CA1339606C - Polypeptides complementary to peptides or proteins having an amino acid sequence or necleotide coding sequence at least partially known - Google Patents

Polypeptides complementary to peptides or proteins having an amino acid sequence or necleotide coding sequence at least partially known

Info

Publication number
CA1339606C
CA1339606C CA000502748A CA502748A CA1339606C CA 1339606 C CA1339606 C CA 1339606C CA 000502748 A CA000502748 A CA 000502748A CA 502748 A CA502748 A CA 502748A CA 1339606 C CA1339606 C CA 1339606C
Authority
CA
Canada
Prior art keywords
amino acid
acid sequence
place
substituting
polypeptide
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.)
Expired - Fee Related
Application number
CA000502748A
Other languages
French (fr)
Inventor
Kenneth L. Bost
Eric M. Smith
J. Edwin Blalock
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.)
University of Texas System
Original Assignee
University of Texas System
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
Priority claimed from US06/708,001 external-priority patent/US4863857A/en
Application filed by University of Texas System filed Critical University of Texas System
Application granted granted Critical
Publication of CA1339606C publication Critical patent/CA1339606C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/18Testing for antimicrobial activity of a material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6878Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids in eptitope analysis
    • 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
    • C07K14/665Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans derived from pro-opiomelanocortin, pro-enkephalin or pro-dynorphin
    • C07K14/695Corticotropin [ACTH]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/803Physical recovery methods, e.g. chromatography, grinding

Abstract

A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein. In one aspect the method involves: (a) determining a first nucleotide sequence of a first nucleic acid coding for the biosynthesis of at least a portion of the original peptide or protein; (b) ascertaining a second nucleotide sequence of a second nucleic acid which base-pairs with the first nucleotide sequence of the first nucleic acid, the first and second nucleic acids pairing in antiparallel directions; and (c) determining the amino acid sequence of the complementary polypeptide by the second nucleotide sequence when read in the same reading frame as the first nucleotide sequence.

The complementary polypeptide whose amino acid sequence is thus determined may be obtained by diverse means such as, for example, chemical synthesis, derivation from a protein or larger polypeptide containing said amino acid sequence, or, when the second nucleic acid is DNA, inserting the second nucleotide sequence into a plasmid to form a recombinant DNA plasmid vector and transforming a unicellular organism therewith to produce a transformant unicellular organism biosynthesizing said complementary polypeptide.

The ascertainment of particular nucleotide sequences may be circumvented, in one aspect, by utilizing the relationships of amino acids having complementary hydropathies for substitutions as generally dictated by base-pairing nucleotide complementarity.

Description

13~g~06 pOLYPEPTIDES COMPLEMENTARY TO PEPTIDES OR
PROTEINS HAVING AN AMINO ACID SEQUENCE
OR NUCLEOTIDE CODING SEQUENCE AT
LEAST PARTIALLY KNOWN AND METHODS
OF DESIGN THEREFOR

The present invention relates to methods for deter-mining the structure of polypeptides having particular structural and biological activities and affinities.

The systematic design of pharmaceutical agents has currently reached a point where medicinal pharmacologists can often predict the activity of a particular pharmaco-logic agent from knowledge of its structure/functionactivity on a chemical level. This knowledqe has been particularly useful in the design of new pharmacologic aqents which are structurally related to a parent com-pound, but which exhibit new pharmacologic properties or activities.

For example, in the area of steroid biochemistry and desi~n, the structure of various steroids has been modi-~C

1339~0~i fied in numerous ways to provide for enhanced or special-ized activities. Another example of systematic druq design is in the medicinal chemistry of the synthetic penicillins: synthetic penicillins have now been designed which exhibit a number of activities not possessed by the non-synthetic penicillins. These improvements include a conference of oral activity, wide-spectrum activity, and activity against penicillinase-producing bacteria.

However, relatively little is known concerning the structure/function activities of macromolecular structures like proteins. For example, while it is known that antibodies bind to antiqens, the underlying attractive interactions are incompletely understood. Even less is known about the underlying mechanism of the response to an antiqenic challenqe of producing a protein, in the form of an antibody, which is capable of binding an antiqen.

Similarly, the interaction of peptide hormones with their hormone receptors is incompletely understood. It is known that in both the binding affinity of the peptide hormone for its receptor and the intrinsic activity of that bound hormone in "stimulating" the receptor, hormonal activity is expressed. From known structure/function relationshiPs of non-protein hormones, it has been postu-lated that binding activity and intrinsic stimulating activity involve separate structural considerations. Cer-tain chemical structures appear to provide for binding of the ligand, for example, a hormone, to its receptor. Yet other chemical structures appear to provide for "stimula-tion" of the receptor once the hormone is bound thereto.

Agents which possess binding activity, but not intrinsic stimulatinq activity, are known as "blockers" or antagonists in that they block the activity of the true hormone. An example of such a blocking agent is isopro-~3~ 1339fiO6 terenol, a well-known catecholamine beta-blocker which was designed based on some knowledge of the structure/function relationships of catecholamines with their receptors.
Similarly, agonists which both bind and activate hormonal receptors have been produced. No such structure/function relationships are entirely known for the polypeptide horm-ones. Thus, there is presently no way to accurately enable the systematic design of polypeptides capable of specifically interacting with a particular protein hormone receptor or with a particular polypeptide hormone.

All orqanisms having an intact immune system possess the biological capability to produce a class of very specialized proteins known as immunoglobulins. Immuno-globulins are produced by specialized cells of an immuno-competent organism in response to the presence of a molecule which is foreign to that organism. These foreign molecules are qenerally termed antiqens. Antigens are operationally defined as being molecules capable of eliciting the formation of a complementary antibody in a given organism. A specific antibody thus formed is capable of binding to the antigen which stimulated its formation. The biological function of a specific antibody is to bind a foreiqn antigen and thus lead to its inactivation.

Scientists have succeeded in manipulating the immune system of various organisms to provide a vast array of antibodies which have proven useful in both therapeutic and diaqnostic medicine. Recently, through the advent of hyhridoma technology, science has developed a capability to produce monoclonal antibodies which wi-ll bind with specificity to a chosen molecular structure termed the determinant. The usefulness of such specific antibodies is immense, ranqing from recent clinical experimentation which sugqest an important future role in combatinq cancer 1~39606 to an everyday clinical role for antibodies in the detec-tion of numerous disease states through blood examination.

One very interesting but largely theoretical applica-tion of antibody technology is in the area of anti-idiotypic antibodies. An anti-idiotypic antibody is a second antibody having binding capability for the idiotype or binding site of a first antibody. Such an anti-idiotypic antibody exhibits features in common with the antigen to which the first antibody binds. For example, if one generates antibodies aqainst insulin and then proceeds to generate anti-idiotypic antibodies directed against the anti-insulin antibodies, a portion (idiotype) of the anti-idiotypic antibodies will exhibit insulin-like properties. This finding lends credence to the theory that the binding site of an antibody is a three-dimen-sional neqative-image of the antiqen and that an anti-idiotypic antibody to a first antibody is therefore a positive image of the original antigen. Such observations suggest that if such interactive structures could be desiqned and produced, a whole new array of bioloqically active substances, for instance, polypeptide hormones or receptors therefor, could be developed which exhibit a wide array of new and useful activities.
Although antibody technology has advanced rapidly, it still has fundamental technological limits. Science and ~edicine, for example, must still rely on an antibody-producinq cell to generate the antibodies. Therefore, scientists have no direct control over antibody produc-tion. Such direct control would be a very important advantaqe. It would allow such advances as the production of man-made "antibodies" that could specifically interact with, or bind, not merely a selected molecule but a pre-selected portion of that molecule. The underlying basis 133960~

of the attractive interaction between the antibody andantiqen is as yet incompletely understood.

From the foregoing discussion, it is evident that antibody-producing cells have a mechanism to ascertain the chemical structure of an antigen and produce a complemen-tary chemical structure in the form of an antibody. Such complementarity results in a capability of binding to the antiqenic structure. Prior to the advent of the present invention, in order to design or construct a protein structure complementary to, and thus capable of binding with another protein structure, a knowledge of the che~-ical interactions which underlie the binding phenomenon was necessary.
All proteins or peptides primarily are polymers of monomeric amino acid units. There are, in general, twenty different amino acids, each possessing a di~ferent chem-ical structure and thus different chemical and physical properties. For example some amino acids tend to be more hydrophobic in nature while others tend to be more hydro-philic in nature. Similarly, some amino acids tend to attract certain other amino acids while repelling yet other amino acids. Therefore, within any given protein, there are a variety of both attractive forces and repul-sive forces exhibited by the individual amino acids of that protein. In addition to these interactive forces between amino acids of a given protein, there are also interactive forces between the amino acids and the sur-rounding environment. The latter forces depend on whetherthe protein resides, for example, in an aqueous or hydro-philic environment or in a non-aqueous or hydrophobic environment.

The interactive forces exhibited by the amino acids of a given protein are a major factor in determining the ~ -6- 1339606 three-dimensional, or "ternary", structure of that pro-tein. Therefore, in one view, certain regions within the protein are binding or attracting certain other regions of the same protein while other regions may be repelling certain regions within the protein. The net result is to give each protein a characteristic shape and, therefore, its functional activity.

Recently, there has been developed a means for characterizing amino acids in terms of hydropathy which reflects relative hydrophilicity and hydrophobicity (Kyte et al, (1982) J. Molec. Biol. Vol. 157, pp 105-132). A
hydropathy scale was therein derived wherein the hydro-philic and hydrophobic properties of each of the twenty amino acid side-chains was taken into consideration. A
computer program was utilized to continuously determine the average hydropathy within a polypeptide sequence of predetermined length. This study demonstrated that proteins have very distinct regions of hydrophobicity and hydrophilicity and that the intramolecular, in addition, of course, to internal disulfide bonding interaction of such reqions, can account for the three dimensional structure of the proteins.

An even more recent study has suggested that amphi-philic protein structures, that is, protein structures which contain both hydrophilic and hydrophobic amino acids and reqions, play an important role in maintaining the activity of both protein hormones and their receptors (Kaiser et al (1984) Science Vol. 223 pp 249-255). This study further su~gests that amphiphilic structures in hor-mone receptors, for example, might be complementary as a mirror-imaae of amphiPhilic structures in the hormones themselves. Therefore, the interaction between a hormone and its receptor could be mediated by a specific inter-action between the amphiphilic structure of the hormone ~7~ 13~9~0~

and a complementary amphiphilic structure of the receptor.
One way in which this concept may be envisioned is to consider the model concept of a lock and its key, with the lock confiquration representing the amphiphilic structure of the receptor and the configuration of the~key rep-resenting the complementary amphiphilic structure of the hormone aqent.

Accordingly, a means of systematically designing polypeptides which are capable of binding or interactinq with known peptides, proteins or proteinaceous receptors would be of great utility. For example, practical knowl-edge concerninq the design of receptor-interactive struc-tures of proteinaceous hormones should lead to the devel-opment of whole new classes of synthetic hormones withgreater specificity of activity. Conversely, one could desiqn and produce polypeptides which are complementary to known proteinaceous hormones and therefore capable of binding to these hormones. Such designed polypeptides may he utilized, for example, to render the complementary hormone inactive.

Similarly, such knowledqe of protein or peptide design could prove very significant for many fields of scientific research. For example, if a synthetic poly-peptide which is complementary to a protein hormone is structurally analogous to the bioloqical receptor for that hormone, then an antibody directed against that comple-mentary protein should also bind the true hormone receptor. Such antibodies would be useful in studying and isolating specific hormone receptors or portions thereof to thereby lead to an even greater understanding of hormone-receptor interactions. In addition, a synthetic protein or peptide which is complementary to a particular protein should be useful in the crystallization of that protein for the purpose, for example, of probing the -8- 1~39~06 protein structure through x-ray crystallography. Further, detoxifying polypeptides could be designed to tightly and specifically bind to toxic peptides found in nature and sometimes ingested.
S

The above illustrations are just a few of the numer-ous possible applications that synthetic protein or peptide design capabilities would enable. The ability to systematically design a polypeptide that will interact with or bind to known proteins, the desiqn being based on structural considerations of the known protein, would clearly constitute a scientific breakthrough of major pro-portions in the field of peptide and/or chemistry and medicinal pharmacology.
For purposes of clarification and consistency, the following terms are defined as to their general meaninq herein.

The term antiparallel, referring to nucleic acid pairings, indicates a directionality as to the paired nucleic acids. The original nucleic acid may be in a 5' to 3' direction where the 5' and 3' refer to positions on the sugar moeities involved in nucleotide coupling. The second nucleic acid strand base-paired or complementary to the ori~inal nucleic acid strand lies in a 3' to 5' direction when linearly aligned with the original strand having a S' to 3' directionality.

The coding nucleic acid contains the sequence of nucleotide triplets (codons) specifying a sequence of amino acids when read in a 5' to 3' direction. The noncoding (complementary) nucleic acid (or nucleic acid strand) is complementary to the coding nucleic acid (or nucleic acid strand), the strands lyin~ or base-pairinq in an antiparallel direction.

- -9- 1339~06 The term hydropathic complementarity, referring to the hydropathic scores ~a relative measure of hydro-philicity and hydrophobicity) of amino acids indicates a low hydropathy corresponding to a high hydropathy and vice versa.

In referring to structures comprising amino acids, they are generally referred to as peptides, polypeptides or proteins, this order designating an increase in size between, for example, dipeptides, oliqopeptides, and proteins containinq many hundred of amino acids.

The term complementary, or complement, as used herein has a meaning based upon its context of usage. For example, complementary bases or nucleotides are those characteristically forming hydrogen bonds (G-C and A-T or A-U), complementary codons nucleic acids or strands thereof are hydrogen bonded polynucleotide components of a double nucleic acid strand such of that in the classically defined double helix for example complementary amino acids usually having hydropathic complementarity are those directed by members of a pair of complementary codons.

Co~plementary peptides or polypeptides and their related original peptide or protein are a pair of peptides directed by complementary nucleotide or amino acid sequences, and characteristically have a binding affinity between members of a pair. Polypeptides complementary to a peptide or at least a portion of a protein, for example, have a binding affinity for the peptide or protein por-tion. While peptide binding affinities are incompletely understood, they may, in part at least, be explained by the concept of amphiphilic secondary structure described by Kaiser et al (Science (1984) Vol. 223 pp. 249-255).

-lO- 133~606 A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an oriqinal peptide or protein has not, before now, been discovered. In one aspect the method involves:
(a) derivinq a first nucleotide sequence of a first nucleic acid potentially coding for the biosynthesis of at least a portion of the oriqinal peptide or protein; tb) determininq a second nucleotide sequence of a second nucleic acid which base-pairs with the first nucleotide sequence of the first nucleic acid, the first and second nucleic acids pairing in antiparallel directions; and (c) determining the amino acid sequence of the complementary polypeptide by the second nucleotide sequence when read in the same reading frame as the first nucleotide sequence.
The complementary polypeptide whose amino acid sequence is thus determined may be obtained by diverse means such as, for example, chemical synthesis, derivation from a protein or larqer polypeptide containing said amino acid sequence, or, when the second nucleic acid is DNA, inserting the second nucleotide sequence into a plasmid to form a recombinant DNA plasmid vector and transforming a unicellular orqanism therewith to produce a transformant unicellular organism biosynthesizinq said complementary polypeptide.

In one aspect the present invention is related to the desiqn and production of polypeptides capable of specifi-cally interacting with selected tarqet peptide structures of known amino acid sequence.

Figure 1 graphically depicts the relationship of hydropathic scores of amino acids specified by a nucleo-tide strand containinq coding information and its anti-parallel base-paired complementary or noncoding nucleotide strand. The triplet nucleotide code of each strand was -ll- 133960fi read in the 5' to 3' direction. The hydropathic scores of the coded amino acids are plotted against the average hydropatic scores of complementary coded amino acifls.

Fiqure 2 graphically depicts the relationship of hydropathic scores of amino acids coded by a coding nucleotide strand and its antiparallel base-paired non-coding nucleotide strand. The triplet nucleotide code of the coding strand was read in the 5' to 3' direction, while that of the noncoding strand, in the 3' to 5' direction. The hydropathic scores of the coded amino acids are plotted against the hydropathic scores of complementarily coded amino acids.

Figure 3 graphically depicts the binding of free ACTH
to microtiter wells coated with HTCA (a complementary peptide to ACTH) or insulin. Bound ACTH was measured by an enzy~e-linked immunoabsorbent assay.

Figure 4 qraphically depicts the bindinq of ACTH to microtiter wells each coated with HTCA (3.7 nmol/well).
Each ACTH addition contained 3.7 nmol soluble ACTH and was premixed with the amounts of soluble HTCA designated on the ahscissa. ~ound ACTH was measured by enzyme-linked immunoabsorbent assay and free ACTH calculated by the difference between total ACTH and bound ACTH.

Figure 5 graphically depicts the binding of antibody for HTCA (anti-HTCA) to affixed mouse adrenal (Y-l) cells and the inhibition of this binding by ACTH.

Figure 6 qraphically depicts the eluent from gel chromatoqraphy of mouse adrenal (Y-l) cell components which had previously bound to anti-HTCA.

-12- 13~960~

Figure 7 qraphically depicts the binding of gamma (~) -endorphin from various amounts added to microtiter wells coated with: a peptide (gamma-odne) coded by the nucleo-tide strand complementary for bovine qamma endorphin, 40uq/well; insulin, 20 units/well; or bovine serum albumin (BSA), 200 ug/well.

Fiqure 8 depicts nucleotide and amino acid sequences for epidermal growth factor (EGF), EGF receptor. The nucleotide sequence complementary to the nucleotide sequence for EGF receptor and the amino acid sequence coded by the complementary nucleotide sequence when read in the 3' to 5' direction are also depicted. For the sequences of EGF and EGF receptor, the lower numbered positions represent the 5' nucleotide direction and the amino-terminal amino acid direction. For the sequences of the complementary message to the EGF receptor, the lower numbered positions represent the 3' nucleotide direction and the amino-terminal amino acid direciton. Homoloqous sequences are boxed.

Figures 9A and 9B depict certain nucleotide and amino acid sequences of: peptide hormones [EGF, interleukin-2 (IL-2) and transferrin (TF)]; peptide hormone receptors [EGF receptor, IL-2 receptor and TF receptor]; and comple-mentary message to the receptors. For the peptide hormone and receptor sequences, the lower numbered positions represent the 5' nucleotide direction and the amino-terminal amino acid direction. For the sequences of the complementary messaqe, the lower numbered positions repre-sent the 3' nucleotide direction and the amino-terminal amino acid direction. The complementary nucleotide was read in the 3' to 5' direction to produce the correspond-ing amino acid sequence.

-13- 13~9606 The interactions of biologically siqnificant mole-cules are a basis of intercellular and interorgan communi-cations. When the particular bioloqically siqnificant communicating molecules are, for example, peptide hormones and peptide-containing cellular receptors therefor, a basis and rational explanation ~or their communicative interactions have long been sought.

A previously unobserved and fundamental relationship has been found, as described herein, to exist between antiparallel base-pairing strands of nucleic acids. In one aspect, this relationship may give rise to pairs of peptides where each member of a particular pair has an affinity for the other member. The basic relationship is demonstrated in Table 1 where the various codons and their complementary (i.e. base pairing) codons are presented.
The codons of a coding strand, (e.g. that strand contain-ing the codinq information describing an amino acid se-quence) are represented as being read from left to right (the 5' to 3' direction). The codons of the complementary (i.e. noncodinq) antiparallel base-paired strand are also read from in the 5' to 3' direction. Noncoding and coding nucleic acid strands pair when lyinq in an antiparallel direction (e.q. coding strand from left to right being 5' to 3' and noncodinq strand from left to riqht being 3' to 5') so that the paired codons are viewed lyinq in an opposite observable direction (e.q. left to riqht vs.
right to left) when read in the 5' to 3' direction. The codons given in Table 1 have been grouped suggestively by hydropathy as defined by Kyte et al. This specific grouping is used for illustrative purposes only and should not be viewed as restrictive of the scope of the present invention. As can be seen in Table 1, the complementary codons pairing with codons for the hydrophobic (high hydropathy) amino acids exhibit a tendency to code for hydrophilic (low hydropathy) amino acids. The reciprocal -14- 13 39~0 situation is shown with codons of the hydrophilic amino acids. For the slightly hydrophilic amino acids ~slightly negative hydropathy), similar amino acids are coded for by the complementary codons. These results are shown in qraphical form in Figure 1. This relationship has great biological siqnificance as described hereinafter.

--1 s--AMINO ACIDS WHOSE CODONS ARE
COMPLEMENTARY TO THOSE OF THE:

Coding Strand Noncoding Strand Codon Amino Acid Codon Amino Acid (1) Hydrophohic Amino Acids AUU Isoleucine AAU Asparagine AUC Isoleucine GAU Aspartic acid ~UA Isoleucine UAU Tyrosine 20 GUU Valine AAC Asparagine GUC Valine GAC Aspartic acid GUG Valine CAC Histidlne GUA Valine UAC Tyrosine 25 CUU Leucine AAG Lysine CIJC Leucine GAG Glutamic acid CUG Leucine CAG Glutamine UUG Leucine CAA Glutamine 30 UUU Phenylalanine AAA Lysine UUC Phenylalanine GAA Glutamic acid UGU Cysteine ACA Threonine UGC Cysteine GCA Alanine AUG Methionine CAU Histidine GCG Alanine CGC Arginine GCU Alanine AGC Serine 40 GCC A1anine GCC Glycine GCA Alanine UGC Cysteine -16- 133960fi TABLE 1 (Continued) Coding Strand Noncodin~ Strand Codon Amino Acid Codon Amino Acid (2) Hydrophilic Amino Acids CGC Arginine GCG Alanine CGU Arginine ACG Threonine CGA Arginine UCG Serine AGA Ar~inine UCU Serine 15 CGG Arginine CCG Proline AGG Ar~inine CCU Proline AAG Lysine CUU Leucine AAA Lysine UUU Phenylalanine AAU Aspara~ine AUU Isoleucine AAC Asparagine GUU Valine GAU Aspartic acid AUC Isoleucine 25 GAC Aspartic acid GUC Valine CAA Glutamine UUG Leucine CAG Glutamine CUG Leucine 30 GAG Glutamic acid UUG Leucine GAA Glutamic acid UUC Phenylalanine CAC Histidine GUG Valine CAU Histidine AUG Methionine -17- 1~3~60fi TABLE 1 (Continued) Codinq Strand Noncoding Strand Codon Amino Acid Codon Amino Acid (3)Slightly Hydrophilic Amino Acid GGU Glycine ACC Threonine GGA Glycine UCC Serine GGG Glycine CCC Proline GGC Glycine GCC Alanine ACC Threonine GGU Glycine ACU Threonine AGU Serine ACG Threonine CGU Arginine ACA Threonine UGU Cysteine UGG Tryptophan CCA Proline UCC Serine GGA Glycine AGU Serine ACU Threonine 25 UCG Serine CGA Arginine UCU Serine AGA Arginine AGC Serine GCU Alanine UAU Tyrosine AUA Isoleucine 30 UAC Tyrosine GUA Valine CCC Proline GGG Glycine CCA Proline UCC Tryptophan CCU Proline AGG Arginine 35 CCG Proline CGG Arginine The paired codons (nucleotide triplets) in Table 1 result from comparing hypothetical coding nucleic acid strands (RNA in this case) and non-coding nucleic acid strands ~RNA paired in an antiparallel direction. Both strands were then read in the 5' to 3' direction and in the same reading frame to obtain the original codons and their complementary (base-paired) codons.

