CN117147824A - Antibody conjugate and application thereof - Google Patents

Antibody conjugate and application thereof Download PDF

Info

Publication number
CN117147824A
CN117147824A CN202310508396.XA CN202310508396A CN117147824A CN 117147824 A CN117147824 A CN 117147824A CN 202310508396 A CN202310508396 A CN 202310508396A CN 117147824 A CN117147824 A CN 117147824A
Authority
CN
China
Prior art keywords
antibody
linker
peg
click chemistry
antibody conjugate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310508396.XA
Other languages
Chinese (zh)
Inventor
钟志荣
陈彬
林芳博
王静文
党海滨
刘青昀
孟媛
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fapon Biotech Inc
Original Assignee
Fapon Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fapon Biotech Inc filed Critical Fapon Biotech Inc
Priority to PCT/CN2023/096265 priority Critical patent/WO2023231889A1/en
Publication of CN117147824A publication Critical patent/CN117147824A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/10General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using coupling agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Analytical Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention discloses an antibody conjugate and application thereof, and relates to the technical field of antibody coupling. The invention provides a technology for directional modification of disulfide bonds of antibodies, which utilizes a linker to efficiently couple the antibodies and conjugation partners, and the prepared antibody conjugate has good uniformity, has obvious advantages of high detection sensitivity, good stability and the like compared with the antibody conjugate prepared by a conventional process, and provides a way for development and application of diagnostic reagents.

