WO1995023813A1 - In vitro antibody affinity maturation using alanine scanning mutagenesis - Google Patents

In vitro antibody affinity maturation using alanine scanning mutagenesis Download PDF

Info

Publication number
WO1995023813A1
WO1995023813A1 PCT/US1995/002492 US9502492W WO9523813A1 WO 1995023813 A1 WO1995023813 A1 WO 1995023813A1 US 9502492 W US9502492 W US 9502492W WO 9523813 A1 WO9523813 A1 WO 9523813A1
Authority
WO
WIPO (PCT)
Prior art keywords
modified
antibody
antibodies
alanine
dna
Prior art date
Application number
PCT/US1995/002492
Other languages
French (fr)
Inventor
Craig M. Lewis
Steven W. Ludmerer
Gregory F. Hollis
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to JP7522980A priority Critical patent/JPH09509835A/en
Priority to EP95911994A priority patent/EP0748338A4/en
Publication of WO1995023813A1 publication Critical patent/WO1995023813A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • C07K16/1045Lentiviridae, e.g. HIV, FIV, SIV
    • C07K16/1063Lentiviridae, e.g. HIV, FIV, SIV env, e.g. gp41, gp110/120, gp160, V3, PND, CD4 binding site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • a method of mutagenizing antibodies to produce 0 modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided.
  • the method of the invention is a systematic means to achieve in vitro antibody maturation and uses alanine scanning mutagenesis.
  • the 5 invention is particularly exemplified with a set of single chain Fv (scFv) antibodies obtained by this technique.
  • the resulting antibodies are directed against the V3 loop of HIV gpl20, and show altered off-rates against the antigen compared to the starting antibody.
  • the altered antibodies which 0 show improved (slower) off-rates to the antigen. Observed improvements have been as high as eleven-fold over wild-type.
  • a method of mutagenizing antibodies to produce 5 modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided.
  • FIG. 1 Alanine-Scanning Mutagenesis. Each of the 27 amino acids in VH CDR3 of scFv P5Q was converted to alanine by site-directed mutagenesis. E. coli clones were induced to express scFv with IPTG. Single chain Fv, which is targeted to the periplasmic space by the fd phage gene3 signal sequence, was extracted with EDTA. Periplasmic extracts were analyzed by BIAcoreTM, which measures antibody-antigen affinity by surface plasmon resonance (Fagerstam, 1991), and off -rates determined against an HIV gpl20 V3 loop peptide.
  • FIG. 1 Amino Acid Randomization: Position 107. Arginine at position 107 was mutated to all amino acids by site- directed mutagenesis. Single chain Fv extracts were analyzed by BIAcore. Percent change in off-rates is shown relative to P5Q.
  • FIG. 1 Amino Acid Randomization: Position 111. Glutamic acid at position 111 was mutated to all amino acids by site- directed mutagenesis. Single chain Fv extracts were analyzed by BIAcore. Percent change in off-rates is shown relative to P5Q.
  • HTV-1 human immunodeficiency virus-1
  • the gpl20 V3 domain of human immunodeficiency virus-1 is a disulfide-linked closed loop of approximately 30 amino acids.
  • the loop in either native or synthetic form, binds to and elicits anti-HIV-1 antibodies.
  • the present invention relates to modified antibodies and methods of making modified.
  • the invention is exemplified with modified HIV-1 immunoglobulins and methods of making these modified HTV-1 immunoglobulins.
  • the modified immunoglobulins of the present invention contain an altered complementary determining region 3 (CDR3) of HTV-l neutralizing antibody.
  • the present invention also comprises a method of treating of preventing infection through the administration of a modified antibody to a suitable host.
  • a method of treating of preventing infection through the administration of a modified antibody to a suitable host In one embodiment of the invention, the treatment or prevention of HIV infection through the administration of the modified HTV-l immunoglobulm is described.
  • the present invention also comprises diagnostic kits useful for the detection or characterization of an antigen.
  • Reagents for the kits may include DNA molecules encoding the modified antibodies or the modified antibodies or combinations thereof.
  • a method of mutagenizing antibodies to produce modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided.
  • the method of the invention is a systematic means to achieve in vitro antibody maturation and uses alanine scanning mutagenesis.
  • the invention is particularly exemplified with a set of single chain Fv (scFv) antibodies obtained by this technique.
  • the resulting antibodies are directed against the V3 loop of HIV gpl20, and show altered off-rates against the antigen compared to the starting antibody.
  • the altered antibodies which show improved (slower) off-rates to the antigen. Observed improvements have been as high as eleven-fold over wild-type.
  • CDR3 complementary determining region 3
  • the underlying principle of the method is the physical and chemical neutrality of alanine.
  • Alanine is substituted throughout a stretch of amino acids, and its effects on binding (such as off -rate and on-rate) are evaluated using conventional methods.
  • the number of positions likely to be identified in this manner is relatively small. Once identified, these key positions may be randomized to all amino acids to identify the best amino acid solution at the position. Because all manipulations and evaluations are conducted in vitro, physiological bias is limited.
  • Alanine scanning maturation enables the rapid identification of residues most likely to be important in binding. Using the example of a twenty-five residue stretch cited above, only twenty-five substitutions would be necessary. From this initial screen, amino acid positions likely to be critical to binding may be identified. The critical residues may then be randomized to identify the amino acids that optimize binding. Using this method, scFv antibodies with dissociation rates greater than ten-fold slower than the original scFv have been created.
  • PCR recombination is used to substitute all or part of the VH and VL genes into libraries of scFv clones.
  • random mutations are made throughout a CDR region of a scFv clone by the use of degenerate oligonucleotides.
  • clones were expressed as a phage fd gene 3 fusion surface protein. Higher affinity clones were identified using a panning assay followed by clonal purification of the phage.
  • the method disclosed herein is systematic, thorough and unlikely to introduce unexpected or undesired mutations. All manipulations are done in vitro, which minimizes bias due to selection steps. Evaluation of clones is quantitative. In some cases, a key amino acid position may display poorer binding with alanine, but subsequent randomization may yield an amino acid solution which enables improved binding. Such mutations would not be detected by previous methods. Because the method of the present invention does not require phage expression for panning, the method can be used on scFVs, Fabs, and full length antibodies. Use is not restricted to a scFv for phage expression. Using the approach of the present invention, an anti-HIV V3 loop antibody was improved approximately eleven-fold.
  • Alanine scanning maturation of antibodies is a general method which may be used to improve binding of antibodies to their cognate antigens.
  • the method has been used to identify critical residues in the scFv 447 which can be introduced into MAb447. Such changes may lead to significant improvement of the binding affinity of MAb447 against multiple species of HTV gpl20 isolates. This improvement may increase the neutralization capability of the antibody, and significantly lower the effective dose.