Of the possible 20 complementary codons for the hydrophobic amino acid-coding codons, only two (GCA and UCG) code for hydrophobic amino acids. Of the possible 18 complementary codons for the hydrophilic amino acid-coding codons, 13 coded for hydrophobic amino acids and 5 coded for slightly hydrophilic amino acids.

Of the possible 20 complementary codons for the slightly hydrophilic amino acid codinq codons, 10 coded for sliqhtly hydrophobic amino acids, 5 coded for strongly hydrophilic amino acids and 5 coded for strongly hydro-phobic amino acids, the net comparative effect being little chanqe in hydropathic character.

Table 2 lists the coded amino acids and their respec-tive complementarily coded amino acids of Table 1 and includes their hydropathic scores (Kyte et al, 1982).

133~6~~

HYDROPATHIC SCORES OF AMINO ACIDS AND

AVERAGE
AMINO ACIDS SCORECOMPLEMENTS SCORES SCORE
ILE +4.5 ASN -3.5 ASP -3.5 TYR -1.3 -2.8 VAL +4.2 ASN -3.5 HIS -3.2 TYR -1.3 -2.9 LEU +3.7 LYS -3.9 GLU -3.5 GLN -3.5 -3.6 PHE +2.7 LYS -3.9 GLU -3.5 -3.7 CYS +2.5 THR -0.7 ALA +1.8 +0.6 MET +1.9 HIS -3.2 ALA +1.8 ARG -4.5 SER -0.9 GLY -0.4 CYS +2.5 -0.8 ARG -4.5 ALA +1.8 THR -0.7 SER -0.9 PRO -1.6 -0.5 LYS -3.9 LEU +3.7 PHE +2.7 +3.2 ASN -3.5 ILE +4.5 VAL +4.2 +4.4 ASP -3.5 ILE +4.5 VAL +4.2 +4.4 GLN -3.5 LEU +3.7 +3.7 GLU -3.5 LEU +3.7 PHE +2.7 +3.2 -20- i339~~ 6 TABLE 2 (Continued) AVERAGE
AMINO ACIDS SCORE COMPLEMENTS SCORES SCORE
HIS -3.2 VAL +4.2 MET +1.9 +3.1 GLY -0.4 THR -0.7 SER -0.9 PRO -1.6 ALA +1.8 -0.1 lS THR -0.7 GLY -0.4 SER _o.g ARG -4.5 CYS +2.5 -0.8 TYP -0.9 PRO -1.6 -1.6 SER -0.9 GLY -0.4 THR -0.7 ARG -4.5 ALA +1.8 -1.6 TYR -1.3 ILE +4.5 VAL +4.2 +4.4 PRO -1.6 GLY -0.4 TRP -0.9 ARG -4.5 -2.5 As shown in Table 2 and qraphically illustrated in Figure 1, a general relationship exists as exemplified by sets of amino acids. For example, a first set of amino acids directed (i.e. coded for ) by a first qroup of codons and a second set (complementarily coded) of amino acids are directed by a second group of codons comple-mentary to the first group of codons. A relationshipbetween the first set of amino acids and the second a set of amino acids is found which may be characterized as hydropathically inverse. In one instance, complementarily coded hydrophilic (low hydropathy) amino acids are directed by codons complementary to those coding for the 1:~39~)06 hydrophobic (high hydropathy)amino acids. This relation-ship may be termed hydropathic complementarity.

Figure 1 shows a plot of data from Table 2 showing the hydropathic scores of the amino acids directed by codons of a coding nucleic acid strand versus the average hydropathic scores of the amino acids complementarily directed by the codons of the the complementary noncoding strand. A linear regression analysis of this data results in a correlation coefficient of -0.77. A similar pattern is observed when calculated by another hydropathic scoring system which has somewhat different values for tryptophan, tyrosine, glutamine and asparagine (data not shown, Hopp et al,, Proc. Natl. Acad. Sci. (1981) Vol. 78 pp. 3824-3828). Thus the noncoding strand-directed amino acid hydropathic scores tend to be inversely related to the coding strand amino acid hydropathic scores and this relationship is not random and could be found with any scoring system reflecting amino acid properties reflecting hydrophobic and hydrophilic tendencies, alone or in combination with other physical properties of amino acids.

Interestingly, a similar relationship also arises when the complementary codons are read in the 3' to 5' direction. The coding relationships of complementary codons read in the 3' to 5' direction are shown in Table 3.

-22- 1~39606 AMINO ACIDS WHOSE CODONS ARE COMPLEMENTARY TO THOSE OF:

Coding Strand Noncoding Strand Codon Amino Acid Codon Amino Acid (1) Hydrophobic Amino Acids AUA Isoleucine UAU Tyrosine GUU Valine CAA Glutamine GUC Valine CAG Glutamine GUG Valine CAC Histidine GUA Valine CAU Histidine UuA Leucine AAU Asparagine UUG Leucine AAC Asparaglne CUU Leucine GAA Glutamic Acid CUC Leucine GAG Glutamic Acid 25 CUA Leucine GAU Aspartic Acid CUG Leucine GAC Aspartic Acid UUU Phenylalanine AAA Lysine UUC Phenylalanine AAG Lysine UGU Cysteine ACA Threonine UGC Cysteine ACG Threonine AUG Methionine UAC Tyrosine GCU Alanine CGA Arginine GCC Alanine CGG- Arginine GCA Alanine CGU Arginine GCG Alanine CGC Arginine (2) Hydrophilic Amino Acids CGU Arginine GCA Alanine CGC Arginine GCG Alanine 45 CGA Arginine GCU Alanine CGG Arginine GCC Alanine AGA Arginine UCU Serine AGG Arginine UCC Serine 50 AAA Lysine UUU Phenylalanine AAG Lysine UUC Phenylalanine 1339~01~

TABLE 3 (Continued) Coding Strand Noncoding Strand Codon Amino Acid Codon Amino Acid Codon Amino Acid Codon Amino Acid AAU Asparagine UUA Leucine AAC Asparagine UUG Leucine GAU Aspartic Acid CUA Leucine 15 GAC Aspartic Acid CUG Leucine CAA Glutamine GUU Valine CAG Glutamine GUC Valine GAG Glutamic Acid CUC Leucine 20 GAA Glutamic Acid CUU Leucine CAC Histidine GUG Valine CAU Histidine GUA Valine (3) Slightly Hydrophilic Amino Acids GGU Glycine CCA Proline GGC Glycine CCG Proline GGA Glycine CCU Proline 30 GGG Glycine CCC Proline ACC Threonine UGG Tryptophan ACG Threonine UCG Cysteine ACA Threonine UGU Cysteine UGG Tryptophan ACC Threonine UCU Serine AGA Arginine UCC Serine AGG Arginine 40 UCA Serine AGU Serine UCG Serine AGC Serine AGU Serine UCA Serine AGC Serine UCG Serine 45 UAU Tyrosine AUA Isoleucine UAC Tyrosine AUG Methionine CCU Proline GGA Glycine CCC Proline GGG Glycine 50 CCA Proline GGU Glycine CCG Proline GGC Glycine As shown in Table 3, of the 20 possible codons complementary to the codons for hydrophobic amino acids, when read in the 3' to 5' direction, none coded for hydrophobic amino acids, 16 coded for hydrophilic amino acids and 4 ( UAU, ACA, ACG and UAC ) coded for slightly hydrophilic amino acids.

Of the 18 possible codons complementary to the codons for the strongly hydrophilic amino acids, when read in the 3' to 5' direction, none coded for strongly hydrophilic amino acids, 16 for hydrophobic amino acids and two (UCU
and UCC) for slightly hydrophilic amino acids.

Table 4 lists the hydropathic scores of amno acids and their complements (i.e. amino acids complementarily coded or coded by respective complementary codons) described in Table 3.

HYDROPATHIC SCORES OF AMINO ACIDS AND

AMINO ACIDSCORE COMPLEMENTS SCORES
ILE +4.5 TYR -1.3 VAL +4.2 GLN -3.5 HIS -3.2 LEU +3.7 ASN -3.5 GLU -3.5 ASP -3.5 PHE +2.7 LYS -3.9 CYS +2.5 THR -0.7 MET +1.9 TYR -1.3 ALA +1.8 ARG -4.5 ARG -4.5 ALA +1.8 SER -0.9 LYS -3.9 PHE +2.7 ASN -3.5 LEU +3.7 ASP -3.5 LEU +3.7 GLN -3.5 VAL +4.2 GLU -3.5 LEU +3.7 HIS -3.2 VAL +4.2 GLY -0.4 PRO -1.6 THR -0.7 TRP -0.9 CYS +2.5 TRP -0.9 THR -0.7 SER -0.9 ARG -4.5 SER _o.g TYR -1.3 ILE +4.5 MET +1.9 PRO -1.6 GLY -0.4 Of the possible complementary codons to the codons coding for slightly hydrophilic amino acids, when read in the 3' to 5' direction, 14 code for slightly hydrophilic amino acids, 2 (ACA and ACG) code for strongly hydrophilic amino acids and 4 (UCG, UGU, AUA and AU~) code for hydro-phobic amino acids. The net effect here being little change in the average hydropathic character of the non-coding strand amino acids.

Figure 2 shows a plot of the hydropathic scores of the coding strand amino acids versus the hydropathic scores of the noncoding strand amino acids. A linear regression analysis of this data results in a correlation coefficient of -0.77. Thus, as was the case for the 5' to 3' direction, in the 3' to 5' direction, the noncoding strand amino acid hydropathic scores are inversely related to those of the coding strand and this relationship is not random.

These relationships of information contained in the genetic code demonstrate a hydropathic complementarity of amino acids. Codons, when read in the 5' to 3' direction, for hydrophilic and hydrophobic amino acids were generally complemented by codons for hydrophobic and hydrophilic amino acids, respectively. The average tendency of codons for "uncharged" (slightly hydrophilic) amino acids was to be complemented by codons for "uncharged" amino acids.

As demonstrated by these observations an almost identical pattern results when the complementary nucleo-tide codon is read in the 3' to 5' rather than the 5' to 3' direction. Since, regardless of the reading direction, the second nucleotide of the complementary codon never changes, this second nucleotide of the triplet codon is the principal determinant for the hydropathic comple-mentarity of amino acids which are specified by comple-1~39606 mentary codons. This seems to largely result from the fact that the preponderance (6 out of 7) of hydrophilic amino acids have adenine as their second nucleotide codon while the complementary nucleotide uridine, is the second nucleotide of the triplet codon for most (5 of 7) hydro-phobic amino acids. One of the 2 exceptions to the above in the hydrophobic group (alanine) does not seriously vitiate the above generality as it has a second base, cytosine, while the second base for the single exception in the hydrophilic group (arginine) has a second base, guanine. Hence, there is a virtually perfect interchange of hydrophobic and hydrophilic amino acids whether the complementary codon is read in the 5' to 3' or 3' to 5' direction. Of the six uncharged (slightly hydrophilic) amino acids with the exception of tyrosine, the second hase of the respective codons is either a G or C. Hence, the codons for this group will usually result in a similar type of amino acid regardless of the direction in which the complementary codon is read.
Table 5 lists amino acids whose codons contain a particular second (middle) base.

-28- ~ 3 3g~06 AMINO ACIDS HAVING A PARTICULAR

-SECOND BASE AMINO ACIDS
oF RNA CODON
U ILE
VAL
LEU
PHE
MET
A LYS
ASN
ASP
GLN
GLU
HIS
TYR
G CYS
ARG
GLY
TRP
SER
C THR
SER
PRO
ALA

1339~06 The group of amino acids (U group) directed by a uridine second base have a complementarily coded group of amino acids (A group) coded by an adenine second base, and vice versa. The cytosine and guanine directed groups tC
group and G group respectively) have the same relationship.
Table 6 lists the hydropathic scores of amino acids directed by codons having a particular second base and, for convenience separately shows corresponding scores for the complementarily coded amino acids (complement).
Again, the hydropathically complementary relationship is illustrated.

_30- 1 3-3 g ~ ~

HYDROPATHIC SCORES OF At1INO ACIDS AND THEIR

SecondAverage Hydropathic Base Scores l0 Group Coded Comple~ent U ILE +4.5 VAL +4.2 LEU +3.7 PHE +2.7 MET +l.9 +3.4 -3.2 A LYS -3.9 ASP -3.5 GLN -3.5 HIS -3.2 TYR -l.3 -3.2 +3.4 G CYS -2.5 ARG -4.5 GLY -0.4 TRP -0.9 SER -0.9 -0.8 -0.4 C THR -0.7 PRO -l.6 ALA +1.8 -0.4 -0.8 -31- 1339~06 Clearly, from Tables 2, 4 and 6 it can be seen that peptides and their complements are related by a general inversion of hydropathic nature on an amino acid by amino acid basis, when the sequences are aligned in a parallel or anti-parallel manner depending on the method of genera-tion. Preferred embodiments of the present invention, as demonstated by utilization of the specific codon relation-ships shown in Table 1 and Table 3 are special cases of a more generally defined method to generate complementary peptides.

When nucleic acid sequences are not known, the general methods based on second base complementarity or hydropathic inversion may be used to generate homologs of the specifically preferred complementary peptides. For example, when an amino acid sequence but not the partic-ular codons for all or a portion of a protein or peptide is known, a complementary peptide may be designed based upon the general relationships shown in Table 6. For an the amino acid in the original protein or peptide sequence having a second codon base of uridine (U group amino acid), an amino acid for the A group is substituted and vice versa. For an amino acid in the protein or peptide sequence having a second codon base of cytosine (C group), an amino acid from the guanine (G group) is substituted and vice versa. After these substitutions the sequence of amino acids thus obtained will be complementarey to repective portions of the original peptide or protein.

Tables 1, 3 and 6 can be used in a general manner when the nucleic acid sequences are not known. In such cases, for an amino acid in the original peptide or protein sequence, an amino acid is substituted from the corresponding set of non-coding strand amino acids. After these substitutions, the sequence of amino acids thus -32- 133960~

obtained will be complementary to the repective portions of the original peptide or protein.

As a further extention of the principles of the present invention, the specific directionality of the complementary amino acid sequence may not be critical. As is clear to one skilled in the art upon study of the entire description presented herein, the juxtaposition of amino acids in construction of complementary polypeptide may be directionally oriented in either of two ways.
Relative to the amino acid sequence directing positioning of amino acids having particular hydropathic character, the amino terminal and carboxy terminal directions are interchangeable, both constructions giving rise to comple-mentary polypeptides. In simpler form, for one example,if the amino terminal end of a particular amino acid sequence contains a valine (second codon base = U), then a complementary amino acid sequence would contain, at the amino terminal end or the carboxy terminal end, an amino acid having a second codon base A (LYS, ASN, ASP, GLN, GLU, HIS, or TYR), using the general method based on Table 6.

The genetic code may have arisen during evolution as a result of the chemical similarity of anticodonic bases and their respective amino acids. Perhaps this similarity resulted in the patterns observed herein. A functional and evolutionary advantage to this phenomenon may reside in the fact that the second base of codons for hydro-pathically similar amino acids is the same. Perhaps,prior to the advent of the directionality of nucleic acid reading, an amino acid from the same hydropathic group would be present and thus the resulting peptides or proteins would be grossly similar in conformation, whether nucleic acids were read 5' to 3' or 3' to 5'.

_33_ 13.39~06 The present invention relates , in a major aspect, to the discovery that polypeptides complementary to at least a portion of an original peptide or protein having known amino acid sequence or nucleotide coding sequence and has binding affinity to the oriqinal peptide or protein may be designed and obtained. If the amino acid sequence of at least a portion (for example four to five amino acids) of an original peptide or protein is known, information of that sequence may be used in several ways to determine the design of a complementary polypeptide.

A preferred way of designing a complementary poly-peptide utilizes the amino acid relationships delineated in Table 3. Accordingly, for any position of isoleucine in an ascertained amino acid sequence of all or part of the original peptide or protein, substitute tyrosine. As one further example, for each valine substitute glutamine or histidine. The residual 18 amino acids are also substituted according to the relationships illustrated in Table 3. As shown subsequently herein (Examples 2A, 2B
and 2C, for example,) when peptide hormone-receptor site amino acid sequences are utilized as original peptides or proteins, statistically significant and unique codon directions are given for portions of the peptide hormones which characteristically bind at those receptor sites and are thus complementary thereto. By further examination of Examples 2A, 2B and 2C and also of Figures 8, 9A and 9B, it is shown that more preferable substitutions were made therein for specific amino acids based on known nucleotide sequence, for example serine was substituted for arginine and serine or cysteine was substituted for threonine.

Another preferred method of designing complementary polypeptides involves usage of the amino acid relation-ships presented in Table 1. Accordingly, however, anamino acid sequence of the original peptide, protein or - 133~60ti portion thereof desired is read from the carboxy terminal direction. This carhoxy terminal direction is to substi-tutinqly correspond (i.e. give the directions for amino acid emplacement) to the amino terminal direction of the complementary polypeptide. Once this reversal of order is attained, substitutions may be made according to the amino acid relationships shown in Table 1. For example, in place of each isoleucine is substituted a tyrosine, asparagine or aspartic acid. Further substitutions for the other 19 amino acids may take place as directed by the amino acid relationships of Table 1.

As subsequently demonstrated in Examples lA to lH, when polypeptides complementary to gamma endorphin and lS ACTH were designed following a preferred variant of the latter method and obtained, specific amino acid substitu-tions based on known nucleotide sequences were made. For example, for valine, aspartic acid or histidine was substituted; for leucine- lysine or glutamic acid was substituted; for phenylalanine- glutamic acid; for arginine-alanine or proline; for lysine-leucine; for histidine-valine; for glycine-proline or alanine; for threonine-glycine or arginine; for serine-glycine, argi-nine or alanine; for tyrosine-valine; and for proline-glycine or arginine.

Another preferred method to select a specific set ofcomplementary amino acids from the general second base grouping is given below. For each amino acid, generate a list of all of the possible complementary amino acids that could be generated from all the possible condons reading 3'-5' and 5'-3' for the selected amino acid. From this list, select the amino acid that occurs most frequently.
For example, for valine there are 4 codons which can be read in both directions to generate a set of possible complements to valine.

133g~06 Valine Codon Complement 5' Translation 3' Translation GUC CAG ASP GLN
GUG CAC HIS HIS
GUA CAU TYR HIS

From this list, HIS is selected as the complement to VAL. Such selections will be referred to herein as consensus complements. The following table gives the selections for each of the amino acids.

-36- 13~ 06 CONSENSUS COMPLEMENT SUBSTITUTIONS

Amino Acid Consensus Complement ILE TYR
VAL HIS
LEU GLU
PHE LYS
MET TYR, HIS
LYS PHE
ASN LEU
ASP LEU
GLN LEU, VAL
GLU LEU
HIS VAL
TYR ILE
CYS THR
ARG ALA
GLY PRO
TRP THR, PRO
SER SER, ARG
THR CYS
PRO GLY
ALA ARG

Those familiar with the production and handling of peptides will recognize that the presence of a cysteine residue in a peptide sequence may lead to problems of oxidation and dimerization in some situations. For those reasons, some investigators may wish to replace cysteine residues in complementary peptide sequences with another hydropathically neutral amino acid such as serine, and such substitutions are contemplated by this invention.

An additional preferred method to select a specific set of complementary amino acids from the general second 1339~06 base grouping is to determine the most frequently used (species specific) codon for each amino acid and to translate the complementary codon either 5' to 3' or 3' to 5' (referred to herein as "frequency-based complements").
Thus, for each amino acid (and each species) two frequency-based complementary amino acids may be derived and used to ~enerate complementary peptide sequences. I~
a stop codon (siqnaling the cessation of translation in cells) is encountered in the above process. Then the second most frequently used codon for the particular amino acifl can be used (and so on). Table 6B below gives the frequency-based complements for the amino acids using human codon frequencies as determined by Grantham et. al.
(Nuc. Acids. Res., 9, p. r43-r75, 1981).

13~06 TABLE 6B:

HUMAN FREPUENCE BASED COMPLEMENT SUBSTITUTIONS

Amino Acid 5'-3' Complement 3'-5' Complement ILE ASP TYR
VAL HIS HIS
LEU GLN ASP
PHE GLN LYS
MET HIS TYR
LYS LEU PHE
AsN VAL LEU
ASP VAL LEU
GLN LEU VAL
GLU LEU VAL
HIS VAL VAL
TYR VAL MET
CYS ALA THR
ARG PRO SER
GLY ALA PRO
TRP PRO THR
SER GLY ARG
THR GLY TRP
PRO GLY GLY
ALA GLY ARG
Another alternative and preferred method to select a specific set of complementary amino acids from the general second base qrouping is to determine the most commonly used codon for each second base group. Since codon usage frequency varies from species to species, this, approach may give different selections for different species.
Using the human codon usage frequencies generated by Grantham et al. (Nuc. Acid. Res., 9, p. r43-r75, 1981), the selected simplified complementary amino acids in the following table were qenerated (said complementary amino acids being referred to herein as "simplified complements").

133960~

HUMAN SIMPLIFIED COMPLEMENT SUBSTITUTIONS

Amino Acid Simplified Complement ILE, VAL, LEU, PHE, MET GLU
LYS, ASN, ASP, GLN, GLU, HIS, TYR LEU
CYS, ARG, GLY, TRP ALA
10 SER, THR, PRO, ALA GLY

In some cases, a peptide may be self-complementary due to special features of its sequence. A sequence is self-complementary when it contains a point of inversion in its second base sequence. For example:

N-terminus Glu-Leu-Glu-Leu Peptide sequence 5' terminus A - U- A - U

3' terminus U - A - U - A Complement sequence C-terminus Leu-Glu-Leu-Glu Clearly, this tetrapeptide is its own complement with antiparallel orientation. This peptide has a number of other antiparallel complements with different sequences of amino acids which still have the same second base sequence. All of the antiparallel complements could be viewed as second base analogs.

The special nature of these sequences can lead to complexities of analysis of certain experimental results when a self-complementary peptide is present along with other antiparallel complements. Other antiparallel complements could have properties of conventional analogs (agonist or antagonist effects) or could have properties normally associated with complements (non-conventional antagonist and antibodies that are conventional agonists 133960~

and antagonists). Regardless of the detailed mode of action of complements in these special cases, the complementary peptide principle can be used to generate new molecules with desirable, bioactive properties.

The complementary polypeptides whose sequence was determined by any of the above described methods based upon the original amino acid sequence or nucleotide codon sequence may then be obtained by chemical synthesis, directed biological synthesis or derivation (e.g. by excision) from peptides or proteins which include the determined amino acid sequences.

The complementary amino acid relationships described herin permit the design, construction and use of many polypeptide structures comprising amino acid sequences complementary to desired sequences of amino acids. The complementary amino acid sequence may be an entire peptide or polypeptide, as, for example shown herein with HTCA and gamma odne, which are respectively complementary to the entire sequence of the target peptides ACTH ( 1-24) or gamma-endorphin.

