WO1997046251A9 - PEPTIDES IMMUNOREACTIFS PAR RAPPORT AUX aPL, LEURS CONJUGUES ET PROCEDES DE TRAITEMENT DES PATHOLOGIES DEPENDANTES DE L'ANTICORPS aPL - Google Patents

PEPTIDES IMMUNOREACTIFS PAR RAPPORT AUX aPL, LEURS CONJUGUES ET PROCEDES DE TRAITEMENT DES PATHOLOGIES DEPENDANTES DE L'ANTICORPS aPL

Info

Publication number
WO1997046251A9
WO1997046251A9 PCT/US1997/010075 US9710075W WO9746251A9 WO 1997046251 A9 WO1997046251 A9 WO 1997046251A9 US 9710075 W US9710075 W US 9710075W WO 9746251 A9 WO9746251 A9 WO 9746251A9
Authority
WO
WIPO (PCT)
Prior art keywords
apl
phage
antibody
peptide
analog
Prior art date
Application number
PCT/US1997/010075
Other languages
English (en)
Other versions
WO1997046251A1 (fr
Filing date
Publication date
Priority claimed from US08/660,092 external-priority patent/US6207160B1/en
Application filed filed Critical
Priority to AU36404/97A priority Critical patent/AU734638B2/en
Priority to EP97933138A priority patent/EP0954531A1/fr
Priority to JP10500927A priority patent/JP2000512981A/ja
Priority to CA002256449A priority patent/CA2256449A1/fr
Publication of WO1997046251A1 publication Critical patent/WO1997046251A1/fr
Publication of WO1997046251A9 publication Critical patent/WO1997046251A9/fr
Priority to NO985636A priority patent/NO985636L/no