Description

Antibody conjugate and application thereof
Technical Field
The invention relates to the technical field of antibody coupling, in particular to an antibody conjugate and application thereof.
Background
The diagnostic field generally uses EDC, NHS or Maleimide (MAL) to couple carboxyl, amino or sulfhydryl groups of proteins, and this protein coupling technology has two problems, affecting the sensitivity and precision of reagents. First, randomness of the coupling sites: for example, an IgG1 type antibody contains an average of 80 lysines, 20 of which are in solvent accessible positions, and some of which are in the antigen recognition region of Fab. Thus, when amino groups of an antibody are used as coupling sites, the antibody conjugate is heterogeneous and the antibody is inactivated due to the random distribution of amino groups in the antibody. Second, the coupling efficiency is low: the secondary reaction kinetics of NHS with amino group was 10 -1 ~10 2 M -1 S -1 When the equilibrium is reached, the yield of the conjugate is 40%, the yield of the product is low, the waste of raw materials is caused, and the sensitivity of the reagent is affected.
The secondary reaction kinetics of maleimide and mercapto reaches 10 2 ~10 3 M -1 S -1 The yield of the conjugate was 80%, but the cysteine residues of the antibody were present as disulfide bonds with little free thiol accessible to the solvent. Another approach is to use Traut's reagent (2-iminothiolanePentane) converts the amino group of an antibody into a thiol group, but there are problems of low conversion efficiency and randomness of coupling sites.
Disclosure of Invention
The invention aims to provide an antibody conjugate and application thereof.
The invention is realized in the following way:
in a first aspect, embodiments of the present invention provide an antibody conjugate suitable for immunodiagnosis comprising the structure:
n, m are integers and are independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24.
In which the coupling means between the conjugation partner and the O atom in the linker is not limited, the conjugation partner may be coupled to the O atom with some of its own groups, or may be coupled to the O atom via other intermediate groups, preferably via pairs of orthogonal reactive groups, such as pairs of click chemistry groups as described in the second aspect of the invention, but this does not mean that the coupling means with click chemistry groups is necessary. The coupling is carried out for the purpose of ligation, however, by other coupling means, including but not limited to pairs of NHS (N-hydroxysuccinimide) and amino, or pairs of MAL (maleimide) and thiol, but at a slower reaction rate and possibly more by-products, but the skilled person will be able to adjust the same end product by subsequent purification and isolation.
In a second aspect, embodiments of the present invention provide an antibody conjugate suitable for immunodiagnosis comprising the structure:
from left to right are conjugation partner, linker and antibody, respectively;
wherein the R is 1 For the first click chemistry group, the R 2 N, m are each integers and are independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24, for the second click chemistry group.
In a third aspect, embodiments of the present invention provide an antibody-linker suitable for immunodiagnosis comprising the structure:
the linker and the antibody are respectively from left to right;
wherein R is 1 N, m are integers and are independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24.
The number of linkers in the structure of the antibody conjugate or antibody-linker of the invention is not limited, and one or more linkers may be present. That is, the antibody may have multiple disulfide bonds, each of which may be independently reduced to a thiol group and then reformed with a linker to a thio-carbon bridge, such that one antibody may be linked to multiple linkers, each of which may in turn be independently linked to a conjugation partner, i.e., form a (conjugation partner-linker) x-antibody structure, x being an integer greater than or equal to 1.
For the "conjugation partner-R" according to the invention 2 (also written as R 2 -conjugation partner) ", or" conjugation partner-R comprised in the structure of the antibody conjugate according to the invention 2 (also written as R 2 -conjugation partner) "unit, R 2 The bar "-" between the conjugate partner is not limited to R 2 Whether or not to be directly bound to the conjugation partner, R 2 Can directly bond and link partners, R 2 The conjugation partner can be linked indirectly via small molecule chemical building blocks, or via macromolecular biological materials, as demonstrated later by experimental dataSome do not affect the detection sensitivity of the final prepared antibody conjugate.
For the antibody conjugates of the invention, the number of conjugation partners attached to the same linker in the structure is not limited, and the same linker may be attached to one or more conjugation partners. That is, the conjugation partner may be indirectly connected to the linker through an amplifying carrier to form a (conjugation partner) y-amplifying carrier-linker-antibody structure, y is an integer greater than or equal to 1, and one amplifying carrier may be connected to a plurality of conjugation partners, so that the same linker may be connected to a plurality of conjugation partners, and the function of amplifying the detection signal of the conjugation antibody to the sample is achieved. If the antibody is also linked to a plurality of linkers at this time, an (((conjugation partner) y-amplified carrier-linker) x-antibody structure is formed, x and y are integers greater than or equal to 1, and the y values of each of the x (conjugation partner) y-amplified carrier-linker structures are independent of each other.
In a fourth aspect, embodiments of the present invention provide a method of preparing an antibody conjugate as described in the previous embodiments, comprising: antibody-linker and R 2 -performing an addition reaction of the conjugation partner to obtain an antibody conjugate; the antibody-linker has the following structure:
n, m are integers and are independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24.
In a fifth aspect, embodiments of the invention provide the use of an antibody conjugate or antibody-linker as described in the previous embodiments for the preparation of a diagnostic reagent or kit.
In a sixth aspect, embodiments of the invention provide a kit comprising an antibody conjugate or antibody-linker as described in the previous embodiments.
The invention has the following beneficial effects:
the invention provides a technology for performing directional modification on disulfide bonds of antibodies for couplingWhich comprises reacting 2- (tosyl) methacrylamide- (PEG) m -amide- (PEG) n -R 1 ,
The following is shownThe antibody conjugate prepared by the method has the obvious advantages of high detection sensitivity, good stability and the like compared with the antibody conjugate prepared by the conventional process, and provides a path for development and application of diagnostic reagents.
Drawings
In order to more clearly illustrate the technical solutions of the embodiments of the present invention, the drawings that are needed in the embodiments will be briefly described below, it being understood that the following drawings only illustrate some embodiments of the present invention and therefore should not be considered as limiting the scope, and other related drawings may be obtained according to these drawings without inventive effort for a person skilled in the art.
FIG. 1 is a schematic diagram of the reconstitution of an antibody disulfide bond after reduction with a reducing agent;
FIG. 2 shows the results of the disulfide reduction amounts RDAR (lanes 1 to 3) and the disulfide reduction amounts (lanes 4 to 6) of the antibodies after reconnection by TSMA based on gel electrophoresis;
FIG. 3 is a graph showing the effect of TCEP concentration on the number of disulfide reductions in antibodies based on gel electrophoresis;
FIG. 4 is a chromatogram of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine;
FIG. 5 is a mass spectrum of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine;
FIG. 6 is a mass spectrometry peak analysis chart of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine;
FIG. 7 is a nuclear magnetic resonance carbon spectrum of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine;
FIG. 8 is a nuclear magnetic resonance hydrogen spectrum of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine.
Detailed Description
In order to make the objects, technical solutions and advantages of the embodiments of the present invention more clear, the technical solutions of the embodiments of the present invention will be clearly and completely described below. The specific conditions are not noted in the examples and are carried out according to conventional conditions or conditions recommended by the manufacturer. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention.
Embodiments of the present invention provide an antibody conjugate comprising the following structure:
wherein the R is 1 For the first click chemistry group, the R 2 N, m are each integers and are independently selected from 0 to 24, preferably m+n=0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24, preferably m is non-zero, preferably n is non-zero, preferably both m and n are non-zero, preferably m+n is ≡4.
The antibody conjugate provided by the invention consists of a conjugation partner, a linker and an antibody, wherein R is modified on the conjugation partner 2 The linker is 2- (tosyl) methacrylamide- (PEG) m -amide- (PEG) n -R 1 It is as follows:
R 1 and R is 2 Is an orthogonal pair of click chemistry groups that can be linked to each other based on a click chemistry reaction, and wherein the 2- (tosyl) methacrylamide structure is a reconstituted cross-linker that can re-bridge the free sulfhydryl groups on the antibody together to form a carbon-sulfur bond of S-C-S, see figure 1. The number of PEG in the linker, i.e., the value of m+n is preferably 0 to 24, the number of PEG in the linker being capable of adjusting the separation distance between the conjugation partner and the antibody,too close a spacing will affect the spatial structure of the antibody and too far a spacing will affect the stability of the antibody conjugate.
In some embodiments, n, m are each integers and are independently selected from 0-24, and may specifically be any one or any range between any two of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24. The commonly used reconstruction crosslinking agent and click chemical groups are organic compounds with strong hydrophobicity, and the disulfide bond position of the water-soluble antibody is difficult to enter. For example, the divinyl sulfonamide disclosed in CN1107400072B as a reconstitution cross-linking agent is poorly water soluble, affecting the efficiency of disulfide reconstitution. According to the invention, PEG molecules are introduced into the reconstruction crosslinking agent through design, so that the water solubility is improved, the disulfide bond reconstruction efficiency is increased, and the detection signal intensity of the final conjugated product is greatly improved.
In some embodiments, the sulfur-carbon bridge in the structural formula of the antibody conjugate is located between antibody hinge regions, between antibody light and heavy chains, or between antibody heavy chains. The heavy chains of the antibody include CH1 and Fc regions.
In some embodiments, the conjugation partner is indirectly linked to the linker through an amplification carrier selected from at least one of glycans, polylysines, protein carriers, PEG (polyethylene glycol), polyethylenimine, and dendrimers. Specifically, glycans include cross-linked polysucroses, glucans; the protein carrier comprises at least one of bovine serum albumin, human serum albumin, ovalbumin, keyhole limpet hemocyanin and thyroglobulin; the dendritic polymer comprises polyethylene glycol and/or polypropylene imine.
The antibodies described herein are to be construed in the broadest sense and may include full length monoclonal antibodies, bispecific or multispecific antibodies, as well as chimeric antibodies and antigen-binding fragments of antibodies, so long as they exhibit the desired biological activity and possess disulfide bonds that crosslink with the 2- (tosyl) methacrylamide structure after being reduced to form a sulfur-carbon bridge.
In some embodiments, R 1 And R is 2 Click chemistry groups that are capable of orthogonal pairing. Point(s)Click chemistry is an orthogonal ligation reaction, and there are two general categories: the first is copper-catalyzed azide-alkyne cycloaddition (CuAAC) or copper-free-catalyzed azide-dibenzocyclooctyne strain-promoted cycloaddition (SPAAC); the second is tetrazine-trans-cyclooctene reverse electron demand addition reaction, which is the most rapid orthogonal reaction found at present, and the second-order reaction kinetics can reach 30000M -1 S -1 . The stability of tetrazine with stronger electron withdrawing group is lower than that of hydrogen substituted tetrazine and lower than that of methyl substituted tetrazine. The hydrogen-substituted tetrazine shows exceptionally fast kinetics (10000M -1 S -1 ) Typically at least 10 times faster than methyl substituted tetrazines. Therefore, a balance needs to be struck in the selection of tetrazine groups in terms of reaction rate and stability. The inventors found that 1mg/mL of methyltetrazine modified IgG antibody and TCO modified AP (alkaline phosphatase) can be completely converted into IgG-AP conjugate within 90min, and that methyltetrazine modified IgG antibody has only 10-20% reduced activity when left for one month at 4 ℃. According to these results and in combination with the application scenario in the IVD field, methyltetrazine (MTz) and trans-cyclooctene (TCO) are preferred as click chemistry orthogonal pairs in the present invention, and other click chemistry orthogonal pairs can achieve similar effects.
In some embodiments, the first click chemistry group and/or the second click chemistry group is selected from the group consisting of: methyl tetrazine (MTz), trans-cyclooctene (TCO), azide (N) 3 ) Any one of Dibenzocyclooctyne (DBCO), tetrazine (Tz), alkyne, cyclopropane cyclooctyne (BCN), and cyclopropene.
Preferably, the first click chemistry group and the second click chemistry group are each selected from any one of methyltetrazine (MTz) and trans-cyclooctene (TCO).
The addition reaction process of TCO and MTz is as follows:
antibodies and conjugation partners modified with click chemistry groups can be rapidly crosslinked to form antibody conjugates. The antibody conjugate prepared by the invention is better than the conventional process, and mainly has the advantages of high yield, clear coupling sites, good uniformity and the like. Furthermore, the antibody conjugate of the present invention is used as a detection kit, and is superior to conventional antibody conjugates in terms of sensitivity and correlation.
In some embodiments, the conjugation partner is a label selected from at least one of a fluorescent dye, an enzyme, a radioisotope, a chemiluminescent reagent, and a nanoparticle-based label; preferably, the fluorescent dye is at least one selected from fluorescein dye and its derivative, rhodamine dye and its derivative, cy series dye and its derivative, alexa series dye and its derivative, and protein dye and its derivative; preferably, the enzyme is selected from any one of horseradish peroxidase, alkaline phosphatase, beta-galactosidase, glucose oxidase, carbonic anhydrase, acetylcholinesterase, and glucose-6-phosphate deoxygenase; preferably, the radioisotope is selected from At least one of 212Bi, 131I, 111In, 90Y, 186Re, 211At, 125I, 1188Re, 153Sm, 213Bi, 32P, 94mTc, 99mTc, 203Pb, 67Ga, 68Ga, 43Sc, 47Sc, 110 msin, 97Ru, 62Cu, 64Cu, 67Cu, 68Cu, 86Y, 88Y, 121Sn, 161Tb, 166Ho, 105Rh, 177Lu, 172Lu and 18F; preferably, the chemiluminescent reagent is selected from at least one of luminol and its derivatives, lucigenin, crustacean fluorescein and its derivatives, ruthenium bipyridine and its derivatives, acridinium esters and its derivatives, dioxane and its derivatives, roflumilast and its derivatives, and peroxyoxalate and its derivatives; preferably, the nanoparticle label is selected from any one of colloid, organic nanoparticle, quantum dot nanoparticle and rare earth complex nanoparticle; preferably, the colloid is selected from at least one of colloidal metal, disperse dye, dye-labeled microsphere and latex; preferably, the colloidal metal is selected from at least one of colloidal gold, colloidal silver and colloidal selenium.
An embodiment of the present invention provides a method for preparing an antibody conjugate according to any of the preceding embodiments, comprising: antibody-linker and R 2 The addition reaction of the conjugation partners to obtain antibody conjugationA material; the antibody-linker has the following structure:
in some embodiments, the antibody-linker is attached to R 2 -the molar ratio of the conjugation partners is (0.5-4): (0.5 to 4), specifically, 0.5: 4. 0.5:3.5, 0.5:3. 0.5:2.5, 0.5:2. 0.5:1.5, 0.5:1. 1:1. 1: 4. 1:3.5, 1:3. 1:2.5, 1:2. 1:1.5, 2: 4. 2:3.5, 2:3. 2:2.5, 2:1.5, 2:1. 2:1. 3: 4. 3:3.5, 3:2.5, 3:2. 3:1.5, 3:1. 3:1. 4:3.5, 4:3. 4:2.5, 4:2. 4:1.5, 4:1 or any one or any two of them.
In some embodiments, the reaction conditions of the addition reaction include: 4-37 ℃ for 0.5-16 h; alternatively, the reaction temperature of the addition reaction may specifically be in a range between any one or any two of 4 ℃, 6 ℃, 8 ℃, 10 ℃, 12 ℃, 14 ℃, 16 ℃, 18 ℃, 20 ℃, 22 ℃, 24 ℃, 26 ℃, 28 ℃, 30 ℃, 32 ℃, 34 ℃, 36 ℃, 37 ℃; the reaction time of the addition reaction may specifically be in a range between any one or any two of 0.5h, 1h, 1.5h, 2h, 2.5h, 3h, 3.5h, 4h, 4.5h, 5h, 5.5h, 6h, 6.5h, 7h, 7.5h, 8h, 8.5h, 9h, 9.5h, 10h, 10.5h, 11h, 11.5h, 12h, 12.5h, 13h, 13.5h, 14h, 14.5h, 15h, 15.5h, and 16 h.
In some embodiments, the method of making further comprises making an antibody-linker: mixing the linker with the antibody with disulfide bond opened by reduction to obtain an antibody-linker; the linker has the following structure:
the mixed reaction of the linker and the antibody with disulfide bond reduced open is as follows:
in some embodiments, the molar ratio of linker to disulfide bond reduced opened antibody is (1-30): 1. the molar ratio can be specifically 1: 1. 2: 1. 4: 1. 6: 1. 8: 1. 10: 1. 12: 1. 14: 1. 16: 1. 18: 1. 20: 1. 22: 1. 24: 1. 26: 1. 28: 1. 30:1 or any one or any two of them.
In some embodiments, the reaction conditions for an antibody in which the linker and disulfide bond are reduced open include: 2-37 ℃ and 0.5-48 h. The reaction temperature may specifically be in a range of any one or any two or more of 2 ℃, 4 ℃, 6 ℃, 8 ℃, 10 ℃, 12 ℃, 14 ℃, 16 ℃, 18 ℃, 20 ℃, 22 ℃, 24 ℃, 26 ℃, 28 ℃, 30 ℃, 32 ℃, 34 ℃, 36 ℃ and 37 ℃; the reaction time of the reaction may specifically be any one of 0.5h, 1h, 2h, 3h, 4h, 5h, 6h, 7h, 8h, 9h, 10h, 11h, 12h, 13h, 14h, 15h, 16h, 17h, 18h, 19h, 20h, 21h, 22h, 23h, 24h, 25h, 26h, 27h, 28h, 29h, 30h, 31h, 32h, 33h, 34h, 35h, 36h, 37h, 38h, 39h, 40h, 41h, 42h, 43h, 44h, 45h, 46h, 47h, 48h or a range between any two of them.
In some embodiments, the method of preparing further comprises preparing an antibody in which the disulfide bond is reduced to open: reducing the antibody under the action of a reducing agent to obtain the antibody with disulfide bonds opened by reduction.
In some embodiments, the reducing agent comprises at least one of 2-MEA and TCEP.
2-Mercaptoethylamine hydrochloride (2-Mercaptotoethylamine. HCl) is 2-MEA for short, is a mild reducing agent, and can specifically reduce disulfide bonds of a hinge region of an antibody, but not disulfide bonds of other sites of the antibody. TCEP is another mild disulfide reducing agent, and at a certain TCEP concentration, for example IgG, the sequence of disulfide reduction of the antibody is: light and heavy chain disulfide bond > upper hinge region disulfide bond > lower hinge region disulfide bond > intra-chain disulfide bond.
In some embodiments, the use concentration of the 2-MEA comprises at least one of 3eq, 5eq, and 10 eq.
In some embodiments, the molar ratio of the reducing agent to the antibody is (0.5-50): 1. the molar ratio may specifically be 0.5: :1. 1: 1. 5: 1. 7: 1. 9: 1. 11: 1. 13: 1. 15: 1. 20: 1. 25: 1. 30: 1. 35: 1. 40: 1. 45: 1. 50:1 or any one or any two of them.
For ease of presentation, "antibody-specific click chemistry groups" are used hereinafter to refer to antibody-linker structures of the invention bearing specific click chemistry groups.
In some embodiments, the method of making further comprises R 2 Preparation of the conjugation partner comprising reacting R 2 Mixed reaction of NHS ester and conjugation partner to obtain R 2 -a conjugation partner.
In some embodiments, R 2 The molar ratio of NHS ester to conjugation partner is (5-25): 1. the molar ratio can be specifically 5: 1. 7: 1. 9: 1. 11: 1. 13: 1. 15: 1. 17: 1. 19: 1. 21: 1. 23: 1. 25: 1. any one or any two of the ranges.
In some embodiments, R 2 The reaction conditions for the NHS ester and the conjugation partner include: 10-30 ℃ and 0.5-1.5 h. The reaction temperature may specifically be in a range between any one or any two of 10 ℃, 12 ℃, 14 ℃, 16 ℃, 18 ℃, 20 ℃, 22 ℃, 24 ℃, 26 ℃, 28 ℃, 30 ℃; the reaction time of the reaction may specifically be in a range between any one or any two of 0.5h, 0.7h, 0.9h, 1.1h, 1.3h, and 1.5h.
In some embodiments, the method of making further comprises making the linker: TSMA and R 1 -(PEG) n -NHS ester mix reaction, TSMA being 2- (tosyl) methacrylamide- (PEG) m -an amide of the formula:
in some embodiments, the TSMA and R 1 -(PEG) n The molar ratio of the NHS ester is (0.5-1.5): (0.5-1.5). The molar ratio may specifically be 0.5:1.5, 0.5:1. 0.5:1:1:0.5, 1.5:1 or any one or any two of them.
In some embodiments, the TSMA and R 1 -(PEG) n The reaction conditions for the NHS esters include: 10-30 ℃ and 10-14 h. The reaction temperature may specifically be in a range between any one or any two of 10 ℃, 12 ℃, 14 ℃, 16 ℃, 18 ℃, 20 ℃, 22 ℃, 24 ℃, 26 ℃, 28 ℃, 30 ℃; the reaction time of the reaction may specifically be in a range between any one or any two of 10h, 10.5h, 11h, 11.5h, 12h, 12.5h, 13h, 13.5h, and 14h.
In some embodiments, the TSMA and R 1 -(PEG) n -reacting NHS ester in organic solvent or water.
In some embodiments, the organic solvent of the organic phase comprises methylene chloride.
In some embodiments, the TSMA and R 1 -(PEG) n The NHS ester is reacted under the action of DIEA and/or triethylamine.
In some embodiments, the molar ratio of DIEA to TSMA is 1: (1-4). The molar ratio can be specifically 1: 1. 1: : 2. 1: 3. 1:4 or a range between any one or any two of them.
In some embodiments, the molar ratio of triethylamine to TSMA is 1: (1-4). The molar ratio can be specifically 1: 1. 1: 2. 1: 3. 1:4 or a range between any one or any two of them.
In some embodiments, the method of preparing further comprises preparing TSMA:
taking a compound A and a compound B as starting materials, and reacting the compound A with the compound B to obtain a compound C;
carrying out substitution reaction on the compound C and the compound D to obtain TSMA;
wherein the compound A is tert-butoxycarbonyl-imino-polyethylene glycol-amine, and has the structural formula:the compound B is methacrylic acid +.>Methacryloyl halide->(wherein X is halogen) and methacrylic anhydride +.>At least one of (a) and (b); the compound C is tert-butyloxycarbonyl-imino-polyethylene glycol-methacrylamide and has the structural formula ofThe compound D is 4-toluenesulfonyl chloride->And sodium 4-toluene sulfinate>At least one of (a) and (b); wherein m is 0 to 24. The specific selection of m is the same as that of the foregoing corresponding embodiment, and will not be repeated.
In some embodiments, the molar ratio of compound a to compound B is (0.35 to 0.85): (0.58-0.98); the molar ratio may specifically be 0.45:0.58, 0.45:0.78, 0.45:0.98, 0.35:0.58, 0.35:0.78, 0.35:0.98, 0.65:0.58, 0.65:0.78, 0.65:0.98, 0.85:0.58, 0.85:0.78, 0.85:0.98, or any one or any two thereof.
In some embodiments, the reaction conditions of compound a and compound B include: stirring for 10-14 h at 20-30 ℃; the reaction temperature may specifically be in a range between any one or any two of 20 ℃, 22 ℃, 24 ℃, 26 ℃, 28 ℃, 30 ℃; the reaction time of the reaction may specifically be in a range between any one or any two of 10h, 10.5h, 11h, 11.5h, 12h, 12.5h, 13h, 13.5h, and 14 h.
In some embodiments, the reaction of compound a and compound B comprises: dissolving the compound A in a first organic solvent, adding the compound B, reacting under the action of a dehydrating agent and/or a hydroxyl activating agent, removing the solvent, filtering to remove the precipitate, and purifying to obtain the compound C.
In some embodiments, the first organic solvent comprises at least one of DMF and dichloromethane;
in some embodiments, the dehydrating agent comprises dicyclohexylcarbodiimide.
In some embodiments, the hydroxyl activator comprises N-hydroxysuccinimide.
In some embodiments, the molar ratio of compound C to compound D is (0.33-0.73): (0.60-1.00). The molar ratio may specifically be 0.33:0.60, 0.33:0.80, 0.33:1.00, 0.53:0.60, 0.53:0.80, 0.53:1.00, 0.73:0.60, 0.73:0.80, 0.73:1.00, or any one or any two thereof.
In some embodiments, the reaction conditions of the substitution reaction include: stirring for 20-28 h at 20-30 ℃. The reaction temperature may specifically be in a range between any one or any two of 20 ℃, 22 ℃, 24 ℃, 26 ℃, 28 ℃, 30 ℃; the reaction time of the reaction may specifically be in a range between any one or any two of 20h, 21h, 22h, 23h, 24h, 25h, 26h, 27h, 28h.
In some embodiments, the substitution reaction comprises: dissolving the compound C in a second organic solvent, and adding the compound D for reaction; adding triethylamine after the reaction, and stirring for 10-14 h; the stirring time may specifically be in a range between any one or any two of 10h, 11h, 12h, 13h, and 14 h.
Washing and drying the stirred product, and removing the solvent to obtain a crude product; dissolving the crude product in ethyl acetate, adding triethylamine for refluxing, removing the solvent by rotary evaporation, and purifying to obtain the compound E.
In some embodiments, the second organic solvent comprises methylene chloride.
In some embodiments, the washing employs a detergent that is at least one of 1M HCl, saturated sodium bicarbonate solution, saturated sodium thiosulfate solution, and saturated sodium chloride solution.
The embodiment of the invention provides a connector compound, which has the structure of 2- (toluenesulfonyl) methacrylamide- (PEG) m -amide- (PEG) n- R 1
Wherein m and n are integers and are independently selected from 0 to 24, preferably m+n=0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24; preferably m+n=4, preferably m is not zero, preferably n is not zero, preferably neither m nor n is zero;
R 1 for the first click chemistry group, it can be combined with a second click chemistry group R 2 Is rapid and convenient to couple; and the 2- (tosyl) methacrylamide structure can be connected with two free sulfhydryl groups in a reaction way to form a sulfur-carbon bridge structure.
It will be appreciated that the specific selection of the linker compound is the same as that described in the corresponding examples above, and will not be repeated.
The present invention provides a method for preparing a linker according to any of the preceding embodiments, comprising the preparation of TSMA according to any of the preceding embodiments and/or the preparation of a linker according to any of the preceding embodiments.
Embodiments of the present invention provide the use of an antibody conjugate as described in any of the preceding embodiments in the preparation of a diagnostic reagent or kit.
Furthermore, embodiments of the present invention provide the use of an antibody conjugate as described in any of the preceding embodiments in the preparation of a reagent or kit for improving detection sensitivity.
The features and capabilities of the present invention are described in further detail below in connection with the examples.
The development thinking of the invention is to anchor disulfide bonds of antibodies, firstly reduce and open the disulfide bonds to obtain two free sulfhydryl groups, and then connect the sulfhydryl groups with a heavy bridging reagent to form sulfur-carbon bridge bonds. The main common heavy bridging reagents used in the prior art are bissulfone compounds, maleimide compounds, pyridazine compounds and divinyl sulfonamide compounds. Also reported in small amounts are allyl sulfones, bromopyridine diones, divinyl pyridines, N-substituted-3-bromo-5-methylenepyrrol-2-ones, and the like. The invention designs an allyl sulfone compound joint, and discovers that the allyl sulfone compound joint is used for coupling an antibody to prepare a labeled antibody, and the obtained labeled antibody has unexpected sensitivity effect in immunodetection.
Example 1
2- (tosyl) methacrylamide- (PEG) m -amide and 2- (tosyl) methacrylamide- (PEG) m -amide- (PEG) n -R 1 Is prepared by the following steps.
The chemical reagents used in the synthesis were purchased from the microphone reagent net unless otherwise specified.
