled in the art that a protein of interest may be expressed using a library of host cells genetically engineered to express various glycosylation enzymes, such that member host cells of the library produce the protein of interest with variant glycosylation patterns. A practitioner may then select and isolate the protein of interest with particular novel glycosylation patterns. Preferably, the protein having a particularly selected novel glycosylation pattern exhibits improved or altered biological properties.
Diagnostic Uses of Anti-Troponin I Antibodies
Given their ability to bind to troponin I, the anti-troponin I antibodies, or portions thereof, of the invention can be used to detect troponin I (e.g., in a biological sample such as serum, whole blood, CSF, brain tissue or plasma), using a conventional immunoassay competitive or non-competitive assay such as, for example, an enzyme linked immunosorbent assay (ELISA), a radioimmunoassay (RIA), an immunometric assay or tissue immunohistochemistry. The invention therefore provides a method for detecting troponin I in a biological sample comprising contacting a biological sample with an antibody, or antibody portion, of the invention and detecting either the antibody (or antibody portion) bound to troponin I or unbound antibody (or antibody portion), to thereby detect troponin I in the biological sample. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetyl-cholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3H, 14C, 35S, 90Y, 99Tc, 111In, 125I, 131I, 177Lu, 166Ho, or 153Sm.
As an alternative to labeling the antibody, troponin I can be assayed in biological fluids by a competition immunoassay utilizing recombinant troponin I standards labeled with a detectable substance and an unlabeled anti-troponin I antibody. In this assay, the biological sample, the labeled recombinant troponin I standards and the anti-troponin I antibody are combined, and the amount of labeled recombinant troponin I standard bound to the unlabeled antibody is determined. The amount of troponin I in the biological sample is inversely proportional to the amount of labeled recombinant troponin I standard bound to the anti-troponin I antibody.
In particular, in one embodiment of the present invention, one or more of the antibodies of the present invention are coated on a solid phase (or are in a liquid phase). The test or biological sample (e.g., serum, plasma, urine, etc.) is then contacted with the solid phase. If troponin I antigens are present in the sample, such antigens bind to the antibodies on the solid phase and are then detected by either a direct or indirect method. The direct method comprises simply detecting presence of the complex itself and thus presence of the antigens. It should be noted that one may use the full antibody or a fragment thereof in connection with the antibodies coated on the solid phase. (For purposes of the present invention, a “fragment” or “portion” of an antibody is defined as a subunit of the antibody which reacts in the same manner, functionally, as the full antibody with respect to binding properties.)
As mentioned above, in the indirect method, a conjugate is added to the bound antigen. The conjugate comprises a second antibody, which binds to the bound antigen, attached to a signal-generating compound or label. Should the second antibody bind to the bound antigen, the signal-generating compound generates a measurable signal. Such signal then indicates presence of the antigen (i.e., troponin I) in the test sample.
Examples of solid phases used in diagnostic immunoassays are porous and non-porous materials, latex particles, magnetic particles, microparticles (see U.S. Pat. No. 5,705,330), beads, membranes, microtiter wells and plastic tubes. The choice of solid phase material and method of labeling the antigen or antibody present in the conjugate, if desired, are determined based upon desired assay format performance characteristics.
As noted above, the conjugate (or indicator reagent) will comprise an antibody (or perhaps anti-antibody, depending upon the assay), attached to a signal-generating compound or label. This signal-generating compound or “label” is itself detectable or may be reacted with one or more additional compounds to generate a detectable product. Examples of signal-generating compounds include chromogens, radioisotopes (e.g., 125I, 131I, 32P, 3H, 35S and 14C), chemiluminescent compounds (e.g., acridinium), particles (visible or fluorescent), nucleic acids, complexing agents, or catalysts such as enzymes (e.g., alkaline phosphatase, acid phosphatase, horseradish peroxidase, beta-galactosidase and ribonuclease). In the case of enzyme use (e.g., alkaline phosphatase or horseradish peroxidase), addition of a chromo-, fluro-, or lumo-genic substrate results in generation of a detectable signal. Other detection systems such as time-resolved fluorescence, internal-reflection fluorescence, amplification (e.g., polymerase chain reaction) and Raman spectroscopy are also useful.
As noted above, examples of biological fluids which may be tested by the above immunoassays include plasma, urine, whole blood, dried whole blood, serum, cerebrospinal fluid, saliva, tears, nasal washes or aqueous extracts of tissues and cells.
Additionally, it should also be noted that the initial capture antibody (for detecting troponin I antigens) used in the immunoassay may be covalently or non-covalently (e.g., ionic, hydrophobic, etc.) attached to the solid phase. Linking agents for covalent attachment are known in the art and may be part of the solid phase or derivatized to it prior to coating.
Further, the assays and kits of the present invention optionally can be adapted or optimized for point of care assay systems, including Abbott's Point of Care (i-STAT™) electrochemical immunoassay system. Immunosensors and methods of manufacturing and operating them in single-use test devices are described, for example in U.S. Pat. No. 5,063,081 and published U.S. Patent Application Nos. 20030170881, 20040018577, 20050054078, and 20060160164 (incorporated by reference herein for their teachings regarding same).
Of course, any of the exemplary formats herein and any assay or kit according to the invention can be adapted or optimized for use in automated and semi-automated systems (including those in which there is a solid phase comprising a microparticle), as described, e.g., in U.S. Pat. Nos. 5,089,424 and 5,006,309, and as, e.g., commercially marketed by Abbott Laboratories (Abbott Park, Ill.) including but not limited to Abbott's ARCHITECT®, AXSYM, IMX, PRISM, and Quantum II platforms, as well as other platforms.
Other assay formats which may be used for purposes of the present invention include, for example, a rapid test, a Western blot, as well as the use of paramagnetic particles in, for example, an ARCHITECT® assay (see Frank Quinn, The Immunoassay Handbook, Second edition, edited by David Wild, pages 363-367, 2001, herein incorporated in its entirety by reference). Such formats are known to those of ordinary skill in the art.
It should also be noted that the elements of the assays described above are particularly suitable for use in the form of a kit. The kit may also comprise one container such as a vial, bottle or strip. These kits may also contain vials or containers of other reagents needed for performing the assay, such as washing, processing and indicator reagents.
Pharmaceutical Compositions
The invention also provides pharmaceutical compositions comprising an antibody, or antigen-binding portion thereof, of the invention and a pharmaceutically acceptable carrier. The pharmaceutical compositions comprising antibodies of the invention are for use in, but not limited to, diagnosing, detecting, or monitoring a disorder, in preventing, treating, managing, or ameliorating of a disorder or one or more symptoms thereof, and/or in research. In a specific embodiment, a composition comprises one or more antibodies of the invention. In another embodiment, the pharmaceutical composition comprises one or more antibodies of the invention and one or more prophylactic or therapeutic agents other than antibodies of the invention for treating a disorder in which troponin I activity is detrimental. Preferably, the prophylactic or therapeutic agents known to be useful for or having been or currently being used in the prevention, treatment, management, or amelioration of a disorder or one or more symptoms thereof. In accordance with these embodiments, the composition may further comprise of a carrier, diluent or excipient.
The antibodies and antibody-portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion.
Various delivery systems are known and can be used to administer one or more antibodies of the invention or the combination of one or more antibodies of the invention and a prophylactic agent or therapeutic agent useful for preventing, managing treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of administering a prophylactic or therapeutic agent of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural administration, intratumoral administration, and mucosal administration (e.g., intranasal and oral routes). In addition, pulmonary administration can be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Pat. Nos. 6,019,968, 5,985,320, 5,985,309, 5,934,272, 5,874,064, 5,855,913, 5,290,540, and 4,880,078; and International Appln. Publication Nos. WO 92/19244, WO 97/32572, WO 97/44013, WO 98/31346, and WO 99/66903, each of which is incorporated herein by reference their entireties. In one embodiment, an antibody of the invention, combination therapy, or a composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.). In a specific embodiment, prophylactic or therapeutic agents of the invention are administered intramuscularly, intravenously, intratumorally, orally, intranasally, pulmonary, or subcutaneously. The prophylactic or therapeutic agents may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
In a specific embodiment, it may be desirable to administer the prophylactic or therapeutic agents of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous or non-porous material, including membranes and matrices, such as sialastic membranes, polymers, fibrous matrices (e.g., Tissuel®), or collagen matrices. In one embodiment, an effective amount of one or more antibodies of the invention antagonists is administered locally to the affected area to a subject to prevent, treat, manage, and/or ameliorate a disorder or a symptom thereof. In another embodiment, an effective amount of one or more antibodies of the invention is administered locally to the affected area in combination with an effective amount of one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than an antibody of the invention of a subject to prevent, treat, manage, and/or ameliorate a disorder or one or more symptoms thereof.
In another embodiment, the prophylactic or therapeutic agent can be delivered in a controlled release or sustained release system. In one embodiment, a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574). In another embodiment, polymeric materials can be used to achieve controlled or sustained release of the therapies of the invention (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, 1983, J. Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neurol. 25:351; Howard et al., 1989, J. Neurosurg. 7 1:105); U.S. Pat. Nos. 5,679,377; 5,916,597; 5,912,015; 5,989,463; 5,128,326; International Appln. Publication No. WO 99/15154; and International Appln. Publication No. WO 99/20253. Examples of polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly (ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. In a preferred embodiment, the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable. In yet another embodiment, a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more therapeutic agents of the invention. See, e.g., U.S. Pat. No. 4,526,938, International Appln. Publication No. WO 91/05548, International Appln. Publication No. WO 96/20698, Ning et al., 1996, “Intratumoral Radioimmunotherapy of a Human Colon Cancer Xenograft Using a Sustained-Release Gel,” Radiotherapy & Oncology 39:179-189, Song et al., 1995, “Antibody Mediated Lung Targeting of Long-Circulating Emulsions,” PDA Journal of Pharmaceutical Science & Technology 50:372-397, Cleek et al., 1997, “Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application,” Proc. Int'l. Symp. Control. Rel. Bioact. Mater. 24:853-854, and Lam et al., 1997, “Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery,” Proc. Int'l. Symp. Control Rel. Bioact. Mater. 24:759-760, each of which is incorporated herein by reference in their entireties.