  • the genetic code is degenerate, more than one codon may be used to encode a particular amino acid, and therefore, the amino acid sequence can be encoded by any of a set of similar DNA oligonucleotides.
  • the cloned DNA molecules obtained may be expressed by cloning the gene encoding the altered antibody into an expression vector containing a suitable promoter and other appropriate transcription regulatory elements, and transferred into prokaryotic or eukaryotic host cells to produce recombinant modified antibodies. Techniques for such manipulations are well-known in the art.
  • Expression vectors are defined herein as DNA sequences that are required for the transcription of cloned copies of genes and the translation of their mRNAs in an appropriate host. Such vectors can be used to express eukaryotic genes in a variety of hosts such as bacteria, bluegreen algae, plant cells, insect cells and animal cells. Expression vectors include, but are not limited to, cloning vectors, modified cloning vectors, specifically designed plasmids or viruses. Specifically designed vectors allow the shuttling of DNA between hosts, such as bacteria-yeast or bacteria-animal cells.
  • DNA encoding antibodies may also be cloned into an expression vector for expression in a host cell.
  • Host cells may be prokaryotic or eukaryotic, including but not limited to bacteria, yeast, mammalian and insect cells and cell lines.
  • the expression vector may be introduced into host cells via any one of a number of techniques including but not limited to transformation, transfection, protoplast fusion, and electroporation.
  • Synthetic mRNA can be efficiently translated in various cell-free systems, including but not limited to wheat germ extracts and reticulocyte extracts, as well as efficiently translated in cell based systems, including but not limited to microinjection into frog oocytes, with micro-injection into frog oocytes being preferred.
  • Plasmid pP5Q was the starting vector for all mutagenic studies. Plasmid pP5Q is a derivative of p5H7 (Cambridge Antibodies). Plasmid pP5Q contains the VH and VL regions originally derived from MAb 447 (Gorney et al.) cloned as a single chain fragment variable (scFv).
  • Table 1 lists some of the oligonucleotide primers used for site-directed mutagenesis of complementary determining region 3 (CDR3) of MAb447.
  • Primers were synthesized on either a model 381 A DNA Synthesizer (Applied Biosystems, Foster City, CA) or a CycloneTM Plus DNA Synthesizer (MilliGen/Biosearch, Marlborough, MA). Mutagenesis was performed with the TransformerTM Mutagenesis Kit (CLONTECH, Palo Alto, CA) according to the manufacturer's instructions. All mutations were verified by DNA sequencing using the Sequenase® V2.0 DNA Sequencing Kit (United States Biochemical, Cleveland, OH).
  • Mutagenized plasmids were introduced by electroporation into bacterial strain Escherichia coli TGI for expression. Single colonies were inoculated into 10 ml of 2X-YT (which contains per liter of water 16 g tryptone, 10 g yeast extract and 5 g sodium chloride) supplemented with 2% glucose. Cells were grown overnight at 30°C with vigorous shaking, collected by centrifugation in a Beckman GPR centrifuge at 2500 rpm, and resuspended in 10 ml of fresh 2X-YT supplemented with 1 mM isopropyl-beta-D-thiogalactopyranoside (IPTG) to induce expression.
  • 2X-YT which contains per liter of water 16 g tryptone, 10 g yeast extract and 5 g sodium chloride
  • IPTG isopropyl-beta-D-thiogalactopyranoside
  • HIV gpl20 V3 loop peptides, Al-1 variant (Ala-1 peptide) were covalently immobilized on a carboxylated dextran/gold matrix via the primary amino group.
  • the carboxyl-dextran matrix was first activated with N-ethyl-N'-(3-diethylaminopropyl)carbondiimide (EDC) and reacted with N-hydroxysuccinimide (NHS).
  • EDC N-ethyl-N'-(3-diethylaminopropyl)carbondiimide
  • NHS N-hydroxysuccinimide
  • HIV g l20 V3 loop peptides such as Ala-1 peptide were covalently immobilized via the free thiol of a cysteine placed at the N-terminus. These peptides were reacted with the EDC-NHS activated matrix which had been reacted with 2-(2-pyridinyldithio)ethaneamine. Remaining unreacted NHS-ester groups were displaced by addition of ethanolamine. EDTA extracts were added in a flow passing over the immobilized antigen. The refractive index changes, in the form of the surface plasmon resonance caused by the binding and subsequent dissociation of the scFv, were monitored continuously. Off -rates were calculated from the automatically collected data using the Pharmacis Kinetics Evaluation software.
  • the two critical class i) positions (107 and 111) were individually randomized to all amino acids, and off -rates against the AL-1 peptide determined.
  • two class ii) positions (112 and 118) were also selected for randomization studies.
  • a double mutant that combined the optimized residues at positions 107 (E) and 111 (W) was constructed to determine whether or not the individual improvements are additive.
  • Figure 5 shows that the double mutant has an off-rate 9-fold slower than wild-type clone P5Q.
  • the off-rate value approximates the product of the fold improvements observed with the individual optimized residues (2.5 for 107E and 4.7 for 111 W).
  • One interpretation of this result is that for these two positions, the contributions to scFv-antigen affinity are independent and additive.
  • An antibody is mutagenized by alanine scanning mutagenesis to produce a modified antibody.
  • the binding of the modified antibody to its .antigen is determined. Binding determinations may be made by conventional methods and include off -rate measurements. Modified antibodies having desired characteristics are selected and maintained.
  • the modified antibodies or pharmaceutical compositions thereof are used for the prophylactic or therapeutic treatment of diseases caused by their antigen.
  • Methods of treatment include, but are not limited to, intravenous or intraperitoneal injection of the modified antibody.
  • the modified antibodies of Example 7 are used as reagents in diagnostic kits.
  • the modified antibody reagents may be further modified through techniques which are well-known in the art, such as radiolabeling or enzyme-labeling.
  • the diagnostic kit may be used to detect or characterize the antigens.
  • the DNA encoding the modified antibody of Example 7 is used as a reagent for the production of modified antibodies.
  • the DNA may be incorporated into an expression vector.
  • the expression vector may be used to transform a host cell. Cultivation of the host cell under conditions suitable for the expression results in the production of modified antibody.
  • the DNA encoding the modified antibody of Example 7 is used to detect DNA encoding the antigen in test samples. Methods of detection include, but are not limited to, hybridization under selective conditions. Test samples include, but are not limited to, samples of blood, cells, and tissues.
  • the light chain of an immunoglobulin is mutagenized by alanine scanning mutagenesis to produce a modified immimoglobulin having modified binding characteristics.
  • the modified immuno ⁇ globulin is used as a reagent for diagnostic kits or as a therapeutic agent.
  • MOLECULE TYPE DNA (genomic)
  • CGCCAGGCCC CAGGGAAGGG GCTGGAGTGG GTCGGCCGTA TTAAAAGCGC CACTGATGGT 180
  • Glu Trp Val Gly Arg lie Lys Ser Ala Thr Asp Gly Gly Thr Thr Asp 50 55 60 Tyr Ala Ala Ser Val Gin Gly Arg Phe Thr lie Ser Arg Asp Asp Ser 65 70 75 80

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • AIDS & HIV (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

A method of mutagenizing antibodies to produce modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided.