Links

Definitions

  • This invention is in the field of immunology and relates to compositions and methods for treating and diagnosing antiphospholipid (aPL) antibody- mediated pathologies. More specifically, the invention relates to conjugates of chemically-defined nonimmunogenic valency platform molecules and immunospecific analogs of aPL-binding epitopes as well as methods and compositions for producing these conjugates. Optimized analogs lack T cell epitopes. In addition, the invention relates to diagnostic assays for detecting the presence of and quantitating the amount of antiphospholipid antibodies in a biological sample. The invention also relates to a method of utilizing random peptide libraries to identify immunospecific analogs of aPL-binding epitopes.
  • Antiphospholipid antibodies occur in autoimmune diseases such as systemic lupus erythematosus (SLE) and antiphospholipid antibody syndrome (APS) as well as in association with infections and drug therapy.
  • SLE systemic lupus erythematosus
  • APS antiphospholipid antibody syndrome
  • APS is characterized by one or more clinical features such as arterial or venous thrombosis, thrombocytopenia and fetal loss.
  • APS may be primary or it may be associated with other conditions, primarily, SLE.
  • POSPHOLIPID-BINDING ANTIBODIES Harris et al., eds., CRC Press, Boca Raton, FL, 1991
  • Transient aPL antibodies such as those detected in a VDRL test, occur during many infections. Approximately 30% of patients possessing persistent aPL antibodies have suffered a thrombic event. The presence of aPL antibodies defines a group of patients within SLE who display a syndrome of clinical features consisting of one or more of thrombosis, TCP, and fetal loss. The risk of this syndrome in SLE overall is around 25%; this risk increases to 40% in the presence of aPL antibodies and decreases to 15% in their absence.
  • aPL antibodies were thought to be directed at phospholipids in plasma membranes, it has been postulated that they may exert direct pathogenic effects in vivo by interfering with hemostatic processes that take place on the phospholipid membranes of cells such as platelets or endothelium.
  • the fact that aPL antibodies appear to be the only risk factor present is further evidence that these antibodies have a direct pathogenic role.
  • Induction of PAPS by immunizing mice with human anticardiolipin antibodies is the best evidence yet that aPL antibodies are directly pathogenic (Bakimer et al. 1992 J. Clin. Invest. 89:1558-1563; Blank et al. 1991 Proc. Natl. Acad. Sci. 88:3069-3073).
  • aPL antibodies recognize an antigenic complex comprised of ⁇ 2-glycoprotein I ( ⁇ 2-GPI) and negatively-charged phospholipid, e.g., cardiolipin (McNeil et al. (1990) Proc. Natl. Acad. Sci. 87:4120-4124; Galli et al. (1990) Lancet 1:1544-1547) (hereinafter "aPL imrnunogen").
  • ⁇ 2-GPI is a minor plasma glycoprotein found free and in association with lipoprotein lipids where it is also known as apolipoprotein H
  • apo H consists of five independently folding domains referred to as Sushi or short consensus repeat domains that resemble similar domains in other proteins.
  • ⁇ 2-GPI has been reported to undergo antigenic and conformational changes upon binding phospholipid (Wagenkneckt et al. (1991) Thromb. Haemostas. 69:361-365; Jones et al. (1992) Proc. 5th Intl. Symp. Antiphospholipid Antibodies
  • the fifth domain contains the putative sites of lipid binding and aPL antibody binding (Hunt J. and S. Krilis, (1994) J. Immunol. 152:653-659; Lauer et al. (1993) Immunol. 80:22-28).
  • the pathological mechanism for aPL is unknown (McNeil et al., supra). Most explanations invoke endothelial cell function or platelet involvement (Haselaar et al. (1990) Thromb. Haemostas. 63:169-173).
  • aPL antibodies may be directly prothrombotic by reducing prostacyclin formation (Vermylen, J. and J. Amout (1992) J. Clin. Lab. Med 120:10-12); by direct interference with the action of coagulation proteins; or by blocking the ability of ⁇ 2-GPI to inhibit the intrinsic blood coagulation pathway, platelet prothrombinase activity, and ADP-mediated platelet aggregation (Arêt et al. (1993) Thromb. Haemostas. 60:336-341).
  • phage libraries are prepared by incorporating randomized oligonucleotide sequences into the phage genome, usually the pill gene, which encode unique peptide sequences on the surface of each phage. Following sequential rounds of affinity purification and amplification, those phage that bind antibody are propagated in E.
  • B cell tolerance entails administering such peptides conjugated to multivalent, stable, non-immunogenic valency platforms in order to abrogate antibody production via B cell anergy or clonal deletion after cross-linking surfare immunoglobulin.
  • This invention resides in the discovery of a method for identifying analogs of key epitopes recognized by aPL antibodies in patients suffering from PAPS, APS and other aPL antibody-mediated diseases, such as recurrent stroke and recurrent fetal loss, using random peptide phage libraries.
  • one aspect of the invention is an improved method for screening random peptide phage libraries in order to identify the peptide sequences which best mimic the epitopes recognized by aPL antibodies.
  • This method comprises the steps of. (a) biopanning the library using methods modified from those known in the art; (b) eliminating very weakly-binding phage by rnicropanning the phage screened from step (a) by (i) incubating the phage in microplate wells coated with aPL antibody bound to Protein G, (ii) washing the microplate wells to remove unbound phage, (iii) eluting the bound phage, and (iv) infecting a microorganism such as E.
  • the invention encompasses a method for identifying analogs of epitopes which specifically bind aPL antibodies isolated from humans suffering from an aPL antibody-mediated disease comprising: (a) preparing phage random peptide libraries; (b) screening said libraries with aPL antibodies to identify aPL mimetic epitopes, wherein said screening comprises (i) screening said libraries by biopanning; (ii) further screening phage isolated by biopanning in (i) by rnicropanning; and (iii) identifying phage containing aPL antibody high-affinity binding peptides recovered in (ii) by immunoassay.
  • the invention also encompasses a method of biopanning phage random peptide libraries to identify and isolate peptides which bind to aPL antibody comprising: (a) reacting affirm ty-purified aPL antibody with phage bearing random peptide inserts; (b) recovering phage bearing random peptide inserts which bind to the aPL antibody; (c) infecting a microorganism with phage recovered in (b); and (d) culturing the infected microorganism in an antibiotic- containing medium in order to isolate the phage.
  • the invention further encompasses a method of rnicropanning phage random peptide libraries to identify and isolate peptides having a high binding affinity to aPL antibodies comprising: (a) isolating phage bearing random peptide inserts by biopanning; (b) incubating the phage recovered in step (a) in microplate wells coated with aPL antibody bound to Protein G; (c) washing the microplate wells to remove unbound phage; (d) eluting bound phage; and (e) infecting a microorganism with phage recovered in (d); and (f) culturing the infected microorganism in an antibiotic-containing medium in order to isolate the phage.
  • the invention also encompasses the above method described wherein the immunoassay is a phage-capture ELISA comprising: (a) incubating phage bearing random peptide inserts isolated by rnicropanning in the microplate wells coated with aPL antibody; (b) washing away unbound phage;(c) incubating an enzyme- labeled anti-phage antibody to the wells; (d) washing away unbound enzyme- labeled anti-phage antibody; (e) adding a calorimetric substrate; and (f) measuring the absorbance of the substrate to identify lilgt-t affinity-binding pliage.
  • the immunoassay is a phage-capture ELISA comprising: (a) incubating phage bearing random peptide inserts isolated by rnicropanning in the microplate wells coated with aPL antibody; (b) washing away unbound phage;(c) incubating an enzyme- labeled anti-phage antibody to the wells; (d
  • Also encompassed by the invention is the method described above and further comprising performing an additional phage-capture ELISA assay of the high affinitybinding phage comprising: (a) coating a uniform amount of the phage on microplate wells; (b) incubating aPL antibody in the wells; (c) washing away unbound antibody; (e) incubating an enzyme-labeled anti-aPL antibody with the bound aPL antibody; (f) washing away unbound enzyme-labeled anti-aPL antibody; (g) adding a calorimetric substrate to the wells; and (h) measuring the absorbance of the substrate to measure the relative binding affinity of the phage.
  • the invention also encompasses the method described above wherein the immunoassay is a colony-blot immunoassay comprising: (a) culturing a microorganism infected with phage bearing random peptide inserts on a nitrocellulose membrane atop an agar-containing culture medium; (b) replicate transferring the microorganism cultured in (a) by blotting the microorganism on a second nitrocellulose membrane atop an agarcontaining culture medium;
  • a method for assaying and ranking, for affinity-binding characteristics, epitopes which specifically bind aPL antibodies isolated from humans suffering from an aPL antibody-mediated disease comprising: (a) coating wells of a microtitration plate with cardiolipin; (b) adding adult bovine or human serum as a source of ⁇ 2-GPI to bind to the cardiolipin and to prevent non-specific binding to the wells of the plate; (c) incubating a solution of monomeric analog and a high-titered aPL antibody for a pre-determined time;
  • Another aspect of the invention encompasses a fluorescence polarization peptide binding assay for determining the dissociation constants for peptides that bind to aPL antibodies. This assay detects direct binding of peptides to aPL antibodies.
  • the invention also encompasses a diagnostic immunoassay, for determining the presence of aPL antibody in body fluids taken from subjects suspected of suffering from an aPL antibody-mediated disease comprising contacting a sample of a body fluid with an analog of an epitope which specifically binds aPL antibodies and determining by methods well known in the art whether aPL antibodies are present in the body fluid and, if present, quantitating the amount of aPL antibodies present in the fluid.
  • One such immunoassay comprises: (a) coating wells of a microtitration plate with an analog of an epitope which specifically binds aPL antibodies; (b) washing the wells to wash away unbound analog; (cladding a test sample of a body fluid to the wells and incubating for a pre-determined time; (d) washing the wells to remove unbound test sample; (e) adding anti-human IgG conjugated with a label to the wells of the plate and incubating for a predetermined time; (f) washing the wells to wash away unbound anti-human IgG conjugate; (g) adding a substrate for the labeled conjugate and developing the substrate/label reaction for a pre-determined time; (h) measuring the end-product of the substrate/label reaction to determine the presence of anti-aPL antibody in the test sample.
  • a diagnostic immunoassay as described above wherein the immunoassay is quantitative is also encompassed.
  • the phage-ELISA assay consists of (i) coating a uniform amount of different clones on wells of a microtitration plate followed by (ii) identifying the peptide inserts which most strongly bind aPL antibody by adding antibody to the wells and developing the reaction with an enzyme-labeled anti-human IgG conjugate.
  • the random peptides displayed by the phage which have a high binding affinity to aPL antibody as measured by phage-ELISA, colony blot or phage-capture-ELISA represent the analogs of the aPL-specific epitope. These peptides are then synthesized and ranked for strength of binding using competition assays.
  • Another aspect of the invention is aPL antibody-binding analogs that bind specifically to B cells to which an aPL epitope binds. Optimized analogs lack T cell epitope(s).
  • a composition for inducing specific B cell tolerance to an aPL immunogen comprising a conjugate of a nonimmunogenic valency platform molecule and an aPL antibody-binding analog that (a) binds specifically to B cells to which an aPL inlinunogen binds and (b) lacks T cell epitope(s).
  • Figure 2 shows that resin-bound analog 5AI2 immunospecifically binds to affinity-purified IgG designated ACA-6501.
  • Figure 3 illustrates that the aPL antibody-binding analogs derived from screening with ACA-6626 bound aPL antisera but did not bind normal sera.
  • Figure 4 also illustrates that the ACA-6501/5A12 analog immunospecifically binds ACA-6501 antiserum and is crossreactive with ACA- 6626.
  • Figures 4 and 5 illustrate that while the ACA-6501/5A12 and ACA-
  • 6626/4D3 aPL antibody-binding analogs derived from screening with methods within the instant invention bind preferentially with the screening antibody, a significant degree of crossreactivity was detected.
  • Figure 6 illustrates method for calculating the GPL value for ACA-6501 aPL antibody. .
  • Figure 7 shows the activity of affinity-isolated ACA-6501 compared to GPL standard sera.
  • Figure 8 illustrates the dramatic drop in sequence diversity of the isolated clones by the fourth round of biopanning.
  • Figure 9 illustrates that three clones (3AI2, 3B3 and 3A5) exhibited a very strong immunospecific signal in the phage-capture ELISA using ACA-6635 whereas all clones tested were unreactive with normal IgG.
  • Figure 10 shows the strong signal exhibited by seven clones in a phage- ELISA using ACA-65 0 1.
  • Figure 11 shows the results of a competitive-binding ELISA obtained with peptides 5A12, CB2 and 3B1O using ACA-6501.
  • 0.16 ⁇ g of Peptide 5AI2 produced 50% inhibition of binding of ACA-6501 aPL antibody to tetravalent peptide ACA6501/3B10 bound to polystyrene microplate wells, whereas 0.08 ⁇ g of CB2 and 0.54 ⁇ g of 3B 10 were required to produce 50% inhibition.
  • Figure 12 illustrates the comparative activity of modified ACA-6641/3G3 analogs.
  • Figure 13 illustrates that the 50% inhibition values for peptides 139, 142 and 143 in a competitive-binding ELISA usi ng ACA-6501 aPL antibody were 6.9, 0.7 and 0.9 ⁇ g, respectively.
  • Figure 14 illustrates the effects of substituting -Me-Pro at positions 3, 9 and both 3 and 9 in peptide 3B10. Substitution of ⁇ -Me-Pro at position 9 increased activity of the peptide four-fold compared to the "native" peptide.
  • Figure 15 shows that peptide 6641/3G3 (LJP 688) is highly cross-reactive with nine affmity-purified ACA antibodies.
  • Figures 16 and 17 show the dose-dependent reduction in anti-685 antibody ABC at 108M using spleen cells from mice immunized with LJP 685-KLH after the spleen cells were incubated with 100, 20 and 4 ⁇ M of LJP 685-MTU-D ABA- PEG conjugate (compound 36 and LJP 685-ATU-MTU-AHAB-TEG conjugate (compound 35), respectively, for 2 hours.
  • Figure 18 displays the NMR structure closest to the centroid of the nine structures elucidated for peptide 925 and is a reasonable representation of the shape of the peptide 925 molecule.
  • Figure 19 compares the structure of peptide 925 (labeled at the bottom of the figure as 3G3) with the structure of peptide 5A12. Both peptides have turns at approximately the same positions in the peptide sequence.
  • Figures 20A and 20B illustrate that the pharmacophore of aPL analogs has been tentatively identified as a small hydrophobic group and a positively charged group.
  • the gem-dimethyl and amino groups of peptide 925 are tentatively identified as the pharmacophore of this peptide as shown in Figure 20A.
  • the lengths of the hydrocarbon linkers that tether the phan-nacophore groups to some scaffold are specified in Figure 20A as well as the distances separating the points at which these linkers are attached to the scaffold.
  • Figure 21 illustrates inhibition of ⁇ 2GPI by 6501 -derived peptides with diluted 6501 serum.
  • Figure 22 shows the ACA-6701 titration of CB2*-F; FITCGPCILLARDRCG.
  • Figure 23 shows the ACA-6501 titration of CB2*-F:
  • Figure 24 shows complete ACA-6501 titration of CB2*-F: FITCGPCILLARDRCG.
  • Figure 25 shows the displacement of CB2*-F from ACA-6701 using 1.04 equivalents of CB2*.
  • Figure 26 shows cFP titration using CB2* to displace CB2*-F from ACA6701.
  • Figure 27 shows cFP titration using 3B10 to displace CB2* -F from ACA6701.
  • Figure 28 shows the dose response of the (LJP685)4/MTU-AHAB-TEG conjugate for tolerance activity.
  • Figure 29 shows the dose response of the (LJP685)4/MTU-D ABA-TEG conjugate for tolerance activity.
  • Figure 30 shows the dose response of tolerance activity of the (LJP685)4/MTU-DABA-TEG conjugate tested in the in vitro model.
  • Figure 31 shows the dose response of tolerance activity of the (LJP685)4/MTU-DABA-TEG conjugate tested in the in vitro model.
  • Figure 32 shows the tolerizing effect of the (LJP685)4/MTU-AHAB-TEG conjugate comparing various administrative routes and dosage ranges.
  • aPL antibody means any antibody which specifically binds ⁇ 2-GPI that mediates disease.
  • B cell anergy intends unresponsiveness of those
  • B cells requiring T cell help to produce and secrete antibody includes, without limitation, clonal deletion of immature and or mature B cells and/or the inability of B cells to produce antibody.
  • Unresponsiveness means a therapeutically effective reduction in the humoral response to an immunogen. Quantitatively the reduction (as measured by reduction in antibody production) is at least 50%, preferably at least 75%, and most preferably 100%.
  • Antibody means those antibodies which are T cell dependent.
  • immunogen means an entity that elicits a humoral immune response comprising aPL antibodies. Immunogens have both
  • aPL immunogens that are involved in aPL antibody-mediated pathologies may be external (foreign to the individual) immunogens such as drugs, including native biological substances foreign to the individual such as therapeutic proteins, peptides and antibodies, and the like or self-immunogens (autoimmunogens) such as those associated with antibody- mediated hypercoagulability (stroke).
  • drugs including native biological substances foreign to the individual such as therapeutic proteins, peptides and antibodies, and the like or self-immunogens (autoimmunogens) such as those associated with antibody- mediated hypercoagulability (stroke).
  • autoimmunogens self-immunogens
  • analog of an immunogen intends a molecule that (a) binds specifically to an antibody to wl-iich the immunogen binds specifically and (b) lacks T cell epitopes.
  • the analog will normally be a fragment or derivative of the immunogen and thus be of the same chemical class as the immunogen (e.g., the immunogen is a polypeptide and the analog is a polypeptide), chemical similarity is not essential. Accordingly, the analog may be of a different chemical class than the inununogen (e.g., the immunogen is a carbohydrate and the analog is a polypeptide) as long as it has the functional characteristics (a) and (b) above.