This example illustrates the preparation of the title compound by taking m=4, whereas in practice the title compound with other values of m (including positive integers of zero) is obtained correspondingly by varying the number of PEG units in compound a used in the preparation (other compounds a with different values of m are also available from the microphone reagent network).
The following example describes the preparation of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine.
(1) 2- (tosyl) methacrylamide- (PEG) 4 Preparation of the amide (TSMA)
a. Boc-imino-tetra-polyethylene glycol-amine (Compound A,200mg,0.65 mmol) was dissolved in 10mL DMF (N, N-dimethylformamide), methacrylic acid (Compound B,67mg,0.78 mmol), DCC (dicyclohexylcarbodiimide, 193mg,0.94 mmol) and NHS (N-hydroxysuccinimide, 119mg,1.03 mmol) were added and magnetically stirred at 25℃for 12 hours.
b. The solvent was removed by rotary evaporation, and the crude product was dissolved in 10mL of deionized water, the precipitate was removed by filtration, and after the solvent was removed by rotary evaporation, it was purified by column chromatography (ethyl acrylate: n-hexane=1:1) to give t-butoxycarbonyl-imino-tetra polyethylene glycol-methacrylamide (compound C) in about 90% yield.
In the above steps, methacrylic acid may be replaced by, but is not limited to, for example, methacrylic acid halides (e.g., methacryloyl chloride, methacryloyl bromide, methacryloyl fluoride, etc.) or methacrylic anhydride. When using methacrylic acid halides or methacrylic anhydride, the synthesis procedure is as follows:
a. boc-imino-tetrapolyethylene glycol-amine (Compound A,200mg,0.65 mmol) was dissolved in 10mL of dichloromethane, triethylamine (78.8 mg, 108. Mu.L, 0.78 mmol), methacryloyl halide or methacrylic anhydride (0.78 mmol) was added at 0deg.C, and magnetically stirred for 12 hours.
b. The solvent was removed by rotary evaporation, the crude product was dissolved in 30mL of dichloromethane, washed with 1M HCl and saturated sodium chloride solution, and the organic layer was dried over anhydrous sodium sulfate, and then the solvent was removed by rotary evaporation. Purification using column chromatography (ethyl acrylate: n-hexane=1:1) gave t-butoxycarbonyl-imino-tetra polyethylene glycol-methacrylamide (compound C) in about 90% yield.
a. Compound C (200 mg,0.53 mmol) was dissolved in 5mL of dichloromethane and 4-toluenesulfonyl chloride (compound D,152mg,0.80 mmol) was added and magnetically stirred at 25℃for 24 hours.
b. Triethylamine (162 mg, 223. Mu.L, 1.60 mmol) was then added and stirred for 12 hours. The crude product was washed with 1M HCl, saturated sodium bicarbonate solution, saturated sodium chloride solution, dried over anhydrous magnesium sulfate and the solvent was removed by rotary evaporation.
c. The crude product was dissolved in 10mL ethyl acetate, triethylamine (162 mg, 223. Mu.L, 1.60 mmol) was added at 0deg.C and refluxed for 12 hours. After the solvent was removed by rotary evaporation, purification was performed by column chromatography (ethyl acrylate: n-hexane=3:1) to give t-butoxycarbonyl-2- (tosyl) methacrylate (compound E) in a yield of about 70%.
In the above step, 4-toluenesulfonyl chloride may be replaced by, but not limited to, sodium 4-toluenesulfonate. When sodium 4-toluene sulfinate is used, steps a and b in the above synthesis steps are as follows:
a. compound C (200 mg,0.53 mmol) was dissolved in 5mL of dichloromethane, sodium 4-toluene sulfinate (143 mg,0.80 mmol) and iodine (204 mg,0.80 mmol) were added and magnetically stirred at 25℃for 72 hours.
b. Triethylamine (162 mg, 223. Mu.L, 1.60 mmol) was then added and stirred for 12 hours. The crude product was washed with 1M HCl, saturated sodium bicarbonate solution, saturated sodium thiosulfate solution, saturated sodium chloride solution, dried over anhydrous magnesium sulfate and the solvent was removed by rotary evaporation.
And c, the step is the same.
(3) Compound E (26.7 mg,0.05 mmol) was dissolved in 5mL of dichloromethane, trifluoroacetic acid (278 mg,2.50 mmol) was added and magnetically stirred at 25℃for 12 hours, and the solvent was removed by rotary evaporation to give TSMA in about 98% yield.
(2) Preparation of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine (TSMA-MTz)
(1) TSMA (13 mg,0.03 mmol) was dissolved in 2mL of dichloromethane, DIEA (N, N-diisopropylethylamine, 7.5mg,0.06 mmol) or triethylamine (6.1 mg, 8.4. Mu.L, 0.06 mmol) was added and stirred to dissolve. Methyl tetrazine-PEG 4-NHS ester (Compound F, purchased from Siam Kang Funuo Biotech Co., ltd., 16.7mg,0.03 mmol) was dissolved in 1mL of methylene chloride and added to the above solution of TSMA after homogenization.
(2) The mixture was magnetically stirred at 25 ℃ for 12 hours, the solvent was removed by rotary evaporation, the crude product was dissolved in 20mL of dichloromethane, washed with saturated sodium chloride solution, dried over anhydrous magnesium sulfate, and purified by column chromatography (dichloromethane: methanol=20:1) after removal of the solvent by rotary evaporation to give 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine (compound G, TSMA-MTz) in about 50% yield.
The detection results of the liquid chromatography-mass spectrometry combined detection and nuclear magnetic resonance detection are shown in figures 4-8, which prove that the method is truly successful in preparing the 2- (toluenesulfonyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine.
The above is an example of a process for the preparation of the title compound with n=4, and in practice methyl tetrazine-PEGn-NHS esters containing varying numbers of PEG units can be used to synthesize title compounds with varying values of n (positive integers including zero). But also the methyltetrazine herein may be replaced by other tetrazine groups such as tetrazine (as used herein, various click chemistry-PEGn-NHS compounds are available from western amp Kang Funuo biotechnology limited unless otherwise indicated). For example, when the preparation is carried out, referring to the previous operation steps, other synthesis steps are unchanged, and only methyl tetrazine-PEG 4-NHS ester is required to be respectively replaced by methyl tetrazine-NHS ester, tetrazine-PEG 4-NHS ester and tetrazine-NHS ester with equal molar weight, so that 2- (toluenesulfonyl) methacrylamide-PEG 4-amide-methyl tetrazine, 2- (toluenesulfonyl) methacrylamide-PEG 4-amide-PEG 4-tetrazine and 2- (toluenesulfonyl) methacrylamide-PEG 4-tetrazine can be respectively synthesized.
The compounds of this example can also be synthesized in the aqueous phase, with the following exemplary steps being performed during the aqueous phase synthesis:
TSMA (13 mg,0.03 mmol) was dissolved in 2mL deionized water, pH adjusted to 7.0-8.0, and stirred well. Methyl tetrazine-PEG 4-NHS ester (Compound F, purchased from Siam Kang Funuo Biotech Co., ltd., 16.7mg,0.03 mmol) or equimolar amount of tetrazine-PEG 4-NHS ester was dissolved in 1mL of deionized water, and added to the above solution of TSMA after homogenization. Magnetically stirring the mixture at 25 ℃ for 12 hours to obtain the aqueous solution of 2- (toluenesulfonyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine or 2- (toluenesulfonyl) methacrylamide-PEG 4-amide-PEG 4-tetrazine.
(3) Preparation of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4- (4E) -trans-cyclooctene (TSMA-TCO).
(1) TSMA (13 mg,0.03 mmol) was dissolved in 2mL of dichloromethane, DIEA (N, N-diisopropylethylamine, 7.5mg,0.06 mmol) or triethylamine (6.1 mg, 8.4. Mu.L, 0.06 mmol) was added and stirred to dissolve. (4E) -trans-cyclooctene-PEG 4-NHS ester (purchased from Siam Kang Funuo Biotech Co., ltd., 15.4mg,0.03 mmol) was dissolved in 1mL of methylene chloride and added to the above solution of TSMA after homogenization.
(2) The mixture was magnetically stirred at 25 ℃ for 12 hours, the solvent was removed by rotary evaporation, the crude product was dissolved in 20mL of dichloromethane, washed with saturated sodium chloride solution, dried over anhydrous magnesium sulfate, and after removal of the solvent by rotary evaporation, purified using column chromatography (dichloromethane: methanol=20:1) to give 2- (tosyl) methacrylamide-PEG 4- (4E) -trans-cyclooctene (TSMA-TCO).
The above is an illustration of the preparation of the title compound with n=4, using (4E) -trans-cyclooctene-PEGn-NHS esters containing different numbers of PEG units to synthesize 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4- (4E) -trans-cyclooctene with different values of n, and (4E) -trans-cyclooctene can also be replaced by (2E) -trans-cyclooctene. For example, when the preparation is carried out, referring to the previous steps, other synthesis steps are unchanged, and only (4E) -trans-cyclooctene-PEG 4-NHS ester is replaced by (4E) -trans-cyclooctene-NHS ester and (2E) -trans-cyclooctene-PEG 4-NHS ester with equal molar weight respectively, so that 2- (tosyl) methacrylamide-PEG 4-amide- (4E) -trans-cyclooctene and 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4- (2E) -trans-cyclooctene can be respectively synthesized.
The compounds of this example can also be synthesized in the aqueous phase. Exemplary steps in the aqueous phase synthesis are as follows:
TSMA (13 mg,0.03 mmol) was dissolved in 2mL deionized water, pH adjusted to 7.0-8.0, and stirred well. (4E) -trans-cyclooctene-PEG 4-NHS ester (purchased from Siam Kang Funuo Biotech Co., ltd., 15.4mg,0.03 mmol) or an equimolar amount of (2E) -trans-cyclooctene-PEG 4-NHS ester was dissolved in 1mL of deionized water and added to the above solution of TSMA after homogenization. Magnetically stirring the mixture at 25 ℃ for 12 hours to obtain an aqueous solution of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4- (4E) -trans-cyclooctene or 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4- (2E) -trans-cyclooctene.
(4) Preparation of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-azide (TSMA-N3)
(1) TSMA (13 mg,0.03 mmol) was dissolved in 2mL of dichloromethane, DIEA (N, N-diisopropylethylamine, 7.5mg,0.06 mmol) or triethylamine (6.1 mg, 8.4. Mu.L, 0.06 mmol) was added and stirred to dissolve. azide-PEG 4-NHS ester (0.03 mmol) was dissolved in 1mL dichloromethane and added to the above solution of TSMA after homogenization.
(2) The mixture was magnetically stirred at 25 ℃ for 12 hours, the solvent was removed by rotary evaporation, the crude product was dissolved in 20mL of dichloromethane, washed with saturated sodium chloride solution, dried over anhydrous magnesium sulfate, and after removal of the solvent by rotary evaporation, purified using column chromatography (dichloromethane: methanol=20:1) to give 2- (tosyl) methacrylamide-PEG 4-azide (TSMA-N3).
The above is an illustration of the preparation of the title compound of this example using n=4, and in practice azide-PEGn-NHS esters containing varying amounts of PEG units (here azide-PEGn-NHS esters are customizable from Shanghai biotechnology company) to synthesize 2- (tosyl) methacrylamide-PEG 4-amide-PEGn-azides with varying values of n. For example, when the preparation is carried out, referring to the previous steps, other synthesis steps are unchanged, and only azide-PEG 4-NHS ester is required to be respectively replaced by azide-PEG 2-NHS ester and azide-PEG 8-NHS ester with equal molar quantity, so that 2- (tosyl) methacrylamide-PEG 4-amide-PEG 2-azide and 2- (tosyl) methacrylamide-PEG 4-amide-PEG 8-azide can be respectively synthesized.
The compounds of this example can also be synthesized in the aqueous phase. Exemplary steps in the aqueous phase synthesis are as follows:
TSMA (13 mg,0.03 mmol) was dissolved in 2mL deionized water, pH adjusted to 7.0-8.0, and stirred well. azide-PEG 4-NHS ester (0.03 mmol) was dissolved in 1mL deionized water and added to the TSMA solution after homogenization. The mixture was magnetically stirred at 25℃for 12 hours to obtain an aqueous solution of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-azide.
(5) Preparation of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-DBCO (TSMA-DBCO)
(1) TSMA (13 mg,0.03 mmol) was dissolved in 2mL of dichloromethane, DIEA (N, N-diisopropylethylamine, 7.5mg,0.06 mmol) or triethylamine (6.1 mg, 8.4. Mu.L, 0.06 mmol) was added and stirred to dissolve. DBCO-PEG4-NHS ester (purchased from Siemens Kang Funuo Biotechnology Co., ltd., 0.03 mmol) was dissolved in 1mL of methylene chloride and added to the above solution of TSMA after homogenization.
(2) The mixture was magnetically stirred at 25 ℃ for 12 hours, the solvent was removed by rotary evaporation, the crude product was dissolved in 20mL of dichloromethane, washed with saturated sodium chloride solution, dried over anhydrous magnesium sulfate, and after the solvent was removed by rotary evaporation, purified using column chromatography (dichloromethane: methanol=20:1) to give 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-DBCO (TSMA-DBCO).
The above is a method for preparing the title compound of this example using n=4 as an example, and actually using DBCO-PEGn-NHS esters with different numbers of PEG units to synthesize 2- (tosyl) methacrylamide-PEG 4-amide-PEGn-DBCO with different values of n. For example, when preparing, referring to the previous steps, other synthesis steps are unchanged, only the DBCO-PEGn-NHS ester is replaced by DBCO-NHS ester and DBCO-PEG2-NHS ester with equal molar amounts respectively, and 2- (tosyl) methacrylamide-PEG 4-DBCO and 2- (tosyl) methacrylamide-PEG 4-amide-PEG 2-DBCO can be obtained by synthesis respectively, and the raw materials can be purchased from Siemens Kang Funuo biotechnology Co. .
The compounds of this example can also be synthesized in the aqueous phase. Exemplary steps in the aqueous phase synthesis are as follows: TSMA (13 mg,0.03 mmol) was dissolved in 2mL deionized water, pH adjusted to 7.0-8.0, and stirred well. DBCO-PEG4-NHS ester (purchased from Siemens Kang Funuo Biotechnology Co., ltd., 0.03 mmol) was dissolved in 1mL deionized water and added to the above solution of TSMA after homogenization. The mixture was magnetically stirred at 25℃for 12 hours to obtain an aqueous solution of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-DBCO.
Example 2
Preparation of click chemistry group modified conjugation partners.
For the click chemistry group modified alkaline phosphatase prepared by way of example in this example, there was a PEG4 spacer between the click chemistry group and the alkaline phosphatase, but in practice the click chemistry group and alkaline phosphatase may be directly linked or may be linked by some spacer, and experimental data later demonstrate that the presence or absence of a spacer between the click chemistry group and alkaline phosphatase has no substantial effect on the detected signal intensity of the final antibody conjugate.