Description

TITLE OF THE INVENTION
IN VITRO ANTIBODY AFFINITY MATURATION USING ALANINE SCANNING MUTAGENESIS
CROSS-RELATED TO OTHER APPLICATIONS This is a continuation of U.S. Serial No. 08/206,076 filed March 4, 1994, now pending.
BRIEF DESCRIPTION OF INVENTION
A method of mutagenizing antibodies to produce 0 modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided. The method of the invention is a systematic means to achieve in vitro antibody maturation and uses alanine scanning mutagenesis. The 5 invention is particularly exemplified with a set of single chain Fv (scFv) antibodies obtained by this technique. The resulting antibodies are directed against the V3 loop of HIV gpl20, and show altered off-rates against the antigen compared to the starting antibody. Of particular interest are the altered antibodies which 0 show improved (slower) off-rates to the antigen. Observed improvements have been as high as eleven-fold over wild-type.
SUMMARY OF THE INVENTION
A method of mutagenizing antibodies to produce 5 modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided.
BRIEF DESCRIPTION OF THE DRAWINGS 0
Figure 1. Alanine-Scanning Mutagenesis. Each of the 27 amino acids in VH CDR3 of scFv P5Q was converted to alanine by site-directed mutagenesis. E. coli clones were induced to express scFv with IPTG. Single chain Fv, which is targeted to the periplasmic space by the fd phage gene3 signal sequence, was extracted with EDTA. Periplasmic extracts were analyzed by BIAcore™, which measures antibody-antigen affinity by surface plasmon resonance (Fagerstam, 1991), and off -rates determined against an HIV gpl20 V3 loop peptide. Results of the alanine scan, relative to P5Q, fall into four classes: i) slower off-rate, ii) faster off-rate, iii) no binding, and iv) minor or no change in off-rate. Standard deviation is ± 25%.
Figure 2. Amino Acid Randomization: Position 107. Arginine at position 107 was mutated to all amino acids by site- directed mutagenesis. Single chain Fv extracts were analyzed by BIAcore. Percent change in off-rates is shown relative to P5Q.
Figure 3. Amino Acid Randomization: Position 111. Glutamic acid at position 111 was mutated to all amino acids by site- directed mutagenesis. Single chain Fv extracts were analyzed by BIAcore. Percent change in off-rates is shown relative to P5Q.
Figure 4. Amino Acid Randomization: Position 112. Aspartic acid at position 112 was mutated to all amino acids by site- directed mutagenesis. Single chain Fv extracts were analyzed by BIAcore. Percent change in off-rates is shown relative to P5Q.
Figure 5. Additive Effect of Combining Optimized Residues. A double mutant, containing the optimized residues, was constructed and analyzed by BIAcore. Percent change in off-rates is shown relative to P5Q.
Figure 6. Nucleotide and amino acid sequences of scFv P5Q with c-myc tail.
DETAILED DESCRIPTION OF THE INVENTION
The gpl20 V3 domain of human immunodeficiency virus-1 (HTV-1) is a disulfide-linked closed loop of approximately 30 amino acids. The loop, in either native or synthetic form, binds to and elicits anti-HIV-1 antibodies.
The present invention relates to modified antibodies and methods of making modified. The invention is exemplified with modified HIV-1 immunoglobulins and methods of making these modified HTV-1 immunoglobulins. The modified immunoglobulins of the present invention contain an altered complementary determining region 3 (CDR3) of HTV-l neutralizing antibody.
The present invention also comprises a method of treating of preventing infection through the administration of a modified antibody to a suitable host. In one embodiment of the invention, the treatment or prevention of HIV infection through the administration of the modified HTV-l immunoglobulm is described.
The present invention also comprises diagnostic kits useful for the detection or characterization of an antigen. Reagents for the kits may include DNA molecules encoding the modified antibodies or the modified antibodies or combinations thereof.
A method of mutagenizing antibodies to produce modified antibodies, modified antibodies, DNA encoding the modified antibodies as well as diagnostic kits and pharmaceutical compositions comprising the antibodies or DNA are provided. The method of the invention is a systematic means to achieve in vitro antibody maturation and uses alanine scanning mutagenesis. The invention is particularly exemplified with a set of single chain Fv (scFv) antibodies obtained by this technique. The resulting antibodies are directed against the V3 loop of HIV gpl20, and show altered off-rates against the antigen compared to the starting antibody. Of particular interest are the altered antibodies which show improved (slower) off-rates to the antigen. Observed improvements have been as high as eleven-fold over wild-type.
Maturation was achieved through an alanine scan of complementary determining region 3 (CDR3) to identify positions critical to antigen binding. Critical positions were then randomized to identify amino acids that provided the slowest off-rates. Finally, clones were optimized through the combining of mutations.
The underlying principle of the method is the physical and chemical neutrality of alanine. Alanine is substituted throughout a stretch of amino acids, and its effects on binding (such as off -rate and on-rate) are evaluated using conventional methods. The number of positions likely to be identified in this manner is relatively small. Once identified, these key positions may be randomized to all amino acids to identify the best amino acid solution at the position. Because all manipulations and evaluations are conducted in vitro, physiological bias is limited.
Present methods of in vitro antibody maturation are essentially random procedures in which the researcher generates clones with amino acid substitutions and evaluates them. The problem is that the number of substitutions necessary for a thorough evaluation is extremely large. For example, if one were to evaluate all random substitutions in CDR3, a region typically twenty-five residues in length, one would have to examine 9»lθ27 possibilities. This is beyond the capabilities of present technologies.
Alanine scanning maturation enables the rapid identification of residues most likely to be important in binding. Using the example of a twenty-five residue stretch cited above, only twenty-five substitutions would be necessary. From this initial screen, amino acid positions likely to be critical to binding may be identified. The critical residues may then be randomized to identify the amino acids that optimize binding. Using this method, scFv antibodies with dissociation rates greater than ten-fold slower than the original scFv have been created.
Previous work in in vitro antibody maturation used one of two general approaches. In one approach, PCR recombination is used to substitute all or part of the VH and VL genes into libraries of scFv clones. In the second approach, random mutations are made throughout a CDR region of a scFv clone by the use of degenerate oligonucleotides. In both cases, clones were expressed as a phage fd gene 3 fusion surface protein. Higher affinity clones were identified using a panning assay followed by clonal purification of the phage.
Each approach has drawbacks. The PCR method is cumbersome, limited to the sequences of the B cell population, is essentially random in nature, and may introduce unwanted mutations through the PCR recombination step. The randomization approach produces only a small fraction of the possible CDR changes. Neither approach allows immediate determination of changes in binding affinity because it is necessary to first generate an enriched population of suitable clones through panning. Both approaches detect only changes which result in improved binding; they do not identify positions for which the change weakened the binding. The latter class of change may include critical binding residues in which the appropriate amino acid solutions leads to improvement.