It should be understood that the antibodies of the invention or antigen binding portion thereof can be used alone or in combination with one or more additional agents, e.g., a therapeutic agent (for example, a small molecule or biologic), said additional agent being selected by the skilled artisan for its intended purpose. The additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent that affects the viscosity of the composition.
It should further be understood that the combinations which are to be included within this invention are those combinations useful for their intended purpose. The agents set forth below are illustrative for purposes and not intended to be limiting. The combinations, which are part of this invention, can be the antibodies of the present invention and at least one additional agent selected from the lists below. The combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
The pharmaceutical compositions of the invention may include a “therapeutically effective amount” or a “prophylactically effective amount” of an antibody or antibody portion of the invention. A “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may be determined by a person skilled in the art and may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody, or antibody portion, are outweighed by the therapeutically beneficial effects. A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included for purposes of illustration only and are not intended to limit the scope of the present invention.
EXAMPLES Example I Generation and Isolation of 19C7
Identification of Immunoglobulin Genes
Messenger RNA was isolated from subcloned anti-TnI 19C7-144 hybridoma cells. (Hybridoma cell line TnI 19C7 is described in U.S. Patent Application Publication No. US2006/0018897.) TnI 19C7 mRNA was utilized in a reverse transcriptase-polymerase chain reaction using a mouse Ig primer set kit purchased from Novagen (Novagen (which is an Affiliate of Merck KGaA, Darmstadt, Germany), Cat No. 69831-3) with immunoglobulin gene specific primers contained in the kit. The resulting PCR products were sequenced and thus the immunoglobulin variable heavy and variable light chain genes were identified (see FIG. 2).
Cloning TnI 19C7 Variable Region Genes into pYD41 Vector
A yeast display system was used to express unmutated or wild-type anti-TnI proteins (described herein infra) and a library of anti-TnI proteins on the yeast surface as a fusion to the yeast protein AGA2. A yeast display vector called pYD (Invitrogen, Carlsbad, Calif.) was used as it allows for cloning of the anti-TnI gene at the C-terminus of the AGA2 gene, a yeast mating factor (See, Boder and Wittrup, Nature Biotechnology, 15:553-557 (June 1997)). Other critical features of the pYD vector include a galactose inducible promoter and an epitope tag, V5, on the C-terminus of the inserted anti-TnI gene (see FIG. 12).
The yeast display platform utilizes an antibody format known as the single-chain variable fragment. In the scFv format, the variable heavy domain is connected to the variable light domain through a flexible linker (variable heavy domain—Linker GPAKELTPLKEAKVS (SEQ ID NO:58)—variable light domain).
PCR single overlap extension (SOE) was used to combine the variable heavy (VH) and the variable light genes (VL) for the TnI 19C7 scFv construct (FIG. 2, and SEQ ID NOs:54 and 55). The TnI 19C7 scFv DNA was cloned into the yeast display vector pYD41 using vector restriction sites SfiI and XhoI. The pYD41-TnI 19C7scFv vector was transformed into DH5α E. coli. Plasmid DNA was then isolated from the E. coli and the TnI 19C7 scFv insert was sequenced to ensure the scFv was cloned in frame with the AGA2 protein.
The cloning site for the scFv into the yeast display vector pYD41 is in an ORF that includes the following genes: AGA2-tether linker 41-X press epitope tag-TnI 19C7 variable heavy chain-Linker 40-TnI 19C7 variable light chain-V5 epitope tag—Six His tag (SEQ ID NO: 29). In addition, the yeast strain EBY100 is a tryptophan auxotroph and the pYD41 vector encodes for tryptophan as the system's selectable marker.
Transformation into Saccharomyces cerevisiae Strain EBY100
Yeast display plasmid, pYD41-TnI 19C7 scFv, was transformed into S. cerevisiae EBY100 using the Gietz and Schiestl Method (See Schiestl and Gietz, Current Genetics, 16(5-6):339-46 (December 1989)). Dilutions of the transformation reaction were plated on selective glucose plates (2% glucose (0.67% yeast nitrogen base, 0.105% HSM-trp-ura, 1.8% bacterial agar, 18.2% sorbitol, 0.86% NaH2PO4 H2O, 1.02% Na2HPO4 7H2O)) and incubated at 30° C. for 48-72 hours. Selective glucose media was inoculated with individual colonies and grown shaking at 30° C. for 16-20 hours. Protein expression was induced in colonies by transferring 0.5 OD600 of cells/ml (1e7 cells/0.50 D/ml) to selective galactose media. Colonies were shaken at 20° C. for 16-24 hours and then analyzed by the FACS Aria flow cytometer for binding to scTnI-C-2 and anti-V5. (It should be noted that scTnI-C-2 is a linked, single-chain TnI (28-110aa)-linker-TnC (1-160aa) from Spectral Diagnostics, Toronto, Canada. ScTnI-C-2 is abbreviated as “scTnI-C” for purposes of the present discussion.) For flow cytometry assays, yeast cells expressing TnI 19C7 scFv incubated with scTnI-C-2 or anti-V5 followed by either anti-troponin mAb and goat anti mouse-phycoerythrin (GAM:PE) (FIG. 3B) or GAM:PE respectively (FIG. 3A). The flow cytometry histograms illustrate the full-length expression of TnI 19C7 scFv as detected by anti-V5 and the ability of TnI 19C7 scFv to bind to scTnI-C-2.
Off-Rate Analysis for TnI 19C7 scFv and TnI 19C7 Variants on Yeast
Off-rate measurements of TnI 19C7 scFv and TnI 19C7 variants on yeast were measured by incubating 0.05OD yeast (1×106 cells) with 50 nM scTnI-C-2 for 30-60 minutes at room temperature. Cells were then washed twice with blocking buffer containing phosphate buffered saline pH 6.8 with 2% bovine serum albumin and 0.2% Standapol ES-1 (PBS/BSA/Standapol) and incubated at room temperature for varying amounts of time (0, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4.25 hr, 25.5 hr, 50 hr 75 hr and 144 hr (see FIG. 4). At each individual time point, yeast cells were transferred to ice to halt the reaction. Cells were then washed twice with blocking buffer and suspended in the next staining reagent, specifically, anti-TnI mAb 8E10 at 100 nM. Cells were incubated on ice for 30 minutes, washed twice and then incubated with goat anti mouse-phycoerythrin (GAM:PE). Finally, the cells were washed and analyzed on the FACS Aria flow cytometer. FIG. 4 shows the off-rate data plotted as mean fluorescence units (“MFU”) versus time (in seconds). A first order decay equation was used to fit the data. The off-rate, m2 in the equation shown in FIG. 4, was fitted to 0.007 sec−1. The TnI 19C7 scFv half-life (t1/2) was approximately 8.5 min (t1/2=ln 2/koff).
An off-rate sorting strategy was used to identify off-rate improved TnI 19C7 variants from mutagenic libraries. Therefore, the TnI 19C7 scFv, unmutated or wildtype (“wt”), half-life was used to determine the appropriate time to sort the mutagenic libraries. TnI 19C7 mutagenic libraries were sorted approximately 9 min after washing cells free of scTnI-C-2 with the same assay conditions described for wt TnI 19C7 scFv.
Equilibrium Disassociation (KD) Analysis for TnI 19C7 scFv and TnI
KD measurements of TnI 19C7 scFv and TnI 19C7 variants on yeast were measured by incubating 0.05 OD yeast (1×106 cells) with varying concentrations of scTnI-C-2 for 45-60 minutes at room temperature. Blocking buffer containing phosphate buffered saline pH 6.8 with 2% bovine serum albumin and 0.2% Standapol ES-1 (PBS/BSA/Standapol) was used for washes and reagent dilutions. Cells were then washed twice and incubated for 30 min with anti-TnI mAb, 8E10. Cells were washed again and incubated with goat anti-mouse phycoerythrin for 30 min. Finally, cells were washed and analyzed on the FACS Aria flow cytometer (see FIG. 5). FIG. 5 shows the KD data plotted as normalized mean fluorescence units (“MFU”) versus concentration scTn-I-C-2 (in Molarity). The antibody-normalized, antigen-binding mean fluorescence intensity was plotted against antigen concentration and a non-linear least squares fit (y=m1+m2*m0/(m3+m0)) was used to determine KD.
Generation of TnI 19C7 Spiked CDR Libraries
Mutagenesis was directed to the three heavy and three light chain complementary determining regions (CDR) of antibody TnI 19C7 since these loops are the major antigen contact sites. CDR loop lengths and numbering were defined using Kabat and Oxford's Molecular AbM modeling nomenclature. Individual libraries were composed such that random mutations are incorporated at each amino acid position of the CDR for a single library. A total of six libraries were generated corresponding to one library per each CDR.
Libraries were generated by combining SfiI/XhoI digested. pYD41 vector and PCR products with chemically competent EBY100 yeast (see FIG. 7). Two PCR products were generated for each CDR library to allow for PCR sorting and homologous recombination into yeast. One PCR product used a primer that was designed, such that for the entire length of the CDR, a 70% wild type to 30% other nucleotide ratio was used in the primer synthesis. This product was called the spiked (sp) product (see FIG. 7). The second PCR product was designed to include the remaining portion of the scFv gene. The two PCR products were combined and used to generate a single-chain variable fragment or a scFv product. Digested vector (1 ug) and the scFv PCR products (5 ug) were combined with EBY100 yeast (5.2e8-6.4e8 cells) and transformed using electroporation. The scFv PCR product and the pYD41 digested vector cyclize during transformation due to homologous recombination facilitated by the nucleotide overlap and the mechanism of yeast endogenous gap repair. Libraries were grown at 30° C. for 48-72 hours in selective glucose media and passed again in selective glucose media prior to induction of protein expression for library sorting.
TnI 19C7 Mutagenic CDR Libraries
TnI 19C7 libraries were sorted based on an off-rate sorting strategy. TnI 19C7 CDR mutagenic libraries were induced in galactose expression media at 20° C. for 18-24 hours. TnI 19C7 scFv and TnI 19C7 libraries on yeast were incubated with 25-50 nM scTnI-C-2 for 10-15 minutes at room temperature. Cells were then washed twice with blocking buffer containing phosphate buffered saline pH 6.8 with 2% bovine serum albumin and 0.2% Standapol ES-1 (PBS/BSA/Standapol) and incubated at room temperature for 8 min. Yeast cells were transferred to ice to halt the reaction. Cells were then washed twice with blocking buffer and suspended in the next staining reagent, specifically, anti-TnI mAb 8E10 at 100 nM and anti-V5 at 1.5-2 ug/ml. Cells were incubated on ice for 30 minutes, washed twice and then incubated with 1:200 goat anti mouseIgG2a-phycoerythrin (GAMIgG2a:PE) and with 1:200 dilution goat anti mouseIgG1-Alexa Fluor488 (GAMIgG1:488). Finally, the cells were washed, analyzed, and sorted on the FACS Aria flow cytometer. Sort gates were set based on unmutated TnI 19C7 binding at 8-9 min with a gate set to sort full-length TnI binding clones. Each sort collected the top 0.1-0.5% of the TnI binding population. Sorted cells were grown in selective glucose media and grown 18-24 hours at 30° C. Sort 1 cells were induced and sorting was repeated for two or three additional rounds.