In particular applications, a complementary peptide may be bonded to a larger molecule by such techniques as chemical cross-linking or incorporation in a larger polypeptide structure. Complementary polypeptides may also be attached to a solid matrix for uses such as affinity chromatography.
In the practice of the present invention, particu-larly when a nucleotide sequence coding for a target peptide is known, it may be utilized to direct the amino acid sequence of complement. In this situation particular codons for isoleucine (AUU, AUC), leucine (UUA, CUA) threonine (ACU) or serine (UCA), may give rise to comple--41- 13.396~6 mentary codons which, when read in the 5' to 3' or 3' to 5' direction, are stop codons coding for cessation of protein synthesis. In this situation, the second base of the stop codon would be used to select an amino acid of appropriate hydropathic complementary character (from the groups shown in Tables 5 and 6). The choice from a particular group may be preferably narrowed by optimizing hydropathic complementarity. For example, with an ILE
(+4.5 hydropathic score) codon (AUU or UUA) a LYS (-3.9 hydropathic score) might be chosen from the complementary second base A-group; with a serine (-0.9 hydropathic score) codon (UCA) or threonine (-0.9 hydropathic score) codon (ACU) a tryptophan (-0.9) or serine (-0.9) may be chosen from the second base G-group.
The scope of the present invention may be further described by the application of particular embodiments.
For example, luteinizing hormone releasing hormone (LH-RH) is a decapeptide whose coding nucleotide sequence is unknown but has the amino acid sequence shown in the topmost line of Table 7.

LH-RH

H2N- Glu- His- Trp~ Ser- Tyr- Gly- Leu- Arg- Pro- Gly- COOH
Table 1 Leu Val Pro Gly Ile Thr Gln Ala Gly Thr Alternatives Phe Met Thr Val Ser Thr Tyr Ser Arg Pro Ser Pro Pro Ala Pro Ala Table 3 Leu Val Thr Arg Ile Pro Asn Ala Gly Pro Alternatives Ser Met Glu Ser Asp Table 6 Ile Thr Thr Ile Thr Lys Thr Cys Thr Alternatives Val Ser Ser Val Ser Asn Pro Arg Ser Leu Pro Pro Leu Pro Asp Ser Gly Pro Phe Ala Ala Phe Ala Gln Ala Trp Ala NMet Met Glu Ser His Tyr o It is confidently predicted, based upon the knowledge and principles described herein that most, if not all, of the complements produced by the methods relating to Table 7 will display an affinity for LH-RH and prove useful in modulating the effects of this hormone.

The selection of the specific complementary peptide or ~eptides according to the invention having the most desirable biological or biochemical properties or effects in any given case will depend on several factors including the nature of the biological system in which the complementary peptide is to be used, whether diagnostic or therapeutic utility is desired and the type of biological effect and mode of action which is anticipated for the complementary peptide in the targeted biological syste~.
Depending on the desired effect, some comPlementary peptides may be preferred over others. The following approach suqgests one of many possible design schemes that can be used to generate complementary peptide sequences that will qive a response in a biological, diagnostic, or physical assay at a concentration of complementary peptide of 10 4 molar or lower. This complementary peptide concentration of 10 molar or lower (herein referred to as the "minimum complementary peptide binding activity") is a useful parameter in selectinq suitable complementary peptidefi accordinq to the invention since from a practical standpoint, concentrations above that established for the minimum complementary peptide binding activity may involve impractical therapeutic dosages and lead to peptides whose bindin~ is not specific enough to be useful in therapeutic or diaqnostic applications.

The selection approach hereinafter described allows those skilled in the art to readily obtain complementary peptides which are characterized by the minimum complementary peptide binding activity. Specifically, if nucleic acid sequences are known, then four complementary peptides can be designed directly from the above disclosure (translating the complementary nucleic acid sequence either 5' to 3' or 3' to 5' and orienting the subsequent sequence of amino acids either amino-terminal to carboxy-terminal or carboxy-terminal to amino-terminal). If none of these complementary peptides has the desired level of activity or if the nucleic acid sequence of the target is not known, then two complementary peptides can be designed from an amino acid target sequence using the consensus complement approach (Table 6A) by orienting the complement amino acid sequence either amino-terminal to carboxy-terminal or carboxy-terminal to amino-terminal. If the desired level of activity has not been reached, then four additional complementary peptides can be designed using the codon usage frequency complement approach (e.g., Table 6B) by translating the species specific, most frequently used codon for each amino acid in the target sequence to its 5' to 3' complement or its 3' to 5' complement and orienting the complement amino acid sequence either amino-terminal to carboxy-terminal or carboxy-terminal to amino-terminal.
Two additional complements can be designed using the simplified complement approach (e.g., Table 6C) by identifying the species specific, most frequently used amino acid for each second base grouping, generating the complementary amino acid sequence on the basis of the second base grouping of each amino acid in the target sequence, and orienting the complement amino acid sequence either amino-terminal to carboxy-terminal or carboxy-terminal to amino-terminal.

It may be observed, in a particular system, that one orientation of peptide bonds produces more desirable complementary peptides than the other, that is, peptides having the minimum complementary peptide binding activity ~33~0~

or peptides which have a range of activities above the minimum comPlementary peptide binding activity. In this case, investigators may wish to limit their efforts to the more desirable orientation to simplify further experimentation. In addition, if the above design scheme has led to a particularly desirable complementary peptide, investigators may, as is commonly done, seek to find still more desirable complementary peptides by selective exchange of one or more amino acids in the complement amino acid sequence with other amino acids within the second base groupinq.

The design of active or desirable complementary peptides may also be aided by the preparation of a mixture of complementary peptides, such as could be obtained in solid-phase peptide synthesis which in one or more couplin~ reactions several amino acid reagents could be added in one step, followed by a gradient eleution of the mixture from an affinity chromatographic column containing the tarqet peptide attached to the solid support. The most ti~htly bound complementary peptide will elute last.
The structures of the peptides in the various elution fractions may be determined by collectinq fractions and sequencinq with conventional means. In this way, many peptides can be evaluated with a minimum of effort, and peptides having the minimum complementary activity can be readily obtained.

An alternative way to determine desirable complementary peptide sequences would be to generate a set of monoclonal antibodies to the target peptide, and to sequence the hypervariable regions of the gene encoding the immunoglobulin. Regions of complementarity found by sequence searching will reveal, when properly assembled, a 3~ complementary peptide sequence.

In designing complementary polypeptides, the natural amino acids may be replaced in part or in whole by analogs having a similar structure or hydropathy. For example, alanine may be replaced by alpha amino isobutyric acid, arginine by canavanine, aspartate by beta-hydroxy aspar-tate, leucine by norleucine, or gamma-chloroleucine, phenylalanine by beta phenylserine or D-phenylalanine, to name but a few of the many structural analogs known to those skilled in the art. The use of these replacements to construct a complementary polypeptide by a method of the present invention is deemed within the scope of the invention.

Peptides complementary to all or parts of natural hormones may be used to generate antibodies that recognize and bind the hormone receptors. Thse antibodies have properties of anti-idiotypic antibodies to the hormone and, thereby, may be used to purify receptors, to stimulate the biological response associated normally with the hormone, or to antagonize the effect of the natural hormone. The biological response due to antibody of a peptide compleméntary to a hormone is governed in part by the manner in which a particular antibody producing cell is presented with the antigen tcomplementary peptide).
Certain presentations may be preferred, depending on the type of response desired.

Generally, peptides are coupled to carrier substrates, such as large proteins, when being used to generate antibodies. A variety of coupling technologies are known to those skilled in the art. The type of coupling used may affect the presentation of complementary peptide antigens.

Depending on the type of immunization, in vitro or in vivo, various other additives (adjuvants) may be combined with the antigen itself to moderate immune response. Such adjuvants may also affect the presentation of complementary peptide antigens.

Antibodies taken from the serum of animals are polyclonal, meaning that they are a mixture of antibodies produced by a number of sets of cells. When animals are immunized with complementary peptides, a subset of the total antibodies will bind to the complementary peptide and to the receptor of the molecule from which the complementary peptide was derived. This subset, also composed of a number of types of antibodies, can be separated from other antibodies by conventional affinity chromatography. This purified subset of antibodies is usually called mono-specific. Mono-specific antibodies to a complementary peptide may be used to purify receptors, to perform diagnostic assays for receptors, or to moderate biological response.

The mono-specific antibodies can be viewed as the set of antibodies that result from the presentation of antigen in a variety of conformations. Since the peptide antigens are somewhat flexible, they may assume many different conformations or three-dimensional structures. Each of these conformations could result in the generation of slightly different antibodies, all of which are members of the mono-specific set.

Using monoclonal techniques that are well-known in the art, it is possible to produce large amounts of a particular antibody of the mono-specific set. Such monoclonal antibodies could be screened for their ability to hind strongly to complementary peptide or receptor, to produce biological responses similar to the molecule from which the complementary peptide was derived, or to inhibit 13~g~0ii the biological response. Thus, it will be possible to select for the type of response desired.

Complementary peptides may be used in combination with adjuvants to generate vaccines. In this way, the immune system of an organism may be used to moderate the biological response of its hormone systems. The presentation of antigen may require special modifications to ensure the highest population of antibodies that give the desired biological response. Different peptides co~plementary to a particular bioactive peptide may yield different biological response upon vaccination due to differences in their three-dimensional conformations.

The complementary peptides of the present invention may be complementary to small peptides or portions of proteins. These complementary peptides may be utilized much as antibodies are currently often utilized. For example, a polypeptide designed according to the present invention may be prepared as complementary to a particular portion of a unique cell surface proteinaceous antigen characterizing a particular neoplasm. Such a comple-mentary polypeptide may be chemically coupled to many materials of interest such as: a biological or chemical toxin such as ricin A chain or cis-platimum compounds; a radio-opaque substance such as a heavy metal; a radio-isotope; or a fluorescent compound, to name but a few of the many possible labels or substances of interest. The polypeptide-material conjugate would specifically bind to the neoplasm and deliver a toxin or label thereto. Drug delivery systems such as Liposomes, biodegradable polymers or other excapsulating substances of interest may have specific pendant complementary polypeptides for delivery to a particular site.

-49- ~339~06 Complementary polypeptides may be utilized to neu-tralize the activity of particular substances by binding, for example, to such as a peptide hormone, the catalytic site of an enzyme, or a peptidaceous toxin. Hormone receptors may be rendered inactive (by an antagonist) or activated (by an agonist) by administration of polypep-tides complementary to a proteinaceous segment of those receptors.

Polypeptides complementary to the active sites of particular enzymes should prove to be pharmacologically effective. For example, many diabetics may benefit by administration of a polypeptide complementary to the catalytic sites of the insulin-deactivating enzymes glutathione-insulin transhydrogenase and/or the protease termed insulinase.

Many hypertensive individuals may be helped by interfering with the angiotensin system through the use of methods of this invention to design and produce peptides which are complementary to at least a portion of angio-tensinogen, angiotensin I and/or angiotensin II.

In the area of endocrinology, polypeptides comple-mentary to at least a portion of a hormone may be used tolessen or obviate hormone biological activity. For example, in Graves disease (exophthalmic goiter) a hyper-function of the thyroid gland appears to be involved.
Polypeptides complementary to thyrotropin releasing hormone (TRH) or to the beta-subunit of thyroid stimulat-ing hormone (TSH or thyrotropin) would bind to these hormones and facilitate deactivation of the thyroid gland.

Among probable applications of the present invention is the facilitation of blood-group identification. Over 100 different blood-group antigens are present on erythro-~50- 133960~

cyte surfaces to distinguish fourteen well-defined, genetically independent human blood-group systems. These antigenic groups are usually identified by erythrocyte agglutination with antibodies to specific antigens.
Polypeptides complementary to specific blood-group anti-gens may be used instead of antibodies for blood-typing purposes. Polypeptides complementary to an amino acid sequence contained by a particular blood-group antigen may be modified to be at least divalent by crosslinking with agents such as glutaraldehyde or may be coupled to fluo-rescent dyes or radioisotopes. Complementary polypeptides with the former modification would aqglutinate erythro-cytes having the blood-group antigen targeted. The fluorescent or radioisotope modified complementary poly-peptides would bind to and label erythrocytes containingthe blood-group antigen targeted.

Analogously, peptides complementary to the beta chain of chorionic gonadotropin, a pregnancy specific component of bioloqical fluids, could be utilized, by attachment of a label such as a fluorescent dye radioisotope or an enzyme yielding chromophorically measurable products, to facilitate pregnancy tests by means well established in this field.
A key to many important aspects of the immune system resides in knowledge of T cell activation by antigen binding to the T cell receptor. The T cell receptor proteins and, in 1984, the genes coding for both proteins were cloned, isolated and defined (Science V25 p859 and Science V25 plO65). By application of the methods described by the present invention, peptides complementary to different segments of the T-cell receptor may be pre-pared and T cell activation mechanisms systematically investigated. Allergic responses are well know to involve immunoglobulin E (IqE) mediation. Peptides complementary -51- L~;~9606 to segments of IgE or proteins containing peptide sequences complementary to IgE may be helpful in the alleviation of IgE mediated allergy symptoms.

The destruction of collagen, the major structural protein of the human body, during inflamation is important in the pathogenisis of a host of disease states. Acti-vated collagenase, a hydrolytic lysosomal metalloenzyme, proteolytically attacks collagen in the initiation of pathological conditions. Polypeptides complementary to the catalytic site of collagenase should be collaqenase deactivating agents. Due to the fact that mammalian collagenase hydrolyzes native type I colla~en at only one particular point in each polypeptide chain, a polypeptide complementary to that same region of native type I col-lagen may be used to protect that collagen from collage-nase-induced degradation.

Polypeptides co~plementary to toxic peptides or proteins serve, when properly administrered, in vivo or in vitro to bind said materials and lessen or obviate their toxicity.

Methods and compositions employing the complementary peptides of the invention, or antibodies thereto, are also afforded for the treatment of diseases associated with the overproduction or underproduction of a proteinaceous substance and for therapies wherein an increase or decrease in the biological response caused by a proteinaceous substance in beneficial. In particular, these therapeutic compositions comprise effective amounts of the complementary peptides or antibodies thereto in admixture with pharmaceutically acceptable carriers. In particular, pharmaceutical compositions that contain the complementary polypeptides of the invention, or antibodies thereto, as an active ingredient will normally be 133~606 formulated with an appropriate solid or liquid carrier depending upon the particular mode of administration being used. For instance, parenteral formulations are usually injectable fluids that use pharmaceutically and physiologically acceptable fluids such as physiological saline, balanced salt solutions, or the like as a vehicle.
Oral formulations, on the other hand, may be solid, e.g., tablet or capsule, or liquid solutions or suspensions.

In the therapeutic methods of the invention, the complementary polypeptides or antibodies thereto may be administered to humans or any other target organism in various manners such as orally, intravenously, intramuscularly, intraperitoneally, intranasally, intradermally, and subcutaneously. The particular mode of administration and dosage regimen will be selected by the skilled artisan taking into account the particulars of the patient, the nature of treatment required, and/or the disease and the disease state involved. For instance, infection by, or exposure to, foreign proteinaceous toxins is usually treated by daily or twice daily dosages over a few days to a few weeks; whereas a proliferative disease treatment like tumor or cancer treatment involves daily or multidaily doses over months or years. The complementary peptide or antibody therapy of the invention may be combined with other treatments and may be combined with or used in association with other chemotherapeutic or chemopreventive agents for providing therapy against those disease states or conditions against which they are effective.

A listing of such potential benefits to mankind of various applications of the present invention could proceed indefintely and include all manners of diagnos-tics, agent delivery, protein and cell cross-linking capabilities, neutralization of toxins from plants, _53_ 1339606 bacteria or insects, and inhibition of tumor growth by numerous mechanisms including neutralization of peptide growth factors essential to certain tumor growth. The utility of the present invention extends far beyond the particular examples expressed herein.

The significance of relationships between pairing nucleotide triplet codon sequences of nucleic acids was further elucidated by studies concerning the functional activities and interrelationships of specific polypep-tides. The following examples are included herein to demonstrate particular preferred embodiments of the pre-sent invention and are not meant to limit the invention unless otherwise specifically indicated by the claims herein.

EXAMPLE lA

ADRENOCORTICOTROPIC HORMONE (ACTH, FRAGMENT CONTAINING AMINO ACIDS 1-24) COMPLEMENTARY POLYPEPTIDE. (HTCA, 1-24) Synthetic ACTH, fragment 1-24 was obtained from Organon (West Orange, NJ). The primary structure of m-RNA
(messenger RNA) coding for ACTH (1-24) was obtained from Nakanishi et al (Nature (1979) Vol. 278, pp. 423-427) and is shown in Table 8. Above the m-RNA sequence the corre-sponding amino acid sequence for ACTH (1-24) is shown.
When the m-RNA was base-paired in an antiparallel direc-tion, the appropriate complementary nucleotide sequence(c-RNA) shown (turned parallel to the m-RNA) in Table 8 resulted. Below the c-RNA sequence is shown the amino acid sequence of HTCA, the complementary polypeptide to ACTH (1-24) resulting from reading the c-RNA sequence in the 5' to 3' direction.

ACTH, HTCA

ACTH: H~N-Ser Tyr Ser Met Glu His Phe Arg Trp Gly Lys mRNA: 5'-UCU UAC UCC AUG GAA CAC UUC CGC UGG GGC AAG
cRNA: 5'-GGG GUA CAC CUU CAC CGG GCG CCG CUU CUU GCC
HTCA:a H2N-Gly Val His Leu His Arg Ala Pro Leu Leu Ala Pro Val Gly Lys Lys Arg Arg Pro Val Lys Val Tyr Pro-COOH
CCG GUG GGC AAG AAG CGG CGC CCG GUG AAG GUG UAC CCC-3' CAC CGG CUU GCC CCA GCG GAA GUG UUC CAU GGA GUA AGA-3' His Arg Leu Ala Pro Ala Glu Val Phe His Gly Val Arg-COOH

A polypeptide having the amino acid sequence of HTCA shown above was synthesized for the inventors by Peninsula Laboratories (San Carlos CA).

~55~ 1339~06 EXAMPLE lB

BINDING OF ACTH (1-24) TO ITS
COMPLEMENTARY POLYPEPTIDE HTCA (1-24) The methods generally described by Johnson et al (J.
Immunol (1982) Vol. 129, pp. 2357-1359) were utilized to demonstrate the binding affinity of the complementary peptides ACTH and HTCA. From 1 to 25 micrograms (ug) per well of HTCA or insulin in carbonate-bicarbonate coatinq buffer were added to 96 well round bottom microtiter plates and incubated at 4~C for 8 hr. The plates were then washed with phosphate buffered saline (PBS)-Tween*20 (Sig~a Chemical Co., St. Louis, MO) buffer.
To the insulin-coated wells and to some of the HTCA-coated wells was added 10 ug synthetic ACTH (1-24) in PBS-Tween buffer. Control wells (HTCA alone) contained only PBS-Tween. The plates were incubated at room temper-ature for 2 hr. and then washed three times with PBS-Tween buffer. Rabbit antisera directed against the amide of synthetic ACTH 1-13 (Accurate Biochemicals, Westbury N.Y.) was added to each well and the plates were incubated for 1 hr at room temperature. Following 3 washes with PBS-Tween buffer, alkaline phosphatase-conjugated goat anti-rabbit Ig G (Miles Laboratories, Elkharbt, IN) in PBS-Tween buffer (1:300 dilution) was added. After inculation at room temperature for 1 hr, the plates were washed three times with PBS-Tween buffer and each well was treated with 200 microliters (ul) of p-nitrophenyl phosphate (lmg/ml in carbonate-bicarbonate buffer) for 1 1/2 hr. at room te~perature. The enzymatic reaction was then stopped by the addition of 3N NaOH (50 ul) to each well and the optical absorbance of p-nitrophenol the alkaline phos-phatase product, was measured at 405 nm. The ACTH bound *Trade mark ;i -56- 13 3g ~0 6 to the coated microtiter wells was measured by this enzy~e-linked immunoabsorbent assay (ELlSA).

The results of this experiment are shown in Figure 3.
Synthetic ACTH ( 1-24) is bound by HTCA coated microtiter wells but not by insulin coated microtiter wells. The antibody specific for ACTH (1-13 amide) does not bind to the coating of HTCA, and nor does ACTH bind to the coating of insulin. The molar amount of ACTH bound was directly proportional to the concentration of HTCA coating which suggests a one to one binding of the two peptides (data analysis not shown).

A microtiter plate was prepared and treated generally as described above but was coated with 3.7 nmol/well HTCA.
To each coated well was added a solution of 3.7 nmol ACTH
combined with the amounts of HTCA designated on the abscissa in Figure 4. When ACTH binding was evaluated by an ELISA, it showed that about 90% of ACTH-HTCA binding was specific. A Scatchard analysis (not shown) of the data from Figure 4 showed a single uniform binding site with a Kd of 1.9 micromolar, comparable to the affinity shown by an antibody-antigen complex.

EXAMPLE lC

Binding of I125 ACTH TO

3'-5' HTCA

Utilizing the complementary RNA (cRNA)sequence shown in Table 8 of Example lA, but reading the cRNA in the 3' to 5' direction, the following amino acid sequence was obtained and the respective peptide (3'-5' HTCA) chemi-cally synthesized:

133~606 H2N- Arg-Met-Arg-Tyr-Leu-Val-Lys-Ala-Thr-Pro-Phe-Gly-His-Pro-Phe-Phe-Ala-Ala-Gly-His-Phe-His-Met-Gly-COOH.

Utilizing techniques described in Example lB, poly-vinyl microtiter wells were coated with 3'-5' HTCA from a lmM solution thereof or with BSA.

The BSA and 3'-5' HTCA coated wells were washed and then treated with one of three solutions of I125 ACTH (1-39) (New England Nuclear Boston, MA) having differentconcentrations. After a forty five minute incubation period the solution was removed and the microtiter wells extensively washed. The microtiter wells were separated and assayed for iodinel25 content in a Beckman Gamma 5500*
gamma counter. The results of these manipulations are shown in Table 9. Bound I-ACTH was measured in duplicate at three concentrations in the absence and presence of excess unlabelled ACTH.

ACTH and 3'-5' HTCA
Bound CPM - 125I_AcTH
BSA 3'-5' HTCA +Excess Coating Coating ACTH
conc 125 I_AcTH 159 1819 616 1:3 dilution 125 I-ACTH 155 601 287 1:9 dilution 125 I-ACTH 133 263 ~ 191 *Trade Mark ~i -58- 13 39~0 ~

As shown by the data in Table 9, 125 I-ACTH binds to coated 3'-5' HTCA but not to coated BSA, a well-known protein with a wide binding capacity. The binding of 125 I-ACTH to 3'-5' HTCA-coated microtiter wells is inhibited by the presence of excess free ACTH. This result demon-strates both the affinity of a complementary peptide to an original peptide and the fact that such complementary polypeptides may be designed and obtained by reading the sequence of complementary nucleic acid codons in the 3'to 5' direction and chemically synthesizing the peptide so directed.