  • the analog may be a peptide, carbohydrate, lipid, lipopolysaccharide, nucleic acid or other biochemical entity. Further, the chemical structure of neither the inununogen nor the analog need be defined for the purposes of this invention.
  • the term "analog" of an immunogen also encompasses the term
  • mimotope intends a molecule which competitively inhibits the antibody from binding the immunogen. Because it specifically binds the antibody, the mimotope is considered to mimic the antigenic determinants of the immunogen.
  • valency platform molecule means a nonimmunogenic molecule containing sites which facilitate the attachment of a discreet number of analogs of immunogens.
  • Nonimmunogenic is used to describe the valency platform molecule and means that the valency platform molecule elicits substantially no immune response when it is administered by itself to an individual.
  • mammals denotes a member of the mammalian species and includes humans, primates, mice and domestic animals such as cattle and sheep, sports animals such as horses, and pets such as dogs and cats.
  • pharmacophore means the three dimensional orientation and chemical properties of key groups involved in binding of an aPL analog to the antibody target.
  • aPL antibody-binding analogs may be identified by screening candidate molecules to determine whether or not they (a) bind specifically to aPL antibodies and (b) lack T cell epitopes. Specific binding to aPL antibodies may be determined using conventional immunoassays such as the ELISA assays described in the examples below and the presence or absence of T cell epitopes may be determined by conventional T cell activation assays also described in the examples. In this regard, an analog which "binds specifically" to serum
  • T cell epitopes 7 1 antibodies to the immunogen exhibits a reasonable affinity thereto, e.g., 10 M " .
  • the presence or absence of T cell epitopes may be determined using a tritiated thymidine incorporation assay disclosed in Serial No. 08/118,055.
  • the presence of T cell epitopes can also be determined by measuring secretion of T cell-derived lymphokines by methods well known in the art. Analogs that fail to induce statistically significant incorporation of thymidine above background are deemed to lack T cell epitopes. It will be appreciated that the quantitative amount of thymidine incorporation may vary with the immunogen. Typically a stimulation index below about 2-3, more usually about 1-2, is indicative of a lack of T cell epitopes.
  • aPL antibody-binding analogs are coupled to a nonimmunogemc valency platform molecule to prepare the conjugates of the invention.
  • Preferred valency platform molecules are biologically stabilized, i. e. , they exhibit an in vivo excretion half-life often of hours to days to months to confer therapeutic efficacy, and are preferably composed of a synthetic single chain of defined composition. They -will normally have a molecular weight in the range of about 200 to about 200,000, usually about 200 to about 20,000.
  • Examples of valency platform molecules within the present invention are polymers such as polyethylene glycol (PEG), poly-D-lysine, polyvinyl alcohol and polyvinylpyrrollidone. Preferred polymers are based on polyethylene glycols (PEGS) having a molecular weight of about 200 to about 8,000.
  • valency platform molecules suitable for use within the present invention are the chemically-defined, non-polymeric valency platform molecules disclosed in co-owned, co-pending U.S. patent application Serial No. 08/152,506, filed November 15, 1993, which is incorporated by reference herein in its entirety.
  • Particularly preferred homogeneous chemically-defined valency platform molecules suitable for use within the present invention are derivatized 2,2'-ethylenedioxydiethylarnine (EDDA) and triethylene glycol (TEG).
  • Suitable valency platform molecules include tetraannnobenzene, heptaarninobetacyclodextrin, tetraaminopentaerytbritol, 1,4,8,11 tetraazacyclotetradecane (Cyclam) and 1,4,7,10-tetraazacyclododecane (Cyclen).
  • Conjugation of the aPL antibody-binding analog to the valency platform molecule may be effected in any number of ways, typically involving one or more crossliriking agents and functional groups on the analog and valency platform molecule.
  • Polypeptide analogs will contain amino acid sid. - ' ..ain moieties containing functional groups such as amino, carboxyl, or sulfhydryl groups that will serve as sites for coupling the analog to the carrier. Residues that have such functional groups may be added to the analog if the analog does not already contain these groups. Such residues may be incorporated by solid phase synthesis techniques or recombinant techniques, both of which are well known in the peptide synthesis arts. In the case of carbohydrate or lipid analogs, functional amino and sulfhydryl groups may be incorporated therein by conventional chemistry.
  • primary amino groups may be incorporated by reaction with ethylenediamine in the presence of sodium cyanoborohydride and sulffiydryls may be introduced by reaction of cysteamine dihydrochloride followed by reduction with a standard disulfide reducing agent.
  • the valency platform molecule may also be derivatized to contain functional groups if it does not already possess appropriate functional groups.
  • Linkers of variable lengths are useful for connecting peptides or other bioactive molecules to valency platform molecules.
  • Suitable linkers include linear oligomers or polymers of ethylene glycol.
  • linkers are useful in connecting a molecule containing a thiol reactive group such as haloaceyl, maleiamide, etc., via a thioether to a second molecule which contains an amino group via an amide bond.
  • linkers are flexible with regard to the order of attachment, i.e., the thioether can be formed first or last.
  • the conjugates will normally be formulated for administration by injection (e.g., intraperitoneally, intravenously, subcutaneously, intramuscularly, etc.). Accordingly, they will typically combined with pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like.
  • the conjugate will normally constitute about 0.01% to 10% by weight of the formulation.
  • the conjugate is administered to an individual in a "therapeutically effective amount", i.e., an amount sufficient to produce B cell anergy to the involved immunogen and effect prophylaxis, improvement or elimination of the antibody-mediated condition being addressed.
  • the particular dosage regimen i. e. , dose, timing and repetition, will depend on the particular individual and that individual's medical history.
  • a dose of about 1 ⁇ g to about 100 mg conjugate/kg body weight, preferably about 100 ⁇ g to about 10 mg/kg body weight, will be given weekly.
  • Other appropriate dosing schedules would be as frequent as daily or 3 doses per week, or one dose per week, or one dose every two to four weeks, or one dose on a monthly or less frequent schedule depending on the individual or the disease state.
  • Repetitive administrations normally timed according to B cell turnover rates, may be required to achieve and/or maintain a state of humoral anergy. Such repetitive administrations will typically involve treatments of about 1 ⁇ g to about 10 mg/kg body weight or higher every 3 0 to 60 days, or sooner, if an increase in antibody titer is detected.
  • sustained continuous release formulations of the conjugates may be indicated for some pathologies.
  • Various formulations and devices for achieving sustained release are known in the art.
  • Anti-helper T cell treatments may be administered together with the conjugates. Such treatments usually employ agents that suppress T cells such as steroids or cyclosporin.
  • NMR nuclear magnetic resonance
  • multilamellar, cardiolipin- containing dispersions liposomes; also containing cholesterol and dicetylphosphate
  • aPL plasma or serum
  • liposomes are pelleted from the serum by centrifugation.
  • the liposome mixture is disrupted by 2% octylglucoside detergent and applied to a protein A-agarose colurrm.
  • IgG aPL antibody is eluted from protein A with mild acid, neutralized, bufferexchanged, and tested in the ACA ELISA.
  • FTJSE 5 filamentous phage random peptide libraries on the p-III protein (five copies of p-III with peptide per phage) are constructed. These libraries provide a vast array of shapes and structures for the discovery of mimetic epitopes.
  • Four libraries, designated “x” libraries have peptide inserts that are 8, 10, 12, and 15 residues in length, respectively, and are flanked by proline residues on both the amino and carboxyl ends. The purpose of these proline residues is to disrupt any contribution to secondary structure that might arise from the native P-III protein and to project the insert into the solvent.
  • the "y' " libraries contain eysteine-bounded inserts that are 6, 7, 8, 9, 1 1, and 13 amino acids long.
  • the “y” library is the same as the “y”' library except that it lacks the 6 and 8 amino acid inserts. These peptide inserts for both “y” and “y”' libraries are flanked by cysteine residues at both the amino and carboxyl ends to form cyclic, more rigid structures. Proline residues are incorporated outside these cysteine residues for reasons similar to those for the "x” libraries above.
  • the " x" is the same as the "y”' library except that it lacks the 6 and 8 amino acid inserts.
  • “y”', and “y” libraries are located five residues from the amino terminus of the native p-III protein.
  • the "z” library consists of random eight amino acid inserts located at the amino terminus of the p-III protein and do not contain any flanking proline or cysteine residues.
  • a combination of the "x,” “y”' and “z” libraries represents eleven different libraries each with approximately one hundred million different peptide inserts.
  • libraries are constructed by incorporation of random oligonucleotide sequences of the length appropriate to give the desired length insert into the p-III gene of fUSE 5 using standard molecular biology techniques. Following restriction endonuclease digestion of the fUSE 5 DNA, an excess of kinased oligonucleotides provided as gapped duplexes is added and ligated. The DNA is then electroporated into E. coli and inserts are selected by culturing in tetracycline-containing media. The phage from this culture (which contain the peptide insert) are isolated from the supernatant, washed and resuspended in buffer. Typically libraries are shown to have 7x10 independent clones at 8x10 transducing units per mL.
  • Biopanning describes the technique wherein affinity-purified aPL antibody and phage bearing random peptide inserts are allowed to mix, following which antibody-specific recovery captures the bound phage.
  • the phage confer tetracycline resistance to E. coli that are propagated in a tetracycline-containing medium and then isolated. Multiple rounds of biopanning enrich the number of immunospecific phage in a sample. Phage are always recovered at the end of three to five rounds of selection but may represent only sequences that are
  • Micropanning is carried out following three or more rounds of biopanning and uses the same antibody as employed in the biopanning method. The method consists of dilution of the phage from the last round of biopanning and analyzing fifty or more of these clones by micropanning. Micropanning is accomplished by growing each clone to a similar density and then incubating dilute phage at an optimal single concentration in microtitration wells previously coated with a constant amount of antibody.
  • the optimal single concentration of phage is that concentration most likely to reveal the widest range of micropanning scores (from 0 to 4+) and, thus, permit the greatest discrimination among the clones being tested. It is based on the 'micropannmg behavior of six randomly selected clones where the score is determined at each of several concentrations of phage obtained by serial dilution. Following the incubation with antibody, the unbound phage are washed away and the amount of bound phage is used as an indication of the affinity of the phage insert for the antibody. The amount of bound phage is determined by elution with mild acid followed by neutralization and infection of E. coli. The number of infected E. coli are then quantitated by plating the microorganisms on agar plates containing tetracycline and then determining colony densities achieved by each clone.
  • Phage-Capture ⁇ LISA The phage-capture ⁇ LISA test was developed to provide an intermediate level assay to bridge the gap between the relatively low stringency of the micropanning assay and the high stringency of the phage- or peptide- ⁇ LISA assays. Preliminary studies show that some antibody preparations give too many positive clones by micropanning but none by phage- ⁇ LISA or peptide- ⁇ LISA. The limitation of the phage-ELISA described below is that only five copies of p-III are located on each phage and even with a large number of phage coated on a well, few copies of the insert are represented and detection requires that the antibody have a very high affinity for the insert. With the phage-capture ELISA, the signal is amplified many times which facilitates the detection of lower affinity, stable interactions between the antibody and the insert.
  • the phage-capture ELISA consists of the following steps. Microtitration wells are coated with aPL antibody and phage clones are added as in the micropanning assay. Unbound phage are washed away and the amount of bound phage is quantitated using an enzyme-conjugated goat antiserum which binds the major coat protein of the phage. Phage screened using phage-capture ELISA react with many aPL antibodies and provide a strong signal in subsequent ELISA assays. This intermediate level of sensitivity allows for greater efficiency in the peptide synthesis effort since few micropanning-positive phage are phage-capture ELISA positive. As a result, peptides synthesized from positive phage-capture
  • This assay allows large-scale colony screening of E. coli infected by biopanned phage.
  • This procedure is an alternative to phage-ELISA for identifying immunoreactive clones and exhibits a comparable level of sensitivity without requiring culturing of individual phage clones prior to testing.
  • E. coli infected with phage from a round of biopanning are spread on a large diameter nitrocellulose (NC) membrane and cultured overnight on the surface of an agar plate containing tetracycline (Barbas et al. (1991) Proc. Natl. Acad. Sci. USA 88:7978-7982). Each colony results from infection by phage containing identical sequences.
  • NC nitrocellulose
  • phage may be probed by the techniques commonly used in Western blotting, i.e., staining or iitm-iunobtotting. Blots that have been blocked may be incubated with the screening aPL antibody. Following washes to remove unbound antibody, an anti-human IgG horseradish peroxidase conjugate is added to bind to any aPL- antibody that is bound to phage. The addition of a calorimetric substrate allows one to localize the discrete colonies in the master plate which represent immunospecific phage that may be cloned for further study. (vi) Peptide-ELISA
  • the peptides are made using standard Fmoc peptide chemistry as is well known in the art.
  • the peptides can be made, for example, as branched tetravalent molecules, / ' . e., each molecule has four copies of the insert. Such a molecule can coat the well of a microtitration plate and still have epitopes exposed to the solution to allow binding by an antibody.
  • the tetravalent peptides are synthesized by incorporating lysines as branch points at the first two couplings analogous to the methods used for Multiple Antigenic Peptides (MAPS) (Posnett et al. (1988) J. Biol. Chem. 263:1719-1725).
  • a spacer consisting of glycine- serine-glycine-serine is added on each arm after the lysines and then the insert, including the framework amino acids found in the phage, proline-glycine at the carboxyl ten-ninus and alanine-glycineproline at the amino terminus. All amino acids in this synthesis are added one at a time using standard Fmoc methods.
  • SUBST ⁇ TUTE SHEET (RULE 26) These peptides are then assayed by ELISA which is carried out by coating the peptides on microtitration wells and then assaying their reactivity with aPL antibody in a standard ELISA format. In practice, the peptides usually bind very strongly to the original screening antibody and show some cross-reactivity with other aPL antibodies. Controls of non-aPL antibodies are included to eliminate nonspecific binding peptides.
  • ELISA-positive peptides are identified, it is necessary to quantitate their relative binding affinity to the aPL antibodies and to determine whether or not two peptides bind the same population of antibodies in a given patient serum via a peptide-competition ELISA assay.
  • various monomeric peptides compete with tetravalent peptides coated on a microtitration plate well.
  • the peptides to be evaluated are synthesized as monomers, i.e., without the lysine branches employed in the synthesis of the tetravalent peptides, using standard Fmoc chemistry. The monomeric peptides are then purified and dissolved at known concentrations.
  • Wells of a microtitration plate are coated with a tetravalent peptide known to bind to the aPL antibody.
  • Serial dilutions of the monomeric peptides are incubated with a constant dilution of the aPL antibody.
  • the dilution of the aPL antibody was previously determined by titering the antibody against the tetravalent peptide and selecting a dilution on the downslope of the titration curve.
  • the antibody/peptide solutions are added to the microtitration wells and a standard calorimetric ELISA is performed.
  • the concentration of each monomeric peptide that decreases binding of the aPL antibody with the tetravalent peptide is determined by plotting the calorimetric readings obtained for each well. The 50% inhibition point is used as the measure of the relative strength of binding for the monomeric peptides.
  • a variation of this assay uses microtitration plates coated with human ⁇ 2 -glycoprotein I/cardiolipin ( ⁇ 2 .GPI/CL) instead of tetravalent peptide and tests the ability of monomeric peptides to block the binding of aPL antibody to the epitope(s) on ⁇ 2 _GPI/CL.
  • IgG-depleted human serum at an optimized concentration is used as a source of ⁇ 2- GPI.
  • the monomeric peptides at several concentrations are incubated with an optimized concentration of aPL antibody in a manner analogous to the assay which employs tetravalent peptide as a plate substrate. Following the incubation of aPL/peptide in ( ⁇ 2 .GPI/CL) plates, antibody binding and the peptide concentration required for 50% inhibition is determined at half-maxim-al absorbance as in the tetravalent assay.
  • An additional variation of this assay tests the ability of monomeric peptides to block the binding of aPL antibody to ⁇ 2- GPI coated directly on the wells of Nunc Maxisorp microtitration plates.