The method disclosed herein is systematic, thorough and unlikely to introduce unexpected or undesired mutations. All manipulations are done in vitro, which minimizes bias due to selection steps. Evaluation of clones is quantitative. In some cases, a key amino acid position may display poorer binding with alanine, but subsequent randomization may yield an amino acid solution which enables improved binding. Such mutations would not be detected by previous methods. Because the method of the present invention does not require phage expression for panning, the method can be used on scFVs, Fabs, and full length antibodies. Use is not restricted to a scFv for phage expression. Using the approach of the present invention, an anti-HIV V3 loop antibody was improved approximately eleven-fold.
Alanine scanning maturation of antibodies is a general method which may be used to improve binding of antibodies to their cognate antigens. The method has been used to identify critical residues in the scFv 447 which can be introduced into MAb447. Such changes may lead to significant improvement of the binding affinity of MAb447 against multiple species of HTV gpl20 isolates. This improvement may increase the neutralization capability of the antibody, and significantly lower the effective dose.
Although the method and antibodies of the present invention are exemplified with scFv antibodies, it is readily apparent to those skilled in the art that the method may be used with other types of antibodies or with antibodies targetted against different epitopes or antigens. Other types of antibodies include but are not limited to fragments of antibodies and full-length antibodies.
The molecular biology and immunological techniques of the present invention can be performed by standard techniques well- known in the art. See, for example, in Mamatis, T., Fritsch, E.F., Sambrook, J., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1982).
Because the genetic code is degenerate, more than one codon may be used to encode a particular amino acid, and therefore, the amino acid sequence can be encoded by any of a set of similar DNA oligonucleotides.
The cloned DNA molecules obtained may be expressed by cloning the gene encoding the altered antibody into an expression vector containing a suitable promoter and other appropriate transcription regulatory elements, and transferred into prokaryotic or eukaryotic host cells to produce recombinant modified antibodies. Techniques for such manipulations are well-known in the art.
In order to simplify the following Examples and the Detailed Description, certain terms will be defined.
Expression vectors are defined herein as DNA sequences that are required for the transcription of cloned copies of genes and the translation of their mRNAs in an appropriate host. Such vectors can be used to express eukaryotic genes in a variety of hosts such as bacteria, bluegreen algae, plant cells, insect cells and animal cells. Expression vectors include, but are not limited to, cloning vectors, modified cloning vectors, specifically designed plasmids or viruses. Specifically designed vectors allow the shuttling of DNA between hosts, such as bacteria-yeast or bacteria-animal cells.
DNA encoding antibodies may also be cloned into an expression vector for expression in a host cell. Host cells may be prokaryotic or eukaryotic, including but not limited to bacteria, yeast, mammalian and insect cells and cell lines.
The expression vector may be introduced into host cells via any one of a number of techniques including but not limited to transformation, transfection, protoplast fusion, and electroporation.
Expression of cloned DNA may also be performed using in vitro produced synthetic mRNA. Synthetic mRNA can be efficiently translated in various cell-free systems, including but not limited to wheat germ extracts and reticulocyte extracts, as well as efficiently translated in cell based systems, including but not limited to microinjection into frog oocytes, with micro-injection into frog oocytes being preferred.
It is also well-known that there is a substantial amount of redundancy in the various codons which code for specific amino acids. Therefore, this invention is also directed to those DNA sequences which contain alternative codons which code for the eventual translation of the identical amino acid. For purposes of this specification, a sequence bearing one or more replaced codons will be defined as a degenerate variant.
The following examples are provided to further define the invention without, however, limiting the invention to the particulars of these examples.
EXAMPLE 1
Construction of mutations
Plasmid pP5Q was the starting vector for all mutagenic studies. Plasmid pP5Q is a derivative of p5H7 (Cambridge Antibodies). Plasmid pP5Q contains the VH and VL regions originally derived from MAb 447 (Gorney et al.) cloned as a single chain fragment variable (scFv).
Table 1 lists some of the oligonucleotide primers used for site-directed mutagenesis of complementary determining region 3 (CDR3) of MAb447. Primers were synthesized on either a model 381 A DNA Synthesizer (Applied Biosystems, Foster City, CA) or a Cyclone™ Plus DNA Synthesizer (MilliGen/Biosearch, Marlborough, MA). Mutagenesis was performed with the Transformer™ Mutagenesis Kit (CLONTECH, Palo Alto, CA) according to the manufacturer's instructions. All mutations were verified by DNA sequencing using the Sequenase® V2.0 DNA Sequencing Kit (United States Biochemical, Cleveland, OH).
Table 1
Primers:
Randomization of position 107:
CTCGGAGACTCCC/GNNAATCATAATAAA
Randomization of position 111 :
GTAGTAGTAGTCC/GNNGGAGACTCCCCG
Randomization of position 112:
GTCGTTGTAGTAGTAGTAGTAC/GNNCTCGGAGAC
EXAMPLE2
Preparation of extracts and BIAcore analysis of scFv Extracts:
Mutagenized plasmids were introduced by electroporation into bacterial strain Escherichia coli TGI for expression. Single colonies were inoculated into 10 ml of 2X-YT (which contains per liter of water 16 g tryptone, 10 g yeast extract and 5 g sodium chloride) supplemented with 2% glucose. Cells were grown overnight at 30°C with vigorous shaking, collected by centrifugation in a Beckman GPR centrifuge at 2500 rpm, and resuspended in 10 ml of fresh 2X-YT supplemented with 1 mM isopropyl-beta-D-thiogalactopyranoside (IPTG) to induce expression. Cells were incubated at 30°C for an additional 5-6 hours with vigorous shaking, collected by centrifugation, resuspended in 1 ml of phosphate buffered saline: ethylenediametetraacetic acid (PBS:EDTA; 10 mM sodium phosphate pH7.0, 150 mM sodium chloride 1 mM EDTA), and incubated on ice for 30 minutes to release periplasmic proteins. Extracts were clarified by centrifugation and stored at 4°C until use.
EXAMPLE 3
Off -rate determinations of the scFv antibodies were determined using the BIAcore system (Pharmacia Biosenser). HIV gpl20 V3 loop peptides, Al-1 variant (Ala-1 peptide) were covalently immobilized on a carboxylated dextran/gold matrix via the primary amino group. The carboxyl-dextran matrix was first activated with N-ethyl-N'-(3-diethylaminopropyl)carbondiimide (EDC) and reacted with N-hydroxysuccinimide (NHS). HIV g l20 V3 loop peptides such as Ala-1 peptide were covalently immobilized via the free thiol of a cysteine placed at the N-terminus. These peptides were reacted with the EDC-NHS activated matrix which had been reacted with 2-(2-pyridinyldithio)ethaneamine. Remaining unreacted NHS-ester groups were displaced by addition of ethanolamine. EDTA extracts were added in a flow passing over the immobilized antigen. The refractive index changes, in the form of the surface plasmon resonance caused by the binding and subsequent dissociation of the scFv, were monitored continuously. Off -rates were calculated from the automatically collected data using the Pharmacis Kinetics Evaluation software.
EXAMPLE 4
Alanine scanning of CDR3 identifies residues which modulate scFv- antigen binding
Alanine scanning mutagenesis was used to identify residues within the VH CDR3 region of scFv clone P5Q critical for binding. It was hypothesized that effects on binding by alanine substitution would lead to four broad classes of effect: class i) slower off-rate; class ii) faster off-rate; class iii) loss of binding; and class iv) minor or no change in off -rate. Class i) and ii) were operationally defined as critical. Class iii) was defined as obligatory. Class iv) was defined as noncritical.
