CA2991384A1 - Bridge linkers for conjugation of a cell-binding molecule - Google Patents

Bridge linkers for conjugation of a cell-binding molecule Download PDF

Info

Publication number
CA2991384A1
CA2991384A1 CA2991384A CA2991384A CA2991384A1 CA 2991384 A1 CA2991384 A1 CA 2991384A1 CA 2991384 A CA2991384 A CA 2991384A CA 2991384 A CA2991384 A CA 2991384A CA 2991384 A1 CA2991384 A1 CA 2991384A1
Authority
CA
Canada
Prior art keywords
cell
receptor
antibody
acid
beta
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.)
Granted
Application number
CA2991384A
Other languages
French (fr)
Other versions
CA2991384C (en
Inventor
Robert Yongxin Zhao
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.)
Hangzhou Dac Biotech Co Ltd
Original Assignee
Sushou M conj Biotech Co Ltd
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 Sushou M conj Biotech Co Ltd filed Critical Sushou M conj Biotech Co Ltd
Publication of CA2991384A1 publication Critical patent/CA2991384A1/en
Application granted granted Critical
Publication of CA2991384C publication Critical patent/CA2991384C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/16Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • C07C233/17Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/20Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to a carbon atom of an acyclic unsaturated carbon skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68031Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being an auristatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68033Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a maytansine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6869Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of the reproductive system: ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6889Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/40Unsaturated compounds
    • C07C59/76Unsaturated compounds containing keto groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/46Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with hetero atoms directly attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Landscapes

  • Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Reproductive Health (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Pyrrole Compounds (AREA)

Abstract

Provided are novel bridge linkers for use in the specific conjugation of a compound, in particular, a cytotoxic agent to a biological molecule. Hence, the present invention relates to methods of making cell-binding agent-drug (cytotoxic agent) conjugates in a specific manner comprising either modification of drugs with these linkers first, followed by reaction with prepared cell-binding agents;
or modification of cell-binding agents with these linkers first, followed by reaction with drugs. Also provided are applications of the conjugates in the treatment of cancers, infections and autoimmune disorders.

Description

SPECIFIC CONJUGATION OF A CELL-BINDING MOLECULE
FIELD OF THE INVENTION
The present invention relates to the preparation of novel linkers used for the specific conjugation of a compound, in particular, a cytotoxic agent to a biological molecule. The present invention also relates to methods of making cell-binding agent-drug (cytotoxic agent) conjugates in a specific manner comprising either modification of drugs with these linkers first, followed by reaction with prepared cell-binding agents; or modification of cell-binding agents with these linkers first, followed by reaction with drugs.
BACKGROUND OF THE INVENTION
Proteins, specifically antibodies have been extensively used in therapeutic applica-tions, in vitro assays as research reagents and in vivo as diagnostic tools or as therapeutic drugs (Gad, S. C. Drug discovery handbook, published by Wiley-Interscience, 2005). For many applications the protein needs to be modified with an interesting group, such as a cytotoxic drug, a radio label element or a chromatographic molecule for use in therapy or a detection agent when used in diagnostics (Teicher, B. A. et al. Clin. Cancer Res. 2011, 17, 6389-97; Elsadek, B. et al., J. Control Release, 2012, 157, 4-28). One of these applica-tions, called antibody¨drug conjugates (ADCs), which is the exquisite targeting ability of antibodies in combination with the cytotoxic action of anticancer agents, enables to target and deliver drugs to cancer cells leaving normal cells largely unaffected, has been ex-tremely intense exploitation in the last two decades. In particular, since US
FDA approvals of Adcetris (brentuximab vedotin) in 2011 and Kadcyla (ado-trastuzumab emtansine) in 2013, the applications of antibody-drug conjugate (ADC) as a promise targeted treatment of cancers have been exploded and almost every major pharmaceutical and biotech com-pany has adopted this approach (Chari, R. et al, Angew. Chem., Int. Ed. 2014, 53, 3796-3827; Sievers, E. L. et al. Annu Rev Med. 2013, 64, 15-29; Mehrling, T.
Future Oncol, 2015, 11, 549). Currently there are more than 50 ADC drugs in the clinic trials according to www.clinictrails.gov.
The first-generation ADCs, including Kadcyla and Adcetris, are produced through nonselective conjugation of native lysine amines or interchain cysteine thiols on an anti-body respectively to a cytotoxic drug. Since there are over 50 surface-exposed lysines and 8 hinge cysteine residues in IgG1 antibodies, this nonselective conjugation results in
2 randomly cross-linkage of cytotoxic drugs to practically all areas of the antibody molecule, particularly having a diverse population of ADCs with a wide distribution of drugs per antibody (DAR) (Wang, L., et al. 2005 Protein Sci. 14, 2436; Hamblett, K. J., et al. 2004 Clin. Cancer Res. 10, 7063). Thus some of the undesired ADC subpopulation could lead to shorter circulation half-life, lower efficacy, potentially increased off-target toxicity and a wide range of in vivo pharmacokinetic (PK) properties (Hamblett, K. J. et al, Clin.
Cancer Res. 2004, 10, 7063-7070; Adem, Y. T. et al, Bioconjugate Chem. 2014, 25, 656-664; Boylan, N. J. Bioconjugate Chem., 2013, 24, 1008-1016; Strop, P., et al 2013 Chem. Biol. 20, 161-167). In addition, with this classical conjugation, the batch-to-batch consistency in ADC production can be challenging and may require diligent manufactur-ing capabilities (Wakankar, A. mAbs, 2011, 3, 161-172).
Therefore, biotechnology companies and academic institutions are intensely focusing on establishing novel reliable methods for site-specific ADC conjugation. So far, there are several approaches developed in recent years for site selective ADC
preparation (Panowski, S, 2014, mAbs 6, 34). They include incorporation of unpaired cysteines, e.g.
engineered reactive cysteine residues, called THIOMAB from Genentech (Junutula, J. R., et al 2010 Clin. Cancer Res. 16, 4769; Junutula, J. R., et al 2008 Nat Biotechnol. 26, 925-32; US Patents 8,309,300; 7,855,275; 7,521,541; 7,723,485, W02008/141044), genetical-ly introduced glutamine tag with Streptoverticillium mobaraense transglutaminase (mTG) (Strop, P., Bioconjugate Chem., 2014, 25, 855-862; Strop, P., et al., 2013, Chem. Biol. 20, 161-167; US Patent 8,871,908 for Rinat-Pfizer) or with Microbial transglutaminase (MTGase) (Dennler, P., et al, 2014, Bioconjug. Chem. 25, 569-578. US pat appl 20130189287 for Innate Pharma; US Pat 7,893,019 for Bio-Ker S.r.l. (IT)), incorporation of thiolfucose (Dennler, P., et al, 2014 Bioconjugate Chemistry 25, 569;
Okeley, N. M., et al 2013 Bioconjugate Chem. 24, 1650), incorporation of unnatural amino acids through mutagenesis (Axup, J.Y., et al., 2012, Proc. Natl. Acad. Sci. 109, 16101-16106; Zimmer-man, ES., et al., 2014, Bioconjug. Chem. 25, 351-361; Wu, P., et al, 2009 Proc. Natl.
Acad. Sci. 106, 3000-3005; Rabuka, D., et al, 2012 Nat. Protoc. 7, 1052-67; US
Pattent 8,778,631 and US Pat Appl. 20100184135, W02010/081110 for Sutro Biopharma;
W02006/069246, 2007/059312, US Patents 7,332,571, 7,696,312, and 7,638,299 for Ambrx; W02007/130453, US patents 7,632,492 and 7,829,659 for Allozyne), Incorpora-tion of selenocysteine into antibodies (Hofer, T., et al 2009, Biochemistry 48, 12047-12057; US Patent 8,916,159 for US National Cancer Institute), Convertion of cysteines
3 located in the CXPXR consensus sequence to formylglycine (FGly) with formylglycine generating enzyme (FGE) (Drake, P.M., et al., 2014, Bioconjug. Chem. 25, 1331-1341.
Carrico; Isaac S. et al 7,985,783; 8,097,701; 8,349,910, and US Pat Appl 20140141025, 20100210543 for Redwood Bioscience), and through glycoengineeringly introduction of sialic acid with the use of galactosyl- and sialytransferases (Thou, Q., et al 2014, Bioconjug. Chem., 25, 510-520, US Pat Appl 20140294867 for Sanofi-Genzyme).
These above methods have produced nearly homogeneous product profiles, but they are required antibody-engineering processes and reoptimization of cell culture conditions.
Moreover, expression yields for genetic encoding of an unnatural amino acid were typically not promisingly high enough (Tian, F., et al, 2014, Proc. Natl. Acad. Sci. U. S.
A. 111, 1766-71) which has a significant impact on the cost of goods of the ADC. In addition, it has been known that ADCs obtained by conjugation to cysteine side chains often display limited stability in circulation, leading to premature disconnection of the cytotoxic payload before the tumor site is reached (Junutula, J. R., et al 2008, Nat.
Biotechnol. 26, 925-32).
The disulfide bond structures of the four subclasses of IgG antibodies were known in the 1960s (Milstein C. Biochem J 1966, 101:338 - 351; Pink JR, Milstein C.
Nature 1967, 214:92-94; Frangione B, Milstein C. Nature 1967, 216:939 - 941; Pink JR, Milstein C.
Nature 1967, 216:941 -942; Frangione B, et al. Biochem J 1968, 106,15 ¨ 21;
Frangione B, Milstein C. J Mol Biol 1968; 33:893 ¨ 906; Edelman GM, et al. Proc Natl Acad Sci USA 1969; 63:78 -85; Frangione B, et al. Nature 196, 221:145 -148, Spiegelberg, H. L. et al Biochemistry, 1975, 10, 2157-63 ). Disulfide bond structure is critical for the structure, stability, and biological functions of IgG molecules. Among the four subclasses of IgG
antibodies, IgGi, IgG2, IgG3 and Igat, each IgG contains a total of 12 intra-chain disulfide bonds; each disulfide bond is associated with an individual IgG domain. The two heavy chains are connected in the hinge region by a variable number of disulfide bonds: 2 for IgGi and Igat, 4 for IgG2 and 11 for IgG3. The light chain of the IgGi is connected to the heavy chain by a disulfide bond between the last cysteine residue of the light chain and the fifth cysteine residue of the heavy chain. But, for IgG2, IgG3 and Igat, the light chain is linked to the heavy chain by a disulfide bond between the last cysteine residue of the light chain and the third cysteine residue of the heavy chain (Liu, H. and May, K., 2012, mAbs
4, 17-23). On the ranks of the susceptibility of disulfide bonds in human IgGi antibodies by experimental reduction, differential alkylation, and LC-MS analysis (Liu, H, et al Anal.
Chem., 2010, 82, 5219-5226), inter chain disulfide bonds are more susceptible to reduc-tion than intra chain disulfide bonds, and the disulfide bonds between the light chain and heavy chain were more susceptible than disulfide bonds between the two heavy chains.
The upper disulfide bond of the two inter heavy chain disulfide bonds was more suscepti-ble than the lower one. Furthermore, disulfide bonds in the CH2 domain were the most susceptible to reduction. Disulfide bonds in VL, CL, VH, and CH1 domains had similar and moderate susceptibility, while disulfide bonds in the CH3 domain were the least susceptible to reduction (Liu, H, et al Anal. Chem., 2010, 82, 5219-5226).
Based on the more susceptibility of inter chain disulfide bonds in human IgG1 anti-bodies, several institutions and companies adopted the chemically specific conjugation strategy through rebridging reduced interchain disulfide bonds of a native antibody, such as, using bromo or dibromo-maleimides, called next generation maleimides (NGMs) (Schumacher, F.F., et al 2014, Org. Biomol. Chem. 12, 7261-7269; UCL Cancer Institute), applying bis-alkylating reagents via a three-carbon bridge (Badescu, G., et al., 2014, Bioconjug. Chem. 25, 1124-1136., W02013/190272, W02014/064424 for PolyTherics Ltd), with di-substituted heteroaryl bridge (US Pat Appl. 2015/0105539 for Concortis Biosystem), or through di-maleimide as a bridge (W02014/114207). We have also used bromo maleimide and dibromomaleimide linkers to conjugate both drugs and antibodies for a quite while (W02014/009774, PCT/IB2012/053554). However, these above bridge linkers were designed in the way to conjugate only one cytotoxic agents to a pair of disul-fide bonds, and therefore at most of time they only produced ADCs at DAR less than 2 (drugs per antibody), due to limited numbers (about two pairs) of reduced disulfide bonds are more accessible for conjugation.
As one of the major issues for ADCs is the limited numbers or amount of cytotoxic compound that ultimately reaches the tumor, and the favorable DAR over 3 is much important factor for improvement of ADC therapeutical index (Epenetos, A. A.
et al, Cancer Res., 1986, 46, 3183-3191; Chari, R. V. Ace. Chem. Res., 2008, 41, 98-107, Zhao, R. Y. 2011 J. Med. Chem. 54, 3606-3623), we therefore disclose novel disulfide bridge linkers of this invention that not only are able to conjugate two or more drugs per linker for achieving higher DARs (>4), but also can selectively bridge inter chain of disulfide bonds on surface of antibody due to the nature of stretch-out triple bond of the acetylenedicarboxyl group, particularly when two cytotoxic agents are linked at both ends of the stretch-out bridge linker, forming a quite large size (>20 A) of molecule which in turn hardly accesses to the other disulfide bond sites, such as intra chain disulfide bonds beneath the complex antibodies. The disulfide bridge linkers of this invention therefore can be used for selective bridging the pairs of free thiols on the inter chain of antibody, which are generated by overloaded TCEP or DTT, and for producing an ADC having DAR (drugs per antibody) over four. And the over reduced disulfide bonds that are mac-
5 cessibly reached by the bridge linkers, particularly by the stretch-out bridge linker contain-ing two cytotoxic agents, can be recoupled (regenerated) by an oxide, e.g.
dehydroascorbic acid (DHAA) or Cu(II), at the end of conjugation. In a word, these bridge linkers of the invention can make homogeneous production of specific ADCs in a simple way.
SUMMARY OF THE INVENTION
The present invention provides linkers containing an acetylenedicarboxylic group to link two drugs to a cell-binding agent (e.g., an antibody). The preferred formula of the cell-binding molecule-linker-drug conjugates can be represented as:
CbLDrugi S Drug2/11, wherein Cb is a cell-binding agent, L is a linker, Drugl and Drug2 are a drug molecule, n is an integer from 1 to 20, and two S (sulfur) elements from Cb bridgely link to L, which covalently connects two or more drugs (per bridge linker L).
The advantages in applying the linker in the cell molecule-drug conjugate are:
a). Retain-ing the stability of the conjugates by covalently cross-linking (re-bridging) the pairs of reduced disulfur bonds of the cell-binding agents, particularly of antibodies;
b). Enabling conjugation of the cytotoxic agents/drugs to specific sites of a cell-binding molecule, e.g.
the inter chain sites of IgG antibodies, resulting in homogeneous production of ADC.
In one aspect of the present invention, the linker is represented by Formula (I) Zi¨Ri Xi ____________________________ X2-R2-Z2 (I) Wherein the acetylenedicarboxyl group on the linker is capable of reacting with a pair of sulfur atoms of the cell-binding agent;
Zi and Z2 are the same or different a function group that enables to react with a cyto-toxic drug. The functional group Z1 or Z2 can link to a cytotoxic drug via a disulfide, ether, ester, thioether, thioester, peptide, hydrazone, carbamate, carbonate, amine (secondary, tertiary, or quartary), imine, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
R1 and R2 are the same or different, and are absent, linear alkyl having from 1-6 car-bon atoms, branched or cyclic alkyl having from 3 to 6 carbon atoms, linear, branched or
6 cyclic alkenyl or alkynyl, or 1-6 carbon atoms of esters, ether, amide, or polyethyleneoxy unit of formula (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combina-tion thereof.
Additionally Ri and R2 are respectively a chain of atoms selected from C, N, 0, S, Si, and P, preferably having 0-500 atoms, which covalently connects to Xi or X2 and Zi or Z2. The atoms used in forming the Ri and R2 may be combined in all chemically relevant ways, such as forming alkylate, alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxylamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
Xi and X2 are independently selected from N(R3), 0, S or CH2; R3 is H, linear alkyl having from 1-6 carbon atoms, branched or cyclic alkyl having from 3 to 6 carbon atoms, linear, branched or cyclic alkenyl or alkynyl, or 1-6 carbon atoms of esters, ether, amide, or polyethyleneoxy unit of formula (0CH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
In another aspect, this invention provides a cell-binding agent-drug conjugate of For-mula (II), in which the cell-binding agent, Cb, and the drug, Drugl and Drug2, have reacted at the ends of the bridge linker:

Drug1¨R1 X1 /
X2 R2 -Drug2 Sµ
_____________________________________________________ In Cb (II) wherein:
Cb represents a cell-binding agent, preferred an antibody;
Inside the bracket (parentheses) are the linker-drug components that are conjugated to the cell-binding molecule via a pair of disulfide bond Drugi and Drug2 represent the same or different cytotoxic agents, which linked to the cell-binding agent via the bridge linker by a disulfide, thioether, thioester, peptide, hydra-zone, ether, ester, carbamate, carbonate, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
n is 1 ¨ 20; RI, R2, Xi and X2 are described the same previously in Formula (I).
7 In a further aspect, the present invention provides a modified cell-binding agent of Formula (III), in which the cell-binding agent, Cb, has reacted with the bridge linker, which has Z1 and Z2, the function groups capable of reacting with a drug:

Z1¨R1 ______________________________ X2-R2-Z2 /S
In Cb (III) Wherein Cb, Zi, Z2, n, R1, R2, X1, and X2 are defined the same as in Formula (I) and (II).
In an even further aspect, the present invention provides a modified drug of Formula (IV), in which the drug, Drugi and Drug2, have reacted with the linker of Formula (I), which still has the acetylenedicarboxyl group capable of reacting with a pair of sulfur atoms of the cell-binding agent:

II ¨
Drug1¨R1¨X1 X2-R2-Drug2 (IV) Wherein Drugi, Drug2, R1, R2, X1, and X2 are defined the same as in Formula (I) and (II).
The present invention further relates to a method of making a cell-binding molecule-drug conjugate of Formula (II), wherein the drugs, Drugi and Drug2 are linked to a cell-binding agent via the bridge linker.
The present invention also relates to a method of making a modified cell-binding mol-ecule of Formula (III), wherein the cell-binding molecule is reacted with the bridge linker of Formula (I).
The present invention also relates to a method of making a modified drug of formula (IV), wherein the drug is reacted with the bridge linker of Formula (I).
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the synthesis of a bridge linker containing polyethylene glycol groups and the application of this linker in the conjugation of an antibody with drugs.
Figure 2 shows the synthesis of a bridge linker and the application of this linker in the conjugation of drugs to an antibody via oxime linkage.
Figure 3 shows the synthesis of a bridge linker containing a peptide and the applica-tion of this linker in the conjugation of drugs to an antibody via amide linkage.
8 Figure 4 shows the synthesis of a bridge linker containing peptides, polyethylene gly-col.
Figure 5 shows the synthesis of a bridge linker containing peptides and polyethylene glycols, and the application in the conjugation of two or four drugs per linker to an anti-body via amide linkage.
Figure 6 shows the synthesis of tubulysin analogs which are modified with the bridge-linker containing peptides and polyethylene glycols.
Figure 7 shows the synthesis of the conjugates of cell-binding molecule-tubulysin analogs via the bridge-linker containing polyethylene glycols.
Figure 8 shows the synthesis of the conjugates of cell-binding molecule-maytansinoids via the bridge-linker.
Figure 9 shows the synthesis of the conjugates of cell-binding molecule-MMAF
ana-logs via the bridge-linker.
Figure 10 shows the synthesis of the conjugates of cell-binding molecule-tubulysin analogs via the bridge-linker.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS
"Alkyl" refers to an aliphatic hydrocarbon group which may be straight or branched having 1 to 8 carbon atoms in the chain. "Branched" means that one or more lower C
numbers of alkyl groups such as methyl, ethyl or propyl are attached to a linear alkyl chain. Exemplary alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl, n-pentyl, 3-pentyl, octyl, nonyl, decyl, cyclopentyl, cyclohexyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, 3,3-dimethylpentyl, 2,3,4-trimethylpentyl, 3-methyl-hexyl, 2,2-dimethylhexyl, 2,4-dimethylhexyl, 2,5-dimethylhexyl, 3,5-dimethylhexyl, 2,4-dimethylpentyl, 2-methylheptyl, 3-methylheptyl, n-heptyl, isoheptyl, n-octyl, and isooctyl. A C1-C8 alkyl group can be unsubstituted or substi-tuted with one or more groups including, but not limited to, -Ci-C8 alkyl,-0-(C1-C8 alkyl), -aryl, -C(0)R', -0C(0)R', -C(0)OR', -C(0)NH2, -C(0)NHR', -C(0)N(R')2, -NHC(0)R', -SR', -S(0)2R', -S(0)R', -OH, -halogen, -N3, -NH2, -NH(R'), -N(R') 2 and -CN;
where each R' is independently selected from -C1-C8 alkyl and aryl."Halogen" refers to fluorine, chlorine, bromine or iodine atom; preferably fluorine and chlorine atom.
"Heteroalkyl" refers to C2-C8 alkyl in which one to four carbon atoms are inde-
9 pendently replaced with a heteroatom from the group consisting of 0, S and N.
"Carbocycle" refers to a saturated or unsaturated ring having 3 to 8 carbon atoms as a monocycle or 7 to 13 carbon atoms as a bicycle. Monocyclic carbocycles have 3 to 6 ring atoms, more typically 5 or 6 ring atoms. Bicyclic carbocycles have 7 to 12 ring atoms, arranged as a bicycle [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicycle [5,6] or [6,6] system. Representative C3-C8 carbocycles include, but are not limited to, -cyclopropyl, -cyclobutyl, -cyclopentyl, -cyclopentadienyl, -cyclohexyl, -cyclohexenyl, -1,3-cyclohexadienyl, -1,4-cyclohexadienyl, -cycloheptyl, -1,3-cycloheptadienyl, -1,3,5-cycloheptatrienyl, -cyclooctyl, and -cyclooctadienyl.
A "C3-C8 carbocycle" refers to a 3-, 4-, 5-, 6-, 7- or 8-membered saturated or unsatu-rated nonaromatic carbocyclic ring. A C3-C8 carbocycle group can be unsubstituted or substituted with one or more groups including, but not limited to, -C1-C8 alkyl,-0-(C1-C8 alkyl), -aryl, -C(0)R', -0C(0)R', -C(0)OR', -C(0)NH2, -C(0)NHR', -C(0)N(R')2, -NHC(0)R', -SW, -S(0)R',-S(0)2R', -OH, -halogen, -N3, -NH2, -NH(R'), -N(R') 2 and -CN;
where each R' is independently selected from -C1-C8 alkyl and aryl.
"Alkenyl" refers to an aliphatic hydrocarbon group containing a carbon-carbon double bond which may be straight or branched having 2 to 8 carbon atoms in the chain. Exem-plary alkenyl groups include ethenyl, propenyl, n-butenyl, i-butenyl, 3-methylbut-2-enyl, n-pentenyl, hexylenyl, heptenyl, octenyl.
"Alkynyl" refers to an aliphatic hydrocarbon group containing a carbon-carbon triple bond which may be straight or branched having 2 to 8 carbon atoms in the chain. Exem-plary alkynyl groups include ethynyl, propynyl, n-butynyl, 2-butynyl, 3-methylbutynyl, 5-pentynyl, n-pentynyl, hexylynyl, heptynyl, and octynyl.
"Alkylene" refers to a saturated, branched or straight chain or cyclic hydrocarbon rad-ical of 1-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Typical alkylene radicals include, but are not limited to: methylene (-CH2-), 1,2-ethyl (-CH2CH2-), 1,3-propyl (-CH2CH2CH2-), 1,4-butyl (-CH2CH2CH2CH2-), and the like.
"Alkenylene" refers to an unsaturated, branched or straight chain or cyclic hydrocar-bon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. Typical alkenylene radicals include, but are not limited to:
1,2-ethylene (-CH=CH-).
"Alkynylene" refers to an unsaturated, branched or straight chain or cyclic hydrocar-bon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a 5 parent alkyne. Typical alkynylene radicals include, but are not limited to: acetylene, propargyl and 4-pentynyl.
"Aryl" or Ar refers to an aromatic or hetero aromatic group, composed of one or sev-eral rings, comprising three to fourteen carbon atoms, preferentially six to ten carbon atoms. The term of "hetero aromatic group" refers one or several carbon on aromatic
10 group, preferentially one, two, three or four carbon atoms are replaced by 0, N, Si, Se, P
or S, preferentially by 0, S, and N. The term aryl or Ar also refers to an aromatic group, wherein one or several H atoms are replaced independently by -R', -halogen, -OR', or -SR', -NR'R", -N=R', -S(0)R', -S(0)2R', -S(0)20R', -0S(0)20R', -PR'R", -P(0)R'R", -P(OR')(OR"), -P(0)(OR')(OR") or -OP(0)(OR')(OR") wherein R', R" are independently H, alkyl, alkenyl, alkynyl, heteroalkyl, aryl, arylalkyl, carbonyl, or pharmaceutical salts.
"Heterocycle" refers to a ring system in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group of 0, N, S, Se, B, Si and P.
Preferable heteroatoms are 0, N and S. Heterocycles are also described in The Handbook of Chemistry and Physics, 78th Edition, CRC Press, Inc., 1997-1998, p. 225 to 226, the disclosure of which is hereby incorporated by reference. Preferred nonaromatic heterocy-clic include, but are not limited to epoxy, aziridinyl, thiiranyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxiranyl, tetrahydrofuranyl, dioxolanyl, tetrahydropyranyl, dioxanyl, dioxolanyl, piperidyl, piperazinyl, morpholinyl, pyranyl, imidazolinyl, pyrrolinyl, pyrazolinyl, thiazolidinyl, tetrahydrothiopyranyl, dithianyl, thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, dihydropyranyl, tetrahydropyridyl, dihydropyridyl, tetrahydropyrimidinyl, dihydrothiopyranyl, azepanyl, as well as the fused systems result-ing from the condensation with a phenyl group.
The term "heteroaryl" or aromatic heterocycles refers to a 5 to 14, preferably 5 to 10 membered aromatic hetero, mono-, bi- or multicyclic ring. Examples include pyrrolyl, pyridyl, pyrazolyl, thienyl, pyrimidinyl, pyrazinyl, tetrazolyl, indolyl, quinolinyl, purinyl, imidazolyl, thienyl, thiazolyl, benzothiazolyl, furanyl, benzofuranyl, 1,2,4-thiadiazolyl, isothiazolyl, triazoyl, tetrazolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl,
11 carbazolyl, benzimidazolyl, isoxazolyl, pyridyl-N-oxide, as well as the fused systems resulting from the condensation with a phenyl group.
"Alkyl", "cycloalkyl", "alkenyl", "alkynyl", "aryl", "heteroaryl", "heterocyclic" and the like refer also to the corresponding "alkylene", "cycloalkylene", "alkenylene", "alkynylene", "arylene", "heteroarylene", "heterocyclene" and the likes which are formed by the removal of two hydrogen atoms.
"Arylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-l-yl, naphthobenzyl, 2-naphthophenylethan-1-y1 and the like.
"Heteroarylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen at-oms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heteroaryl radical. Typical heteroarylalkyl groups include, but are not limited to, 2-benzimidazolylmethyl, 2-furylethyl and the like.
Examples of a "hydroxyl protecting group" include, but are not limited to, methoxymethyl ether, 2-methoxyethoxymethyl ether, tetrahydropyranyl ether, benzyl ether, p-methoxybenzyl ether, trimethylsilyl ether, triethylsilyl ether, triisopropylsilyl ether, t-butyldimethylsilyl ether, triphenylmethylsilyl ether, acetate ester, substituted acetate esters, pivaloate, benzoate, methanesulfonate and p-toluenesulfonate.
"Leaving group" refers to a functional group that can be substituted by another func-tional group. Such leaving groups are well known in the art, and examples include, but are not limited to, a halide (e.g., chloride, bromide, and iodide), methanesulfonyl (mesyl), p-toluenesulfonyl (tosyl), trifluoromethylsulfonyl (triflate), and trifluoromethylsulfonate.
The following abbreviations may be used herein and have the indicated definitions:
Boc, tert-butoxy carbonyl; BroP, bromotrispyrrolidinophosphonium hexafluorophosphate;
CDI, 1,1'-carbonyldiimidazole; DCC, dicyclohexylcarbodiimide; DCM, dichloromethane;
DIAD, diisopropylazodicarboxylate; DIBAL-H, diisobutylaluminium hydride;
D1PEA, diisopropylethylamine; DEPC, diethyl phosphorocyanidate; DMA, N,N-dimethyl acetamide; DMAP, 4-(N, N-dimethylamino)pyridine; DMF, N,N-dimethylformamide;
DMSO, dimethylsulfoxide; DTT, dithiothreitol; EDC, 1-(3-dimethylaminopropy1)-3-ethylcarbodiimide hydrochloride; ESI-MS, electrospray mass spectrometry; HATU, 0-(7-azabenzotriazol-1-y1)-N, N, N', N'-tetramethyluronium hexafluorophosphate;
HOBt, 1-
12 hydroxybenzotriazole; HPLC, high pressure liquid chromatography; NHS, N-Hydroxysuccinimide; MMP, 4-methylmorpholine; PAB, p-aminobenzyl; PBS, phosphate-buffered saline (pH 7.0-7.5); PEG, polyethylene glycol; SEC, size-exclusion chromatog-raphy; TCEP, tris(2-carboxyethyl)phosphine; TFA, trifluoroacetic acid; THF, tetrahydrofuran; Val, valine.
"Pharmaceutically" or "pharmaceutically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
"Pharmaceutically acceptable solvate" or "solvate" refer to an association of one or more solvent molecules and a disclosed compound. Examples of solvents that form phar-maceutically acceptable solvates include, but are not limited to, water, isopropanol, etha-nol, methanol, DMSO, ethyl acetate, acetic acid and ethanolamine.
"Pharmaceutically acceptable excipient" includes any carriers, diluents, adjuvants, or vehicles, such as preserving or antioxidant agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
Supplementary active ingredients can also be incorporated into the compositions as suitable therapeutic combi-nations.
As used herein, "pharmaceutical salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
The phar-maceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, tartaric, citric, methanesulfonic, benzenesulfonic, glucuronic, glutamic, benzoic, salicylic, toluenesulfonic, oxalic, fumaric, maleic, lactic and the like.
Further addition salts include ammonium salts such as tromethamine, meglumine, epolamine, etc., metal salts such as sodium, potassium, calcium, zinc or magnesium.
13 The pharmaceutical salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
Generally, such salts can be prepared via reaction the free acidic or basic forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
The novel conjugates disclosed herein use the bridge linkers. Examples of some suita-ble linkers and their synthesis are shown in Figures 1 to 10.
THE BRIDGE LINKERS
The synthetic routes to produce bridge linkers as well as the preparation of the conju-gates of drugs to a cell binding molecules of the present invention are shown in Figures 1-9. The bridge linkers possess two elements: a) A Substituent that is acetylenedicarboxyl group that can react to a pair of thiols to form covalent thioether bonds, and b) A group, such as but not limited to, a disulfide, maleimide, haloacetyl, aldehyde, ketone, azide, amine, alkoxyamine and hydrazide, capable of reaction with a drug. The bridge substitu-ents of acetylenedicarboxyl can be introduced by direct condensation of acetylenedicarboxylic acid with an amine, an alcohol, or a thiol group to form amide, ester or thioester bonds at acetylenedicarboxyl sites. The synthesis of these bridge linkers is exampled in the Figures 1, 3, 4, 5, 6, 7, 8 and 9. The bridge substituents of acetylenedicarboxyl can be introduced by condensation of acetylene with acid halides or acid anhydrides to form carbon-carbon bonds acetylenedicarboxyl sites. The synthesis of these bridge linkers is exampled in the Figure 2 and 10.
Preferably, the bridge linkers are compounds of the Formula (I) below:

Zi¨Ri Xi ____________________________ X2¨R2¨Z2 (I) Wherein the acetylenedicarboxyl group on the linker is capable of reacting with a pair of sulfur atoms of the cell-binding agent;
Z1 and Z2 are the same or different a function group that enables to react with a cyto-toxic drug. The functional group Z1 or Z2 can link to a cytotoxic drug via a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, amine (second-
14 ary, tertiary, or quarter), imine, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
Ri and R2 are the same or different, and are H, linear alkyl having from 1-6 carbon at-oms, branched or cyclic alkyl having from 3 to 6 carbon atoms, linear, branched or cyclic alkenyl or alkynyl, or 1-6 carbon atoms of esters, ether, amide, or polyethyleneoxy unit of formula (OCH2CH2)p, or polypropyleneoxy unit of formula (OCH2(CH3)CH2)p wherein p is an integer from 0 to about 1000, or combination thereof.
Additionally Ri and R2 are respectively a chain of atoms selected from C, N, 0, S, Si, and P, preferably having 0-500 atoms, which covalently connects to Xi or X2 and Zi or Z2. The atoms used in forming the Ri and R2 may be combined in all chemically relevant ways, such as forming alkylate, alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxylamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
Xi and X2 are independently selected from N(R3), 0, S or CH2; Wherein R3 is H, line-ar alkyl having from 1-6 carbon atoms, branched or cyclic alkyl having from 3 to 6 carbon atoms, linear, branched or cyclic alkenyl or alkynyl, or 1-6 carbon atoms of esters, ether, amide, or polyethyleneoxy unit of formula (0CH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
In another embodiment, R1, R2, and R3, can be respectively a chain of atoms selected from C, N, 0, S, Si, and P that covalently connects the cell-surface binding molecule and the conjugated drug. The atoms used in forming the bridge linker may be combined in all chemically relevant ways, such as forming alkylate, alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxylamines, urethanes, amino acids, acyloxylamines, hydroxamic acids, and many others. In addition, it is to be understood that the atoms forming the linker (L) may be either saturated or unsaturated, or may be radicals, or may be cyclized upon each other to form divalent cyclic structures, including cyclo alkanes, cyclic ethers, cyclic amines, arylenes, heteroarylenes, and the like in the linker.
Examples of the functional group, Zi and Z2, which enable linkage of a cytotoxic drug, include groups that enable linkage via a disulfide, thioether, thioester, peptide, hydrazone, ester, carbamate, carbonate, alkoxime or an amide bond. Such functional groups include, but are not limited to, thiol, disulfide, amino, carboxy, aldehydes, ketone, maleimido, haloacetyl, hydrazines, alkoxyamino, and/or hydroxy.
Examples of the functional group, Zi and Z2, that enable reaction with the terminal of amine of a drug/ cytotoxic agent can be, but not limited to, N-hydroxysuccinimide esters, 5 p-nitrophenyl esters, dinitrophenyl esters, pentafluorophenyl esters;
with the terminal of thiol can be, as but not limited to, pyridyldisulfides, nitropyridyldisulfides, maleimides, haloacetates and carboxylic acid chlorides; with the terminal of ketone or aldehyde can be, as but not limited to, amines, alkoxyamines, hydrazines, acyloxylamine; with the terminal of azide can be, as but not limited to, alkyne.
10 In preferred embodiments, R1, R2, and R3, are linear alkyl having from 1-6 carbon at-oms, or polyethyleneoxy unit of formula (OCH2CH2)p, p = 1-100.
The key step of synthesis of the bridge linker containing acetylenedicarboxyl groups is the condensation of the acetylenedicarboxylic acid, or its acid derivatives, with the other components containing an amine (1 or 2 amines), alcohol, or thiol on their terminal, as
15 shown in the following scheme (Ia):

II ¨ . ¨
II X
+ X ¨ R ¨s55 cSSRX ¨
Lvi uv2 (Ia) Wherein X is X1 or X2 described in Formula (I) as N(R3), 0, or S; R is 121 and/or R2 that described in Formula (I); R3 is the same in Formula (I).
Lv1 and Lv2 are the same or independently OH; F; Cl; Br; I; nitrophenol; N-hydroxysuccinimide (NHS); phenol; dinitrophenol; pentafluorophenol;
tetrafluorophenol;
difluorophenol; monofluorophenol; pentachlorophenol; triflate; imidazole;
dichlorophenol;
tetrachlorophenol; 1-hydroxybenzotriazole; tosylate; mesylate; 2-ethy1-5-phenylisoxazolium-3'-sulfonate, anhydrides formed its self, or formed with the other anhydride, e.g. acetyl anhydride, formyl anhydride; or a intermediate molecule generated with a condensation reagent for peptide coupling reactions, or for Mitsunobu reactions, e.g.
condensation reagents are: EDC (N-(3-Dimethylaminopropy1)-N'-ethylcarbodiimide), DCC (Dicyclohexyl-carbodiimide), N,N'-Diisopropylcarbodiimide (DIC), N-Cyclohexyl-N'-(2-morpholinoethyl)carbodiimide metho-p-toluenesulfonate (CMC,or CME-CDI), 1,1'-Carbonyldiimidazole (CDI),TBTU (0-(Benzotriazol-1-y1)-N,N,N',N'-tetramethyluronium tetrafluoroborate), N,N,N',N'-Tetramethy1-0-(1H-benzotriazol-1-y1)uronium hexafluorophosphate (HBTU), (Benzotriazol-1-yloxy)tris(dimethylamino)phosphonium hexafluorophosphate (BOP), (Benzotriazol-1-yloxy)tripyrrolidinophosphonium
16 hexafluorophosphate (PyB OP), Diethyl cyanophosphonate (DEPC), Chloro-N,N,N',N'-tetramethylformamidinium hexafluorophosphate, 1-[Bis(dimethylamino)methylene]-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (HATU), 1-[(Dimethylamino)(morpholino) methylene]-1H-[1,2,3]triazolo[4,5-b]pyridine-l-ium 3-oxide hexafluorophosphate (HDMA), 2-Chloro-1,3-dimethylimidazolidinium hexafluorophosphate (CIP), Chlorotripyrrolidinophosphonium hexafluorophosphate (PyCloP), Fluoro-N,N,N',N'-bis(tetramethylene)formamidinium hexafluorophosphate (BTFFH), N,N,N',N'-Tetramethyl-S-(1-oxido-2-pyridyl)thiuronium hexafluorophosphate, 0-(2-0xo-1(2H)pyridy1)-N,N,N',N'-tetramethyluronium tetrafluoroborate (TPTU), S-(1-Oxido-2-pyridy1)-N,N,N',N1-tetramethylthiuronium tetrafluoroborate, 0-[(Ethoxycarbonyl) cyano-methylenamino]-N,N,N',N'-tetramethyluronium hexafluorophosphate (HOTU), (1-Cyano-2-ethoxy-2-oxoethylidenaminooxy) dimethylamino-morpholino-carbenium hexafluorophosphate(COMU), 0-(Benzotriazol-1-y1)-N,N,N',N'-bis(tetramethylene) uronium hexafluorophosphate (HBPyU), N-Benzyl-N'-cyclohexylcarbodiimide (with, or without polymer-bound), Dipyrrolidino(N-succinimidyloxy)carbenium hexafluoro-phosphate (HSPyU), Chlorodipyrrolidinocarbenium hexafluorophosphate (PyClU), 2-Chloro-1,3-dimethylimidazolidinium tetrafluoroborate(CIB), (Benzotriazol-l-yloxy) dipiperidinocarbenium hexafluorophosphate (HBPipU), 0-(6-Chlorobenzotriazol-1-y1)-N,N,N',N'-tetramethyluronium tetrafluoroborate (TCTU), Bromotris(dimethylamino)-phosphonium hexafluorophosphate (BroP), Propylphosphonic anhydride (PPACA, T3P
), 2-Morpholinoethyl isocyanide (MET), N,N,N',N'-Tetramethy1-0-(N-succinimidyeuronium hexafluorophosphate (HSTU), 2-Bromo-1-ethyl-pyridinium tetrafluoroborate (BEP), 0-[(Ethoxycarbonyl)cyanomethylenamino]-N,N,N',N'-tetramethyluronium tetrafluoroborate (TOTU), 4-(4,6-Dimethoxy-1,3,5-triazin-2-y1)-4-methylmorpholinium chloride (MMTM, DMTMM), N,N,N',N1-Tetramethy1-0-(N-succinimidyl)uronium tetrafluoroborate (TS
TU), 0-(3,4-Dihydro-4-oxo-1,2,3-benzotriazin-3-y1)-N,N,N',N'-tetramethyluronium tetrafluoro-borate (TDBTU), 1,1'-(Azodicarbonyl)dipiperidine (ADD), Di-(4-chlorobenzyl) azodicar-boxylate (DCAD), Di-tert-butyl azodicarboxylate (DBAD), Diisopropyl azodicarboxylate (DIAD), Diethyl azodicarboxylate (DEAD).
When X is CH2, wherein the acetylenedicarboxyl group on the bridge linker connects to the other components of linker through C-C bonds, then the key step of synthesis of the bridge linker containing acetylenedicarboxyl groups is the condensation of bis(trimethylsilyl)acetylene, or acetylene bis-magnesiums (Grignard reagent), or acetylene
17 bis-lithiums (difl tiiioacet.ylene), or other di-metal acetylide with acid halides or acid anhy-drides, depicted as following reaction equations (Ib), (Ic), (Id), (1e), (If), (Ig) and (Ih):
0 ,0 + s(¨ csS/ =
CI (Ib) O-\ )2 + ¨Si __________________________ ¨01" =
/ \ (Ic) )c)??? + BrMg---MgBr (le) jt 0 5)2 BrMg ___________________ = MgBr (If) 0 0, ,0 M ________________________________ = M css._}µ __ =
CI
(Ig) M M ¨110. =
2 (Ih) Wherein M is Na, K, Li, Cu, CuLi, Sn, Ti, Ca, Mg or Zn.
The detail examples of the synthesis of the bridge linkers are shown in the figures 1-10. Normally the bridge substituents of acetylenedicarboxyl can be condensated with linker components containing function groups capable to react to drugs of desired conjuga-tion.
CELL-BINDING AGENT-DRUG CONJUGATES
The conjugates of the present invention can be represented by the following formula, Cb4DDrugi rug2 11, wherein Cb is a cell-binding agent, L is a linker, Drugl and Drug2 are a drug molecule, n is an integer from 1 to 20, and two S (sulfur) elements from Cb bridgely link to L, which covalently connects two or more drugs (per bridge linker L).
The bridge linker L may be composed of one or more linker components.
Exemplary linker components include 6-maleimidocaproyl ("MC"), maleimidopropanoyl ("MP"), valine-citrulline ("val-cit" or "vc"), alanine-phenylalanine ("ala-phe" or "af"), p-aminobenzyloxycarbonyl ("PAB"), 4-thiopentanoate ("SPP"), 4-(N-maleimidomethyl)-cyclohexane-1 carboxylate ("MCC"), (4-acetyl)aminobenzoate ("SIAB"), 4-thio-butyrate
18 (SPDB), 4-thio-2-hydroxysulfonyl-butyrate (2-Sulfo-SPDB), ethyleneoxy ¨CH2CH20-- as one or more repeating units ('EO" or "PEO"). Additional linker components are known in the art and some are described herein.
Example structures of these components containing linkers are:

H H
0 ; (MC, 6-maleimidocaproyl containing) N,/'\
H NS
H 0 (MP, maleimidopropanoyl containing) (24, HN tit H
01N1.,r-..NS
IT H ?
0 ,(PAB , p-aminobenzyloxycarbonyl containing) H Ni\LIII¨ril?\S 0 0 5L-N \r-rs õe IN
HN.,/===..
0 , 0 0 , .11?\s )21 I 0 0 ig,õ NH p tv....1?\ ".c.,.., '/..........r.-S ==2z, H...
N./..""-1 0 (ME, , , maleimidoethyl containing).

Q
H2N N H H õI C i'LlNI/\/YL0 li If vil N H H2 II HN H1µ1 \T
orS5S. 0" t*--(valine-citrulline containing) t...& ,c00_4<O\"NI ......4.

gls:?--.N1¨NH I* N jUl S N

H

(MCC, 4-(N-maleimidomethyl)cyclohexane-1 carboxylate)
19 0 0fi 0 0 k_11 NH N
1/N N )e221 OH H H
((4-acetyl)aminobenzoate containing) H H
Ap-N..,css H03 S HO3S it (4-thio-2-hydroxysulfonyl-butyrate, 2-sulfo-SPDB) Preferably, the conjugates have the following Formula (II):

Drug-I¨RI X1 /
X2-R2-Drug2 /s _____________________________________________________ In Cb (II) wherein:
Cb represents a cell-binding agent, preferably an antibody;
Drugi and Drug2 represent the same or different cytotoxic agents, linked to the cell-binding agent via the bridge linker through an alkyl, alkylene, alkenylene, alkynylene, ether, polyoxyalkylene, ester, amine, imine, polyamine, hydrazine, hydrazone, amide, urea, semicarbazide, carbazide, alkoxyamine, urethanes, amino acid, peptide, acyloxylamine, hydroxamic acid, disulfide, thioether, thioester, carbamate, carbonate, heterocyclic ring, heteroalkyl, heteroaromatic, or alkoxime bond, or combination thereof.
n is 1 ¨ 20; Ri, R2, Xi and X2 are described the same previously in Formula (I).
As described in more detail below, Drug' and Drug2 can be any of many small mole-cule drugs, including, but not limited to, tubulysins, calicheamicins, auristatins, maytansinoids, CC-1065 analogs, morpholinos doxorubicins, taxanes, cryptophycins, epothilones, and benzodiazepine dimers (e.g., dimmers of pyrrolobenzodiazepine (PBD) or tomaymycin), indolinobenzodiazepines, imidazobenzothiadiazepines, or oxazolidinobenzodiazepines).
To synthesize the conjugate, the cell-binding agent can be first modified with the bridge linkers of the present invention through reduction of disulfide bonds of the cell-binding molecule. The yielded a pair of free thiols can react to the bridge linker of Formula (I) at pH 5-9 aqueous media with or without addition of 0-30% of water mixable (miscible) organic solvents, such as DMA, DMF, ethanol, methanol, acetone, acetonitrile, THE, isopropanol, dioxane, propylene glycol, or ethylene diol, to introduce the reactive groups of Z1 and Z2, whose reactive groups can be a disulfide, maleimido, haloacetyl, azide, 1-yne, ketone, aldehyde, alkoxyamino, or hydrazide. Then the reactive group of a cytotoxic 5 agent reacts to the modified cell-binding molecule accordingly. For example, synthesis of the cell-binding agent-drug conjugates linked via disulfide bonds is achieved by a disulfide exchange between the disulfide bond in the modified cell-binding agent and a drug con-taining a free thiol group. Synthesis of the cell-binding agent-drug conjugates linked via thioether is achieved by reaction of the maleimido or haloacetyl or ethylsulfonyl modified 10 cell-binding agent and a drug containing a free thiol group. Synthesis of conjugates bearing an acid labile hydrazone can be achieved by reaction of a carbonyl group with the hydrazide moiety in the linker, by methods known in the art (see, for example, P. Hamann et al., Hinman, L. M., et al, Cancer Res. 53, 3336-334, 1993; B. Laguzza et al., J. Med.
Chem., 32; 548-555, 1959; P. Trail et al., Cancer Res., 57; 100-105, 1997).
Synthesis of 15 conjugates bearing triazole linkage can be achieved by reaction of a 1-yne group of the drug with the azido moiety in the linker, through the click chemistry (Huisgen cycloaddition) (Lutz, .1-F. ei al, 2008, Adv. Drug Del. Rev, 60, 958-970;
Sletten, E. M. et al 2011, Acc Chem, Research 44, 666-676).
Alternatively, the drug can react with the bridge linkers of the present invention that
20 have conjugated to a cell-binding molecule to give a modified cell-binding molecule linker of Formula (III) bearing functionalities. For example, a thiol-containing drug can be reached with the modified cell-binding molecule bridge linker of Formula (III) bearing a maleimdo, or a haloacetyl, or an ethylsulfonyl substituent at pH 5.5-9.0 in aqueous buffer to give a cell-binding molecule-drug conjugate via a thioether linkage. A
thiol-containing drug can undergo disulfide exchange with a modified bridge linker of Formula (III) bear-ing a pyridyldithio moiety to give a conjugate a disulfide bond linkage. A
drug bearing a hydroxyl group or a thiol group can be reacted with a modified bridge linker of Formula (III) bearing a halogen, particularly the alpha halide of carboxylates, in the presence of a mild base, e.g. pH 8.0-9.5, to give a modified drug bearing an ether or thiol ether link. A
hydroxyl group containing drug can be condensed with a bridge cross linker of Formula (I) bearing a carboxyl group, in the presence of a dehydrating agent, such as EDC
or DCC, to give ester linkage, then the subject drug modified bridge linker undergoes the conjugation with a cell-binding molecule. A drug containing an amino group can condensate with a
21 carboxyl ester of NHS, imidazole, nitrophenol; N-hydroxysuccinimide (NHS);
phenol;
dinitrophenol; pentafluorophenol; tetrafluorophenol; difluorophenol;
monofluorophenol;
pentachlorophenol; triflate; imidazole; dichlorophenol; tetrachlorophenol; 1-hydroxybenzotriazole; tosylate; mesylate; 2-ethyl-5-phenylisoxazolium-3'-sulfonate on the cell-binding molecule-bridge linker of Formula (III) to give a conjugate via amide bond linkage.
The conjugate may be purified by standard biochemical means, such as gel filtration on a Sephadex G25 or Sephacryl S300 column, adsorption chromatography, and ion ex-change or by dialysis. In some cases, a small molecule as a cell-binding agent (e.g. folic acid, melanocyte stimulating hormone, EGF etc) conjugated with a small molecular drugs can be purified by chromatography such as by HPLC, medium pressure column chroma-tography or ion exchange chromatography.
MODIFIED CELL-BINDING AGENTS/MOLECULES
The cell-binding agent modified by reaction with linkers of the present invention are preferably represented by the Formula (III) Z1¨R1 / X2¨R2¨Z2 /S
In Cb (III) Wherein Cb, Zi, Z2, n, R1, R2, X1, and X2 are defined the same as in Formula (I) and (II).
In preferred embodiments, Zi and Z2 are a disulfide substituent, maleimido, haloacetyl, alkoxyamine, azido, ketone, aldehyde, hydrazine group, an N-hydroxysuccinimide ester, or a carboxyl ester formed with phenol; dinitrophenol; pentafluorophenol;
tetrafluorophenol;
difluorophenol; monofluorophenol; pentachlorophenol; triflate; imidazole;
dichlorophenol;
tetrachlorophenol; 1-hydroxybenzotriazole; tosylate; mesylate; 2-ethy1-5-phenylisoxa-zolium-3'-sulfonate. Zi and Z2 can then react with a cytotoxic agent through thioether, hydrazone, amide, alkoxime, carbamate, ester, ether or disulfide bond. The modified cell-binding agent can be prepared via a reaction of the cell-binding agent with the bridge linkers of Formula (I) as described in Formula (II) above.
In order to achieve a higher conjugation yield of the alkyne group on the bridge link-ers with a pair of free thiols on the cell-binding molecule, preferably on an antibody, a
22 small percentage of organic co-solvent may be required to add to the reaction mixture, as well in the solution after the reaction to maintain solubility of the Formula (III) in aqueous solution. To modify the cell-binding agents, the cross-linking reagent (bridge linker) of Formula (I) can be first dissolved in a polar organic solvent that is miscible with water, for example different alcohols, such as methanol, ethanol, and propanol, acetone, acetonitrile, tetrahydrofuran (THF), 1,4-dioxane, dimethyl formamide (DMF), dimethyl acetamide (DMA), or dimethylsulfoxide (DMSO) at a high concentration, for example 1-500 mM.
Meanwhile, the cell-binding molecule, such as antibody dissolved in an aqueous buffer pH
5-9.5, preferably pH 6-8.5, at 1-35 mg/ml concentration was treated with 1-20 equivalent of TCEP or DTT for 20 min to 12 hour. After the reduction, DTT can be removed by SEC
chromatographic purification. TCEP can be optionally removed by SEC
chromatography too, or staying in the reaction mixture for the next step reaction without purification.
Furthermore, the reduction of antibodies or the other cell-binding agents with TCEP can be performed with a bridge linker of Formula (I), for which the cross-linking conjugation for the cell-binding molecules can be achieved simultaneously along with the TCEP
reduction.
The aqueous solutions for the modification of cell-binding agents are buffered be-tween pH 6 and 9, preferably between 6.5 and 7.5 and can contain any non-nucleophilic buffer salts useful for these pH ranges. Typical buffers include phosphate, triethanolamine HC1, HEPES, and MOPS buffers, which can contain additional components, such as cyclodextrins, sucrose and salts, for examples, NaC1 and KC1. After the addition of the bridge linker of Formula (I) into the solution containing the reduced cell-binding molecules, the reaction mixture is incubated at a temperature of from 4 C to 45 C, preferably at ambient temperature. The progress of the reaction can be monitored by measuring the decrease in the absorption at 254 nm, or the other appropriate wavelength.
After the reaction is complete, isolation of the modified cell-binding agent can be performed in a routine way, using for example gel filtration chromatography, or adsorptive chromatog-raphy.
The extent of modification can be assessed by measuring the absorbance of the nitropyridine thione, dinitropyridine dithione, pyridine thione, carboxamidopyridine dithione and dicarboxamidopyridine dithione group released via UV spectra. For the conjugation without a chromophore group, the modification or conjugation reaction can be monitored by LC-MS, preferably by UPLC-QTOF mass spectrometry, or Capil-lary electrophoresis¨mass spectrometry (CEMS). The bridge cross-linkers described herein
23 have diverse functional groups that can react with any drugs, preferably cytotoxic agents that possess a suitable substituent. For examples, the modified cell-binding molecules bearing an amino or hydroxyl substituent can react with drugs bearing an N-hydroxysuccinimide (NHS) ester, the modified cell-binding molecules bearing a thiol substituent can react with drugs bearing a maleimido or haloacetyl group.
Additionally, the modified cell-binding molecules bearing a carbonyl (ketone or aldehyde) substituent can react with drugs bearing a hydrazide or an alkoxyamine. One skilled in the art can readily determine which linker to use based on the known reactivity of the available func-tional group on the linkers.
MODIFIED CYTOTOXIC DRUGS
The cytotoxic drugs modified by reaction with cross-linkers of the present invention are preferably represented by the Formula (IV):

¨
Drug1¨R1 X1 n X2¨ R2 ¨Drug2 (IV) Wherein Drug', Drug2, Zi, Z2, n, R1, R2, Xi, and X2 are defined the same as in Formu-la (I) and (II).
The modified drugs can be prepared via reaction of the drug with the linkers of the Formula (I) to give a modified drug of Formula (IV) bearing functionality of an acetylenedi-carboxyl group capable of reacting with a pair of thiol groups of a cell-binding agent. The acetylenedicarboxyl group is synthesized through condensation with acetylene via the methods described in reaction equation (Ia), (lb), (Ic), (Id), (le), (If), (Ig) and (111).
But for drugs containing a thiol, or the drugs undergoing to link a cell-binding molecule via the bridge linkers through thioether, thioester or disulfide bond, it is therefore preferred that the Drug' or Drug2 may be synthesized to connect to Ri, or R2 in a piece of compo-nents via the linkage of thioether, thioester or disulfide bond first. Then the synthesized R1-Drug' or R2-Drug2 component is assembled to an acetylenedicarboxyl group to form the bridge linker modified drugs of Formula (IV).
For examples of the synthesis, a thiol-containing drug can be reacted with the linker of components R1 or R2 bearing a maleimdo substituent at neutral pH in aqueous buffer to give a RI-Drug' or R2-Drug2 compartment bearing thioether linkage, and following by condensation with a compartment of acetylenedicarboxyl group to give a modified drug of Formula (IV) bearing thioether linkage. A drug bearing a hydroxyl group can be reacted
24 with a linker component R1 or R2 bearing a halogen, or a tosylate, or a mesylate, in the presence of a mild base, to give a Ri-Drugi or R2-Drug2 compartment bearing ether linkage, and following by condensation with a compartment of acetylenedicarboxyl group to give a modified drug of Formula (IV) bearing thioether linkage. A hydroxyl group containing drug can be condensed with a linker of Formula (I) bearing a carboxyl group, in the pres-ence of a dehydrating agent, such as EDC or dicyclohexylcarbodiimide (DCC), to give a modified drug of Formula (IV) via ester linkage. A drug bearing a thiol group can also react the linker of components R1 or R2 bearing a maleimido or a vinylsulfonyl, or a haloacetyl group, give a RI-Drug' or R2-Drug2 compartment bearing thioether linkage, and following by condensation with a compartment of acetylenedicarboxyl group to give a modified drug of Formula (IV) bearing thioether linkage. An amino group containing drug can similarly undergo condensation with a carboxyl group on the bridge linker of Formula (I) to give a modified drug of Formula (IV) bearing amide bonds. The modified drug can be purified by standard methods such as column chromatography over silica gel or alumi-na, crystallization, preparatory thin layer chromatography, ion exchange chromatography, or HPLC.
CELL-BINDING AGENTS
The cell-binding molecule that comprises the conjugates and the modified cell-binding agents of the present invention may be of any kind presently known, or that become known, molecule that binds to, complexes with, or reacts with a moiety of a cell population sought to be therapeutically or otherwise biologically modified.
The cell binding agents include, but are not limited to, large molecular weight proteins such as, for example, full-length antibodies (polyclonal antibodies, monoclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies);
single chain antibodies; fragments of antibodies such as Fab, Fab', F(ab')2, Fv, [Parham, J. Immunol.
131, 2895-2902 (1983)], fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, CDR's, diabody, triabody, and epitope-binding fragments of any of the above which immuno-specifically bind to cancer cell antigens, viral antigens, microbial antigens or a protein generated by the immune system that is capable of recognizing, binding to a specific antigen or exhibiting the desired biological activity (Miller et al (2003) J. of Immunology 170:4854-4861); interferons (such as type I, II, III);
peptides;
lymphokines such as IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, GM-CSF, interferon-gamma (IFN-y); hormones such as insulin, TRH (thyrotropin releasing hormones), MSH
(melano-cyte-stimulating hormone), steroid hormones, such as androgens and estrogens, melano-cyte-stimulating hormone (MSH); growth factors and colony-stimulating factors such as epidermal growth factors (EGF), granulocyte-macrophage colony-stimulating factor (GM-5 CSF), transforming growth factors (TGF), such as TGFa, TGFO, insulin and insulin like growth factors (IGF-I, IGF-II) G-CSF, M-CSF and GM-CSF [Burgess, Immunology Today, 5, 155-158 (1984)[; vaccinia growth factors (VGF); fibroblast growth factors (FGFs); smaller molecular weight proteins, poly-peptide, peptides and peptide hormones, such as bombesin, gastrin, gastrin-releasing peptide; platelet-derived growth factors;
10 interleukin and cytokines, such as interleukin-2 (IL-2), interleukin-6 (IL-6), leukemia inhibitory factors, granulocyte-macrophage colony-stimulating factor (GM-CSF);
vitamins, such as folate; apoproteins and glycoproteins, such as transferrin [O'Keefe et al, 260 J.
Biol. Chem. 932-937 (1985)]; sugar-binding proteins or lipoproteins, such as lectins; cell nutrient-transport molecules; and small molecular inhibitors, such as prostate-specific 15 membrane antigen (PS MA) inhibitors and small molecular tyrosine kinase inhibitors (TKI), non-peptides or any other cell binding molecule or substance, such as bioactive polymers (Dhar, et al, Proc. Natl. Acad. Sci. 2008, 105, 17356-61); bioactive dendrimers (Lee, et al, Nat. Biotechnol. 2005, 23, 1517-26; Almutairi, et al; Proc. Natl.
Acad. Sci.
2009, 106, 685-90); nanoparticles (Liong, et al, ACS Nano, 2008, 19, 1309-12;
Medarova, 20 et al, Nat. Med. 2007, 13, 372-7; Javier, et al, Bioconjugate Chem.
2008, 19, 1309-12);
liposomes (Medinai, et al, Curr. Phar. Des. 2004, 10, 2981-9); viral capsides (Flenniken, et al, Viruses Nanotechnol. 2009, 327, 71-93).
In general, a monoclonal antibody is preferred as a cell-surface binding agent if an ap-propriate one is available. And the antibody may be murine, human, humanized, chimeric,
25 or derived from other species.
Production of antibodies used in the present invention involves in vivo or in vitro pro-cedures or combinations thereof. Methods for producing polyclonal anti-receptor peptide antibodies are well-known in the art, such as in U.S. Pat. No. 4,493,795 (to Nestor et al). A
monoclonal antibody is typically made by fusing myeloma cells with the spleen cells from a mouse that has been immunized with the desired antigen (Kohler, G.;
Milstein, C.
(1975). Nature 256: 495-497). The detailed procedures are described in "Antibodies--A
Laboratory Manual", Harlow and Lane, eds., Cold Spring Harbor Laboratory Press, New York (1988), which is incorporated herein by reference. Particularly monoclonal antibodies
26 are produced by immunizing mice, rats, hamsters or any other mammal with the antigen of interest such as the intact target cell, antigens isolated from the target cell, whole virus, attenuated whole virus, and viral proteins. Splenocytes are typically fused with myeloma cells using polyethylene glycol (PEG) 6000. Fused hybrids are selected by their sensitivity to HAT (hypoxanthine-aminopterin-thymine). Hybridomas producing a monoclonal anti-body useful in practicing this invention are identified by their ability to immunoreact specified receptors or inhibit receptor activity on target cells.
A monoclonal antibody used in the present invention can be produced by initiating a monoclonal hybridoma culture comprising a nutrient medium containing a hybridoma that secretes antibody molecules of the appropriate antigen specificity. The culture is main-tained under conditions and for a time period sufficient for the hybridoma to secrete the antibody molecules into the medium. The antibody-containing medium is then collected.
The antibody molecules can then be further isolated by well-known techniques, such as using protein-A affinity chromatography; anion, cation, hydrophobic, or size exclusive chromatographies (particularly by affinity for the specific antigen after protein A, and sizing column chromatography); centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
Media useful for the preparation of these compositions are both well-known in the art and commercially available and include synthetic culture media. An exemplary synthetic medium is Dulbecco's minimal essential medium (DMEM; Dulbecco et al., Virol.
8, 396 (1959)) supplemented with 4.5 gm/1 glucose, 0-20 mM glutamine, 0-20% fetal calf serum, several ppm amount of heavy metals, such as Cu, Mn, Fe, or Zn, etc, or/and the heavy metals added in their salt forms, and with an anti-foaming agent, such as polyoxyethylene-polyoxypropylene block copolymer.
In addition, antibody-producing cell lines can also be created by techniques other than fusion, such as direct transformation of B lymphocytes with oncogenic DNA, or transfec-tion with an oncovirus, such as Epstein-Barr virus (EBV, also called human herpesvirus 4 (HHV-4)) or Kaposi's sarcoma-associated herpesvirus (KSHV). See, U.S. Pat.
Nos.
4,341,761; 4,399,121; 4,427,783; 4,444,887; 4,451,570; 4,466,917; 4,472,500;
4,491,632;
4,493,890. A monoclonal antibody may also be produced via an anti-receptor peptide or peptides containing the carboxyl terminal as described well-known in the art.
See Niman et al., Proc. Natl. Acad. Sci. USA, 80: 4949-4953 (1983); Geysen et al., Proc.
Natl. Acad. Sci.
USA, 82: 178-182 (1985); Lei et al. Biochemistry 34(20): 6675-6688, (1995).
Typically,
27 the anti-receptor peptide or a peptide analog is used either alone or conjugated to an im-munogenic carrier, as the immunogen for producing anti-receptor peptide monoclonal antibodies.
There are also a number of other well-known techniques for making monoclonal anti-bodies as binding molecules in this invention. Particularly useful are methods of making fully human antibodies. One method is phage display technology which can be used to select a range of human antibodies binding specifically to the antigen using methods of affinity enrichment. Phage display has been thoroughly described in the literature and the construction and screening of phage display libraries are well known in the art, see, e.g., Dente et al, Gene. 148(1):7-13 (1994); Little et al, Biotechnol Adv. 12(3):539-55 (1994);
Clackson et al., Nature 352:264-628 (1991); Huse et al., Science 246:1275-1281 (1989).
Monoclonal antibodies derived by hybridoma technique from another species than human, such as mouse, can be humanized to avoid human anti-mouse antibodies when infused into humans. Among the more common methods of humanization of antibodies are complementarity-determining region grafting and resurfacing. These methods have been extensively described, see e.g. U.S. Pat. Nos. 5,859,205 and 6,797,492; Liu et al, Immunol Rev. 222:9-27 (2008); Almagro et al, Front Biosci. 13: 1619-33 (2008); Lazar et al, Mol Immunol. 44(8):1986-98 (2007); Li et al, Proc. Natl. Acad. Sci. U S A.
103(10):3557-62 (2006) each incorporated herein by reference. Fully human antibodies can also be prepared by immunizing transgenic mice, rabbits, monkeys, or other mammals, carrying large portions of the human immunoglobulin heavy and light chains, with an immunogen.
Examples of such mice are: the Xenomouse. (Abgenix/Amgen), the HuMAb-Mouse (Medarex/BMS), the VelociMouse (Regeneron), see also U.S. Pat. No. 6,596,541, 6,207,418, No. 6,150,584, No. 6,111,166, No. 6,075,181, No. 5,922,545, Nos.
5,661,016, 5,545,806, 5,436,149 and 5,569,825. In human therapy, murine variable regions and hu-man constant regions can also be fused to construct called "chimeric antibodies" that are considerably less immunogenic in man than murine mAbs (Kipriyanov et al, Mol Biotechnol. 26:39-60 (2004); Houdebine, Curr Opin Biotechnol. 13:625-9 (2002) each incorporated herein by reference). In addition, site-directed mutagenesis in the variable region of an antibody can result in an antibody with higher affinity and specificity for its antigen (Brannigan et al, Nat Rev Mol Cell Biol. 3:964-70, (2002)); Adams et al, J
Immunol Methods. 231:249-60 (1999)) and exchanging constant regions of a mAb can improve its ability to mediate effector functions of binding and cytotoxicity.
28 Antibodies immunospecific for a malignant cell antigen can also be obtained commer-cially or produced by any method known to one of skill in the art such as, e.g., chemical synthesis or recombinant expression techniques. The nucleotide sequence encoding anti-bodies immunospecific for a malignant cell antigen can be obtained commercially, e.g., from the GenBank database or a database like it, the literature publications, or by routine cloning and sequencing.
Apart from an antibody, a peptide or protein that bind/block/target or in some other way interact with the epitopes or corresponding receptors on a targeted cell can be used as a binding molecule. These peptides or proteins could be any random peptide or proteins that have an affinity for the epitopes or corresponding receptors and they don't necessarily have to be of the immunoglobulin family. These peptides can be isolated by similar tech-niques as for phage display antibodies (Szardenings, J Recept Signal Transduct Res. 2003;
23(4):307-49). The use of peptides from such random peptide libraries can be similar to antibodies and antibody fragments. The binding molecules of peptides or proteins may be conjugated on or linked to a large molecules or materials, such as, but is not limited, an albumin, a polymer, a liposome, a nano particle, a dendrimer, as long as such attachment permits the peptide or protein to retain its antigen binding specificity.
Examples of antibodies used for conjugation of drugs via the bridge linkers of this prevention for treating cancer, autoimmune disease, and/or infectious disease include, but are not limited to, 3F8 (anti-0D2), Abagovomab (anti CA-125), Abciximab (anti (integrin alpha-IIb), Adalimumab (anti-TNF-a), Adecatumumab (anti-EpCAM, CD326), Afelimomab (anti-TNF-a); Afutuzumab (anti-CD20), Alacizumab pegol (anti-VEGFR2), ALD518 (anti-IL-6), Alemtuzumab (Campath, MabCampath, anti- CD52), Altumomab (anti-CEA), Anatumomab (anti-TAG-72), Anrukinzumab (IMA-638, anti-IL-13), Apolizumab (anti-HLA-DR), Arcitumomab (anti-CEA), Aselizumab (anti-L-selectin (CD62L), Atlizumab (tocilizumab, Actemra, RoActemra, anti-IL-6 receptor), Atorolimumab (anti-Rhesus factor), Bapineuzumab (anti-beta amyloid), Basiliximab (Simulect, antiCD25 (a chain of IL-2 receptor), Bavituximab (anti-phosphatidylserine), Bectumomab (LymphoScan, anti-CD22), Belimumab (Benlysta, LymphoStat-B, anti-BAFF), Benralizumab (anti-CD125), Bertilimumab (anti-CCL11 (eotaxin-1)), Besilesomab (Scintimun, anti-CEA-related antigen), Bevacizumab (Avastin, anti-VEGF-A), Biciromab (FibriScint, anti-fibrin II beta chain), Bivatuzumab (anti-CD44 v6), Blinatumomab (BiTE, anti-CD19), Brentuximab (cAC10, anti-CD30 TNFRSF8),
29 Briakinumab (anti-IL-12, IL-23) Canakinumab (Ilaris, anti-IL-1), Cantuzumab (C242, anti-CanAg), Capromab, Catumaxomab (Removab, anti-EpCAM, anti-CD3), CC49 (anti-TAG-72), Cedelizumab (anti-CD4), Certolizumab pegol (Cimzia anti-TNF-a), Cetuximab (Er-bitux, IMC-C225, anti-EGFR), Citatuzumab bogatox (anti-EpCAM), Cixutumumab (anti-IGF-1), Clenoliximab (anti-CD4), Clivatuzumab (anti-MUC1), Conatumumab (anti-TRAIL-R2), CR6261 (anti-Influenza A hemagglutinin), Dacetuzumab (anti-CD40), Daclizumab (Zenapax, anti-CD25 (a chain of IL-2 receptor)), Daratumumab (anti-(cyclic ADP ribose hydrolase), Denosumab (Prolia, anti-RANKL), Detumomab (anti-B-lymphoma cell), Dorlimomab, Dorlixizumab, Ecromeximab (anti-GD3 ganglioside), Eculizumab (Soliris, anti-CS), Edobacomab (anti-endotoxin), Edrecolomab (Panorex, MAb17-1A, anti-EpCAM), Efalizumab (Raptiva, anti-LEA-1 (CD11a), Efungumab (Mycograb, anti-Hsp90), Elotuzumab (anti-SLAMF7), Elsilimomab (anti-IL-6), Enlimomab pegol (anti-ICAM-1 (CD54)), Epitumomab (anti-episialin), Epratuzumab (anti-CD22), Erlizumab (anti-ITGB2 (CD18)), Ertumaxomab (Rexomun, anti-HER2/neu, CD3), Etaracizumab (Abegrin, anti-integrin a03), Exbivirumab ( anti-hepatitis B surface antigen), Fanolesomab (NeutroSpec, anti-CD15), Faralimomab (anti-interferon receptor), Farletuzumab (anti-folate receptor 1), Felvizumab (anti-respiratory syncytial virus), Fezakinumab (anti-IL-22), Figitumumab (anti-IGF-1 receptor), Fontolizumab (anti-IFN-y), Foravirumab (anti-rabies virus glycoprotein), Fresolimumab (anti-TGF-13), Galiximab (anti-CD80), Gantenerumab (anti- beta amyloid), Gavilimomab (anti-CD147 (basigin)), Gemtuzumab (anti-CD33), Girentuximab (anti-carbonic anhydrase 9), Glembatumumab (CR011, anti-GPNMB), Golimumab (Simponi, anti-TNF-a), Gomiliximab (anti-CD23 (IgE receptor)), lbalizumab (anti-CD4), Ibritumomab (anti-CD20), Igovomab (Indimacis-125, anti-CA-125), Imciromab (Myoscint, anti-cardiac myosin), Infliximab (Remicade, anti-TNF-a), Intetumumab (anti-CD51), Inolimomab (anti-CD25 (a chain of IL-2 recep-tor)), Inotuzumab (anti-CD22), Ipilimumab (anti-CD152), Iratumumab (anti- CD30 (TNFRSF8)), Keliximab (anti-CD4), Labetuzumab (CEA-Cide, anti-CEA), Lebrikizumab (anti- IL-13), Lemalesomab (anti-NCA-90 (granulocyte antigen)), Lerdelimumab (anti-TGF beta 2), Lexatumumab (anti-TRAIL-R2), Libivirumab (anti-hepatitis B
surface antigen), Lintuzumab (anti-CD33), Lucatumumab (anti-CD40), Lumiliximab (anti-(IgE receptor), Mapatumumab (anti-TRAIL-R1), Maslimomab (anti- T-cell receptor), Matuzumab (anti-EGFR), Mepolizumab (Bosatria, anti-IL-5), Metelimumab (anti-TGF
beta 1), Milatuzumab (anti-CD74), Minretumomab (anti-TAG-72), Mitumomab (BEC-2, anti-GD3 ganglioside), Morolimumab (anti-Rhesus factor), Motavizumab (Numax, anti-respiratory syncytial virus), Muromonab-CD3 (Orthoclone OKT3, anti-CD3), Nacolomab (anti-C242), Naptumomab (anti-5T4), Natalizumab (Tysabri, anti-integrin an), Nebacumab (anti-endotoxin), Necitumumab (anti-EGFR), Nerelimomab (anti-TNF-a), Nimotuzumab 5 (Theracim, Theraloc, anti-EGFR), Nofetumomab, Ocrelizumab (anti-CD20), Odulimomab (Afolimomab, anti-LFA-1 (CD11 a)), Ofatumumab (Arzerra, anti-CD20), Olaratumab (anti-PDGF-R a), Omalizumab (Xolair, anti-IgE Fc region), Oportuzumab (anti-EpCAM), Oregovomab (OvaRex, anti-CA-125), Otelixizumab (anti-CD3), Pagibaximab (anti-lipoteichoic acid), Palivizumab (Synagis, Abbosynagis, anti-respiratory syncytial virus), 10 Panitumumab (Vectibix, ABX-EGF, anti-EGFR), Panobacumab (anti-Pseudoinonas aeruginosa), Pascolizumab (anti-IL-4), Pemtumomab (Theragyn, anti-MUC1), Pertuzumab (Omnitarg, 2C4, anti-HER2/neu), Pexelizumab (anti-05), Pintumomab (anti-adenocarcinoma antigen), Priliximab (anti-CD4), Pritumumab (anti-vimentin), (anti-CCR5), Racotumomab (1E10, anti-(N-glycolylneuraminic acid (NeuGc, NGNA)-15 gangliosides GM3)), Rafivirumab (anti-rabies virus glycoprotein), Ramucirumab (anti-VEGFR2), Ranibizumab (Lucentis, anti-VEGF-A), Raxibacumab (anti-anthrax toxin, protective antigen), Regavirumab (anti-cytomegalovirus glycoprotein B), Reslizumab (anti-IL-5), Rilotumumab (anti-HGF), Rituximab (MabThera, Rituxanmab, anti-CD20), Robatumumab (anti-IGF-1 receptor), Rontalizumab (anti-IFN-a), Rovelizumab 20 (LeukArrest, anti-CD11, CD18), Ruplizumab (Antova, anti-CD154 (CD4OL)), Satumomab (anti-TAG-72), Sevirumab (anti-cytomegalovirus), Sibrotuzumab (anti-FAP), Sifalimumab (anti-IFN-a), Siltuximab (anti-IL-6), Siplizumab (anti-CD2), (Smart) MI95 (anti-CD33), Solanezumab (anti-beta amyloid), Sonepcizumab (anti-sphingosine-l-phosphate), Sontuzumab (anti-episialin), Stamulumab (anti-myostatin), Sulesomab (LeukoScan, (anti-25 NCA-90 (granulocyte antigen), Tacatuzumab (anti-alpha-fetoprotein), Tadocizumab (anti-integrin a1b133), Talizumab (anti-IgE), Tanezumab (anti-NGF), Taplitumomab (anti-CD19), Tefibazumab (Aurexis, (anti-clumping factor A), Telimomab, Tenatumomab (anti-tenascin C), Teneliximab (anti-CD40), Teplizumab (anti-CD3), TGN1412 (anti-CD28), Ticilimumab (Tremelimumab, (anti-CTLA-4), Tigatuzumab (anti-TRAIL-R2), TNX-650
30 (anti-IL-13), Tocilizumab (Atlizumab, Actemra, RoActemra, (anti-IL-6 receptor), Toralizumab (anti-CD154 (CD4OL)), Tositumomab (anti-CD20), Trastuzumab (Herceptin, (anti-HER2/neu), Tremelimumab (anti-CTLA-4), Tucotuzumab celmoleukin (anti-EpCAM), Tuvirumab (anti-hepatitis B virus), Urtoxazumab (anti- Escherichia coli),
31 Ustekinumab (Stelara, anti-IL-12, IL-23), Vapaliximab (anti-A0C3 (VAP-1)), Vedolizumab, (anti-integrin 47), Veltuzumab (anti-CD20), Vepalimomab (anti-(VAP-1), Visilizumab (Nuvion, anti-CD3), Vitaxin (anti-vascular integrin avb3), Volociximab (anti-integrin co3,), Votumumab (HumaSPECT, anti-tumor antigen CTAA16.88), Zalutumumab (HuMax-EGFr, (anti-EGFR), Zanolimumab (HuMax-CD4, anti-CD4), Ziralimumab (anti-CD147 (basigin)), Zolimomab (anti-CD5), Etanercept (Enbre110), Alefacept (AmeviveC)), Abatacept (Orencia ), Rilonacept (Arcalyst), 14F7 [anti-MP-2 (Iron Regulatory Protein 2)], 14G2a (anti-GD2 ganglioside, from Nat. Cancer Inst. for melanoma and solid tumors), J591 (anti-PSMA, Weill Cornell Medical School for prostate cancers), 225.285 [anti-HMW-MAA (High molecular weight-melanoma-associated antigen), Sorin Radiofarmaci S.R.L. (Milan, Italy) for melanoma], COL-1 (anti-CEACAM3, CGM1, from Nat. Cancer Inst. USA for colorectal and gastric cancers), CYT-356 (Oncoltad , for prostate cancers), HNK20 (OraVax Inc. for respiratory syncytial virus), ImmuRAIT (from Immunomedics for NHL), Lym-1 (anti-HLA-DR10, Peregrine Pharm. for Cancers), MAK-195F [anti-TNF (tumor necrosis factor; TNFA, TNF-alpha;
TNFSF2), from Abbott / Knoll for Sepsis toxic shock], MEDI-500 [T10B9, anti-CD3, TRaf3 (T cell receptor alpha/beta), complex, from MedImmune Inc for Graft-versus-host disease], RING SCAN [ anti-TAG 72 (tumour associated glycoprotein 72), from Neoprobe Corp. for Breast, Colon and Rectal cancers], Avicidin (anti-EPCAM (epithelial cell adhe-sion molecule), anti-TACSTD1 (Tumor-associated calcium signal transducer 1), anti-GA733-2 (gastrointestinal tumor-associated protein 2), anti-EGP-2 (epithelial glycoprotein 2); anti-KSA; KS1/4 antigen; M4S; tumor antigen 17-1A; CD326, from NeoRx Corp.
for Colon, Ovarian, Prostate cancers and NHL]; LymphoCide (Immunomedics, NJ), Smart ID10 (Protein Design Labs), Oncolym (Techniclone Inc, CA), Allomune (BioTransplant, CA), anti-VEGF (Genentech, CA); CEAcide (Immunomedics, NJ), IMC-1C11 (ImClone Systems, NJ) and Cetuximab (ImClone, NJ) .
Other antibodies as cell binding molecules/ligands include, but are not limited to, are antibodies against the following antigens: Aminopeptidase N (CD13), Annexin Al, B7-H3 (CD276, various cancers), CA125 (ovarian), CA15-3 (carcinomas), CA19-9 (carcinomas), L6 (carcinomas), Lewis Y (carcinomas), Lewis X (carcinomas), alpha fetoprotein (carci-nomas), CA242 (colorectal), placental alkaline phosphatase (carcinomas), prostate specific antigen (prostate), prostatic acid phosphatase (prostate), epidermal growth factor (carcino-mas), CD2 (Hodgkin's disease, NHL lymphoma, multiple myeloma), CD3 epsilon (T
cell
32 lymphoma, lung, breast, gastric, ovarian cancers, autoimmune diseases, malignant ascites), CD19 (B cell malignancies), CD20 (non-Hodgkin's lymphoma), CD22 (leukemia, lym-phoma, multiple myeloma, SLE), CD30 (Hodgkin's lymphoma), CD33 (leukemia, auto-immune diseases), CD38 (multiple myeloma), CD40 (lymphoma, multiple myeloma, leukemia (CLL)), CD51 (Metastatic melanoma, sarcoma), CD52 (leukemia), CD56 (small cell lung cancers, ovarian cancer, Merkel cell carcinoma, and the liquid tumor, multiple myeloma), CD66e (cancers), CD70 (metastatic renal cell carcinoma and non-Hodgkin lymphoma), CD74 (multiple myeloma), CD80 (lymphoma), CD98 (cancers), mucin (car-cinomas), CD221 (solid tumors), CD227 (breast, ovarian cancers), CD262 (NSCLC
and other cancers), CD309 (ovarian cancers), CD326 (solid tumors), CEACAM3 (colorectal, gastric cancers), CEACAM5 (carcinoembryonic antigen; CEA, CD66e) (breast, colorectal and lung cancers), DLL4 (delta-like-4), EGFR (Epidermal Growth Factor Receptor, vari-ous cancers), CTLA4 (melanoma), CXCR4 (CD184, Heme-oncology, solid tumors), Endoglin (CD105, solid tumors), EPCAM (epithelial cell adhesion molecule, bladder, head, neck, colon, NHL prostate, and ovarian cancers), ERBB2 (Epidermal Growth Factor Receptor 2; lung, breast, prostate cancers), FCGR1 (autoimmune diseases), FOLR
(folate receptor, ovarian cancers), GD2 ganglioside (cancers), G-28 (a cell surface antigen glyvolipid, melanoma), GD3 idiotype (cancers), Heat shock proteins (cancers), (lung, stomach cancers), HER2 (breast, lung and ovarian cancers), HLA-DR10 (NHL), HLA-DRB (NHL, B cell leukemia), human chorionic gonadotropin (carcinoma), (insulin-like growth factor 1 receptor, solid tumors, blood cancers), IL-2 receptor (interleu-kin 2 receptor, T-cell leukemia and lymphomas), IL-6R (interleukin 6 receptor, multiple myeloma, RA, Castleman's disease, IL6 dependent tumors), Integrins (avf33, a5431, cc6f34, cd1133, a5135, av135, for various cancers), MAGE-1 (carcinomas), MAGE-2 (carcinomas), MAGE-3 (carcinomas), MAGE 4 (carcinomas), anti-transferrin receptor (carcinomas), p97 (melanoma), MS4A1 (membrane-spanning 4-domains subfamily A member 1, Non-Hodgkin's B cell lymphoma, leukemia), MUC1 or MUC1-KLH (breast, ovarian, cervix, bronchus and gastrointestinal cancer), MUC16 (CA125) (Ovarian cancers), CEA
(colorec-tal), gp100 (melanoma), MART 1 (melanoma), MPG (melanoma), MS4A1 (membrane-spanning 4-domains subfamily A, small cell lung cancers, NHL), Nucleolin, Neu oncogene product (carcinomas), P21 (carcinomas), Paratope of anti-(N-glycolylneuraminic acid, Breast, Melanoma cancers), PLAP-like testicular alkaline phosphatase (ovarian, testicular cancers), PSMA (prostate tumors), PSA (prostate), ROB04, TAG 72 (tumour associated
33 glycoprotein 72, AML, gastric, colorectal, ovarian cancers), T cell transmembrane protein (cancers), Tie (CD202b), TNFRSF1OB (tumor necrosis factor receptor superfamily mem-ber 10B, cancers), TNFRSF13B (tumor necrosis factor receptor superfamily member 13B, multiple myeloma, NHL, other cancers, RA and SLE), TPBG (trophoblast glycoprotein, Renal cell carcinoma), TRAIL-R1 (Tumor necrosis apoprosis Inducing ligand Receptor 1,1ymphoma, NHL, colorectal, lung cancers), VCAM-1 (CD106, Melanoma), VEGF, VEGF-A, VEGF-2 (CD309) (various cancers). Some other tumor associated antigens recognized by antibodies have been reviewed (Gerber, et al, mAbs 1:3, 247-253 (2009);
Novellino et al, Cancer Immunol Immunother. 54(3), 187-207 (2005). Franke, et al, Cancer Biother Radiopharm. 2000, 15, 459-76).
The cell-binding agents, more preferred antibodies, can be any agents that are able to against tumor cells, virus infected cells, microorganism infected cells, parasite infected cells, autoimmune cells, activated cells, myeloid cells, activated T-cells, B
cells, or mela-nocytes. More specifically the cell binding agents can be any agent/molecule that is able to against any one of the following antigens or receptors: CD3, CD4, CD5, CD6, CD7, CD8, CD9, CD10, CD11a, CD11b, CD11c, CD12w, CD14, CD15, CD16, CDw17, CD18, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD36, CD37, CD38, CD39, CD40, CD41, CD42, CD43, CD44, CD45, CD46, CD47, CD48, CD49b, CD49c, CD51, CD52, CD53, CD54, CD55, CD56, CD58, CD59, CD61, CD62E, CD62L, CD62P, CD63, CD66, CD68, CD69, CD70, CD72, CD74, CD79, CD79a, CD79b, CD80, CD81, CD82, CD83, CD86, CD87, CD88, CD89, CD90, CD91, CD95, CD96, CD98, CD100, CD103, CD105, CD106, CD109, CD117, CD120, CD125, CD126, CD127, CD133, CD134, CD135, CD138, CD141, CD142, CD143, CD144, CD147, CD151, CD147, CD152, CD154, CD156, CD158, CD163, CD166, .CD168, CD174, CD180, CD184, CDw186, CD194, CD195, CD200, CD200a, CD200b, CD209, CD221, CD227, CD235a, CD240, CD262, CD271, CD274, CD276 (B7-H3), CD303, CD304, CD309, CD326, 4-1BB, SAC, 5T4 (Trophoblast glycoprotein, TPBG, 5T4, Wnt-Activated Inhibitory Factor 1 or WAIF1), Adenocarcinoma antigen, AGS-5, AGS-22M6, Activin receptor-like kinase 1, AFP, AKAP-4, ALK, Alpha intergrin, Alpha v beta6, Amino-peptidase N, Amyloid beta, Androgen receptor, Angiopoietin 2, Angiopoietin 3, Annexin Al, Anthrax toxin protective antigen, Anti-transferrin receptor, A0C3 (YAP-1), B7-H3, Bacillus anthracis anthrax, BAFF (B-cell activating factor), B-lymphoma cell, bcr-abl, Bombesin, BORIS, C5, C242 antigen, CA125
34 (carbohydrate antigen 125, MUC16), CA-IX (or CAIX, carbonic anhydrase 9), CALLA, CanAg, Canis lupus familiaris IL31, Carbonic anhydrase IX, Cardiac myosin, CCL11(C-C
motif chemokine 11), CCR4 (C-C chemokine receptor type 4, CD194), CCR5, CD3E
(epsilon), CEA (Carcinoembryonic antigen), CEACAM3, CEACAM5 (carcinoembryonic antigen), CFD (Factor D), Ch4D5, Cholecystokinin 2 (CCK2R), CLDN18 (Claudin-18), Clumping factor A, CRIPTO, FCSF1R (Colony stimulating factor 1 receptor, CD115), CSF2 (colony stimulating factor 2, Granulocyte-macrophage colony-stimulating factor (GM-CSF)), CTLA4 (cytotoxic T-lymphocyte-associated protein 4), CTAA16.88 tumor antigen, CXCR4 (CD184), C-X-C chemokine receptor type 4, cyclic ADP ribose hydrolase, Cyclin Bl, CYP1B1, Cytomegalovirus, Cytomegalovirus glycoprotein B, Dabigatran, DLL4 (delta-like-ligand 4), DPP4 (Dipeptidyl-peptidase 4), DR5 (Death receptor 5), E.
coli shiga toxin type-1, E. coli shiga toxin type-2, ED-B, EGFL7 (EGF-like domain-containing protein 7), EGFR, EGFRII, EGFRvIII, Endoglin (CD105), Endothelin B
receptor, Endotoxin, EpCAM (epithelial cell adhesion molecule), EphA2, Episialin, ERBB2 (Epidermal Growth Factor Receptor 2), ERBB3, ERG (TMPRSS2 ETS fusion gene), Escherichia coli, ETV6-AML, FAP (Fibroblast activation protein alpha), FCGR1, alpha-Fetoprotein, Fibrin II, beta chain, Fibronectin extra domain-B, FOLR
(folate recep-tor), Folate receptor alpha, Folate hydrolase, Fos-related antigen 1.F protein of respiratory syncytial virus, Frizzled receptor, Fucosyl GM1, GD2 ganglioside, G-28 (a cell surface antigen glyvolipid), GD3 idiotype, GloboH, Glypican 3, N-glycolylneuraminic acid, GM3, GMCSF receptor a-chain, Growth differentiation factor 8, GP100, GPNMB
(Transmembrane glycoprotein NMB), GUCY2C (Guanylate cyclase 2C, guanylyl cyclase C(GC-C), intestinal Guanylate cyclase, Guanylate cyclase-C receptor, Heat-stable entero-toxin receptor (hSTAR)), Heat shock proteins, Hemagglutinin, Hepatitis B
surface antigen, Hepatitis B virus, HER1 (human epidermal growth factor receptor 1), HER2, HER2/neu, HER3 (ERBB-3), IgG4, HGF/SF (Hepatocyte growth factor/scatter factor), HHGFR, HIV-1, Histone complex, HLA-DR (human leukocyte antigen), HLA-DR10, HLA-DRB , HMWMAA, Human chorionic gonadotropin, HNGF, Human scatter factor receptor kinase, HPV E6/E7, Hsp90, hTERT, ICAM-1 (Intercellular Adhesion Molecule 1), Idiotype, IGF1R (IGF-1, insulin-like growth factor 1 receptor), IGHE, IFN-y, Influeza hemag-glutinin, IgE, IgE Fc region, IGHE, IL-1, IL-2 receptor (interleukin 2 receptor), IL-4, IL-5, IL-6, IL-6R (interleukin 6 receptor), IL-9, IL-10, IL-12, IL-13, IL-17, IL-17A, IL-20, IL-22, 1L-23, IL31RA, ILGF2 (Insulin-like growth factor 2), Integrins (a4, a1b133, avI33, a4137, a5131, a6134, a7[37, a11133, a5135, avI35), Interferon gamma-induced protein, ITGA2, ITGB2, KIR2D, LCK, Le, Legumain, Lewis-Y antigen, LFA-1(Lymphocyte function-associated antigen 1, CD11a), LHRH, LINGO-1, Lipoteichoic acid, LIV1A, LMP2, LTA, MAD-CT-1, MAD-CT-2, MAGE-1, MAGE-2, MAGE-3, MAGE Al, MAGE A3, MAGE 4, 5 MARTI, MCP-1, MW (Macrophage migration inhibitory factor, or glycosylation-inhibiting factor (GIF)), MS4A1 (membrane-spanning 4-domains subfamily A
member 1), MSLN (mesothelin), MUC1(Mucin 1, cell surface associated (MUC1) or polymorphic epithelial mucin (PEM)), MUC1-KLH, MUC16 (CA125), MCP1(monocyte chemotactic protein 1), MelanA/MART1, ML-IAP, MPG, MS4A1 (membrane-spanning 4-domains 10 subfamily A), MYCN, Myelin-associated glycoprotein, Myostatin, NA17, NARP-1, NCA-90 (granulocyte antigen), Nectin-4 (ASG-22ME), NGF, Neural apoptosis-regulated pro-teinase 1, NOGO-A, Notch receptor, Nucleolin, Neu oncogene product, NY-BR-1, NY-ES0-1, OX-40, OxLDL (Oxidized low-density lipoprotein), 0Y-TES1, P21, p53 nonmutant, P97, Page4, PAP, Paratope of anti-(N-glycolylneuraminic acid), PAX3, PAX5, 15 PCSK9, PDCD1 (PD-1, Programmed cell death protein 1,CD279), PDGF-Ra (Alpha-type platelet-derived growth factor receptor), PDGFR-13, PDL-1, PLAC1, PLAP-like testicular alkaline phosphatase, Platelet-derived growth factor receptor beta, Phosphate-sodium co-transporter, PMEL 17, Polysialic acid, Proteinase3 (PR1), Prostatic carcinoma, PS
(Phosphatidylserine), Prostatic carcinoma cells, Pseudomonas aeruginosa, PSMA, PSA, 20 PSCA, Rabies virus glycoprotein, RHD (Rh polypeptide 1 (RhPI), CD240), Rhesus factor, RANKL, RhoC, Ras mutant, RGS5, ROB04, Respiratory syncytial virus, RON, Sarcoma translocation breakpoints, SART3, Sclerostin, SLAMF7 (SLAM family member 7), Selectin P, SDC1 (Syndecan 1), sLe(a), Somatomedin C, SIP (Sphingosine-l-phosphate), Somatostatin, Sperm protein 17, SSX2, STEAP1 (six-transmembrane epitheli-25 al antigen of the prostate 1), STEAP2, STn, TAG-72 (tumor associated glycoprotein 72), Survivin, T-cell receptor, T cell transmembrane protein, TEM1 (Tumor endothelial marker 1), TENB2, Tenascin C (TN-C), TGF-a, TGF-f3 (Transforming growth factor beta), TGF-131, TGF-132 (Transforming growth factor-beta 2), Tie (CD202b), Tie2, TIM-1 (CDX-014), Tn, TNF, TNF-a, TNFRSF8, TNFRSF1OB (tumor necrosis factor receptor superfamily 30 member 10B), TNFRSF13B (tumor necrosis factor receptor superfamily member 13B), TPBG (trophoblast glycoprotein), TRAIL-R1 (Tumor necrosis apoprosis Inducing ligand Receptor 1), TRAILR2 (Death receptor 5 (DR5)), tumor-associated calcium signal trans-ducer 2, tumor specific glycosylation of MUC1, TWEAK receptor, TYRP1(glycoprotein 75), TRP-2, Tyrosinase, VCAM-1 (CD106), VEGF, VEGF-A, VEGF-2 (CD309), VEGFR-1, VEGFR2, or vimentin, WT1, XAGE 1, or cells expressing any insulin growth factor receptors, or any epidermal growth factor receptors.
In another specific embodiment, the cell-binding ligand-drug conjugates via the bridge linkers of this invention are used for the targeted treatment of cancers. The targeted cancers include, but are not limited, Adrenocortical Carcinoma, Anal Cancer, Bladder Cancer, Brain Tumor (Adult, Brain Stem Glioma, Childhood, Cerebellar Astrocytoma, Cerebral Astrocytoma, Ependymoma, Medulloblastoma, Supratentorial Primitive Neuroectodermal and Pineal Tumors, Visual Pathway and Hypothalamic Glioma), Breast Cancer, Carcinoid Tumor, Gastrointestinal, Carcinoma of Unknown Primary, Cervical Cancer, Colon Cancer, Endometrial Cancer, Esophageal Cancer, Extrahepatic Bile Duct Cancer, Ewings Family of Tumors (PNET), Extracranial Germ Cell Tumor, Eye Cancer, Intraocular Melanoma, Gallbladder Cancer, Gastric Cancer (Stomach), Germ Cell Tumor, Extragonadal, Gesta-tional Trophoblastic Tumor, Head and Neck Cancer, Hypopharyngeal Cancer, Islet Cell Carcinoma, Kidney Cancer (renal cell cancer), Laryngeal Cancer, Leukemia (Acute Lym-phoblastic, Acute Myeloid, Chronic Lymphocytic, Chronic Myelogenous, Hairy Cell), Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer (Non-Small Cell, Small Cell, Lym-phoma (AIDS-Related, Central Nervous System, Cutaneous T-Cell, Hodgkin's Disease, Non-Hodgkin's Disease, Malignant Mesothelioma, Melanoma, Merkel Cell Carcinoma, Metasatic Squamous Neck Cancer with Occult Primary, Multiple Myeloma, and Other Plasma Cell Neoplasms, Mycosis Fungoides, Myelodysplastic Syndrome, Myeloproli-ferative Disorders, Nasopharyngeal Cancer, Neuroblastoma, Oral Cancer, Oropharyngeal Cancer, Osteosarcoma, Ovarian Cancer (Epithelial, Germ Cell Tumor, Low Malignant Potential Tumor), Pancreatic Cancer (Exocrine, Islet Cell Carcinoma), Paranasal Sinus and Nasal Cavity Cancer, Parathyroid Cancer, Penile Cancer, Pheochromocytoma Cancer, Pituitary Cancer, Plasma Cell Neoplasm, Prostate Cancer Rhabdomyosarcoma, Rectal Cancer, Renal Cell Cancer (kidney cancer), Renal Pelvis and Ureter (Transitional Cell), Salivary Gland Cancer, Sezary Syndrome, Skin Cancer, Skin Cancer (Cutaneous T-Cell Lymphoma, Kaposi's Sarcoma, Melanoma), Small Intestine Cancer, Soft Tissue Sarcoma, Stomach Cancer, Testicular Cancer, Thymoma (Malignant), Thyroid Cancer, Urethral Cancer, Uterine Cancer (Sarcoma), Unusual Cancer of Childhood, Vaginal Cancer, Vulvar Cancer, Wilms' Tumor.
In another specific embodiment, the cell-binding-drug conjugates via the bridge likers of this invention are used in accordance with the compositions and methods for the treat-ment or prevention of an autoimmune disease. The autoimmune diseases include, but are not limited, Achlorhydra Autoimmune Active Chronic Hepatitis, Acute Disseminated Encephalomyelitis, Acute hemorrhagic leukoencephalitis, Addison's Disease, Agammaglobulinemia, Alopecia areata, Amyotrophic Lateral Sclerosis, Ankylosing Spon-dylitis, Anti-GBM/TBM Nephritis, Antiphospholipid syndrome, Antisynthetase syndrome, Arthritis, Atopic allergy, Atopic Dermatitis, Autoimmune Aplastic Anemia, Autoimmune cardiomyopathy, Autoimmune hemolytic anemia, Autoimmune hepatitis, Autoimmune inner ear disease, Autoimmune lymphoproliferative syndrome, Autoimmune peripheral neuropathy, Autoimmune pancreatitis, Autoimmune polyendocrine syndrome Types I, II, & III, Autoimmune progesterone dermatitis, Autoimmune thrombocytopenic purpura, Autoimmune uveitis, Balo disease/Balo concentric sclerosis, Bechets Syndrome, Berger's disease, Bickerstaff s encephalitis, Blau syndrome, Bullous Pemphigoid, Castleman's disease, Chagas disease, Chronic Fatigue Immune Dysfunction Syndrome, Chronic in-flammatory demyelinating polyneuropathy, Chronic recurrent multifocal ostomyelitis, Chronic lyme disease, Chronic obstructive pulmonary disease, Churg-Strauss syndrome, Cicatricial Pemphigoid, Coeliac Disease, Cogan syndrome, Cold agglutinin disease, Com-plement component 2 deficiency, Cranial arteritis, CREST syndrome, Crohns Disease (a type of idiopathic inflammatory bowel diseases), Cushing's Syndrome, Cutaneous leukocytoclastic angiitis, Dego's disease, Dercum's disease, Dermatitis herpetiformis, Dermatomyositis, Diabetes mellitus type 1, Diffuse cutaneous systemic sclerosis, Dress-ler's syndrome, Discoid lupus erythematosus, Eczema, Endometriosis, Enthesitis-related arthritis, Eosinophilic fasciitis, Epidermolysis bullosa acquisita, Erythema nodosum, Essential mixed cryoglobulinemia, Evan's syndrome, Fibrodysplasia ossificans progressiva, Fibromyalgia, Fibromyositis, Fibrosing aveolitis, Gastritis, Gastrointestinal pemphigoid, Giant cell arteritis, Glomerulonephritis, Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, Haemolytic anaemia, Henoch-Schonlein purpura, Herpes gestationis, Hidradenitis suppurativa, Hughes syndrome (See Antiphospholipid syndrome), Hypogamma-globulinemia, Idiopathic Inflammatory Demyelinating Diseases, Idiopathic pulmonary fibrosis, Idiopathic thrombocytopenic purpura (See Autoimmune thrombocytopenic purpura), IgA nephropathy (Also Berger's disease), Inclusion body myositis, Inflammatory demyelinating polyneuopathy, Interstitial cystitis, Irritable Bowel Syndrome , Juvenile idiopathic arthritis, Juvenile rheumatoid arthritis, Kawasaki's Disease, Lambert-Eaton myasthenic syndrome, Leukocytoclastic vasculitis, Lichen planus, Lichen sclerosus, Linear IgA disease (LAD), Lou Gehrig's Disease (Also Amyotrophic lateral sclerosis), Lupoid hepatitis, Lupus erythematosus, Majeed syndrome, Meniere's disease, Microscopic polyangiitis, Miller-Fisher syndrome, Mixed Connective Tissue Disease, Morphea, Mucha-Habermann disease, Muckle¨Wells syndrome, Multiple Myeloma, Multiple Sclerosis, Myasthenia gravis, Myositis, Narcolepsy, Neuromyelitis optica (Devic's Disease), Neuromyotonia, Occular cicatricial pemphigoid, Opsoclonus myoclonus syndrome, Ord thyroiditis, Palindromic rheumatism, PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus), Paraneoplastic cerebellar degeneration, Parox-ysmal nocturnal hemoglobinuria, Parry Romberg syndrome, Parsonnage-Turner syndrome, Pars planitis, Pemphigus, Pemphigus vulgaris, Pernicious anaemia, Perivenous encephalo-myelitis, POEMS syndrome, Polyarteritis nodosa, Polymyalgia rheumatica, Polymyositis, Primary biliary cirrhosis, Primary sclerosing cholangitis, Progressive inflammatory neu-ropathy, Psoriasis, Psoriatic Arthritis, Pyoderma gangrenosum, Pure red cell aplasia, Rasmussen's encephalitis, Raynaud phenomenon, Relapsing polychondritis, Reiter's syn-drome, Restless leg syndrome, Retroperitoneal fibrosis, Rheumatoid arthritis, Rheumatoid fever, Sarcoidosis, Schizophrenia, Schmidt syndrome, Schnitzler syndrome, Scleritis, Scleroderma, Sjogren's syndrome, Spondyloarthropathy, Sticky blood syndrome, Still's Disease, Stiff person syndrome, Subacute bacterial endocarditis, Susac's syndrome, Sweet syndrome, Sydenham Chorea, Sympathetic ophthalmia, Takayasu's arteritis, Temporal arteritis (giant cell arteritis), Tolosa-Hunt syndrome, Transverse Myelitis, Ulcerative Colitis (a type of idiopathic inflammatory bowel diseases), Undifferentiated connective tissue disease, Undifferentiated spondyloarthropathy, Vasculitis, Vitiligo, Wegener's granulomatosis, Wilson's syndrome, Wiskott-Aldrich syndrome In another specific embodiment, a binding molecule used for the conjugate via the bridge linkers of this invention for the treatment or prevention of an autoimmune disease can be, but are not limited to, anti-elastin antibody; Abys against epithelial cells antibody;
Anti-Basement Membrane Collagen Type IV Protein antibody; Anti-Nuclear Antibody;
Anti ds DNA; Anti ss DNA, Anti Cardiolipin Antibody IgM, IgG; anti-celiac antibody;
Anti Phospholipid Antibody IgK, IgG; Anti SM Antibody; Anti Mitochondrial Antibody;
Thyroid Antibody; Microsomal Antibody, T-cells antibody; Thyroglobulin Antibody, Anti SCL-70; Anti-Jo; Anti-U1RNP; Anti-La/SSB; Anti SSA; Anti SSB; Anti Perital Cells Antibody; Anti Histones; Anti RNP; C-ANCA; P-ANCA; Anti centromere; Anti-Fibrillarin, and Anti GBM Antibody, Anti-ganglioside antibody; Anti-Desmogein 3 anti-body; Anti-p62 antibody; Anti-sp100 antibody; Anti-Mitochondrial(M2) antibody;
Rheu-matoid factor antibody; Anti-MCV antibody; Anti-topoisomerase antibody; Anti-neutrophil cytoplasmic(cANCA) antibody.
In certain preferred embodiments, the binding molecule for the conjugate in the pre-sent invention, can bind to both a receptor and a receptor complex expressed on an activat-ed lymphocyte which is associated with an autoimmune disease. The receptor or receptor complex can comprise an immunoglobulin gene superfamily member (e.g. CD2, CD3, CD4, CD8, CD19, CD20, CD22, CD28, CD30, CD33, CD37, CD38, CD56, CD70, CD79, CD79b, CD90, CD125, CD147, CD152/CTLA-4, PD-1, or ICOS), a TNF receptor super-family member (e.g. CD27, CD40, CD95/Fas, CD134/0X40, CD137/4-1BB, INF-R1, TNFR-2, RANK, TACT, BCMA, osteoprotegerin, Apo2/TRAIL-R1, TRAIL-R2, TRAIL-R3, TRAIL-R4, and APO-3), an integrin, a cytokine receptor, a chemokine receptor, a major histocompatibility protein, a lectin (C-type, S-type, or I-type), or a complement control protein.
In another specific embodiment, useful cell binding ligands that are immunospecific for a viral or a microbial antigen are humanized or human monoclonal antibodies. As used herein, the term "viral antigen" includes, but is not limited to, any viral peptide, polypep-tide protein (e.g. HIV gp120, HIV nef, RSV F glycoprotein, influenza virus neuramimi-dase, influenza virus hemagglutinin, HTLV tax, herpes simplex virus glycoprotein (e.g.
gB, gC, gD, and gE) and hepatitis B surface antigen) that is capable of eliciting an immune response. As used herein, the term "microbial antigen" includes, but is not limited to, any microbial peptide, polypeptide, protein, saccharide, polysaccharide, or lipid molecule (e.g., a bacteria, fungi, pathogenic protozoa, or yeast polypeptides including, e.g., LPS and capsular polysaccharide 5/8) that is capable of eliciting an immune response.
Examples of antibodies available 1 for the viral or microbial infection include, but are not limited to, Palivizumab which is a humanized anti-respiratory syncytial virus monoclonal antibody for the treatment of RSV infection; PR0542 which is a CD4 fusion antibody for the treatment of HIV infection; Ostavir which is a human antibody for the treatment of hepatitis B virus;
PROTVIR which is a humanized IgG1 antibody for the treatment of cytomegalovirus;
and anti-LPS antibodies.

The cell binding molecules¨drug conjugates via the bridge linkers of this invention can be used in the treatment of infectious diseases. These infectious diseases include, but are not limited to, Acinetobacter infections, Actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (Acquired immune deficiency syndrome), Amebiasis, 5 Anaplasmosis, Anthrax, Arcanobacterium haemolyticum infection, Argentine hemorrhagic fever, Ascariasis, Aspergillosis, Astrovirus infection, Babesiosis, Bacillus cereus infection, Bacterial pneumonia, Bacterial vaginosis, Bacteroides infection, Balantidiasis, Baylisascaris infection, BK virus infection, Black piedra, Blastocystis hominis infection, Blastomycosis, Bolivian hemorrhagic fever, Borrelia infection, Botulism (and Infant 10 botulism), Brazilian hemorrhagic fever, Brucellosis, Burkholderia infection, Buruli ulcer, Calicivirus infection (Norovirus and Sapovirus), Campylobacteriosis, Candidiasis (Moniliasis; Thrush), Cat-scratch disease, Cellulitis, Chagas Disease (American trypanosomiasis), Chancroid, Chickenpox, Chlamydia, Chlamydophila pneumoniae infec-tion, Cholera, Chromoblastomycosis, Clonorchiasis, Clostridium difficile infection, 15 Coccidioidomycosis, Colorado tick fever, Common cold (Acute viral rhinopharyngitis;
Acute coryza), Creutzfeldt-Jakob disease, Crimean-Congo hemorrhagic fever, Cryptococ-cosis, Cryptosporidiosis, Cutaneous larva migrans, Cyclosporiasis, Cysticercosis, Cyto-megalovirus infection, Dengue fever, Dientamoebiasis, Diphtheria, Diphyllobothriasis, Dracuneuliasis, Ebola hemorrhagic fever, Echinococcosis, Ehrlichiosis, Enterobiasis 20 (Pinworm infection), Enterococcus infection, Enterovirus infection, Epidemic typhus, Erythema infectiosum (Fifth disease), Exanthem subitum, Fasciolopsiasis, Fasciolosis, Fatal familial insomnia, Filariasis, Food poisoning by Clostridium perfringens, Free-living amebic infection, Fusobacterium infection, Gas gangrene (Clostridial myonecrosis), Geotrichosis, Gerstmann-Straussler-Scheinker syndrome, Giardiasis, Glanders, Gnathosto-25 miasis, Gonorrhea, Granuloma inguinale (Donovanosis), Group A
streptococcal infection, Group B streptococcal infection, Haemophilus influenzae infection, Hand, foot and mouth disease (HFMD), Hantavirus Pulmonary Syndrome, Helicobacter pylori infection, Hemo-lytic-uremic syndrome, Hemorrhagic fever with renal syndrome, Hepatitis A, Hepatitis B, Hepatitis C, Hepatitis D, Hepatitis E, Herpes simplex, Histoplasmosis, Hookworm infec-30 tion, Human bocavirus infection, Human ewingii ehrlichiosis, Human granulocytic anaplasmosis, Human metapneumovirus infection, Human monocytic ehrlichiosis, Human papillomavirus infection, Human parainfluenza virus infection, Hymenolepiasis, Epstein-Barr Virus Infectious Mononucleosis (Mono), Influenza, Isosporiasis, Kawasaki disease, Keratitis, Kingella kingae infection, Kuru, Lassa fever, Legionellosis (Legionnaires' disease), Legionellosis (Pontiac fever), Leishmaniasis, Leprosy, Leptospirosis, Listeriosis, Lyme disease (Lyme borreliosis), Lymphatic filariasis (Elephantiasis), Lymphocytic choriomeningitis, Malaria, Marburg hemorrhagic fever, Measles, Melioidosis (Whitmore's disease), Meningitis, Meningococcal disease, Metagonimiasis, Microsporidiosis, Molluscum contagiosum, Mumps, Murine typhus (Endemic typhus), Mycoplasma pneu-monia, Mycetoma, Myiasis, Neonatal conjunctivitis (Ophthalmia neonatorum), (New) Variant Creutzfeldt-Jakob disease (vCJD, nvCJD), Nocardiosis, Onchocerciasis (River blindness), Paracoccidioidomycosis (South American blastomycosis), Paragonimiasis, Pasteurellosis, Pediculosis capitis (Head lice), Pediculosis corporis (Body lice), Pediculosis pubis (Pubic lice, Crab lice), Pelvic inflammatory disease, Pertussis (Whooping cough), Plague, Pneumococcal infection, Pneumocystis pneumonia, Pneumonia, Poliomyelitis, Prevotella infection, Primary amoebic meningoencephalitis, Progressive multifocal leukoencephalopathy, Psittacosis, Q fever, Rabies, Rat-bite fever, Respiratory syncytial virus infection, Rhinosporidiosis, Rhinovirus infection, Rickettsial infection, Rickettsial-pox, Rift Valley fever, Rocky mountain spotted fever, Rotavirus infection, Rubella, Sal-monellosis, SARS (Severe Acute Respiratory Syndrome), Scabies, Schistosomiasis, Sepsis, Shigellosis (Bacillary dysentery), Shingles (Herpes zoster), Smallpox (Variola), Sporotrichosis, Staphylococcal food poisoning, Staphylococcal infection, Strongyloidiasis, Syphilis, Taeniasis, Tetanus (Lockjaw), Tinea barbae (Barber's itch), Tinea capitis (Ring-worm of the Scalp), Tinea corporis (Ringworm of the Body), Tinea cruris (Jock itch), Tinea manuum (Ringworm of the Hand), Tinea nigra, Tinea pedis (Athlete's foot), Tinea unguium (Onychomycosis), Tinea versicolor (Pityriasis versicolor), Toxocariasis (Ocular Larva Migrans), Toxocariasis (Visceral Larva Migrans), Toxoplasmosis, Trichinellosis, Trichomoniasis, Trichuriasis (Whipworm infection), Tuberculosis, Tularemia, Ureaplasma urealyticum infection, Venezuelan equine encephalitis, Venezuelan hemorrhagic fever, Viral pneumonia, West Nile Fever, White piedra (Tinea blanca), Yersinia pseudotuber-culosis infection, Yersiniosis, Yellow fever, Zygomycosis.
The cell binding molecule, which is more preferred to be an antibody described in this patent that are against pathogenic strains include, but are not limit, Acinetobacter baumannii, Actinomyces israelii, Actinomyces gerencseriae and Propionibacterium propionicus, Trypanosoma brucei, HIV (Human immunodeficiency virus), Entamoeba histolytica, Anaplasma genus, Bacillus anthracis, Arcanobacterium haemolyticum, Junin virus, Ascaris lumbricoides, Aspergillus genus, Astroviridae family, Babesia genus, Bacil-lus cereus, multiple bacteria, Bacteroides genus, Balantidium coli, Baylisascaris genus, BK
virus, Piedraia hortae, Blastocystis hominis, Blastomyces dermatitides, Machupo virus, Borrelia genus, Clostridium botulinum, Sabia, Brucella genus, usually Burkholderia cepacia and other Burkholderia species, Mycobacterium ulcerans, Caliciviridae family, Campylobacter genus, usually Candida albicans and other Candida species, Bartonella henselae, Group A Streptococcus and Staphylococcus, Trypanosoma cruzi, Haemophilus ducreyi, Varicella zoster virus (VZV), Chlamydia trachomatis, Chlamydophila pneumoniae, Vibrio cholerae, Fonsecaea pedrosoi, Clonorchis sinensis, Clostridium difficile, Coccidioides immitis and Coccidioides posadasii, Colorado tick fever virus, rhinoviruses, coronaviruses, CJD prion, Crimean-Congo hemorrhagic fever virus, Crypto-coccus neoformans, Cryptosporidium genus, Ancylostoma braziliense; multiple parasites, Cyclospora cayetanensis, Taenia solium, Cytomegalovirus, Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN-4) ¨ Flaviviruses, Dientamoeba fragilis, Corynebacterium diphtheriae, Diphyllobothrium, Dracunculus medinensis, Ebolavirus, Echinococcus genus, Ehrlichia genus, Enterobius vermicularis, Enterococcus genus, Enterovirus genus, Rickettsia prowazekii, Parvovirus B19, Human herpesvirus 6 and Human herpesvirus 7, Fasciolopsis buski, Fasciola hepatica and Fasciola gigantica, FFI prion, Filarioidea superfamily, Clos-tridium perfringens, Fusobacterium genus, Clostridium perfringens; other Clostridium species, Geotrichum candidum, GSS prion, Giardia intestinalis, Burkholderia mallei, Gnathostoma spinigerum and Gnathostoma hispidum, Neisseria gonorrhoeae, Klebsiella granulomatis, Streptococcus pyogenes, Streptococcus agalactiae, Haemophilus influenzae, Enteroviruses, mainly Coxsackie A virus and Enterovirus 71, Sin Nombre virus, Helico-bacter pylori, Escherichia coli 0157:H7, Bunyaviridae family, Hepatitis A
Virus, Hepatitis B Virus, Hepatitis C Virus, Hepatitis D Virus, Hepatitis E Virus, Herpes simplex virus 1, Herpes simplex virus 2, Histoplasma capsulatum, Ancylostoma duodenale and Necator americanus, Hemophilus influenzae, Human bocavirus, Ehrlichia ewingii, Anaplasma phagocytophilum, Human metapneumovirus, Ehrlichia chaffeensis, Human papilloma-virus, Human parainfluenza viruses, Hymenolepis nana and Hymenolepis diminuta, Ep-stein-Barr Virus, Orthomy-xoviridae family, Isospora belli, Kingella kingae, Klebsiella pneumoniae, Klebsiella ozaenas, Klebsiella rhinoscleromotis, Kuru prion, Lassa virus, Legionella pneumophila, Legionella pneumophila, Leishmania genus, Mycobacterium leprae and Mycobacterium lepromatosis, Leptospira genus, Listeria monocytogenes, Borrelia burgdorferi and other Borrelia species, Wuchereria bancrofti and Brugia malayi, Lymphocytic choriomeningitis virus (LCMV), Plasmodium genus, Marburg virus, Measles virus, Burkholderia pseudomallei, Neisseria meningitides, Metagonimus yokagawai, Microsporidia phylum, Molluscum contagiosum virus (MCV), Mumps virus, Rickettsia typhi, Mycoplasma pneumoniae, numerous species of bacteria (Actinomycetoma) and fungi (Eumycetoma), parasitic dipterous fly larvae, Chlamydia trachomatis and Neisseria gonorrhoeae, vCJD prion, Nocardia asteroides and other Nocardia species, Onchocerca volvulus, Paracoccidioides brasiliensis, Paragonimus westermani and other Paragonimus species, Pasteurella genus, Pediculus humanus capitis, Pediculus humanus corporis, Phthirus pubis, Bordetella pertussis, Yersinia pestis, Streptococcus pneumoniae, Pneumo-cystis jirovecii, Poliovirus, Prevotella genus, Naegleria fowleri, JC virus, Chlamydophila psittaci, Coxiella burnetii, Rabies virus, Streptobacillus moniliformis and Spirillum minus, Respiratory syncytial virus, Rhinosporidium seeberi, Rhinovirus, Rickettsia genus, Rick-ettsia akari, Rift Valley fever virus, Rickettsia rickettsii, Rotavirus, Rubella virus, Salmo-nella genus, SARS coronavirus, Sarcoptes scabiei, Schistosoma genus, Shigella genus, Varicella zoster virus, Variola major or Variola minor, Sporothrix schenckii, Staphylococ-cus genus, Staphylococcus genus, Staphylococcus aureus, Streptococcus pyogenes, Strongyloides stercoralis, Treponema pallidum, Taenia genus, Clostridium tetani, Trichophyton genus, Trichophyton tonsurans, Trichophyton genus, Epidermophyton floccosum, Trichophyton rubrum, and Trichophyton mentagrophytes, Trichophyton rubrum, Hortaea werneckii, Trichophyton genus, Malassezia genus, Toxocara canis or Toxocara cati, Toxoplasma gondii, Trichinella spiralis, Trichomonas vaginalis, Trichuris trichiura, Mycobacterium tuberculosis, Francisella tularensis, Ureaplasma urealyticum, Venezuelan equine encephalitis virus, Vibrio colerae, Guanarito virus, West Nile virus, Trichosporon beigelii, Yersinia pseudotuberculosis, Yersinia enterocolitica, Yellow fever virus, Mucorales order (Mucormycosis) and Entomophthorales order (Entomophthora-mycosis), Pseudomonas aeruginosa, Campylobacter (Vibrio) fetus, Aeromonas hydrophila, Edwardsiella tarda, Yersinia pestis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Salmonella typhimurium, Treponema pertenue, Treponema carateneum, Borrelia vincentii, Bonelia burgdorferi, Leptospira icterohemorrhagiae, Pneumocystis carinii, Brucella abortus, Brucella suis, Brucella melitensis, Mycoplasma spp., Rickettsia prowazeki, Rick-ettsia tsutsugumushi, Clamydia spp.; pathogenic fungi (Aspergillus fumigatus, Candida albicans, Histoplasma capsulatum); protozoa (Entomoeba histolytica, Trichomonas tenas, Trichomonas hominis, Tryoanosoma gambiense, Trypano soma rhodesiense, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Pneumocystis pneumonia, Plasmo-dium vivax, Plasmodium falciparum, Plasmodium malaria); or Helminiths (Schistosoma japonicum, Schistosoma mansoni, Schistosoma haematobium, and hookworms).
Other antibodies as cell binding ligands used in this invention for treatment of viral disease include, but are not limited to, antibodies against antigens of pathogenic viruses, including as examples and not by limitation: Poxyiridae, Herpesviridae, Adenoviridae, Papovaviridae, Enteroviridae, Picomaviridae, Parvoviridae, Reoviridae, Retroviridae, influenza viruses, parainfluenza viruses, mumps, measles, respiratory syncytial virus, rubella, Arboviridae, Rhabdoviridae, Arenaviridae, Non-A/Non-B Hepatitis virus, Rhinoviridae, Coronaviridae, Rotoviridae, Oncovirus [such as, HBV
(Hepatocellular carcinoma), HPV (Cervical cancer, Anal cancer), Kaposi's sarcoma-associated herpesvirus (Kaposi's sarcoma), Epstein-Barr virus (Nasopharyngeal carcinoma, Burkitt's lymphoma, Primary central nervous system lymphoma), MCPyV (Merkel cell cancer), SV40 (Simian virus 40), HCV (Hepatocellular carcinoma), HTLV-I (Adult T-cell leukemia/lymphoma)], Immune disorders caused virus: [such as Human Immunodeficiency Virus (AIDS)];
Cen-tral nervous system virus: [such as, JCV (Progressive multifocal leukoencephalopathy), MeV (Subacute sclerosing panencephalitis), LCV (Lymphocytic choriomeningitis), Arbovirus encephalitis, Orthomyxoviridae (probable) (Encephalitis lethargica), RV (Ra-bies), Chandipura virus, Herpesviral meningitis, Ramsay Hunt syndrome type II;
Po-liovirus (Poliomyelitis, Post-polio syndrome), HTLV-I (Tropical spastic paraparesis)];
Cytomegalovirus (Cytomegalovirus retinitis, HSV (Herpetic keratitis));
Cardiovascular virus [such as CBV (Pericarditis, Myocarditis)]; Respiratory system/acute viral nasopharyngitis/viral pneumonia: [Epstein-Barr virus (EBV infection/Infectious mononu-cleosis), Cytomegalovirus; SARS coronavirus (Severe acute respiratory syndrome) Orthomyxoviridae: Influenzavirus A/B/C (Influenza/Avian influenza), Paramyxovirus:
Human parainfluenza viruses (Parainfluenza), RSV (Human respiratory syncytial virus), hMPV]; Digestive system virus [MuV (Mumps), Cytomegalovirus (Cytomegalovirus esophagitis); Adenovirus (Adenovirus infection); Rotavirus, Norovirus, Astrovirus, Coro-navirus; HBV (Hepatitis B virus), CBV, HAY (Hepatitis A virus), HCV (Hepatitis C
virus), HDV (Hepatitis D virus), HEY (Hepatitis E virus), HGV (Hepatitis G
virus)];
Urogenital virus [such as, BK virus, MuV (Mumps)].

According to a further object, the present invention also concerns pharmaceutical compositions comprising the conjugate via the bridge linkers of the invention together with a pharmaceutically acceptable carrier, diluent, or excipient for treatment of cancers, infec-tions or autoimmune disorders. The method for treatment of cancers, infections and auto-5 immune disorders can be practiced in vitro, in vivo, or ex vivo. Examples of in vitro uses include treatments of cell cultures in order to kill all cells except for desired variants that do not express the target antigen; or to kill variants that express undesired antigen. Exam-ples of ex vivo uses include treatments of hematopoietic stem cells (HSC) prior to the performance of the transplantation (HSCT) into the same patient in order to kill diseased or 10 malignant cells. For instance, clinical ex vivo treatment to remove tumour cells or lym-phoid cells from bone marrow prior to autologous transplantation in cancer treatment or in treatment of autoimmune disease, or to remove T cells and other lymphoid cells from allogeneic bone marrow or tissue prior to transplant in order to prevent graft-versus-host disease, can be carried out as follows. Bone marrow is harvested from the patient or other 15 individual and then incubated in medium containing serum to which is added the conjugate of the invention, concentrations range from about 1 pM to 0.1 mM, for about 30 minutes to about 48 hours at about 37 C. The exact conditions of concentration and time of incuba-tion (=dose) are readily determined by the skilled clinicians. After incubation, the bone marrow cells are washed with medium containing serum and returned to the patient by i.v.
20 infusion according to known methods. In circumstances where the patient receives other treatment such as a course of ablative chemotherapy or total-body irradiation between the time of harvest of the marrow and reinfusion of the treated cells, the treated marrow cells are stored frozen in liquid nitrogen using standard medical equipment.
For clinical in vivo use, the conjugate via the linkers of the invention will be supplied 25 as solutions or as a lyophilized solid that can be redissolved in sterile water for injection.
Examples of suitable protocols of conjugate administration are as follows.
Conjugates are given weekly for 8-20 weeks as an i.v. bolus. Bolus doses are given in 50 to 500 ml of normal saline to which human serum albumin (e.g. 0.5 to 1 mL of a concentrated solution of human serum albumin, 100 mg/mL) can be added. Dosages will be about 501..tg to 20 30 mg/kg of body weight per week, i.v. (range of 10 lag to 200 mg/kg per injection). 4-20 weeks after treatment, the patient may receive a second course of treatment.
Specific clinical protocols with regard to route of administration, excipients, diluents, dosages, times, etc., can be determined by the skilled clinicians.

Examples of medical conditions that can be treated according to the in vivo or ex vivo methods of killing selected cell populations include malignancy of any types of cancer, autoimmune diseases, graft rejections, and infections (viral, bacterial or parasite).
The amount of a conjugate which is required to achieve the desired biological effect, will vary depending upon a number of factors, including the chemical characteristics, the potency, and the bioavailability of the conjugates, the type of disease, the species to which the patient belongs, the diseased state of the patient, the route of administration, all factors which dictate the required dose amounts, delivery and regimen to be administered.
In general terms, the conjugates via the linkers of this invention may be provided in an aqueous physiological buffer solution containing 0.1 to 10% w/v conjugates for parenteral administration. Typical dose ranges are from 1 g/kg to 0.1 g/kg of body weight per day; a preferred dose range is from 0.01 mg/kg to 20 mg/kg of body weight per day, or per week, or an equivalent dose in a human child. The preferred dosage of drug to be administered is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, the formulation of the compound, the route of administration (intravenous, intramuscular, or other), the pharmacokinetic properties of the conjugates by the chosen delivery route, and the speed (bolus or continuous infusion) and schedule of administrations (number of repetitions in a given period of time).
The conjugates via the linkers of the present invention are also capable of being ad-ministered in unit dose forms, wherein the term "unit dose" means a single dose which is capable of being administered to a patient, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose comprising either the active conjugate itself, or as a pharmaceutically acceptable composition, as described hereinafter.
As such, typical total daily/weekly/biweekly/monthly dose ranges are from 0.01 to 100 mg/kg of body weight. By way of general guidance, unit doses for humans range from 1 mg to 3000 mg per day, or per week, per two week or per month. Preferably the unit dose range is from 1 to 500 mg administered one to four times a week, and even more prefera-bly from 1 mg to 100 mg, once a week. Conjugates provided herein can be formulated into pharmaceutical compositions by admixture with one or more pharmaceutically acceptable excipients. Such unit dose compositions may be prepared for use by oral administration, particularly in the form of tablets, simple capsules or soft gel capsules; or intranasal, particularly in the form of powders, nasal drops, or aerosols; or dermally, for example, topically in ointments, creams, lotions, gels or sprays, or via trans-dermal patches.
DRUGS/CYTOTOXIC AGENTS
Drugs that can be conjugated to a cell-binding molecule in the present invention are small molecule drugs including cytotoxic agents, which can be linked to or after they are modified for linkage to the cell-binding agent. A "small molecule drug" is broadly used herein to refer to an organic, inorganic, or organometallic compound that may have a molecular weight of for example 100 to 1800, more suitably from 120 to 1400.
Small molecule drugs are well characterized in the art, such as in W005058367A2, and in U.S.
Patent No. 4,956,303, among others and are incorporated in their entirety by reference. The drugs include known drugs and those that may become known drugs.
Drugs that are known include, but not limited to, 1). Chemotherapeutic agents: a). Alkylating agents: such as Nitrogen mustards:

chlorambucil, chlornaphazine, cyclophosphamide, dacarbazine, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, mannomustine, mitobronitol, melphalan, mitolactol, pipobroman, novembichin, phenesterine, prednimustine, thiotepa, trofosfamide, uracil mustard; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); Duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); Benzodiazepine dimers (e.g., dimmers of pyrrolobenzodiazepine (PBD) or tomaymycin, indolinobenzodiazepines, imidazobenzothiadiazepines, or oxazolidino-benzodiazepines); Nitrosoureas: (carmustine, lomustine, chlorozotocin, fotemustine, nimustine, ranimustine); Alkylsulphonates: (busulfan, treosulfan, improsulfan and piposulfan); Triazenes: (dacarbazine); Platinum containing compounds:
(carboplatin, cisplatin, oxaliplatin); aziridines, such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemel-amine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomel-amine]; b). Plant Alkaloids: such as Vinca alkaloids: (vincristine, vinblastine, vindesine, vinorelbine, navelbin); Taxoids: (paclitaxel, docetaxol) and their analogs, Maytansinoids (DM1, DM2, DM3, DM4, maytansine and ansamitocins) and their analogs, cryptophycins (particularly cryptophycin 1 and cryptophycin 8); epothilones, eleutherobin, discodermo-lide, bryostatins, dolostatins, auristatins, tubulysins, cephalostatins;
pancratistatin; a sarcodictyin; spongistatin; c). DNA Topoisomerase Inhibitors: such as [Epipodophyllins:

(9-aminocamptothecin, camptothecin, crisnatol, daunomycin, etoposide, etoposide phos-phate, irinotecan, mitoxantrone, novantrone, retinoic acids (retinols), teniposide, topotecan, 9-nitrocamptothecin (RFS 2000)); mitomycins: (mitomycin C)]; d). Anti-metabolites: such as { [Anti-folate: DHFR inhibitors: (methotrexate, trimetrexate, denopterin, pteropterin, aminopterin (4-aminopteroic acid) or the other folic acid analogues); IMP
dehydrogenase Inhibitors: (mycophenolic acid, tiazofurin, ribavirin, EICAR); Ribonucleotide reductase Inhibitors: (hydroxyurea, deferoxamine)]; [Pyrimidine analogs: Uracil analogs:

(ancitabine, azacitidine, 6-azauridine, capecitabine (Xeloda), carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, 5-Fluorouracil, floxuridine, ratitrexed (Tomudex)); Cytosine analogs: (cytarabine, cytosine arabinoside, fludarabine);
Purine analogs: (azathioprine, fludarabine, mercaptopurine, thiamiprine, thioguanine)]; folic acid replenisher, such as frolinic acid}; e). Hormonal therapies: such as {Receptor antagonists:
[Anti-estrogen: (megestrol, raloxifene, tamoxifen); LHRH agonists: (goscrclin, leuprolide acetate); Anti-androgens: (bicalutamide, flutamide, calusterone, dromostanolone propio-nate, epitiostanol, goserelin, leuprolide, mepitiostane, nilutamide, testolactone, trilostane and other androgens inhibitors)]; Retinoids/Deltoids: [Vitamin D3 analogs: (CB
1093, EB
1089 KH 1060, cholecalciferol, ergocalciferol); Photodynamic therapies:
(verteporfin, phthalocyanine, photosensitizer Pc4, demethoxy-hypocrellin A); Cytokines:
(Interferon-alpha, Interferon-gamma, tumor necrosis factor (TNFs), human proteins containing a TNF
domain)]}; f). Kinase inhibitors, such as BMW 2992 (anti-EGFR/Erb2), imatinib, gefitinib, pegaptanib, sorafenib, dasatinib, sunitinib, erlotinib, nilotinib, lapatinib, axitinib, pazopanib. vandetanib, E7080 (anti-VEGFR2), mubritinib, ponatinib (AP24534), bafetinib (INNO-406), bosutinib (SKI-606), cabozantinib, vismodegib, iniparib, ruxolitinib, CYT387, axitinib, tivozanib, sorafenib, bevacizumab, cetuximab, Trastuzumab, Ranibizumab, Panitumumab, ispinesib; g). antibiotics, such as the enediyne antibiotics (e.g. calicheamicins, especially calicheamicin y1,61, al and 131, see, e.g., J. Med. Chem., 39 (11), 2103-2117 (1996), Angew Chem Intl. Ed. Engl. 33:183-186 (1994);
dynemicin, including dynemicin A and deoxydynemicin; esperamicin, kedarcidin, C-1027, maduropeptin, as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin;
chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, nitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; f). Others: such as Polyketides (acetogenins), especially bullatacin and bullatacinone; gemcitabine, epoxomicins (e. g. carfilzomib), bortezomib, thalidomide, lenalidomide, pomalidomide, tosedostat, zybrestat, PLX4032, STA-9090, Stimuvax, allovectin-7, Xegeva, Provenge, Yervoy, Isoprenylation inhibitors (such as Lovastatin), Dopaminergic neurotoxins (such as 1-methy1-4-phenylpyridinium ion), Cell cycle inhibi-tors (such as staurosporine), Actinomycins (such as Actinomycin D, dactinomycin), Bleomycins (such as bleomycin A2, bleomycin B2, peplomycin), Anthracyclines (such as daunorubicin, doxorubicin (adriamycin), idarubicin, epirubicin, pirarubicin, zorubicin, mtoxantrone, MDR inhibitors (such as verapamil), Ca2+ATPase inhibitors (such as thapsigargin), Histone deacetylase inhibitors (Vorinostat, Romidepsin, Panobinostat, Valproic acid, Mocetinostat (MGCD0103), Belinostat, PCI-24781, Entinostat, SB939, Resminostat, Givinostat, AR-42, CUDC-101, sulforaphane, Trichostatin A) ;
Thapsigargin, Celecoxib, glitazones, epigallocatechin gallate, Disulfiram, Salinosporamide A.; Anti-adrenals, such as aminoglutethimide, mitotane, trilostane; aceglatone;
aldophosphamide glycoside; aminolevulinic acid; amsacrine; arabinoside, bestrabucil;
bisantrene; edatraxate;
defofamine; demecolcine; diaziquone; eflornithine (DFMO), elfomithine;
elliptinium acetate, etoglucid; gallium nitrate; gacytosine, hydroxyurea; ibandronate, lentinan;
lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin;
phenamet;
pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK ;
razoxane; rhizoxin;
sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2, 2',2"-trichlorotriethylamine;
trichothecenes (especially T-2 toxin, verrucarin A, roridin A and anguidine);
urethane, siRNA, antisense drugs, and a nucleolytic enzyme.
2). An anti-autoimmune disease agent includes, but is not limited to, cyclosporine, cy-closporine A, aminocaproic acid, azathioprine, bromocriptine, chlorambucil, chloroquine, cyclophosphamide, corticosteroids (e.g. amcinonide, betamethasone, budesonide, hydro-cortisone, flunisolide, fluticasone propionate, fluocortolone danazol, dexamethasone, Triamcinolone acetonide, beclometasone dipropionate), DHEA, enanercept, hydroxychloroquine, infliximab, meloxicam, methotrexate, mofetil, mycophenylate, pred-nisone, sirolimus, tacrolimus.

3). An anti-infectious disease agent includes, but is not limited to, a).
Aminoglyco-sides: amikacin, astromicin, gentamicin (netilmicin, sisomicin, isepamicin), hygromycin B, kanamycin (amikacin, arbekacin, bekanamycin, dibekacin, tobramycin), neomycin (framycetin, paromomycin, ribostamycin), netilmicin, spectinomycin, streptomycin, to-5 bramycin, verdamicin; b). Amphenicols: azidamfenicol, chloramphenicol, florfenicol, thiamphenicol; c). Ansamycins: geldanamycin, herbimycin; d). Carbapenems:
biapenem, doripenem, ertapenem, imipenem/cilastatin, meropenem, panipenem; e). Cephems:
carbacephem (loracarbef), cefacetrile, cefaclor, cefradine, cefadroxil, cefalonium, cefaloridine, cefalotin or cefalothin, cefalexin, cefaloglycin, cefamandole, cefapirin, 10 cefatrizine, cefazaflur, cefazedone, cefazolin, cefbuperazone, cefcapene, cefdaloxime, cefepime, cefminox, cefoxitin, cefprozil, cefroxadine, ceftezole, cefuroxime, cefixime, cefdinir, cefditoren, cefepime, cefetamet, cefmenoxime, cefodizime, cefonicid, cefoperazone, ceforanide, cefotaxime, cefotiam, cefozopran, cephalexin, cefpimizole, cefpiramide, cefpirome, cefpodoxime, cefprozil, cefquinome, cefsulodin, ceftazidime, 15 cefteram, ceftibuten, ceftiolene, ceftizoxime, ceftobiprole, ceftriaxone, cefuroxime, cefuzonam, cephamycin (cefoxitin, cefotetan, cefmetazole), oxacephem (flomoxef, latamoxef); f). Glycopeptides: bleomycin, vancomycin (oritavancin, telavancin), teicoplanin (dalbavancin), ramoplanin; g). Glycylcyclines: e. g. tigecycline;
g). 13-Lactamase inhibitors: penam (sulbactam, tazobactam), clavam (clavulanic acid);
i).
20 Linco s amides: clindamycin, lincomycin; j). Lipopeptides: daptomycin, A54145, calcium-dependent antibiotics (CDA); k). Macrolides: azithromycin, cethromycin, clarithromycin, dirithromycin, erythromycin, flurithromycin, josamycin, ketolide (telithromycin, cethromycin), midecamycin, miocamycin, oleandomycin, rifamycins (rifampicin, rifampin, rifabutin, rifapentine), rokitamycin, roxithromycin, spectinomycin, spiramycin, tacrolimus 25 (FK506), troleandomycin, telithromycin; 1). Monobactams: aztreonam, tigemonam; m).
Oxazolidinones: linezolid; n). Penicillins: amoxicillin, ampicillin (pivampicillin, hetacillin, bacampicillin, metampicillin, talampicillin), azidocillin, azlocillin, benzylpenicillin, benzathine benzylpenicillin, benzathine phenoxymethyl-penicillin, clometocillin, procaine benzylpenicillin, carbenicillin (carindacillin), cloxacillin, dicloxacillin, epicillin, 30 flucloxacillin, mecillinam (pivmecillinam), mezlocillin, meticillin, nafcillin, oxacillin, penamecillin, penicillin, pheneticillin, phenoxymethylpenicillin, piperacillin, propicillin, sulbenicillin, temocillin, ticarcillin; o). Polypeptides: bacitracin, colistin, polymyxin B; p).
Quinolones: alatrofloxacin, balofloxacin, ciprofloxacin, clinafloxacin, danofloxacin, difloxacin, enoxacin, enrofloxacin, floxin, garenoxacin, gatifloxacin, gemifloxacin, grepafloxacin, kano trovafloxacin, levofloxacin, lomefloxacin, marbofloxacin, moxifloxacin, nadifloxacin, norfloxacin, orbifloxacin, ofloxacin, pefloxacin, trovafloxacin, grepafloxacin, sitafloxacin, sparfloxacin, temafloxacin, tosufloxacin, trovafloxacin; q).
Streptogramins: pristinamycin, quinupristin/dalfopristin); r). Sulfonamides:
mafenide, prontosil, sulfacetamide, sulfamethizole, sulfanilimide, sulfasalazine, sulfisoxazole, trime-thoprim, trimethoprim-sulfamethoxazole (co-trimoxazole); s). Steroid antibacterials: e.g.
fusidic acid; t). Tetracyclines: doxycycline, chlortetracycline, clomocycline, demeclocycline, lymecycline, meclocycline, metacycline, minocycline, oxytetracycline, penimepicycline, rolitetracycline, tetracycline, glycylcyclines (e.g.
tigecycline); u). Other types of antibiotics: annonacin, arsphenamine, bactoprenol inhibitors (Bacitracin), DADAL/AR inhibitors (cycloserine), dictyostatin, discodermolide, eleutherobin, epothilone, ethambutol, etoposide, faropenem, fusidic acid, furazolidone, isoniazid, laulimalide, metronidazole, mupirocin, mycolactone, NAM synthesis inhibitors (e. g.
fosfomycin), nitrofurantoin, paclitaxel, platensimycin, pyrazinamide, quinupristin/dalfopristin, rifampicin (rifampin), tazobactam tinidazole, uvaricin;
4). Anti-viral drugs: a). Entry/fusion inhibitors: aplaviroc, maraviroc, vicriviroc, gp41 (enfuvirtide), PRO 140, CD4 (ibalizumab); b). Integrase inhibitors:
raltegravir, elvitegravir, globoidnan A; c). Maturation inhibitors: bevirimat, vivecon; d).
Neuramini-dase inhibitors: oseltamivir, zanamivir, peramivir; e). Nucleosides &nucleotides: abacavir, aciclovir, adefovir, amdoxovir, apricitabine, brivudine, cidofovir, clevudine, dexelvucitabine, didanosine (ddI), elvucitabine, emtricitabine (FTC), entecavir, famciclovir, fluorouracil (5-FU), 3'-fluoro-substituted 2', 3'-dideoxynucleoside analogues (e.g. 3'-fluoro-2',3'-dideoxythymidine (FLT) and 3'-fluoro-2',3'-dideoxyguanosine (FLG), fomivirsen, ganciclovir, idoxuridine, lamivudine (3TC), 1-nucleosides (e.g. fi-l-thymidine and /3-1-2'-deoxycytidine), penciclovir, racivir, ribavirin, stampidine, stavudine (d4T), taribavirin (viramidine), telbivudine, tenofovir, trifluridine valaciclovir, valganciclovir, zalcitabine (ddC), zidovudine (AZT); f). Non-nucleosides:
amantadine, ateviridine, capravirine, diarylpyrimidines (etravirine, rilpivirine), delavirdine, docosanol, emivirine, efavirenz, foscarnet (phosphonoformic acid), imiquimod, interferon alfa, loviride, lodenosine, methisazone, nevirapine, NOV-205, peginterferon alfa, podophyllotoxin, rifampicin, rimantadine, resiquimod (R-848), tromantadine;
g). Protease inhibitors: amprenavir, atazanavir, boceprevir, darunavir, fosamprenavir, indinavir, lopinavir, nelfinavir, pleconaril, ritonavir, saquinavir, telaprevir (VX-950), tipranavir; h).
Other types of anti-virus drugs: abzyme, arbidol, calanolide a, ceragenin, cyanovirin-n, diarylpyrimidines, epigallocatechin gallate (EGCG), foscarnet, griffithsin, taribavirin (viramidine), hydroxyurea, KP-1461, miltefosine, pleconaril, portmanteau inhibitors, ribavirin, seliciclib.
5). The drugs used for conjugates via a bridge linker of the present invention also 3-11,-.
in-clude radioisotopes. Examples of radioisotopes (radionuclides) are 1-1, t-, 14,-. 18- 32-
35 64 68 86 99 111 123 124 125 131 133 177 211 213 =
S, Cu, Ga, Y, Tc, In, I, L L L Xe, Lu, At, or Bt. Radioiso-tope labeled antibodies are useful in receptor targeted imaging experiments or can be for targeted treatment such as with the antibody-drug conjugates of the invention (Wu et al (2005) Nature Biotechnology 23(9): 1137-1146). The cell binding molecules, e.g. an antibody can be labeled with ligand reagents through the bridge linkers of the present patent that bind, chelate or otherwise complex a radioisotope metal, using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991). Chelating ligands which may complex a metal ion include DOTA, DOTP, DOTMA, DTPA and TETA (Macrocyclics, Dallas, Tex.).
6). The pharmaceutically acceptable salts, acids or derivatives of any of the above drugs.
In another embodiment, the drug in the Formula (II) and (IV) can a chromophore mol-ecule, for which the conjugate can be used for detection, monitoring, or study the interac-tion of the cell binding molecule with a target cell. Chromophore molecules are a com-pound that have the ability to absorb a kind of light, such as UV light, florescent light, IR
light, near IR light, visual light; A chromatophore molecule includes a class or subclass of xanthophores, erythrophores, iridophores, leucophores, melanophores, and cyanophores; a class or subclass of fluorophore molecules which are fluorescent chemical compounds re-emitting light upon light; a class or subclass of visual phototransduction molecules; a class or subclass of photophore molecules; a class or subclass of luminescence molecules; and a class or subclass of luciferin compounds.
The chromophore molecule can be selected from, but not limited, Non-protein organic fluorophores, such as: Xanthene derivatives (fluorescein, rhodamine, Oregon green, eosin, and Texas red); Cyanine derivatives: (cyanine, indocarbocyanine, oxacarbocyanine, thiacarbocyanine, and merocyanine); Squaraine derivatives and ring-substituted squaraines, including Seta, SeTau, and Square dyes; Naphthalene derivatives (dansyl and prodan derivatives); Coumarin derivatives; Oxadiazole derivatives (pyridyloxazole, nitrobenzoxadiazole and benzoxadiazole); Anthracene derivatives (anthraquinones, includ-ing DRAQ5, DRAQ7 and CyTRAK Orange); Pyrene derivatives (cascade blue, etc);
Oxazine derivatives (Nile red, Nile blue, cresyl violet, oxazine 170 etc).
Acridine deriva-tives (proflavin, acridine orange, acridine yellow etc). Arylmethine derivatives (auramine, crystal violet, malachite green). Tetrapyrrole derivatives (porphin, phthalocyanine, biliru-bin).
Or a chromophore molecule can be selected from any analogs and derivatives of the following fluorophore compounds: CF dye (Biotium), DRAQ and CyTRAK probes (BioStatus), BODIPY (Invitrogen), Alexa Fluor (Invitrogen), DyLight Fluor (Thermo Scientific, Pierce), Atto and Tracy (Sigma Aldrich), FluoProbes (Interchim), Abberior Dyes (Abberior), DY and MegaStokes Dyes (Dyomics), Sulfo Cy dyes (Cyandye), HiLyte Fluor (AnaSpec), Seta, SeTau and Square Dyes (SETA BioMedicals), Quasar and Cal Fluor dyes (Biosearch Technologies), SureLight Dyes (APC, RPEPerCP, Phycobilisomes)(Columbia Biosciences), APC, APCXL, RPE, BPE (Phyco-Biotech);
Examples of the widely used fluorophore compounds which are reactive or conjugatable with the linkers of the invention are: Allophycocyanin (APC), Aminocou-marin, APC-Cy7 conjugates, BOD1PY-FL, Cascade Blue, Cy2, Cy3, Cy3.5, Cy3B, Cy5, Cy5.5, Cy7, Fluorescein, FluorX, Hydroxycoumarin, Lissamine Rhodamine B, Lucifer yellow, Methoxycoumarin, NBD, Pacific Blue, Pacific Orange, PE-Cy5 conjugates, PE-Cy7 conjugates, PerCP, R-Phycoerythrin(PE), Red 613, Seta-555-Azide, Seta-555-DBCO, Seta-555-NHS, Seta-580-NHS, Seta-680-NHS, Seta-780-NHS, Seta-APC-780, Seta-PerCP-680, Seta-R-PE-670, SeTau-380-NHS, SeTau-405-Maleimide, SeTau-405-NHS, SeTau-425-NHS, SeTau-647-NHS, Texas Red, TRUC, TruRed, X-Rhodamine.
The fluorophore compounds that can be linked to the linkers of the invention for study of nucleic acids or proteins are selected from the following compounds or their derivatives:
7-AAD (7-aminoactinomycin D, CG-selective), Acridine Orange, Chromomycin A3, CyTRAK Orange (Biostatus, red excitation dark), DAPI, DRAQ5, DRAQ7, Ethidium Bromide, Hoechst33258, Hoechst33342, LDS 751, Mithramycin, PropidiumIodide (PI), SYTOX Blue, SYTOX Green, SYTOX Orange, Thiazole Orange, TO-PRO: Cyanine Monomer, TOTO-1, TO-PRO-1, TOTO-3, TO-PRO-3, YOSeta-1, YOYO-1. The fluorophore compounds that can be linked to the linkers of the invention for study cells are selected from the following compounds or their derivatives: DCFH
(2'7'Dichorodihydro-fluorescein, oxidized form), DHR (Dihydrorhodamine 123, oxidized form, light catalyzes oxidation), Fluo-3 (AM ester. pH > 6), Fluo-4 (AM ester. pH 7.2), Indo-1 (AM
ester, low/high calcium (Ca2+)), SNARF(pH 6/9). The preferred fluorophore compounds that can be linked to the linkers of the invention for study proteins/antibodies are selected from the following compounds or their derivatives: Allophycocyanin(APC), AmCyanl (tetram-er, Clontech), AsRed2 (tetramer, Clontech), Azami Green (monomer, MBL), Azurite, B-phycoerythrin(BPE), Cerulean, CyPet, DsRed monomer (Clontech), DsRed2 ("RFP", Clontech), EBFP, EBFP2, ECFP, EGFP (weak dimer, Clontech), Emerald (weak dimer, Invitrogen), EYFP (weak dimer, Clontech), GFP (565A mutation), GFP (S65C
mutation), GFP (S65L mutation), GFP (S65T mutation), GFP (Y66F mutation), GFP (Y66H muta-tion), GFP (Y66W mutation), GFPuv, HcRed 1, J-Red, Katusha, Kusabira Orange (mono-mer, MBL), mCFP, mCherry, mCitrine, Midoriishi Cyan (dimer, MBL), mKate (TagFP635, monomer, Evrogen), mKeima-Red (monomer, MBL), mKO, mOrange, mPlum, mRaspberry, mRFP1 (monomer, Tsien lab), mStrawberry, mTFP1, mTurquoise2, P3 (phycobilisome complex), Peridinin Chlorophyll (PerCP), R-phycoerythrin(RPE), T-Sapphire, TagCFP (dimer, Evrogen), TagGFP (dimer, Evrogen), TagRFP (dimer, Evrogen), TagYFP (dimer, Evrogen), tdTomato (tandem dimer), Topaz, TurboFP602 (dimer, Evrogen), TurboFP635 (dimer, Evrogen), TurboGFP (dimer, Evrogen), TurboRFP
(dimer, Evrogen), TurboYFP (dimer, Evrogen), Venus, Wild Type GFP, YPet, ZsGreenl (tetramer, Clontech), ZsYellowl (tetramer, Clontech).
In yet another embodiment, the preferred cytotoxic agents that conjugated to a cell-binding molecule via a bridge linker of this patent are tubulysins, maytansinoids, taxanoids (taxanes), CC-1065 analogs, daunorubicin and doxorubicin compounds, benzodiazepine dimers (e.g., dimers of pyrrolobenzodiazepine (PBD), tomaymycin, anthramycin, indolinobenzodiazepines, imidazobenzothiadiazepines, or oxazolidino-benzodiazepines), calicheamicins and the enediyne antibiotics, actinomycin, azaserines, bleomycins, epirubicin, tamoxifen, idarubicin, dolastatins, auristatins (e.g. monomethyl auristatin E, MMAE , MMAF, auristatin PYE, auristatin TP, Auristatins 2-AQ, 6-AQ, EB (AEB), and EFP (AEFP)), duocarmycins, thiotepa, vincristines, hemiasterlins, nazumamides, microginins, radiosumins, alterobactins, microsclerodermins, theonellamides, esperamicins, PNU-159682, and their analogues and derivatives above thereof.

Tubulysins that are preferred for conjugation in the present invention are well known in the art and can be isolated from natural sources according to known methods or prepared synthetically according to known methods (e. g. Balasubramanian, R.; et al. J.
Med.
Chem., 2009, 52, 238-240. Wipf, P.; et al. Org. Lett., 2004, 6, 4057-4060.
Pando, O.; et al.
5 J. Am. Chem. Soc., 2011, 133, 7692-7695. Reddy, J. A.; et al. Mol.
Pharmaceutics, 2009, 6, 1518-1525. Raghavan, B.; et al. J. Med. Chem., 2008, 51, 1530-1533.
Patterson, A. W.;
et al. J. Org. Chem., 2008, 73, 4362-4369. Pando, 0.; et al. Org. Lett., 2009, 11(24), pp 5567-5569. Wipf, P.; et al. Org. Lett., 2007, 9 (8), 1605-1607. Friestad, G.
K.; Org. Lett., 2004, 6, pp 3249-3252. Hillary M. Peltier, H. M.; et al. J. Am. Chem. Soc., 2006, 128, 10 16018-16019. Chandrasekhar, S.; et al. J. Org. Chem., 2009, 74, 9531-9534. Liu, Y.; et al.
Mol. Pharmaceutics, 2012, 9, 168-175. Friestad, G. K.; et al. Org. Lett., 2009, 11, 1095-1098. Kubicek, K.; et al., Angew Chem Int Ed Engl, 2010. 49: p. 4809-12. Chai, Y.; et al., Chem Biol, 2010, 17: 296-309. Ullrich, A.; et al., Angew Chem Int Ed Engl, 2009, 48, 4422-5. Sani, M.; et al. Angew Chem Int Ed Engl, 2007, 46, 3526-9. Domling, A.; et al., 15 Angew Chem Int Ed Engl, 2006. 45, 7235-9. Patent applications: Zanda, M.
; et al, Can.
Pat. Appl. CA 2710693 (2011). Chai, Y.; et al. Eur. Pat. Appl. 2174947 (2010), PCT WO
2010034724. Leamon, C.; et al, PCT WO 2010033733, WO 2009002993. Ellman, J.;
et al, PCT WO 2009134279; PCT WO 2009012958, US appl. 20110263650, 20110021568, Matschiner, G.; et al, PCT WO 2009095447.Vlahov, I.; et al, PCT WO 2009055562, WO
20 2008112873. Low, P.; et al, PCT WO 2009026177. Richter, W., PCT WO
2008138561.
Kjems, J.; et al, PCT WO 2008125116. Davis, M.; et al, PCT WO 2008076333.
Diener, J.;
et al, U.S. Pat. Appl. 20070041901, WO 2006096754. Matschiner, G.; et al, PCT
WO
2006056464. Vaghefi, F.; et al, 5 PCT WO 2006033913. Doemling, A., Ger. Offen.
DE
102004030227; PCT WO 2004005327; WO 2004005326; W02004005269. Stanton, M.; et 25 al, U.S. Pat. Appl. Publ. 20040249130. Hoene, G.; et al, Ger. Offen. DE
10254439 ; DE
10241152; DE 10008089. Leung, D.; et al, WO 2002077036. Reichenbach, H.; et al, Ger.
Offen. DE 19638870; Wolfgang, R.; US 20120129779, Chen, H.,US appl.
20110027274.
The preferred structure of tubulysins for conjugation of cell binding molecules are de-scribed in the patent application of PCT/1B2012/053554.
30 Examples of the structures of the conjugates of the antibody-tubulysin analogs via the bridge linker are T01, T02, T03, T04, T05, and T06 as following:

¨ * Z3 ¨
H 0 OAc 0 v 0 Cy:4 Ni), H 1%1 NS N /
OH s\mAb N

= Z3 s ri g :ii)k OAc N 0 N¨R2--X2 0 H
*I' 4 N 0A
Nlf _ I r, / N OH n v, 1 I S H 0 ¨

_ 0 H 0X OAc OH S\mAb N.EN/41µf)L.rN_ I
I 01 I sir µ1A 0 ilt OµRcxr\cs/
NiortI(H 0 NVOAc k OH
_I 0 µN'SI'L I / N
S H 0 n _ ¨ H
* N,Ri....xi..........s cii(g 0 OAc LL.

/õ,k. INT..4 NI
I 0 µ0 X R2'S
H 0 OAc OH
XiC
, I ki 1 1 S H 0 n ¨

_ -NIC(N, 0c. '...
õ
N ===Y Ri S\mAb I 0 µ , 0 * Z3 ri _II 0 OAc 0 XeS/

N ...2 -j 0 ,,IL i S H 0 _ n X .--======-'"-Ri _______________________ 0 =Z3 ¨

mAb7 OH
/ N
\ I 0 µ0 I S H 0 S
X2 ----- R2 -------- 0tit Z3 N / N

¨ . Z3 ¨
N ='' i mAb I* Z3 NS
OH
¨ ¨ 11 TO6 ¨ = Z3 -14 0 OAc 0 mAb . Z3 \ 0/NIIIVT( i 4 N ;17A OH
Of R2 ¨ ¨ n Wherein mAb is an antibody; Z3 is H, OP(0)(0M1)(0M2), OCH2OP(0)(01\41)(0M2), 0S03M1, or 0-, NH-, S- or CH2- glycoside (glucoside, galactoside, mannoside, glueuronoside, alloside, fructoside, etc); M1 and M2 are independently H. Na, K, Ca, Mg, NR1R2R3; n is 1-20; X1, X2, R1, R2 and R3 are the same defined in Formula (I).
Calicheamicins and their related enediyne antibiotics that are preferred for cell-binding molecule-drug conjugates of this patent are described in: Nicolaou, K.
C. et al, Science 1992, 256, 1172-1178; Proc. Natl. Acad. Sci USA. 1993, 90, 5881-5888), U.S.
Patent Nos. 4,970,198; 5,053,394; 5,108,912; 5,264,586; 5,384,412; 5,606,040;
5,712,374;
5,714,586; 5,739,116; 5,770,701; 5,770,710; 5,773,001; 5,877,296; 6,015,562;
6,124,310;

8,153,768. An Example of the structure of the conjugate of the antibody-Calicheamicin analog via the bridge linker is CO1 as the following:
I
¨
___..----Rr-------S HO
:y...............ii : CH3 0 ik40 ¨
, ii,c 0 H3C H3 I.1 H

C 17-1.-.
Z H..o32_:42. OCH3 2I-% j.....0 H
mAb H3C0 H3YHtrs-j3 0 OH

HC

______________________________________________ S HO 0 3 0 1.14 CH
0 , ari s.)--C....,,..Ø.. H3c \ , 0cH3 N--..4....
0 w sub.
HHO N.
H3...C4.2.71 8 CH3 C2..Li 5 0 H
HO
¨ H3C0 OH H3CNT.9.j n 8 H3c C01.
Wherein mAb is an antibody; n is 1-20; Xi, X2, R1, R2 and R3 are the same defined in Formula (I).
Maytansinoids that are preferred to be used in the present invention including maytansinol and its analogues are described in U.S. Patent Nos. 4,256,746, 4,361,650, 4,307,016, 4,294,757, 4,294,757, 4,371,533, 4,424,219, 4,331,598, 4,450,254, 4,364,866, 4,313,946, 4,315,929 4,362,663, 4,322,348, 4,371,533, 4,424,219, 5,208,020, 5,416,064, 5,208,020; 5,416,064; 6,333.410; 6,441,163; 6,716,821, 7,276,497, 7,301,019, 7,303,749, 7,368,565, 7,411,063, 7,851,432, and 8,163,888 . An example of the structure of the conjugate of the antibody- Maytansinoids via the bridge linker is as the following M01:
mAb s/ \
S

i 1,41¨Xiir.X2-R12)....ro \ 0 0 ----CI \Me e o . o Mel .11 * A
o . .0' . . .0".
. --- . .--NIOe il NI 'µO
H3C0 HO H H3C0 Ho H n , M01.
Wherein mAb is an antibody; n is 1-20; Xi, X2, Ri, R2 and R3 are the same defined in Formula (I).

Taxanes, which includes Paclitaxel (Taxol), a cytotoxic natural product, and docetaxel (Taxotere), a semi-synthetic derivative, and their analogs which are preferred for conjuga-tion via the bridge linkers of the present patent are exampled in:. K C.
Nicolaou et al., J.
Am. Chem. Soc. 117, 2409-2420, (1995); Ojima et al, J. Med. Chem. 39:3889-3896 (1996); 40:267-278 (1997); 45, 5620-5623 (2002); Ojima et al., Proc. Natl.
Acad. Sci., 96:4256-4261 (1999; Kim et al., Bull. Korean Chem. Soc., 20, 1389-1390 (1999);
Miller, et al. J. Med. Chem., 47, 4802-4805(2004); U.S. Patent No. 5,475,011 5,728,849, 5,811,452; 6,340,701; 6,372,738; 6,391,913, 6.436,931; 6,589,979; 6,596,757;
6,706,708;
7,008,942; 7,186,851; 7,217,819; 7,276,499; 7,598,290; and 7,667,054.
Examples of the structures of the conjugate of the antibody- taxanes via the bridge linker are as the following Tx01, Tx02 and Tx03.
mAb I S/ \ S

R,..--Xf-Tr) 47/--X2-.....,R I2 0 Q.
...1 "..?%'0ils NH
77. 0 AV*NH 0 r eV*
e Nei" . 5. o --. .., - A %-.. . µ . E.
.., 0 \ -OHO OH 8 H OAc \ 0 H 6 OH el I"
r=A
OAc I Me0 #
OMe Me0 lir OMe I
n Tx01 mAb I \
S/
S
/ I
R1¨X1 AA)---477--X2¨R2 0 Q.

er 0 ii4h6 lik ..... Nilo milk.
WVAW 0 CYLp 0 IF
I i dr I H S 0 till OH 0 0Ae OH OH li 1T Ac Me0 10 Me0 lip I OMe OMe I n Tx02 mAb S/ \S
0 0/ o CYk S,.. c cw,ANH 0As ;111 o .40*
t.

' OH HO 0 HA

Me0 Me0 OMe OMe I
n Tx03 Wherein mAb is an antibody; n is 1-20; Xi, X2, Ri and R2 are the same defined in Formu-la (I).
CC-1065 analogues and doucarmycin analogs are also preferred to be used for a con-5 jugate with the bridge linkers of the present patent. The examples of the CC-1065 ana-logues and doucarmycin analogs as well as their synthesis are described in:
e.g.Warpehoski, et al, J. Med. Chem. 31:590-603 (1988), D. Boger et al., J.
Org. Chem;
66; 6654-6661, 2001; U. S. Patent Nos: 4169888, 4391904, 4671958, 4816567, 4912227, 4923990, 4952394, 4975278, 4978757, 4994578, 5037993, 5070092, 5084468, 5101038, 10 5117006, 5137877, 5138059, 5147786, 5187186, 5223409, 5225539, 5288514, 5324483, 5332740, 5332837, 5334528, 5403484, 5427908, 5475092, 5495009, 5530101, 5545806, 5547667, 5569825, 5571698, 5573922, 5580717, 5585089, 5585499, 5587161, 5595499, 5606017, 5622929, 5625126, 5629430, 5633425, 5641780, 5660829, 5661016, 5686237, 5693762, 5703080, 5712374, 5714586, 5739116, 5739350, 5770429, 5773001, 5773435, 15 5786377 5786486, 5789650, 5814318, 5846545, 5874299, 5877296, 5877397, 5885793, 5939598, 5962216, 5969108, 5985908, 6060608, 6066742, 6075181, 6103236, 6114598, 6130237, 6132722, 6143901, 6150584, 6162963, 6172197, 6180370, 6194612, 6214345, 6262271, 6281354, 6310209, 6329497, 6342480, 6486326, 6512101, 6521404, 6534660, 6544731, 6548530, 6555313, 6555693, 6566336, 6,586,618, 6593081, 6630579, 20 6,756,397, 6759509, 6762179, 6884869, 6897034, 6946455, 7,049,316, 7087600, 7091186, 7115573, 7129261, 7214663, 7223837, 7304032, 7329507, 7,329,760, 7,388,026, 7,655,660, 7,655,661, 7,906,545, and 8,012,978. Examples of the structures of the conjugate of the antibody- CC-1065 analogs via the bridge linker are as the following CC01, CCO2, and CC03.

_ _ Cl" X3.-R_.
N /

0 1INI /1-1 1161 1 Xi VD
...-=S
110 o ITI mAb /
OZ4 'S
Cl/AX3 f__ 40 g / 40 µR2-"Xl -2 U
N /

0 110 o H
n _ OZ4 ¨

Cl , s 0,, Cl/A N / 0 o 1 N /
10 # o 1T/ 0._ 400 0 III 0-0, 0 x, 0 0 v_.,µ IL ,X3 R2---..,2 / \ --'sx1----R1 S n \ VS
mAb CCO2 ¨ 401 Cl... H
N . X3¨Ri...xi _ * N i * 0 Z40 H.- S,µ
N mAb . X\3 i Z40 * N IIP 0 R2¨ X2 ,.. N n _ tp H ¨

Wherein mAb is an antibody; n is 1-20; Z4 is H, P0(01\41)(0M2), S03M1, 5 CH2P0(01\41)(0M2), CH3N(CH2CH2)2NC(0)-, 0(CH2CH2)2NC(0)-, or glycoside;
X3 is 0, NH, NHC(0), OC(0), CO, or absent; Xi, X2, RI, R2, MI, and M2 are the same defined in Formula (I).
Daunorubicin/Doxorubicin Analogues are also preferred for conjugation via the bridge linkers of the present patent. The preferred structures and their synthesis are exam-10 pled in: Hurwitz, E., et al., Cancer Res. 35, 1175-1181(1975). Yang, H.
M., and Reisfeld, R. A., Proc. Natl. Acad. Sci. 85, 1189-1193 (1988); Pietersz, C. A., E., et al., E., et al.,"
Cancer Res. 48, 926-9311(1988); Trouet, et al., 79, 626-629 (1982); Z. Brich et al., J.

Controlled Release, 19, 245-258 (1992); Chen et al., Syn. Comm., 33, 2377-2390, 2003;
King et al., Bioconj. Chem., 10, 279-288, 1999; King et al., J. Med. Chem., 45, 4336-4343, 2002; Kratz et al., J Med Chem. 45, 5523-33. 2002; Kratz et al., Biol Pharm Bull.
Jan. 21, 56-61 , 1998; Lau et al., Bioorg. Med. Chem. 3, 1305-1312, 1995;
Scott et al., Bioorg. Med.1 Chem. Lett. 6, 1491-1496; 1996; Watanabe et al., Tokai J.
Experimental Clin. Med. 15, 327-334, 1990; Zhou et al., J. Am. Chem. Soc. 126, 15656-7, 2004; WO
01/38318; U.S. Patent No.). 5,106,951; 5,122,368; 5,146,064; 5,177,016;
5,208,323;
5,824,805; 6,146,658; 6,214,345; 7569358; 7,803,903; 8,084,586; 8,053,205.
Examples of the structures of the conjugate of the antibody- CC-1065 analogs via the bridge linker are as the following Da01, Da02, Da03 and Da04.
0 = H 0 0 = H 0 I H = H
011101.11111 OH .11111.11110 OH
Ai a H3co 0 OH 0--c H3C0 I OH 0 :C
OH --ClOH
HN,..
-112¨Xrir¨c¨LX1¨RVNH
S S n N , mAb Da01 mAb I
X3 \ 0 N I 0 0 H \ I H
/N
I
0 = 40 . 446 HOH *1111110. µ90H. II

0 OH 6 ey0H H3C0 I H t = _ , o I k_y-n Da02 ,nruhe Sr ity'X2 ______________________________________ o SH
= OH
/N
I = HH
4040 ...OH
0111111110t. S0011 OH OvnN OH
n Da03 Me0v mAb S/ XS
xr-Ri_ OH

Has jI
/er HOJ OH
$1.01.1 n OHO OMe 0 OHO OMe volk .4 I0µ,"ThNoi0 Me Me0 n Da04 Wherein mAb is an antibody; n is 1-20; X3 is H, 0, NH, NHC(0), NHC(0)NH, C(0), or OC(0); X1, X2, R1, and R2 are the same defined in Formula (I).
Auristatins and dolastatins are preferred in conjugation via the bridge linkers of this patent. The auristatins (e. g. auristain E (AE) auristatin EB (AEB), auristatin EFP (AEFP), monomethyl auristatin E (MMAE), Monomethylauristatin (MMAF), Auristatin F
phenylene diamine (AFP) and a phenylalanine variant of MMAE) which are synthetic analogs of dolastatins, are described in Int. J. Oncol. 15:367-72 (1999);
Molecular Cancer Therapeutics, vol. 3, No. 8, pp. 921-932 (2004); U.S. Application Nos.
11134826, 20060074008, 2006022925. U.S. Patent Nos. 4414205, 4753894, 4764368, 4816444, 4879278, 4943628, 4978744, 5122368, 5165923, 5169774,5286637, 5410024, 5521284, 5530097, 5554725, 5585089, 5599902, 5629197, 5635483, 5654399, 5663149, 5665860, 5708146, 5714586, 5741892, 5767236, 5767237, 5780588, 5821337, 5840699, 5965537, 6004934, 6033876, 6034065, 6048720, 6054297, 6054561, 6124431, 6143721, 6162930, 6214345, 6239104, 6323315, 6342219, 6342221, 6407213, 6569834, 6620911, 6639055, 6884869, 6913748, 7090843, 7091186, 7097840, 7098305, 7098308, 7498298, 7375078, 7462352, 7553816, 7659241, 7662387, 7745394, 7754681, 7829531, 7837980, 7837995, 7902338, 7964566, 7964567, 7851437, 7994135. Examples of the structures of the conju-gate of the antibody-auristatins via the bridge linker are as the following Au01, Au02, Au03, Au04, and Au05.
_ H ill H OH -/s\ rXr-RIµN)ryN,Nry....I.Prii,N
I 0 A I *
....-0 0 ....-0 0 mAb Xs,/)).. ,R2 H 12/ ryThAlIcH OH

(101 n - I 0 ,./\ I _....0 0 _--0 0 -Au01 s = N

mAb X2 R N a NifrIrN
#

---0 0 = H -Au02 - H _ %.)cr. rNIRIIrkl * N'Ilf-X3 0 N : N
I 0 j' OH Z3 mAb H
,/
\ )crik0( N * _ 2 0 N z N..)nyThrirli :3R2-X 0 0 OH _ n AuO3 c - H H -ryN.,....0rrlcVyN k Thr *
I0 A 7 ......0 0 .....0 0 X4 "iv-iv NT S
* /
ri ,......
mAb N
: N S
......0 0 .....co 0 x4.,...Rer.--X2 0 _ n Au04 -NrNt''.1CNIN 110 1 0 ....0 0 014 X3-'111--X i S
mAb \ Xrki . 9 _iciiiikrII
N * /
S
I H Xr112...X2 0 n _ Au05 Wherein mAb is an antibody; n is 1-20; X3 is CH2, 0, NH, NHC(0), NHC(0)NH, C(0), OC(0) or absent; X4 is CH2, C(0), C(0)NH, C(0)N(R1), or C(0)0; X1, X2, R1, R2 5 and R3 are the same defined in Formula (I).
The benzodiazepine dimers (e. g. dimmers of pyrrolobenzodiazepine (PBD) or (tomaymycin), indolinobenzodiazepines, imidazobenzothiadiazepines, or oxazolidinobenzodiazepines) which are preferred cytotoxic agents according to the present invention are exampled in the art: US Patent Nos . 8,163,736; 8,153,627;
8,034,808;
10 7,834,005; 7,741,319; 7,704,924; 7,691,848; 7,678,787; 7,612,062;
7,608,615; 7,557,099;
7,528,128; 7,528,126; 7,511,032; 7,429,658; 7,407,951; 7,326,700; 7,312,210;
7,265,105;
7,202,239; 7,189,710; 7,173,026; 7,109,193; 7,067,511; 7,064,120; 7,056,913;
7,049,311;
7,022,699; 7,015,215; 6,979,684; 6,951,853; 6,884,799; 6,800,622; 6,747,144;
6,660,856;
6,608,192; 6,562,806; 6,977,254; 6,951,853; 6,909,006; 6,344,451; 5,880,122;
4,935,362;
15 4,764,616; 4,761,412; 4,723,007; 4,723,003; 4,683,230; 4,663,453;
4,508,647; 4,464,467;
4,427,587; 4,000,304; US patent appl. 20100203007, 20100316656, 20030195196.
Exam-ples of the structures of the conjugate of the antibody- benzodiazepine dimers via the bridge linker are as the following PB01, PB02, PB03, PB04, PB05, PB06, PB07 and PB08.
_ H03 H ,k1 03H R I ....x1 _ WI
yol IMe MeI f&I i 0s N¨X3 ¨
N N
R3 i =
14, H03 H 114 03H 1 .mAb _ I I iii,h /
N 14 IIV __ ¨N XI
20 ¨
R3 OMe Me0 rr R2---X 0 n N¨
_ -----Ri---------Xi 0 ¨
F.:Cr 0111 l'ri 1,\,00 74.-- \¨js ...
R3 OMe 0 Me R3 /mAb ---N--------R2--x2 S
r=CrN * e 0\/ * 441µ1 \.o R3 OMe Me0 R3 _ 0 0 _ n ¨ H03 g ki a 1,^A,00 03H , N_x3 i S/
S
N N
R3 e Me Me0 H03 g ,kii 03H

I
R( %N i Me Me R2¨x2 0 n ¨ 0 0 _PB03 ¨ H03 Ikif ,,c4-H,¨x1 0 _ ah I I
R3 N "P OMe Mek,r, R3 S>Ab X4¨R2¨ X2 S

N
¨R( 1141 1 Me Me0* R3 ¨ n _ X3 _______________________________________________ _ H03 ikl ,v 03H
iiiii = osi , eg,i, ,.
s., # N
"11 OMe Me0 IV N At -inAb H03 ki H 03 H0 N
N4 0 14111 = *
OMe Mel N *0 _ n X4-R1¨ XI 0 _ - H03 ig N' rai I =

* N 0 WI 0 Me Me0 * N AO, 1103 114 , ¨ * N 4N 410 - n OMe Me0 .

X3 ______ Ri------__ v _ H03 g g 03H s _ N.

mAb N 41:1 * N R3 /
R3 I Me Me0 3 ,x2 -- X2 S
H03 ki H 03H 0 R3 - N . I 101111 I 3 Me Me: n - _ X3 ____________________________________ Ri,..,x1 I 0 ¨
H...g C H......e.:Hj. *
OMe101111 Me0 ,mAb 1442,1,3.4-11 H.....ef 0 _ . Me Me I _ n Wherein mAb is an antibody; n is 1-20; X3 is CH2, 0, NH, NHC(0), NHC(0)NH, C(0), OC(0) or absent; X4 is CH2, C(0), C(0)NH, C(0)N(R1), or C(0)0; X1, X2, R1, R2 and R3 are the same defined in Claim 1. In addition, R1 and/or R2 can be absent.
The drugs/ cytotoxic agents used for conjugation via a bridge linker of the present patent can be any analogues and/or derivatives of drugs/molecules described in the present patent. One skilled in the art of drugs/cytotoxic agents will readily understand that each of the drugs/cytotoxic agents described herein can be modified in such a manner that the resulting compound still retains the specificity and/or activity of the starting compound. The skilled artisan will also understand that many of these compounds can be used in place of the drugs/cytotoxic agents described herein. Thus, the drugs/cytotoxic agents of the present invention include analogues and derivatives of the compounds described herein.
All references cited herein and in the examples that follow are expressly incorporated by reference in their entireties.
EXAMPLES
The invention is further described in the following examples, which are not intended to limit the scope of the invention. Cell lines described in the following examples were maintained in culture according to the conditions specified by the American Type Culture Collection (ATCC) or Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, Braunschweig, Germany (DMSZ), or The Shanghai Cell Culture Institute of Chinese Acadmy of Science, unless otherwise specified. Cell culture reagents were ob-tained from Invitrogen Corp., unless otherwise specified. All anhydrous solvents were commercially obtained and stored in Sure-seal bottles under nitrogen. All other reagents and solvents were purchased as the highest grade available and used without further purification. The preparative HPLC separations were performed with Varain PreStar HPLC. NMR spectra were recorded on Varian Mercury 400 MHz Instrument. Chemical shifts (.delta.) are reported in parts per million (ppm) referenced to tetramethylsilane at 0.00 and coupling constants (J) are reported in Hz. The mass spectral data were acquired on a Waters Xevo Qtof mass spect equipped with Waters Acquity UPLC separations module and Acquity TUV detector.
Example 1: tert-Butyl 3-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)propanoate (34) HO011) =C .k 3 -30- HO0-)....,/=y Nf....

Na/THF 0 To 350 mL of anhydrous THF was added 80 mg (0.0025 mol) of sodium metal and triethylene glycol 2 (150.1 g, 1.00 mol) with stirring. After the sodium had completely dissolved, tert-butyl acrylate (24 mL, 0.33 mol) was added. The solution was stirred for 20 h at room temperature and neutralized with 8 mL of 1.0 M HC1. The solvent was removed in vacuo and the residue was suspended in brine (250 mL) and extracted with ethyl acetate (3 x 125 mL). The combined organic layers were washed with brine (100 mL) then water (100 mL), dried over sodium sulfate, and the solvent was removed. The resulting colorless oil was dried under vacuum to give 69.78 g (76% yields) of product 34. 1H NMR:
1.41 (s, 9H), 2.49 (t, 2H, J=6.4 Hz), 3.59-3.72 (m, 14H). ESI MS m/z- C13H2506 (M-H), cacld.
277.17, found 277.20.
Example 2. tert-Butyl 3-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)propanoate (35) TsO'TsC1 0 f"-Pyr 0 A solution of 34 (10.0 g, 35.95 mmol) in acetonitrile (50.0 mL) was treated with pyri-dine (20.0 mL). A solution of tosyl chloride (7.12 g, 37.3 mmol) in 50 mL
acetonitrile was added dropwise via an addition funnel over 30 minutes. After 5 h TLC analysis revealed that the reaction was complete. The pyridine hydrochloride that had formed was filtered off and the solvent was removed. The residue was purified on silica gel by eluting from with 20% ethyl acetate in hexane to with neat ethyl acetate to give 11.2 g (76% yield) of compound 35.
NMR: 1.40 (s, 9H), 2.40 (s, 3H), 2.45 (t, 2H, J=6.4 Hz), 3.52-3.68 (m, 14H), 4.11 (t, 2H, J=4.8 Hz), 7.30(d, 2H, J=8.0 Hz), 7.75 (d, 2H, J=8.0 Hz);
ESI MS m/z+
C20113308S (M+H), cacld. 433.18, found 433.30.
Example 3. tert-Butyl 3-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)propanoate (36) NaN3 Ts0 -31" N34='*. 1-3-"*Y µ"\e--To 50 mL of DMF was added tert-butyl 3-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)-propanoate 35 (4.0 g, 9.25 mmol) and sodium azide (0.737 g, 11.3 mmol) with stirring.
The reaction was heated to 80 C. After 4 h TLC analysis revealed that the reaction was complete. The reaction was cooled to room temperature and quenched with water (25 mL). The aqueous layer was separated and extracted into ethyl acetate (3 x 35 mL). The combined organic layers were dried over anhydrous magnesium sulfate, filtered, and the solvent removed in vacuo. The crude azide (about 90% pure by TLC) was used without further purification. 1H NMR (CDC13): 1.40 (s, 9H), 2.45 (t, 2H, J=6.4 Hz), 3.33 (t, 2H, J=5.2 Hz), 3.53-3.66 (m, 12H). ESI MS m/z+ C13H26N308 (M+H), cacld. 304.18, found 304.20.
Example 4. 13-Amino-4,7,10-trioxadodecanoic acid tert-butyl ester, 37; 13-Amino-bis(4,7,10-trioxadodecanoic acid tert-Butyl Ester), 38.

37 \(%/0 38 The crude azide material 36 (5.0 g, -14.84 mmol) was dissolved in ethanol (80 mL) and 300 mg of 10% Pd/C was added. The system was evacuated under vacuum and placed under 2 atm of hydrogen gas via hydrogenation reactor with vigorous stirring. The 5 reaction was then stirred overnight at room temperature and TLC showed that the starting materials disappeared. The crude reaction was passed through a short pad of celite rinsing with ethanol. The solvent was removed and the amine purified on silica gel using a mixture of methanol (from 5% to 15%) and 1% triethylamine in methylene chloride as the eluant to give 13-amino-4,7,10-trioxadodecanoic acid tert-butyl ester 37 (1.83 g, 44%
10 yield, ESI MS m/z+ C13H27N05 (M+H), cacld. 278.19, found 278.30) and 13-amino-bis(4,7,10-trioxadodecanoic acid tert-butyl ester), 38 (2.58 g, 32% yield, ESI
MS m/z+
C26H52N010 (M+H), cacld. 538.35, found 538.40).
Example 5. 3-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)propanoic acid, HC1 salt, 39.

15 To 13-amino-4,7,10-trioxadodecanoic acid tert-butyl ester, 37 (0.80 g, 2.89 mmol) in 30 mL of dioxane was 10 ml of HC1 (36%) with stirring. After 0.5 h TLC
analysis re-vealed that the reaction was complete, the reaction mixture was evaporated, and co-evaporated with Et0H and Et0H/Toluene to form the title product in HC1 salt (>90%
pure, 0.640 g, 86% yield) without further purification. ESI MS m/z+ C9H20N05 (M+H), 20 cacld. 222.12, found 222.20.
Example 6. 13-Amino-bis(4,7,10-trioxadodecanoic acid, HC1 salt, 40.

\(%/0-OH 40 To 13-amino-bis(4,7,10-trioxadodecanoic acid tert-butyl ester), 38 (1.00 g, 1.85 mmol) in 30 mL of dioxane was 10 ml of HC1 (36%) with stirring. After 0.5 h TLC
25 analysis revealed that the reaction was complete, the reaction mixture was evaporated, and co-evaporated with Et0H and Et0H/Toluene to form the title product in HC1 salt (>90% pure, 0.71 g, 91% yield) without further purification. ESI MS m/z+

(M+H), cacld. 426.22, found 426.20.
Example 7. bis(2,5-dioxopyrrolidin-1-y1) but-2-ynedioate, 9.

I -HO 118 OH -)0' N,o_ JI - I
DMA 0' To but-2-ynedioic acid 8 (2.0 g, 17.54 mmol) in DMA (100 ml) was added NHS
(5.0 g, 43.4 mmol) and EDC (12.0 g, 62.5 mmol). The mixture was stirred under dark over-night, evaporated and purified on Si02 column eluted with Et0Ac/DCM (1:10) to afford the title compound 9 (4.10 g, 76% yield). ESI MS m/z+ C12H9N208 (M+H), cacld.
309.03, found 309.20.
Example 8. 4,7-dioxodec-5-ynedioic acid, 15.
I0 12 (5 mol%) JO ________ 0 =
C
\ 0 C-RT, DCM
0 c:10H HO 0 To a stirred solution of bis(trimethylsilyeacetylene (5.0 g, 29.34 mmol) and iodine (0.37 g, 1.45 mmol) in dichloromethane (100 mL), was added succinyl chloride (18.11 g, 116.83 mmol) slowly in a dropwise manner at 0 C. After addition, the mixture was allowed to stir at room temperature until complete conversion as indicated by TLC (-2 h).
The reaction mixture was quenched with water (15 mL) and extracted with dichloro-methane (3 x 70 mL). The combined extracts were washed with 15% solution of sodium thiosulphate, dried over anhydrous Na2SO4, and concentrated in vacuo. The resulting product was purified by column chromatography on silica gel (100-200 mesh, aqueous form, from 5% to 10% of H20 in acetonitrile) to afford the pure title product (5.50 g, yield 85%). ESI MS m/z- C10H906 (M-H), cacld. 226.05, found 226.10.
Example 9. (R,R,S,S,R,4R,4'R)-5,5'-(44,7-dioxodec-5-ynedioyObis(azanediy1))bis(4-hydroxy-3,1-phenylene))bis(4-(24(1R,3R)-1-acetoxy-3-((2S,3S)-N,3-dimethyl-2-((R)-1-methylpiperidine-2-carboxamido)pentanamido)-4-methylpentyl)thiazole-4-carboxamido)-2-methylpentanoic acid), 79 _,1,1 0 OAc 0 4.4k1N1 OHH

I = S IN1 alki OH
jki 0 OAc 0 g= P N
\ +elf 44k1.1 0 0 I = S H 0 The compound 9 (25 mg, 0.081 mmol) in THF (3.0 ml) was added (4R)-4-(2-((1R,3R)-1-acetoxy-3-((2S,3S)-N,3-dimethy1-2-((R)-1-methylpiperidine-2-carboxamido)pentanamido)-4-methylpentyl)thiazole-4-carboxamido)-5-(3-amino-4-hydroxypheny1)-2-methylpentanoic acid, 51 (Huang Y. et al, Med Chem. #44, 249th ACS
National Meeting, Denver, CO, Mar. 22-26, 2015; W02014009774) (151 mg, 0.199 mmol) in THF (4.0 ml) and buffer (4 ml, 100 mM Na2HPO4, pH 7.0). After stirred at RT
for 4 h, the mixture was concentrated and purified with C-18 preparative HPLC
(250 mm x ID 20 mm), eluted with water/ethanol (90% water to 50% water in 55 min, v =

ml/min ). The fractions containing the product were pooled, concentrated and crystallized with Et0H/Hexane to afford the title compound (73 mg, 53% yield). ESI MS m/z+
C86H122N12Na020S2 (M+Na), cacld. 1729.83, found 1730.10.
Example 10. 14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioic acid, 86.

HOy\ jr,o, As_ LN4."

3-(2-(2-(2-Aminoethoxy)ethoxy)ethoxy)propanoic acid, HC1 salt, 39 (601 mg, 2.33 mmol) in the mixture of THF (6 ml) and a buffer (150 mM NaH2PO4, pH 7.2, 4 ml) was added bis(2,5-dioxopyrrolidin-1-yl)but-2-ynedioate, 9 (350 mg, 1.13 mmol).
After stirred at RT under dark for 4 h, the mixture was concentrated and purified with Si02 column, eluted with water/acetonitrile (1:9). The fractions containing the product were pooled and concentrated to afford the title compound (345 mg, 59% yield). ESI MS m/z-(M-H), cacld. 519.22, found 519.30.
Example 11. 13,18-bis(2-(2-(2-(2-carboxyethoxy)ethoxy)ethoxy)ethyl)-14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioic acid, 87.

13-Amino-bis(4,7,10-trioxadodecanoic acid, HC1 salt, 40 (650 mg, 1.40 mmol) in the mixture of THF (6 ml) and a buffer (150 mM NaH2PO4, pH 7.2, 4 ml) was added bis(2,5-dioxopyrrolidin-1-yl)but-2-ynedioate, 9 (190 mg, 0.61 mmol). After stirred at RT under dark for 4 h, the mixture was concentrated and purified with C-18 preparative HPLC (250 mm x ID 30 mm), eluted with water/ethanol (90% water to 50% water in 55 min, v = 35 ml/mm). The fractions containing the product were pooled and concentrated to afford the title compound (287 mg, 51% yield). ESI MS m/z- C40H67N2022 (M-H), cacld.
927.42, found 928.30.
Example 12. Bis(2,5-dioxopyrrolidin-l-y1) 14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioate, 88.

To 14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioic acid, 86 (340 mg, 0.653 mmol) in DMA (6 ml) was added NHS (225 mg, 1.96 mmol) and EDC
(401 mg, 2.08 mmol). The mixture was stirred under dark overnight, evaporated and purified on Si02 column eluted with Et0Ac/DCM (5:1) to afford the title compound 88 (330 mg, 71% yield). ESI MS m/z+ C30I-143N4016 (M+H), cacld. 715.26, found 715.20.
Example 13. Bis(2,5-dioxopyrrolidin-l-y1) 13,18-bis(2-(2-(2-(3-((2,5-dioxopyrrolidin-1-yl)oxy)-3-oxopropoxy)ethoxy)ethoxy)ethyl)-14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioate, 89 rice c: 1***(N--011...%.õ,0\/....00 0 0.
..,... _.....-_ 0¨<-===/' o''\.=0'-'==----N--1, A-...... 0 N.11. J.1 N=0--rr\,..-00/N...0 0 To 13,18-bis(2-(2-(2-(2-carboxyethoxy)ethoxy)ethoxy)ethyl)-14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioic acid, 87 (280 mg, 0.301 mmol) in DMA (6 ml) was added NHS (105.0 mg, 0.913 mmol) and EDC (200 mg, 1.04 mmol). The mixture was stirred under dark overnight, evaporated and purified on Si02 column eluted with Et0H/DCM (1:10-1:5) to afford the title compound 89 (249 mg, 63%
yield). ESI MS m/z+ C56H81N6030 (M+H), cacld. 1317.49, found 1317.80.
Example 14. (R,R,S ,S,R,4R,4'R)-5,5'-(((14,17-dioxo-4,7,10,21,24,27-hexaoxa-13,18-diazatriacont-15-yne-1,30-dioyebis(azanediy1))bis(4-hydroxy-3,1-phenylene))bis(4-(2-((1R,3R)-1-acetoxy-3-((2S,3S)-N,3-dimethy1-2-((R)-1-methylpiperidine-2-carboxami-do)pentanamido)-4-methylpentyl)thiazole-4-carboxamido)-2-methylpentanoic acid), 90 rai OH

H
("1 .1kil 0 OAc 0IsW1 N.,..C.(.., \ *el( 4f4N ,Nyk OH 0 I = 3 H I
le OH

\ -self N 1YA/ N OH 0 0 N 0 oSik I S H
1 = 0 90 The compound 88 (30 mg, 0.042 mmol) in THF (3.0 ml) was added (4R)-4-(2-((1R,3R)-1-acetoxy-3-((2S,3S)-N,3-dimethy1-2-((R)-1-methylpiperidine-2-carboxamido)pentanamido)-4-methylpentyl)thiazole-4-carboxamido)-5-(3-amino-4-hydroxypheny1)-2-methylpentanoic acid, 51 (Huang Y. et al, Med Chem. #44, 249th ACS
National Meeting, Denver, CO, Mar. 22-26, 2015; W02014009774) (80 mg, 0.107 mmol) in THF (4.0 ml) and buffer (4 ml, 100 mM Na2HPO4, pH 7.0). After stiffed at RT
for 4 h, the mixture was concentrated and purified with C-18 preparative HPLC (250 mm x mm), eluted with water/ethanol (95% water to 50% water in 55 min, v = 15 ml/min ). The fractions containing the product were pooled, concentrated and crystallized with Et0H/Hexane to afford the title compound (48 mg, 56% yield). ESI MS m/z-C98H1471\11402652 (M-H), cacld. 2000.01, found 2000.40.
Example 15. Conjugated compound 90 to an antibody for 91.
dim OH

H_ -r") .1kil,4t OAcNjyt WI N,C(, \ 4e1( N
N 0 .= I / N
S H 0 0,,c)-3.111 I =
dah OH mAb 14 0 OAc 0 WI N---iN'43-H N

0.1IP < 41kN .'N.IAN

N 0 _ n - I =S1 S H 0 91 5 To a mixture of 2.0 mL of 10 mg/ml Herceptin in pH 6.0-8.0, were added of 0.70 -2.0 mL PBS buffer of 100 mM NaH2PO4, pH 6.5-7.5 buffers, TCEP (28 [tI4, 20 mM
in water) and the compound 90 (14 idt, 20 mM in DMA). The mixture was incubated at RT
for 2-16 h, then DHAA (135 L, 50 mM) was added in. After continuous incubation at RT overnight, the mixture was purified on G-25 column eluted with 100 mM
NaH2PO4, 50 10 mM NaC1 pH 6.0-7.5 buffer to afford 16.8-17.9 mg of the conjugate compound 91 (-87%
yield) in 13.1-14.9 ml buffer. The drug/antibody ratio (DAR) was 4.0, which was deter-mined via UPLC-Qtof mass spectrum. It was 96-99% monomer analyzed by SEC HPLC
(Tosoh Bioscience, Tskgel G3000SW, 7.8 mm ID x 30 cm, 0.5 ml/min, 100 min) and a single band measured by SDS-PAGE gel.
15 Example 16. Compound 92 (containing 4 tubulysin analogs per bridge linker).
a OH
0 OAc N 0 4.1.' rP 43----\
JA/ N
N 0 .. I OH
S H
I = 0 1,01 OH

H
(NI iNt OAcNjA l'W N.t0i.../ 0 OH
/ N
NO
I =
Avi OH II
--.) OAc 0 WI N -(\,r/'k \ 441( 411bN DA/ N H

.<

I = 04 OH
n H0 OAc N 0 JAI N H

N 0 ., I S H
I = 0 92 The compound 89 (35 mg, 0.026 mmol) in THF (3.0 ml) was added (4R)-4-(2-((1R,3R)-1-acetoxy-3-((2S,3S)-N,3-dimethy1-2-((R)-1-methylpiperidine-2-carboxamido)pentanamido)-4-methylpentyl)thiazole-4-carboxamido)-5-(3-amino-4-hydroxyphenyl)-2-methylpentanoic acid, 51 (Huang Y. et al, Med Chem. #44, 249th ACS
National Meeting, Denver, CO, Mar. 22-26, 2015; W02014009774) (100.6 mg, 0.132 mmol) in THF (4.0 ml) and buffer (4 ml, 100 mM Na2HPO4, pH 7.0). After stirred at RT
for 4 h, the mixture was concentrated and purified with C-18 preparative HPLC
(250 mm x ID 20 mm), eluted with water/ethanol (95% water to 50% water in 50 min, v =

ml/min ). The fractions containing the product were pooled, concentrated and crystallized with Et0H/Hexane to afford the title compound 92 (47.6 mg, 47% yield). ESI MS
miz-C192H291N26050S4 (M-H), cacld. 3890.00, found 3890.30.
Example 17. Conjugated compound 92 to an antibody for 93 a OH
N
n OAcyt N / N

OHH

I =
n t OAcN} Nt0+õ 0 OH
16( N Cc( s / N
I = \mAb OH
H 0 OAc 0 C?...141,k;X)r..seyt "1-st S H
= 0 OH
\O

/ N 0.9"\0 OH

S H
I = 0 93 To a mixture of 2.0 mL of 10 mg/ml Herceptin in pH 6.0-8.0, were added of 0.70 ¨
2.0 mL PBS buffer of 100 mM NaH2PO4, pH 6.5-7.5 buffers, TCEP (28 L, 20 mM in water) and the compound 92 (14 ut, 20 mM in DMA). The mixture was incubated at RT
for 2-16 h, then DHAA (135 L, 50 mM) was added in. After continuous incubation at RT overnight, the mixture was purified on G-25 column eluted with 100 mM
NaH2PO4, 50 mM NaC1 pH 6.0-7.5 buffer to afford 16.9-17.5 mg of the conjugate compound (-85% yield) in 13.1-14.9 ml buffer. The drug/antibody ratio (DAR) was 8.0, which was determined via UPLC-Qtof mass spectrum. It was 96-99% monomer analyzed by SEC
HPLC (Tosoh Bioscience, Tskgel G3000SW, 7.8 mm ID x 30 cm, 0.5 ml/min, 100 min) and a single band measured by SDS-PAGE gel.

Example 18. In vitro cytotoxicity evaluation of conjugates 91 and 93 in comparison withT-DM1:
The cell lines used in the cytotoxicity assays were HL-60, a human promyelocytic leukemia cell line; NCI-N87, a human gastric carcinoma cell line; BT-474, a human invasive ductal carcinoma cell line; and SKOV3, a human ovarian carcinoma cell line. For HL-60, NCI-N87, and BT-474 cells, the cells were grown in RPMI-1640 with 10%
FBS.
For SKOV3 cells, the cells were grown in McCoy's 5A Medium with 10% FBS. To run the assay, the cells (180 1, 6000 cells) were added to each well in a 96-well plate and incubated for 24 hours at 37 C with 5% CO2. Next, the cells were treated with test corn-pounds (20 ul) at various concentrations in appropriate cell culture medium (total volume, 0.2 mL). The control wells contain cells and the medium but lack the test compounds. The plates were incubated for 120 hours at 37 C with 5% CO2. MTT (5mg/m1) was then added to the wells (20 ul) and the plates were incubated for 1.5hr at 37 C. The medium was carefully removed and DMSO (180 ul) was added afterward. After it was shaken for 15min, the absorbance was measured at 490nm and 570nm with a reference filter of 620nm. The inhibition% was calculated according to the following equation:
inhibition%
= [1-(assay-blank)/(control-blank)] x 100.
The cytotoxicity results:
IC50 (nM) N87 cell (Ag+) SK-OV-3 cell (Ag+) HL60 cell (Ag-) Conjugate 91 0.108 nM 0.089 nM >20 nM
Conjugate 93 0.037 nM 0.029nM >10 nM
T-DM1 0.270nM 0.191M >15 nM
Specificity of conjugate 91 for N87 cell was over 185 (IC50> 20/ IC50= 0.108), and for SK-OV-3 cell was over 225; Specificity of conjugate 93 for N87 cell was over 270 (IC50>
10/ IC50= 0.037), and for SK-OV-3 cell was over 344; Specificity of conjugate T-DM1 for N87 cell was over 55 (IC50> 15/ IC50= 0.27), and for SK-OV-3 cell was over 78.
Both conjugate 91 and conjugate 93 were extremely more potent than the commercial conjugate T-DM1. The conjugate 93 having DAR=8 was three-fold more potent than conjugate 91 having DAR=4.

Claims (29)

What is claimed is:
1. A bridge linker compound of the Formula (I) Wherein:
The acetylenedicarboxyl group on the linker is capable of reacting with a pair of sulfur atoms of the cell-binding agent;
Z1 and Z2 are the same or different a function group that enables to react with a cytotoxic drug. The functional group Z1 or Z2 can link to a cytotoxic drug via a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, amine (secondary, tertiary, or quartary), imine, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
R1 and R2 are the same or different, and are absent, linear alkyl having from 1-6 carbon atoms, branched or cyclic alkyl having from 3 to 6 carbon atoms, linear, branched or cyclic alkenyl or alkynyl, or 1~6 carbon atoms of esters, ether, amide, or polyethyleneoxy unit of formula (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
Additionally R1 and R2 are respectively a chain of atoms selected from C, N, O, S, Si, and P, preferably having 0~500 atoms, which covalently connects to X1 or X2 and Z1 or Z2. The atoms used in forming the R1 and R2 may be combined in all chemically relevant ways, such as forming alkylate, alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxyamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
X1 and X2 are independently selected from N(R3), O, S or CH2; R3 is H, linear alkyl hav-ing from 1-6 carbon atoms, branched or cyclic alkyl having from 3 to 6 carbon atoms, linear, branched or cyclic alkenyl or alkynyl, or 1~6 carbon atoms of esters, ether, amide, or polyethyleneoxy unit of formula (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
2. A cell-binding agent-drug conjugate compound of Formula (II) Wherein:

Cb represents a cell-binding agent, preferably an antibody;
Inside the bracket (parentheses) are the linker-drug components that are conjugated to the cell-binding molecule via a pair of disulfide bond Drug1 and Drug2 represent the same or different cytotoxic agents or drugs, linked to the cell-binding agent via the bridge linker through an alkyl, alkylene, alkenylene, alkynylene, ether, polyoxyalkylene, ester, amine, imine, polyamine, hydrazine, hydrazone, amide, urea, semicarbazide, carbazide, alkoxyamine, urethanes, amino acid, peptide, acyloxylamine, hydroxamic acid, disulfide, thioether, thioester, carbamate, carbonate, heterocyclic ring, heteroalkyl, heteroaromatic, or alkoxime bond, or combination thereof.
n is 1 ~ 20; R1, R2, X1 and X2 are described the same previously in Claim 1.
3. A compound of Formula (III):
Wherein:
Cb, Z1, Z2, n, R1, R2, X1, and X2 are defined the same as in Claims 1 and 2.
4. A compound of Formula (IV):
Wherein:
Drug1, Drug2, Z1, Z2, n, R1, R2, X1, and X2 are defined the same as in Claims 1 and 2.
5. The bridge linker compound of Formula (I) of claims 1, wherein the acetylenedicarboxyl groups is synthesized through the condensation of the acetylenedicarboxylic acid, or its acid derivatives, with the other components containing an amine (1° or 2° amines), alcohol, or thiol on their terminal, as shown in the following scheme (Ia):
Wherein X is X1 or X2 as described in Claim 1 as N(R3), O, or S; R is R1 and/or R2 that are described in Claim 1;
Lv1 and Lv2 are the same or independently OH; F; Cl; Br; I; nitrophenol; N-hydroxy-succinimide (NHS); phenol; dinitrophenol; pentafluorophenol;
tetrafluorophenol; difluoro-phenol; monofluorophenol; pentachlorophenol; triflate; imidazole;
dichlorophenol;
tetrachlorophenol; 1-hydroxybenzotriazole; tosylate; mesylate; 2-ethyl-5-phenylisoxazolium-3'-sulfonate, anhydrides formed its self, or formed with the other anhydride, e.g. acetyl anhy-dride, formyl anhydride; or a intermediate molecule generated with a condensation reagent for peptide coupling reactions, or for Mitsunobu reactions, e.g. condensation reagents are: EDC
(N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide), DCC (Dicyclohexyl-carbodiimide), N,N'-Diisopropylcarbodiimide (DIC), N-Cyclohexyl-N'-(2-morpholinoethyl)carbodiimide metho-p-toluenesulfonate (CMC,or CME-CDI), 1,1'-Carbonyldiimidazole (CDI),TBTU (O-(Benzotria-zol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate), N,N,NP,NP-Tetramethyl-O-(1H-benzotriazol-1-yl)uronium hexafluorophosphate (HBTU), (Benzotriazol-1-yloxy)tris(dimethyl-amino)phosphonium hexafluorophosphate (BOP), (Benzotriazol-1-yloxy)tripyrrolidinophos-phonium hexafluorophosphate (PyBOP), Diethyl cyanophosphonate (DEPC), Chloro-N,N,N',N'tetramethylformamidinium hexafluorophosphate, 1-[Bis(dimethylamino)-methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (HATU), 1-[(Dimethylamino)(morpholino) methylene]-1H-[1,2,3]triazolo[4,5-b]pyridine-1-ium 3-oxide hexafluorophosphate (HDMA), 2-Chloro-1,3-dimethylimidazolidinium hexafluorophosphate (CIP), Chlorotripyrrolidinophosphonium hexafluorophosphate (PyCloP), Fluoro-N,N,N',N'-bis(tetramethylene)formamidinium hexafluorophosphate (BTFFH), N,N,N',N'-Tetramethyl-S-(1-oxido-2-pyridyl)thiuronium hexafluorophosphate, O-(2-Oxo-1(2H)pyridyl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (TPTU), S-(1-Oxido-2-pyridyl)-N,N,N',N'tetramethyl-thiuronium tetrafluoroborate, O-[(Ethoxycarbonyl) cyano-methylenamino]-N,N,N',N'-tetramethyluronium hexafluorophosphate (HOTU), (1-Cyano-2-ethoxy-2-oxoethylidenamino-oxy)dimethylamino-morpholino-carbenium hexafluorophosphate(COMU), O-(Benzotriazol-1-yl)-N,N,N',N'-bis(tetramethylene) uronium hexafluorophosphate (HBPyU), N-Benzyl-N'-cyclohexylcarbodiimide (with, or without polymer-bound), Dipyrrolidino(N-succinimidyloxy)-carbenium hexafluoro-phosphate (HSPyU), Chlorodipyrrolidinocarbenium hexafluorophos-phate (PyC1U), 2-Chloro-1,3-dimethylimidazolidinium tetrafluoroborate(CIB), (Benzotriazol-1-yloxy) dipiperidinocarbenium hexafluorophosphate (HBPipU), O-(6-Chlorobenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (TCTU), Bromotris(dimethylamino)-phosphonium hexafluorophosphate (BroP), Propylphosphonic anhydride (PPACA, T3P
®), 2-Morpholinoethyl isocyanide (MEI), N,N,N',N'-Tetramethyl-O-(N-succinimidyl)uronium hexafluorophosphate (HSTU), 2-Bromo-1-ethyl-pyridinium tetrafluoroborate (BEP), O-[(Ethoxycarbonyl)cyanomethylenamino]-N,N,N',N'-tetramethyluronium tetrafluoroborate (TOTU), 4-(4,6-Dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium chloride (MMTM, DMTMM), N,N,N',N'-Tetramethyl-O-(N-succinimidyl)uronium tetrafluoroborate (TSTU), O-(3,4-Dihydro-4-oxo-1,2,3-benzotriazin-3-yl)-N,N,N',N'-tetramethyluronium tetrafluoro-borate (TDBTU), 1,1'-(Azodicarbonyl)dipiperidine (ADD), Di-(4-chlorobenzyl) azodicar-boxylate (DCAD), Di-tert-butyl azodicarboxylate (DBAD), Diisopropyl azodicarboxylate (DIAD), Di-ethyl azodicarboxylate (DEAD).
6. The bridge linker compound of Formula (I) of claims 1, wherein the acetylenedicarboxyl groups is synthesized through the condensation of bis(trimethylsilyl)acetylene, or acetylene bis-magnesiums (Grignard reagent), or acetylene bis-lithiums (dilithioacetylene), or other di-metal acetylide with acid halides or acid anhydrides, depicted as following reaction equations (Ib), (Ic), (Id), (Ie), (If), (Ig) and (Ih):
Wherein M is Na, K, Li, Cu, CuLi, Sn, Ti, Ca, Mg or Zn.
7. The Formula (II) and (IV) of claims 2, and 4 wherein the Drug1 and Drug2 are the same or independently selected from:
1). Chemotherapeutic agents: a). Alkylating agents: such as Nitrogen mustards:

chlorambucil, chlornaphazine, cyclophosphamide, dacarbazine, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, mannomustine, mitobronitol, melphalan, mitolactol, pipobroman, novembichin, phenesterine, prednimustine, thiotepa, trofosfamide, uracil mustard; CC-1065 (including its adozelesin, carzelesin and bizelesin syn-thetic analogues); duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI);
benzodiazepine dimers (e.g., dimmers of pyrrolobenzodiazepine (PBD) or tomaymycin, indolinobenzodiazepines, imidazobenzothiadiazepines, or oxazolidinobenzodiazepines);
Nitrosoureas: (carmustine, lomustine, chlorozotocin, fotemustine, nimustine, ranimustine);

Alkylsulphonates: (busulfan, treosulfan, improsulfan and piposulfan);
Triazenes: (dacarbazine);
Platinum containing compounds: (carboplatin, cisplatin, oxaliplatin);
aziridines, such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines in-cluding altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaor-amide and trimethylolomelamine]; b). Plant Alkaloids: such as Vinca alkaloids:
(vincristine, vinblastine, vindesine, vinorelbine, navelbin); Taxoids: (paclitaxel, docetaxol) and their analogs, Maytansinoids (DM1, DM2, DM3, DM4, DM5, DM6, DM7, maytansine and ansamitocins) and their analogs, cryptophycins (particularly cryptophycin 1 and cryptophycin 8);
epothilones, eleutherobin, discodermolide, bryostatins, dolostatins, auristatins, tubulysins, cephalostatins;
pancratistatin; a sarcodictyin; spongistatin; c). DNA Topoisomerase Inhibitors: such as [Epipodophyllins: (9-aminocamptothecin, camptothecin, crisnatol, daunomycin, etoposide, etoposide phosphate, irinotecan, mitoxantrone, novantrone, retinoic acids (retinols), teniposide, topotecan, 9-nitrocamptothecin (RFS 2000)); mitomycins: (mitomycin C)]; d).
Antimetabolites:
such as {[Anti-folate: DHFR inhibitors: (methotrexate, trimetrexate, denopterin, pteropterin, aminopterin (4-aminopteroic acid) or the other folic acid analogues); IMP
dehydrogenase Inhib-itors: (mycophenolic acid, tiazofurin, ribavirin, EICAR); Ribonucleotide reductase Inhibitors:
(hydroxyurea, deferoxamine)]; [Pyrimidine analogs: Uracil analogs:
(ancitabine, azacitidine, 6-azauridine, capecitabine (Xeloda), carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, 5-Fluorouracil, floxuridine, ratitrexed (Tomudex)); Cytosine analogs: (cytarabine, cytosine arabinoside, fludarabine); Purine analogs: (azathioprine, fludarabine, mercaptopurine, thiamiprine, thioguanine)]; folic acid replenisher, such as frolinic acid};
e). Hormonal therapies:
such as {Receptor antagonists: [Anti-estrogen: (megestrol, raloxifene, tamoxifen); LHRH ago-nists: (goscrclin, leuprolide acetate); Anti-androgens: (bicalutamide, flutamide, calusterone, dromostanolone propionate, epitiostanol, goserelin, leuprolide, mepitiostane, nilutamide, testolactone, trilostane and other androgens inhibitors)]; Retinoids/Deltoids:
[Vitamin D3 ana-logs: (CB 1093, EB 1089 KH 1060, cholecalciferol, ergocalciferol);
Photodynamic therapies:
(verteporfin, phthalocyanine, photosensitizer Pc4, demethoxy-hypocrellin A);
Cytokines: (Inter-feron-alpha, Interferon-gamma, tumor necrosis factor (TNFs), human proteins containing a TNF
domain)] }; f). Kinase inhibitors, such as BIBW 2992 (anti-EGFR/Erb2), imatinib, gefitinib, pegaptanib, sorafenib, dasatinib, sunitinib, erlotinib, nilotinib, lapatinib, axitinib, pazopanib.
vandetanib, E7080 (anti-VEGFR2), mubritinib, ponatinib (AP24534), bafetinib (INNO-406), bosutinib (SKI-606), cabozantinib, vismodegib, iniparib, ruxolitinib, CYT387, axitinib, tivozanib, sorafenib, bevacizumab, cetuximab, Trastuzumab, Ranibizumab, Panitumumab, ispinesib; g). antibiotics, such as the enediyne antibiotics (e.g.
calicheamicins, especially calicheamicin ..gamma.1, .delta.1, .alpha.1 and .beta.1; dynemicin, including dynemicin A and deoxydynemicin;
esperamicin, kedarcidin, C-1027, maduropeptin, as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin;
chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxoru-bicin, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, nitomycins, mycophe-nolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; f). Others:
such as Polyketides (acetogenins), especially bullatacin and bullatacinone;
gemcitabine, epoxomicins (e. g. carfilzomib), bortezomib, thalidomide, lenalidomide, pomalidomide, tosedostat, zybrestat, PLX4032, STA-9090, Stimuvax, allovectin-7, Xegeva, Provenge, Yervoy, Isoprenylation inhibitors (such as Lovastatin), Dopaminergic neurotoxins (such as 1-methyl-4-phenylpyridinium ion), Cell cycle inhibitors (such as staurosporine), Actinomycins (such as Actinomycin D, dactinomycin), Bleomycins (such as bleomycin A2, bleomycin B2, peplomycin), Anthracyclines (such as daunorubicin, doxorubicin (adriamycin), idarubicin, epirubicin, pirarubicin, zorubicin, mtoxantrone, MDR inhibitors (such as verapamil), Ca2+ ATPase inhibitors (such as thapsigargin), Histone deacetylase inhibitors (Vorinostat, Romidepsin, Panobinostat, Valproic acid, Mocetinostat (MGCD0103), Belinostat, PCI-24781, Entinostat, SB939, Resminostat, Givinostat, AR-42, CUDC-101, sulforaphane, Trichostatin A) ;
Thapsigargin, Celecoxib, glitazones, epigallocatechin gallate, Disulfiram, Salinosporamide A.;
Anti-adrenals, such as aminoglutethimide, mitotane, trilostane; aceglatone;
aldophosphamide glycoside; aminolevulinic acid; amsacrine; arabinoside, bestrabucil;
bisantrene; edatraxate;
defofamine; demecolcine; diaziquone; eflornithine (DEMO), elfomithine;
elliptinium acetate, etoglucid; gallium nitrate; gacytosine, hydroxyurea; ibandronate, lentinan;
lonidamine;
mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet;
pirarubicin;
podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK. .; razoxane;
rhizoxin; sizofiran;
spirogermanium; tenuazonic acid; triaziquone; 2, 2',2"-trichlorotriethylamine;
trichothecenes (especially T-2 toxin, verrucarin A, roridin A and anguidine); urethane, siRNA, antisense drugs;
2). Anti-autoimmune disease agents: cyclosporine, cyclosporine A, aminocaproic acid, azathioprine, bromocriptine, chlorambucil, chloroquine, cyclophosphamide, corticosteroids (e.g.
amcinonide, betamethasone, budesonide, hydrocortisone, flunisolide, fluticasone propionate, fluocortolone danazol, dexamethasone, Triamcinolone acetonide, beclometasone dipropionate), DHEA, enanercept, hydroxychloroquine, infliximab, meloxicam, methotrexate, mofetil, mycophenylate, prednisone, sirolimus, tacrolimus.
3). Anti-infectious disease agents, a). Aminoglyco sides: amikacin, astromicin, gentami-cin (netilmicin, sisomicin, isepamicin), hygromycin B, kanamycin (amikacin, arbekacin, bekanamycin, dibekacin, tobramycin), neomycin (framycetin, paromomycin, ribostamycin), netilmicin, spectinomycin, streptomycin, tobramycin, verdamicin; b).
Amphenicols:
azidamfenicol, chloramphenicol, florfenicol, thiamphenicol; c). Ansamycins:
geldanamycin, herbimycin; d). Carbapenems: biapenem, doripenem, ertapenem, imipenem/cilastatin, meropenem, panipenem; e). Cephems: carbacephem (loracarbef), cefacetrile, cefaclor, cefradine, cefadroxil, cefalonium, cefaloridine, cefalotin or cefalothin, cefalexin, cefaloglycin, cefamandole, cefapirin, cefatrizine, cefazaflur, cefazedone, cefazolin, cefbuperazone, cefcapene, cefdaloxime, cefepime, cefminox, cefoxitin, cefprozil, cefroxadine, ceftezole, cefuroxime, cefixime, cefdinir, cefditoren, cefepime, cefetamet, cefmenoxime, cefodizime, cefonicid, cefoperazone, ceforanide, cefotaxime, cefotiam, cefozopran, cephalexin, cefpimizole, cefpiramide, cefpirome, cefpodoxime, cefprozil, cefquinome, cefsulodin, ceftazidime, cefteram, ceftibuten, ceftiolene, ceftizoxime, ceftobiprole, ceftriaxone, cefuroxime, cefuzonam, cephamycin (cefoxitin, cefotetan, cefmetazole), oxacephem (flomoxef, latamoxef); f). Glyco-peptides: bleomycin, vancomycin (oritavancin, telavancin), teicoplanin (dalbavancin), ramoplanin; g). Glycylcyclines: e. g. tigecycline; g). .beta.-Lactamase inhibitors: penam (sulbactam, tazobactam), clavam (clavulanic acid); i). Lincosamides: clindamycin, lincomycin; j).
Lipopeptides: daptomycin, A54145, calcium-dependent antibiotics (CDA); k).
Macrolides:
azithromycin, cethromycin, clarithromycin, dirithromycin, erythromycin, flurithromycin, josamycin, ketolide (telithromycin, cethromycin), midecamycin, miocamycin, oleandomycin, rifamycins (rifampicin, rifampin, rifabutin, rifapentine), rokitamycin, roxithromycin, spectinomycin, spiramycin, tacrolimus (FK506), troleandomycin, telithromycin;
l). Monobac-tams: aztreonam, tigemonam; m). Oxazolidinones: linezolid; n). Penicillins:
amoxicillin, ampi-cillin (pivampicillin, hetacillin, bacampicillin, metampicillin, talampicillin), azidocillin, azlocillin, benzylpenicillin, benzathine benzylpenicillin, benzathine phenoxymethylpenicillin, clometocillin, procaine benzylpenicillin, carbenicillin (carindacillin), cloxacillin, dicloxacillin, epicillin, flucloxacillin, mecillinam (pivmecillinam), mezlocillin, meticillin, nafcillin, oxacillin, penamecillin, penicillin, pheneticillin, phenoxymethylpenicillin, piperacillin, propicillin, sulbenicillin, temocillin, ticarcillin; o). Polypeptides: bacitracin, colistin, polymyxin B; p).
Quinolones: alatrofloxacin, balofloxacin, ciprofloxacin, clinafloxacin, danofloxacin, difloxacin, enoxacin, enrofloxacin, floxin, garenoxacin, gatifloxacin, gemifloxacin, grepafloxacin, kano trovafloxacin, levofloxacin, lomefloxacin, marbofloxacin, moxifloxacin, nadifloxacin, norfloxacin, orbifloxacin, ofloxacin, pefloxacin, trovafloxacin, grepafloxacin, sitafloxacin, sparfloxacin, temafloxacin, tosufloxacin, trovafloxacin; q). Streptogramins:
pristinamycin, quinupristin/dalfopristin); r). Sulfonamides: mafenide, prontosil, sulfacetamide, sulfamethizole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim, trimethoprim-sulfamethoxazole (co-trimoxazole); s). Steroid antibacterials: e.g. fusidic acid; t).
Tetracyclines: doxycycline, chlortet-racycline, clomocycline, demeclocycline, lymecycline, meclocycline, metacycline, minocycline, oxytetracycline, penimepicycline, rolitetracycline, tetracycline, glycylcyclines (e.g. tigecycline);
u). Other types of antibiotics: annonacin, arsphenamine, bactoprenol inhibitors (Bacitracin), DADAL/AR inhibitors (cycloserine), dictyostatin, discodermolide, eleutherobin, epothilone, ethambutol, etoposide, faropenem, fusidic acid, furazolidone, isoniazid, laulimalide, metronida-zole, mupirocin, mycolactone, NAM synthesis inhibitors (e. g. fosfomycin), nitrofurantoin, paclitaxel, platensimycin, pyrazinamide, quinupristin/dalfopristin, rifampicin (rifampin), tazobactam tinidazole, uvaricin;
4). Anti-viral drugs: a). Entry/fusion inhibitors: aplaviroc, maraviroc, vicriviroc, gp41 (enfuvirtide), PRO 140, CD4 (ibalizumab); b). Integrase inhibitors:
raltegravir, elvitegravir, globoidnan A; c). Maturation inhibitors: bevirimat, vivecon; d). Neuraminidase inhibitors:
oseltamivir, zanamivir, peramivir; e). Nucleosides &nucleotides: abacavir, aciclovir, adefovir, amdoxovir, apricitabine, brivudine, cidofovir, clevudine, dexelvucitabine, didanosine (ddI), elvucitabine, emtricitabine (FTC), entecavir, famciclovir, fluorouracil (5-FU), 3'-fluoro-substituted 2', 3'-dideoxynucleoside analogues (e.g. 3'-fluoro-2',3'-dideoxythymidine (FLT) and 3' -fluoro-2',3' -dideoxyguanosine (FLG), fomivirsen, ganciclovir, idoxuridine, lamivudine (3TC), 1-nucleosides (e.g. .beta.-1-thymidine and .beta.-1-2'-deoxycytidine), penciclovir, racivir, ribavi-rin, stampidine, stavudine (d4T), taribavirin (viramidine), telbivudine, tenofovir, trifluridine valaciclovir, valganciclovir, zalcitabine (ddC), zidovudine (AZT); f). Non-nucleosides: aman-tadine, ateviridine, capravirine, diarylpyrimidines (etravirine, rilpivirine), delavirdine, docosanol, emivirine, efavirenz, foscarnet (phosphonoformic acid), imiquimod, interferon alfa, loviride, lodenosine, methisazone, nevirapine, NOV-205, peginterferon alfa, podophyllotoxin, rifampicin, rimantadine, resiquimod (R-848), tromantadine; g). Protease inhibitors: amprenavir, atazanavir, boceprevir, darunavir, fosamprenavir, indinavir, lopinavir, nelfinavir, pleconaril, ritonavir, saquinavir, telaprevir (VX-950), tipranavir; h). Other types of anti-virus drugs:
abzyme, arbidol, calanolide a, ceragenin, cyanovirin-n, diarylpyrimidines, epigallocatechin gallate (EGCG), foscarnet, griffithsin, taribavirin (viramidine), hydroxyurea, KP-1461, miltefosine, pleconaril, portmanteau inhibitors, ribavirin, seliciclib.

5). A radioisotope that can be selected from (radionuclides) 3H, 11C, 14C, 18F, 32P, 35S, 64Cu, 68Ga, 86Y, 99Tc, 111In, 123I, 124I, 125I, 131I, 133I, 177Lu, 211At, 213Bi.
6). A chromophore molecule, which can be one that has the ability to absorb a kind of light, such as UV light, florescent light, IR light, near IR light, visual light; A class or subclass of xanthophores, erythrophores, iridophores, leucophores, melanophores, cyanophores, fluorophore molecules which are fluorescent chemical compounds re-emitting light upon light, visual phototransduction molecules, photophore molecules, luminescence molecules, luciferin compounds; Non-protein organic fluorophores, such as: Xanthene derivatives (fluorescein, rhodamine, Oregon green, eosin, and Texas red); Cyanine derivatives: (cyanine, indocarbocyanine, oxacarbocyanine, thiacarbocyanine, and merocyanine);
Squaraine derivatives and ring-substituted squaraines, including Seta, SeTau, and Square dyes;
Naphthalene deriva-tives (dansyl and prodan derivatives); Coumarin derivatives; Oxadiazole derivatives (pyridyloxazole, nitrobenzoxadiazole and benzoxadiazole); Anthracene derivatives (anthraquinones, including DRAQ5, DRAQ7 and CyTRAK Orange); Pyrene derivatives (cas-cade blue, etc); Oxazine derivatives (Nile red, Nile blue, cresyl violet, oxazine 170 etc).
Acridine derivatives (proflavin, acridine orange, acridine yellow etc).
Arylmethine derivatives (auramine, crystal violet, malachite green). Tetrapyrrole derivatives (porphin, phthalocyanine, bilirubin); Any analogs and derivatives of the following fluorophore compounds: CF dye (Biotium), DRAQ and CyTRAK probes (BioStatus), BODIPY (Invitrogen), Alexa Fluor (Invi-trogen), DyLight Fluor (Thermo Scientific, Pierce), Atto and Tracy (Sigma Aldrich), FluoProbes (Interchim), Abberior Dyes (Abberior), DY and MegaStokes Dyes (Dyomics), Sulfo Cy dyes (Cyandye), HiLyte Fluor (AnaSpec), Seta, SeTau and Square Dyes (SETA
BioMedicals), Quasar and Cal Fluor dyes (Biosearch Technologies), SureLight Dyes (APC, RPEPerCP, Phycobilisomes)(Columbia Biosciences), APC, APCXL, RPE, BPE (Phyco-Biotech), Allophycocyanin (APC), Aminocoumarin, APC-Cy7 conjugates, BODIPY-FL, Cas-cade Blue, Cy2, Cy3, Cy3.5, Cy3B, Cy5, Cy5.5, Cy7, Fluorescein, FluorX, Hydroxycoumarin, Lissamine Rhodamine B, Lucifer yellow, Methoxycoumarin, NBD, Pacific Blue, Pacific Or-ange, PE-Cy5 conjugates, PE-Cy7 conjugates, PerCP, R-Phycoerythrin(PE), Red 613, Seta-555-Azide, Seta-555-DBCO, Seta-555-NHS, Seta-580-NHS, Seta-680-NHS, Seta-780-NHS, Seta-APC-780, Seta-PerCP-680, Seta-R-PE-670, SeTau-380-NHS, SeTau-405-Maleimide, SeTau-405-NHS, SeTau-425-NHS, SeTau-647-NHS, Texas Red, TRITC, TruRed, X-Rhodamine, AAD (7-aminoactinomycin D, CG-selective), Acridine Orange, Chromomycin A3, CyTRAK
Orange (Biostatus, red excitation dark), DAPI, DRAQ5, DRAQ7, Ethidium Bromide, Hoechst33258, Hoechst33342, LDS 751, Mithramycin, PropidiumIodide (PI), SYTOX
Blue, SYTOX Green, SYTOX Orange, Thiazole Orange, TO-PRO: Cyanine Monomer, TOTO-1, TO-PRO-1, TOTO-3, TO-PRO-3, YOSeta-1, YOYO-1. The fluorophore compounds that can be linked to the linkers of the invention for study cells are selected from the following compounds or their derivatives: DCFH (2'7'Dichorodihydro-fluorescein, oxidized form), DHR
(Dihydrorhodamine 123, oxidized form, light catalyzes oxidation), Fluo-3 (AM
ester. pH > 6), Fluo-4 (AM ester. pH 7.2), Indo-1 (AM ester, low/high calcium (Ca2+)), SNARF(pH 6/9), Allophycocyanin(APC), AmCyan1 (tetramer, Clontech), AsRed2 (tetramer, Clontech), Azami Green (monomer, MBL), Azurite, B-phycoerythrin(BPE), Cerulean, CyPet, DsRed monomer (Clontech), DsRed2 ("RFP", Clontech), EBFP, EBFP2, ECFP, EGFP (weak dimer, Clontech), Emerald (weak dimer, Invitrogen), EYFP (weak dimer, Clontech), GFP (S65A
mutation), GFP
(S65C mutation), GFP (S65L mutation), GFP (S65T mutation), GFP (Y66F
mutation), GFP
(Y66H mutation), GFP (Y66W mutation), GFPuv, HcRed1, J-Red, Katusha, Kusabira Orange (monomer, MBL), mCFP, mCherry, mCitrine, Midoriishi Cyan (dimer, MBL), mKate (TagFP635, monomer, Evrogen), mKeima-Red (monomer, MBL), mKO, mOrange, mPlum, mRaspberry, mRFP1 (monomer, Tsien lab), mStrawberry, mTFP1, mTurquoise2, P3 (phycobilisome complex), Peridinin Chlorophyll (PerCP), R-phycoerythrin(RPE), T-Sapphire, TagCFP (dimer, Evrogen), TagGFP (dimer, Evrogen), TagRFP (dimer, Evrogen), TagYFP (di-mer, Evrogen), tdTomato (tandem dimer), Topaz, TurboFP602 (dimer, Evrogen), TurboFP635 (dimer, Evrogen), TurboGFP (dimer, Evrogen), TurboRFP (dimer, Evrogen), TurboYFP (dimer, Evrogen), Venus, Wild Type GFP, YPet, ZsGreen1 (tetramer, Clontech), ZsYellow1 (tetramer, Clontech).
7). The pharmaceutically acceptable salts, acids or derivatives of any of the above drugs.
8. The compound of Formula (II) and (IV) of claims 2 and 4, wherein "Drug1"
and "Drug2"
are a chromophore molecule, the conjugate compounds of Formula (II) and (IV) of claims 2, and 4, can be used for detecting, monitoring, or studying the interactions and/or functions of the cell binding molecule, and/or of the conjugate with a target, particularly, a targeted cell.
9. The conjugate compound of claims 2 and 4, wherein "Drug1" and "Drug2" are preferred se-lected from tubulysins, calicheamicins, auristatins, maytansinoids, CC-1065 analogs, daunorubicin and doxorubicin compounds, taxanoids (taxanes), cryptophycins, epothilones, benzodiazepine dimers (e.g., dimmers of pyrrolobenzodiazepine (PBD), tomaymycin, anthramycin, indolinobenzodiazepines, imidazobenzothiadiazepines, or oxazolidinobenzodiaze-pines), calicheamicins and the enediyne antibiotics, actinomycin, azaserines, bleomycins, epirubicin, tamoxifen, idarubicin, dolastatins/auristatins (e.g. monomethyl auristatin E, MMAE , MMAF, auristatin PYE, auristatin TP, Auristatins 2-AQ, 6-AQ, EB (AEB), and EFP
(AEFP)), duocarmycins, thiotepa, vincristine, hemiasterlins, nazumamides, microginins, radiosumins, alterobactins, microsclerodermins, theonellamides, esperamicins, siRNA, nucleolytic enzymes, and/or pharmaceutically acceptable salts, acids, or/and their analogues derivatives of any of the above molecules.
10. The conjugate compounds of claims 2 and 3 wherein the cell binding agent/molecule is selected from an antibody, a protein, a vitamin (e.g. folate), peptides, a polymeric micelle, a liposome, a lipoprotein-based drug carrier, a nano-particle drug carrier, a dendrimer, and a mol-ecule above coating with cell-binding ligands, or a combination of above thereof.
11. The cell-binding molecule/agent according to claims 2, 3, and 10 is preferred an antibody, a single chain antibody, an antibody fragment that binds to the target cell, a monoclonal anti-body, a single chain monoclonal antibody, or a monoclonal antibody fragment that binds the target cell, a chimeric antibody, a chimeric antibody fragment that binds to the target cell, a do-main antibody, a domain antibody fragment that binds to the target cell, a resurfaced antibody, a resurfaced single chain antibody, or a resurfaced antibody fragment that binds to the target cell, a humanized antibody or a resurfaced antibody, a humanized single chain antibody, or a human-ized antibody fragment that binds to the target cell, a lymphokine, a hormone, a vitamin, a growth factor, a colony stimulating factor, or a nutrient-transport molecule.
12. The cell-binding molecule/agent according to claims 2, 3, and 10 can be any agent that is able to target against a tumor cell, a virus infected cell, a microorganism infected cell, a parasite infected cell, an autoimmune disease cell, an activated tumor cells, a myeloid cell, an activated T-cell, an affecting B cell, or a melanocyte.
13. The cell-binding molecule/agent according to claims 2, 3, and 10 can be any agent/molecule that is able to against any one of the following antigens or receptors: CD3, CD4, CD5, CD6, CD7, CD8, CD9, CD10, CD11a, CD11b, CD11c, CD12w, CD14, CD15, CD16, CDw17, CD18, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD36, CD37, CD38, CD39, CD40, CD41, CD42, CD43, CD44, CD45, CD46, CD47, CD48, CD49b, CD49c, CD51, CD52, CD53, CD54, CD55, CD56, CD58, CD59, CD61, CD62E, CD62L, CD62P, CD63, CD66, CD68, CD69, CD70, CD72, CD74, CD79, CD79a, CD79b, CD80, CD81, CD82, CD83, CD86, CD87, CD88, CD89, CD90, CD91, CD95, CD96, CD98, CD100, CD103, CD105, CD106, CD109, CD117, CD120, CD125, CD126, CD127, CD133, CD134, CD135, CD138, CD141, CD142, CD143, CD144, CD147, CD151, CD147, CD152, CD154, CD156, CD158, CD163, CD166, .CD168, CD174, CD180, CD184, CDw186, CD194, CD195, CD200, CD200a, CD200b, CD209, CD221, CD227, CD235a, CD240, CD262, CD271, CD274, CD276 (B7-H3), CD303, CD304, CD309, CD326, 4-1BB, 5AC, 5T4 (Trophoblast glycoprotein, TPBG, 5T4, Wnt-Activated Inhibitory Factor 1 or WAIF1), Adenocarcinoma antigen, AGS-5, AGS-22M6, Activin receptor-like ki-nase 1, AFP, AKAP-4, ALK, Alpha intergrin, Alpha v beta6, Amino-peptidase N, Amyloid beta, Androgen receptor, Angiopoietin 2, Angiopoietin 3, Annexin Al, Anthrax toxin protective anti-gen, Anti-transferrin receptor, AOC3 (VAP-1), B7-H3, Bacillus anthracis anthrax, BAFF (B-cell activating factor), B-lymphoma cell, bcr-abl, Bombesin, BORIS, C5, C242 antigen, CA125 (carbohydrate antigen 125, MUC16), CA-IX (or CAIX, carbonic anhydrase 9), CALLA, CanAg, Canis lupus familiaris IL31, Carbonic anhydrase IX, Cardiac myosin, CCL11(C-C
motif chem-okine 11), CCR4 (C-C chemokine receptor type 4, CD194), CCR5, CD3E (epsilon), CEA
(Carcinoembryonic antigen), CEACAM3, CEACAM5 (carcinoembryonic antigen), CFD
(Fac-tor D), Ch4D5, Cholecystokinin 2 (CCK2R), CLDN18 (Claudin-18), Clumping factor A, CRIPTO, FCSF1R (Colony stimulating factor 1 receptor, CD115), CSF2 (colony stimulating factor 2, Granulocyte-macrophage colony-stimulating factor (GM-CSF)), CTLA4 (cytotoxic T-lymphocyte-as sociated protein 4), CTAA16.88 tumor antigen, CXCR4 (CD184), C-X-C chem-okine receptor type 4, cyclic ADP ribose hydrolase, Cyclin B1, CYP1B1, Cytomegalovirus, Cy-tomegalovirus glycoprotein B, Dabigatran, DLL4 (delta-like-ligand 4), DPP4 (Dipeptidyl-peptidase 4), DR5 (Death receptor 5), E. coli shiga toxin type-1, E. coli shiga toxin type-2, ED-B, EGFL7 (EGF-like domain-containing protein 7), EGFR, EGFRII, EGFRvIII, Endoglin (CD105), Endothelin B receptor, Endotoxin, EpCAM (epithelial cell adhesion molecule), EphA2, Episialin, ERBB2 (Epidermal Growth Factor Receptor 2), ERBB3, ERG
(TMPRSS2 ETS fusion gene), Escherichia coli, ETV6-AML, FAP (Fibroblast activation protein alpha), FCGR1, alpha-Fetoprotein, Fibrin II, beta chain, Fibronectin extra domain-B, FOLR (folate receptor), Folate receptor alpha, Folate hydrolase, Fos-related antigen 1.F
protein of respiratory syncytial virus, Frizzled receptor, Fucosyl GM1, GD2 ganglioside, G-28 (a cell surface antigen glyvolipid), GD3 idiotype, GloboH, Glypican 3, N-glycolylneuraminic acid, GM3, GMCSF re-ceptor a-chain, Growth differentiation factor 8, GP100, GPNMB (Transmembrane glycoprotein NMB), GUCY2C (Guanylate cyclase 2C, guanylyl cyclase C(GC-C), intestinal Guanylate cyclase, Guanylate cyclase-C receptor, Heat-stable enterotoxin receptor (hSTAR)), Heat shock proteins, Hemagglutinin, Hepatitis B surface antigen, Hepatitis B virus, HER1 (human epider-mal growth factor receptor 1), HER2, HER2/neu, HER3 (ERBB-3), IgG4, HGF/SF
(Hepato-cyte growth factor/scatter factor), HHGFR, HIV-1, Histone complex, HLA-DR
(human leuko-cyte antigen), HLA-DR10, HLA-DRB , HMWMAA, Human chorionic gonadotropin, HNGF, Human scatter factor receptor kinase, HPV E6/E7, Hsp90, hTERT, ICAM-1 (Intercellular Ad-hesion Molecule 1), Idiotype, IGF1R (IGF-1, insulin-like growth factor 1 receptor), IGHE, IFN-.gamma., Influenza hemagglutinin, IgE, IgE Fc region, IGHE, IL-1, IL-2 receptor (interleukin 2 recep-tor), IL-4, IL-5, IL-6, IL-6R (interleukin 6 receptor), IL-9, IL-10, IL-12, IL-13, IL-17, IL-17A, IL-20, IL-22, IL-23, IL31RA, ILGF2 (Insulin-like growth factor 2), Integrins (.alpha.4, .alpha.IIb.beta.3, .alpha.v.beta.3, .alpha.4.beta.7, .alpha.5.beta.1, .alpha.6.beta.4, .alpha.7.beta.7, .alpha.11.beta.3, .alpha.5.beta.5, .alpha.v.beta.5), Interferon gamma-induced protein, ITGA2, ITGB2, KIR2D, LCK, Le, Legumain, Lewis-Y antigen, LFA-1(Lymphocyte function-associated antigen 1, CD11a), LHRH, LINGO-1, Lipoteichoic acid, LIV1A, LMP2, LTA, MAD-CT-1, MAD-CT-2, MAGE-1, MAGE-2, MAGE-3, MAGE A1, MAGE A3, MAGE 4, MART1, MCP-1, MIF
(Macrophage migration inhibitory factor, or glycosylation-inhibiting factor (GIF)), MS4A1 (membrane-spanning 4-domains subfamily A member 1), MSLN (mesothelin), MUC1(Mucin 1, cell surface associated (MUC1) or polymorphic epithelial mucin (PEM)), MUC1-KLH, MUC16 (CA125), MCP1(monocyte chemotactic protein 1), MelanA/MART1, ML-IAP, MPG, (membrane-spanning 4-domains subfamily A), MYCN, Myelin-associated glycoprotein, Myostatin, NA17, NARP-1, NCA-90 (granulocyte antigen), Nectin-4 (ASG-22ME), NGF, Neu-ral apoptosis-regulated proteinase 1, NOGO-A, Notch receptor, Nucleolin, Neu oncogene prod-uct, NY-BR-1, NY-ESO-1, OX-40, OxLDL (Oxidized low-density lipoprotein), OY-TES1, P21, p53 nonmutant, P97, Page4, PAP, Paratope of anti-(N-glycolylneuraminic acid), PAX3, PAX5, PCSK9, PDCD1 (PD-1, Programmed cell death protein 1,CD279), PDGF-R.alpha. (Alpha-type platelet-derived growth factor receptor ), PDGFR-.beta., PDL-1, PLAC1, PLAP-like testicular alkaline phosphatase, Platelet-derived growth factor receptor beta, Phosphate-sodium co-transporter, PMEL 17, Polysialic acid, Proteinase3 (PR1), Prostatic carcinoma, PS
(Phosphatidylserine), Prostatic carcinoma cells, Pseudomonas aeruginosa, PSMA, PSA, PSCA, Rabies virus glycoprotein, RHD (Rh polypeptide 1 (RhPI), CD240), Rhesus factor, RANKL, RhoC, Ras mutant, RGS5, ROBO4, Respiratory syncytial virus, RON, Sarcoma translocation breakpoints, SART3, Sclerostin, SLAMF7 (SLAM family member 7), Selectin P, (Syndecan 1), sLe(a), Somatomedin C, SIP (Sphingosine-1-phosphate), Somatostatin, Sperm protein 17, SSX2, STEAP1 (six-transmembrane epithelial antigen of the prostate 1), STEAP2, STn, TAG-72 (tumor associated glycoprotein 72), Survivin, T-cell receptor, T
cell transmembrane protein, TEM1 (Tumor endothelial marker 1), TENB2, Tenascin C
(TN-C), TGF-.alpha., TGF-.beta. (Transforming growth factor beta), TGF-.beta.1, TGF-.beta.2 (Transforming growth fac-tor-beta 2), Tie (CD202b), Tie2, TIM-1 (CDX-014), Tn, TNF, TNF-.alpha., TNFRSF8, TNFRSF10B
(tumor necrosis factor receptor superfamily member 10B), TNFRSF13B (tumor necrosis factor receptor superfamily member 13B), TPBG (trophoblast glycoprotein), TRAIL-R1 (Tumor ne-crosis apoprosis Inducing ligand Receptor 1), TRAILR2 (Death receptor 5 (DR5)), tumor-associated calcium signal transducer 2, tumor specific glycosylation of MUC1, TWEAK recep-tor, TYRP1(glycoprotein 75), TRP-2, Tyrosinase, VCAM-1 (CD106), VEGF, VEGF-A, VEGF-2 (CD309), VEGFR-1, VEGFR2, or vimentin, WT1, XAGE 1, or cells expressing any insulin growth factor receptors, or any epidermal growth factor receptors.
14. The tumor cell according to claim 12 is selected from lymphoma cells, myeloma cells, re-nal cells, breast cancer cells, prostate cancer cells, ovarian cancer cells, colorectal cancer cells, gastric cancer cells, squamous cancer cells, small-cell lung cancer cells, none small-cell lung cancer cells, testicular cancer cells, or any cells that grow and divide at an unregulated, quick-ened pace to cause cancers.
15. The linkage components R1 and/or R2 according to claims 1, 2, 3, and/or 4, can be com-posed of one or more linker components of: 6-maleimidocaproyl (MC), maleimido propanoyl (MP), valine-citrulline (val-cit), alanine-phenylalanine (ala-phe), lysine-phenylalanine (lys-phe), p-aminobenzyloxycarbonyl (PAB), 4-thio-pentanoate (SPP), 4-(N-maleimidomethyl)cyclo-hexane-1-carboxylate (MCC), 4-thio-butyrate (SPDB), maleimidoethyl (ME), 4-thio-2-hydroxysulfonyl-butyrate (2-Sulfo-SPDB), pyridinyl-dithiol (PySS), alkoxy amino (AOA), ethyleneoxy (EO), 4-methyl-4-dithio-pentanoic (MPDP), azido (N3), alkynyl, dithio, peptide, and/or (4-acetyl)aminobenzoate (SIAB).
16. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a Tubulysin ana-log, the conjugate compound of Formula (II) is preferred structures of T01, T02, T03, T04, T05, and T06 as following:
Wherein mAb is an antibody; Z3 is H, R1, OP(O)(OM1)(0M2), OCH2OP(O)(OM1)(OM2), OSO3M1, or O-, NH-, S- or CH2- glycoside (gluco side, galactoside, mannoside, giucuronoside, alloside, fructoside); M1 and M2 are independently H, Na, K, Ca, Mg, NR1R2R3;
n is 1~20; X1, X2, R1, R2 and R3 are the same defined in Claim 1.
17. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a Calicheamicin analog, the conjugate compound of Formula (II) is preferred structures of C01 as following:
Wherein mAb is an antibody; n is 1-20; X1, X2, R1, R2 and R3 are the same defined in Claim 1.
18. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a Maytansinoid analog, the conjugate compound of Formula (II) is preferred structures of M01 as following:
Wherein mAb is an antibody; n is 1~20; X1, X2, R1, R2 and R3 are the same defined in Claim 1.
19. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a Taxane analog, the conjugate compound of Formula (II) is preferred structures of Tx01, Tx02 and Tx03 as fol-lowing:
Wherein mAb is an antibody; n is 1-20; X1, X2, R1 and R2 are the same defined in Claim 1.
20. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a CC-1065 ana-logue and/or doucarmycin analog, the conjugate compound of Formula (II) is preferred struc-tures of CC01, CC02, and CC03 as following:

Wherein mAb is an antibody; n is 1~20; Z4 is H, PO(OM1)(OM2), CH2PO(OM1)(OM2), SO3M1, CH3N(CH2CH2)2NC(O)-, O(CH2CH2)2NC(O)-, or glycoside; X3 is O, NH, NHC(O), OC(O), -C(O)O, or absent; X1, X2, R1, R2, Ml, and M2 are the same defined in Claim 1.
21. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a Daunorubicin or Doxorubicin analogue, the conjugate compound of Formula (II) is preferred structures of Da01, Da02, Da03 and Da04 as following:

Wherein mAb is an antibody; n is 1-20; X3 iS H, O, NH, NHC(O), NHC(O)NH, C(O), or OC(O); X1, X2, R1, and R2 are the same defined in Claim 1.
22. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are an Auristatin and dolastatin analogue, the conjugate compound of Formula (II) is preferred structures of Au01, Au02, Au03, Au04, and Au05 as following:
Wherein mAb is an antibody; n is 1~20; X3 iS CH2, O, NH, NHC(O), NHC(O)NH, C(O), OC(O) or absent; X4 is CH2, C(O), C(O)NH, C(O)N(R1), or C(O)O; Z3 is H, R1, OP(O)(OM1)(OM2), OCH2OP(O)(OM1)(OM2), OSO3M1, or O-, NH-, S- or CH2- glycoside (glucoside, galactoside, mannoside, glucuronoside, alloside, fructoside); X1, X2, R1, R2 and R3 are the same defined in Claim 1.
23. The conjugate compound of claim 2, wherein "Drug1" and "Drug2" are a benzodiazepine dimer analogues, the conjugate compound of Formula (II) is preferred structures of PB01, PB02, PB03, PB04, PB05, PB06, PB07 and PB08 as following:
Wherein mAb is an antibody; n is 1~20; X3 is CH2, O, NH, NHC(O), NHC(O)NH, C(O), OC(O) or absent; X4 is CH2, C(O), C(O)NH, C(O)N(R1), or C(O)O; X1, X2, R1, R2 and R3 are the same defined in Claim 1. In addition, R1 and/or R2 can be absent.
24. A pharmaceutical composition comprising a therapeutically effective amount of the con-jugate compounds of claims 2, 16, 17, 18, 19, 20, 21, 22 and/or 23, and a pharmaceutically ac-ceptable salt, carrier, diluent, or excipient therefore, or a combination thereof, for the treatment or prevention of a cancer, or an autoimmune disease, or an infectious disease.
25. The conjugate of claim 2, 16, 17, 18, 19, 20, 21, 22, 23 or 24, having in vitro, in vivo or ex vivo cell killing activity.
26. The conjugate compound of claim 2, 16, 17, 18, 19, 20, 21, 22 or 23, can comprise either a peptides of 1~20 units of natural or unnatural amino acids, or a p-aminobenzyl unit, or a 6-maleimidocaproyl unit, or a disulfide unit, or a thioether unit, or a hydrozone unit, a triazole unit, or an alkoxime unit, inside the linkage components R1 and/or R2.
27. The conjugate of claim 2, 16, 17, 18, 19, 20, 21, 22 or 23, wherein the linkage components R1 and/or R2 can be cleavable by a protease.
28. A pharmaceutical composition comprising a therapeutically effective amount of the con-jugate of claim 2, 16, 17, 18, 19, 20, 21, 22, 23 or/and 24, administered concurrently with the other therapeutic agents such as the chemotherapeutic agent, the radiation therapy, immunother-apy agents, autoimmune disorder agents, anti-infectious agents or the other conjugates for syn-ergistically effective treatment or prevention of a cancer, or an autoimmune disease, or an infec-tious disease.
29. The synergistic agents according to claim 28 are preferably selected from one or several of the following drugs: Abatacept (Orencia), Abiraterone acetate (Zytiga ®), Acetaminophen /hydrocodone, Adalimumab, afatinib dimaleate (Gilotrif ®), alemtuzumab (Campath ®), Alitretinoin (Panretin ®), ado-trastuzumab emtansine (Kadcyla .TM.), Amphetamine mixed salts (Amphetamine/dextroamphetamine, or Adderall XR), anastrozole (Arimidex ®), Aripiprazole, Atazanavir, Atezolizumab (MPDL3280A), Atorvastatin, axitinib (Inlyta ®), belinostat (Beleodaq .TM.), Bevacizumab (Avastin ®), Cabazitaxel (Jevtana ®), Cabozantinib (Cometriq .TM.), bexarotene (Targretin ®), blinatumomab (Blincyto .TM.), Bortezomib (Velcade ®), bosutinib (Bosulif ®), brentuximab vedotin (Adcetris ®), Budesonide, Budesonide/formoterol, Buprenorphine, Capecitabine, carfilzomib (Kyprolis ®), Celecoxib, ceritinib (LDK378/Zykadia), Cetuximab (Erbitux ®), Ciclosporin, Cinacalcet, crizotinib (Xalkori ®), Dabigatran, dabrafenib (Tafinlar ®), Darbepoetin alfa, Darunavir, imatinib mesylate (Gleevec ®), dasatinib (Sprycel ®), denileukin diftitox (Ontak ®), Denosumab (Xgeva ®), De-pakote, Dexlansoprazole, Dexmethylphenidate, Dinutuximab (Unituxin .TM.), Doxycycline, Du-loxetine, Emtricitabine/Rilpivirine/Tenofovir disoproxil fumarate, Emtricitabine/tenofovir/efavirenz, Enoxaparin, Enzalutamide (Xtandi ®), Epoetin alfa, erlotinib (Tarceva ®), Esomeprazole, Eszopiclone, Etanercept, Everolimus (Afinitor ®), exemestane (Aromasin ®), everolimus (Afinitor ®), Ezetimibe, Ezetimibe/simvastatin, Fenofibrate, Filgrastim, fingolimod, Fluticasone propionate, Fluticasone/salmeterol, fulvestrant (Faslodex ®), gefitinib (Iressa ®), Glatiramer, Goserelin acetate (Zoladex), Imatinib (Gleevec), Ibritumomab tiuxetan (Zevalini ®), ibrutinib (Imbruvica .TM.), idelalisib (Zydelig ®), Infliximab, Insulin aspart, Insulin detemir, Insulin glargine, Insulin lispro, Interferon beta 1a, Interferon beta 1b, lapatinib (Tykerb ®), Ipilimumab (Yervoy ®), Ipratropium bromide/salbutamol, Lanreotide acetate (Somatuline ® Depot), lenaliomide (Revlimid ®), lenvatinib mesylate (Lenvima .TM.), letrozole (Femara ®), Levothyroxine, Levothyroxine, Lidocaine, Linezolid, Liraglutide, Lisdexamfetamine, MEDI4736 (AstraZeneca, Celgene), Memantine, Methylphenidate, Metoprolol, Modafinil, Mometasone, Nilotinib (Tasigna ®), Nivolumab (Opdivo ®), ofatumumab (Arzerra ®), obinutuzumab (Gazyva .TM.), olaparib (Lynparza .TM.), Olmesartan, Olmesartan/hydrochlorothiazide, Omalizumab, Omega-3 fatty acid ethyl esters, Oseltamivir, Oxycodone, palbociclib (Ibrance ®), Palivizumab, panitumumab (Vectibix ®), panobinostat (Farydak ®), pazopanib (Votrient ®), pembrolizumab (Keytruda ®), Pemetrexed (Alimta), pertuzumab (Perjeta .TM.), Pneumococcal conjugate vaccine, pomalidomide (Pomalyst ®), Pregabalin, Quetiapine, Rabeprazole, radium 223 chloride (Xofigo ®), Raloxifene, Raltegravir, ramucirumab (Cyramza ®), Ranibizumab, regorafenib (Stivarga ®), Rituximab (Rituxan ®), Rivaroxaban, romidepsin (Istodax ®), Rosuvastatin, ruxolitinib phosphate (Jakafi .TM.), Salbutamol, Sevelamer, Sildenafil, siltuximab (Sylvant .TM.), Sitagliptin, Sitagliptin/metformin, Solifenacin, Sorafenib (Nexavari ®), Sunitinib (Sutent ®),Tadalafil, tamoxifen, Telaprevir, temsirolimus (Torisel ®), Tenofovir/emtricitabine, Testosterone gel, Thalidomide (Immunoprin, Talidex), Tiotropium bromide, toremifene (Fareston ®), trametinib (Mekinist ®), Trastuzumab, Tretinoin (Vesanoid ®), Ustekinumab, Valsartan, vandetanib (Caprelsa ®), vemurafenib (Zelboraf ®), vorinostat (Zolinza ®), ziv-aflibercept (Zaltrap ®), Zostavax., and their analogs, derivatives, pharmaceutically acceptable salts, carriers, diluents, or excipients therefore, or a combination above thereof,
CA2991384A 2015-07-04 2015-07-04 Bridge linkers for conjugation of a cell-binding molecule Active CA2991384C (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2015/055051 WO2015151080A2 (en) 2015-07-04 2015-07-04 Specific conjugation of a cell-binding molecule

Publications (2)

Publication Number Publication Date
CA2991384A1 true CA2991384A1 (en) 2015-10-08
CA2991384C CA2991384C (en) 2022-11-08

Family

ID=54241397

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2991384A Active CA2991384C (en) 2015-07-04 2015-07-04 Bridge linkers for conjugation of a cell-binding molecule

Country Status (5)

Country Link
EP (1) EP3317246A4 (en)
JP (1) JP6700321B2 (en)
CN (2) CN108811499B (en)
CA (1) CA2991384C (en)
WO (1) WO2015151080A2 (en)

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10441654B2 (en) 2014-01-24 2019-10-15 Children's Hospital Of Eastern Ontario Research Institute Inc. SMC combination therapy for the treatment of cancer
WO2016001485A1 (en) 2014-06-30 2016-01-07 Glykos Finland Oy Saccharide derivative of a toxic payload and antibody conjugates thereof
CN115300640A (en) * 2015-08-10 2022-11-08 杭州多禧生物科技有限公司 Novel linkers and their use for specific coupling of drugs and biomolecules
JP7330515B2 (en) * 2015-08-10 2023-08-22 ハンジョウ ディーエーシー バイオテック シーオー.,エルティディ. Novel conjugates and their use in specific conjugation of biomolecules and drugs
US11129910B2 (en) * 2016-02-04 2021-09-28 Hangzhou Dac Biotech Co., Ltd. Specific conjugation linkers, specific immunoconjugates thereof, methods of making and uses such conjugates thereof
TWI808055B (en) 2016-05-11 2023-07-11 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-1 inhibitors
TWI794171B (en) 2016-05-11 2023-03-01 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-l1 inhibitors
KR20230149857A (en) 2016-07-07 2023-10-27 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Antibody adjuvant conjugates
CN106474147B (en) * 2016-10-14 2022-07-05 上海颉隆投资管理有限公司 Dressing for preventing and controlling human papilloma virus infection and preparation method thereof
US20210308277A1 (en) 2016-11-14 2021-10-07 Hangzhou Dac Biotech Co., Ltd. Conjugation linkers, cell binding molecule-drug conjugates containing the linkers, methods of making and uses such conjugates with the linkers
JP7300394B2 (en) 2017-01-17 2023-06-29 ヘパリジェニックス ゲーエムベーハー Protein kinase inhibition to promote liver regeneration or reduce or prevent hepatocyte death
CN109444349B (en) * 2018-12-26 2020-07-07 中国科学院地球化学研究所 Method for measuring metabolic water utilization share and actual water demand of indoor plant
WO2020190725A1 (en) 2019-03-15 2020-09-24 Bolt Biotherapeutics, Inc. Immunoconjugates targeting her2
CN111773183B (en) * 2019-04-04 2022-02-01 沈阳药科大学 Cu (DDC)2Nano nuclear liposome and preparation method and application thereof
JP2022532342A (en) 2019-05-14 2022-07-14 ニューベイション・バイオ・インコーポレイテッド Anti-cancer nuclear hormone receptor targeting compound
KR20220029725A (en) 2019-06-29 2022-03-08 항저우 디에이씨 바이오테크 씨오, 엘티디 Cell-binding molecule-tubulin derivative conjugate and method for preparing same
CN110308145A (en) * 2019-07-10 2019-10-08 迪瑞医疗科技股份有限公司 A kind of chlamydia trachomatis glycogen detection reagent, chlamydia trachomatis glycogen test strip and preparation method thereof
CN112341521B (en) * 2019-08-09 2024-06-25 上海翰森生物医药科技有限公司 Small molecule active compound and antibody conjugate thereof, preparation method and medical application thereof
EP4058464A1 (en) 2019-11-13 2022-09-21 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
CN110835405B (en) * 2019-11-28 2020-09-15 安阳师范学院 Polymer biosensor for tumor detection and preparation method and application thereof
CN111879684B (en) * 2020-06-18 2021-12-21 山东大学 Phagocyte function detection method based on flow cytometry
CN113925973A (en) * 2020-07-14 2022-01-14 辽宁中健医药科技有限公司 Polypeptide coupled drug, preparation method and application thereof
CN112409242B (en) * 2020-09-25 2022-07-12 华南农业大学 Amlodipine hapten, artificial antigen, antibody and preparation method and application thereof
CN112379029B (en) * 2020-11-10 2022-03-08 苏州艾迪迈医疗科技有限公司 Method for detecting concentration of anti-novel coronavirus drug in blood
US20240148888A1 (en) * 2021-02-23 2024-05-09 Children's Medical Center Corporation Intercellular adhesion molecule 1 (icam1) antibody drug conjugate and uses thereof
MX2023011241A (en) 2021-03-23 2023-10-03 Nuvation Bio Inc Anti-cancer nuclear hormone receptor-targeting compounds.
CN113214403B (en) * 2021-04-25 2023-06-09 重庆威斯腾前沿生物研究院有限责任公司 Streptavidin magnetic bead and preparation method thereof
EP4334314A1 (en) 2021-05-03 2024-03-13 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
CN113913535A (en) * 2021-11-30 2022-01-11 江西农业大学 Causal gene for identifying blue peacock white feather character and application thereof
WO2023125349A1 (en) * 2021-12-27 2023-07-06 山东先声生物制药有限公司 Anti-gucy2c antibody and application thereof
CN116239513B (en) * 2023-05-05 2023-08-18 天津凯莱英制药有限公司 Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug
CN116813710B (en) * 2023-07-05 2024-07-12 重庆药友制药有限责任公司 Preparation method of carfilzomib

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0228358D0 (en) * 2002-12-05 2003-01-08 Unilever Plc Improvements relating to fabric treatment
KR20100137585A (en) * 2008-04-30 2010-12-30 이뮤노젠 아이엔씨 Potent conjugates and hydrophilic linkers
CN101723961A (en) * 2009-12-08 2010-06-09 刘超美 Beta-lactam twin antibiotic compound, preparation method thereof and use thereof
EP2610263A1 (en) * 2011-12-30 2013-07-03 Brossmer, Reinhard Sialic acid dimers
AU2012395148B2 (en) * 2012-11-24 2016-10-27 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
CN103933575B (en) * 2013-01-23 2017-09-29 上海新理念生物医药科技有限公司 A kind of three flute profile connexons and its application
US11229711B2 (en) * 2013-06-06 2022-01-25 Magenta Therapeutics, Inc. Linkers for antibody-drug conjugates and related compounds, compositions, and methods of use
CN103467590B (en) * 2013-09-02 2016-04-13 深圳大学 Bioconjugation body, the purine compound preparing it, synthetic method, medicinal preparations and its application in immunomodulatory
NZ717003A (en) * 2013-09-02 2019-05-31 Hangzhou Dac Biotech Co Ltd Novel cytotoxic agents for conjugation of drugs to cell binding molecule

Also Published As

Publication number Publication date
CA2991384C (en) 2022-11-08
JP6700321B2 (en) 2020-05-27
EP3317246A2 (en) 2018-05-09
EP3317246A4 (en) 2019-02-27
WO2015151080A3 (en) 2016-06-02
JP2018523639A (en) 2018-08-23
CN113698335A (en) 2021-11-26
CN108811499B (en) 2021-09-24
WO2015151080A2 (en) 2015-10-08
CN108811499A (en) 2018-11-13

Similar Documents

Publication Publication Date Title
AU2022206809B2 (en) Bridge Linkers for Conjugation of Cell-Binding Molecules
US10328157B2 (en) Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
US20220323602A1 (en) Conjugation linkers, cell binding molecule-drug conjugates containing the linkers, methods of making and uses such conjugates with the linkers
US20220378951A1 (en) Specific conjugation linkers, specific immunoconjugates thereof, methods of making and uses such conjugates thereof
US10307391B2 (en) Disulfur bridge linkers for conjugation of a cell-binding molecule
CA2991384C (en) Bridge linkers for conjugation of a cell-binding molecule
CA2989269C (en) Hydrophilic linkers for conjugation of a cytotoxic agent or chromophore molecule to a cell-binding molecule
US10975112B2 (en) Linkers for conjugation of cell-binding molecules
WO2015155753A2 (en) Novel linkers and their uses in specific conjugation of drugs to a biological molecule

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20180104

EEER Examination request

Effective date: 20180104

EEER Examination request

Effective date: 20180104