  • the use of cardiolipin is omitted and instead of fish gelatin, the reagent diluent and blocker used is nonfat milk/Tween.
  • (viii) Fluorescence Polarization Peptide Binding Assay This assay detects direct binding of the peptide to aPL antibody. Since aPL antibodies bind to ⁇ 2 .GPI (the antigen), the ELISA competitive inhibition assay can show inhibition due to binding to ⁇ 2 -GPI as well inhibition due to binding of the peptide to the aPL antibody. Because binding to antibodies is required in order for the peptide to function as a Toleragen, it is essential to establish that a peptide can directly bind to an aPL antibody. This assay is used to determine the dissociation constants for peptides that bind to two aPL antibodies, ACA-6501 and ACA-6701.
  • the desired epitope for tolerance induction should have as strong an interaction with as many of the aPL antibodies as possible but not contain any unnecessary residues.
  • analogs of each peptide are made (i) that lack given residues, for example, the framework residues at the carboxyl and/or amino termini are deleted, or (ii) in which amino acid substitutions have been made which differ from sequences found in the epitope library screen.
  • These amino acid substitutions may be either natural, e.g. , isoleucine for leucine, or unnatural, e.g. , alpha methyl proline for proline.
  • the effect of these deletions and/or substitutions are then measured via peptide-competition ELISA.
  • a Tolerogen For a Tolerogen to be generally effective, it must bind a major portion of the aPL antibodies in the majority of patients. It is important to determine if several antibodies from different patients bind identical residues within the eighty- four amino acid 5th domain of ⁇ 2 -GPI which has been suggested by others to contain the target epitope. If several antibodies bind identical residues, a single mimotope derived from the structural data of the peptides can be constructed which will react with all the antibodies. On the other hand, if the antibodies bind to different residues, a unique tolerogen would be required for each antibody.
  • Antibody ACA-6501 from a patient with a GPL score of 151 (high titer) and a historyof recurrent stroke, fetal loss, lupus and three aortic valve replacements was immunoaffinity purified on dardiolipin liposomes.
  • the antibody was used in four separate phage library screens, using the xy, the xyz, the xy'z, and a special pro/cysbounded 7-mer library where, based on the previous screens, Arg was fixed at the seventh position.
  • Table 1 36 sequences were obtained in phage that micropanned (out of 140 tested). These appear quite homologous and the conserved DR residues at positions 6 and 7 are notably striking.
  • the consensus sequence is CLLLAPDRC. Despite this homology, only seven phage (see Table 2) were positive after phage-ELISA calorimetric testing. Screening the xy'z phage with affinity purified ACA-6626 (from a patient with a high, but lower than ACA-6501, titer) yielded five unique sequences that were phage-ELISA tested. None were coi ⁇ r positive but two were positive when the ELISA immunoconjugate was developed with a chemiluminescent substrate. The sequence motif associated with ACA-6626 appears related but is different from that seen with ACA-6501 (see Table 3). Both antibodies preferred the cys-bounded (probably cyclized and constrained) epitopes over the open pro-bounded sequences.
  • ACA-6644 is another high-titered aPL antibody that was used to screen thepooled P-III phage libraries according to methods described herein. The following sequences were discovered:
  • ACA-66644/CBf GILALDYV These sequences all were derived from the component "z" epitope library that lacks phage framework residues at the N-terminus. When synthesized as peptides the sequences were im.munoreactive with several ACA sera including ACA-6644 and ACA-6501. Analysis revealed unsuspected homologies with the sequences previously obtained with ACA-6501 as illustrated in Table 4.
  • sequence homology from two very dissimilar source libraries screened by these two aPL antibodies suggests that the sequences may mimic a major, perhaps immunodominant, region in the native target antigen.
  • the sequences were strongly immunoreactive with the parent serum (ACA-670 1) but were minimally cross-reactive with other aPL antibodies.
  • ACA-6641/3G3 AGP CLGVLGKLC PG ACA-6501/3BIO: AGP CLLLAPDRC PG ACA-6626/4C7: AGPDNIADPRC PG ACA-6701/3B1: AGP LSDPGYVRNIFH PG ACA-6644/CBf: GILALDYV GG Subsequent testing determined that this peptide, ACA-6641/3G3 having the sequence AGP-CLGVLGKLC-PG (LJP 688) was cross-reactive with a number of ACA antisera. Chemical optimization of this peptide was pursued by truncation, systematic amino acid substitution, and non-disulfide cyclization studies.
  • Phage sequences obtained with affinity-purified ACA-6501 and found to be phage-ELISA-posit ive were synthesized on a solid support recently developed for combinatorial synthetic peptide libraries.
  • This support a Rapp resin, has a high peptide density and uses a hydrophilic polyethylene glycol spacer before the first amino acid is coupled.
  • the synthesis resulted in resin-bound peptide that was ideally suited for antibody binding studies.
  • peptide 5AI2 sequence CLILAPDRC
  • Similar results were obtained with the other phageELISA-positive peptides tested.
  • resin peptide-bound affinity-purified ACA-6501 aPL antibody was detected by an immunoconjugate color reaction.
  • 6641/3G3 was tested against several high titer ACA antisera. This peptide appeared to be crossreactive with the vast majority of ACA antisera tested as illustrated in Figure 14. 6641/3G3 demonstrated dose-dependent cross-reactivity with 10 of 10 ACA antibodies with complete inhibition (>80%) at 1.8 mg/mL and appeared to be specific for ACA antibodies as demonstrated in Table 7 below.
  • RFL recurrent fetal loss
  • Peptide synthesis allows the molecular dissection of the mimotope by selective synthesis. This includes the modification of each amino acid along the
  • SUBST ⁇ UTE SHEET (RULE 26) chain with the goal of enhancing antibody binding.
  • Selective synthesis reveals the relative importance of each amino acid in the sequence. If necessary, selective substitution at particular residue locations can be designed to maintain B cell reactivity while abolishing any T cell proliferative reactivity discovered during T cell assays.
  • Peptide 6641/3G3 (LJP 688) was subjected to a number of analyses.
  • the analyses included truncation at both the N-terminus and the C-terminus, disulfide substitution, substitution of alanine and glycine for amino acids in positions 2 through 8, substitution of branched aliphatic amino acids in positions 2, 4, 5 and 8, substitution of amino acids affecting conformation of the peptide, including substitution of a-methyl amino acids, substitution of basic amino acids in position 7, substitution of D-amino acids in positions 2 through 9, and the substitution of N-a-methyl amino acids in positions 1 through 9.
  • Table 9 The structure/activity relationship of these substitutions and truncations is shown in Table 8.
  • Disulfide surrogates c L G V L G K L C 1 10 #952 100
  • Gly scan c G G V L G K L C 310 #952 120 c L G G L G K L C inf c L G V G G K L C inf c L G v L G G L C inf c L G v L G K G C 500 Sequence optim.
  • Branched aliphatic amino acids c 1 G V L G K L C 60 #910 40 c V G V L G K L C 130 c M G V L G K L C c Cy G V L G K L C c tL G V L G K L C c mL G V L G K L C c Iv G V L G K L C c L G L L G K L C
  • Proline residues have a special significance due to their influence on the chain conformation of polypeptides. They often occur in reverse turns on the surface of globular proteins. In the phage epitope libraries of the present invention, all random peptide inserts are flanked by boundary prolines. In addition, most of the mimotopes discovered with ACA-6501 have a third proline which, based on computer-based predictions, likely exists as part of a ⁇ -turn. ⁇ -turn mimetics can be used to enhance the stability of reverse turn confon-
  • SUBST ⁇ U ⁇ E SHEET (RULE 26) nations in small peptides.
  • Such a mimetic is (S - ⁇ -methyl proline ( ⁇ - ePro), a proline analog that, in addition to stabilizing turn conformations, confers resistance to protease degradation.
  • Protease resistance is a desirable property for a potential drug designed to act in the plasma.
  • Peptide ACA-6501/3B10 AGP CLLLAPDRC? G is a consensus peptide. It has a sequence featuring the most prevalent residue at each position based on a comparison with 35 other homologous sequences. Due to its representative character, the sequence was subjected to a number of systematic modifications and deletions and its activity subsequently evaluated by aPL antibody binding.
  • Proline-3 is derived from the phage framework and is not part of the random insert.
  • the most dramatic effect was obtained by the substitution of a-MePro for proline at the 9-position. This substitution led to a six-fold enhancement in immunoreactivity.
  • the results of a-methyl and ⁇ -methyl amino acid substitution studies carried out on peptide 6641/3G3 are shown above in Table 8.
  • alpha amino acids In addition to the twenty naturally-occurring amino acids and their homoanalogs and noranalogs, several other classes of alpha amino acids can be employed in the present invention. Examples of these other classes include d-amino acids, /V-alkyl amino acids, alpha-alkyl amino acids, cyclic amino acids, chirrieric amino acids, and miscellaneous amino acids. These non-natural amino acids have been widely used to modify bioactive polypeptides to enhance resistance to proteolytic degradation and or to impart conformational constraints to improve biological activity (Hruby et al. (1990) Biochem. J 268:249-262; Hruby and Bonner (1995) Methods in Molecular Biology 35:201-240).
  • N ⁇ alkyl amino acids are the iV ⁇ -methyl amino acids, such as N'-methyl cysteine (nC), N ⁇ methyl glycine (nG), .V ' methyl leucine (nL), N ⁇ methyl lysine (nK), and N ⁇ methyl valine (nV).
  • alpha-alkyl amino acids examples include alpha-methyl alanine (niA), alpha-aminoisobutyric acid (aiB), alpha- methyl proline (mP), alpha-methyl leucine (mL), alpha-methyl valine (mV), alpha-methylalpha-aminobutyric acid (tv), diethylglycine (deG), diphenylglycine (dpG), and dicyclohexyl glycine (dcG) (Balararn (1992) Pure & Appl. Chem. 64:1061-1066; Toniolo et al. (1993) Biopolymers 33:1061-1072; Hinds et al.
  • cyclic amino acids include 1 -amino-1-cyclopropane carboxylic acid (cG), 1 -amino- 1-cyclopentane carboxylic acid (Ac5 c), 1 -amino- 1 -cyclohexane carboxylic acid (Ac6c), aminoin.dane carboxylic acid (ind), tetrahydroisoquinoline carboxylic acid (Tic), and pipecolinic acid (Pip) (C. Toniolo (1990) Int'l J. Peptide Protein Res. 35:287-300; Burgess et al. (1995) J. Am. Chem. Soc.
  • chimeric amino acids include penicillamine (Pe), combinations of cysteine with valine, 4R- and 4S- mercaptoprolines (Mpt), combinations of homocysteine and proline and 4R- and 4S-hydroxyprolines (hyP) and a combination of homoserine and proline.
  • miscellaneous alpha amino acids include basic amino acid analogs such as omithine (Or), N f -methyl lysine (mK), 4-pyridyl alanine(pyA), 4- piperidino alanine (piA), and 4-aminophenylalanine; acidic amino acid analogs such as citrulline (Cit), and 3 -hydroxy valine; aromatic amino acid analogs such as 1-naphthylalanine (1- ⁇ al), 2-naphthylalanine (2-Nal), phenylglycine (pG), 3,3- diphenylalanine (dpA), 3-(2-thienyl)alanine (TE).
  • basic amino acid analogs such as omithine (Or), N f -methyl lysine (mK), 4-pyridyl alanine(pyA), 4- piperidino alanine (piA), and 4-aminophenylalanine
  • halophenylalanines e.g., 2- fluorophenylalanine and 4-chlorophenylalanine
  • hydrophobic amino acid analogs such as t-butylglycine (i.e., tertiary leucine (tL)), 2-aminobutyric acid (Abu), cyclohexylalanine (Cy), 4-tetrahydropyranyl alanine (tpA), 3, 3 -dicyclohexyl alanine (dcA), and 3,4-dehydroproline.
  • beta amino acids can also be used in the present invention.
  • these other amino acids include 2- aminobenzoic acid (Abz), ⁇ -aminopropanoic acid ( ⁇ -Apr), ⁇ -aminobutyric acid ( ⁇ -Abu), and 6-aminohexanoic acid ( ⁇ -Ahx).
  • Carboxylic acids such as 4- chlorobutyric acid (By) and 3-chloropropionic acid (Pp) have also been used as the first residue on the N-terminal in the synthesis of cyclic thioether peptides.
  • T'he mimetic peptides identified by the methods of the instant invention can be further modified to contain thioether substitutions. Modification of cyclic disulfide analogs to cyclic thioether analogs will extend the plasma half-life of the analog conjugates and, therefore, require a lower dosage. Cyclic thioether analogs also eliminate the problem of disulfide bond exchange which often occurs with cyclic disulfide polypeptides. In addition, the cyclic thioether analogs may also interfere with MHC Class 1 1 presentation to T cells and, thus, facilitate induction of anergy. Finally, the cyclic thioether analogs are useful in the thiol-dependent conjugation reactions used in the production of valency platform molecule conjugates.
  • cyclic thioether analogs were prepared according to the methodology described in co-owned, co-pending patent application, attorney docket number 252312006600, which is incorporated herein in its entirety. Using either a RinkTM amide 4-methyl benzhydrylamino resin or MBHA resin, fall length peptide analogs of 6641/3 G3 were prepared, converted into chloro-peptides, cleaved from a solid support and then cyclized. See Thioether Reaction Scheme below. Suitable cyclic thioether analogs include the analogs shown below.
  • thioether analogs are prepared according to the reaction scheme below.
  • the aPL standards (APL Diagnostics, Inc., Louisville, KY) were reconstituted according to the manufacturer's instructions and diluted 1 :50 with 10% ABS-PBS.
  • the plate was washed five times with TBS and 100 ⁇ L of 1 : 1 ,000 goat-anti-human-IgG/alkaline phosphatase conjugate (Zymed,
  • the optical density was read at 550 nm in a microplate autoreader (Bio-Tek Instruments, Winooski, VT, Model EL311). The optical density of the odd-numbered control wells (blank, without cardiolipin (CL)) was subtracted from the optical density of the evennumbered wells.
  • the absorbance readings of the aPL standards were plotted using Graph Pad Prizm (Graph Pad Software, Inc., San Diego, CA) to generate the GPL (IgG phospholipid) standard curve.
  • the diluted 6501 test serum absorbance readings were used to calculate GPL scores based on the GPL standard curve.
  • the precipitate was washed 3 times with 25 mL of cold 0.96% NaCl using the RC3 centrifuge.
  • the pellet was dissolved in 1 mL of 2% (wt/vol) solution of n-octyl-p-D-glucopyranoside (Calbiochem, La Jolla, CA) in TBS and applied to a 0.6 mL protein A/cross- linked agarose (Repligen Corporation, Cambridge, MA) column which had been pre- washed with 15 times bed volume of 1 M acetic acid and equilibrated with 15 times bed volumes of TBS.
  • the antibody-protein A/agarose column was washed with 40 times bed volume of 2% octylglucopyranoside to remove lipids, followed by extensive washings with TBS until the o tical density of the eluate at 280 nm approached the baseline.
  • the bound antibody was eluted with 1 M acetic acid.
  • Example 3 Construction of a p-III Library Vector Preparation fUSE 5 (Scott, J.K. and G. Smith, supra) was used as the vector for the construction of p-III libraries, and a variation of the method of Holmes, D.S. and M. Quigley (1981), Anal. Biochem. 144:193) was employed to generate the doublestranded replicative form (RF). Briefly, an 800 ml culture of E. coli K802, harboring fUSE 5, was grown in 2YT medium (Difco Labs, Ann Arbor, NU) containing 20 micrograms/mL tetracycline for 18 hours at 37 degrees with vigorous shaking. Cells were collected by centrifugation and resuspended in 75 mL STET.
  • 2YT medium Difco Labs, Ann Arbor, NU
  • STET consists of 8% sucrose in 50 mM Tris/HCI pH 8.0, 50 mM EDTA and contains 0.5% Triton X-100. Lysozyme, 10 mg/mL in STET. was added to a final concentration of 1 mg/mL. After 5 minutes at RT, three equal aliquots were placed in a boiling water bath with occasional shaking for 3.5 minutes. The viscous slurry was centrifuged for 30 minutes at 18000 x G and an equal volume of isopropanol was added to the supernatant. The solution was cooled to -20°C and the nucleic acids were collected by .centrifugation. The RF was isolated from a CsCl gradient as described by Sambrook et al. MOLECULAR CLONING: A LABORATORY MANUAL (Cold Spring Harbor Laboratory Press,
  • the DNA for insertion was generated by the "gapped duplex" method described by Cwirla et al. (1990) Proc. Natl. Acad. Sci. USA 87:6378-6382.
  • an oligonucleotide containing a degenerate region in the middle is surrounded by short constant regions on each end of the oligonucleotide.
  • Two shorter, complementary oligonucleotides are annealed to form a "gapped duplex" possessing overhangs that are complementary to the sticky ends produced by the restriction endonuclease used to digest the vector.
  • EpiGS2 was designed to base pair to the 5' end of the degenerate oligonucleotide and has the sequence 5' GGGTCCAGCCCCGT 3'.
  • EpiGS3 was designed to anneal to the 3 'end of the degenerate oligonucleotide and has the sequence 5'CAGCCCCCGG 3'. When correctly annealed, the three oligonucleotides form a "gapped duplex" which, when inserted into fUSE 5 digested with Sfil, restores the reading ftme of P-III with the random insert near the 5' end.
  • Oligonucleotides described above were prepared by excision from polyacrylainide gels. One nanomole of EpiGS2, EpiGS3, and 50 picomoles of the degenerate oligonucleotide were kinased separately in 66 microliter volumes. The three oligonucleotides were then pooled, NaCl was added to 50 mM and the mixture was heated to 65 °C for 5 minutes followed by slow cooling to RT. The annealed oligonucleotides were then cooled on ice and used immediately in the Iigation reaction with fUSE 5.
  • the Iigation reaction consisted of 10 micrograms of fUSE 5 DNA digested to completion with Sfil, 30 ⁇ L of the "gapped duplex" solution and 1000 U of T4 ligase in a total volume of 450 microliters. The Iigation was incubated for 18 hours at 16°C. The mixture was then phenol- chloroform extracted, precipitated with ethanol and the precipitate dissolved in 20 ⁇ L of water.
  • the ligated DNA was introduced into E. coli by electroporation (Dower et al. (1988) Nucleic Acid Res. 16:6127-6145). Frozen electrocompetent MC 1061 cells (0.1 mL) were mixed with 4 ⁇ L of ligated DNA in a cold 2 mm cuvette and subjected to 2.5 kV, 5.2 mS pulse by a BTX electroporation device (BTX Corp., San Diego, CA). Immediately after the pulse, 1 mL of SOC, a cell growth medium (see Dower et al, supra) was added. Five separate electroporations were carried out pooled, and incubated at 37°C for 1 hour.
  • BSA bovine serum albumin