EXAMPLE lD

CO~IPONENT PEPTIDE SEQUENCES OF HTCA

Utilizing the cRNA sequence and HTCA sequence shown in Table 8 of Example lA, a series of peptides having a carboxy-terminal portion of the HTCA amino acid were synthesized. A pentamer (5-mer) contained the amino acid sequence: H2N-Phe-His-Gly-Val-Arg-COOH. A decamer ~10-mer) contained the amino acid sequence: H2N-Ala-Pro-Ala-Glu-Val-Phe-His-Gly-Val-Arg-COOH. A twenty membered peptide (20-mer) contained the amino acids sequence :
H2N-His-Arg-Ala-Pro-Leu-Leu-Ala-His-Arg-Leu-Ala-Pro-Ala-Glu-Val-Phe-His-Gly-Val-Arg-COOH. Each of thes peptides was used to coat microtiter wells and the coated wells tested for the ability to bind 1 5 I-ACTH as described in Example lC. The results of these manipulations are shown in Table 10.

13~960~

I-ACTH Binding to HTCA Components CPM bound 125I-ACTH
BSA 5-mer +excess 10-mer +excess 20-mer +excess Coating Coating ACTH Coating ACTH Coating ACTH
onc 125I ACTH 159 10,252 233 8,927 345 1978 229 160 11,388 245 9,072 350 1768 213 1:3 dilution I-ACTH 155 3 65515 97 3391 132 66253 l8l33 1:9 dilution 5I-ACTH 133 995 52 1062 5682 221385 4389 As shown by the data in Table 10, the peptides from the HTCA sequence all exhibit the ability of binding 125 I-ACTH.

EXAMPLE lE

HTCA with Reversed Directionality and Binding of 12 I-ACTH (1-39) thereto A 24-mer HTCA variant (R-HTCA) with reversed amino acid sequence directionality (amino-terminal and carboxy-terminal ends being reversed) was synthesized and had the following sequence: H2N-Arg-Val-Gly-His-Phe-Val-Glu-Ala-Pro-Ala-Leu-Arg-His-Ala-Leu-Leu-Pro-Ala-Arg-His-Leu-His-Val-Gly-COOH. The well-coating and 125I-ACTH binding procedures were performed as described in Example 1-C and the resultant data shown in Table 11.

I-ACTH Bind~ to R-HTCA
CPM Bound I-ACTH

BSA R-HTCA +Excess ACTH
conc 125I_ACTH 159 10,392 303 160 10,709 325 1 3 dil 125I ACTH 155 2,997 140 165 3,209 121 1:9 dil 125I ACTH 133 1,101 65 114 1,161 69 -61- 1~39~06 As demonstrated in Table 11, the R-HTCA polypeptide, complementary to ACTH, has a si~nificant affinity for ACTH. This exhibits yet another aspect of the present invention, the reversed directionality specifically tested here permits a further variance in the desiqn of polypep-tides where an optimal set of chemical, physical and biological effects may be sought for a particular circum-stance or application. If HTCA is understood as having an amino acid sequence antiparallel to the ACTH sequence, then the R-HTCA sequence is parallel to the ACTH amino acid sequence. Thus both parallel and antiparallel peptides or polypeptides complementary to an original or target amino acid sequence effectively have affinities therefor. The complementarity or affinity of a comple-mentary polypeptide to an original peptide or protein isretained, regardless of the amino-terminal and carboxy-terminal directionality of said complementary polypeptide.

EXAMPLE lF
PREPARATION AND PROPERTIES OF ANTIBODY TO HTCA.

An antigenic form of HTCA was prepared by coupling 200 ug HTCA to 200 ug keyhole limpet hemocyanin (KLH) with 6.7 mM glutaraldehyde according to the methods of Avrameas et al (Immunochemistry (1969) Vol. 6, pp. 53-66).
Excess glutaraldehyde was removed from the HTCA-KLH
conjugate by passage through a Bio-Rad*P-10 column (Bio-Rad, Richmond, CA).
Three injections, containing 25 ug, HTCA-KLH in 0.5 ml complete Freunds adjuvant were administered to a rabbit at two-week intervals. Total immunoglobulin from the resulting rabbit antiserum was isolated by immunoaffinity 3S chromatography on a column of Sepharose*4B (Pharmacia Fine Chemicals, Uppsala, Sweden,) coupled to goat anti-rabbit * Trade Mark 133~60~

immunoglobulin. To purify the anti-HTCA antibody, KLH
antibody was removed from the total immunoglobulin by passage through a column of Sepharose 4B coupled to KLH.
A 1:300 dilution of the purified antibody preparation (anti-HTCA) would detect at least 100 ng of the HTCA in an indirect ELISA.

An induction of glucocorticoid hormone production by ACTH and anti-HTCA was found with cultured mammalian cells. Duplicate cultures of mouse adrenal tumor (Y-l) cells in microtiter plates were treated with culture media, ACTH ~10 microunits/well) or various dilutions of the anti-HTCA. The results of this experiment are shown in Table 12.

13~960~

Additiona Corticosterone equivalents ~ug/ml)b Experiment 1 Experiment 2 ACTH 1.08 1.42 anti-HTCA 1:3 1.19 1.03 1:19 N.D. 0.78 1:30 N.D. 0.62 Media 0.66 0.68 aDuplicate cultures of mouse adrenal (Y-l) cells in microtiter plates were treated with culture media, ACTH (10 microunits/well) or the indicated dilutions of antibody to HTCA.
After 18 hrs incubation at 37~ in 4% CO2, replicate cultures were pooled and assayed for glucocorticoid hormone production by a radio-immunoassay (RIA) for corticosterone (Smith et al, Science (1982~, Vol. 218, pp. 1311-1312) bParallel dose responses for experimental samples and the corticosterone standard were obtained over a ten fold range. Interassay variation was 8.8 percent.
CNot done.

The activation of the mouse adrenal tumor cell ACTH
receptor by anti-HTCA indicates a configurational analogy of the antibody and ACTH. Neither normal rabbit serum nor 133~606 antibody to KLH caused a steroidogenic response (data not shown). The activation of receptors for insulin (Sege et al, Proc. Nat'l. Aca. Sci. (1978) and for beta-adrenergic agents (Schreiber et al, Proc. Nat'l. Acad. Sci. (1980), Vol. 77, pp. 7385-7389) by anti-idiotypic antibodies raised against antibodies for insulin or beta adrenergic agents has been described. Thus, a relationship of analogy exists between to complementary peptide ligands and anti-idiotypic antibodies.
EXAMPLE lG

BINDING OF ANTI-HTCA TO MOUSE
ADRENAL TUMOR (Y-l) CELLS
Mouse adrenal tumor (Y-1) cells were affixed by glutaraldehyde in flat bottom wells of a microtiter plate.
The affixed cells were then treated with rabbit anti-KLH
or rabbit anti-HTCA alone or in the presence of several levels of ACTH. After washing, the microtiter wells were treated with goat antibody to rabbit immunoglobulin the goat antibody being coupled to the enzyme alkaline phos-phatase. After washing away unbound antibody-phosphatase complex, p-nitrophenyl phosphate was added and the enzyme dependent development of absorbancy at 405 nM was moni-tored. As shown in Figure 5, ACTH blocked binding of the anti-HTCA in a dose dependent manner. ACTH and anti-HTCA
appeared competitive for the same binding site on the mouse adrenal tumor (Y-l) cells. Rabbit anti KLH had no effect (shaded region) 13~960~
EXAMPLE lH

PURIFICATION OF ACTH RECEPTOR.

Purified anti-HTCA was covalently coupled to cyanogen bromide-activated Sepharose 4B. Approximately 108 mouse adrenal tumor (Y-l) cells were sonicated for 5 min. at 40 KH~ (Branson E Module 8ath Sonicator) in the presence of 2mM phenylmethylsulfonyl fluoride. After removal of cell debris by centrifugation, the supernatant fluid was passed through a chromatographic column containing Sepharose 48 coupled to anti HTCA. After extensive washing, the residual binding material was eluted from the column with 0.1M glycine, pH 2Ø The eluted material was neutralized and concentrated by dialysis against dry polyethylene glycol. The concentrated material was then subjected to gel chromatography on a calibrated column of Sephacryl*S-200 (Pharmacia, Fine Chemicals, Uppsala, Sweden). Ali-quots of the gel chromatography fractions were assayed for ACTH receptor activity by a radio receptor procedure.
Briefly, this procedure involved incubation of the above aliquots in 96-well polyvinyl plates for 18 hr, followed by removal of unbound materials by washing. Radio-iodinated ACTH ( I-ACTH, 70 microcuries/ug, New England Nuclear, ~oston, MA) was then added to the wells in the presence or absence of an unlabeled ACTH excess (10 ug/well). The plates were then extensively washed and the wells were excised from the plate and measured for 125I-ACTH with a Beckman Gamma 5500 gamma counter. The chro-matographic fractions were also monitored for absorbanceat 280 nM. The results of this Example are shown in Figure 6.

Specifically bound 1 5I-ACTH was found by subtracting the radioactivity bound in the presence of excess un-labeled ACTH (~enerally less than 10~) from radioactivity ~~; *Trade Mark c ' :

1:~39~06 bound in the absence of unlabeled ACTH. The elution points for the 158 kilodalton (K), 67K, 45K and 14.4K
molecular weight standards are indicated by the arrows.

The ACTH receptor activity had a molecular weight of about 80 to 100K, which was similar to that previously reported for an ACTH receptor identified by a photo-affinity labeling technique (Ramachandran et al, Proc.
Nat'l. Acad. Sci. (1980), Vol. 77, pp. 3697-3970).
The procedure described in this example demonstrates a general method of the invention for obtaining components of any peptide or protein ligand receptor site. A poly-peptide complementary to at least a portion of the peptide or protein is first provided. An antibody against said complementary polypeptide is then prepared. The antibody is then coupled by chemical or adsorptive means to a solid matrix. A receptor-containing sample is then treated with the antibody-coupled matrix to specifically bind compo-nents of the receptor site. Finally the bound components are eluted.

EXAMPLE lI

GAMMA ENDORPHIN, THE DESIGN AND OBTAINING
OF ITS COMPLEMENTARY POLYPEPTIDE.

To further illustrate the general applicability and significance of the present invention, a second pair of interactive complementary peptides was studied. Positions 104 to 120 of the amino acid sequence of bovine gamma endorphin precursor and mRNA sequence coding therefor were shown by Nakanishi et al (Nature (1979), Vol. 278, pp.
423-427). Table 13 shows this gamma endorphin amino acid sequence (designated endo)and corresponding m-RNA se-quence. The complementary strand of RNA (c-RNA) base-pairing in an antiparallel direction with the m-RNA for gamma-endo is shown beneath the m-RNA, the cRNA is shown in Table 5 parallel to the m-RNA. The polypeptide (gamma-odne) whose sequence is directed by reading the c-RNA in the 5' to 3' direction is shown beneath the c-RNA
~designated gamma-odne).

-endo: H2N-tyr-gly-gly-phe-met-thr-ser-glu m-RNA: 5'-UAC-GGC-GGG-UUC-AUG-ACC-UCC-GAG
cRNA: 5'-CAG-CGU-GAC-AAG-GGG-CGU-UUG-GCU
-odne: H2N-gln-arg-asp-lys-gly-arg-leu-ala lys-ser-gln-thr-pro-leu-val-thr-leu-COOH
AAG-AGC-CAA-ACG-CCC-CUU-GUC-ACG-CUG3' CUU-CUC-GGA-GGU-CAU-GAA-CCC-GCC-GUA3' leu-leu-gly-gly-his-glu-pro-ala-val-COOH

A polypeptide having the amino acid sequence of gamma-odne was synthesized for the inventors by Peninsula Labora-tories (San Carlos, CA).

EXAMPLE lJ

PROPERTIES OF GAMMA (~)-ODNE, THE POLYPEPTIDE
COMPLEMENTARY TO GAMMA-ENDORPHIN (~-ENDO).

Synthetic bovine gamma-endorphin was obtained from Boehringer Mannheim (Indianapolis, IN) and rabbit antibody for synthetic gamma-endo was obtained from Accurate Biochemicals (Westbury, NY).

The wells of a 96-well round-bottomed microtiter plate were coated (by the procedure described in Example lB) with gamma-odne (40 ug/well), insulin (20U/well) or -68- 13~9~06 bovine serum albumin (BSA,200 ug/wèll). Varying concen-trations of gamma-endorphin (as shown on the abscissa in Figure 7) were incubated in the coated wells for 1 hr, after which the plates were thrice washed with PBS-Tween buffer. The wells were then treated with rabbit antibody for gamma-endorphin, washed, and then treated with goat antibody against rabbit immunoglobulin the goat antibody being conjugated to alkaline phosphatase. After washing away unbound goat antibody conjugate, the bound alkaline phosphatase activity was measured with p-nitrophenylphos-phate as earlier described. The results of the above manipulations are shown in Figure 7 where the extent of absorbance at 405 nM reflects the degree of gamma-endorphin binding to wells coated with gamma-odne, insulin or BSA. It was clear that gamma-endorphin significantly bound to the affixed gamma-odne as compared to its binding to affixed insulin or BSA. The shaded area in Figure 7 represents the extent of gamma-endorphin binding in the presence of soluble excess of gamma-odne. Thus it is shown that a second pair of complementary peptides inter-acts in a manner showing affinity and apparent congruence.

EXAMPLE lK

GENERAL APPLICABILITY FOR PRODUCTION
OF COMPLEMENTARY POLYPEPTIDES

The results of Examples lA through lJ demonstrate particular applications of a general method for designing and obtaining polypeptides complementary for proteins or peptides having on at least partially known nucleotide coding sequence. For example, the complementary nucleo-tide sequence which codes for a polypeptide complementary to at least a portion of a first protein or peptide when read in a 5' to 3' direction may be DNA. Said DNA may be inserted into a plasmid to form a recombinant DNA transfer - 1339~06 vector. A unicellular organism, suitably a bacteria yeast or mammalian cell may then be transformed with the recom-binant DNA vector to produce a transformant unicellular organism biosynthesizing said complementary polypeptide.
The techniques for such insertions and transformations arewell known in the relevant fields.

Techniques of chemical polypeptide synthesis from amino acids, as well as methods of obtaining polypeptides for example by a proteolytic excision from proteins having of a larger amino acid sequence but containing the comple-mentary amino acid sequence desired are also known. Thus, many ways of obtaining polypeptides complementary to peptide or protein ligand are available.
EXAMPLE lL

Blocking of stress response in mice by a peptide complementary to Adrenocorticotropic Hormone (ACTH).
In response to various stresses, animals produce adrenocorticotropic hormone (ACTH), which acts on adrenal cells to produce elevated serum levels of corticosterone.
To test the effectiveness of the inhibition of activity of ACTH in vivo by a complementary peptide, HTCA (see Example _), the followin~ sets of experiments were performed.
Mice (BALC/c) were injected with the complementary peptide HTCA, held for a period of time, stressed, decapitated, and serum corticosterone levels were determined by conventional immunoassay.

In the first experiment, 1 mg HTCA dissolved in PBS
(phosphate buffered saline) was injected into 8 sets of 3 BAL~/c mice each. Two sets of 3 BALB/c mice were injected only with PBS. The mean corticosterone levels of each set of mice was determined after various delay and stress 1339~0~

regimens. The results are presented in Table 14, where the stress is a 30 second immersion in ice water.

TABLE 14:

* Corticosterone Injection Route Delay (hr) Stress (ug/dl) PBS IM 0 none 18 HTCA IM 0.5 YES 90 HTCA IM 1.0 YES 55 HTCA IM 2.0 YES 40 HTCA IM 4.0 YES 60 HTCA IP 0.5 YES 95 HTCA IP 1.0 YES 85 HTCA IP 2.0 YES 75 HTCA IP 4.0 YES 65 *IM - intramuscular injuection IP - intraperitoneal injection The maximum effect observed, about a 50~ reduction in serum corticosterone, occurred with stress occuring 2 hours after intramuscular injection of 1 mg of HTCA. In a second experiment, various amounts of HTCA were injected (IM) into sets of 3 BALB/c mice. Mice were held for 2 hours and stressed with immersion in ice water. Table 15 shows the mean serum corticosterone levels for the various sets of mice.

35 Injection Stress Corticosterone (ug/dl) 0.01 mg HTCA YES 44 0.1 mg HTCA YES 37 1.0 mg HTCA YES 30 -71- 13 3g ~06 Combined, these experiments show the ability of a peptide ccnplementary to ACTH to lower the corticosterone stress response in mice.

EXAMPLE lM

Blocking of Binding of 125I-beta-endorphin to NG108-15 cells by a peptide complementary to gamma-endorphin.

NG108-15 cells are known to contain receptors for beta-endorphin called opiate receptors. In addition to beta-endorphin, gamma-endorphin and several analogs of beta-endorphin bind to these opiate receptors. The peptide, gamma-ODNE, complementary to gamma-endorphin described in Example II was examined for its ability to inhibit the binding of 125I-beta-endorphin to opiate receptors on NG108-15 cells. Gamma-endorphin is a fragment of the larger peptide beta-endorphin.

In separate experiments, the specific binding of 1 I-beta-endorphin to NG108-15 cells plated in microtiter wells was determined by competition with unlabelled beta-endorphin. Seventy percent of the total binding was specific by conventional criteria.
For this experiment, 125I-beta-endorphin was pre-incubated 30 minutes with various amounts of the complementary peptide, gamma-ODNE, before a 30 minute (37~C) incubation with NG108-15 cells in microtiter wells.
After the incubation, cells were washed 3 times and cell associated radioactivity wad determined with a Packard Multi-Prias gamma counter. Table 16 demonstrates the ability of gamma-ODNE to inhibit the binding of 125I-beta-endorphin (10 1 M,2000 Cl/mmol) to NG108-15 cells.

*Trade Mark ~3 --72- 13~5~6 ~ Inhibition of Specific Gamma-ODNE Concentration 125I-beta-endorphin M 24.7 + 3.9 _4 36.0 + 2.1 M 75.0 + 0.6 EXAMPLE lN

Induction of catatonia in mice by antibodies to a peptide complementary to gamma-endorphin.

It is well known that larqe doses of opiate-like suhstances can induce a catatonic state in animals which is inhibited or reversed by treatment with the opiate antagonist naloxone. In analogy with the results in earlier examples, the ability of a peptide (gamma-ODNE) complementary to an opiate peptide, gamma-endorphin, to induce antibodies that have opiate properties was examined.

Gamma-ODNE (Example II) was conju~ated to Keyhole Limpet Hemocyanin (KLN) and used with Freund's adjuvant to generate rabbit antibodies in a conventional manner (see Example lF). Both normal and induced rabbit sera were injected intracerebroventicularly (i.c.) into female ICR
mice. Injection of 50 ul of normal rabbit serum (diluted 1:100) resulted in no induction of opiate-like catatonia.
Injection of 50 ul of complementary peptide (gamma-ODNE) induced rabbit serum (diluted 1:100) resulted in an excellent induction of catatonia, which was reversed by the simultaneous i.c. injection 0.2 mg of the opiate antaqonist naloxone.

~73~ 13 39 ~0 ~

These results demonstrate the ability of an antibody to a complementary peptide to an opiate to itself show opiate activity.

Antigenic Relationship Between Two Peptides Complementary to Adrenocorticotropic Hormone (ACTH) Two peptides complementary to ACTH, described in Examples lB and lC as HTCA and 3'-5' HTCA, were each coupled to Keyhole Limpet Hemocyanin (KLH) with glutaraldeyde (1 mg peptide, 1 mq KLH, 30 mM
qlutaraldehyde for 30 minutes at room temperature followed by dialysis) to prepare antigens for rabbit immunization.
Two rabbits were injected separately with 250 ug of peptide-KLH antigens weekly for four weeks. Each rabbit was bled biweekly for three weeks followinq the last injection. Antibodies representing the total immunoglobulin from serum of each rabbit was purified by precipitation from serum with 50% saturated ammonium sulfate at 4~C followed by standard ion exchange chromatography on a DEAE column.

To examine the binding of each of the immunoglobulins to each of the peptides, enzyme-linked immunosorbant assays were performed. In these assays, one of the complementary peptides was coated onto polycarbonate plates in a carhonate coatinq buffer at pH 9.0 overnight.
Unhound peptide was removed by washing three times with phosphate buffered saline (PBS) - TWEEN 20. Various amounts of immunoqlobulins were added to peptide-coated wells in PBS-TWEEN 20 solution and incubated for one hour.
Unhound immunoglobulin was removed by washinq with PBS-TWEEN 20. To each well was added a 1:300 dilution of goatanti-rabbit IqG coupled to alkaline phosphatase enzyme.
*T~e Mark .
~j ..

1~3960fi After one hour the wells were washed with PBS-TWEEN 20 to remove unbound secondary antibodies. The amount of remaininq secondary antibody was determined by reacting p-nitrophenyl phosphate with the remaining aklaline phosphatase enzyme for 15 minutes, stopping the reaction with 3M NaOH, and measuring nitrophenol produced spectrophotometrically at 490 nm.

Table 17 qives the results of the assay. Not shown are three controls (ACTH plates + immunized rabbit immunoglobulin, HTCA plates + normal rabbit immunoglobulin, and 3'-5' HTCA plates + normal rabhit immunoglobulin) all of which showed less than 0.01 absorbance in the assay.

Antibody Added Immunogen Absorbance at 490nm mq/ml Plates coated with HTCA
0.012 3'-5' HTCA 0.08 + 0.04 0.04 3'-5' HTCA 0.24 + 0.08 0.11 3'-5' HTCA 0.37 + 0.09 0.3 3'-5' HTCA 0.91 + 0.11 0.11 HTCA 0.98 + 0.18 Plates coated with 3'-5' HTCA
0.012 HTCA 0.05 + 0.01 0.04 HTCA 0.12 + 0.06 0.11 HTCA 0.44 + 0.06 0.3 HTCA 1.39 + 0.21 0.11 3'-5' HTCA 0.89 + 0.23 These results demonstrate that antibodies to one complementary peptide recognize and bind to another complementary peptide. Thus, the two complementary peptides are antigenically related even though their 13~9606 sequences have only one position where the same amino acid occurred in the same absolute position.

EXAMPLE lP
Idiotype:Anti-idiotype Relationship between Antibodies to Adrenocorticotropic Hormone (ACTH) and Antibodies to a Peptide Complementary to ACTH

Using a standard Radioimmunoassay (RIA) for ACTH
(Immuno Nuclear Corporation, CAT. #2400), the ability of antibodies to a peptide (HTCA) complementary to ACTH to bind with antibodies to ACTH was determined. In a Standard RIA for ACTH, unlabelled ACTH is added in known amounts to a solution of radiolabelled ACTH and antibody to ACTH to compete away the binding of radiolabelled ACTH
to its antibody. The amount of radiolabel bound is determined by immunoprecipitating the antibody and counting the radioactivity of the precipitiate.
In the experiment, unlabelled ACTH, immunoglobulin from rabbits immunized with the complementary peptide HTCA, and immunoglobulins from normal rabbits were used to compete with radiolabelled ACTH in the RIA.
Immunoglobulin (35 mg/ml) solutions were prepared in phosphate buffered saline, and ACTH and 5I-ACTH were prepared as specified in the RIA kit. Table 18 gives the results of the competion assay.