(1) Preparation of alkaline phosphatase-trans-cyclooctene (AP-TCO)
Zeba for alkaline phosphatase (5 mg/mL) TM The desalting column (10K MWCO) was replaced into 100mM PBS buffer pH 7.4. 10mM TCO-PEG4-NHS (10 eq, DMSO dissolved) was added to 40uM alkaline phosphatase solution and reacted at 25℃for 1 hour. Then, the excess TCO-PEG4-NHS was used as Zeba TM Desalting treatment was performed with a desalting column (10K MWCO) and replaced with PB (50 mM, pH 7.4) buffer. AP-TCO was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4) buffer, and stored at 4℃for further use.
(2) Preparation of alkaline phosphatase-methyltetrazine (AP-MTz)
Zeba for alkaline phosphatase (5 mg/mL) TM The desalting column (10K MWCO) was replaced into 100mM PBS buffer pH 7.4. 10mM methyltetrazine-PEG 4-NHS (10 eq, DMSO dissolved) was added to 40uM alkaline phosphatase solution and reacted at 25℃for 1 hour. Then, the excess methyltetrazine-PEG 4-NHS was treated with Zeba TM Desalting treatment was performed with a desalting column (10K MWCO) and replaced with PB (50 mM, pH 7.4) buffer. AP-MTz was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4) buffer, and stored at 4℃for further use.
(3) Preparation of alkaline phosphatase-dibenzocyclooctyne (AP-DBCO)
Zeba for alkaline phosphatase (5 mg/mL) TM The desalting column (10K MWCO) was replaced into 100mM PBS buffer pH 7.4. 10mM DBCO-PEG4-NHS (10 eq, DMSO dissolved) was added to 40uM alkaline phosphatase solution and reacted at 25℃for 1 hour. Then, the excess DBCO-PEG4-NHS was treated with Zeba TM Desalting treatment was performed with a desalting column (10K MWCO) and replaced with PB (50 mM, pH 7.4) buffer. AP-DBCO was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4) buffer, and stored at 4deg.C for further use.
(4) Preparation of acridinium ester-trans-cyclooctene (AE-TCO)
In the embodiment of the invention, the acridinium ester and the trans-cyclooctene are simultaneously modified on a protein or chemical macromolecule carrier, and the acridinium ester is indirectly connected with the trans-cyclooctene through the carrier, however, the acridinium ester-trans-cyclooctene (AE-TCO) can also be directly prepared through a chemical synthesis mode. In this embodiment, the protein carrier may be Bovine Serum Albumin (BSA), alkaline Phosphatase (AP), chicken Ovalbumin (OVA), hemocyanin (KLH), etc.; the polymeric carrier may be dextran or polylysine. Further, the inventors have improved the crosslinking ratio of AE on the carrier, and can improve the sensitivity of the reagent. Meanwhile, by controlling the amount of TCEP, it is possible to achieve one antibody linked to 4 AEs or carrier-AEs, enabling a significant improvement in sensitivity. The modification of acridinium esters is described below with BSA as an example, and the modification of other carriers is similar.
Bovine serum albumin (5 mg/mL) was dissolved in 100mM PBS buffer pH7.4, 10mM TCO-PEG4-NHS (10 eq, DMSO in solution) was added, and reacted at 25℃for 1 hour. Excess TCO-PEG4-NHS Zeba TM Desalting was performed with a desalting column (10K MWCO) and replaced with 100mM PBS buffer pH 7.4. Then 10mM acridinium ester-NHS (20 eq, DMSO dissolved) was added and reacted at 25℃for 1 hour. Zeba for excess acridinium ester-NHS TM Desalting treatment was performed with a desalting column (10K MWCO) and replaced with PB (50 mM, pH 7.4) buffer. BSA-TCO-AE was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4) buffer, and stored at 4℃for further use.
Example 3
Reduction of disulfide bonds of antibodies.
Examples of the invention IgG1, igG2a, igG2b murine monoclonal antibodies were selected for investigation, wherein IgG 1-type antibodies include AFP antibodies, CA153 antibodies, CA724 antibodies, CA199 antibodies, and HIV P24 antibodies; the IgG2a type antibody is CA125 antibody; the IgG2b antibody is cTnI antibody.
Two free sulfhydryl groups can be obtained by reduction at the disulfide bond of the antibody, then a linker molecule with a 2- (tosyl) methacrylamide structure is added, and the two free sulfhydryl groups are connected again into a sulfur-carbon bridge bond through the connection reaction of the 2- (tosyl) methacrylamide and the two free sulfhydryl groups. The above antibodies have 2 to 4 disulfide bonds in the hinge region and disulfide bonds in front of the light and heavy chains, and the reduction process can be adjusted so that disulfide bonds are more likely to be reduced in the hinge region of the antibody or between the light and heavy chains of the antibody.
(1) Reduction of disulfide bonds in antibody hinge regions
Zeba for antibody (5 mg/mL) TM The desalting column (10K MWCO) was replaced in PBS (100 mM PB,50mM sodium chloride, pH7.4,1mM EDTA) buffer. 2-MEA (2-mercaptoethylamine hydrochloride) was prepared as a 10mM solution in PBS (100 mM,50mM sodium chloride, pH7.4,1mM EDTA). 10eq of 2-MEA was added to the antibody solution and reacted at 25℃for 1 hour with gentle shaking (400 rpm). The reduced antibody was kept at 4℃for further use.
(2) Reduction of disulfide bonds between light and heavy chains
Zeba for antibody (5 mg/mL) TM The desalting column (10K MWCO) was replaced into PB (50mM,pH7.4,1mM EDTA) buffer. TCEP (Sigma) was prepared as a 10mM solution with PB (50mM,pH7.4,1mM EDTA). To the antibody solution was added 2eq of TCEP and reacted at 37℃for 2 hours with gentle shaking (400 rpm). The reduced antibody was kept at 4℃for further use.
Example 4
Reconstruction of antibody disulfide bonds.
(1) Introduction of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-methyltetrazine (TSMA-MTz) linker molecules
TSMA-MTz was formulated under dry conditions with DMSO as a 10mM solution. To the reduced antibody solution was added 20eq of TSMA-MTz and reacted at 4℃for 16 hours. Zeba for excess chemical reagent in antibody solutions TM The desalting column (10K MWCO) was replaced into PB (50 mM, pH 7.4) buffer. The antibody-methyltetrazine (antibody-MTz) was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4), and stored at 4℃for further use.
(2) Introduction of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-trans-cyclooctene (TSMA-TCO) linker molecules
TSMA-TCO is formulated in dry condition with DMSO as a 10mM solution. To the reduced antibody solution was added 20eq of TSMA-TCO and reacted at 4℃for 16 hours. Excess of antibody solution Zeba for chemical reagents of (a) TM The desalting column (10K MWCO) was replaced into PB (50 mM, pH 7.4) buffer. The antibody-trans-cyclooctene (antibody-TCO) was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4), and stored at 4℃for further use.
(3) Introduction of 2- (tosyl) methacrylamide-PEG 4-amide-PEG 4-azide (TSMA-N3) linker molecules
TSMA-N3 was formulated under dry conditions with DMSO as a 10mM solution. To the reduced antibody solution was added 20eq of TSMA-N3 and reacted at 4℃for 16 hours. Zeba for excess chemical reagent in antibody solutions TM The desalting column (10K MWCO) was replaced into PB (50 mM, pH 7.4) buffer. Antibody-azide (antibody-N3) was prepared, diluted to 4mg/mL with PB (50 mM, pH 7.4), and stored at 4℃for further use.
Example 5
Preparation of conjugation partner-directed cross-linked antibody conjugates.
(1) Antibody-methyltetrazine (4 mg/mL) to alkaline phosphatase-trans-cyclooctene (4 mg/mL), molar ratio 1:1, and reacting at 25 ℃ for one hour to obtain the alkaline phosphatase-labeled antibody conjugate of the invention. After the reaction, the mixture is kept at 4 ℃ for standby.
(2) Antibody-trans-cyclooctene (4 mg/mL) to alkaline phosphatase-methyltetrazine (4 mg/mL), molar ratio of 1:1, and reacting at 25 ℃ for one hour to obtain the alkaline phosphatase-labeled antibody conjugate of the invention. After the reaction, the mixture is kept at 4 ℃ for standby.
(3) Antibody-azide (4 mg/mL) to alkaline phosphatase-DBCO (4 mg/mL), molar ratio of 1:1, and reacting at 25 ℃ for one hour to obtain the alkaline phosphatase-labeled antibody conjugate of the invention. After the reaction, the mixture is kept at 4 ℃ for standby.
(4) Antibody-methyltetrazine (4 mg/mL) to acridinium ester-trans-cyclooctene (4 mg/mL), molar ratio 1:1, and reacting at 25 ℃ for one hour to obtain the acridinium ester-labeled antibody conjugate of the invention. After the reaction, the mixture is kept at 4 ℃ for standby.
Comparative example 1
A method for preparing a conventional process antibody conjugate is provided, in particular as follows.
The conventional preparation principle of antibody conjugates in the IVD field is to use amino or sulfhydryl groups of amino acid side chains of proteins as coupling sites.
The method for preparing alkaline phosphatase antibody conjugate by conventional process is as follows:
(1) Thiol modification of antibodies (IgG-SH):
zeba for antibody (5 mg/mL) TM The desalting column (10K MWCO) was replaced with PBS (100 mM PB,50mM sodium chloride, pH8.0,5mM EDTA). Traut's reagent (Pierce) was prepared as a 10mM solution in PBS (100 mM PB,50mM sodium chloride, pH7.4,1mM EDTA). To the antibody solution, 10eq of Traut's reagent was added and the reaction was allowed to proceed for 2 hours with gentle shaking (400 rpm) at 25 ℃. Desalting to remove excessive reagent, and storing the sulfhydrylation modified antibody at 4deg.C for use.
(2) Maleimide modification of alkaline phosphatase (AP-MAL):
zeba for alkaline phosphatase (5 mg/mL) TM The desalting column (10K MWCO) was replaced in PBS (10 mM PB,50mM sodium chloride, pH7.4,5mM EDTA) buffer. 10mM SMCC (10 eq, DMSO dissolved) was added to 40uM alkaline phosphatase solution and reacted at 25℃for 1 hour. Desalting to remove excessive reagent, and storing maleimide modified AP at 4deg.C for use.
(3) IgG-SH and AP-MAL crosslinking:
thiol-modified antibodies (4 mg/mL) and maleimide-modified AP (4 mg/mL) were reacted in PBS (10 mM PB,50mM sodium chloride, pH 7.4) buffer at 25℃for 1 hour. After the reaction, the mixture is kept at 4 ℃ for standby.
The method for preparing the acridinium ester antibody conjugate by the conventional process is as follows:
zeba for antibody (5 mg/mL) TM The desalting column (10K MWCO) was replaced in PBS (10 mM PB,50mM sodium chloride, pH7.4,5mM EDTA) buffer. 10mM NHS-AE (10 eq, DMSO dissolved) was added to 30uM antibody solution and reacted at 25℃for 1 hour. After the reaction was completed, the excess reagent was removed by desalting and stored at 4℃for further use.
Verification example 1
Reduction and modification of the antibody hinge region was verified.
2-Mercaptoethylamine hydrochloride (2-Mercapoethylamine. HCl), abbreviated as 2-MEA, is a mild reducing agent that can specifically tend to reduce disulfide bonds in the hinge region of an antibody, but not in other sites of the antibody.
AFP monoclonal antibody (murine IgG1 type) was reduced with three concentrations of 2-MEA 10eq, 5eq, 3eq (procedure see example 3). The reduction specificity of 3eq of 2-MEA was found to be optimal, only one hinge disulfide was opened, and the disulfide reduction number RDAR of the antibody was 1.1 (FIG. 2 and Table 1).
Table 1 disulfide bond reduction number RDAR of antibodies
Under the reduction of 10eq and 5eq of 2-MEA, disulfide bonds between light and heavy chains of AFP antibody are opened to different degrees, and the disulfide bond reduction amount RDAR of the antibody is 3.4 and 2.5 (sequence numbers 1 to 2).
The three reduced AFP antibodies are subjected to TSMA-MTz linker molecules, and opened disulfide bonds are reconnected to form a sulfur-carbon bridge (the process is described in example 4), so that a complete antibody is formed again, the reduction number of the disulfide bonds of the antibodies is 0.01-0.02 (sequence numbers 4-6), and the fact that most of the broken disulfide bonds of the antibodies form sulfur-carbon bridge again is shown.
Verification example 2
Three antibodies labeled with MTz in verification example 1 were each crosslinked with alkaline phosphatase (procedure of example 5) and prepared as AFP detection reagents, and a sensitivity test was performed. The detection results showed that the sensitivity of the low value samples (CL) was 23.2% and 26.7% higher for 3eq of the 2-MEA prepared antibody conjugate compared to 5eq and 10eq of the 2-MEA, respectively (table 2).
TABLE 2AFP project sensitivity test
This result suggests that the disulfide bond in the antibody hinge region is a better coupling site than the disulfide bond between the light and heavy chains of the antibody. The conjugation partner is coupled at the disulfide bond position of the antibody hinge region with less impact on the structure and function of the antibody.
Compared with the conventional process, the antibody conjugate prepared by the embodiment of the invention has the advantages that the sensitivity of a low-value sample (CL) is 127%, 120% and 179% respectively, and the antibody conjugate has obvious advantages.
Verification example 3
Control of disulfide bond reduction sites.
Mouse IgG has three subtypes, igG1, igG2a and IgG2b, with the different subtypes differing in the number of interchain disulfide bonds. The number of interchain disulfide bonds for both IgG2a and IgG2b is 6, while IgG1 is 4. Murine IgG antibodies of both the CA153 (IgG 1) and cTnI (IgG 2 b) subtypes were selected for study.
CA153 and cTnI antibodies at a concentration of 1mg/mL were split into 7 aliquots. TCEP was formulated with PB (50mM,pH7.4,1mM EDTA) as a 10mM solution. To 7 parts of antibody solution, 0eq,5eq,10eq,15eq,20eq,30eq and 50eq of TCEP solution were added, respectively, and the reaction was carried out at 37℃for 2 hours with gentle shaking (400 rpm). After the reaction, protein samples were taken for SDS-PAGE and quantitative analysis of disulfide bond reduction of antibodies. The results are shown in FIG. 3.
The results show that TCEP concentration and the number of antibody disulfide reductions (RDAR) are positively correlated, the higher the TCEP concentration, the greater the number of antibody disulfide reductions. By adjusting the concentration of TCEP, the number of reduction of disulfide bonds between antibody chains can be controlled. At a TCEP concentration of 10eq, the number of disulfide open of the IgG1 antibody was 3.1 and the number of disulfide open between IgG2b antibody chains was 1.6.
Further, the CA153 antibody and the cTnI antibody reduced with 10eq of TECP were crosslinked with alkaline phosphatase (TSMA-MTz of example 1 was used). The resulting antibody conjugates were subjected to sensitivity tests, the results of which are shown in table 3.
TABLE 3 sensitivity of different items