  • Protein G-agarose beads were washed 2x in TBS/BSA and 2 times in TBS/0.5% Tween-20 and stored at 4°C as a 50% suspension in TBS/Tween. Two hundred gL of the protein G-agarose bead suspension was then added to the ACA-6501 /phage mixture and incubation continued for an additional 1 hour at RT. At this point, the mixture was chilled and washed 3 times with cold TBS/Tween and the precipitate was collected in a microfuge in a cold room. The washed beads were transferred to new microfuge tubes prewashed with TBS/BSA and TBS/Tween to prevent non-s ecific adherence.
  • the beads with bound ACA-6501 -bearing phage were eluted with 300 ⁇ L 0.2 N HCI/glycine, pH 2.1 by tumbling for 10 minutes at RT. Following centrifugation at 16,000 X g, the acidic eluate supernatant was collected and an additional 100 ⁇ L of elution solution was added to the bead pellet and the procedure repeated. After 10 minutes, the phage-containing eluates
  • the suspension was centrifuged at 12,000 x g for 10 minutes in polycarbonate tubes and the pellet discarded. After heating the supernatant at 70°C for 30 minutes in polypropylene tubes, the material was spun again in polymbonate tubes and the supernatant saved. To the supernatant, 1/4 volume of 20% (w/v) polyethylene glycol, molecular weight 8000 (PEG 8000) was added to precipitate phage. The solution was mixed by inversion 100 times and then incubated at 4°C for 2 hours.
  • PEG 8000 polyethylene glycol, molecular weight 8000
  • the phage-containing pellet was resuspended in -0.5 mL of TBS/BSA and transferred to a 1.4 mL microfuge tube. After a 1 minute spin in a microfuge at 16,000 x g, the supernatant was transferred to a clean tube and labeled first round amplified phage.
  • Immulon type 2 plates were coated with protein G.
  • Protein G was prepared at 10 ⁇ g/mL in 0.1 M NaHCO 3 and 100 ⁇ L per well was added to the wells of microtitration plates and incubated overnight at 4°C. After discarding excess protein G solution from plates, each well was blocked with 250-300 ⁇ L 2YT for 1 hour at RT with agitation on an oscillating platform. Tris-buffered saline, pH 7.4/0.5%) Tween 20 (TBS/Tween), was used with an automatic plate washer to wash the wells 4 times with 200 ⁇ L.
  • affACA-6501 or control normal IgG
  • 2YT was added to washed wells.
  • the plate was transferred to a cold room rotator near the end of a 1 hour incubation at RT on a rotating platform.
  • Phage to be tested by micropanning were obtained from the agar plates generated by biopanning.
  • Each clone to be tested was transferred using sterile toothpicks to a separate well of a round-bottom 96-well microtitration plate (Corning, NY) containing 250 ⁇ L 2 YT/Tet per well and cultured overnight at 37°C.
  • ACA6501/3B10 refers to the clone isolated by ACA-6501 in the third round located in the well designated BIO on the microtitration plate.
  • the incubation of dilute phage with aPL antibody or control IgG was carried out for 2 hours at 4°C on a flat rotator. After 9 washes with TBS/Tween in an automated plate washer, the IgG-bound phage was eluted with 20 ⁇ L of 0.2 N HCIglycine/0.1% BSA, pH 2.2. The elution incubation continued for 10 minutes at RT, during which time a new Corning microtitration plate was prepared containing 20 ⁇ L of freshly starved
  • E. coli per well and kept chilled.
  • One hundred forty ⁇ L of 29 mM Tris was added to the plate containing the phage eluates in order to neutralize the pH, following which 20 ⁇ L of phage suspension was transferred from each well to the corresponding well in the plate containing starved E. coli.
  • 200 ⁇ L 2YT/dil Tet was added and incubation carried out an
  • SUBSTTTUTE SHEET (RULE 26) additional 30 minutes at 37°C.
  • 10 ⁇ L from each well was spotted on a large 2YT/Tet agar plate while retaining the original 8x12 well pattern and orientation from the last microtitration plate. After allowing the spots to dry for 30 minutes, the plate was incubated overnight at 37°C. The following day, colonies were semiquantitatively scored from 0 to 4+, with 0 symbolizing ⁇ 10 colonies; +/-, 10-20; 1+, 20-50; 2+, 50-70% confluent; 3+, 70- 90% confluent; and 4+ representing >90% confluent colonies.
  • Single-stranded viral DNA was isolated from cultures incubated overnight at 37°C.
  • 2YT/Tet either as 2 mL in tubes or 250 ⁇ L in microtitration plate round-bottom wells, was inoculated with individual phage from spread plates of previously grown cultures in microtitration plates.
  • the purification of phage by 20% PEG/2.5 M NaCl precipitation of culture supernatants as well as the isolation or release of virion DNA by phenol- chloroform extraction or by alkali denaturation was performed as described in Smith, G.P. and J.K. Scott, "Libraries of peptides and proteins displayed on filamentous phage" (1993) Meth. Enzymol.
  • Phage-Capture ELISA Clones ACA-6635/3A12, 3B3, 3C8, 3A5, 3C9, and 3B7 were grown as 3 mL cultures.
  • Affinity purified ACA-6635 was diluted to 2.5 ⁇ g/mL in phosphate- buffered saline, pH 7.2, and 100 ⁇ L added to Immulon-2 microtitration plate wells. After 2 hours, the plates were washed 3 times with TBS/Tween in an automated plate washer with no shaking. The plate was then blocked with 150 ⁇ L
  • SUBSTTTUTE SHEET (RULE 26) 0.1% BSA (globulin-free) in PBS per well. After 1 hour at 4°C, the plate was washed 3 times as previously described. After centrifuging each phage culture 3 minutes at 17,000 x g, each supernatant was diluted 1 : 10 in 0.1% BSA/PBS and 100 ⁇ L added to each of the wells coated with affinity purified ACA-6635 and then incubated for 2 hours at 4°C. Plates were then washed with TBS/Tween as before.
  • Horseradish peroxidase-conjugated sheep IgG anti-M13 phage antibody (Pharmacia, Inc., Piscataway, NJ) was diluted 1 :5,000 in 0.1% BSA PBS and 100 ⁇ L applied to each well. Following incubation for 1 hour at 4°C, the plate was washed 4 times as before. One hundred ⁇ L of substrate prepared according to the conjugate manufacturer's instructions was added to each well. After 19 minutes, the absorbance at 405 nm of each well was read in the automated microplate absorbance reader (Biotek, Winooski, VT). The seven clones tested from the ACA-6635 phage library screen were selected because of their high micropanning scores and negative phage-ELISA scores (see below). As shown in Figure 9, three clones (3A12, 3B3, and 3A5) out of the seven tested exhibited a very strong immunospecific signal in the phage-capture ELISA.
  • Peptide-ELISA In the standard protocol, stock solutions of tetrameric peptide in dimethylformamide were diluted 1000 times to 10 ⁇ g/mL in pH 9.5 carbonate buffer. Each microtitration plate well was coated overnight at 40°C with 100 ⁇ L of dilute peptide followed by blocking with buffered albumin. Peptide-coated microtitration plates were incubated 1 -2 hours at room temperature with aPL sera at several dilutions starting at 1 :50. Following washes, the presence of peptide- bound human IgG was determined with enzyme-conjugated anti -human IgG according to standard ELISA procedures.
  • Chantilly VA was coated with 100 ⁇ L of a solution containing 10 ⁇ g tetravalent peptide ACA 6501/3B10 in 50 mM sodium carbonate, pH 9.5, containing 35 mM sodium bicarbonate (Fisher Scientific, Pittsburgh, PA, reagent grade) for at least 1 hour at RT, except for three wells used as blank controls.
  • the liquid was then removed from the wells and 200 ⁇ L of 0.5% (wt/vol) BSA (Sigma Chemical,
  • SUBS ⁇ TUTE SHEET (RULE 26) ⁇ L of a stock solution of approximately 400-500 ⁇ g/mL of monomeric peptide (ACA-5A12 or -CB2 or -3B10 or scrambled -3B10 as negative control) in TBS; and 8.2 ⁇ L of 1 :10 diluted serum 6501 in 0.5% BSA-TBS.
  • the following reagents were mixed in the second microfuge tube: 30 ⁇ L of 5%> BSA; 290 ⁇ L TBS; 2 ⁇ L of a stock solution of approximately 400-500 ⁇ g/mL of monomeric peptide (ACA-5A12 or -CB2 or -3B10 or scrambled -3B10 as negative control) in TBS; and 8.2 gL of 1 :10 diluted serum 6501 in 0.5% BSA-TBS.
  • the following reagents were mixed in the third microfuge tube: 30 ⁇ L of 5% BSA, 287 ⁇ L of a 1 : 10 dilution of approximately 400-500 ⁇ g/mL of monomeric peptides (5A12, CB2.
  • the CL-coated, blocked plate was washed 5 times with TBS and then to each well was added ⁇ 2-GPI as 100 ⁇ L of 2.3% (v/v in PBS) IgG-depleted human serum (Sigma Chemical Co., St. Louis, MO) and incubated 2 hours at RT.
  • variable amounts of each of six peptides were mixed with 22 ⁇ L ACA-6501 serum diluted with 3% fish gelatin in 1 :1 TBS/PBS (final dilution of 1 :400) in a final volume of 220 ⁇ L using Eppendorf microcentrifuge tubes.
  • tube #1 were mixed 181.3 ⁇ L of 3% fish gelatin in TBS-PBS, 16.7 ⁇ L of peptide stock solution plus 22 ⁇ L of ACA-6501 serum diluted 40 times in 3% fish gelatin/TBS-PBS.
  • Stock solutions ranged from 450-800 ⁇ g/mL for peptides #951 (diserine non-cyclized negative control), #952 (a lot of LJP 690), and thioethers CCTE-3G3, CHTE-3G3, HCTE-3G3 and HHTE-3G3.
  • peptides #951 (diserine non-cyclized negative control)
  • #952 (a lot of LJP 690)
  • thioethers CCTE-3G3, CHTE-3G3, HCTE-3G3 and HHTE-3G3.
  • tube #2 the following were added: 148 ⁇ L fish gelatin/TBS-PBS, 50 ⁇ L of peptide stock solution, plus 22 ⁇ L of 40 times diluted ACA6501 serum.
  • Tube #3 contained 48 ⁇ L fish gelatin/TBS-PBS, 50 ⁇ L of peptide stock solution, plus 22 ⁇ L of 40 times diluted ACA-6501 serum.
  • the control tube #4 received 396 ⁇ L fish gelatin TBS-PBS and 44 ⁇ L of 40 times diluted ACA-6501 serum (no peptide).
  • Each of the tubes incubated for approximately 1 hour at RT.
  • the CL/ ⁇ 2-GPI microtitration plate was washed 5 times with TBS and 100 ⁇ L aliquots in duplicate from Eppendorf tubes #1, 2, 3, and 4 containing the antibody-peptide (or no peptide mixtures) were added to the wells.
  • a volume of 100 ⁇ L from tube #4 was added to the duplicate control wells containing no cardiolipin.
  • the microtitration plate was incubated for 1 hour at RT with agitation
  • variable amounts of each of four test peptides were mixed with 22 ⁇ L of ACA-6501 serum diluted with sample diluent in a final volume of 220 ⁇ L using Eppendorf microcentrifuge tubes.
  • tube #1 188 ⁇ L of sample diluent, 10 ⁇ L of peptide stock solution (2 mg - 4 mg/mL diluent), and 22 ⁇ L of AC-6501 serum diluted 1 :35 in sample diluent were added.
  • tube #2 158 ⁇ L sample diluent, 40 ⁇ L peptide stock solution and 22 ⁇ L of 1 :35 diluted ACA-6501 serum were added.
  • Tube #3
  • SUBSTTTUTE SHEET contained 38 HL sample dituent, 160 ⁇ L peptide stock solution and 22 ⁇ L of 1 :35 diluted ACA-6501 sertim.
  • the control tube, tube 94 contained 396 ⁇ L sample diluent and 44 ⁇ L of 1:35 diluted ACA-6501 serum (no peptide). Each of the tubes was incubated for approximately 1 hours at room temperature.
  • the ⁇ 2 -GPI microplate was washed 5 times with TBS and 100 ⁇ L of the mixtures contained in Eppendorf tubes 91, 2, 3 and 4 was added to duplicate wells of the microplate.
  • the plate was again washed 5 times with TBS and the calorimetric enzyme detection developed by adding 100 ⁇ L PPMP solution (7.8 g phenolphthalein monophosphate and 69.5 g 2-amino-2-methyl-l-propanol in 100 mL water stock solution diluted 1 :26 with water). After 22 minutes, the reaction was stopped with
  • each soluble, monomer peptide to be tested was set up in three test tubes (Eppendorf micro test tubes, Brinkmann Instruments,
  • the concentration of the stock solution was approximately 340-400 ⁇ g/mL TBS and aliquots of 19 ⁇ L, 75 ⁇ L. and 292 ⁇ L were removed to prepare the three peptide concentration tubes.
  • stock solution concentrations were 400-500 ⁇ g/mL in TBS. Aliquots from each stock solution of 1 ⁇ L, 4 ⁇ L, and 16 ⁇ L were removed to set up the three concentration tubes for each peptide.
  • a tube with a final volume of 660 ⁇ L was prepared containing 60 ⁇ L 5% BSA, 16.5 ⁇ L of a 1 :10 dilution of ACA-6501 and 583.5 ⁇ L
  • TBS i.e., the same final concentrations (0.5% BSA and ACA-6501 serum at 1 :400) as the 330 ⁇ L tubes but with twice the volume and without peptide.
  • the plate coated with tetravalent peptide was washed 5 times with TBS. From each 330 ⁇ L tube containing peptides 139, 142 and 143 at different concentrations, 100 ⁇ L was added to coated triplicate wells. Three 100 ⁇ L aliquots from the 660 ⁇ L control tube without peptide were each added to coated wells and three additional 100 ⁇ L aliquots were each added to uncoated blank wells. The plate was incubated at 40 rev/min on a rotary orbital shaker (Rotator V, American Dade. Miami. FL) for 1 hour at RT and then washed 5 times with TBS.
  • a rotary orbital shaker Rostator V, American Dade. Miami. FL
  • SUBSTTTUTE SHEET (RULE 26) IgG/alkaline phosphatase conjugate (Cat. no. 62-8422, Zymed, South San Francisco, CA) diluted 1 : 1000 in BSA/TBS was added to the microplate. After incubation for 1 hour at RT, the plate again was washed 5 times with TBS. Color development followed the addition of 100 ⁇ L/well of freshly prepared dilute PPMP substrate solution. A dilute solution of PPMP (phenolphthalein monophosphate, cat. no. P-5758, Sigma Chemical Co., St. Louis, MO) was prepared by making a 1 :26 dilution with water of the PPMP stock solution (0.
  • Example 6 Substitution of alpha methyl proline into 3B10 and the resulting ELISAs Testing of peptides ACA-6501/3B10 and analogs in which prolines at the
  • Microtitration plates were coated with tetramer 3B10 peptide as described for Example 5. For each of the four peptides tested, three peptide concentrations were prepared in tubes. As in Example 5, these twelve tubes had final concentrations of 0.5% BSA/TBS and ACA-6501 serum at a final dilution of 1 :400 in a final volume of 330 ⁇ L. All peptide stock solutions were at 400-500 ⁇ g/mL TBS.
  • Example 7 Abbreviated Description of Screen with 6626 Antibody and the Corresponding Sequences
  • Affinity purified ACA-6626 (AffACA-6626) was isolated by affinity purification from 8 mL of ACA-6626 plasma as previously described.
  • AffACA- 6626 (10 ⁇ g) was incubated with the epitope xy'z phage library consisting of a pool of all p-III component libraries in a final volume of 100 ⁇ L as previously described for ACA-6501 biopanning. Following three rounds of biopanning, randomly selected phage from the second and third rounds were tested by micropanning. Only a few clones were weakly immunopositive at a 1 : 1000 dilution. An additional 4th round of biopanning was carried out.
  • Micropanning of 94 fourth round clones revealed 43 immunopositives, some at phage dilutions as high as 1 : 100,000.
  • G-Tracking DNA sequencing of the 43 immunopositive clones carried out as previously described for ACA-6501 revealed 5 unique sequences. After conventional four base DNA sequencing, the translated amino acid sequences of Table 3 were obtained.
  • the epi xy z phage display library was screened using methods similar to those in Example 4 with ACA affinity purified antibody from patient number 6644.
  • a colony blot assay as described previously was employed as the final identification step prior to peptide synthesis. Approximately 150 colonies were plated on the original nitrocellulose membrane and assayed. Antibody from patient 6644 was used at a concentration of 1 ⁇ g/mL. Of the 150 colonies plated on the nitrocellular membrane and assayed, only 4 were strongly positive and 2 weakly positive in this screen. Sequencing of the inserts of the six positive phage selected by this screen revealed that the inserts were all derived from the 8-mer library with a free amino-terminus (epi z ):
  • AffACA-6641 was isolated from 4 mL of plasma taken from patient number 6641.
  • AffACA-6641 (10 ⁇ g) was incubated with the pooled p-III phage libraries in a final volume of 100 ⁇ L as described previously.
  • 45 clones from the 3rd and 4th rounds were tested by micropanning. Of the 45, 23 scored negative.
  • the 3rd round phage yielded two clones that scored 4+, two that scored 3+ and two that scored 2+. From the 4th round, one clone scored 4+, one scored 3+ and three scored 2+.
  • G-tracking DNA sequencing revealed six unique sequences. Only one, clone 3G3, was strongly positive in the phage-capture ELISA. Four base DNA sequencing gave the following translated peptide sequence:
  • Example 10 Peptide conjugation to non-immunogenic. multivalent carriers
  • Several tetravalent platforms for the development of B cell tolerogens have been developed as described in co-owned and co-pending U.S. Patent Applications, Serial Nos., 08/1 18, 055, filed September 8, 1993, 08/152,506, filed November 15, 1993, and U.S. Patents Nos. 5,268,454, 5,276,013 and 5.162,515 which are incorporated by reference herein in their entirety.
  • Candidate peptides selected by aPL antibody screens of epitope libraries are conjugated and tested for modifications of immunochemical behavior such as antibody binding.
  • Non-inununogenic multivalent platforms with amine groups are synthesized as shown in the following scheme.
  • a peptide is synthesized with standard solid phase methods using FMOC chemistry on a Wang (p-alkoxybenzyl) resin, using trifluoroacetic acid (TFA) stable protecting groups (benzyl ester or cyclohexyl ester on carboxyl groups and carbobenzyloxy (CBZ) on amino groups). Amino acid residues are added sequentially to the amino terminus.
  • the peptide is removed from the resin with TFA to provide a peptide with one free carboxyl group at the carboxy terminus and all the other carboxyls and amines blocked.
  • the protected peptide is purified by reverse phase HPLC.
  • the protected peptide (0.3 mmol) is dissolved in 1 mL of dimethylformamide (DMF), and to the solution is added 0.3 mmol of diisopropylcarbodiimide and 0.3 mmol of 1 -hydroxybenzotriazole hydrate
  • conjugate 3 is treated with hydrofluoric acid (HF) in the presence of anisole for 1 hour at 0° to give conjugate 4. Purification is accomplished by preparative reverse phase HPLC.
  • the following scheme shows the attachment of an amino group of a peptide to a carboxy group on a platform.
  • pep unprotected p ⁇ _d ⁇
  • Succinic anhydride (1.0 g, 10 mmol) is added to a solution of 861 mg (1.0 mmol) of 2_and 252 mg (3.0 mmol) of NaHCO 3 in 20 mL of 1/1 dioxane/H 2 O, and the mixture is stirred for 16 hours at RT.
  • the mixture is acidified with IN HCl and concentrated.
  • the concentrate is purified by silica gel chromatography to provide 5.
  • a peptide is synthesized with standard solid phase methods on an amide resin, which resulted in a carboxy terminal amide after cleavage from the resin, using TFA stable protecting groups (benzyl ester or cyclohexyl ester on carboxyl groups and CBZ on amino groups). Amino acid residues are added sequentially to the amino terminus using standard FMOC chemistry.
  • the peptide is removed from the resin with trifluoroacetic acid to provide a protected peptide with a free amine linker.
  • the protected peptide is purified by reverse phase HPLC.
  • Compound 7 is purified by preparative reverse phase HPLC.
  • pep cyclic norwfisuftida peptide
  • N-BOC-glycylproline-4-nitrophenyl ester compound 15: A solution of 3.0 g (1 1.6 mmol) of N-BOC-glycylproline and 1.93 g (13.9 mmol) of 4-nitrophenol in 82 mL of dry THF was cooled to 0°C and 3.34 g (16.2 mmol) of DCC was added.