-76- 13~60~

Competitor Added Percent Specific Binding none 100 20 pq/ml ACTH go 50 pg/ml ACTH 77.5 100 pg/ml ACTH 62.2 200 pg/ml ACTH 42 500 pg/ml ACTH 21.6 IgG HTCA (1:10 dilution) 0 IqG HTCA (1:30 dilution) 10 IgG HTCA (1:100 dilution) 26.3 IqG HTCA (1:300 dilution) 73.6 IgG HTCA (1:1000 dilution) 100 Normal (1:100 dilution) 89.3 Normal (1:1000 dilution) 94.4 These results demonstrate that antibodies to a peptide complementary to ACTH recognize and bind to antibodies to ACTH and thus define an idiotype:anti-idiotype relationship between the antibodies.

EXAMPLE lQ
In Vivo Activity of Antibodies to a Peptide Complementary to Adrenocorticotropic Hormone (ACTH) Generated by Animal Vaccination A peptide comPlementary to ACTH (see HTCA example lA) was conjugated to Keyhole Limpet Hemocyanin (KLH) using the procedure described in Example 10. Three BALB/c mice per time point were immunized (100 ug conjuqate/0.2 ml complete Freund's adjuvant on day 0 injected intraperitoneal and subcutaneous, 100 ug conjugate/0.2 ml incomplete Freund's adjuvant at weeks 1, 2, 3, and 4 injected intraperitoneal and subcutaneous). Four rabbits were immunized (300 ug conjugated/l.0 ml complete Freund's adjuvant injected on day 0, 200 uq conjuqate/1.0 ml -77_ 133~06 incomplete Freund's adjuvant injected on days 10, 20, 30, 40 and 60).

At each time point in the mouse experiment, three 5 mice were sacrificed, their blood collected and allowed to clot, and their antihody titers were determined by the following solid phase radio immunoassay (RIA). HTCA was coated in wells of a polyvinyl microtiter plate from solution (0.25 mq/ml). Wells were washed with phosphate buffered saline (PBS) containing TWEEN 20 and serum in 1: 5 serial dilutions was added. After one hour, 125I rabbit anti-mouse immunoglobulin was added and wells were washed with PBS-TWEEN 20 to remove unbound radiolabel. Wells were punched and counted to determine titers. For the 15 molJse study, titer is defined as the serum dilution that produces 100 counts per minute above backqround. Also at each time point, serum was analyzed by commercial RIA for corticosterone levels.

At each time point in the rabbit experiment, 2 ml of blood was drawn, allowed to clot, and frozen until analyses. Antibody titers were determined by an enzyme-linked immunosorhant assay (ELISA) as follows. HTCA was coated from a 0.25 mg/ml solution onto wells of a 25 polycarbonate plate overnight. Wells were then washed and serum added in 1: 5 serial dilutions. After one hour goat anti-rabbit immunoqlobulin conjugated with alkaline phosphatase (1:300 dilution) was added. In one hour, wells were washed with PBS-TWEEN 20. p-nitrophenol phosphate substrate was added and allowed to react for 15 minutes before quenchinq with 3M NaOH. Titers were determined spectrophotometrically by absorbance at 490nm.
For the rabbit study, titer is defined as the serum dilution that produces an absorbance of 0. 05 (background e~uals 0.01). Also at each time point, serum was analyzed by commercial RIA for corticosterone levels.

13~9~0~

Tables 19 and 20 present and results of these experiments in terms of antibody to HTCA titers and serum corticosterone levels as a percent of controls (animals immunized with KLH alone and bled on the same schedule as those receiving HTCA).

Mouse Experiment Day HTCA TiterCorticosterone (~ of Control) 0 3.6 100 Rabbit Experiment Day HTCA TiterCorticosterone (~ of Control) 0 1.4 86 3.8 66 In the mouse experiment, corticosterone levels of controls were near normal except for the last time point which was elevated to about three times normal.
Corticosterone levels in control rabbits were below normal ~79~ 1~3~ 0~

and falling between days 0 and 30 and recovered to normal levels by day 60.

In both experiments, a burst of hormone-like (ACTH) response was generated and was followed with a general depression of response. These experiments demonstrate the ability of complementary peptide antigen to moderate hormone response in vivo by stimulating antibody production with receptor binding activity. No effort was made to alter antigen presentation for the production of either agonistic or antagonistic antibodies such as might be desirable in other situations.

EXAMPLE lR
Potential Diagnostic Assay for Adrenocorticotropic Hormone (ACTH) Based on Complementary Peptide Binding The potential for use of peptide complementary to ACTH, specifically HTCA of Example lA, in determining ACTH
levels in serum was investigated using a solid phasé
radiobinding assay.

Complementary peptide (HTCA) was dissolved in phosphate buffered saline (PBS) at 1 mg/ml and 200 ul aliquots were placed in wells of a 96 well polyvinyl assay plate (Microtes~ III, Falcon Cat. #3911). HTCA was allowed to coat the wells overnight at 4~C. The coating solution was removed and the wells were washed three times with PBS. Radiolabeled ACTH ~125I-ACTH, New England Nuclear NEX-65) was diluted into P8S containing 0.0005%
TWEEN-20 so as to deliver 50 pg (or about 30,000 cpm) in a 20 ul volume.

Serum samples were prepared in a 96 well tissue culture plate by serial dilution of serum into PBS/TWEEN
*Trade Mark -80- i3~06 20 to give a final volume of 180 ul/well. The serum samples were then transferred to the HTCA coated a ssay plate and allowed to incubate. Finally, 20 ul of 125I-ACTH solution was added, incubated, and the total sample of 200 ul was removed. After washing three times with PBS, the wells were cut from the plate and counted in a gamma counter. Nonspecific binding was determined by adding 5 ug of unlabeled ACTH to some of the samples before transferring them to the assay plate.
Serum samples used in this study contained either 50 pq/ml or 500 pg/ml of ACTH added to fetal calf serum (FCS).

The data in Table 21 shows the percent inhibition of specific 12 I-ACTH binding as a function of serum dilution for both samples.

50 pq/ml 500 pg/ml % Inhibition of % Inhibition of Log10 Dilution Specific Binding Specific 8inding -0.5 73 90 -1.0 40 77 -1.5 44 57 -2.0 9 53 30 -2.5 1 35 -3.0 0 32 _3.5 0 20 Total specific binding in this assay was determined to be 1000 counts per minute and, based on specific activity, 50% specific binding is equivalent to 0.85 pg of ACTH in a serum dilution.

For the 50 pg/ml serum sample, as determined by commercial radio-immuno assay techniques, 50% inhibition -81- ~3 3~ 60 6 occurred at a Logl0 Dilution of -1.1 (determined by qraphic interpolation). Thus, this assay measures the serum sample as 54 pg/ml, in good agreement with standard methods.

~ y graphic interpolation, 50% inhibition for the 500 pg/ml serum sample occurred at a Logl0 Diulution of -2.0 as expected.

These results demonstrate the feasibility of developing diagnostic assays based on complementary peptide binding.

EXAMPLE lS
Comparison of Binding of I-Adrenocorticotropic Hormone to 5'-3' and 3'-5' Complementary Peptides A solid-phase binding assay was used to determine the ability of both the 5'-to-3' and the 3'-to-5'-complementary-RNA-strand peptides (see Examples lA and lC) to bind 12 I-ACTH. The peptides were diluted in phosphate-buffered saline (140 mM-NaCl/3mM-KCl/0.1%
NaN3/20 mM-phosphate buffer, pH 7.4) to 2 mg/ml, and 0.2 ml/well was placed into poly(vinyl chloride) micro-titre plates (Becton and Dickinson, Oxnard, CA, U.S.A.). After 18 h at 4~C, unbound peptide was removed by washing three times with phosphate-buffered saline containing 0.1%
bovine serum albumin (Sigma Chemical Co., St. Louis, MO, U.S.A.). 125I_ACTH was diluted to various concentrations with phosphate-buffered saline containing 0.1~ bovine serum albumin and incubated in peptide-coated wells for 60 minutes at 4~C. To some wells, various concentrations of soluble peptides were added in addition to the 125I_ACTH
in order to demonstrate the specificity of binding. After incubation, unbound radiolabel was washed out with 1339~0~

phosphate-buffered saline containinq 0.1~ bovine serum albumin, and the radioactivity of individual wells was measured by gamma-radiation counting. The amount of specifically bound radioactivity was determined by blocking I-ACTH binding with unlabelled ACTH.
Additional controls included coating wells with insulin or bovine serum albumin ~2 mg/ml) before the addition of the 125I-ACTH.

In these experiments, half-maximal binding of I-ACTH occurred at 0.3 mM ACTH for both co~plementary peptides. Greater than 8~% of binding was found to be specific and less than 5% of binding to complementary peptide plates was observed for insulin or bovine serum albumin plates. ~oth complementary peptides, when present in solution, competed to an equal extend to block 125I-ACTH binding to their respective sorbed counterparts.

These data demonstrate that, for the ACTH system, 5'-3' and 3'-5' comPlementary peptides are equally effective in binding ACTH.

HOMOLOGIES BETWEEN PEPTIDE HORMONES AND
POLY-PEPTIDES CODED BY REVERSELY-READ
NUCLEIC ACIDS COMPLEMENTARY TO NUCLEIC ACIDS
CODING FOR THE PEPTIDE HORMONE RECEPTOR PROTEINS.

Subtle but significant functional and structural relationships exist between peptides codingly specified by complementary strands of nucleic acids. This relationship was reflected by readinq the complementary nucleic acid in the normally transcribed 5' to 3' direction (See, for example, Table 1, Fiqure 1, and ExamPles lA to lH). When the complementary nucleic acids are read in the reverse or 3' to 5' direction, unique relationships of the resultant -83- 13~960~

coded amino acid sequences are similarly apparent as shown in the following examples.

EPIDERMAL GROWTH FACTOR (EGF), EGF RECEPTOR
AND COMPLEMENTARY MESSAGE TO THE EGF RECEPTOR

The amino acid sequence of EGF and its coding nucleo-tide (mRNA) sequence are shown in Figure 8 as taken from Gray et al (Nature (London, 1983), Vol. 303, p. 722) and ~cott et al (Science (1983), Vol. 221, p. 236). Also shown in Figure 8 are a partial amino acid sequence and partial coding nucleotide sequence (c-DNA) for EGF recep-tor as taken from Ulrich et al (Nature London, 1984), Vol.309, p. 418) The final column of Figure 8 shows the nucleotide (RNA) sequence complementary to the RNA sequence which codes for the EGF receptor. An antiparallel base-pairing alignment of the EGF receptor nucleotide sequence and its complementary nucleotide sequence, was assumed. The complementary nucleotide sequence was read in the same reading frame as the coding sequence but and in the 3' to 25 5 ' direction. The coded amino acid sequence shown above the complementary nucleotide sequence was thus obtained.
The XXX codon symbolizes termination. When the amino terminal directions of the amino acid sequences shown in Figure 8, are in the lower numbered direction two homolo-gous regions (appearing in boxes) of the EGF receptor-complementary polypeptide and EGF appear. The entire EGF
receptor complementary sequence (not shown) was analyzed to yield only these two complementary amino acid regions.
EGF arrino acid sequences 11-16 and 24-29 were found to be 35 homologous to amino acid sequences 111-116 and 149-154 -84- 13~9~0~

respectively coded by the nucleotide sequence comple-mentary to that of the EGF receptor.

As shown in Figure 8, with the two homologous regions in EGF consisting of six amino acids, five amino acids are identical in each sequence (83% homology). Furthermore, with the nucleotide sequences there is 67 and 78% nucleo-tide homology, respectively, between the two regions, with most of the nucleotide differences not affecting the encoded amino acids (e.g. third base changes). The two homologous amino acid regions include approximately 23% of the total EGF amino acid sequence (12 of 53 residues), and the homology is so striking that it is highly unlikely that it represents a random event. A non-random basis for this amino acid homology is strongly supported by the observation that when the sequences ASP-GLY-TYR-X-LEU-ASN
and GLU-SER-LEU-X-SER-TYR (where X is any amino acid) were screened against 3060 proteins in the protein sequence bank at the National Biomedical Research Foundation (NBRF), only EGF contained these sequences. The protein sequence database at the National Biomedical Research Foundation searched was the SIAO: [Blomquist] NEW. PRO:
80 file. In total, 3,060 protein sequences were searched, which included 616,748 test segments of 6 amino acids in length or 619,803 test segments of 5 amino acids in length. So as not to bias the search for homologous sequences at positions of difference between the ligand and receptor complement sequences, any amino acid (X) was accepted as a match. Thus, the search for homologous se-quences was not limited to the specific ligand or receptorcomplement sequences shown in Figure 2, but rather allowed any amino acid substitution at positions of difference.
Of 616,748 segments of 6 amino acids in length tested for homology, only EGF contained either of these sequences.
Therefore, the relationship between these particular amino - 13~960O

acid sequences reflects a significant relationship of EGF
and its receptor.

STATISTICAL SIGNIFICANCE OF AMINO ACID AND
NUCLEOTIDE HOMOLOGIES BETWEEN PEPTIDE HORMONES
AND POLY-PEPTIDES CODED BY NUCLEOTIDE SEQUENCES
COMPLEMENTARY TO NUCLEOTIDE SEQUENCES CODING
10FOR PROTEINS OF THE PEPTIDE HORMONE RECEPTORS.

The statistical siqnificance of homology between any two nucleotide sequences was determined by calculating P
values, which are the probabilities that a particular homolo~y occurred accidentally. The equation used was a summation of the Poisson distribution.

20N e~Np (Np)i Pa = ~ i!

Where N is the length of nucleotides in the homologous sequence, i is the number of matches over the sequence and p is the probability that any qiven nucleotide will match.
For ideal randomness, p=0.25 if there is no preference for any nucleotide at any position. To determine if there was any siqnificant deviation from randomness, p values were empirically determined for all three receptors. In actuality, p values were always between 0.25 and 0.27, therefore for simplicity we assumed p=0.25 for calculating the Pa values. For N=18 and N=15 in sequences with ideal randomness, the number of nucleotide matches ti) equals 4.5 and 3.75, respectively. To determine the deviation from randomness of the receptor sequences for N=18 and -86- 133960~

N=15, i values were empirically determined for each receptor and found to be 4.7S ( 1.41) and 3.88 ( 1.45), respectively. To be considered statistically significant, i values had to be ~reater than two standard deviations from the mean (i.e. i 7.57 for N=18 and i 6.78 for N=15).
Thus, Pa values 4.63 x 10 for N=18 or 4.87 x 10 for N=15 were considered statistically significant. Pa values that were less than or equal to 4.63xlO for 18 nucleo-tides and 4.87xlO for 15 nucleotides were determined empirically to be statistically significant. Figure 9A
shows that the nucleotide sequence homologies between EGF
and the two EGF receptor complements have calculated Pa values of 1.60 x 10 3 and 1.78 x 10 , respectively.
Thus, the homologies between these sequences are highly siqnificant with the number of base matches being greater than five standard deviations from the means for ideal randomness.

In further analyses, performed generally according to the procedure of Example 2B, the relationships o~ other~
peptide hormones and their receptors and receptor comple-mentary polypeptides were elucidated.

The amino acid and nucleotide sequences shown in Figure 9B were obtained from the following sources:

Interleukin -2 (IL-2) from Taniquchi et al (Nature (Landon, 1984) Vol. 302, p. 305) and Devos et al (Nucl. Acid Res. (1983) Vol. VII, p. 4307).
Interleukin -2 Receptor (IL-2 Receptor) from Nikaido et al (Nature (London, 1984) Vol. 311, p. 631).
Transferrin (TF) from Yang et al (Proc. Nat'l. Acad.
35Sci. (1984) Vol. 81, p. 2752.
Transferrin Receptor (TF Receptor) from Schneider et al (Nature (London, 1984) Vol. 311, p. 675).

13~60~

As shown in Figures 9A and 9B, results similar to those ~ound with the EGF system were found when IL-2 and TF were searched for homology with their corresponding receptor complements. For IL-2, two homologous regions of 6 and 5 amino acids were found (Figure 9A) with 83 and 80%
amino acid homology, respectively. In addition, the nucleotide homology between the two sequences (61 and 67~, respectively) was highly significant (Pa = 4.26 x 10 and 3.56 x 10 3, respectively). Both amino acid sequences (LEU-GLU-X-LEU-LEU-LEU and TYR-ARG-MET-X-LEU, where X is a~ amino acid) were screened for homoloqies with 3060 proteins in the NBRF sequence bank.

For 616,748 test segments of six amino acids in length, only 7 proteins, including IL-2, were found to have homoloqy with LEU-GLU-X-LEU-LEU-LEU. When the sequence LEU-GLU-X-LEU-LEU-LEU (where X is any amino acid) was screened for homologies against 616,748 test segments of 6 amino acids in length, seven proteins contained homoloqous sequences. These included human IL-2, human and mouse Iq alpha heavy chain, arabinose operon regula-tory protein from E. coli and S. typhimurium, gene k protein of OX-174 and protein 4 from Asperqillus amstelo-dami mitochondria. However, only one protein (IL-2) contained complete homoloqy with IL-2 where X=HIS and only one protein (Protein 4 from Aspergillus amstelodami mitochondria) contained complete homology with the IL-2 receptor complement where X=THR. ~hen the sequence TYR-ARG-MET-X-LEU was screened against 619,803 segments of five amino acids in length, only IL-2 and the hemoglobin alpha chain of the South African toad contained homologous sequences. Taken together, the two homologous sequences were found to be uniquely associated with IL-2.

For TF and its receptor complement, there were many reqions of siqnificant sequence homology, however it should be noted that, due to space limitations, not all regions of homology are shown. The representative sequences shown in Figure 9B have at least 53% nucleotide homology and have Pa values below those considered statis-tically significant. When the amino acid sequences ILE-PRO-X-GLY-LEU-LEU and GLU-PHE-X-LEU-PHE-SER (where X is any amino acid) were screened for homologies against 3,060 proteins in the NBRF sequence bank, only TF contained both sequences. The latter sequence was only found in trans-ferrin while ILE-PRO-X-GLY-LEU-LEU was found in trans-ferrin, lactotransferrin and only three unrelated proteins (bacterial tryptophan synthase, E. coli colicin El immu-nity protein and influenza C hemagglutinin precursor).

From the results presented in Example 2C there can be little doubt that the nucleotide sequences for ligands and receptors contain highly significant regions of comple-mentarity. At the present time these were the onlyliqand-rece~tor pairs for which the complete amino acid and nucleotide sequences were known. Thus, all the sequence data available to date supports the hypothesis that receptor and ligand bindin~ sites could have evolved from complementary strands of nucleic acid. There are several observations supporting the idea that the comple-mentary regions shown here may in fact code for amino acid sequences in the binding site of the receptor. First, the complementary nucleotide sequences were always detected in 30 the portion of the receptor external to the cytoplasmic membrane. For example, the two homologous sequences detected in the EGF receptor complement were in the 100,000 dalton external domain (the domain which binds EGF
in the receptor) whereas no homologies were detected in the 60,000 dalton cytoplasmic domain (the domain with protein kinase activity). This finding was also true for -89- ~33~06 the IL-2 and TF receptors sequences, since in all instances homoloqies were in the external portion which contributes to ligand binding. Secondly, for the ligand, their size ~5-6 amino acids) approximates what one might expect to fill a complete receptor site if one used antibody combinina sites for an example as shown in Nisonoff et al (The Antibody Molecule (Academic Press.
N.Y. 1984) pp. 29-38). These sequences appear to repre-sent binding sites, one of which would be expected to be at each point of contact between the receptor and ligand.
Third, and most importantly, it has been demonstrated, as earlier described herein, that the hormones ACTH and gamma-endorphin bind with high affinity to synthetically derived peptides encoded by RNA complementary to the respective hormone mRNA. This observation demonstrates that amino acid sequences complementary to a peptide do in fact bind that peptide, and therefore the sequence comple-mentary to the peptide must contain a receptor-like hin~ing site. Furthermore, the "synthetic" binding site for ACTH was antigenically related to an ACTH adrenal cell receptor. In total, these observations indicate that peptide-receptor binding sites may ultimately be derived from complementary strands of nucleic acid.

If protein-protein binding interactions evolving from complementary strands of nucleic acids prove to be as general a phenomenon in biology as discerned, there are many potential applications for this concept. For example, the knowledge of ligand sequences would allow easy purification and characterization of receptors usinq methodology similar to that previously described herein.
Valuable information concerning ligand conformations in binding site environments may be obtained by constructing well defined liqand-"binding site" pairs. Ultimately, knowledge of the binding site sequences for receptor-ligand pairs will allow construction of small, well 1339~06 defined receptor aqonists, and/or antagonists valuable for manipulatinq biological responses. These findings may also be important in the investigation and understandinq of differentiation and embryogenesis. For instance, the mere transcription of a DNA sequence by one cell and its comPlement by another could allow for cellular recognition and communication via the resulting peptides or proteins which interact. The concepts described herein may, for instance, provide a qenetic and molecular basis for internal imaqing in the immune system and circuit forma-tion in the central nervous system.

The discoveries described herein, particularly in Examples 2A to 2C, describe a process for preparing polypeptides having an affinity for cellular receptor sites of particular peptide hormones. Said process comprises a series of steps. First, a second nucleotide sequence of a second nucleotide strand base-pairing with a first nucleotide strand coding for at least a portion of a proteinoceous component of a peptide hormone receptor site is ascertained. Homoloqous amino acid sequences between the peptide hormone and the amino acid sequence coded by the second nucleotide sequence, when read in the 3' to 5' direction, are then determined.
Having found these homologous amino acid sequences, which appear responsible for the characteristic binding of peptide hormones to their receptor sites, polypeptides comprising at least a portion of at least one of said homologous sequences may be prepared for example, by routine chemical or biological synthetic methods. These polypeptides, containing key regions of homoloqy and receptor binding affinity with a peptide hormone or liqand, may be screened by commonly utilized techniques as aqonists or antagonists for the peptide hormone or ligand.

1339~06 Angiotensin II and the Design and Obtaining of Complementary Peptides Based on 5Nucleic Acid Sequences The Angiotensin system is shown below:

An~iotensinoqen----> Angiotensin I----> Anqiotensin II

Reaction 1 is an enzymatic cleavage by the enzyme renin and reaction 2 is an enzymatic cleavage by angiotensin converting enzyme (ACE). Anqiotensinogen contains 453 amino acid residues, Angiotensin I consists of the 10 N-terminal residues of Anqiotensinogen, and Angiotensin II contains the 8 N-terminal residues of Angiotensin I. Anqiotensin II is the active molecule that controls blood pressure regulation.
The nucleic acid sequence for rat Angiotensin II was obtained from Ohkubo et al. (Proc. Nat. Acad. Sci. USA, 80, p. 2197-2200, 1983) by translating the entire mRNA for angiotensino~en and observing the amino acid sequence (from base 134 through 157) known to be Angiotensin II.
The sequence, its translation, its complement, and the complementary amino acids determined by 3' and 5' reaading are shown in the following table:

30 Angiotensin II ASP ARG VAL TYR ILE HIS PR~ ~E Carboxy Terminus mRNA GAC CGC GUA UAC AUC CAC CCC UUU 3' Terminus cRNA CUG GCG CAU AUG UAG GUG GGG AAA 5' Terminus 3' Reading LEU ALA HIS MET END VAL GLY LYS
5' Reading VAL ALA TYR VAL ASP VAL GLY LYS

Since 3' reading gave a termination siqnal (UAG/END), ~SP was substituted for EN~. Complementary peptides were ~3~606 ohtained by conventional solid-phase synthesis from Triton Biosciences Inc. (Alameda, CA) for the following complementary peptides. Note that two peptides have parallel peptide bond orientations and two have anti-parallel.