After the last sort, sorted cells were plated onto selective glucose plates and placed at 30° C. for 72 hours. Individual yeast colonies from these libraries were inoculated in selective glucose media, cryopreserved and induced in selective galactose media. Individual colonies were then characterized and ranked in an off-rate assay. TnI 19C7 AM4 was isolated and identified from this sorting output.
Generation and Analysis of TnI 19C7 Combinatorial Mutant Clones
Clones that were characterized for off-rate from each master CDR library or the total master CDR library output were used to construct scFv genes containing different pairings of the individual mutations. This approach enabled determination of whether the binding properties were further enhanced upon combining individual mutations. Combinatorial clones containing various mutations in each CDR region were constructed by PCR amplification and combined using routine techniques known to those skilled in the art. Combinatorial mutant libraries were transformed into yeast as described above and sorted two times using off-rate and KD selection pressures. For KD selection, 100 pM (round 1) and 50 pM (round 2) scTnI-C were used in the KD experiment as described above (FIG. 5). For off-rate sorting, sorting was conducted as described above with incubation times following washing away of antigen of 3 hr 40 min (round 1) and 4 hrs 25 min (round 2). Sort gates were set based on unmutated TnI 19C7 binding for each condition with a gate set to sort full-length TnI binding clones. Each sort collected the top 0.1% of the TnI binding population. Sorted cells were grown in selective glucose media for 18-24 hours at 30° C. Sort 1 cells were induced and sorting was repeated for one additional round.
After the last sort, sorted cells were plated onto selective glucose plates and placed at 30° C. for 72 hours. Individual yeast colonies from these libraries were inoculated in selective glucose media, cryopreserved and induced in selective galactose media. Individual colonies were then characterized and ranked in an off-rate assay. TnI 19C7 AM1, AM2, and AM3 were isolated and identified from this combinatorial library.
Analysis of Selected TnI 19C7 Variants
Selected clones were initially characterized for improvements in KD as described above for wild type TnI 19C7 scFv. FIG. 9 shows the scFv KD values determined for four selected clones. The TnI 19C7 AM1 clone exhibited the most improved binding at 0.36 nM compared to the wild-type TnI 19C7 antibody 1.7 nM.
Selected TnI 19C7 scFv variants were sequenced to determine the amino acid mutations being expressed. Initially, plasmid DNA was isolated from yeast suspension cultures using a yeast mini-prep kit (Cat No. D2001, Zymo Research Orange, Calif.). In order to obtain sequencing grade plasmid DNA, plasmid from the yeast mini-prep kit was transformed into DH5α E. coli, and then purified from culture using E. coli mini-prep kits (Qiagen). Pure plasmid DNA was then sequenced using pYD41 vector specific primers (pYD41 for-TAGCATGACTGGTGGACAGC (SEQ ID NO:59) and pYD41rev-CGTAGAATCGAGACCGAG (SEQ ID NO:60)) Amino acid sequence data for TnI 19C7 scFv variants is shown in FIG. 13. Position numbers refers to amino acid position in the respective CDR (as numbered using Kabat method).
Overall the source of sequence diversity from the wild-type clone was found in the CDR L2 and CDR H1, whereas CDR L1 and H3 folded into a consensus motif. The CDR L3 and CDR H2 remained unmutated. The sequence data for CDR H1 indicated a preference for a conservative change at position 34 from isoleucine to leucine as identified in the 3 clones isolated from the combinatorial library. The consensus sequence for CDR H3 indicated a strong preference at position 100a for tyrosine instead of tryptophan, at position 101 for threonine instead of alanine, and at position 102 for aspartate instead of tyrosine as identified in the 3 clones isolated from the combinatorial library. From the master CDR sorting in which clone 19C7 AM4 was identified, the CDR H3 was the only CDR with mutations that were different than the combinatorial consensus set of sequences. Specifically the mutations were Ala96Phe, Tyr99Ser, Trp100aAla, and Tyr102Asp.
The consensus sequence data for CDR L1 indicated a preference for threonine at position 25, lysine at position 27, asparagine at position 28, valine at position 29, and histidine at position 34. For the CDR L2, each clone has unique or no mutations with only TnI 19C7 AM1 and AM2 sharing only the Ser54Arg mutation.
Cloning and Soluble Expression of TnI 19C7 Chimeric Antibodies in a Transient or Stable Expression System
Selected TnI 19C7 variants were converted to chimeric mouse-mouse IgG/mouse kappa and/or mouse-human IgG1/human kappa antibodies through cloning of the TnI 19C7 variable domains into the transient expression vector system called pBOS (Abbott Bioresearch Center, Worcester, Mass.). More specifically, PCR was used to amplify the variable heavy and variable light chain genes with restriction sites for cloning into separate pBOS vectors (Mizushima and Nagata, Nucleic Acids Research, 18:5322 (1990)). The variable heavy and variable light genes were ligated in digested and dephosphorylated vector and transformed into DH5α E. coli. Plasmid DNA was purified from E. coli and transfected into 293H cells using PEI (1 mg/ml). Transient antibody was expressed for the following TnI 19C7 variants: TnI 19C7 wt, AM1, AM2, AM3 and AM4.
For example, using the pBOS-TnI 19C7 AM1 heavy and light vectors, a stable CHO cell line plasmid was created in a two-step cloning procedure. First, variable heavy chain and variable light genes were ligated in frame to the human constant genes in pBV and pJV plasmids (Abbott Bioresearch Center, Worcester, Mass.), respectively, using the restriction enzymes SrfI/NotI. Ligation reactions were transformed into DH5α E. coli and plasmid DNA was subsequently isolated from individual colonies. The pBV-TnI 19C7 mouse variable heavy-human IgG1 and pJV-TnI 19C7 mouse variable light-human kappa were sequenced at the cloning sites.
The second cloning step involved combining the heavy chain IgG1 genes and the light chain kappa genes into a single stable cell line vector. The pBV-TnI 19C7 AM1 human IgG1 and pJV-TnI 19C7 AM1 human kappa vectors were digested with AscI/PacI. The VL-human kappa constant and the VH-human IgG1 constant DNA fragments were gel purified and ligated to produce the stable cell line vector called pBJ-TnI 19C7 AM1. The pBJ-TnI 19C7 AM1 human heavy/light chimeric plasmid was transformed into CHO cells using a lipofectamine (Invitrogen) protocol. Stable cell lines were sub-cloned from initial transformation. A stable CHO cell line has been developed for the clone AM1 (also referred to as “TnI 19C7AM1 hG1kCHO204”) and was deposited with the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209 on Feb. 11, 2009 and received deposit designation PTA-9816.
Example II Relative Affinity of Troponin I Clone 19C7 Wild Type and Affinity Matured Antibodies
Troponin I clone 19C7 wild type (full mouse construct) and affinity matured (human constant region) antibodies were evaluated for relative affinity in a microtiter enzyme immunoassay. 96-well assay plates (NUNC Corporation, Rochester, N.Y.) were coated by adding 100 uL/well of a 2 ug/mL solution of either sheep anti-mouse IgG Fcγ specific antibody (Jackson ImmunoResearch, West Grove, Pa.) or donkey antihuman IgG Fcγ fragment specific antibody (Jackson ImmunoResearch). Both antibodies were diluted in phosphate buffered saline (PBS, Abbott Laboratories, Abbott Park, Ill.). The assay plates were incubated overnight at 15-30 deg C. The next day the coating reagent was removed and 200 uL/well of BSA solution (bovine serum albumin [Abbott Laboratories] diluted in PBS) was added. The BSA solution was incubated in the assay wells for 30 minutes at 15-30 deg C., removed and the assay wells washed by adding 300 uL/well distilled water (dH2O, Abbott Laboratories) and aspirating for three wash cycles. Next, 100 uL/well test samples were added. Test samples were prepared by creating an initial 2 ug/mL solution (in BSA solution) of each antibody, followed by log 3 dilutions, in BSA solution. The test samples were incubated for 2-3 hours at 15-30 deg C. after which they were aspirated away and the wells washed with dH2O as described above. Next, 100 uL/well of test antigen solutions were added to each assay well. The test antigen solutions were created by first preparing a 1000 ng/mL solution of scTnI-C-2 (aa 28-100 of cardiac troponin I linked to full length cardiac troponin C, Spectral Diagnostics) in BSA solution, followed by log 2 dilutions. The antigen solutions were incubated in the assay wells for 10 minutes at 15-30 deg C. and then removed by slapping out the solutions. The assay plates were then washed with dH2O as previously described. Next, 100 uL/well of biotin labeled goat anti-troponin I antibody (HyTest, diluted to 500 ng/mL in BSA solution) was added to each assay well and incubated for 30 minutes at 15-30 deg C. The antibody was then aspirated away and the wells washed with dH2O as described. Next, 100 uL/well of a 200 ng/mL solution (in BSA solution) of horse radish peroxidase labeled streptavidin (SA-HRPO, Jackson ImmunoResearch) was added and incubated for 30 minutes at 15-30 deg C. The SA-HRPO reagent was then aspirated away and the plates washed as described. Next, substrate solution was prepared by dissolving 1 OPD tablet per 10 mL OPD diluent (o-phenylenediamine, both Abbott Laboratories). 100 uL/well of the prepared substrate solution was added to the assay plates, incubated for about 4-5 minutes and then the reaction quenched by adding 100 uL/well 1N sulfuric acid (Abbott Laboratories). The resulting signal was read at 492 nm using an optical fluorometer. Results from the experiment were plotted using kaleidagraph software. The Ag50 value (the concentration of antigen at 50% of maximal binding) was determined and used to compare the antibodies for relative affinity to the tested antigen.
Example III Use of Monoclonal Antibody 19C7 in an Immunoassay
Troponin I clone 19C7 wild type (full mouse construct) and affinity matured (human constant region) antibodies were evaluated for relative affinity on the ARCHITECT® immunoassay analyzer (Abbott Laboratories). The assay was fully automated, and the analyzer performs all steps. Magnetic microparticles coated with a mouse anti-troponin I antibody (Abbott Laboratories, Abbott Park, Ill.) were mixed with varying levels of scTnI-C-2 (aa 28-100 of cardiac troponin I linked to full length cardiac troponin C (Spectral Diagnostics) and incubated for 18 minutes at 15-30 deg C. During this time, the microparticle coated antibody bound the scTnI-C-2. The microparticles were then attracted to a magnet, the remaining assay solution was aspirated, and the particles washed with assay diluent (Abbott Laboratories, Abbott Park, Ill.). Next, the wild type or affinity matured 19C7 antibodies, all of which were labeled with acridinium (Abbott Laboratories, Abbott Park, Ill.) were added to the microparticles and incubated for 4 minutes at 15-30 deg C. Next, the microparticles were attracted to a magnet, the remaining assay solution was aspirated, and the particles were washed with assay diluent. Signal (relative light units) was generated by the addition of pre-trigger and trigger solutions (both Abbott Laboratories). Signal ratios were calculated and used to compare antibodies. As can be established based upon the results shown in FIG. 11, AM1 antibody gave a better signal than wild-type antibody.

Claims (12)

What is claimed is:
1. A Chinese Hamster Ovary (CHO) cell line, referred to as TnI 19C7 AM1 hG1CHO 204, designated by American Type Culture Collection (ATCC) deposit Number PTA-9816.
2. An isolated nucleic acid molecule encoding a binding protein, wherein the binding protein comprises a variable heavy chain, and wherein the amino acid sequence of the variable heavy chain of said binding protein has at least 90% identity to SEQ ID NO:25 and wherein said binding protein maintains the binding activity of SEQ ID NO:25.
3. The isolated nucleic acid molecule of claim 2, wherein the encoded binding protein further comprises a variable light chain, and wherein the amino acid sequence of the variable light chain of said binding protein has at least 90% identity to SEQ ID NO:28 and wherein said binding protein maintains the binding activity of SEQ ID NO:28.
4. An isolated nucleic acid molecule encoding a binding protein, wherein the binding protein comprises a variable heavy chain, and wherein the amino acid sequence of the variable heavy chain of said binding protein is SEQ ID NO:25.
5. An isolated nucleic acid molecule encoding a binding protein, wherein the binding protein comprises a variable light chain, and wherein the amino acid sequence of the variable light chain of said binding protein is SEQ ID NO:28.
6. The isolated nucleic acid molecule of claim 5, wherein said encoded binding protein further comprises a variable heavy chain, and wherein the amino acid sequence of the variable heavy chain of said binding protein is SEQ ID NO:25.
7. A vector comprising said isolated nucleic acid molecule wherein said isolated nucleic acid molecule encodes a binding protein, wherein said binding protein comprises the variable heavy chain of claim 4 and the variable light chain of claim 5.
8. An isolated host cell comprising said vector of claim 7.
9. A method of producing a binding protein capable of binding to Troponin I, comprising culturing said host cell of claim 8 for a time and under conditions sufficient to produce said binding protein.
10. A method of producing a Troponin I binding protein comprising culturing a host cell referred to as TnI 19C7 AM1 hG1 CHO 204, designated by American Type Culture Collection (ATCC) deposit Number PTA-9816 under conditions sufficient to produce said binding protein.
11. A method of producing a Troponin I binding protein comprising culturing a host cell that comprises a nucleic acid that encodes an antibody heavy chain that comprises a CDR1 sequence of SEQ ID NO:52, a CDR2 sequence of SEQ ID NO: 53, and a CDR3 sequence of SEQ ID NO:54, and a light chain that comprises a CDR1 sequence of SEQ ID NO:55, a CDR2 sequence of SEQ ID NO: 56, and a CDR3 sequence of SEQ ID NO:57 under conditions sufficient to produce said binding protein.
12. A method of producing a Troponin I binding protein comprising culturing a host cell that comprises a nucleic acids that encode the same heavy and light chains as the Chinese Hamster ovary cell referred to as TnI 19C7 AM1 hG1 CHO 204, designated by American Type Culture Collection (ATCC) deposit Number PTA-9816 under conditions sufficient to produce said binding protein.
US14/035,420 2009-02-24 2013-09-24 Antibodies to troponin I and methods of use thereof Active USRE45763E1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/035,420 USRE45763E1 (en) 2009-02-24 2013-09-24 Antibodies to troponin I and methods of use thereof
US14/461,082 US20150024457A1 (en) 2009-02-24 2014-09-05 Antibodies to troponin i and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12/391,937 US8030026B2 (en) 2009-02-24 2009-02-24 Antibodies to troponin I and methods of use thereof
US14/035,420 USRE45763E1 (en) 2009-02-24 2013-09-24 Antibodies to troponin I and methods of use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/391,937 Reissue US8030026B2 (en) 2009-02-24 2009-02-24 Antibodies to troponin I and methods of use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/461,082 Continuation US20150024457A1 (en) 2009-02-24 2014-09-05 Antibodies to troponin i and methods of use thereof

Publications (1)

Publication Number Publication Date
USRE45763E1 true USRE45763E1 (en) 2015-10-20

Family

ID=42102721

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/391,937 Ceased US8030026B2 (en) 2009-02-24 2009-02-24 Antibodies to troponin I and methods of use thereof
US13/319,657 Abandoned US20120076803A1 (en) 2009-02-24 2010-02-23 Antibodies to troponin i and methods of use thereof
US14/035,420 Active USRE45763E1 (en) 2009-02-24 2013-09-24 Antibodies to troponin I and methods of use thereof
US14/461,082 Abandoned US20150024457A1 (en) 2009-02-24 2014-09-05 Antibodies to troponin i and methods of use thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US12/391,937 Ceased US8030026B2 (en) 2009-02-24 2009-02-24 Antibodies to troponin I and methods of use thereof
US13/319,657 Abandoned US20120076803A1 (en) 2009-02-24 2010-02-23 Antibodies to troponin i and methods of use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/461,082 Abandoned US20150024457A1 (en) 2009-02-24 2014-09-05 Antibodies to troponin i and methods of use thereof

Country Status (9)

Country Link
US (4) US8030026B2 (en)
EP (2) EP2401292B1 (en)
JP (3) JP6055593B2 (en)
CN (1) CN102421795B (en)
BR (1) BRPI1007867B8 (en)
CA (1) CA2753541C (en)
ES (1) ES2525799T3 (en)
MX (2) MX359949B (en)
WO (1) WO2010099079A1 (en)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8030026B2 (en) * 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof
EP2655415A4 (en) * 2010-12-22 2016-03-09 Abbvie Inc Tri-variable domain binding proteins and uses thereof
US8603759B2 (en) 2011-12-06 2013-12-10 Panasonic Corporation Recombinant protein capable of binding specifically and quickly to troponin I derived from human myocardium
WO2013084371A1 (en) * 2011-12-06 2013-06-13 パナソニック株式会社 Recombinant protein capable of binding specifically and rapidly to human cardiac troponin i
US8663933B2 (en) * 2011-12-16 2014-03-04 Panasonic Corporation Recombinant protein capable of binding specifically and quickly to troponin I derived from human myocardium
US8603760B2 (en) * 2011-12-20 2013-12-10 Panasonic Corporation Recombinant protein capable of binding specifically and quickly to troponin I derived from human myocardium
WO2013175678A1 (en) * 2012-05-24 2013-11-28 パナソニック株式会社 Variant protein capable of binding specifically and rapidly to human cardiac troponin i
US20160116472A1 (en) * 2013-02-04 2016-04-28 The General Hospital Corporation Biomarkers for stroke diagnosis
US9790478B2 (en) 2013-03-14 2017-10-17 Abbott Laboratories HCV NS3 recombinant antigens and mutants thereof for improved antibody detection
CA2906417C (en) 2013-03-14 2022-06-21 Robert Ziemann Hcv core lipid binding domain monoclonal antibodies
US9194873B2 (en) 2013-03-14 2015-11-24 Abbott Laboratories HCV antigen-antibody combination assay and methods and compositions for use therein
WO2014192202A1 (en) * 2013-05-29 2014-12-04 パナソニックヘルスケア株式会社 Recombinant protein capable of bonding specifically and quickly to troponin i from human myocardium
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US20190025328A1 (en) 2015-10-27 2019-01-24 Abbott Laboratories Troponin i and soluble urokinase receptor detection for determining the risk of cardiovascular disease
EP3436822B1 (en) 2016-03-31 2023-10-18 Abbott Laboratories Decision tree based systems and methods for estimating the risk of acute coronary syndrome
CN109844540A (en) 2016-04-13 2019-06-04 雅培制药有限公司 Cardiac muscle troponin I during pregnancy for cardiovascular disease identification and risk assessment detects
WO2017218911A1 (en) 2016-06-17 2017-12-21 Abbott Laboratories BIOMARKERS TO PREDICT NEW ONSET HEART FAILURE WITH PRESERVED EJECTION FRACTION (HFpEF)
EP3279667A1 (en) 2016-08-02 2018-02-07 Abbott Laboratories Cardiac troponin i and copeptin as biomarkers for short-term mortality in acute exacerbation of chronic obstructive pulmonary disease (aecopd)
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
JP2021515567A (en) * 2018-03-14 2021-06-24 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft New anti-troponin T antibody
WO2019213624A1 (en) * 2018-05-04 2019-11-07 The Regents Of The University Of California Spiked primers for enrichment of pathogen nucleic acids among background of nucleic acids
CN111018980B (en) * 2018-10-10 2021-12-03 东莞市朋志生物科技有限公司 Anti-human cardiac troponin I antibody and application thereof
CN111018976B (en) * 2018-10-10 2022-04-01 东莞市朋志生物科技有限公司 Recombinant antibody of anti-human cardiac troponin I
KR20220042367A (en) * 2019-08-09 2022-04-05 에프. 호프만-라 로슈 아게 Novel anti-troponin T antibody
WO2024044288A1 (en) 2022-08-26 2024-02-29 Abbott Laboratories Use of cardiac troponin and galectin-3 to differentiate myocardial infarction type i and type ii

Citations (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0086631A2 (en) 1982-02-11 1983-08-24 E.I. Du Pont De Nemours And Company Spinneret plate
US4526938A (en) 1982-04-22 1985-07-02 Imperial Chemical Industries Plc Continuous release formulations
EP0229246A2 (en) 1986-01-15 1987-07-22 ANT Nachrichtentechnik GmbH Method for decoding digital signals, as well as a Viterbi decoder and applications
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4880078A (en) 1987-06-29 1989-11-14 Honda Giken Kogyo Kabushiki Kaisha Exhaust muffler
WO1990001443A1 (en) 1988-08-08 1990-02-22 Iffiu Michael A Collapsible recumbent bicycle with streamlined three-part folding canopy
WO1990005370A1 (en) 1988-11-12 1990-05-17 Cambridge Capacitors Limited Electrical capacitors
WO1990005144A1 (en) 1988-11-11 1990-05-17 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
WO1990014424A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation Method for isolating receptors having a preselected specificity
WO1990014430A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation A new method for tapping the immunological repertoire
WO1990014443A1 (en) 1989-05-16 1990-11-29 Huse William D Co-expression of heteromeric receptors
US5006309A (en) 1988-04-22 1991-04-09 Abbott Laboratories Immunoassay device with liquid transfer between wells by washing
WO1991005939A1 (en) 1989-10-11 1991-05-02 Ide Russell D Progressive cavity drive train
WO1991005548A1 (en) 1989-10-10 1991-05-02 Pitman-Moore, Inc. Sustained release composition for macromolecular proteins
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
WO1991009630A1 (en) 1989-12-22 1991-07-11 Essilor International Method and solution for decontaminating a flexible contact lens, particularly of the hydrophilic type
US5063081A (en) 1988-11-14 1991-11-05 I-Stat Corporation Method of manufacturing a plurality of uniform microfabricated sensing devices having an immobilized ligand receptor
WO1991017271A1 (en) 1990-05-01 1991-11-14 Affymax Technologies N.V. Recombinant library screening methods
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5089424A (en) 1988-06-14 1992-02-18 Abbott Laboratories Method and apparatus for heterogeneous chemiluminescence assay
WO1992009690A2 (en) 1990-12-03 1992-06-11 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
US5128326A (en) 1984-12-06 1992-07-07 Biomatrix, Inc. Drug delivery systems based on hyaluronans derivatives thereof and their salts and methods of producing same
WO1992015679A1 (en) 1991-03-01 1992-09-17 Protein Engineering Corporation Improved epitode displaying phage
WO1992018619A1 (en) 1991-04-10 1992-10-29 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
WO1992019244A2 (en) 1991-05-01 1992-11-12 Henry M. Jackson Foundation For The Advancement Of Military Medicine A method for treating infectious respiratory diseases
WO1992020791A1 (en) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
WO1993001288A1 (en) 1991-07-08 1993-01-21 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Phagemide for screening antibodies
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
WO1994001234A2 (en) 1992-07-10 1994-01-20 Horst Warneke Assembly line for producing steel coffers for ceiling and/wall constructions from sheet metal plates
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
WO1996001878A1 (en) 1994-07-08 1996-01-25 The Gillette Company Aqueous correction fluids
WO1996018978A1 (en) 1994-12-14 1996-06-20 The Secretary Of State For Defence Method of authenticating watermarked paper
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1996020698A2 (en) 1995-01-05 1996-07-11 The Board Of Regents Acting For And On Behalf Of The University Of Michigan Surface-modified nanoparticles and method of making and using same
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5565352A (en) 1993-11-24 1996-10-15 Arch Development Corporation Deubiquitinating enzyme: compositions and methods
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
WO1997029131A1 (en) 1996-02-09 1997-08-14 Basf Aktiengesellschaft HUMAN ANTIBODIES THAT BIND HUMAN TNF$g(a)
WO1997032572A2 (en) 1996-03-04 1997-09-12 The Penn State Research Foundation Materials and methods for enhancing cellular internalization
US5679377A (en) 1989-11-06 1997-10-21 Alkermes Controlled Therapeutics, Inc. Protein microspheres and methods of using them
WO1997044013A1 (en) 1996-05-24 1997-11-27 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5705330A (en) 1995-04-14 1998-01-06 Abbott Laboratories Chemiluminescent immunoassay for antibody detection
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5714352A (en) 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
US5723323A (en) 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
WO1998016280A1 (en) 1996-10-16 1998-04-23 Decathlon Sa Roller skate with reinforcing leggings and in particular for braking
US5763192A (en) 1986-11-20 1998-06-09 Ixsys, Incorporated Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
WO1998031346A1 (en) 1997-01-16 1998-07-23 Massachusetts Institute Of Technology Preparation of particles for inhalation
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5855913A (en) 1997-01-16 1999-01-05 Massachusetts Instite Of Technology Particles incorporating surfactants for pulmonary drug delivery
WO1999015154A1 (en) 1997-09-24 1999-04-01 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release preparations
WO1999020253A1 (en) 1997-10-23 1999-04-29 Bioglan Therapeutics Ab Encapsulation method
US5912120A (en) 1992-04-09 1999-06-15 The United States Of America As Represented By The Department Of Health And Human Services, Cloning, expression and diagnosis of human cytochrome P450 2C19: the principal determinant of s-mephenytoin metabolism
US5912015A (en) 1992-03-12 1999-06-15 Alkermes Controlled Therapeutics, Inc. Modulated release from biocompatible polymers
US5916597A (en) 1995-08-31 1999-06-29 Alkermes Controlled Therapeutics, Inc. Composition and method using solid-phase particles for sustained in vivo release of a biologically active agent
US5934272A (en) 1993-01-29 1999-08-10 Aradigm Corporation Device and method of creating aerosolized mist of respiratory drug
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US5985309A (en) 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
FR2779526A1 (en) 1998-06-09 1999-12-10 Pasteur Sanofi Diagnostics NEW METHOD FOR DETERMINING THE CARDIAC TROPONIN I
WO1999066903A2 (en) 1998-06-24 1999-12-29 Advanced Inhalation Research, Inc. Large porous particles emitted from an inhaler
US6019968A (en) 1995-04-14 2000-02-01 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
US6194222B1 (en) * 1998-01-05 2001-02-27 Biosite Diagnostics, Inc. Methods for monitoring the status of assays and immunoassays
US6204023B1 (en) 1985-11-01 2001-03-20 Xoma Ltd. Modular assembly of antibody genes, antibodies prepared thereby and use
WO2001083525A2 (en) 2000-05-03 2001-11-08 Amgen Inc. Modified peptides, comprising an fc domain, as therapeutic agents
EP1176195A1 (en) 1999-04-09 2002-01-30 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
WO2002072636A2 (en) 2000-12-28 2002-09-19 Altus Biologics Inc. Crystals of whole antibodies and fragments thereof and methods for making and using them
US20020137134A1 (en) 2000-06-28 2002-09-26 Gerngross Tillman U. Methods for producing modified glycoproteins
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US20030170881A1 (en) 2002-03-05 2003-09-11 I-Stat Corporation Apparatus and methods for analyte measurement and immuno assay
US20030186374A1 (en) 2001-10-01 2003-10-02 Hufton Simon E. Multi-chain eukaryotic display vectors and uses thereof
US20040018590A1 (en) 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20040018577A1 (en) 2002-07-29 2004-01-29 Emerson Campbell John Lewis Multiple hybrid immunoassay
US6699658B1 (en) 1996-05-31 2004-03-02 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
WO2005000901A2 (en) 2003-05-09 2005-01-06 Duke University Cd20-specific antibodies and methods of employing same
US20050054078A1 (en) 2003-09-10 2005-03-10 Miller Cary James Immunoassay device with improved sample closure
WO2005100584A2 (en) 2004-04-15 2005-10-27 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
US20060018897A1 (en) 2004-06-28 2006-01-26 Transtarget Inc. Bispecific antibodies
US20060160164A1 (en) 2003-09-10 2006-07-20 Miller Cary J Immunoassay device with immuno-reference electrode
US7285418B2 (en) 1996-06-25 2007-10-23 Hytest Ltd. Method and kit for the diagnosis of troponin I
US7371383B2 (en) * 2002-04-12 2008-05-13 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
EP2014302A1 (en) 2007-07-12 2009-01-14 Institut Curie An antibody specific for the Tn antigen for the treatment of cancer
US8030026B2 (en) 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8901334D0 (en) 1989-01-21 1989-03-15 Oakland Design Products Ltd Improvements relating to broadhead arrows
US5006209A (en) * 1990-02-13 1991-04-09 Electrochemical Technology Corp. Electrolytic reduction of alumina
GB9101134D0 (en) 1991-01-18 1991-02-27 R B S Improvements in and relating to accommodation for animals
GB9201755D0 (en) 1992-01-28 1992-03-11 British Bio Technology Compounds
CA2615918C (en) * 1997-02-21 2013-11-26 Genentech, Inc. Antibody fragment-polymer conjugates and humanized anti-il-8 monoclonal antibodies
JPH10324699A (en) * 1997-03-21 1998-12-08 Sankyo Co Ltd Anti-human fas humanized antibody
ZA982371B (en) * 1997-03-21 1998-09-28 Sankyo Co Humanized anti-human fas antibody
MXPA01009645A (en) * 1999-03-25 2003-09-04 Abbott Gmbh & Co Kg Human antibodies that bind human il-12 and methods for producing.
CN1247615C (en) * 2000-03-21 2006-03-29 上海润东生物科技有限公司 Purification of human myocardium troponin I and preparation method of its monoclonal anti-body
CN1644685A (en) * 2004-12-23 2005-07-27 复旦大学 Monoclone antibody against human cardiac troponin I and its use
CN1982337A (en) * 2005-12-14 2007-06-20 中国医学科学院放射医学研究所 Synthesis of polypeptide antibody from anti-human myocardial troponin I, its production and use
JP5470817B2 (en) 2008-03-10 2014-04-16 日産自動車株式会社 Battery electrode, battery using the same, and manufacturing method thereof
JP6136279B2 (en) 2013-01-15 2017-05-31 株式会社ジェイテクト Rolling bearing device
TWI503850B (en) 2013-03-22 2015-10-11 Polytronics Technology Corp Over-current protection device
TWI510996B (en) 2013-10-03 2015-12-01 Acer Inc Methods for controlling a touch panel and portable computers using the same
US9816280B1 (en) 2016-11-02 2017-11-14 Matthew Reitnauer Portable floor

Patent Citations (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0086631A2 (en) 1982-02-11 1983-08-24 E.I. Du Pont De Nemours And Company Spinneret plate
US4526938A (en) 1982-04-22 1985-07-02 Imperial Chemical Industries Plc Continuous release formulations
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5128326A (en) 1984-12-06 1992-07-07 Biomatrix, Inc. Drug delivery systems based on hyaluronans derivatives thereof and their salts and methods of producing same
US5723323A (en) 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US5817483A (en) 1985-03-30 1998-10-06 Stuart Kauffman Process for the production of stochastically-generated peptides,polypeptides or proteins having a predetermined property
US5976862A (en) 1985-03-30 1999-11-02 Ixsys Corporation Process for obtaining DNA, RNA, peptides, polypeptides, or proteins, by recombinant DNA technique
US5814476A (en) 1985-03-30 1998-09-29 Stuart Kauffman Process for the production of stochastically-generated transcription or translation products
US5824514A (en) 1985-03-30 1998-10-20 Stuart A. Kauffman Process for the production of expression vectors comprising at least one stochastic sequence of polynucleotides
US6204023B1 (en) 1985-11-01 2001-03-20 Xoma Ltd. Modular assembly of antibody genes, antibodies prepared thereby and use
EP0229246A2 (en) 1986-01-15 1987-07-22 ANT Nachrichtentechnik GmbH Method for decoding digital signals, as well as a Viterbi decoder and applications
EP0239400B1 (en) 1986-03-27 1994-08-03 Medical Research Council Recombinant antibodies and methods for their production
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5763192A (en) 1986-11-20 1998-06-09 Ixsys, Incorporated Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US4880078A (en) 1987-06-29 1989-11-14 Honda Giken Kogyo Kabushiki Kaisha Exhaust muffler
US5006309A (en) 1988-04-22 1991-04-09 Abbott Laboratories Immunoassay device with liquid transfer between wells by washing
US5089424A (en) 1988-06-14 1992-02-18 Abbott Laboratories Method and apparatus for heterogeneous chemiluminescence assay
WO1990001443A1 (en) 1988-08-08 1990-02-22 Iffiu Michael A Collapsible recumbent bicycle with streamlined three-part folding canopy
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
WO1990005144A1 (en) 1988-11-11 1990-05-17 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
WO1990005370A1 (en) 1988-11-12 1990-05-17 Cambridge Capacitors Limited Electrical capacitors
US5063081A (en) 1988-11-14 1991-11-05 I-Stat Corporation Method of manufacturing a plurality of uniform microfabricated sensing devices having an immobilized ligand receptor
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
WO1990014443A1 (en) 1989-05-16 1990-11-29 Huse William D Co-expression of heteromeric receptors
WO1990014424A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation Method for isolating receptors having a preselected specificity
WO1990014430A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation A new method for tapping the immunological repertoire
WO1991005548A1 (en) 1989-10-10 1991-05-02 Pitman-Moore, Inc. Sustained release composition for macromolecular proteins
WO1991005939A1 (en) 1989-10-11 1991-05-02 Ide Russell D Progressive cavity drive train
US5679377A (en) 1989-11-06 1997-10-21 Alkermes Controlled Therapeutics, Inc. Protein microspheres and methods of using them
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
WO1991009630A1 (en) 1989-12-22 1991-07-11 Essilor International Method and solution for decontaminating a flexible contact lens, particularly of the hydrophilic type
WO1991017271A1 (en) 1990-05-01 1991-11-14 Affymax Technologies N.V. Recombinant library screening methods
WO1992020791A1 (en) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1992009690A2 (en) 1990-12-03 1992-06-11 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
WO1992015679A1 (en) 1991-03-01 1992-09-17 Protein Engineering Corporation Improved epitode displaying phage
WO1992018619A1 (en) 1991-04-10 1992-10-29 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
WO1992019244A2 (en) 1991-05-01 1992-11-12 Henry M. Jackson Foundation For The Advancement Of Military Medicine A method for treating infectious respiratory diseases
US5290540A (en) 1991-05-01 1994-03-01 Henry M. Jackson Foundation For The Advancement Of Military Medicine Method for treating infectious respiratory diseases
EP0519596B1 (en) 1991-05-17 2005-02-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
WO1993001288A1 (en) 1991-07-08 1993-01-21 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Phagemide for screening antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US6350861B1 (en) 1992-03-09 2002-02-26 Protein Design Labs, Inc. Antibodies with increased binding affinity
US5912015A (en) 1992-03-12 1999-06-15 Alkermes Controlled Therapeutics, Inc. Modulated release from biocompatible polymers
US5912120A (en) 1992-04-09 1999-06-15 The United States Of America As Represented By The Department Of Health And Human Services, Cloning, expression and diagnosis of human cytochrome P450 2C19: the principal determinant of s-mephenytoin metabolism
WO1994001234A2 (en) 1992-07-10 1994-01-20 Horst Warneke Assembly line for producing steel coffers for ceiling and/wall constructions from sheet metal plates
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
US5934272A (en) 1993-01-29 1999-08-10 Aradigm Corporation Device and method of creating aerosolized mist of respiratory drug
US5565352A (en) 1993-11-24 1996-10-15 Arch Development Corporation Deubiquitinating enzyme: compositions and methods
WO1996001878A1 (en) 1994-07-08 1996-01-25 The Gillette Company Aqueous correction fluids
WO1996018978A1 (en) 1994-12-14 1996-06-20 The Secretary Of State For Defence Method of authenticating watermarked paper
WO1996020698A2 (en) 1995-01-05 1996-07-11 The Board Of Regents Acting For And On Behalf Of The University Of Michigan Surface-modified nanoparticles and method of making and using same
US6019968A (en) 1995-04-14 2000-02-01 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
US5705330A (en) 1995-04-14 1998-01-06 Abbott Laboratories Chemiluminescent immunoassay for antibody detection
US5916597A (en) 1995-08-31 1999-06-29 Alkermes Controlled Therapeutics, Inc. Composition and method using solid-phase particles for sustained in vivo release of a biologically active agent
WO1997029131A1 (en) 1996-02-09 1997-08-14 Basf Aktiengesellschaft HUMAN ANTIBODIES THAT BIND HUMAN TNF$g(a)
US5985320A (en) 1996-03-04 1999-11-16 The Penn State Research Foundation Materials and methods for enhancing cellular internalization
WO1997032572A2 (en) 1996-03-04 1997-09-12 The Penn State Research Foundation Materials and methods for enhancing cellular internalization
US5714352A (en) 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
US5985309A (en) 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
WO1997044013A1 (en) 1996-05-24 1997-11-27 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6699658B1 (en) 1996-05-31 2004-03-02 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US7285418B2 (en) 1996-06-25 2007-10-23 Hytest Ltd. Method and kit for the diagnosis of troponin I
WO1998016280A1 (en) 1996-10-16 1998-04-23 Decathlon Sa Roller skate with reinforcing leggings and in particular for braking
US5855913A (en) 1997-01-16 1999-01-05 Massachusetts Instite Of Technology Particles incorporating surfactants for pulmonary drug delivery
WO1998031346A1 (en) 1997-01-16 1998-07-23 Massachusetts Institute Of Technology Preparation of particles for inhalation
WO1999015154A1 (en) 1997-09-24 1999-04-01 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release preparations
US5989463A (en) 1997-09-24 1999-11-23 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release devices
WO1999020253A1 (en) 1997-10-23 1999-04-29 Bioglan Therapeutics Ab Encapsulation method
US6194222B1 (en) * 1998-01-05 2001-02-27 Biosite Diagnostics, Inc. Methods for monitoring the status of assays and immunoassays
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
FR2779526A1 (en) 1998-06-09 1999-12-10 Pasteur Sanofi Diagnostics NEW METHOD FOR DETERMINING THE CARDIAC TROPONIN I
WO1999066903A2 (en) 1998-06-24 1999-12-29 Advanced Inhalation Research, Inc. Large porous particles emitted from an inhaler
EP1176195A1 (en) 1999-04-09 2002-01-30 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
WO2001083525A2 (en) 2000-05-03 2001-11-08 Amgen Inc. Modified peptides, comprising an fc domain, as therapeutic agents
US20040018590A1 (en) 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20020137134A1 (en) 2000-06-28 2002-09-26 Gerngross Tillman U. Methods for producing modified glycoproteins
WO2002072636A2 (en) 2000-12-28 2002-09-19 Altus Biologics Inc. Crystals of whole antibodies and fragments thereof and methods for making and using them
WO2002072636A3 (en) 2000-12-28 2003-04-17 Altus Biologics Inc Crystals of whole antibodies and fragments thereof and methods for making and using them
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
WO2003016466A3 (en) 2001-08-17 2003-10-23 Lilly Co Eli ANTI-Aβ ANTIBODIES
US20030186374A1 (en) 2001-10-01 2003-10-02 Hufton Simon E. Multi-chain eukaryotic display vectors and uses thereof
WO2003035835A3 (en) 2001-10-25 2003-10-16 Genentech Inc Glycoprotein compositions
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US20030170881A1 (en) 2002-03-05 2003-09-11 I-Stat Corporation Apparatus and methods for analyte measurement and immuno assay
US7371383B2 (en) * 2002-04-12 2008-05-13 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
US20040018577A1 (en) 2002-07-29 2004-01-29 Emerson Campbell John Lewis Multiple hybrid immunoassay
WO2005000901A3 (en) 2003-05-09 2005-06-23 Univ Duke Cd20-specific antibodies and methods of employing same
WO2005000901A2 (en) 2003-05-09 2005-01-06 Duke University Cd20-specific antibodies and methods of employing same
US20060160164A1 (en) 2003-09-10 2006-07-20 Miller Cary J Immunoassay device with immuno-reference electrode
US20050054078A1 (en) 2003-09-10 2005-03-10 Miller Cary James Immunoassay device with improved sample closure
WO2005100584A2 (en) 2004-04-15 2005-10-27 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
WO2005100584A3 (en) 2004-04-15 2006-12-21 Glycofi Inc Production of galactosylated glycoproteins in lower eukaryotes
US20060018897A1 (en) 2004-06-28 2006-01-26 Transtarget Inc. Bispecific antibodies
EP2014302A1 (en) 2007-07-12 2009-01-14 Institut Curie An antibody specific for the Tn antigen for the treatment of cancer
US8030026B2 (en) 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof

Non-Patent Citations (123)

* Cited by examiner, † Cited by third party
Title
Altshchul, et al., Nucleic Acids Research, vol. 25 pp. 3389-3402 (1997).
Azzazy, H., et al., Clin. Biochem., vol. 35 pp. 425-445 (2002).
Barbas, et al., PNAS, vol. 88 pp. 7978-7982 (1991).
Bird, et al., Science, vol. 242 pp. 423-426 (1988).
Bodar, et al., Clinical Chemistry, vol. 38 pp. 2203-2214 (1992).
Boder, et al., Nature Biotechnology, vol. 15 pp. 553-557 (Jun. 1997).
Buchwald, et al., Surgery, vol. 88 p. 507 (1980).
Carter, et al., Proc. Natl. Acad. Sci., vol. 89 p. 4285, USA (1992).
Chothia, et al., J. Mol. Biol., vol. 196 pp. 901-917 (1987).
Chothia, et al., J. Mol. Biol., vol. 227 p. 799 (1992).
Chothia, et al., Nature, vol. 342 pp. 877-883 (1989).
Clackson, et al., Nature, vol. 352 pp. 624-628 (1991).
Cleek, et al., "Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application," Pro. Int'l. Symp. Control. Rel. Bioact. Mater., vol. 24 pp. 853-854 (1997).
Co, M.S., et al., Mol. Immunol., vol. 30 pp. 1361-1367 (1993).
Colman P. M. Research in Immunology, 145:33-36, 1994. *
Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds), Wiley, New York (1984).
Cummins, et al., Am. Heart Journal, vol. 113 pp. 1333-1344 (1987).
During, et al., Ann. Neurol., vol. 25 p. 351 (1989).
Eriksson et al. (2005) "Comparison of Cardiac Troponin I Immunoassays Variably Affected by Circulating Autoantibodies," Clin. Chem. 51(5):848-855.
Filatov V. L., et al., "Epitope Mapping of Anti-Troponin I Monoclonal Antibodies, XP000941112," Biochemistry and Molecular Biology International, 36039, 45 (6), Academic Press, London, GB, 1179-1187.
Foote, et al., J. Mol. Biol., vol. 224 pp. 487-499 (1992).
Fuchs, et al., Bio/Technology, vol. 9 pp. 1370-1372 (1991).
Garrad, et al., Bio/Technology, vol. 9 pp. 1373-1377 (1991).
Gavilondo, J.V., et al., BioTechniques, vol. 29 pp. 128-145 (2000).
Giege, R., et al., Crystallization of Nucleic Acids and Proteins , a Practical Approach, 2.sup.nd Edition, pp. 20 1-16, Oxford University Press, New York, New York (1999).
Giege, R., et al., Crystallization of Nucleic Acids and Proteins , a Practical Approach, 2nd Edition, pp. 20 1-16, Oxford University Press, New York, New York (1999).
Gillies, et al., J. Immunol. Methods, vol. 125 pp. 191-202 (1989).
Goodson, et al., Medical Applications of Controlled Release, supra, vol. 2 pp. 115-138 (1984).
Gram, et al., PNAS, vol. 89 pp. 3576-3580 (1992).
Griffiths, et al., EMBO J., vol. 12 pp. 725-734 (1993).
Hawkins, et al., J. Mol. Biol., vol. 226 pp. 889-896 (1992).
Hay, et al., Hum. Antibod. Hybridomas, vol. 3 pp. 81-85 (1992).
Higgins, et al., CABIOS, 5L151-5L153 (1989).
Holliger, P., et al., Proc. Natl. Acad. Sci., vol. 90 pp. 6444-6448, USA (1993).
Hoogenboom, et al., Nuc. Acid Res., vol. 19 pp. 4133-4137 (1991).
Hoogenboom, H., et al., Immunology Today, vol. 21 pp. 371-378 (2000).
Hoogenboom, H.R., et al., TIB Tech., vol. 15 pp. 62-70 (1997).
Howard, et al., J. Neurosurg., vol. 71 p. 105 (1989).
Huse, et al., Science, vol. 246 pp. 1275-1281 (1989).
Huston, et al., Proc. Natl. Acad. Sci., vol. 85 pp. 5879-5883, USA (1988).
Hytest 1999 Product Catalog.
Hytest 2000 General Product Catalog.
Hytest 2001 Cardiac Markers Panel.
Hytest 2001-2002 General Product Catalog.
Hytest 2003 General Product Catalog.
Hytest 2004 Cardiac Markers Panel.
Hytest 2004-2005 General Product Catalog.
Hytest 2005 Markers of Cardiovascular Diseases Catalog.
Hytest 2005-2006 General Product Catalog.
Hytest 2006-2007 General Product Catalog.
Hytest 2007 Markers of Cardiovascular Diseases Catalog.
Hytest 2007-2008 General Product Catalog.
Hytest 2008 Markers of Cardiovascular Diseases and Metabolic Syndrome.
Hytest 2008-2009 General Product Catalog.
Jefferis, R., et al., Biotechnol. Prog., vol. 21 pp. 11-16 (2005).
Johnsson, B., et al., Anal. Biochem., vol. 198 pp. 268-277 (1991).
Johnsson, B., et al., J. Mol. Recognit., vol. 8 pp. 125-131 (1995).
Jones, et al., Nature, vol. 321 pp. 522 (1986).
Jonsson, U., et al. Ann. Biol. Clin. 51: 19-26 (1993).
Jönsson, U., et al., Biotechniques, vol. 11 pp. 620-627 (1991).
Kabat, et al., Ann. NY Acad. Sci., vol. 190 pp. 382-391 (1971).
Kabat, et al., Sequences of Proteins of Immunological Interest, 5.sup.th Edition, Department of Health and Human Services, NIH Publication No. 91/3242, USA (1991).
Kabat, et al., Sequences of Proteins of Immunological Interest, National Institutes of Health, Bethesda, Maryland (1987) and (1991).
Katrukha (2006) "Troponin I measurement: the concept of a precise immunoassay," Clin. Lab. Internat. 30(5):14-16.
Katrukha et al. (1998) "Degradation of cardiac troponin I: implication for reliable immunodetection," Clin. Chem. 44 (12):2433-2440.
Kaufman, R.J., et al., Mol. Biol., vol. 159 pp. 601-621 (1982).
Kellermann, S.A., et al., Current Opinion in Biotechnology, vol. 13 pp. 593-597 (2002).
Kipriyanov, S.M., et al., Human Antibodies and Hybridomas, vol. 6 pp. 93-101 (1995).
Kipriyanov, S.M., et al., Mol. Immunol., vol. 31 pp. 1047-1058 (1994).
Kontermann, Antibody Engineering, p. 790, Springer-Verlag, New York (2001).
Lam, et al., "Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery," Proc. Int'l Symp. Control Rel. Bioact. Mater., vol. 24 pp. 759-760 (1997).
Langer, et al,. J. Macromol. Sci. Rev. Macromol. Chem., vol. 23 p. 61 (1983).
Langer, et al., Science, vol. 249 pp. 1527-1533 (1990).
Langer, supra, Sefton, et al., CRC Crit. Ref. Biomed. Eng., vol. 14 p. 20 (1987).
Levy, et al., Science, vol. 228 p. 190 (1985).
Little, M., et al., Immunology Today, vol. 21 pp. 364-370 (2000).
MacCallum, et al., J. Mol. Biol., vol. 262 (5) pp. 732-745 (1996).
Marchalonis, et al., Adv. Exp. Med. Biol., vol. 484 pp. 13-30 (2001).
McCafferty, et al., Nature, vol. 348 pp. 552-554 (1990).
Medical Applications of Controlled Release, Langer and Wise (eds), CRC Press, Boca Raton, Florida (1974).
Mizushima, et al, Nucleic Acids Research, vol. 18 p. 5322 (1990).
Morrison, et al., Proc. Natl. Acad. Sci., vol. 81 pp. 851-855 (1984).
Morrison, et al., Science, vol. 229 p. 1202 (1985).
Needleman, et al., J. Mol. Biol., vol. 48 p. 443 (1970).
Neuberger, et al., Nature, vol. 312 pp. 604-608 (1984).
Ning, et al., "Intratumoral Radioimmunotherapy of a Human Colon Cancer Xenograft Using a Sustained-Released Gel," Radiotherapy and Oncology, vol. 39 pp. 179-189 (1996).
Oi, et al., BioTechniques, vol. 4 p. 214 (1986).
Padlan, et al., Faseb J., vol. 9 pp. 133-139 (1995).
Padlan, et al., Molecular Immunology, vol. 28 (4/5) pp. 489-498 (1991).
Paul, Fundamental Immunology, 3rd Edition, 1993, pp. 292-295, under the heading "Fv Structure and Diversity in Three Dimensions". *
PCT International Search Report and Written Opinion of International Application No. PCT/US2010/024979 mailed on May 6, 2010, 21 pages.
Pearson, et al., Proc. Natl. Acad. Sci. vol. 85 p. 2444, USA, (1988).
Peronnet, et al., "Isoelectric point determination of cardiac troponin I forms present 1n plasma from patients with myocardial Infarction, NL LNKDD0I: 10.1016/J.CCA.2006.10.006, XP005805375," Clinica Chimica Acta, 39066, 377 (1-2), Elsevier BV, Amsterdam, 243-247.
Poljak, R.J., et al., Structure, vol. 2 pp. 1121-1123 (1994).
Presta, et al., J. Immunol., vol. 151 p. 2623 (1993).
Quinn, F., et al., The Immunoassay Handbook, 2.sup.nd Edition, pp. 363-367 (2001). Riechmann, et al., Nature, vol. 332 p. 323 (1988).
Rama D., et al., "Epitope Localization of Monoclonal Antibodies Used in Human Troponin I Immunoenzymometric Assay, XP009033257," Hybridoma, 35431, 16 (2), Liebert, New York, NY, US, 153-157.
Reverse Translate a Protein (www.vivo.colostae.edu/molkit/translate/index.html), Last updated on Jun. 20, 1998 (Exhibit AA).
Riechmann, et al., Nature, vol. 332 p. 323 (1988).
Roguska, et al., PNAS, vol. 91 pp. 969-973 (1994).
Rudikoff et al Proc. Natl. Acad. Sci.USA, 76(6):1979-1983, Mar. 1982. *
Rudikoff et al Proc. Natl. Acad. Sci.USA, 79(6):1979-1983, Mar. 1982.
Sambrook, et al., A Laboratory Manual, Molecular Cloning, 2.sup.nd Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989).
Saudek, et al., N. Engl. J. Med., vol. 321 p. 574 (1989).
Schiestl, et al., Current Genetics, vol. 16 (5-6) pp. 339-346 (Dec. 1989).
Shapiro, et al., Crit. Rev. Immunol., vol. 22 (3) pp. 183-200 (2002).
Shields, R.L., et al,. J. Biol. Chem., vol. 277 pp. 26733-26740 (2002).
Sims, et al., J. Immunol., vol. 151 p. 2296 (1993).
Smith, et al., Appl. Math., vol. 2 p. 482 (1981).
Song, et al., "Antibody Mediated Lung Targeting of Long-Circulating Emulsions," PDA Journal of Pharmaceutical Science and Technology, vol. 50 pp. 372-397 (1995).
Studnicka, et al., Protein Engineering, vol. 7 (6) pp. 805-814 (1994).
Table showing Codon-amino acid Abbreviations (www.hgmd.cf.ac.uk/docs/cd-amino.html) (Exhibit BB).
Takeda, et al., Nature, vol. 314 pp. 452-454 (1985).
Taylor, L.D., et al., Nucl. Acids Res., vol. 20 pp. 6287-6295 (1992).
Umana, et al., Nat. Biotech., vol. 17 pp. 176-181 (1999).
Urlaub, et al., Proc. Natl. Acad. Sci., vol. 77 pp. 4216-4220, USA (1980).
Verhoeyen, et al., Science, vol. 239 p. 1534 (1988).
Wallick, S.C., et al., Exp. Med., vol. 168 pp. 1099-1109 (1988).
Ward, et al., Nature, vol. 341 pp. 544-546 (1989).
Winnaker, et al., From Genes to Cones, Verlagsgesellschaft, Weinheim, Germany (1987).
Wright, A,. et al., EMBO J., vol. 10 pp. 2717-2723 (1991).
Wu, et al., J. Biol. Chem., vol. 262 pp. 4429-4432 (1987).
Ylikotila et al. (2006) "Utilization of Recombinant Fab Fragments in a cTnl Immunoassay Conducted in Spot Wells," Clin. Biochem. 39:843-850.

Also Published As

Publication number Publication date
EP2853540A1 (en) 2015-04-01
MX2011008938A (en) 2012-04-02
CN102421795A (en) 2012-04-18
CN102421795B (en) 2015-11-25
WO2010099079A1 (en) 2010-09-02
US20100216720A1 (en) 2010-08-26
JP2012518418A (en) 2012-08-16
EP2401292B1 (en) 2014-10-29
CA2753541A1 (en) 2010-09-02
US20120076803A1 (en) 2012-03-29
JP6199935B2 (en) 2017-09-20
CA2753541C (en) 2018-07-17
BRPI1007867A2 (en) 2018-03-06
BRPI1007867B8 (en) 2021-05-25
US20150024457A1 (en) 2015-01-22
JP2018038392A (en) 2018-03-15
EP2401292A1 (en) 2012-01-04
JP6055593B2 (en) 2016-12-27
JP2016039804A (en) 2016-03-24
ES2525799T3 (en) 2014-12-30
MX359949B (en) 2018-10-17
US8030026B2 (en) 2011-10-04
BRPI1007867B1 (en) 2021-01-26

Similar Documents

Publication Publication Date Title
USRE45763E1 (en) Antibodies to troponin I and methods of use thereof
JP6948415B2 (en) Anti-GP73 monoclonal antibody and how to obtain it
US20190359725A1 (en) Anti-cd40 antibodies and uses thereof
CN105037543B (en) Therapeutic DLL4 binding proteins
US8283162B2 (en) Antibodies relating to PIVKAII and uses thereof
KR101640520B1 (en) Monoclonal antibodies to progastrin and their uses
US11866487B2 (en) Filamin A binding proteins and uses thereof
US20140024050A1 (en) Antibodies which detect pivkaii and methods of use thereof
US20200024335A1 (en) Filamin b binding proteins and uses thereof
US20130217145A1 (en) Antibodies which detect pivkaii and methods of use thereof
WO2014039372A1 (en) Antibodies which detect pivkaii and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BROPHY, SUSAN E.;TU, BAILIN;HUANG, DAGANG;AND OTHERS;SIGNING DATES FROM 20150608 TO 20150610;REEL/FRAME:035886/0790

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12