  • N-FMOC-S-t-butylthiocysteineamide A solution of 5.0 g (11.6 mmol) of FMOC-S-t-butylthiocysteine and 1.33 g (11.6 mmol) of N-
  • the mixture was stirred with 200 mL of CH 2 C1 2 until most of the solid dissolved, then was shaken with 100 mL of IN HCl solution.
  • the CH 2 C1 2 layer was washed with 100 mL of saturated NaHCO 3 solution, dried (Na 2 SO 4 ), and filtered.
  • N-FMOC-L-Alanyl-L-2-methylproline N-FMOC-L-Alanyl-L-2-methylproline.
  • compound 19 A solution of 2- methylproline (Seebach et al. (1983) J Am. Chem. Soc. 105:5390-5398) (1.00 g5 4.76 mmol), 4.00 g (47.6 mmol) of NaHCO 3 , and 31 mg (0.23 mmol) of HOBT in 6.9 mL of DMF was cooled to 0°C and 3.18 g (6.66 mmol) of N-FMOC-L-alanine was added. The 'faction was stirred for 1 hour at 0°C, then at room temperature for 18 hours.
  • N-FMOC-L-Leucinyl-HMPB-MBHA resin A solution of N-FMOC-L-leucine in 22.5 mL of CH 2 Cl 2 and a few drops of DMF was prepared and cooled to 0°C. To the solution was added 1.71 ml (1.38 g, 10.9 mmol) of diisopropylcarbodiimide (DIC) and the mixture was stirred for 20 minutes at 0°C. The mixture was concentrated to an oil; meanwhile, enough DMF approximately 3 mL) was added to 2.5 g (0.87 mmol/g, 2.18 mmol) HMPB-MBHA resin (Nova Biochem) to swell the resin. The concentrated oil was dissolved in a minimal amount of DMF
  • N-FMOC-linear peptide with t-butyl ester on aspartic acid and Pmc group on arginine and with thioether insert compound 20: This peptide was prepared by standard FMOC synthesis on N-FMOC-L-leucinyl-HMPb-MBRA resin. Three equivalents of amino acid HOBT and (DIC) were used for each coupling step with the exception of the coupling step of compound 18. Two equivalents of compound 18 were used with three equivalents of HOBT and diisopropylcarbodiimide. Each step was monitored by using 10 ⁇ L of bromophenol blue indicator. Completeness of the reaction was also assessed with a ninhydrin test (beads turn blue for incomplete reaction with 1 mg is heated at
  • LJP 685 also referred to as compound 21, was treated with the PNP ester of 3-tritylmercaptopropionic acid and the resulting product was detritylated to give compound 22, the peptide with the free thiol linker. Reaction of an excess of compound 22 with valency platform molecule 12 produced tetravalent conjugate 25. Treatment of compound 21 with the longer linker, compound 3,3 (see reaction scheme below), followed by detritylation, gave compound 24. Compound 24 reacted with valency platform molecule 12 to give the tetraconjugate 26. Both conjugation reactions appeared very clean by HPLC.
  • the thiobenzoate ester, compound 28, was prepared from compound 27. Compound 28 was converted to compound 29 in portions. The thiobenzoate was
  • SUBSTTTUTE SHEET (RULE 26) removed by ethanolysis and the resulting thiol was tritylated. The ethyl ester was then hydrolyzed to give compound 29.
  • the nitrophenyl phosphate (PNP) ester, compound 30, was prepared from compound 2__- Aminotrioxoudecanoicacid ethylester, compound 3_1, was prepared by treatment of compound 2Z with sodium azide and reduction to the amine. Amine 3-1 was acylated with compound 3-0 to provide compound 2. Hydrolysis of compound 32 was achieved by treatment with sodium hydroxide to give a free carboxylic acid.
  • Tetravalent conjugate 35 was prepared as shown below. The peptide with linkers attached, compound 2A was dissolved in He sparged, pH 8.5, 200 niM borate buffer. To the mixture was added 0.3 mol equivalents of platform compound 1-2. The mixture was stirred for I hour and the product was purified by HPLC.
  • Tritiated thymidine uptake by peptide-stimulated T cells was monitored in 96well round bottom plates.
  • a single-cell suspension of draining lymph node cells (mice) or isolated peripheral blood lymphocytes (human), 5 x 10 were mixed with between I and 30 jig of peptide in a final volume of 150 ⁇ L per well and incubated for 5 days at 37°C in 5% C0 2 . At that point, 1 micro curie of labeled thymidine was added and incubated for an additional 15-24 hours.
  • the harvested cells were collected on filters and counted by liquid scintillation spectrometry.
  • the cell suspension was divided into aliquots of 2.5 mL /petri dish and incubated for 2 hours at 37°C with (LJP685) 4 -DABA-PEG, compound 36, and (LJP685) -TEG, compound 35, in concentrations of 100 ⁇ M, 20 ⁇ M and 4 ⁇ M.
  • One group of cells was incubated without toleragen and acted as the positive control.
  • the cells were then washed with large volumes of balanced salt solution and resuspended in 2.5 mL of balanced salt solution.
  • the cells were then injected into 650R irradiated syngeneic recipient mice in such a manner that all of the cells from a given treatment group were divided evenly into five recipients.
  • mice All of the recipient mice, including the positive controls, were then given a booster immunization of 10 ⁇ g of LJP 685-KLH in saline, intraperitoneally. Seven days after the booster immunization, the mice were bled and their sera tested for the presence of anti-LJP 685 antibody.
  • ROE Overhauser Effect
  • coupling data are consistent with one structure.
  • Distance geometry calculations yielded a family of 50 structures. The 15 best had a mean root mean square deviation (RMSD) for all atoms of 2.1 ⁇ 0.2 angstroms.
  • RMSD mean root mean square deviation
  • RMSD for all non-hydrogen atoms was 2.45 ⁇ 0.36 angstroms when each structure was compared to the centroid.
  • Figure 18 displays the structure closest to the centroid of the ensemble and, therefore, is a reasonable representation of the shape of the peptide 925 molecule.
  • Figure 19 compares the structure of peptide 925 (labeled at the bottom of the figure as 3G3) with the structure of peptide 5A12.
  • Both peptides have turns at approximately the same positions in the peptide sequence.
  • the pharmaeophore of the peptides has been tentatively identified as a small hydrophobic group and a positively charged group.
  • the gem-dimethyl and amino groups of peptide 925 are tentatively identified as the pharmaeophore of this peptide as shown in Figure 20.
  • the hydrocarbon linkers that tether the pharmacophore groups to some scaffold have the lengths specified in Figure 20 and the points at which these linkers are attached to the scaffold are separated by the distance specified. Finally, the dihedral angle defining the relative orientation of the two linkers was determined to be 22°.
  • Bis-N-ft-butoxycarbonylVdiaminobenzoic acid, compound 40 A solution of 7.18 g (32.9 mmol of di-t-butyldicarbonate in 5.5 mL of MeOH was slowly added to a solution of 2.5 g (16.4 mmol) of 3,5-diaminobenzoic acid and 2.76 g (32.9 mmol) of NaHCO 3 in 44.5 mL of H 2 O and 22.5 mL of MeOH, and the mixture was stirred at room temperature for 24 h. The mixture was cooled to 0°, and 6.53 g of citric acid was added, and the mixture was extracted with EtOAc. The combined EtOAc layers were dried (MgSO 4 ), filtered, and concentrated.
  • N-hydroxysuccinimidyl ester of compound 40 N-hydroxysuccinimidyl ester of compound 40.
  • compound 41 Dicyclohexylcarbodiimide (3.34 g, 16.2 mmol) was added to a solution of 3.8 g (10. 8 mmol) of compound A0 and 1.24 g (10.8 mmol) of N-hydroxysuccinimide in 55 mL of EtOAc which had been cooled to 0°, and the resulting mixture was stirred for 18 h allowing to come to room temperature. To the mixture was added 0.55 mL of acetic acid. The mixture was stirred for 30 min and placed in the freezer for 2 h. The mixture was filtered to remove solids, and the filtrate was concentrated to give 5.80 g of pink foamy solid. Purification by silica gel chromatography (60/40/1 hexane/EtOAc/HOAc) gave 4.30 g (89%) of compound 41 as a slightly pink solid.
  • Diamino-TEG bis-trifluoroacetate salt, compound 44 300 mg (0.57 mmol) of compound 43. was.dissolved in 3.5 g of CH 2 C1 2 , and 3.5 mL of trifluoroacetic acid was added. The mixture was stirred for 3 h at room temperature, and the solution was concentrated to give 398 mg of crude compound 44 which was used directly in the next step.
  • EtOAc layer was washed successively with 200 mL of 0.1 N HCl and 200 mL of H 2 O, dried (Na 2 SO 4 ), filtered, and concentrated to yield 3.0 g (58%) of compound 47 as a white solid.
  • SUBSTTTUTE SHEET (RULE 26) accomplished by silica gel chromatography (gradient, 80/20 to 70/30 CH 3 Cl/MeOH).
  • Compound 50 A solution of 135 uL (0.66 mmol) of triethyleneglycol bis- chloro formate in 0.3 mL of THF was added to a 0° solution of 855 mg (1.58 mmol) of compound 49 and 275 uL (1. mmol) of diisopropylethylamine in 13 mL of THF. The cloudy mixture cleared when the ice bath was removed. An additional 70 uL of dilsopropyethylamine was added to maintain a basic pH. The mixture was stirred at room temperature for a total of 3 h and partitioned between 25 mL of H 2 O and 25 mL of EtOAc.
  • the resulting HBr salt was dissolved in 10 mL of H 2 O, and 191 mg (2.27 mmol) of NaHCO 3 was added. A solution of 591 mg (2.27 mmol) of bromoacetic anhydride in 10 mL dioxane was added to the mixture. An additional 2 mL of dioxane was used to rinse. More NaHCO 3 was added as needed to maintain a basic pH. The mixture was stirred for 2 h at room temperature, and acidified with
  • SUBSTTTUTE SHEET (RULE 26) uL of triethyleneglycol biscmoroforinate in 4 mL of CH 2 C1 2 , and the mixture was stirred for 3 h at room temperature, mixture was partitioned between 80 mL of CH 2 C1 2 and 80 mL of 1 N HCl. The CH L layer was washed with two 80 mL portions of water, dried (MgSO 4 ), filtered, and concentrated to give 736 mg (93%o) of compound 58 as a crystalline solid.
  • Compound 60 Compound 58 (61 mg, 0.48 mmol) was dissolved in 3 mL of 30% HBr/HOAc and the resulting mixture was stirred at room temperature for 1 h at which time 5 mL of Et 2 O added. The mixture was placed in the freezer for 1 h and centrifuged. The resulting pellet was washed with Et 2 O and dried to give the tetralaydrobromide salt 59 which was dissolved in 1 mL of H 2 O. To the mixture is added 49 mg (0.58 mmol) of NaHCO 3 and 3 mL of dioxane. More NaHCO 3 is added, if needed, to make the mixture basic.
  • Example 25 Synthesis of the Tetravalent Platform tetrakis-BAIPIZ PMA. 63.
  • Example 26 Synthesis of the Tetravalent Platform BA/PIZ/IDA/HB/TEG. 68.
  • Compound 66 Compound ⁇ £ is prepared similarly to the method of preparing compound 56 in example 23.
  • Compound 65 is used instead of N-BOC- iminodiacetic acid, and compound 5_7 is used instead of mono-CBZ-piperazine. Purification is accomplished using silica gel chromatography.
  • Compound 68 Compound 66 is converted to compound 68 in essentially the same manner as described for the conversion of 58 to 60 in Example 23. Purification is accomplished using silica gel chromatography.
  • Compound 69 Compound 53 is hydrolyzed with LiOH in essentially the same manner as described for the hydrolysis of 64 in example 25 with the exception that 4.4 equivalents of LiOH is used.
  • Compound 70 The tetra-acid, compound 69_, is converted to compound 70 in essentially the same manner as described for the conversion of pyromellitic acid to 61 in example 24 with the exception that 69 is used instead of pyromellitic acid.
  • Compound 72 Compound 70 is converted to compound 72 in essentially the same manner described for the conversion of 58 to 60 in Example 23. Purification is accomplished using silica gel chromatography.
  • Example 28 Synthesis of Conjugates of Haloacetylated Tetravalent Compounds Synthesis of fLJP685yMTU/ A/DAB A/ATEG conjugate compound 73 :
  • a solution was prepared of 21 mg (15.5 umol) of compound 24 in Helium sparged pH 8.5 200 mM borate buffer. To the solution was added a second solution consisting of 3.25 mg * (2.6 umol) of I A/DAB A/ATEG, compound 46, dissolved in 396 uL of MeOH. A precipitate formed, and 1 mL of MeOH was added to dissolve all the solids. The mixture was stirred for 18 h at room temperature, and 6 mL of 9/1 H 2 O/HOAc was added. The mixture was diluted with 50 ML of H O/CH 3 CN and loaded onto an HPLC preparative column. Purification was accomplished by preparative HPLC (25 cm X 22.4 mm C, 8 .
  • Example 29 Synthesis of Conjugates Tetravalent Platform 55.
  • Pep can be LJP685 or other relevant peptide.
  • Pep can be LJP685 or other relevant peptide.
  • Pep Pep LJP 685
  • the FITC peptide was obtained as a bright yellow powder after lyophilization (3.7 mg, 15% yield): MS (ESI): m/e (M+l) Calcd. for C 73 H 102 N 19 0 20 S 3 : 1661, obsd.: 1661.
  • the above described dbFP assay provides binding constants for FITC- labeled peptides and requires on the order of 0.5 mg of purified antibody.
  • the cFP assay provides binding constants for peptides that lack the FITC group and it consumes less antibody, on the order of 10 ⁇ g.
  • the cFP assay is modified from that reported in PanVera Applications Guide (1994) PanVera Corporation such that it consumes 50-fold less antibody. Briefly, antibody (ACA 6701) is mixed with trace FITC labeled peptide (CB2*-F) and enough time is allowed for equilibrium to be reached. This was 1 hour for ACA 6701 and CB2*-F.
  • (mP) value should be 20 or more mP units to insure reliable results.
  • P L is the same as in Equation 1
  • 1 is the concentration of unlabeled peptide competitor
  • Kj' is the apparent dissociation constant for that peptide. Values for these parameters were obtained by fitting cFP titration data to the above equation.
  • the true dissociation constant for I is obtained from Equation 4.
  • Equation 4 K,'/(l .0 + R/K D )
  • R and K D are defined as in Equation 1.
  • the R K D ratio is obtained from the values of P H ' (from Equation 3) and P H and P L (from Equation 1) and using
  • Equation 5 can be used to determine aPL antibody concentrations once the titration defined by Equation 1 is performed as a "standard curve.”
  • this method provides a means of standardizing all aPL antibody stock solution concentrations and of analyzing their binding activity/stability over time using only 5-10 ⁇ g of antibody per cFP assay.
  • mice were immunized with the peptide conjugated to the immunogenic carrier Keyhole Limpet Hemocyanin (KLH) to generate peptide-specific memory B cells.
  • KLH Keyhole Limpet Hemocyanin
  • mice later, groups of 5 mice per group were treated with various doses of the test conjugates, one group of mice was not treated and acted as the control. Five days later, all of the mice, including the control group, were boosted with the peptide conjugated to KLH and seven days later all of the mice were bled and their sera assayed for anti-peptide antibodies using a modified Farr assay.
  • ABSORB Binding Capacity
  • G.M. Iverson "Assay for in vivo adoptive immune response," Volume II, Chapter 67, HANDBOOK OF EXPERIMENTAL IMMUNOLOGY (Blackwell Scientific Publications, Weir et al, eds., 4th ed., Oxford, 1986). These values were then used to determine a mean and standard deviation for all of the individuals of a group.
  • the cells were washed, transferred to irradiated syngeneic recipients and boosted with the peptide conjugated to KLH. Seven days later all of the mice were bled and their sera assayed for anti-peptide antibodies.
  • the conjugate that did not induce any detectable tolerance when tested in the in vivo model did induce tolerance when tested in this in vitro model. This result supports the assumption that the difference between conjugates is due to a short half-life of the conjugate.
  • the conjugate was administered continuously by an implanted osmotic pump over a prolonged period of time. The results clearly show that this conjugate induced tolerance when administered by sustained release but not when administered as a bolus as shown in Figure 32.
  • mice were primed with LJP-685-KLH on alum plus pertussis as an adjuvant. Three weeks later, the mice were treated with a range of doses of the
  • (LJP685) 4 /MTU-AHAB-TEG conjugate One group was not treated and acted as a control group. Five days later, all of the mice, including the control group, were boosted with 10 ⁇ g LJP685-KLH and seven days later the mice were bled. Their sera were analyzed for anti-LJP685 antibodies by a modified Fair assay as described above. The results as shown in Figure 28 demonstrate that the treatment with the (LJP685) /MTU-AHAB-TEG conjugate, over a dose range of 1 to 50 nmoles, had no detectable effect on the anti-LJP685 response.
  • mice were primed with LJP685-KLH on alum Three weeks later, the mice were treated with 5, 10 or 50 nmoles of the (LJP685) 4 /MTU- D ABA-TEG conjugate. One group was not treated and acted as a control group. Five days later, all of the mice, including the control group, were boosted with 10 ⁇ g LJP685-KLH and seven days later the mice were bled. Their sera were analyzed for anti-LJP685 antibodies by a modified Fair assay. The results as shown in Figure 29 demonstrate that (LJP685) 4 /MTU-DABA-TEG conjugate induces tolerance in the in vivo model with an ED 50 of 5 nmoles.
  • SUBSTTTUTE SHEET (RULE 26) cells were washed, transferred to irradiated recipients and boosted with 10 ⁇ g of LJP685-K-LH. Seven days later, the mice were bled and their sera were analyzed for anti-LJP685 antibodies by a modified Farr assay.
  • the results as shown in Figure 30 clearly illustrate that the (LJP685) 4 /MTU-AHAB-TEG conjugate can induce tolerance when tested in the in vitro model achieving an IC 50 of ⁇ 4 ⁇ M.
  • Figure 31 demonstrate that (LJP685) 4 /MTU-D ABA-TEG conjugate can induce tolerance when tested in the in vitro model, achieving an IC 50 of ⁇ 4 ⁇ M.
  • mice were primed with LJP685-KLH on alum plus pertussis. Three weeks later, the mice were divided into 5 groups of five mice per group. On day 1 , one group was treated with a bolus of saline and another group was treated with a bolus containing 50 nmoles of the (LJP685) 4 /MTU-AHAB-TEG conjugate. The three remaining groups were implanted with osmotic pumps. In one group, the pumps were filled with saline and delivered at 1 ⁇ L/hour for 3 days. The two remaining groups received pumps filled with the (LJP685) 4 /MTU-AHAB-TEG conjugate (50 nmoles). One group received pumps that deliver at 1 ⁇ L/hour for three days and the other received pumps that deliver at 0.5 ⁇ L/hour for seven days. On day 5, the pumps that deliver for three days were surgically removed. On day 1, the pumps that deliver for three days were surgically removed. On day
  • SUBSTTTUTE SHEET (RULE 26) 7 all of the mice, including the control group, were boosted with 10 ⁇ g of LJP685-KLH. On day 10, the pumps that deliver for seven days were surgically removed. On day 14, all of the mice were bled. Their sera were analyzed for anti- LJP685 antibodies by a modified Farr assay. The results are shown in Figure 32.
  • mice are primed with peptide-KLH on alum plus pertussis. Three weeks later, the mice are treated with the (LJP-peptide) 4 /MTU-BMP-TEG conjugate, one group is not treated and acts as the control group. Five days later, all of the mice, including the control group, are boosted with 10 ⁇ g of peptide-KLH and seven days later the mice are bled. Their sera are analyzed for anti-peptide antibodies by a modified Farr assay.

Abstract

Analogues de l'anticorps antiphospholipide (aPL) qui (a) se lient spécifiquement aux lymphocytes B auxquels un épitope d'aPL se lie. Des analogues optimisés qui sont exempts du ou des épitopes de lymphocytes T sont utiles comme conjugués pour traiter des maladies dépendantes de l'anticorps aPL. Des conjugués comprenant des analogues d'aPL et des molécules à plate-forme de valence non immunogènes sont décrits, ainsi que de nouvelles molécules à plate-forme de valence non immunogènes et des séquences de liaison, des procédés de préparation et d'identification desdits analogues, de procédés de traitement au moyen desdits analogues, des procédés et des compositions destinés à la préparation de conjugués desdits analogues, et des dosages immunologiques et diagnostiques pour les anticorps aPL.
PCT/US1997/010075 1996-06-06 1997-06-06 PEPTIDES IMMUNOREACTIFS PAR RAPPORT AUX aPL, LEURS CONJUGUES ET PROCEDES DE TRAITEMENT DES PATHOLOGIES DEPENDANTES DE L'ANTICORPS aPL WO1997046251A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU36404/97A AU734638B2 (en) 1996-06-06 1997-06-06 aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies
EP97933138A EP0954531A1 (fr) 1996-06-06 1997-06-06 PEPTIDES IMMUNOREACTIFS PAR RAPPORT AUX aPL, LEURS CONJUGUES ET PROCEDES DE TRAITEMENT DES PATHOLOGIES DEPENDANTES DE L'ANTICORPS aPL
JP10500927A JP2000512981A (ja) 1996-06-06 1997-06-06 aPL免疫応答性ペプチド、その結合体およびaPL抗体媒介病理のための処置方法
CA002256449A CA2256449A1 (fr) 1996-06-06 1997-06-06 Peptides immunoreactifs par rapport aux apl, leurs conjugues et procedes de traitement des pathologies dependantes de l'anticorps apl
NO985636A NO985636L (no) 1996-06-06 1998-12-03 aPL immunoreaktive peptider, konjugater derav og metoder for behandling av aPL antistoff-medierte patologier

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/660,092 1996-06-06
US08/660,092 US6207160B1 (en) 1995-06-07 1996-06-06 aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies
US76050896A 1996-12-05 1996-12-05
US08/760,508 1996-12-05

Publications (2)

Publication Number Publication Date
WO1997046251A1 WO1997046251A1 (fr) 1997-12-11
WO1997046251A9 true WO1997046251A9 (fr) 1998-06-04

Family

ID=27097975

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/010075 WO1997046251A1 (fr) 1996-06-06 1997-06-06 PEPTIDES IMMUNOREACTIFS PAR RAPPORT AUX aPL, LEURS CONJUGUES ET PROCEDES DE TRAITEMENT DES PATHOLOGIES DEPENDANTES DE L'ANTICORPS aPL

Country Status (7)

Country Link
EP (1) EP0954531A1 (fr)
JP (1) JP2000512981A (fr)
KR (1) KR20000016414A (fr)
AU (1) AU734638B2 (fr)
CA (1) CA2256449A1 (fr)
NO (1) NO985636L (fr)
WO (1) WO1997046251A1 (fr)

Families Citing this family (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE164395T1 (de) * 1990-12-03 1998-04-15 Genentech Inc Verfahren zur anreicherung von proteinvarianten mit geänderten bindungseigenschaften
US5874409A (en) 1995-06-07 1999-02-23 La Jolla Pharmaceutical Company APL immunoreactive peptides, conjugates thereof and methods of treatment for APL antibody-mediated pathologies
US7888466B2 (en) 1996-01-11 2011-02-15 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US6812327B1 (en) 1996-10-25 2004-11-02 Human Genome Sciences, Inc. Neutrokine-alpha polypeptides
US8212004B2 (en) 1999-03-02 2012-07-03 Human Genome Sciences, Inc. Neutrokine-alpha fusion proteins
US6858210B1 (en) 1998-06-09 2005-02-22 La Jolla Pharmaceutical Co. Therapeutic and diagnostic domain 1 β2GPI polypeptides and methods of using same
IL140918A0 (en) 1998-07-27 2002-02-10 Genentech Inc Improved transformation efficiency in phage display through modification of a coat protein
US6238860B1 (en) 1998-11-05 2001-05-29 Dyax Corp. Binding moieties for human parvovirus B19
US6458953B1 (en) 1998-12-09 2002-10-01 La Jolla Pharmaceutical Company Valency platform molecules comprising carbamate linkages
US6399578B1 (en) 1998-12-09 2002-06-04 La Jolla Pharmaceutical Company Conjugates comprising galactose α1,3 galactosyl epitopes and methods of using same
US6197526B1 (en) 1999-01-04 2001-03-06 Dyax Corp. Polypeptides for binding human factor VIII and fragments of human factor VIII
US7112438B2 (en) 1999-01-04 2006-09-26 Dyax Corp. Binding molecules for human factor VIII and factor VIII-like proteins
GB9919452D0 (en) * 1999-08-17 1999-10-20 Osteometer Biotech As Specific autoimmune reactions against isomerised/optically inverted epitopes:application for diagnosis of autoimmune diseases
AU7646100A (en) * 1999-11-05 2001-05-14 Novozymes A/S A high throughput screening (hts) method
KR20020059808A (ko) 1999-11-28 2002-07-13 와이즈먼 앤드루 항체 친화도에 기초한 루프스 치료방법 및 그것의 사용을위한 스크리닝 방법과 조성물
US20030031675A1 (en) 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
KR101287395B1 (ko) 2000-06-16 2014-11-04 휴먼 게놈 사이언시즈, 인코포레이티드 면역특이적으로 BLyS에 결합하는 항체
US7879328B2 (en) 2000-06-16 2011-02-01 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator
US20030091565A1 (en) 2000-08-18 2003-05-15 Beltzer James P. Binding polypeptides and methods based thereon
AU2001285066B2 (en) 2000-08-18 2006-06-29 Human Genome Sciences, Inc. Binding polypeptides for B lymphocyte stimulator protein (BLYS)
EP1366058B1 (fr) 2001-02-09 2011-01-26 Human Genome Sciences, Inc. Recepteur de chemokine de la g-proteine humaine (ccr5) hdgnr10
AU2002257142B2 (en) 2001-04-13 2008-09-04 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
DK2000545T3 (da) 2001-06-20 2011-11-28 Genentech Inc Sammensætninger og fremgangsmåder til diagnose og behandling af lunge-tumor
DK2423335T3 (da) 2001-06-21 2014-08-18 Dynavax Tech Corp Kimæriske immunmodulatoriske forbindelser og fremgangsmåder til anvendelse deraf
NZ573740A (en) 2001-09-18 2010-07-30 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor, particularly kidney tumor - TAT184
AU2002367318B2 (en) 2002-01-02 2007-07-12 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
AU2003230874A1 (en) 2002-04-16 2003-11-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
NZ538628A (en) 2002-08-12 2008-06-30 Dynavax Tech Corp Immunomodulatory compositions, methods of making, and methods of use thereof
ES2751414T5 (es) 2003-07-08 2024-04-23 Novartis Pharma Ag Anticuerpos antagonistas a polipéptidos heterólogos IL-17 A/F
ES2472690T3 (es) 2003-11-17 2014-07-02 Genentech, Inc. Anticuerpo contra CD22 para el tratamiento de tumores de origen hematopoy�tico
KR101268877B1 (ko) 2004-09-01 2013-05-31 다이나박스 테크놀로지 코퍼레이션 선천성 면역반응 및 자가면역의 억제를 위한 방법 및조성물
KR20070110077A (ko) 2005-03-10 2007-11-15 제넨테크, 인크. 혈관 완전성을 조정하기 위한 방법 및 조성물
US9168286B2 (en) 2005-10-13 2015-10-27 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
WO2007123765A2 (fr) 2006-03-31 2007-11-01 Human Genome Sciences Inc. NEUTROKINE-ALPHA et variant d'epissage de la neutrokine-alpha
AU2007243946B2 (en) 2006-04-05 2012-11-29 Curis, Inc. Method for using BOC/CDO to modulate hedgehog signaling
WO2008021290A2 (fr) 2006-08-09 2008-02-21 Homestead Clinical Corporation Protéines spécifiques d'organes et procédés d'utilisation
EP2061904A2 (fr) 2006-09-06 2009-05-27 Ortho-McNeil Pharmaceutical, Inc. Biomarqueurs pour l'évaluation d'une réponse au traitement de c-met
US20100297103A1 (en) 2006-09-14 2010-11-25 Medical & Biological Laboratories Co., Ltd. Antibody having enhanced adcc activity and method for production thereof
AU2008218199B2 (en) 2007-02-22 2013-10-31 Genentech, Inc. Methods for detecting inflammatory bowel disease
WO2009055076A2 (fr) 2007-10-26 2009-04-30 Dynavax Technologies Corporation Procédés et compositions pour inhiber les réponses immunitaires et l'auto-immunité
EP2304439A4 (fr) 2008-05-29 2012-07-04 Nuclea Biotechnologies Llc Anticorps anti-phospho-akt
KR20110038047A (ko) 2008-06-20 2011-04-13 고꾸리츠 다이가꾸 호우징 오까야마 다이가꾸 산화 LDL/β2GPI복합체에 대한 항체 및 그 용도
US9182406B2 (en) 2008-08-04 2015-11-10 Biodesy, Inc. Nonlinear optical detection of molecules comprising an unnatural amino acid possessing a hyperpolarizability
MX2012004617A (es) 2009-10-22 2012-05-08 Genentech Inc Metodos y composiciones para modular activacion de hepsina de proteina que estimula macrofago.
MX338694B (es) 2009-11-30 2016-04-27 Genentech Inc Composiciones y metodos para el diagnostico y el tratamiento de tumores.
PE20130214A1 (es) 2010-02-23 2013-03-11 Genentech Inc Composiciones y metodos para el diagnostico y tratamiento de tumores
EP2566893A1 (fr) 2010-05-03 2013-03-13 F. Hoffmann-La Roche AG Compositions et méthodes de diagnostic et de traitement d'une tumeur
CA2802873C (fr) 2010-06-16 2018-09-18 Dynavax Technologies Corporation Methodes et traitements a l'aide d'inhibiteurs de tlr7 et/ tlr9
EP2654772B1 (fr) * 2010-12-22 2018-09-26 The Salk Institute for Biological Studies Peptides cycliques antagonistes de la corticolibérine
GB2503604B (en) 2011-03-21 2020-04-22 Biodesy Llc Classification of kinase inhibitors using second harmonic optical techniques
RU2014109395A (ru) 2011-09-15 2015-10-20 Дженентек, Инк. Способы стимуляции дифференциации
JP6254087B2 (ja) 2011-10-15 2017-12-27 ジェネンテック, インコーポレイテッド 癌を治療するためのscd1アンタゴニスト
EP2804630B1 (fr) 2012-01-18 2017-10-18 F. Hoffmann-La Roche AG Procédés d'utilisation de modulateurs fgf19
US20130209473A1 (en) 2012-02-11 2013-08-15 Genentech, Inc. R-spondin translocations and methods using the same
RU2014141617A (ru) 2012-03-16 2016-05-10 Дженентек, Инк. Сконструированные конформационно-стабилизированные белки
US9139863B2 (en) 2012-03-16 2015-09-22 Genentech, Inc. Engineered conformationally-stabilized proteins
BR112014020173A8 (pt) 2012-03-16 2017-07-11 Hoffmann La Roche Métodos para o tratamento de um melanoma, utilizações de um inibidor, composições, conjunto, método de inibição, método de identificação, método de ajuste do tratamento e invenção
CN104583776B (zh) 2012-04-25 2016-09-07 比奥德赛公司 用于检测蛋白质的变构调节剂的方法
WO2013170191A1 (fr) 2012-05-11 2013-11-14 Genentech, Inc. Procédés d'utilisation d'antagonistes de biosynthèse de nicotinamide adénine dinucléotide à partir de nicotinamide
US9228184B2 (en) 2012-09-29 2016-01-05 Dynavax Technologies Corporation Human toll-like receptor inhibitors and methods of use thereof
EP4356960A2 (fr) 2013-03-15 2024-04-24 F. Hoffmann-La Roche AG Biomarqueurs et méthodes de traitement d'états associés à pd-1 et pd-l1
WO2015116902A1 (fr) 2014-01-31 2015-08-06 Genentech, Inc. Récepteurs couplés aux protéines g dans la signalisation hedgehog
KR20170005016A (ko) 2014-05-23 2017-01-11 제넨테크, 인크. MiT 바이오마커 및 그의 사용 방법
CN114057857A (zh) 2014-06-20 2022-02-18 豪夫迈·罗氏有限公司 基于chagasin的支架组合物、方法和应用
WO2016106286A1 (fr) 2014-12-23 2016-06-30 Biodesy, Inc. Fixation de protéines à des interfaces destinées à être utilisées en détection optique non linéaire
EP3509616A1 (fr) 2016-09-09 2019-07-17 H. Hoffnabb-La Roche Ag Inhibiteurs peptidiques sélectifs de frizzled
WO2018152496A1 (fr) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions et méthodes de diagnostic et de traitement de l'infection par le virus zika
EP4230649A3 (fr) 2017-04-25 2023-10-25 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Anticorps et procédés de diagnostic et de traitement d'infection par le virus d'epstein barr
US11827669B2 (en) 2017-07-19 2023-11-28 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis and treatment of hepatitis b virus infection
CN113795514A (zh) 2019-05-09 2021-12-14 豪夫迈·罗氏有限公司 制备抗体的方法
CN112480209B (zh) * 2020-11-16 2022-03-04 昆明学院 一种抗皮肤光损伤保护活性多肽rl-pl9及其应用

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991006006A1 (fr) * 1989-10-19 1991-05-02 Yamasa Shoyu Kabushiki Kaisha Excipient de liaison d'anticorps antiphospholipides, immuno-analyse utilisant cet excipient et kit associe
US5552391A (en) * 1990-01-16 1996-09-03 La Jolla Pharmaceutical Company Chemically-defined non-polymeric valency platform molecules and conjugates thereof
US5268454A (en) * 1991-02-08 1993-12-07 La Jolla Pharmaceutical Company Composition for inducing humoral anergy to an immunogen comprising a t cell epitope-deficient analog of the immunogen conjugated to a nonimmunogenic carrier
US5498538A (en) * 1990-02-15 1996-03-12 The University Of North Carolina At Chapel Hill Totally synthetic affinity reagents
ATE127925T1 (de) * 1990-04-06 1995-09-15 Yamasa Shoyu Kk Verfahren zur bestimmung von antiphospholipid antikörpern.
WO1991015722A1 (fr) * 1990-04-10 1991-10-17 Masur, Walter Processus de fonctionnement d'une thermopompe a compression et thermopompe a compression
EP1808183A3 (fr) * 1993-09-08 2009-07-01 La Jolla Pharmaceutical Company Molécules à plate-forme de valence non polymères, définies chimiquement et leurs conjugués
US5874409A (en) * 1995-06-07 1999-02-23 La Jolla Pharmaceutical Company APL immunoreactive peptides, conjugates thereof and methods of treatment for APL antibody-mediated pathologies

Similar Documents

Publication Publication Date Title
AU734638B2 (en) aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies
US6207160B1 (en) aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies
WO1997046251A9 (fr) PEPTIDES IMMUNOREACTIFS PAR RAPPORT AUX aPL, LEURS CONJUGUES ET PROCEDES DE TRAITEMENT DES PATHOLOGIES DEPENDANTES DE L'ANTICORPS aPL
WO1996040197A9 (fr) PEPTIDES A IMMUNOREACTION aPL, LEURS CONJUGUES, ET METHODES DE TRAITEMENT DES PATHOLOGIES INDUITES PAR LES ANTICORPS aPL
AU772851B2 (en) Therapeutic and diagnostic domain 1 beta2GPI polypeptides and methods of using same
US7517657B2 (en) Peptides for detecting anti-double stranded DNA antibody and uses thereof
US6410775B1 (en) APL immunoreactive peptides, conjugates thereof and methods of treatment for APL antibody-mediated pathologies
CZ277198A3 (cs) Imunogenní peptidy, způsob jejich přípravy, farmaceutický prostředek, který je obsahuje a jejich použití
CN1950394B (zh) 在受试者中抑制免疫复合物形成的方法
US6455244B1 (en) Methods for the detection of antibodies associated with autoimmune disorders and infectious agents employing immunoretroid peptides derived from antigens associated with said disorders and agents
US7053178B2 (en) Synthetic peptides and kits for diagnosis of anti-phospholipid syndrome
US7074888B1 (en) Mimotopes and anti-mimotopes of human platelet glycoprotein Ib/IX
EP1106625A1 (fr) Séquences péptidiques spécifiques pour rhesus D
AU2004201699A1 (en) Therapeutic and diagnostic domain 1 Beta2GPI polypeptides and methods of using same
CN1225015A (zh) aPL免疫反应性肽、其缀合物以及aPL抗体介导的疾病的治疗方法
MXPA00012040A (en) THERAPEUTIC AND DIAGNOSTIC DOMAIN 1&bgr;2