Table 3 shows that the sensitivity of the low value samples (CL) was 96% and 220% higher, respectively, using cTnI antibody conjugates and CA153 antibody conjugates prepared by the inventive technique as compared to antibody conjugates prepared by conventional techniques. Meanwhile, the difference in the improvement of the sensitivity of the two antibodies suggests that the sensitivity and the number of disulfide bond reduction (RDAR) of the antibodies are positively correlated. I.e., the greater the number of disulfide reductions, the greater the sensitivity.
Verification example 4
Click chemistry reaction orthogonality assessment.
AFP monoclonal antibody (IgG 1), 10eq TCEP reduced AFP monoclonal antibody, 20eq TSMA-MTz after 10eq TCEP reduction (example 1) reconstituted AFP monoclonal antibody was crosslinked with AP-TCO, molar ratio 1: after 1, 25℃for one hour, 4℃was kept ready for use.
Meanwhile, 20eq TSMA-MTz reconstructed AFP monoclonal antibody after 10eq TCEP reduction is crosslinked with AP, and the molar ratio is 1: after 1, 25℃for one hour, 4℃was kept ready for use.
IgG-SH and AP-MAL were prepared according to the conventional procedure, in a molar ratio of 1:1, after reacting for one hour at 25 ℃,4 ℃ is kept for standby.
5 AFP monoclonal antibody conjugates are prepared, diluted into conjugate working solution according to a certain proportion, and matched with magnetic bead working solution to form a detection reagent for sensitivity test. From the sensitivity test results (table 4).
TABLE 4 evaluation of click chemistry reaction orthogonality
The antibody conjugate obtained by crosslinking the AP-TCO with the IgG and the reduced IgG of the TCEP has no activity, the antibody conjugate obtained by preparing the AP and the antibody-MTz has no signal, and only the antibody conjugate obtained by preparing the AP-TCO and the antibody-MTz has activity, so that the result shows the orthogonality of click chemistry groups. Meanwhile, the click chemical group does not react with amino, sulfhydryl or carboxyl and other biological active groups of the protein, and has good biocompatibility.
Verification example 5
Disulfide bond reconstruction oriented crosslinker technology versatility evaluation.
2 IgG 1-type antibodies and 1 IgG2a project were tested to assess the versatility of the technology provided by the examples of the present invention in the field of immunodiagnosis.
The monoclonal antibodies of the two items CA724 and HIV P24 are selected by IgG1 type, the monoclonal antibody of the item CA125 is selected by IgG2a type, the alkaline phosphatase antibody conjugate (adopting a linker TSMA-MTz) is prepared by the technology of the invention, and the alkaline phosphatase antibody conjugate and the respective magnetic bead coating are matched to form a detection reagent for sensitivity test. From the sensitivity test results (table 5).
TABLE 5 evaluation of disulfide bond reconstruction Directional crosslinking technique commonality
TABLE 6 evaluation of disulfide bond reconstruction Directional crosslinking technique commonality
The IgG2a type antibody CA125 project can improve the sensitivity of a low-value sample (CL) by 108.1% by applying the technology of the invention. The IgG1 antibody CA724 program can improve the sensitivity of low-value sample (CL) by 63.9% by applying the technology of the invention. The detection sensitivity of the P24 antigen of the HIV fourth-generation detection reagent can improve the window period of detection, and has important significance for clinical diagnosis of HIV. As can be seen from Table 6, the technology of the invention is applied to detect 1.25IU/mL international standard (NIBSC, national institute of biological products, UK), the P/N ratio reaches 30.3, and the sensitivity can be improved by 105.3% compared with the conventional technology.
Verification example 6
Disulfide bond reduction sites are related to sensitivity and correlation.
From the results of the foregoing verification, it is clear that the hinge region disulfide bond is a better coupling site than the light-heavy interchain disulfide bond. CA153 antibody was reduced with TCEP and 2-MEA at 10eq concentration, respectively, and then after reconstitution of disulfide bonds with 20eq TSMA-MTz (example 1), crosslinking was performed by adding AP-TCO, the molar ratio of antibody-MTz to AP-TCO being 1:1. the 2 directionally cross-linked conjugates were prepared and subjected to SEC-HPLC analysis, sensitivity and correlation tests along with conjugates prepared by conventional techniques.
TABLE 7CA153 project sensitivity
The conjugates prepared by both TCEP and 2-MEA reduction, i.e., light and heavy chain disulfide and hinge region disulfide reconstitution, were prepared with lower value samples (CL) having sensitivities 189% and 200% higher, respectively, than conventional processes (table 7). Hinge region disulfide bond reconstruction is less sensitive than light-heavy chain disulfide bond reconstruction.
The antibody conjugate is prepared by the three different processes to form detection reagents, 57 cases of Roche clinical samples with the concentration range of 4.22-367.4U/mL are respectively tested, the samples with each concentration are detected for 1 time, and the test data are shown as the supplementary material 1. And (3) performing linear regression analysis and data fitting by taking the RLU as a y-axis variable and taking the concentration value of the Roche clinical sample as an x-axis variable through testing by a Fenpeng instrument. The conjugates obtained with TCEP reducing antibody and 2-MEA reducing antibody were of the conventional technology and had correlation coefficients 0.9524, 0.9257, 0.9789, respectively (Table 7). Hinge region disulfide reconstruction has significant advantages over light-heavy chain disulfide reconstruction in correlation.
SEC-HPLC analysis shows that the yields of the conventional process, TCEP reduction process and 2-MEA reduction process are 43.78%, 87.67% and 91.16%, respectively. Antibody conjugate yields are consistent with the sensitivity results. The antibody conjugates obtained by conventional processes are a mixture of dimers, trimers and tetramers, and the antibody conjugates obtained by the technique of the invention are a trimer-based homogeneous mass.
Verification example 7
2- (tosyl) methacrylamide- (PEG) m -amide- (PEG) n MTz linker has the effect of varying amounts of PEG on detection sensitivity.
Six values of m+n=0, 2, 3, 4, 8, and 16 were synthesized, and the linker of the title compound was coupled to a CA153 antibody to give CA153 antibody-MTz, and then coupled to trans-cyclooctene-AP to give a CA153 antibody-AP conjugate (see example 5), and then a detection sensitivity test was performed on a CA153 sample. The test results showed that the P/N ratios of the m+n=0, 2, 3, 4, 8, 16 six CA153 antibody-AP conjugates in the test CA153 low value samples (CL) were 1.4, 1.9, 3.6, 3.1, 2.1 (table 8), respectively, where C0 represents the background value samples, CL represents the low value samples, CH represents the high value samples, and the same applies.
TABLE 8 sensitivities of the linker to different PEG amounts (CA 153)
In the absence of PEG molecules, the linker has low solvent accessibility, low disulfide bond reconstitution efficiency, and significantly reduced sensitivity. Too many PEG molecules affect the structure and stability of the antibody conjugate, which is detrimental to the immune response and reduces the sensitivity. The most suitable number of PEG molecules for the linker is between 4 and 8, with 4 being optimal.
Example 8 is demonstrated the effect of the presence of a spacer arm between the conjugation partner and the click chemistry group on the detection sensitivity.
TCO modification is carried out on alkaline phosphatase by TCO-NHS and TCO-PEG4-NHS respectively to obtain AP-PEG4-TCO and AP-TCO, and crosslinking is carried out on the AP-TCO and the AP-TCO respectively with CA153 antibody-MTz of verification example 7, wherein the molar ratio is 1:1. two antibody-AP conjugates were prepared with PEG molecules between the conjugation partner and click chemistry groups in numbers of 0 and 4, respectively, and sensitivity tests were performed on CA153 samples. Table 9 the test results show that the sensitivity of the antibody-AP conjugates with PEG molecules numbers 0 and 4 between the conjugation partner and the click chemistry group, respectively, was substantially consistent when testing CA153 samples at different levels, indicating whether there was a spacer between the conjugation partner and the click chemistry group that did not significantly affect the detection sensitivity of the antibody conjugate.
TABLE 9 influence of the presence or absence of spacer arms between conjugation partners and click chemistry groups on sensitivity (CA 153)
Verification example 9
Evaluation of the thermostability of the antibody conjugate obtained by disulfide bond reconstruction technique.
CA242 antibody was reduced with 10eq of 2-MEA and 10eq of TCEP, respectively, followed by disulfide bond reconstitution with 20eq of TSMA-MTz (same as in example 4) to give antibody-MTz, which was then reacted with AP-TCO in a molar ratio of 1:1 to obtain the antibody-AP conjugate. The two antibody-AP conjugates prepared by the invention and the antibody-AP conjugate prepared by the conventional process are respectively diluted by a conjugation partner diluent to prepare the enzyme-labeled working solution. Three different enzyme-labeled working solutions are respectively and evenly divided into three parts, and one part is preserved at the temperature of 2-8 ℃ to be used as a control sample. The other two parts are placed in a constant temperature box at 37 ℃ to be used as test samples, and the test samples are taken out and stored at 2-8 ℃ on the 3 rd day and the 7 th day. On the last day, all samples and the magnetic bead working solution form detection reagents, three samples with different concentrations of CA242 are tested, each sample is tested for 2 times, a relative luminescence value (RLU) is obtained, and a relative luminescence value average value is calculated. The relative deviation of the test and control groups was calculated.
Table 10 evaluation of the thermal stability of antibody conjugates obtained by disulfide reconstruction technique (CA 242)
From the experimental results (table 10), the conventional process antibody conjugates were accelerated for 3 days and 7 days, with low value sample (CL) luminescence value deviations of-12% and 16%, respectively. The directional crosslinking antibody conjugate prepared by the 2-MEA and the TCEP is accelerated for 3 days and the luminous value deviation of 7 days is less than 10 percent. The antibody conjugate obtained by the technology has better thermal acceleration stability than the conventional technology.
Verification example 10
Acridinium ester-labeled antibody conjugate detection sensitivity evaluation.
Sensitivity tests were carried out on the prepared acridinium ester antibody conjugate 1 (no amplifying carrier exists between the acridinium ester and the linker) and the prepared acridinium ester antibody conjugate 2 (amplifying carrier exists between the acridinium ester and the linker), and the sensitivity of the low-value sample (CL) is 7.4, 17.1 and 26.8 respectively. Compared with the conventional process, the sensitivity of the acridinium ester marker 1 and the acridinium ester marker 2 prepared by the method is respectively 130% and 262%. The 57% difference in sensitivity between the acridinium ester label 1 and the acridinium ester label 2 for detecting the low-value sample shows that the performance of the final antibody conjugate can be further improved obviously by adding the minor scheme of carrier amplification to the major technical scheme of the invention.
TABLE 11 sensitivity of acridinium ester markers (CA 153)
Performance testing
The antibody conjugate of the invention and the antibody conjugate of the conventional process are diluted by enzyme-labeled diluent to prepare enzyme-labeled working solution which is matched with magnetic bead working solution to form detection reagents, and the detection reagents are tested on a Phpenishine series full-automatic chemiluminescence immunoassay analyzer to evaluate the sensitivity, the correlation and the stability of the detection reagents.
(1) Sensitivity of
Internal references C0, CL, CH for different projects were tested, each sample was tested twice to obtain relative luminescence values (RLUs), and RLU mean values were calculated. Sensitivity (P/N) was calculated by the ratio of the RLU mean of CL and CH to the RLU mean of CO.
(2) Correlation of
The cobas 411 and the fei pengshine analyzers were calibrated with their own calibrants, and then at least 40 correlated samples were tested, once for each sample, to obtain relative luminescence values (RLUs). The result of the Fit system is taken as a Y-axis variable, and the result of the Roche system is taken as an X-axis variable. The data were fitted using linear regression analysis to obtain the equation y=kx+b, correlation coefficient r.
Analysis of physicochemical Properties
(1)SDS-PAGE
A5 Xloading Buffer (2.5 mM pH7.8 Tris-HCl,0.1%SDS,0.005% bromophenol blue) was added to the protein sample to prepare a protein electrophoresis sample. Using surePAGE TM Protein preps (gold) and Tris-MOPS-SDS Running Buffer Powder (gold) were subjected to SDS-PAGE of protein electrophoresis samples.
(2)SEC-HPLC
And performing size exclusion analysis on the protein sample by using a Waters high performance liquid chromatography platform. The operation is carried out according to the instructions of the manufacturer instrument, the concentration of the protein sample is not lower than 0.1mg/mL, and the total protein is not lower than 50ug.
(3) Determination of the amount of disulfide bond reduction of antibody
The molar concentration of free thiol of the antibody was quantitatively determined using Ellman reagent (DTNB, pierce) and the number of disulfide bond reductions RDAR (Reduced disulfide bonds antibody ratio) of the antibody was determined by the ratio to the molar concentration of the antibody. OD value was measured at a wavelength of 412nm, and molar extinction coefficient ε412 of thiol was 10mM-1cm-1. The molar extinction coefficient ε280 of the antibody was 210mM-1cm-1 when the 0D value was measured at a wavelength of 280 nm. According to Lambert-Beer law: aλ=epsilonλbc, aλ is the ultraviolet absorbance of the sample at λ wavelength, epsilonλ is the molar extinction coefficient of the sample at λ wavelength, b is the optical path, and C is the molar concentration of the sample. The number of disulfide reduction RDAR of the antibody can be calculated by the following formula:
the above description is only of the preferred embodiments of the present invention and is not intended to limit the present invention, but various modifications and variations can be made to the present invention by those skilled in the art. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (10)