The 27 positions that comprise VH CDR3 of scFv clone P5Q were individually changed to alanine by site-directed mutagenesis. Periplasmic extracts were prepared from the alanine replacement clones and assayed for off-rate determinations against the AL-1 gpl20 V3 loop peptide (Fig. 1). Alanine substitutions at positions 107 and 111 resulted in 1.7 and 2.7 fold improvements in off-rate, respectively. These positions (class i) were judged critical and subsequently randomized to identify optimal residues. Alanine substitutions at positions 102, 112, 113, 114, and 118 led to faster off-rates (class ii); two of these positions were selected for further evaluation. Alanine substitution at positions 98, 101, 115, 116, 117, and 121 resulted in no binding (class iii). Alanine substitution at the remaining fourteen positions had only a minor effect on the off-rate (class iv). The class iii and iv positions were not evaluated further.
EXAMPLE 5
Randomization at critical positions to identify optimal amino acid solutions
The two critical class i) positions (107 and 111) were individually randomized to all amino acids, and off -rates against the AL-1 peptide determined. In addition, two class ii) positions (112 and 118) were also selected for randomization studies.
The results for position 107 are shown in Fig. 2. The slowest off-rate was observed with the negatively-charged glutamic acid, which decreased dissociation 2.5-fold. Substitution of other polar and charged amino acids had no significant effect on dissociation. With the exception of alanine, substitution with hydrophobic amino acid resulted in complete loss of binding. These results are consistent with the preponderance of surface ligand- contact residues being hydrophilic. Randomization of position 111 (Fig. 3) showed that the aromatic residues tyrosine and tryptophan produced the slowest off- rates (dissociation rates decreased 4.2 and 4.7-fold, respectively). However, substitution with any hydrophobic amino acids increased affinity relative to wild-type clone P5Q.
Class ii) positions 112 and 118 (faster off-rate upon alanine substitution) were also selected for amino acid randomization. For both position 112 (Fig. 4) and 118, the residues present in the original scFv P5Q, aspartic acid and asparagine, were the best solutions.
EXAMPLE 6
Improvements at positions 107 and 111 are additive
A double mutant that combined the optimized residues at positions 107 (E) and 111 (W) was constructed to determine whether or not the individual improvements are additive. Figure 5 shows that the double mutant has an off-rate 9-fold slower than wild-type clone P5Q. The off-rate value approximates the product of the fold improvements observed with the individual optimized residues (2.5 for 107E and 4.7 for 111 W). One interpretation of this result is that for these two positions, the contributions to scFv-antigen affinity are independent and additive.
EXAMPLE 7
Method of making modified antibodies
An antibody is mutagenized by alanine scanning mutagenesis to produce a modified antibody. The binding of the modified antibody to its .antigen is determined. Binding determinations may be made by conventional methods and include off -rate measurements. Modified antibodies having desired characteristics are selected and maintained. EXAMPLE 8
Method of using modified antibodies
The modified antibodies or pharmaceutical compositions thereof are used for the prophylactic or therapeutic treatment of diseases caused by their antigen. Methods of treatment include, but are not limited to, intravenous or intraperitoneal injection of the modified antibody.
EXAMPLE 9
Diagnostic kit employing modified antibodies
The modified antibodies of Example 7 are used as reagents in diagnostic kits. The modified antibody reagents may be further modified through techniques which are well-known in the art, such as radiolabeling or enzyme-labeling. The diagnostic kit may be used to detect or characterize the antigens.
EXAMPLE 10
DNA encoding modified antibodies
The DNA encoding the modified antibody of Example 7 is used as a reagent for the production of modified antibodies. The DNA may be incorporated into an expression vector. The expression vector may be used to transform a host cell. Cultivation of the host cell under conditions suitable for the expression results in the production of modified antibody. EX AMPLE 11
DNA encoding modified antibodies
The DNA encoding the modified antibody of Example 7 is used to detect DNA encoding the antigen in test samples. Methods of detection include, but are not limited to, hybridization under selective conditions. Test samples include, but are not limited to, samples of blood, cells, and tissues.
EXAMPLE 12
Preparation of modified light chain immunoglobulins
The light chain of an immunoglobulin is mutagenized by alanine scanning mutagenesis to produce a modified immimoglobulin having modified binding characteristics. The modified immuno¬ globulin is used as a reagent for diagnostic kits or as a therapeutic agent.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: LEWIS, CRAIG M.
LUDMERER, STEVEN W. HOLLIS, GREGORY F.
(ii) TITLE OF INVENTION: IN VITRO ANTIBODY MATURATION
(iii) NUMBER OF SEQUENCES: 2
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: CHRISTINE E. CARTY
(B) STREET: P.O. BOX 2000, 126 E. LINCOLN AVENUE
(C) CITY: RAHWAY
(D) STATE: NJ
(E) COUNTRY: USA
(F) ZIP: 07065
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/206,079
(B) FILING DATE: 04-MAR-1994
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: CARTY, CHRISTINE E.
(B) REGISTRATION NUMBER: 36,090
(C) REFERENCE/DOCKET NUMBER: 19190P
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (908) 594-6734
(B) TELEFAX: (908) 594-4720
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 816 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
GCCATGGCCG AGGTGCAGCT GGTGGAGTCT GGGGGAGGCT TGGTAAAGCC TGGGGGGTCC 60
CTCAGACTCA CCTGTGTAGC CTCTGGCTTC ACGTTCAGTG ATGTCTGGCT GAACTGGGTC 120
CGCCAGGCCC CAGGGAAGGG GCTGGAGTGG GTCGGCCGTA TTAAAAGCGC CACTGATGGT 180
GGGACAACAG ACTACGCTGC ATCCGTGCAA GGCAGATTCA CCATCTCAAG AGATGACTCA 240
AAAAACACGC TATATCTGCA AATGAATAGC CTGAAAACCG AGGACACAGC CGTTTATTCC 300
TGCAACACAG ATGGTTTTAT TATGATTCGG GGAGTCTCCG AGGACTACTA CTACTACTAC 360
AACGACGTTT GGGGCAAAGG GACCACGGTC ACCGTCTCCT CAGGTGCAGG CGGTTCAGGC 420
GGAGGTGGCT CTGGCGGTGG CGGATCGCAG TCTGTGTTGA CGCAGCCGCC CTCAGTGTCT 480
GCGGCCCCAG GACAGAAGGT CACCATCTCC TGCTCTGGAA GCAGCTCCAA CATTGGGAAT 540
AATTATGTAT TGTGGTACCA GCAGTTCCCA GGAACAGCCC CCAAACTCCT CATTTATGGC 600
AATAATAAGC GACCCTCAGG GATTCCTGAC CGATTCTCTG GCTCCAAGTC TGGCACGTCA 660
GCCACCCTGG GCATCACCGG ACTCCAGACT GGGGACGAGG CCGATTATTT CTGCGCAACA 720
TGGGATAGCG GCCTGAGTGC TGATTGGGTG TTCGGCGGAG GGACCAAGCT GACCGTCCTA 780
GGTGCGGCCG CAGAACAAAA ACTCATCTCA GAAGAG 816 (2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 272 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Ala Met Ala Glu Val Glx Leu Val Glu Ser Gly Gly Gly Leu Val Lys 1 5 10 15
Pro Gly Gly Ser Leu Arg Leu Thr Cys Val Ala Ser Gly Phe Thr Phe
20 25 30
Ser Asp Val Trp Leu Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu 35 40 45
Glu Trp Val Gly Arg lie Lys Ser Ala Thr Asp Gly Gly Thr Thr Asp 50 55 60 Tyr Ala Ala Ser Val Gin Gly Arg Phe Thr lie Ser Arg Asp Asp Ser 65 70 75 80
Lys Asn Thr Leu Tyr Leu Glx Met Asn Ser Leu Lys Thr Glu Asp Thr 85 90 95
Ala Val Tyr Ser Cys Asn Thr Asp Gly Phe lie Met lie Arg Gly Val 100 105 110
Ser Glu Asp Tyr Tyr Tyr Tyr Tyr Asn Asp Val Trp Gly Lys Gly Thr 115 120 125
Thr Val Thr Ala Ser Ser Gly Ala Gly Gly Ser Gly Gly Gly Gly Ser 130 135 140
Gly Gly Gly Ser Gin Ser Val Leu Thr Gin Pro Pro Ser Val Ser Ala 145 150 155 160
Ala Pro Gly Gin Lys Val Thr lie Ser Cys Ser Gly Ser Ser Ser Asn 165 170 175 lie Gly Asn Asn Tyr Val Leu Trp Tyr Gin Gin Phe Pro Gly Thr Ala 180 185 190
Pro Lys Leu Leu lie Tyr Gly Asn Asn Lys Arg Pro Ser Gly lie Pro 195 200 205
Asp Arg Phe Ser Gly Ser Lys Leu Leu lie Tyr Gly Ala Thr Leu Gly 210 215 220 lie Thr Gly Leu Gin Thr Gly Asp Gin Ala Asp Tyr Phe Cys Ala Thr 225 230 235 240
Trp Asp Ser Gly Leu Ser Ala Asp Trp Val Phe Gly Gly Gly Thr Lys 245 250 255
Leu Thr Val Leu Gly Ala Ala Ala Glu Gin Lys Leu lie Ser Glu Glu 260 265 270

Claims

WHAT IS CLAIMED IS:
1. A DNA molecule encoding a modified antibody, the modified antibody being derived from a native antibody by alanine scanning mutagenesis and the modified antibody having binding characteristics different than binding characteristics of the native antibody.
2. The DNA molecule of Claim 1 wherein the native antibody is MAb447.
3. The DNA molecule of Claim 2, the DNA molecule being selected from the group consisting of P5Q, DNA encoding modified antibodies of Figures 1, 2, 3, 4, 5, combinations thereof, derivatives thereof and degenerate variants thereof.
4. A method of modifying an antibody to make an modified antibody comprising replacing at least one amino acid of the antibody with alanine to produce a modified antibody.
5. The method of Claim 4 wherein the modified antibody has improved binding characteristics.
6. Modified antibodies produced by the method of Claim 4 or homologues thereof.
7. The method of Claim 4 wherein the antibody is MAb447.
8. The method of Claim 7 wherein the amino acid replaced with alanine is located in complementary determining region 1, complementary determining region 2 or complementary determining region 3 of MAb447.
9. The modified antibodies of Claim 6 selected from the group consisting of P5Q, the antibodies of Figures 1, 2, 3, 4, 5, combinations thereof, derivatives thereof, and homologues thereof.
10. Diagnostic kits comprising the modified antibodies produced by the method of Claim 6.
11. Diagnostic kits comprising the DNA molecules of Claim 1.
12. A pharmaceutical composition comprising at least one modified antibody of Claim 6 or DNA encoding at least one modified antibody of Claim 6 or combinations thereof.
PCT/US1995/002492 1994-03-04 1995-02-27 In vitro antibody affinity maturation using alanine scanning mutagenesis WO1995023813A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP7522980A JPH09509835A (en) 1994-03-04 1995-02-27 In vitro antibody affinity maturation by alanine scanning mutagenesis
EP95911994A EP0748338A4 (en) 1994-03-04 1995-02-27 In vitro antibody affinity maturation using alanine scanning mutagenesis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US20607994A 1994-03-04 1994-03-04
US206,079 1994-03-04

Publications (1)

Publication Number Publication Date
WO1995023813A1 true WO1995023813A1 (en) 1995-09-08

Family

ID=22764890

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/002492 WO1995023813A1 (en) 1994-03-04 1995-02-27 In vitro antibody affinity maturation using alanine scanning mutagenesis

Country Status (4)

Country Link
EP (1) EP0748338A4 (en)
JP (1) JPH09509835A (en)
CA (1) CA2183550A1 (en)
WO (1) WO1995023813A1 (en)

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999001475A2 (en) * 1997-07-04 1999-01-14 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Human antibody against a fusion (poly) peptide or protein presenting a part having at least six histidines
US6025158A (en) * 1997-02-21 2000-02-15 Genentech, Inc. Nucleic acids encoding humanized anti-IL-8 monoclonal antibodies
US6037454A (en) * 1996-11-27 2000-03-14 Genentech, Inc. Humanized anti-CD11a antibodies
US6090382A (en) * 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US6133426A (en) * 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US6258562B1 (en) 1996-02-09 2001-07-10 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US6458355B1 (en) 1998-01-22 2002-10-01 Genentech, Inc. Methods of treating inflammatory disease with anti-IL-8 antibody fragment-polymer conjugates
US6468532B1 (en) 1998-01-22 2002-10-22 Genentech, Inc. Methods of treating inflammatory diseases with anti-IL-8 antibody fragment-polymer conjugates
WO2003023032A2 (en) * 2001-08-27 2003-03-20 Nautilus Biotech High throughput directed evolution by rational mutagenesis
US6870033B1 (en) 1997-02-21 2005-03-22 Genentech, Inc. Antibody fragment-polymer conjugates and humanized anti-IL-8 monoclonal antibodies
US7005504B2 (en) 1998-01-22 2006-02-28 Genentech, Inc. Antibody fragment-peg conjugates
US7214776B2 (en) 1999-01-21 2007-05-08 Genentech, Inc. Antibody fragment-polymer conjugates and uses of same
US7650243B2 (en) 2002-09-09 2010-01-19 Hanall Pharmaceutical Co., Ltd. Rational evolution of cytokines for higher stability, the cytokines and encoding nucleic acid molecules
US8231876B2 (en) 2006-04-05 2012-07-31 Abbott Biotechnology Ltd. Purified antibody composition
US8846046B2 (en) 2002-10-24 2014-09-30 Abbvie Biotechnology Ltd. Low dose methods for treating disorders in which TNFα activity is detrimental
US8889135B2 (en) 2001-06-08 2014-11-18 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8889136B2 (en) 2004-04-09 2014-11-18 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US8906373B2 (en) 2002-07-19 2014-12-09 Abbvie Biotechnology Ltd. Use of TNF-alpha inhibitor for treatment of psoriasis
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9127052B2 (en) 2010-06-29 2015-09-08 Centre National De La Recherche Scientifique LLT-1 antibodies with new functional properties
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
EP1660655B1 (en) * 2003-06-27 2016-11-02 Bioren, LLC Look-through mutagenesis
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
US9610301B2 (en) 2008-01-15 2017-04-04 Abbvie Deutschland Gmbh & Co Kg Powdered protein compositions and methods of making same
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2814780C (en) * 2010-09-22 2017-05-16 Amgen Inc. Carrier immunoglobulins and uses thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ASHKENAZI A., ET AL.: "MAPPING THE CD4 BINDING SITE FOR HUMAN IMMUNODEFICIENCY VIRUS BY ALANINE-SCANNING MUTAGENESIS.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, US, vol. 87., 1 September 1990 (1990-09-01), US, pages 7150 - 7154., XP000578388, ISSN: 0027-8424, DOI: 10.1073/pnas.87.18.7150 *
CUNNINGHAM B. C., WELLS J. A.: "HIGH-RESOLUTION EPITOPE MAPPING OF HGH-RECEPTOR INTERACTIONS BY ALANINE-SCANNING MUTAGENESIS.", SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE, US, vol. 244., no. 4908., 2 June 1989 (1989-06-02), US, pages 1081 - 1085., XP000025308, ISSN: 0036-8075, DOI: 10.1126/science.2471267 *
M. GORNY ET AL.: "Repertoire of neutralizing human monoclonal antibodies specific for the V3 domain of HIV-1 gp120.", THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 2, no. 150, 15 January 1993 (1993-01-15), US, pages 635 - 643, XP002076058, ISSN: 0022-1767 *
See also references of EP0748338A4 *

Cited By (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8372401B2 (en) 1996-02-09 2013-02-12 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα
US8414894B2 (en) 1996-02-09 2013-04-09 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα and methods of using same
US7588761B2 (en) 1996-02-09 2009-09-15 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα
US7223394B2 (en) 1996-02-09 2007-05-29 Abbott Biotechnology Ltd Human antibodies that bind human TNFα
US6090382A (en) * 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US6509015B1 (en) 1996-02-09 2003-01-21 Basf Aktiengesellschaft Human antibodies that bind human TNFa
US6258562B1 (en) 1996-02-09 2001-07-10 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US8197813B2 (en) 1996-02-09 2012-06-12 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα
US8753633B2 (en) 1996-02-09 2014-06-17 Abbvie Biotechnology Ltd. Human antibodies that bind human TNFα
US7541031B2 (en) 1996-02-09 2009-06-02 Abbott Biotechnology Ltd. Methods for treating rheumatoid arthritis using human antibodies that bind human TNFα
US8206714B2 (en) 1996-02-09 2012-06-26 Abbott Biotechnology Ltd. Methods for treating rheumatoid arthritis using human antibodies that bind human TNFa
US8372400B2 (en) 1996-02-09 2013-02-12 Abbott Biotechnology Ltd. Methods of treating disorders using human antibodies that bind human TNFα
US6703018B2 (en) 1996-11-27 2004-03-09 Genentech, Inc. Method of treatment using humanized anti-CD11a antibodies
US6037454A (en) * 1996-11-27 2000-03-14 Genentech, Inc. Humanized anti-CD11a antibodies
US6133426A (en) * 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US6870033B1 (en) 1997-02-21 2005-03-22 Genentech, Inc. Antibody fragment-polymer conjugates and humanized anti-IL-8 monoclonal antibodies
US6025158A (en) * 1997-02-21 2000-02-15 Genentech, Inc. Nucleic acids encoding humanized anti-IL-8 monoclonal antibodies
WO1999001475A2 (en) * 1997-07-04 1999-01-14 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Human antibody against a fusion (poly) peptide or protein presenting a part having at least six histidines
WO1999001475A3 (en) * 1997-07-04 1999-05-14 Deutsches Krebsforsch Human antibody against a fusion (poly) peptide or protein presenting a part having at least six histidines
US6468532B1 (en) 1998-01-22 2002-10-22 Genentech, Inc. Methods of treating inflammatory diseases with anti-IL-8 antibody fragment-polymer conjugates
US6458355B1 (en) 1998-01-22 2002-10-01 Genentech, Inc. Methods of treating inflammatory disease with anti-IL-8 antibody fragment-polymer conjugates
US7005504B2 (en) 1998-01-22 2006-02-28 Genentech, Inc. Antibody fragment-peg conjugates
US8147830B2 (en) 1999-01-21 2012-04-03 Genentech, Inc. Antibody fragment-polymer conjugates and uses of same
US7842789B2 (en) 1999-01-21 2010-11-30 Genentech, Inc. Antibody fragment-polymer conjugates and uses of same
US7214776B2 (en) 1999-01-21 2007-05-08 Genentech, Inc. Antibody fragment-polymer conjugates and uses of same
US8652468B2 (en) 1999-01-21 2014-02-18 Genentech, Inc. Methods of binding TNF-α using anti-TNF-α antibody fragment-polymer conjugates
US7507405B2 (en) 1999-01-21 2009-03-24 Genentech, Inc. Antibody fragment-polymer conjugates and uses of same
US8974790B2 (en) 2001-06-08 2015-03-10 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8992926B2 (en) 2001-06-08 2015-03-31 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8889135B2 (en) 2001-06-08 2014-11-18 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US9546212B2 (en) 2001-06-08 2017-01-17 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8911737B2 (en) 2001-06-08 2014-12-16 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US9073987B2 (en) 2001-06-08 2015-07-07 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US9017680B2 (en) 2001-06-08 2015-04-28 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
WO2003023032A2 (en) * 2001-08-27 2003-03-20 Nautilus Biotech High throughput directed evolution by rational mutagenesis
US7647184B2 (en) 2001-08-27 2010-01-12 Hanall Pharmaceuticals, Co. Ltd High throughput directed evolution by rational mutagenesis
WO2003023032A3 (en) * 2001-08-27 2003-10-23 Nautilus Biotech High throughput directed evolution by rational mutagenesis
US9085620B1 (en) 2002-07-19 2015-07-21 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriatic arthritis
US9090689B1 (en) 2002-07-19 2015-07-28 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriasis
US8906373B2 (en) 2002-07-19 2014-12-09 Abbvie Biotechnology Ltd. Use of TNF-alpha inhibitor for treatment of psoriasis
US8057787B2 (en) 2002-09-09 2011-11-15 Hanall Biopharma Co., Ltd. Protease resistant modified interferon-beta polypeptides
US7650243B2 (en) 2002-09-09 2010-01-19 Hanall Pharmaceutical Co., Ltd. Rational evolution of cytokines for higher stability, the cytokines and encoding nucleic acid molecules
US7998469B2 (en) 2002-09-09 2011-08-16 Hanall Biopharma Co., Ltd. Protease resistant interferon beta mutants
US8052964B2 (en) 2002-09-09 2011-11-08 Hanall Biopharma Co., Ltd. Interferon-β mutants with increased anti-proliferative activity
US8105573B2 (en) 2002-09-09 2012-01-31 Hanall Biopharma Co., Ltd. Protease resistant modified IFN beta polypeptides and their use in treating diseases
US8846046B2 (en) 2002-10-24 2014-09-30 Abbvie Biotechnology Ltd. Low dose methods for treating disorders in which TNFα activity is detrimental
EP1660655B1 (en) * 2003-06-27 2016-11-02 Bioren, LLC Look-through mutagenesis
US8961973B2 (en) 2004-04-09 2015-02-24 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US8961974B2 (en) 2004-04-09 2015-02-24 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US9512216B2 (en) 2004-04-09 2016-12-06 Abbvie Biotechnology Ltd. Use of TNFα inhibitor
US8986693B1 (en) 2004-04-09 2015-03-24 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriasis
US9061005B2 (en) 2004-04-09 2015-06-23 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating idiopathic inflammatory bowel disease
US9187559B2 (en) 2004-04-09 2015-11-17 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating idiopathic inflammatory bowel disease
US8889136B2 (en) 2004-04-09 2014-11-18 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US9499615B2 (en) 2004-04-09 2016-11-22 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating idiopathic inflammatory bowel disease
US9067992B2 (en) 2005-05-16 2015-06-30 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriatic arthritis
US8231876B2 (en) 2006-04-05 2012-07-31 Abbott Biotechnology Ltd. Purified antibody composition
US11083792B2 (en) 2006-04-05 2021-08-10 Abbvie Biotechnology Ltd Purified antibody composition
US9913902B2 (en) 2006-04-05 2018-03-13 Abbvie Biotechnology Ltd. Purified antibody composition
US9273132B2 (en) 2006-04-05 2016-03-01 Abbvie Biotechnology Ltd Purified antibody composition
US9328165B2 (en) 2006-04-05 2016-05-03 Abbvie Biotechnology Ltd. Purified antibody composition
US8906372B2 (en) 2006-04-05 2014-12-09 Abbvie Biotechnology Ltd. Purified antibody composition
US8895009B2 (en) 2006-04-05 2014-11-25 Abbvie Biotechnology Ltd. Purified antibody composition
US8916153B2 (en) 2006-04-05 2014-12-23 Abbvie Biotechnology Ltd. Purified antibody composition
US9096666B2 (en) 2006-04-05 2015-08-04 Abbvie Biotechnology Ltd Purified antibody composition
US9102723B2 (en) 2006-04-05 2015-08-11 Abbvie Biotechnology Ltd Purified antibody composition
US8883156B2 (en) 2006-04-05 2014-11-11 Abbvie Biotechnology Ltd. Purified antibody composition
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
US9284370B1 (en) 2007-06-11 2016-03-15 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis
US9669093B2 (en) 2007-06-11 2017-06-06 Abbvie Biotechnology Ltd Methods for treating juvenile idiopathic arthritis
US9610301B2 (en) 2008-01-15 2017-04-04 Abbvie Deutschland Gmbh & Co Kg Powdered protein compositions and methods of making same
US9127052B2 (en) 2010-06-29 2015-09-08 Centre National De La Recherche Scientifique LLT-1 antibodies with new functional properties
US9376496B2 (en) 2010-06-29 2016-06-28 Centre National De La Recherche Scientifique LLT-1 antibodies with new functional properties
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9365645B1 (en) 2011-04-27 2016-06-14 Abbvie, Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9090688B2 (en) 2011-04-27 2015-07-28 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9346879B2 (en) 2012-04-20 2016-05-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9359434B2 (en) 2012-04-20 2016-06-07 Abbvie, Inc. Cell culture methods to reduce acidic species
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9290568B2 (en) 2012-09-02 2016-03-22 Abbvie, Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9315574B2 (en) 2013-10-18 2016-04-19 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9266949B2 (en) 2013-10-18 2016-02-23 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200069B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions

Also Published As

Publication number Publication date
EP0748338A4 (en) 2001-03-28
EP0748338A1 (en) 1996-12-18
CA2183550A1 (en) 1995-09-08
JPH09509835A (en) 1997-10-07

Similar Documents

Publication Publication Date Title
EP0748338A1 (en) In vitro antibody affinity maturation using alanine scanning mutagenesis
US6818748B2 (en) Cloning, expression, sequencing, and functional enhancement of monoclonal ScFv antibody against Venezuelan equine encephalitis virus (VEE)
Liu et al. Fine mapping of the antigen–antibody interaction of scFv215, a recombinant antibody inhibiting RNA polymerase II from Drosophila melanogaster
US7118915B2 (en) Muteins of apolipoprotein D
EP0894135B1 (en) Multivalent and multispecific antigen-binding protein
US7999081B2 (en) Anti-C5 alpha antibodies
AU682206B2 (en) Intracellular immunization
CA2471570A1 (en) Focussed antibody technology
WO2003105782A2 (en) Specificity grafting of a murine antibody onto a human framework
JPH06510671A (en) Production of chimeric antibodies - combinatorial approach
WO1997049429A1 (en) Genetically engineered immunoglobulins with specificity for glycated albumin
WO1992015682A1 (en) Serine-rich peptide linkers
WO2002055668A2 (en) Nucleotide sequences encoding variable regions of heavy and light chains of monoclonal antibody 1f7
JP2006523086A (en) Intracellularly expressed antibodies to carcinogenic forms of RAS
CA2395041A1 (en) Stabilizing peptides, polypeptides and antibodies which include them
JP2006521088A (en) Anti-activated RAS antibody
JPH0920798A (en) Human anti-hbs antibody
Watkins et al. Evidence for distinct contributions of heavy and light chains to restriction of antibody recognition of the HIV-1 principal neutralization determinant.
CA2269103A1 (en) Peptides binding to bone marrow stromal cell antigen
CN114349854B (en) Antibody aiming at N1 subtype NA protein of influenza A virus and application thereof
JPH06125783A (en) Recombinant anti-hiv antibody and its preparation
Böldicke et al. Human Monoclonal Antibodies to Cytomegalovirus: Characterization and Recombinant Expression of a Glycoprotein‐B‐Specific Antibody
JP2003516145A (en) Molecular display on multimeric protein backbone
CN113880947A (en) Small molecule antibody and its coding gene, preparation method and application and pharmaceutical composition
CN114805562A (en) Anti-novel coronavirus humanized nano antibody and application thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1995911994

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2183550

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 1996 702501

Country of ref document: US

Date of ref document: 19960904

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 1995911994

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995911994

Country of ref document: EP