SEQUENCE

~esi~nation N Terminus C Terminus 5CA-AII Rat LYS GLY VAL ASP VAL TYR ALA VAL
3CA-AII Rat (4ASP) LYS GLY VAL ASP MET HIS ALA LEU
5CP-AII Rat VAL ALA TYR VAL ASP VAL GLY LYS
3CP-AII Rat (5ASP) LEU ALA HIS MET ASP VAL GLY LYS

Angiotensin II and the Desiqn and Obtaininq of Complementary Peptides Based on Amino Acid Sequences (Consensus Complements) The amino acid sequence of human Angiotensin II was taken from the sequence of a fragment of human Angiotensinogen determined by Tewksbury et al. (BBRC, 99, p. 1311-1315, 1981).

Using the substitutions given in Table 6A two consensus complements of Angiotensin II were designed, one with parallel peptide bond orientation and one with anti-parallel orientation as shown below:

CCA-AII LYS-GLY-VAL-TYR-ILE-HIS-ALA-LEU
CCP-AII LEU-ALA-HIS-ILE-TYR-VAL-GLY-LYS

These complementary peptides were obtained by conventional solid-phase synthesis from Triton Biosciences ~nc. (Alameda, CA).

133~506 Angiotensin II and Design and Obtaining of Conplementary Peptides Based on Amino Acid Sequences 5(Simplified Complements) Using the amino acid sequence for Angiotensin II from the prior example and the substitutions given in Table 6C, the following (anti-parallel) simplified complementary peptide to Angiotensin II (hu~an) was designed.

SCA-AII GLU-GLY-LEU-GLU-LEU-GLU-ALA-LEU

This complementary peptide was obtained by conventional solid-phase synthesis from Triton Biosciences Inc. (Alameda, CA).

EXA~IPLE 3D

Angiotensin and Design and Obtaining of Related Complementary Peptides Based on the known network of reactions in the angiotensin system, complementary peptides were prepared that could reveal multiple beneficial effects.

~ lolecules are designed, based on the complementary peptide binding observations, that can specifically interact with one or more of the angiotensin system molecules. As a result of such interactions, and angiotensin molecule will not undergo enzymatic reaction.
Thus, the conversion of, for example, angiotensin I to Angiotensin II could be reduced without actually inhibiting ACE or its other functions.

~94~ 1339~06 Based on the human angiotensinogen fragment sequence for Example 3B, the following two peptides were desiqned using the consensus complenment method.

5 CCA-AI GLU-VAL-LYS-GLY-VAL-IYR-I~-HIS-ALA-~U
CCA-A(1-13) VAL-TYR-HIS-GLU-VAL-LYS-GLY-VAL-TYR-I~ ~IS-ALA-~U

These complementary peptides were obtained by solid-phase synthesis from Triton Biosciences Inc. (Alameda, CA).

The molecule CCA-A(1-13) miqht be expected to bind all three members of the anqiotensin family shown above, and thereby inhibit reactions 1 and 2 and inhibit the ability of Angiotensin II to activate its receptor.

Angiotensin II and the Design and Obtaining of Potentially Metabolically Stable Complementary Peptides Based on the structure of one of the complementary peptides of an~iotensin II (CCA-AII, Example 3B) several derivatives were designed that might show improved metabol;c stability to various peptidases. Both amino-and carboxy- peptidases are known to be present in many organisms. It is known that many modifications to peptide structures can protect peptides from the action of these enzymes. Acylation (Ac) of terminal amino groups and prolyation and amidation (Am) of terminal carboxy groups are common methods to protect peptides from amino- and carboxy- peptidases, respectively.

The following molecules were designed based on these principles and the structure of the active complementary peptide CCA-AII.

Desiqnation Structure ~ 3 ~ ~ ~ 0 6 CCA-AII (~m) LYS-GLY-VAL-TYR-ILE-HIS-ALA-LEU-Am CCA-AII (PR09) LYS-GLY-VAL-TYR-ILE-HIS-ALA-LEU-PRO
5 CCA-AII (Ac, Am) Ac-LYS-GLY-V~L-TYR-ILE-HIS-ALA-LEU-Am CCA-AII (Ac, PR09) Ac-LYS-GLY-VAL-TYR-ILE-HIS-ALA-LEU-PRO
The molecules were obtained by conventional solid-phase chemistry from Triton Biosciences Inc. (Alameda, CA). Amide resins were used in solid-phase synthesis to obtain admidated peptides. Acylation was carried out by reacting acetic anhydride with fully protected peptide while still on the resin of the solid-phase synthesis.

15Another method to protect peptides from enzymatic attack is to substitute D-amino acids for the naturally occurinq L-amino acids. For this reason, an all D
complementary peptide was desiqned based on the sequence of CCA-AII (Example 3B).
CCA-AII (all D) LYS-GLY-VAL-TYR-ILE-HIS-ALA-LEU

This molecule was obtained by conventional solid-phase synthesis from Triton Biosciences Inc. (Alameda, 25 CA).

Effect of Peptides Complementary to 30Angiotensin II on the Binding of Radiolabelled Angiotensin II to its Receptor Inhibition of angiotensin II binding to anqiotensin II receptors by complementary peptides was tested with measurement through the use of radioactive angiotensin II.
Rabbit livers were homogenized, and were centifuqed in order to isolate particles sedimenting between 1,000 and 100,000 xq. Binding activity was solublized with 1%

~ ~ 3 9 ~

diqitonin, followed by amrnonium sulfate fractionation between 49 and 65% satuation followed by DEAE- cellulose chromatoaraphy at pH 7.5 usinq a linear gradient between 0.0 and 0.3 M KCl. The partially purified, solubilized 5 receptor preparation bound 17 pmoles of angiotensin II per ng protein when analyzed by Scatchard analysis, indicatinq a purity of approximately 0.1%.

The standard assay for hindinq of anqiotensin II to 10 the receptor is as follows: The complete system (150 ul) contains 30 ~M Tris--HCl, pH 7-5, 2-5 mM K2EDTA, 0.2 mM
PCMS, 0.25 nM [125 I] angiotensin II (ca. 100,000 cpm), 100 ug BSA, 0.25% (V/V) Brij 99*and 30 ug of partially purified receptor. The reaction is initiated by addition 15 of the receptor and samples are incubated for 60 minutes at 20~C. The reaction is terminated with 1 ml of cold 0.5% charcoal/0.05% Dextran in 100 mM Tris-HCl, pH 7.5.
Tubes are vortexed and then allowed to stand 10 minutes at 4~C, after which they are centrifuged and their 20 supernatants, containing protein-bound angiotensin II, are counted.

The complete systerl under these conditions regularly yields about 10,000 cpm of bound radioactivity. A control 25 lacking receptor yields values of 50-200 cpm which were subtracted from these data. Values of 50-2000 cpm were obtained when 120 uM cold antiotensin II is present in the reaction mixture indicating that virtually all binding is specific. A sample including 20 nM cold angiotensin II
30 was also run. Residual binding of radioactivity in this control was 35-4596. Complementary peptides to angiotensin II were dissolved in water, except for CCA-A (1-13) which was dissolved in 3% DMSO, 0.04 M acetic acid and 0.05 M
HCl. Results of these assays are given in Table 22. ID50 35 is the concentration of peptide that inhibits binding of radiolabelled angiotensin II by 50%.
*Trade Mark 1~39~0~

Inhibition by Complementary Peptides of Angiotensin II Binding to Isolated Hepatic Receptor Peptide ID50 (nM) Anqiotensin II 15 CCA-A (1-13) 40 CCA-AII (Am) 160 CCA-AII (Ac, Am) 3,100 CCA-AII (Pro 9) 40 CCA-AII (Ac, Pro 9) 1,350 CCA-AII (all D) >10,000 5CA-AII Rat 4,000 3CA-AII Rat (4 ASP) 5,000 5CP-AII Rat >10,000 3CP-AII Rat (5 ASP) >10,000 Luteininzing Hormone Releasing Hormone (LHRH) and the Design and Obtaininq of a Complementary Peptide Luteinizing hormone releasinq hormone has a wide variety of biological effects and receptors for it occurs presumably on many cell types (Miller, et al. Nature, 313, p. 231-233, 1985). The nucleic acid sequence for the precursor form of LHRH has been reported (Seeburg and Adelman, Nature, 311, p 666-668, 1984) and was used in the following design of a complementary peptide. The sequence for LHRH, its translation, its complement, and the 5'-3' translation of its complement are shown below.

-98- 13 3~ ~0 6 L~ GLN-HIS-T ~ SER-TYR-GLY-L~-ARG~PRO-G~ C terminus mRNA C ~ CAC-T&G-TCC-TAT~X~X~-CCr~ 3' terminus cRNA GTC~ACC-~K~ATA-CCT ~ C-3~{~-CCr 5' termi~s 5CA-~ ~ -V~L-~}GLY-I~-SER-GLN-ALA-A ~ SER N te~i~s The c~mplementary peptide 5CA-LHRH was obtained by solid-phase synthesis from lriton Biosciences Inc.
(Alameda, CA).

Effect of a Peptide Complementary to LHRH
(5CA-LHRH~ on LHRH-Stimulated LH Release from Pituitary Cells A reverse hemolytic plaque assay (Smith, et al.), Method in Enzymology, 124, p. 443) was used to examine the effect of a peptide (5CA-LHRH) complementary to LHRH on LHRH-stimulated release of LH from pituitary cells. The assay was performed as referenced, except that in the experiments labelled as (Pre), the complementary peptide and LHRH were preincuhated for 1 hour before addition to dispersed pituitary cells.

Plaques were analyzed by determining plaque area with an imaqe analysis (Bioquant*or Image Technology Corp.
Model 300) of 125x-500x microscopic enlargements. Results are presented as the percentage response of a particular assay to control assay (plaques formed under stimulation by 5xlO 10 M LHRH).

The effect of the complementary peptide (5CA-LHRH) is presented in Table 23:

*Trade Mark -99- 1339~0~

~ABLE 23 Exp. 1 Exp. 2 Exp. 3 Exp. 4 Exp. 5 Treatment (Pre) (No) (Pre) (Pre) (No) None 7 5 0.03 2 2 Control 5x10 M LHRH100 100 100 100 100 10 5x10 M LH~+5CA-LHRH

10-5~ 82 52 68 63 70 1510_8~ 101 74 88 78 115 10 gM 105 83 NT 96 92 5x10 M LHRH+Somatostatin 2510 M NT NT 2.7 NT NT
~6M NT NT 0.5 NT NT
10 M NT NI 0.5 NT NT
The results show a clear inhibition of the effects of LHRH in 30 this assay ~ the co~plementary peptide 5CA-LHRH.

EFFECT OF ANTIBODY TO 5CA-LHRH (A5CALHRH) ON

The reverse hemolytic plaque assay for LH secretion by dispersed pituitary cells of proestrous rats were performed essentially as described in Example 4B, except that the pituitary cells were preincubated with A5CALHRH
or the normal rabbit serum (NRS) for two hours. After washinq, reverse hemolytic plaque formations was initiated with the addition of LHRH and anti-LH antiserum. Two hours later, com~lement was added for 30 minutes.
A5CALHRH at the same concentration was also added during -lOO- 133~06 reverse hemolytic plaque formation. A5CALHRHBl is the IqG
fraction of antiserum from the first bleed at 40 days after immunization of a male rabbit with 5CA-LHRH coupled to Keyhole Limpet Hemocyanin (300ug antigen in lml complete Freund's adjuvant, initial injection Day O, 200uq antiqen in lml incomplete Freund's adjuvant, injections on days 10, 20, 30, 40, 50 and 60). A5CALHRHB3 is from the third bleed of the rabbit at day 60. F(ab)2 fragments of A5CALHRHBl were produced by conventional methods (Methods In Immunology, 3rd ed., p. 256, 1977). Results are shown ~ in Table 24.

1333~06 Treatment ~ Control None 10 20 LHRH 5xlO M 100 (Control) A5CALHRHBl 1:10 + LHRH 5xlO 1O M 140 1:25 + LHRH 5xlO 1 oM 158 1:100 + LHRH 5xlO M 178 A5CALHRHBl F9Ab)2*
1:10 + LHRH 5xlO 0 M 75 1:25 + LHRH 5xlO 110 M 132 1:100 + LHRH 5xlO M 146 NRS
1:10 + LHRH 5xlO 10 M 78 1:25 + LHRH 5xlO -10 M 118 1:100 + LHRH 5xlO M 93 1:5 5 1:10 3 1:25 10 3S 1:100 28 1:5 + LHRH 5xlO 110M 27 1:10 + LHRH 5xlO 10 M 25 1:25 + LHRH 5xlO 10M 46 1:100 + LHRH 5xlO M 184 * Pituitary cells are reported to have receptors for the FC portion of antibody molecules. (Pou~lane et al, Nature 261, 142 (1976), Buffa et al Histochem 63 15 (1979)) F(Ab)2 frag~ents of A5CALHRH were prepared to ensure that any effect on LHRH stimulation of pituitary 1~3~6~

cells was not due to interaction of A5CALHRH with non-specific FC receptors.

These results show the presence of both antagonistic and agonistic antibodies in the immune serum of rabbits immunized with a peptide comple~entary to LHRH.

EXAMPLE 4n Immunocytochemical Assay for LHRH Cell Surface Receptors Based on Antibodies to Peptides Complementary to LHRH

Various cells have receptors for LHRH on their surfaces. It is well-known that 5% of pituitary cells have such receptors since they respond to LHRH by releasing LH. The followinq experiments were performed to demonstrate the ability of antibodies to peptides complementary to LHRH to specifically label those pituitary cells containing LHRH receptors.

After standard plaque assays were performed (as in Example 4B), the chambers were infused with sodium acetate to elute the bound antibodies and fixed sequentially in B5 fixative, ethanol, Lugol's iodide and sodium thiosulfate (as described in ~mith et al., Methods in Enzymology, 124, p. 443). The slides were treated with hydrogen peroxide and normal goat serum before application of suitable dilutions of the antibodies. Immunocytochemistry was performed with the ABC method (Vector Labs, Burlingame, CA) with diaminobenzidine as substrate.

Cells containing LH were selectively stained with antibodies to LH. In general, plaques formed in the assays of earlier examples contained one LH-containing cell near the center of each circular plaque. Cells -103- 13~9~0~

presenting LHRH receptors were stained using IgG fractions of i~mune serum from rabbits immunized with a peptide complementary to LHRH (see Example 4C). Again, one cell per circular aque was stained and these were the same - 5 cells that contained LH. Control experiments demonstrated that receptor staininq by antibody to the complement of LHRH could be blocked with both LHRH (by binding to receptor) and by the comple~ent to LHRH (by binding to the antihody).
These experiments demonstrate that the antibodies to the complement of LHRH selectively stain pituitary cells that present LHRH receptors.

Ribonuclease A S-Peptide and the Design and Obtaining of a Complementary Peptide Treatment of bovine ribonuclease A (RNase), a 124 amino acid protein, with the proteolytic enyzme, subtilisin, cleaves RNase into two fragments designated S-peptide, amino acid residues 1-20, and S-protein, amino acid residues 21-124. The m-RNA sequence for rat RNase was obtained from MacDonald et al. (J. Biol. Chem., 257, p. 14582-14585, 19~2), whic~ shows substantial sequence homology with hovine RNase. Residues 4 through 23 share ho~ology with the S-peptide of bovine S-peptide. A
putative M-RNA structure for bovine RNase was derived by making the minimal number of base changes in the rat m-RNA
sequence and is shown in the followinq table:

Rat RNase amino acid residues 4-23 ARG GLU SER SER
ALA ASP LYS PHE LYS ARG GLN HIS MET ASP THR GLU GLY PRO
SER LYS

~33~06 Rat m-RNA sequence for rat RNase residues 4-23 reading 5' to 3' AGG GAA UCA UCG GCG GAU AAG UUU AAG AGG
CAG CAC AUG GAC ACA GAG GGU CCC UCC AGG

Putative m-RNA for bovine S-peptide reading 5' to 3' AAG GAA ACA GCG GCG GCU AAG UUU GAG AGG GAG CAC AUG GAC
UCA UCG ACU UCC GCC GCG

Amino acid sequence of bovine S-peptide LYS GLU THR
ALA ALA ALA LYS PHE GLU ARG GLU HIS MET ASP SER SER THR
SER ALA ALA

The amino acids for the complementary peptide to bovine S-peptide were determined from the nucleic acid structure by readinq the com~lementary strand in the 5' to 3' reading frame as shown in the followinq table:

Complementary nucleic acid structure for bovine S-peptide CGC GGC GGA AGU CGA UGA GUC CAU GUG CUC CCU CUC
AAA CUU AGC CGC CGC UGU UUC CUU

The parallel complementary amino acid sequence for bovine S-peptide LEU PHE SER ARG ARG SER LEU LYS LEU PRO
LEU VAL HIS VAL SER ARG SER GLY GLY ARG
Since the sixth codon gave a termination signal (UGA/END), SER was substituted for END. For codon 18 (UGU/CYS), SER was substituted for CYS. These amino acid substitutions maintain second base complementarity with the bovine S-peptide. The complementary peptide was obtained by conventional solid-phase synthesis from Triton Biosciences Inc. (Alameda, CA).

~33g~06 Ribonuclease A S-Peptide and the Binding and Purification of a Complementary Peptide The interaction of the S-peptide complementary peptide with S-peptide was examined by covalently coupling S-peptide to a N-hydroxysuccinimide activated silica gel bonded phase using conventional chemistry. The peptide mixture obtained from hydrogen fluoride treatment of the complementary peptide usinq conventional chemistry was applied directly to the S-peptide chromatographic column (3mm by 44mm), previously equilibrated at either 0.10 M
Tris-acetate, pH 7.0, or 0.10 M Tris-acetate, pH 5.1, at a flow rate of 1 ml/min. The column effluent was monitored by absorhance at 226 nm. Under these conditions, a large peak composed of peptide and non-peptide material was obtained at the solvent front. The column was washed with 45 ml of buffer, followed by 30 ml of water, and then 0.10 M acetic acid was applied to the column which eluted 226 nm absorbing material. Chromatography of this eluted material on a ~aters*reversed phase C-18 column usinq a flow rate of 1 ml/min and a gradient elution for 10~
solution A to 40% solution B over 35 min (solution A, 0.1%
trifluroacetic acid/water: solution B, 0.1%
trifluoroacetic acid/100% acetonitrile) gave a single peak with a retention time of approximately 26 minutes.
Sequencin~ this material using a gas phase sequenator (Applied Biosystems, Foster City, CA) confirmed that its structure was identical to the synthesized complementary peptide. Control peptides did not bind to the S-peptide affinity column. Thesç results demonstrate a specific bindinq between the S-peptide and one of its complements and also demonstrates that the S-peptide affinity column can be used to puri~y crude preparations of peptide.

*Trade Mark ~'' 1339i~06 * * * * * * * *

Chan~es may be made in the construction, operation and arrange~ent of the various amino acids, elements, steps and procedures described herein without departing from the concept and scope of the invention as defined in the followinq claims.

Claims (110)

1. A method for determining the amino acid sequence of a polypeptide having complementary binding affinity for at least a portion of an original peptide or protein, comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine;
substituting in place of each valine of the ascertained amino acid sequence, glutamine or histidine;
substituting in place of each leucine of the ascertained amino acid sequence, asparagine, aspartic acid or glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, lysine;
substituting in place of each cysteine of the ascertained amino acid sequence, threonine;
substituting in place of each methionine of the ascertained amino acid sequence, tyrosine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine or serine;
substituting in place of each lysine of the ascertained amino acid sequence, phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, leucine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, leucine;
substituting in place of each glutamine of the ascertained amino acid sequence, valine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each glycine of the ascertained amino acid sequence, proline;
substituting in place of each threonine of the ascertained amino acid sequence, tryptophan or cysteine;
substituting in place of each tryptophan of the ascertained amino acid sequence, threonine;
substituting in place of each serine of the ascertained amino acid sequence, arginine or retaining said serine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine or methionine;
substituting in place of each proline of the ascertained amino acid sequence, glycine; and determining the amino acid sequence obtained after the above substitutions.
2. The method of claim 1 defined further wherein:
serine is substituted in place of each arginine of the ascertained amino acid sequence;
serine is retained in place of each serine of the ascertained amino acid sequence; and cysteine is substituted in place of each threonine of the ascertained amino acid sequence.
3. The method of claim 2 wherein the complementary polypeptide is defined further as retaining complementarity or binding affinity for the original peptide or protein regardless of the amino terminal and carboxy-terminal directionality of said complementary polypeptide.
4. A method for determining a polypeptide having complementary binding affinity for at least a portion of an original peptide or protein, comprising the steps of:

ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine;
substituting in place of each valine of the ascertained amino acid sequence, glutamine or histidine;
substituting in place of each leucine of the ascertained amino acid sequence, asparagine, aspartic acid or glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, lysine;
substituting in place of each cysteine of the ascertained amino acid sequence, threonine;
substituting in place of each methionine of the ascertained amino acid sequence, tyrosine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine or serine;
substituting in place of each lysine of the ascertained amino acid sequence, phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, leucine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, leucine;
substituting in place of each glutamine of the ascertained amino acid sequence, valine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each glycine of the ascertained amino acid sequence, proline;
substituting in place of each threonine of the ascertained amino acid sequence, tryptophan or cysteine;
substituting in place of each tryptophan of the ascertained amino acid sequence, threonine;
substituting in place of each serine of the ascertained amino acid sequence, arginine or retaining said serine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine or methionine;
substituting in place of each proline of the ascertained amino acid sequence, glycine; and obtaining a polypeptide comprising the amino acid sequence determined by the above substitutions.
5. The method of claim 4 defined further wherein:
serine is substituted in place of each arginine of the ascertained amino acid sequence;
serine is retained in place of each serine of the ascertained amino acid sequence; and cysteine is substituted in place of each threonine of the ascertained amino acid sequence.
6. The method of claim 4 wherein the obtaining step comprises chemically synthesizing said polypeptide.
7. The method of claim 4 wherein the obtaining step comprises excising said polypeptide from a protein or larger polypeptide including said amino acid sequence.
8. The method of claim 4 wherein the obtaining step is defined further as comprising insertion of a DNA nucleotide sequence coding for said polypeptide into a plasmid to form a recombinant DNA vector and transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said polypeptide.
9. The method of claim 8 wherein the unicellular organism is selected from the group consisting of bacterial cells and yeast cells.
10. A method for determining the amino acid sequence of a polypeptide having complementary binding affinity for at least a portion of an original protein or peptide, comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original protein or peptide;
reading the ascertained amino acid sequence, starting from the carboxy-terminal direction thereof, to substitutingly correspond to the amino-terminal direction of the complementary polypeptide;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine, asparagine or aspartic acid;
substituting in place of each leucine of the ascertained amino acid sequence, lysine, glutamine or glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, threonine or alanine;
substituting in place of each methionine of the ascertained amino acid sequence, histidine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine, serine, glycine or cysteine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine, serine, threonine or proline;
substituting in place of each lysine of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each glutamine of the ascertained amino acid sequence, leucine;

substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each histidine of the ascertained amino acid sequence, valine or methionine;
substituting in place of each glycine of the ascertained amino acid sequence, proline, serine, threonine or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine, serine, arginine or cysteine;
substituting in place of each tryptophan of the ascertained amino acid sequence, proline;
substituting in place of each serine of the ascertained amino acid sequence, glycine, threonine, alanine or arginine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each proline of the ascertained amino acid sequence, glycine, arginine or tryptophan; and determining the amino acid sequence found after the above substitutions.
11. The method of claim 10 defined further wherein:
aspartic acid or histidine is substituted in place of each valine of the ascertained amino acid sequence;
lysine or glutamine is substituted in place of each leucine of the ascertained amino acid sequence;
glutamic acid is substituted in place of each phenylalanine of the ascertained amino acid sequence;
alanine or proline is substituted in place of each arginine of the ascertained amino acid sequence;
leucine is substituted in place of each lysine of the ascertained amino acid sequence;
valine is substituted in place of each histidine of the ascertained amino acid sequence;

proline or alanine is substituted in place of each glycine of the ascertained amino acid sequence;
glycine or arginine is substituted in place of each threonine of the ascertained amino acid sequence;
glycine, arginine or alanine is substituted in place of each serine of the ascertained amino acid sequence;
valine is substituted in place of each tyrosine of the ascertained amino acid sequence; and glycine or arginine is substituted in place of each proline of the ascertained amino acid sequence.
12. A method of obtaining a polypeptide having complementary binding affinity for at least a portion of an original peptide or protein, comprising the steps of ascertaining the amino acid sequence of at least a portion of the original protein or peptide;
reading the ascertained amino acid sequence starting from the carboxy-terminal direction thereof to substitutingly correspond to the amino-terminal direction of the complementary polypeptide;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine, asparagine or aspartic acid;
substituting in place of each leucine of the ascertained amino acid sequence, lysine, glutamine or glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, threonine or alanine;
substituting in place of each methionine of the ascertained amino acid sequence, histidine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine, serine, glycine or cysteine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine, serine, threonine or proline;

substituting in place of each lysine of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each glutamine of the ascertained amino acid sequence, leucine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each histidine of the ascertained amino acid sequence, valine or methionine;
substituting in place of each glycine of the ascertained amino acid sequence, proline, serine, threonine or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine, serine, arginine or cysteine;
substituting in place of each tryptophan of the ascertained amino acid sequence, proline;
substituting in place of each serine of the ascertained amino acid sequence, glycine, threonine, alanine or arginine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each proline of the ascertained amino acid sequence, glycine, arginine or tryptophan; and obtaining a polypeptide comprising the amino acid sequence determined by the above substitutions.
13. The method of claim 12 wherein the obtaining step comprises chemically synthesizing said polypeptide.
14. The method of claim 12 wherein the obtaining step further comprising excising said polypeptide from a protein or larger polypeptide including said amino acid sequence.
15. The method of claim 12 wherein the obtaining step comprises insertion of a DNA nucleotide sequence coding for said polypeptide into a plasmid to form a recombinant DNA
vector and transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said polypeptide.
16. The method of claim 12 wherein the unicellular organism is selected from the group consisting of bacterial cells and yeast cells.
17. The method of claim 12 defined further wherein:
substituting in place of each valine of the ascertained amino acid sequence, aspartic acid or histidine;
substituting in place of each leucine of the ascertained amino acid sequence, lysine or glutamine;
substituting in place of each phenylalanine of the ascertained amino acid sequence, glutamic acid;
substituting in place of each arginine of the ascertained amino acid sequence, alanine or proline;
substituting in place of each lysine of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each glycine of the ascertained amino acid sequence, proline or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine or arginine;
substituting in place of each serine of the ascertained amino acid sequence, glycine, arginine or alanine;
substituting in place of each tyrosine of the ascertained amino acid sequence, valine; and substituting in place of each proline of the ascertained amino acid sequence, glycine or arginine.
18. A method for determining the amino acid sequence of a polypeptide having complementary binding affinity for at least a portion of an original peptide or protein wherein the amino acids of the polypeptide, original peptide and original protein are defined as being in groups according to the second base of their codons, comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each A group amino acid, an amino acid of the U group;
substituting in place of each U group amino acid, an amino acid of the A group;
substituting in place of each C group amino acid, an amino acid of the G group;
substituting in place of each G group amino acid, an amino acid of the C group; and determining, after the above substitutions, the resulting amino acid sequence.
19. A method for obtaining a polypeptide having complementary binding affinity for at least a portion of an original peptide or protein wherein the amino acids of the polypeptide, original peptide or original protein are defined as contained in groups (U, A, C or G) according to the second base of their codons, comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each A group amino acid, an amino acid of the U group;
substituting in place of each U group amino acid, an amino acid of the A group;
substituting in place of each C group amino acid, an amino acid of the G group; and substituting in place of each G group amino acid, an amino acid of the C group; and obtaining a polypeptide comprising the amino acid sequence defined by the above substitutions.
20. The method of claim 19 wherein the obtaining step comprises chemically synthesizing said polypeptide.
21. The method of claim 19 wherein the obtaining step comprises excising said polypeptide from a protein or larger polypeptide including said amino acid sequence.
22. The method of claim 19 wherein the obtaining step comprises insertion of a DNA nucleotide sequence including the code of said polypeptide into a plasmid to form a recombinant DNA vector and transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said polypeptide.
23. The method of claim 22 wherein the unicellular organism is selected from the group consisting of bacterial cells and yeast cells.
24. A method for determining the amino acid sequence of a polypeptide having a minimum complementary peptide binding activity for at least a portion of an original peptide or protein comprising the steps of:
(a) determining a first nucleotide sequence of a first nucleic acid, said first nucleotide sequence coding for an amino acid sequence of at least a portion of an original peptide or protein;
(b) ascertaining a second nucleotide sequence of a second nucleic acid which base-pairs with the first nucleotide sequence of the first nucleic acid, the first and second nucleic acids pairing in antiparallel directions;
(c) determining the amino acid sequence of the complementary polypeptide by finding the amino acid sequence coded by the second nucleotide sequence when read in the same reading frame as the first nucleotide sequence.
25. The method of claim 24 wherein the second nucleotide sequence is read in the 5' to 3' direction.
26. The method of claim 24 wherein the second nucleotide sequence is read in the 3' to 5' direction.
27. The method of claim 24 wherein the first nucleic acid is defined further as being DNA.
28. The method of claim 24 wherein the first nucleic acid is defined further as being messenger RNA.
29. A method for obtaining a polypeptide having a minimum complementary peptide binding activity for at least a portion of an original peptide or protein comprising the steps of:
(a) determining a first nucleotide sequence of a first nucleic acid, said first nucleotide sequence coding for an amino acid sequence of at least a portion of an original peptide or protein;
(b) ascertaining a second nucleotide sequence of a second nucleic acid which base-pairs with the first nucleotide sequence of the first nucleic acid, the first and second nucleic acids pairing in antiparallel directions;
(c) determining the amino acid sequence of the complementary polypeptide by finding the amino acid sequence coded by the second nucleotide sequence when read in the same reading frame as the first nucleotide sequence; and (d) obtaining a polypeptide comprising the amino acid sequence determined in step (c).
30. The method of claim 29 wherein step (d) is defined further as comprising chemically synthesizing said polypeptide.
31. The method of claim 29 wherein step (d) is defined further as obtaining said polypeptide from a protein or larger polypeptide including said amino acid sequence.
32. The method of claim 29 wherein the second nucleic acid is defined further as being DNA and step (d) is defined further as comprising insertion of the second nucleotide sequence into a plasmid to form a recombinant DNA vector and transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said complementary polypeptide.
33. The method of claim 32 wherein the unicellular organism is selected from the group consisting of bacteria, and yeast.
34. The method of claim 29 wherein the second nucleotide sequence is read in the 5' to 3' direction.
35. The method of claim 29 wherein the second nucleotide sequence is read in the 3' to 5' direction.
36. The method of claim 29 wherein the first nucleic acid is defined further as being DNA.
37. The method of claim 31 wherein the first nucleic acid is defined further as being messenger RNA.
38. A method for preparing a polypeptide having a minimum complementary peptide binding activity for the cellular receptor site for a particular peptide ligand, comprising the steps of:
ascertaining a second nucleotide sequence of a second nucleotide strand base-pairing with a first nucleotide strand coding for at least a part of a protein portion of a peptide ligand receptor site;
determining any amino acid sequences in the peptide ligand which are homologous to amino acid sequences coded by the second nucleotide sequence when said second sequence is read in a 3' to 5' direction; and preparing a polypeptide comprising at least a portion of at least one of said homologous amino acid sequences.
39. A method for preparing polypeptides having a minimum complementary peptide binding activity for a polypeptide ligand based on its cellular receptor site, comprising the steps of:
ascertaining a second nucleotide sequence of a second nucleotide strand base-pairing with a first nucleotide strand coding for at least a part of a protein portion of a peptide ligand receptor site;
determining any amino acid sequences in the peptide ligand that are homologous to amino acid sequences coded by the second nucleotide sequence when said second sequence is read in a 3' to 5' or 5' to 3' direction;
determining the amino acid sequences of the receptor site for the polypeptide ligand that correspond to the homologous amino acid sequences of the preceding step;
and preparing a polypeptide comprising at least a portion of at least one of said amino acid sequences of the receptor site.
40. A polypeptide complementary to at least a portion of an original peptide or protein, said polypeptide being produced by a process comprising the steps of:

(a) determining a first nucleotide seguence of a first nucleic acid, said first nucleotide sequence coding for an amino acid sequence of at least a portion of the original peptide or protein;
(b) ascertaining a second nucleotide sequence of a second nucleic acid which base-pairs with the first nucleotide sequence of the first nucleic acid, the first and second nucleic acids pairing in antiparallel directions;
(c) determining the amino acid sequence of the complementary polypeptide by finding the amino acid sequence coded by the second nucleotide sequence when read in the same reading frame as the first nucleotide sequence; and (d) producing a polypeptide comprising the amino acid sequence determined in step (c).
41. The polypeptide of claim 40 wherein step (d) is defined further as comprising chemically synthesizing said polypeptide.
42. The polypeptide of claim 40 wherein step (d) is defined further as obtaining said polypeptide from a protein or larger polypeptide including said amino acid sequence.
43. The polypeptide of claim 40 wherein the second nucleic acid is defined further as being DNA and step (d) is defined further as comprising insertion of the second nucleotide sequence into a plasmid to form a recombinant DNA
vector and transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said complementary polypeptide.
44. The polypeptide of claim 43 wherein the unicellular organism is defined further as selected from a class consisting of bacteria and yeast.
45. The polypeptide of claim 40 wherein the second nucleotide sequence is read in the 5' to 3' direction.
46. The polypeptide of claim 40 wherein the second nucleotide sequence is read in the 3' to 5' direction.
47. The polypeptide of claim 40 wherein step (a) is defined further as determining a sequence of nucleotide triplet codons of the first nucleotide sequence coding for at least a portion of the amino acid sequence of the original peptide.
48. The polypeptide of claim 40 wherein the first nucleic acid is defined further as being DNA.
49. The polypeptide of claim 40 wherein the first nucleic acid is defined further as being messenger RNA.
50. The polypeptide of claim 40 wherein the first nucleotide sequence and second nucleotide sequence are defined further as comprising triplet nucleotide codons.
51. The polypeptide of claim 40 defined further as retaining complementary or binding affinity for the original peptide or protein regardless of the amino terminal and carboxy-terminal directionality of said complementary polypeptide.
52. The polypeptide of claim 40 wherein the original peptide or protein is defined further as being a cell surface component and the complementary polypeptide is defined further as being conjugated to a toxin or diagnostic agent such as a fluorescent label and the conjugate specifically adheres to said cell surface component.
53. The polypeptide of claim 40 wherein the original peptide or protein is defined further as being an enzyme and the polypeptide is defined further as inhibiting catalytic activity of said enzyme.
54. The polypeptide of claim 40 wherein the original peptide or protein is defined further as having toxic effects and the polypeptide is defined further as lessening or obviating said toxic effects, when administered in vivo or in vitro.
55. The polypeptide of claim 40 wherein the original peptide or protein is defined further as being at least a portion of a hormone and the polypeptide is defined further as binding to said hormone and thereby lessening or obviating its biological activity.
56. The polypeptide of claim 40 wherein the original peptide or protein is defined further as being a blood-group antigen and the polypeptide is defined further as being at least divalent or having a label attached thereto to facilitate identification of said blood-group antigen.
57. The polypeptide of claim 40 wherein the original peptide or protein is defined further as being a pregnancy-specific component of biological fluids and the polypeptide is defined further as being coupled to a fluorescent dye, radioisotope or enzyme yielding chromophorically measurable products to facilitate pregnancy testing.
58. A method for detecting and determining a peptide or protein in a sample containing said peptide or protein to be determined which comprises:
(a) obtaining a polypeptide of claim 40 complementary to at least a portion of said peptide or protein;
(b) chemically coupling said complementary polypeptide to a label to form a labelled conjugate of the polypeptide;
(c) contacting the sample containing the peptide or protein to be determined with the labelled conjugate of the complementary polypeptide and thereby forming a chemical complex or couple in which the peptide or protein to be determined is bound to the labelled conjugate of the complementary polypeptide; and (d) detecting and determining the peptide or protein to be determined by assaying for the labelled conjugate bound to the peptide or protein.
59. The method of claim 58 wherein the label is selected from the group consisting of enzymes, heavy metals, radioisotopes and fluorescent compounds.
60. A polypeptide according to claim 40 for use in the treatment of a disease state characterized by the overproduction or presence of an unwanted proteinaceous substance in an organism to be treated;
the polypeptide being complementary to at least a portion of said unwanted proteinaceous substance;
the complementary polypeptide being introduced into the organism to be treated whereby the complementary polypeptide comes into contact with and binds to the unwanted proteinaceous substance, thereby rendering said substance biologically inactive.
61. An antibody for use in the treatment of a disease state characterized by the underproduction or absence of a proteinaceous substance in an organism to be treated;
wherein said antibody is an antibody to the complementary peptide of claim 40;
said polypeptide being complementary to at least a portion of said proteinaceous substance;
said antibody being introduced into the organism to be treated whereby the antibody functions to replace or supplement the proteinaceous substance in its designated biological function in the organism.
62. A method for detecting a peptide or protein in an organism containing said peptide or protein which comprises:
(a) obtaining a polypeptide of claim 40 complementary to at least a portion of said peptide or protein;
(b) chemically coupling said complementary polypeptide to a label to form a labelled conjugate of the polypeptide;
(c) administering the conjugate to the organism so that the conjugate contacts said peptide or protein contained therein to form a chemical complex or couple in which the peptide or protein is bound to the conjugate; and (d) detecting the peptide or protein to be detected by assaying for the labelled conjugate bound to the peptide or protein.
63. A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
ascertaining the amino acid corresponding to the most frequently used codon for the species of interest for each group of codons with second bases G, C, A and U or T;
substituting in place of each amino acid in the ascertained sequence, the ascertained amino acid from the group of amino acids with second base complementary to the second base of the most frequently used codon in the species of interest for the amino acid in the ascertained sequence.
64. A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
ascertaining the most frequently used codon for each amino acid in the sequence for the species of interest such that the 5' to 3' reading of the complementary codon does not result in a stop codon;
substituting in place of each amino acid of the ascertained amino acid sequence, the amino acid defined by the 5' to 3' translation of the complement of the ascertained most frequently used codon.
65. A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
ascertaining the most frequently used codon for each amino acid in the sequence for the species of interest such that the 3' to 5' reading of the complementary codon does not result in a stop codon;
substituting in place of each amino acid of the ascertained amino acid sequence, the amino acid defined by the 3' to 5' translation of the complement of the ascertained most frequently used codon.
66. A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original protein or peptide, comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original protein or peptide;
reading the ascertained amino acid sequence from carboxy terminus to amino terminus aligning the carboxy terminus of the ascertained amino acid sequence with the amino terminus of the determined sequences;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine, asparagine or aspartic acid;
substituting in place of each valine of the ascertained amino acid sequence, asparagine, aspartic acid, histidine or tyrosine;
substituting in place of each leucine of the ascertained amino acid sequence, lysine, glutamine or glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, lysine or glutamic acid;
substituting in place of each cysteine of the ascertained amino acid sequence, threonine or alanine;
substituting in place of each methionine of the ascertained amino acid sequence, histidine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine, serine, glycine or cysteine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine, serine, threonine or proline;
substituting in place of each lysine of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each glutamine of the ascertained amino acid sequence, leucine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each histidine of the ascertained amino acid sequence, valine or methionine;
substituting in place of each glycine of the ascertained amino acid sequence, proline, serine, threonine or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine, serine, arginine or cysteine;
substituting in place of each tryptophan of the ascertained amino acid sequence, proline;
substituting in place of each serine of the ascertained amino acid sequence, glycine, threonine, alanine or arginine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine or valine; and substituting in place of each proline of the ascertained amino acid sequence, glycine, arginine or tryptophan.
67. The method of claim 66 defined further wherein:
substituting in place of each valine of the ascertained amino acid sequence, aspartic acid or histidine;
substituting in place of each leucine of the ascertained amino acid sequence, lysine or glutamine;
substituting in place of each phenylalanine of the ascertained amino acid sequence, glutamic acid;
substituting in place of each arginine of the ascertained amino acid sequence, alanine or proline;
substituting in place of each lysine of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each glycine of the ascertained amino acid sequence, proline or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine or arginine;
substituting in place of each serine of the ascertained amino acid sequence, glycine, arginine or alanine;
substituting in place of each tyrosine of the ascertained amino acid sequence, valine; and substituting in place of each proline of the ascertained amino acid sequence, glycine or arginine.
68. A method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein comprising the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine;
substituting in place of each valine of the ascertained amino acid sequence, histidine;
substituting in place of each leucine of the ascertained amino acid sequence, glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, lysine;
substituting in place of each methionine of the ascertained amino acid sequence, tyrosine or histidine;
substituting in place of each lysine of the ascertained amino acid sequence, phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, leucine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, leucine;
substituting in place of each glutamine of the ascertained amino acid sequence, leucine or valine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine;
substituting in place of each cysteine of the ascertained amino acid sequence, threonine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine;
substituting in place of each glycine of the ascertained amino acid sequence, proline;
substituting in place of each tryptophan of the ascertained amino acid sequence, threonine or proline;
substituting in place of each serine of the ascertained amino acid sequence, serine or arginine;
substituting in place of each threonine of the ascertained amino acid sequence, cysteine or serine;
substituting in place of each proline of the ascertained amino acid sequence, glycine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine;
69. The method of any one of claims 63 to 68 wherein the determined amino acid sequence is defined further as retaining complementary or binding affinity for the original peptide regardless of the amino-terminal and carboxy-terminal directionality of the determined amino acid sequence.
70. A method of obtaining a polypeptide complementary to at least a portion of an original peptide or protein characterized by obtaining a polypeptide comprising the amino acid sequence determined in any one of claims 63 to 68.
71. The method of claim 70 wherein the polypeptide is obtained by chemically synthesizing said polypeptide.
72. The method of claim 70 wherein the polypeptide is obtained from a protein or larger polypeptide including the amino acid sequence of the obtained polypeptide.
73. The method of claim 70 wherein the polypeptide is obtained by a) inserting a DNA sequence which encodes, at least in part, the amino acid sequence of said polypeptide into a plasmid to form a recombinant DNA vector and b) transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said polypeptide.
74. The method of claim 73 wherein the unicellular organism is selected from the group comprising bacteria and yeast.
75. The method of claim 72 wherein the polypeptide is obtained by excising said polypeptide from a protein or larger polypeptide including the amino acid sequence of the obtained polypeptide.
76. A polypeptide complementary to gamma-endorphin and comprising the amino acid sequence: H2N-Gln-Arg-Asp-Lys-Gly-Arg-Leu-Ala-Leu-Leu-Gly-Gly-His-Glu-Pro-Ala-Val-COOH.
77. A polypeptide complementary to luteinizing hormone releasing hormone (LHRH) comprising the sequence H2N-Ser-Arg-Ala-Gln-Ser-Ile-Gly-Pro-Val-Leu-COOH.
78. Polypeptides complementary to adrenocorticotrophic hormone (ACTH) and selected from the class:
a) NH2-Arg-Met-Arg-Tyr-Leu-Val-Lys-Ala-Thr-Pro-Phe-Gly-His-Pro-Phe-Phe-Ala-Ala-Gly-His-Phe-His-Met-Gly-COOH;
b) NH2-Arg-Val-Gly-His-Phe-Val-Glu-Ala-Pro-Ala-Leu-Arg-His-Ala-Leu-Leu-Pro-Ala-Arg-His-Leu-His-Val-Gly-COOH;
c) NH2-Phe-His-Gly-Val-Arg-COOH;
d) NH2-Ala-Pro-Ala-Glu-Val-Phe-His-Gly-Val-Arg-COOH;
e) NH2-His-Arg-Ala-Pro-Leu-Leu-Ala-His-Arg-Leu-Ala-Pro-Ala-Glu-Val-Phe-His-Gly-Val-Arg-COOH; and f) H2N-Gly-Val-His-Leu-His-Arg-Ala-Pro-Leu-Leu-Ala-His-Arg-Leu-Ala-Pro-Ala-Glu-Val-Phe-His-Gly-Val-Arg-COOH.
79. A method of purifying peptides or proteinaceous substances which comprises:
a) obtaining a polypeptide complementary to at least a portion of said peptide or proteinaceous substance where said polypeptide is defined by any one of claims 40, 45 and 46;
b) affixing said polypeptide to a suitable solid or polymeric support material;
c) contacting the support material having bound polypeptide with a solution containing the peptide or proteinaceous substance, thereby selectively binding said peptide or proteinaceous substance to said bound polypeptide;
d) washing or eluting the support material having bound polypeptide to which the peptide or proteinaceous substance is also bound with a solvent to remove unbound material;
e) washing or eluting the support material having bound polypeptide to which the peptide or proteinaceous substance is also bound with a second solvent to remove, in purified form in solution, the desired peptide or proteinaceous substance.
80. An antibody to a polypeptide , whereby said polypeptide:
a) is complementary, sequence position by sequence position, to an original peptide ligand when oriented either amino terminus to carboxy terminus or carboxy terminus to amino terminus with respect to the original peptide ligand;
b) is encoded by a DNA strand which is complementary to a portion of a DNA strand encoding the original peptide ligand as defined by the complementary DNA
codons, or the second base of said complementary codons, for each strand base pairing in an antiparallel direction, in each case being read in either the 3' to 5' or 5' to 3' direction in the same reading frame as the reading frame for the DNA codons encoding the original peptide ligand, and c) has binding affinity for a portion of the original peptide ligand, said antibody being characterised by having an affinity for a receptor site at which the peptide ligand binds and transmits its hormonal effects.
81. The antibody of claim 80 wherein the peptide ligand is adrenocorticotrophic hormone (ACTH) and the polypeptide has the amino acid sequence;
H2N-Gly-Val-His-Leu-His-Arg-Ala-Pro-Leu-Leu-Ala-His-Arg-Leu-Ala-Pro-Ala-Glu-Val-Phe-His-Gly-Val-Arg-COOH.
82. The antibody of claim 80 characterized in that the antibody is generated against antigen comprising in whole or in part a complementary polypeptide complementary to at least a portion of an original peptide or protein, said polypeptide being produced by a process characterized by the steps of:
a) determining a first nucleotide sequence of a first nucleic acid, said first nucleotide sequence coding for an amino acid sequence of at least a portion of the original peptide or protein;
b) ascertaining a second nucleotide sequence of a second nucleic acid which base-pairs with the first nucleotide sequence of the first nucleic acid, the first and second nucleic acids pairing in antiparallel directions;
c) determining the amino acid sequence of the complementary polypeptide by finding the amino acid sequence coded by the second nucleotide sequence when read in the 5' to 3' direction or the 3' to 5' direction and in the same reading frame as the first nucleotide sequence; and d) producing a polypeptide comprising the amino acid sequence determined in step (c).
83. The antibody of claim 82 characterized in that the second nucleotide sequence is read in the 5' to 3' direction.
84. The antibody of claim 82 characterized in that the second nucleotide sequence is read in the 3' to 5' direction.
85. The antibody of claim 80 characterized in that the antibody is generated against antigen comprising in whole or in part a complementary polypeptide obtained by a method for obtaining a polypeptide complementary to at least a portion of an original peptide or protein, characterized by the steps of ascertaining the amino acid sequence of at least a portion of the original protein or peptide; reading the ascertained amino acid sequence from carboxy terminus to amino terminus;
aligning the carboxy terminus of the amino acid sequence with the amino terminus of the determined sequence; substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine, asparagine or aspartic acid;
substituting in place of each valine of the ascertained amino acid sequence, asparagine, aspartic acid, histidine or tyrosine;
substituting in place of each leucine of the ascertained amino acid sequence, lysine, glutamine or glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, lysine or glutamic acid;
substituting in place of each cysteine of the ascertained amino acid sequence, threonine or alanine;
substituting in place of each methionine of the ascertained amino acid sequence, histidine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine, serine, glycine or cysteine;
substituting in place of each arginine of the ascertained amino acid sequence, alanine, serine, threonine or proline;
substituting in place of each lysine of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each aspartic acid of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each glutamine of the ascertained amino acid sequence, leucine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine or phenylalanine;
substituting in place of each histidine of the ascertained amino acid sequence, valine or methionine;
substituting in place of each glycine of the ascertained amino acid sequence, proline, serine, threonine or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine, serine, arginine or cysteine;
substituting in place of each tryptophan of the ascertained amino acid sequence, proline;
substituting in place of each serine of the ascertained amino acid sequence, glycine, threonine, alanine or arginine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine or valine;
substituting in place of each proline of the ascertained amino acid sequence, glycine, arginine or tryptophan; and obtaining a polypeptide comprising the amino acid sequence determined by the above substitutions.
86. The antibody of claim 85 characterized in that the polypeptide is obtained by chemical synthesis.
87. The antibody of claim 85 characterized in that the polypeptide is obtained by excising it from a protein or larger polypeptide including said amino acid sequence.
88. The antibody of claim 85 characterized in that the polypeptide is obtained by inserting a DNA sequence encoding it into a plasmid and transforming a unicellular organism or mammalian cell with said plasmid, whereupon said unicellular organism or mammalian cell biosynthesizes said polypeptide.
89. The antibody of claim 88 characterized in that the unicellular organism is selected from bacterial cells and yeast cells.
90. The antibody of claim 85 characterized in that the sequence of the complementary polypeptide is obtained by:
substituting in place of each valine of the ascertained amino acid sequence, aspartic acid or histidine;
substituting in place of each leucine of the ascertained amino acid sequence, lysine or glutamine;
substituting in place of each phenylalanine of the ascertained amino acid sequence, glutamic acid;
substituting in place of each arginine of the ascertained amino acid sequence, alanine or proline;
substituting in place of each lysine of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each glycine of the ascertained amino acid sequence, proline or alanine;
substituting in place of each threonine of the ascertained amino acid sequence, glycine or arginine;
substituting in place of each serine of the ascertained amino acid sequence, glycine, arginine or alanine;
substituting in place of each tyrosine of the ascertained amino acid sequence, valine; and substituting in place of each proline of the ascertained amino acid sequence, glycine or arginine.
91. The antibody of claim 80 characterised in that the antibody is generated against antigen comprising in whole or in part a complementary polypeptide obtained by a method for obtaining a polypeptide complementary to at least a portion of an original peptide or protein, wherein the amino acids of the polypeptide, original peptide or original protein are defined as contained in groups (U, A, C or G) according to the second base of their codons, characterised by the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each A group amino acid, an amino acid of the U group;
substituting in place of each U group amino acid, an amino acid of the A group;
substituting in place of each C group amino acid, an amino acid of the G group; and substituting in place of each G group amino acid, an amino acid of the C group; and obtaining a polypeptide comprising the amino acid sequence defined by the above substitutions.
92. The antibody of claim 91 characterised in that the polypeptide is obtained by chemical synthesis.
93. The antibody of claim 91 characterised in that the polypeptide is obtained by excising it from a protein or larger polypeptide including said amino acid sequence.
94. The antibody of claim 91 characterised in that the polypeptide is obtained by inserting a DNA sequence encoding said polypeptide into a plasmid and transforming a unicellular organism or mammilian cell with said plasmid whereupon said unicellular organism or mammalian cell biosynthesizes said polypeptide.
95. The antibody of claim 91 characterised in that the unicellular organism is selected from bacteria cells and yeast cells.
96. The antibody of claim 80 characterized in that the antibody is generated against antigen comprising in whole or part a complementary polypeptide complementary to at least a portion of an original peptide or protein, said polypeptide being produced by a process characterised by the steps of:
(a) determining the amino acid sequence of said polypeptide by a method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein characterised by the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
substituting in place of each isoleucine of the ascertained amino acid sequence, tyrosine;
substituting in place of each valine of the ascertained amino acid sequence, histidine;
substituting in place of each leucine of the ascertained amino acid sequence, glutamic acid;
substituting in place of each phenylalanine of the ascertained amino acid sequence, lysine;
substituting in place of each methionine of the ascertained amino acid sequence, tyrosine or histidine;
substituting in place of each lysine of the ascertained amino acid sequence, phenylalanine;
substituting in place of each asparagine of the ascertained amino acid sequence, leucine;

substituting in place of each aspartic acid of the ascertained amino acid sequence, leucine;
substituting in place of each glutamine of the ascertained amino acid sequence, leucine or valine;
substituting in place of each glutamic acid of the ascertained amino acid sequence, leucine;
substituting in place of each histidine of the ascertained amino acid sequence, valine;
substituting in place of each tyrosine of the ascertained amino acid sequence, isoleucine;
substituting in place of each cysteine of the ascertained amino acid sequence, threonine;
substituting in place of each arginine (acid) of the ascertained amino acid sequence, alanine;
substituting in place of each glycine of the ascertained amino acid sequence, proline;
substituting in place of each tryptophan of the ascertained amino acid sequence, threonine or proline;
substituting in place of each serine of the ascertained amino acid sequence, serine, or arginine;
substituting in place of each threonine of the ascertained amino acid sequence, cysteine or serine;
substituting in place of each proline of the ascertained amino acid sequence, glycine;
substituting in place of each alanine of the ascertained amino acid sequence, arginine;
(b) producing the polypeptide comprising the amino acid sequence determined in step (a).
97. The antibody of claim 80 characterised in that the antibody is generated against antigen comprising in whole or in part a complementary polypeptide complementary to at least a portion of an original peptide or protein, said polypeptide being produced by a process characterised by the steps of:
(a) determining the amino acid sequence of said polypeptide by a method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein characterised by the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
ascertaining the most frequently used codon for each amino acid in the sequence of the species of interest such that the 5 to 3 reading of the complementary codon does not result in a stop codon;
substituting in place of each amino acid of the ascertained amino acid sequence, the amino acid defined by the 5 to 3 translation of the complement of the ascertained most frequently used codon;
(b) producing the polypeptide comprising the amino acid sequence determined in step (a).
98. The antibody of claim 80 characterised in that the antibody is generated against antigen comprising in whole or in part a complementary polypeptide complementary to at least a portion of an original peptide or protein, said polypeptide being produced by a process characterised by the steps of:
(a) determining the amino acid sequence of said polypeptide by a method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein characterised by the steps of:
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
ascertaining the most frequently used codon for each amino acid in the sequence of the species of interest such that the 3 to 5 reading of the complementary codon does not result in a stop codon;
substituting in place of each amino acid of the ascertained amino acid sequence, the amino acid defined by the 3' to 5' translation of the complement of the ascertained most frequently used codon;
(b) producing the polypeptide comprising the amino acid sequence determined in step (a).
99. The antibody of claim 80 characterised in that the antibody is generated against antigen comprising in whole or in part a complementary polypeptide complementary to at least a portion of an original peptide or protein, said polypeptide being produced by a process characterised by the steps of:
(a) determining the amino acid sequence of said polypeptide by a method for determining the amino acid sequence of a polypeptide complementary to at least a portion of an original peptide or protein characterised by the steps of :
ascertaining the amino acid sequence of at least a portion of the original peptide or protein;
ascertaining the amino acid corresponding to the most frequently used codon for the species of interest for each group of codons with second bases G, C, A and U/T;
substituting in place of each amino acid in the ascertained sequence, the ascertained amino acid from the group of amino acids with second base complementary to the second base of the most frequently used codon in the species of interest for the amino acid in the ascertained sequence;
(b) producing the polypeptide comprising the amino acid sequence determined in step (a).
100. The antibody of any one of claims 96 to 99 characterised in that the polypeptide is produced by chemical synthesis.
101. The antibody of any one of claims 96 to 99 characterised in that the polypeptide is obtained by excising it from a protein or larger polypeptide including said amino acid sequence.
102. The antibody of any one of claims 96 to 99 characterised in that the polypeptide is obtained by inserting a DNA acid sequence encoding it into a plasmid and transforming a unicellular organism or mammalian cell therewith to produce a transformant unicellular organism or mammalian cell biosynthesizing said complementary peptide.
103. The antibody of claim 102 characterised in that the unicellular organism is selected from bacteria, and yeast.
104. The antibody of any one of claims 96 to 99 characterised in that the complementary polypeptide retains complementarity or binding affinity for the original peptide or protein regardless of the amino-terminal and carboxy-terminal directionality of said complementary polypeptide.
105. The antibody of any one of claims 82 to 99 characterised in that the antibody is a monoclonal antibody.
106. The antibody of claim 80 characterised in that the peptide ligand is gamma-endorphin and the polypeptide comprises the amino acid sequence:

H2N-Gln-Arg-Asp-Lys-Gly-Arg-Leu-Ala-Leu-Leu-Gly-Gly-His-Glu-Pro-Ala-Val-COOH.
107. Antibody to polypeptide prepared by a method for preparing a polypeptide having an affinity for the cellular receptor site for a particular peptide ligand, characterised by the steps of:
ascertaining a second nucleotide sequence of a second nucleotide strand base-pairing with a first nucleotide strand coding for at least a portion of a proteinaceous component of a peptide ligand receptor site;

determining any amino acid sequences in the peptide ligand which are homologous to amino acid sequences coded by the second nucleotide sequence when said second sequence is read in a 3' to 5' direction; and preparing a polypeptide comprising at least a portion of at least one of said homologous amino acid sequences.
108. Antibody to a polypeptide prepared by a method for preparing polypeptides having an affinity for a polypeptide ligand, said method based on the sequence of its cellular receptor site said method being characterised by the steps of:
ascertaining a second nucleotide sequence of a second nucleotide strand base-pairing with a first nucleotide strand coding for at least a portion of a proteinaceous component of a peptide ligand receptor site;
determining any amino acid sequences in the peptide ligand that are homologous to amino acid sequences coded by the second nucleotide sequence when said second sequence is read in a 3' to 5' or 5' to 3' direction;
determining the amino acid sequences of the proteinaceous receptor for the polypeptide ligand that correspond to the homologous regions of the preceding step;
and preparing a polypeptide comprising at least a portion of at least one of said amino acid sequences of the proteinaceous receptor.
109. A method for obtaining components of a peptide ligand receptor site from a mixture, characterised by the steps of:
preparing the antibody of any one of claims 80 to 99 and 106;
coupling the antibody to a solid matrix;
treating the mixture with the antibody-coupled matrix to specifically bind components of the receptor site;
and eluting the bound components.
110. A method of detecting receptors for proteinaceous substances on cell surfaces or fluids of an organism which is characterised by:
a) obtaining the antibody of any one of claims 80 to 99 and 106;
b) introducing said antibody to cell surfaces or fluids of an organism;
c) detecting the presence of said antibody bound to the receptors of said proteinaceous substance.
CA000502748A 1985-03-01 1986-02-26 Polypeptides complementary to peptides or proteins having an amino acid sequence or necleotide coding sequence at least partially known Expired - Fee Related CA1339606C (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US708,001 1985-03-01
US06/708,001 US4863857A (en) 1985-03-01 1985-03-01 Polypeptide complementary to peptides or proteins having an amino acid sequence or nucleotide coding sequence at least partially known
US06/829,709 US5077195A (en) 1985-03-01 1986-02-19 Polypeptides complementary to peptides or proteins having an amino acid sequence or nucleotide coding sequence at least partially known and methods of design therefor
US829,709 1986-02-19

Publications (1)

Publication Number Publication Date
CA1339606C true CA1339606C (en) 1997-12-30

Family

ID=27108003

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000502748A Expired - Fee Related CA1339606C (en) 1985-03-01 1986-02-26 Polypeptides complementary to peptides or proteins having an amino acid sequence or necleotide coding sequence at least partially known

Country Status (18)

Country Link
US (2) US5077195A (en)
EP (2) EP0332225B1 (en)
JP (1) JPH07215999A (en)
KR (1) KR920002315B1 (en)
AT (2) ATE88572T1 (en)
AU (1) AU592781B2 (en)
BR (1) BR8606615A (en)
CA (1) CA1339606C (en)
DE (2) DE3679017D1 (en)
DK (1) DK521386A (en)
ES (3) ES8705919A1 (en)
FI (1) FI90255C (en)
GR (1) GR860576B (en)
IE (1) IE58837B1 (en)
IL (1) IL96043A0 (en)
NO (1) NO174971C (en)
PT (1) PT82132B (en)
WO (1) WO1986005208A1 (en)

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU592324B2 (en) * 1984-10-29 1990-01-11 Roche Diagnostics Corporation Mehods for protein binding enzyme complementation assays
DE3527568A1 (en) * 1985-08-01 1987-02-05 Bissendorf Peptide Gmbh SYNTHETIC PEPTIDE RECEPTORS, METHOD FOR THE PRODUCTION AND USE THEREOF
CA1297003C (en) * 1985-09-20 1992-03-10 Jack H. Nunberg Composition and method for treating animals
EP0294390A1 (en) * 1986-02-19 1988-12-14 Triton Biosciences Inc. Peptides affecting blood pressure regulation
US4772684A (en) * 1987-01-20 1988-09-20 Triton Biosciences, Inc. Peptides affecting blood pressure regulation
JPH02104595A (en) * 1988-03-04 1990-04-17 Univ Alabama Peptide corresponding substance of non-peptide and design method thereof
DE69033395D1 (en) * 1989-07-31 2000-01-20 Tecnogen Scpa Process for the identification and production of binding sites of interacting proteins
US5324512A (en) * 1990-12-26 1994-06-28 The Population Council [Gln']-luteinizing hormone releasing hormone conjugate of tetanus vaccine and its uses
JPH05262788A (en) * 1991-09-03 1993-10-12 Hitachi Chem Co Ltd Peptides, designing and production thereof, and determination of the bonding position of physiologically active peptide on the receptor
US5620675A (en) 1992-06-23 1997-04-15 Diatech, Inc. Radioactive peptides
US6017512A (en) * 1992-06-23 2000-01-25 Diatide, Inc. Radiolabeled peptides
WO1995009643A1 (en) * 1993-10-07 1995-04-13 Kurashiki Boseki Kk Endothelin activity inhibitor
US6051206A (en) * 1994-06-03 2000-04-18 Diatide, Inc Radiolabeled somatostatin-derived peptides for imaging and therapeutic uses
IL115177A0 (en) * 1994-09-16 1995-12-31 Immunomedics Inc Phosphorus-32 labeling of antibodies for cancer therapy
US5523208A (en) * 1994-11-30 1996-06-04 The Board Of Trustees Of The University Of Kentucky Method to discover genetic coding regions for complementary interacting proteins by scanning DNA sequence data banks
EP0835446B1 (en) 1995-06-26 2003-04-09 Perseptive Biosystems, Inc. High speed, automated, continuous flow, multi-dimensional molecular selection and analysis
US5807552A (en) * 1995-08-04 1998-09-15 Board Of Regents, The University Of Texas System Compositions for conferring immunogenicity to a substance and uses thereof
DK1039931T3 (en) * 1997-12-01 2005-08-08 Fang Fang Multivalent recombinant antibodies to treat HRV infections
DE19805334A1 (en) * 1998-02-11 1999-08-12 Otogene Biotechnologische Fors Process for the development of an active pharmaceutical ingredient
EP1078099A4 (en) * 1998-04-24 2005-02-09 Fang Fang Identifying peptide ligands of target proteins with target complementary library technology (tclt)
US20030035798A1 (en) * 2000-08-16 2003-02-20 Fang Fang Humanized antibodies
EP1560590B1 (en) * 1999-03-09 2008-10-22 Fornix Biosciences N.V. Synthetic complementary peptides and ophthalmologic uses thereof
GB2356401A (en) 1999-11-19 2001-05-23 Proteom Ltd Method for manipulating protein or DNA sequence data
GB9929469D0 (en) * 1999-12-13 2000-02-09 Proteom Ltd Complementary peptide ligands generated from plant genomes
GB9929464D0 (en) * 1999-12-13 2000-02-09 Proteom Ltd Complementary peptide ligande generated from the human genome
GB9929466D0 (en) * 1999-12-13 2000-02-09 Proteom Ltd Complementary peptide ligands generated from microbial genome sequences
AU2001294822A1 (en) * 2000-09-26 2002-04-08 Geneformatics, Inc. Methods for determining aproximations of three dimensional polypeptide structure
GB0025251D0 (en) * 2000-10-14 2000-11-29 Proteom Ltd Antisense peptides to Fc receptors and their uses
DE60238308D1 (en) * 2001-02-20 2010-12-30 Univ Georgia QUICK PRODUCTION MONOCLONAL ANTIBODY
JP2005522192A (en) * 2001-07-19 2005-07-28 パーラン セラピューティクス, インコーポレイテッド Multimeric proteins and methods of making and using multimeric proteins
AU2008266120B2 (en) * 2007-06-14 2013-06-20 The Board Of Regents For Oklahoma State University Vaccines containing canine parvovirus genetic variants
US8227583B2 (en) 2007-06-14 2012-07-24 The Board Of Regents For Oklahoma State University Vaccines containing canine parvovirus genetic variants
WO2019046634A1 (en) * 2017-08-30 2019-03-07 Peption, LLC Method of generating interacting peptides

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6936694B1 (en) * 1982-05-06 2005-08-30 Intermune, Inc. Manufacture and expression of large structural genes

Also Published As

Publication number Publication date
IE58837B1 (en) 1993-11-17
ATE63138T1 (en) 1991-05-15
EP0332225A2 (en) 1989-09-13
EP0332225A3 (en) 1989-09-27
PT82132B (en) 1989-05-31
FI90255C (en) 1994-01-10
DK521386A (en) 1986-12-29
EP0214232B1 (en) 1991-05-02
KR920002315B1 (en) 1992-03-21
NO174971C (en) 1994-08-10
EP0332225B1 (en) 1993-04-21
ES8800266A1 (en) 1987-11-01
BR8606615A (en) 1987-08-11
NO864369D0 (en) 1986-10-31
EP0214232A1 (en) 1987-03-18
ES8800267A1 (en) 1987-11-01
NO864369L (en) 1987-01-02
DE3679017D1 (en) 1991-06-06
KR870700100A (en) 1987-02-28
ES552534A0 (en) 1987-05-16
FI90255B (en) 1993-09-30
NO174971B (en) 1994-05-02
DE3688333T2 (en) 1993-10-28
FI864427A0 (en) 1986-10-30
GR860576B (en) 1986-07-01
WO1986005208A1 (en) 1986-09-12
DE3688333D1 (en) 1993-05-27
AU5510286A (en) 1986-09-24
PT82132A (en) 1986-04-01
FI864427A (en) 1986-10-30
IL96043A0 (en) 1991-07-18
US5077195A (en) 1991-12-31
ES557189A0 (en) 1987-11-01
ES557188A0 (en) 1987-11-01
ES8705919A1 (en) 1987-05-16
JPH07215999A (en) 1995-08-15
US5212072A (en) 1993-05-18
IE860527L (en) 1986-09-01
AU592781B2 (en) 1990-01-25
ATE88572T1 (en) 1993-05-15
DK521386D0 (en) 1986-10-31

Similar Documents

Publication Publication Date Title
CA1339606C (en) Polypeptides complementary to peptides or proteins having an amino acid sequence or necleotide coding sequence at least partially known
US4863857A (en) Polypeptide complementary to peptides or proteins having an amino acid sequence or nucleotide coding sequence at least partially known
US5314992A (en) Lipocortin-1 receptor protein and its uses
EP0605489A1 (en) Modified peptide derivatives
Meo et al. Monoclonal antibody to the message sequence Tyr-Gly-Gly-Phe of opioid peptides exhibits the specificity requirements of mammalian opioid receptors.
US5686597A (en) Thrombin receptor homolog
CA2224799A1 (en) A human edg-2 receptor homolog
Terskiy et al. Search of the human proteome for endomorphin-1 and endomorphin-2 precursor proteins
Roth et al. Nonopiate active proenkephalin‐derived peptides are secreted by T helper cells
Arai et al. Identical structural changes in inherited albumin variants from different populations.
Nyberg et al. Identification of high molecular weight enkephalin percursor forms in human cerebrospinal fluid
US5352772A (en) 75 Kilodalton interleukin-2 receptor proteins and their uses
US6682896B1 (en) Peptides representing epitopic sites on r-IFN-β, antibodies thereto and uses thereof
JP2717319B2 (en) Peptide representing R-IFN-beta epitope site, antibody thereto, and use thereof
Cockle et al. Processing of the thyrotropin releasing hormone (TRH) precursor in Xenopus skin and bovine hypothalamus: evidence for the existence of extended forms of TRH
Nag et al. Separation of complexes of major histocompatibility class II molecules and known antigenic peptide by metal chelate affinity chromatography
Oppenheim et al. Molecular basis of salivary proline-rich protein and peptide synthesis: cell-free translations and processing of human and macaque statherin mRNAs and partial amino acid sequence of their signal peptides
Yan et al. Neutrophil-activating intercrine secreted by porcine platelets is active without proteolytic processing
Katamura et al. Biochemical identification of glycosylation inhibiting factor.
Glass et al. Antibodies against Synthetic Phosphorylation Site Peptides
Putnam Electrophoretic and structural characterization of genetic variants of human serum albumin
MXPA97009746A (en) Homologo de receptor de tromb

Legal Events

Date Code Title Description
MKLA Lapsed