1. An antibody conjugate comprising the structure:
wherein n and m are integers and are independently selected from 0 to 24.
2. An antibody conjugate comprising the structure:
wherein R is 1 R is a first click chemistry group 2 And n and m are integers and are independently selected from 0 to 24.
3. An antibody-linker comprising the structure:
wherein R is 1 N and m are integers and are independently selected from 0 to 24.
4. The antibody conjugate of claim 1 or 2 or the antibody-linker of claim 3, wherein m+n = 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24;
preferably, m is non-zero, preferably n is non-zero, preferably both m and n are non-zero; preferably, m+n+.4;
preferably, the sulphur-carbon bridge in the structure is located between antibody hinge regions, between antibody light and heavy chains or between antibody heavy chains;
preferably, the first click chemistry group is capable of pairing with the second click chemistry group; the first click chemistry group and/or the second click chemistry group is selected from the group consisting of: any one of methyltetrazine, trans-cyclooctene, azide, dibenzocyclooctyne, tetrazine, alkyne, cyclopropane cyclooctyne and cyclopropene;
Preferably, the first click chemistry group and the second click chemistry group are both selected from any one of methyltetrazine and trans-cyclooctene;
preferably, the sulfur-carbon bridge in the structure is formed by reducing disulfide bonds of the antibody itself and then reconstructing the disulfide bonds;
alternatively, conjugation partner and R 2 An amplifying carrier is connected between the two.
5. The antibody conjugate of claim 1, wherein the conjugation partner is selected from at least one of a fluorescent dye, an enzyme, a radioisotope, a chemiluminescent reagent, a nanoparticle-based label.
6. A method of preparing an antibody conjugate according to any one of claims 2, 3, 4, 5, characterized in that it comprises: antibody-linker and R 2 -performing an addition reaction of the conjugation partner to obtain an antibody conjugate;
the antibody-linker has the following structure:
the preparation method of the antibody-linker comprises the following steps:
mixing the linker with the antibody with disulfide bond opened by reduction to obtain an antibody-linker; the linker has the following structure:
7. use of the antibody conjugate of any one of claims 1, 2, 4, 5 in the preparation of a diagnostic reagent or kit.
8. Use of the antibody-linker of claim 3 in the preparation of a diagnostic reagent or kit.
9. A kit comprising the antibody conjugate of any one of claims 1, 2, 4, 5, 6.
10. A kit comprising the antibody-linker of claim 3 or 4.
CN202310508396.XA 2022-05-29 2023-05-06 Antibody conjugate and application thereof Pending CN117147824A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2023/096265 WO2023231889A1 (en) 2022-05-29 2023-05-25 Antibody conjugate and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2022105954453 2022-05-29
CN202210595445 2022-05-29
CN202310471792 2023-04-25
CN202310471792X 2023-04-25

Publications (1)

Publication Number Publication Date
CN117147824A true CN117147824A (en) 2023-12-01

Family

ID=88883015

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202310508396.XA Pending CN117147824A (en) 2022-05-29 2023-05-06 Antibody conjugate and application thereof
CN202310601112.1A Pending CN117147826A (en) 2022-05-29 2023-05-25 Antibody conjugate and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202310601112.1A Pending CN117147826A (en) 2022-05-29 2023-05-25 Antibody conjugate and application thereof

Country Status (2)

Country Link
CN (2) CN117147824A (en)
WO (1) WO2023231889A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2561164A1 (en) * 2004-04-01 2005-10-20 Elan Pharmaceuticals, Inc. Steroid sparing agents and their use
NO2789793T3 (en) * 2012-10-24 2018-01-27
WO2015179734A1 (en) * 2014-05-23 2015-11-26 Novartis Ag Methods for making conjugates from disulfide-containing proteins
US10098960B2 (en) * 2015-04-03 2018-10-16 Ucl Business Plc Polymer conjugate
US11666658B2 (en) * 2018-12-21 2023-06-06 Regeneran Pharmaceuticals, Inc. Rifamycin analogs and antibody-drug conjugates thereof
EA202192137A1 (en) * 2019-01-31 2021-10-25 Ханчжоу Дэк Биотек Ко., Лтд AMATOXY ANALOGUE CONJUGATE WITH BRANCHED LINKERS

Also Published As

Publication number Publication date
WO2023231889A1 (en) 2023-12-07
CN117147826A (en) 2023-12-01

Similar Documents

Publication Publication Date Title
CA2609702C (en) Antibody conjugates via heterobifunctional peg linkers
CN104704366B (en) Azaindole quinoline-the cyanine dye and its bioconjugates of hydroxamic acid substitution
JP4896959B2 (en) Doxorubicin immunoassay
US6632929B1 (en) Avidin derivatives and uses thereof
JPS63238100A (en) Production of antibody complex
JPH06322000A (en) Immunologically active conjugates and their production
JPH03504645A (en) Cross-linked antibody and its production method
JPH0123063B2 (en)
JPS62120378A (en) Protain label and medicine release system
WO2011109326A2 (en) Crosslinking reagents, methods, and compositions for studying protein-protein interactions
Kirley et al. Selective disulfide reduction for labeling and enhancement of Fab antibody fragments
US4975532A (en) Method to derivatize dextran
DE10048417A1 (en) Branched linker connections
Luescher et al. Binding of photoreactive lysozyme peptides to murine histocompatibility class II molecules.
JPH0115827B2 (en)
JP4085443B2 (en) Amino acid analysis reagent and amino acid analysis method
CN117147824A (en) Antibody conjugate and application thereof
US11236386B2 (en) Method for labeling of aldehyde containing target molecules
DE10111224A1 (en) Analogs of ecstasy class and use thereof in detecting ecstasy-class compounds
CN117147825A (en) Antibody conjugate and application thereof
US20210380635A1 (en) Compositions and methods for protein labeling, modification, analysis, and targeted delivery
KR101919427B1 (en) Antibody-functionalization Method and Method for Manufacturing Nanoparticle-Antibody Conjugated Nano-platform
JP2594803B2 (en) Lectin complex, method for producing the same and probe
US5869348A (en) Methods for the preparation of antibodies directed against dithiocarbamates and the use thereof for detection of nitric oxide in body fluids
JP3399598B2 (en) Labeling reagent for immunoassay using N-partially substituted aminoglycan

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination