WO2014080251A1 - Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules - Google Patents

Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules Download PDF

Info

Publication number
WO2014080251A1
WO2014080251A1 PCT/IB2012/056700 IB2012056700W WO2014080251A1 WO 2014080251 A1 WO2014080251 A1 WO 2014080251A1 IB 2012056700 W IB2012056700 W IB 2012056700W WO 2014080251 A1 WO2014080251 A1 WO 2014080251A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
drug
inhibitors
cells
antibody
Prior art date
Application number
PCT/IB2012/056700
Other languages
French (fr)
Inventor
R. Yongxin ZHAO
Original Assignee
Hangzhou Dac 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=50775615&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014080251(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Hangzhou Dac Biotech Co., Ltd. filed Critical Hangzhou Dac Biotech Co., Ltd.
Priority to PCT/IB2012/056700 priority Critical patent/WO2014080251A1/en
Priority to ES12888899T priority patent/ES2701076T3/en
Priority to CN201280076481.3A priority patent/CN105849086B/en
Priority to AU2012395148A priority patent/AU2012395148B2/en
Priority to EP12888899.7A priority patent/EP2922818B1/en
Priority to CA2891280A priority patent/CA2891280C/en
Priority to JP2015543532A priority patent/JP6133431B2/en
Priority to US14/432,073 priority patent/US10131682B2/en
Publication of WO2014080251A1 publication Critical patent/WO2014080251A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/572Five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • A61K31/10Sulfides; Sulfoxides; Sulfones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • 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
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/30Phosphinic acids R2P(=O)(OH); Thiophosphinic acids, i.e. R2P(=X)(XH) (X = S, Se)
    • C07F9/301Acyclic saturated acids which can have further substituents on alkyl
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/30Phosphinic acids R2P(=O)(OH); Thiophosphinic acids, i.e. R2P(=X)(XH) (X = S, Se)
    • C07F9/302Acyclic unsaturated acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/30Phosphinic acids R2P(=O)(OH); Thiophosphinic acids, i.e. R2P(=X)(XH) (X = S, Se)
    • C07F9/32Esters thereof
    • C07F9/3205Esters thereof the acid moiety containing a substituent or a structure which is considered as characteristic
    • C07F9/3235Esters of poly(thio)phosphinic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/58Pyridine rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the preparation of novel hydrophilic linkers used for the conjugation of a drug, 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 comprising either modification of drugs with these hydrophilic linkers first, followed by reaction with cell-binding agents; or modification of cell-binding agents 10 with these hydrophilic linkers first, followed by reaction with drugs.
  • anticancer drugs such as Albumin-based drug carriers; Carbohydrate-enhanced
  • Vitamins e.g. folates as carriers; Nanoparticle carriers,; Liposome carriers, e. g.
  • pegylated liposomes enclosed in a polyethylene glycol bilayer
  • An ideal drug delivery vehicle must be non-toxic, biocompatible, non-immunogenic, and biodegradable (Scott, R; Crabbe, D; et al (2008) Expert Opin. Drug Deli. 5, 459) and avoid recognition by the host's defense mechanisms (Saltzman, W.; Torchilin, V. (2008). "Drug delivery systems” Access Science. McGraw-Hill Co.).
  • the link between the delivery vehicles, in particular, antibodies and the cell-killing agent plays a critical role in the development of targeted drug delivery systems, as the nature of the linker significantly affects the potency, selectivity and the pharmacokinetics of the resulting conjugates (Zhao, R.;
  • linkers had been used for preparation of cell binding agent-drug conjugates that have entered the clinic: (a) acid-labile linkers, exploiting the acidic endosomal and lysosomal intracellular microenvironment; (b) linkers cleavable by lysosomal proteases; (c) chemically stable thioether linkers that release a lysyl adduct after proteolytic degradation of the antibody inside the cell; and (d) disulfide - containing linkers, which are cleaved upon exposure to an intracellular thiol (Zhao, R.; Wilhelm, S. et al, 2011 J. Med. Chem. 36, 5404).
  • the cell-binding agents are first modified with a bifunctional agent such as SPDP (N-succinimidyl 3-(2-pyridyldithio) propionate), or SMCC (succinimidyl-4-(N- maleimidomethyl)cyclohexane-l-carboxylate), or SPDB (N-succinimidyl 4-(2- pyridyldithio)butanoate), to introduce an active disulfide or a maleimido moiety.
  • a thiol-containing cytotoxic drug provides a conjugate in which the cell-binding agent, such as a monoclonal antibody, and drug are linked via disulfide bonds or thioether bonds
  • the use of the cell binding molecule-drug conjugates, such as antibody- drug conjugates (ADCs), in developing therapies for a wide variety of cancers has been limited both by the availability of specific targeting agents (carriers) as well as the conjugation methodologies which result in the formation of protein aggregates when the amount of the drugs that are conjugated to the carrier (i.e., the drug loading) is increased.
  • the tendency for cytotoxic drug conjugates to aggregate is especially problematic when the conjugation reactions are performed with the hydrophobic linkers.
  • higher drug loading increases the inherent potency of the conjugate, it is desirable to have as much drug loaded on the carrier as is consistent with retaining the affinity of the carrier protein.
  • aggregated protein which may be nonspecifically toxic and immunogenic, and therefore must be removed for therapeutic applications, makes the scale -up process for the production of these conjugates more difficult and decreases the yield of the products.
  • the present invention provides hydrophilic linkers containing phosphinate, sulfonyl, and/or sulfoxide groups to link drugs to a cell-binding agent (e.g., an antibody).
  • a cell-binding agent e.g., an antibody
  • the preferred formula of the cell binding molecule - hydrophilic linker- drug conjugates can be represented as: Cb-(-L-Drug) n , wherein Cb is a cell-binding agent, L is a hydrophilic linker, Drug is a drug molecule, and n is an integer from 1 to 20.
  • the advantages in applying the hydrophilic linker in the cell molecule-drug conjugate are: a), reducing the aggregation of the conjugates in water based media; b).
  • the hydrophilic linker is represented by formula (I) wherein Y can react with a cell-binding agent and Z can react with a cytotoxic drug: Wherein:
  • Y represents a functional group that enables reaction with a cell-binding agent
  • n and n are integer from 0 to 5, but not 0 at the same time;
  • Z represents a functional group that enables linkage of 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;
  • Ri, P 2 , P 3 , PM, P5 and P 6 are the same or different and are 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 (OCH 2 CH 2 ) p , wherein p is an integer from 0 to about 1000, or combination thereof.
  • R 2 , R 3 and R 4 are respectively a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl group, the conjugated drug and among themselves (Ri, R 2 , R 3 and R 4 ).
  • the atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxylamines, urethanes, amino acids, peptides,
  • M is H, or Na, or K, or N + RiR 2 R 3 or a pharmaceutical salt.
  • Ri, R 2 and R 3 are described above.
  • this invention provides a cell-binding agent-drug conjugate of formula (II), in which the cell-binding agent, Cb, and the drug, Drug, have reacted at the two end ( ⁇ )
  • Cb represents a cell-binding agent
  • Drug represents the drug linked to the cell-binding agent via the hydrophilic linker by a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
  • the present invention provides a modified cell-binding agent of formula (III), in which the cell-binding agent, Cb, has reacted with the hydrophilic linker, which s : (III)
  • the present invention provides a modified drug of formula (IV), in which the drug, Drug, has reacted with the hydrophilic linker, which still has Y, a group c : (IV)
  • the present invention further relates to a method of making a cell-binding molecule-drug conjugate of formula (II), wherein the drug is linked to a cell-binding agent via the hydrophilic linker.
  • the present invention also relates to a method of making a modified cell-binding molecule of formula (III), wherein the cell-binding molecule is reacted with the hydrophilic linker.
  • the present invention also relates to a method of making a modified drug of formula (IV), wherein the drug is reacted with the hydrophilic linker.
  • Figure 1 shows the synthesis of diphosphinate-containing cross-linking reagents that contain a pyridyldisulfide group and a reactive carboxylic acid ester, and the linker used for the conjugation of an antibody.
  • Ammonium phosphinate are first converted into bis(trimethylsilyl) phosphonite, followed by Michael addition with an acrylate and then substitution reaction with excess amount of 1,2-dibromo ethane to form (2-bromoethyl)(3- ethoxy-3-oxopropyl)phosphinic acid (4).
  • the bromoethyl phosphinic acid moiety (5) was then substituted with bis(trimethylsilyl) phosphonite (2) and 1 ,2-dibromo ethane to generate (2-bromoethyl)(2-(ethoxy(3-ethoxy-3-oxopropyl)phosphoryl)ethyl)phosphinic acid (8), whose bromide group was then replaced by potassium ethyl xanthogenate, followed by basic hydrolysis and a substitution reaction with an excess of 2,2'- dithiobispyridine, as well as condensation reaction of the acid 10 with N-hydroxysuccimide (NHS) in an acid medium using the carbodiimide coupling agent EDC to give the diphosphinate linker (11).
  • the linker 11 can be used for the conjugation of drugs to a cell binding molecule (such as antibody).
  • Figure 2 shows the synthesis of a triphosphinate-containing linker 18 that contains a pyridyldisulfide group and a reactive carboxylic acid ester.
  • the linker can be used for the conjugation of a drug to cell binding molecule via a disulfide bond.
  • Figures 3 shows the synthesis of tetraphosphinate-containing cross linkers 23 that contain a reactive carboxylic acid ester and a pyridyldisulfide group, enabling linkage of a drug to a cell binding molecule via a disulfide bond.
  • Figure 4 shows the synthesis of diphosphinate-containing cross-linker 26 that contain a maleimido group and a reactive carboxylic acid ester enabling linkage of a drug to a cell binding molecule via a thioether bond
  • Figures 5 shows the synthesis of monophosphinate, diphosphinate, triphosphinate and tetraphosphinate-containing linkers that contain a pyridyldisulfide group and a reactive carboxylic acid ester via substitution of ethyl 2-bromoacetate with bis(trimethylsilyl) phosphonite (2).
  • Figure 6 shows the synthesis of linking reagents that contain a vinylsulfonyl (55), a monosulfonyl (61) and disulfonyl (69) groups.
  • Figures 7 shows the synthesis of mono-, and di-sulfonyl linkers
  • Figure 8 shows the synthesis of mono-sulfonyl linkers containing polyethylene glycols.
  • Figure 9 shows the synthesis of di-sulfonyl linkers containing polyethylene glycols.
  • the linker can be used for the conjugation of an antibody or a protein via a disulfide bond.
  • Figure 10 shows the synthesis of a hydrophilic linker containing a phosphinic acid and a sulfonyl group.
  • Figure 11 shows the synthesis of a hydrophilic linker containing contain a phosphinate, a sulfonyl and a ketone groups that can be used for the conjugation of a drug to an antibody or a protein via a hydrazone bond.
  • Figure 12 shows the synthesis of hydrophilic linkers that contain a disulfonyl, polyethylene glycols, a much hindered pyridyldisulfide and a reactive carboxylic acid ester groups via substitution of l,3-dibromo-3-methylbutane.
  • the linker is used for the conjugation of drugs to an antibody or a protein via a hinder disulfide bond.
  • Figure 13 shows the synthesis of hydrophilic linkers containing a phosphinate and a sulfonyl groups that are used for the conjugation of drugs to a cell binding molecule either via a triazole or via a thiolether bond.
  • Figures 14 shows synthesis of hydrophilic linkers that contain a disulfonyl, phosphinate, a pyridyldisulfide group, a polyethylene glycol (PEG) chain and a reactive carboxylic acid ester.
  • the linkers are used for the conjugation of a cell binding molecule via cell thioether bond
  • Figures 15 shows synthesis of hydrophilic linkers that contain a disulfonyl, phosphinate, an alkoxylamino and a maleimido substituents, enabling ketone or aldehyde- containing drug to link to an antibody via a thioether and an alkoxime bond.
  • Figure 16 shows the synthesis of disulfonyl-containing linkers that contains an alkoxylamino and a maleimido substituent, enabling ketone or aldehyde- containing drug to link to an antibody via a thioether and an alkoxime bond.
  • Figure 17 shows the synthesis of sulfoxide-containing and disulf oxide-containing linkers that are used for the conjugation of drugs to a cell binding molecule via a disulfide bond.
  • Figures 18 show the synthesis of a disulfonyl-containing linker (221) that used for conjugation of amine-containing cytotoxic drugs to an antibody via the Val-Cit-PABC linkage.
  • Figure 19 (19-a ⁇ 19-z) shows the antibody-drug conjugate (ADC) structures of the typical cytotoxic agents (the analogs of tubulysins, calicheamicins, maytansinoids, auristatins, doxorubicin, daunorubicin, CC-1065, pyrrolobenzodiazepine dimmers) via the hydrophilic linkers of this patent.
  • ADC antibody-drug conjugate
  • Figure 20 shows the use of a hydrophilic linker (86) in modifying a cell-binding agent (antiHer2 antibody) and following by production of a cell-binding agent-drug (TZ03) conjugate containing the hydrophilic linker.
  • Figure 21 shows 5 days in vito assays of the cytotoxicity of the antiCD22-TZ041 (tubulysin analog) conjugate with different drug load ratios via a hydrophilic linker (86).
  • Alkyl means an aliphatic hydrocarbon group which may be straight or branched having 1 to 8 carbon atoms in the chain or cyclic. "Branched” means that one or much lower alkyl groups such as methyl, ethyl or propyl are attached to a linear alkyl chain.
  • Exemplary alkyl groups include methyl, ethyl, n-propyl, /-propyl, «-butyl, Z-butyl, «-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-methylhexyl, 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 d-Cs alkyl group can be unsubstituted or substituted with one or more groups including, but not limited to, -Ci ⁇ C 8 alkyl, -0-(Ci ⁇ C 8 alkyl), - aryl, -C(0)R', -OC(0)R', -C(0)OR', -C(0)NH 2 , -C(0)NHR', -C(0)N(R') 2 -NHC(0)R', - S(0) 2 R', -S(0)R', -OH, -halogen (F, CI, Br or I), -N 3 , -NH 2 , -NH(R'), -N(R') 2 and -CN; where each R' is independently selected from -Ci ⁇ Cs alkyl and aryl.
  • C3 ⁇ Cs carbocycle means a 3-, 4-, 5-, 6-, 7- or 8-membered saturated or unsaturated non-aromatic carbocyclic ring.
  • Representative C3 ⁇ Cs 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 ⁇ Cs carbocycle group can be unsubstituted or substituted with one or more groups including, but not limited to, -Ci ⁇ C 8 alkyl, -0-( Ci ⁇ C 8 alkyl), -aryl, -C(0)R', -OC(0)R', -C(0)OR', - C(0)NH 2 , -C(0)NHR', -C(0)N(R') 2 -NHC(0)R', -S(0) 2 R', -S(0)R', -OH, -halogen, -N 3 , - NH 2 , -NH(R'), -N(R') 2 and -CN; where each R' is independently selected from -Ci ⁇ C 8 alkyl and aryl.
  • Cs-Cs carbocyclo refers to a C3 ⁇ Cs carbocycle group defined above wherein one of hydrogen atoms on the carbocycle is replaced with a bond.
  • Heterocycle refers to an aromatic or non-aromatic C3 ⁇ Ci 4 carbocycle in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group of O, N, S Se, and P.
  • Preferable heteroatoms are oxygen, nitrogen and sulphur.
  • Suitable heterocyclics are also disclosed in The Handbook of Chemistry and Physics, 76 th Edition, CRC Press, Inc., 1995-1996, p. 2-25 to 2-26, the disclosure of which is hereby incorporated by reference.
  • Preferred non aromatic heterocyclic include, but are not limited to pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxiranyl, tetrahydrofuranyl, dioxolanyl, tetrahydro-pyranyl, dioxanyl, dioxolanyl, piperidyl, piperazinyl, morpholinyl, pyranyl, imidazolinyl, pyrrolinyl, pyrazolinyl, thiazolidinyl, tetrahydrothiopyranyl, dithianyl, thiomorpholinyl, dihydro- pyranyl, tetrahydropyranyl, dihydropyranyl, tetrahydro-pyridyl, dihydropyridyl, tetrahydropyrinidinyl, dihydrothiopyranyl, azepanyl, as well as the fused systems resulting from the condensation
  • Alkyl refers also to the corresponding "alkylene”, “cycloalkylene”, “alkenylene”,
  • alkynylene "arylene”, “heteroarylene” , “heterocyclene” and the likes which are formed by the removal of two hydrogen atoms.
  • Halogen atom refers to fluorine, chlorine, bromine or iodine atom; preferably bromine and chlorine atom.
  • “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 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.
  • preserving or antioxidant agents 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 combinations.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • the pharmaceutically 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.
  • 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, glucoronic, 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.
  • 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.
  • 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.
  • non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17 th ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
  • novel conjugates disclosed herein use hydrophilic cross-linkers. Examples of some suitable cross-linkers and their synthesis are shown in Figures 1 to 18.
  • hydrophilic cross linkers possess three elements: a) substituents that are either phosphinate, or sulfonyl or sulfoxide groups, or mixed of these groups, b) a group, such as a N-hydroxysuccimimide ester, maleimido group, haloacetyl group, and hydrazide, capable of reaction with a cell-binding agent, and c) a group, such as but not limited to, a disulfide, maleimide, haloacetyl, aldehyde, ketone, azide, amine, alkoxylamino and hydrazide, capable of reaction with a drug.
  • the hydrophilic substituents can be introduced by methods described herein.
  • the phosphinate substituents they can be introduced by first treating a commercially available ammonium phosphinate with an acrylate via Michael addition and followed by substitution of excess amount of dibromo alkane to a phosphinate group.
  • the sulfonyl and sulfoxide substituents they can be introduced by first generation of thioether components, followed by oxidization of these components. More detail synthesis of the hydrophilic linkers and their uses for the preparation of cell binding ligand-drug conjugates of this invention are disclosed in the figures 1-19.
  • Y represents a functional group that enables reaction with a cell-binding agent
  • n and n are integer from 0 to 5, but not 0 at the same time;
  • Z represents a functional group that enables linkage of 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;
  • Ri, P2, P3, P4, P5 and P6 are the same or different and are 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 (OCH 2 CH 2 ) p , wherein p is an integer from 0 to about 1000, or combination thereof.
  • Ri, R 2 , R3, and R4 can be respectively a chain of atoms selected from C, ⁇ , O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl or sulfoxide group, the conjugated drug and themselves (Ri, R 2, R 3 and R 4 ).
  • the atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming 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.
  • 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.
  • M is H, or Na, or K, or N + RiR 2 R 3 or a pharmaceutical salt.
  • Ri, R 2 and R 3 are described above.
  • Examples of the functional group, Y, that enables reaction with a cell-binding agent include amine reacting agents such as but not limited to N-hydroxysuccinmide esters, p- nitrophenyl esters, dinitrophenyl esters, pentafluorophenyl esters; thiol reactive agents such as but not limited to pyridyldisulfides, nitropyridyldisulfides, maleimides, haloacetates and carboxylic acid chlorides.
  • amine reacting agents such as but not limited to N-hydroxysuccinmide esters, p- nitrophenyl esters, dinitrophenyl esters, pentafluorophenyl esters
  • thiol reactive agents such as but not limited to pyridyldisulfides, nitropyridyldisulfides, maleimides, haloacetates and carboxylic acid chlorides.
  • Examples of the functional group, Z, which enables linkage of a cytotoxic drug include groups that enable linkage via an either disulfide, or thioether, thioester, peptide, hydrazone, ester, carbamate, carbanate, alkoxime or amide bond.
  • Such functional groups include, but are not limited to, thiol, disulfide, amino, carboxy, aldehydes, maleimido, haloacetyl, hydrazines, and hydroxy.
  • Huisgen 1,3-dipolar cycloaddition of azides to alkynes is shown, for example, in Figures 13.
  • the synthesis of hydrophilic cross linkers of formula (I) bearing a hydrazide or ketone moieties enabling linkage via acid-labile bonds is shown, for example, in Figures 11, 16and 18.
  • the synthesis of hydrophilic cross linkers of formula (I) bearing an alkoxylamino moiety enabling linkage via alkoxime bonds is shown, for example, in Figure 15 andl6.
  • the synthesis of dipeptide -containing cross linkers of the formula (I) is shown, for example, in Figure 18.
  • the conjugates of the present invention can be represented by the following formula, Cb-(-L-Drug) n , wherein Cb is a cell-binding agent, L is a hydrophilic linker, Drug is a drug molecule, and n is an integer from 1 to 20.
  • the hydrophilic 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
  • SPDB 4-thio-2-hydroxysulfonyl-butyrate
  • EO ethyleneoxy -CH 2 CH 2 0— as one or more repeating units
  • PEO repeating units
  • linkers are: -maleimidocaproyl containing
  • Cb represents a cell-binding agent
  • Drug represents the drug linked to the cell-binding agent via the hydrophilic linkers of this invention by a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, heterocyclic ring, amine, imine, alkoxime or amide bond; q is 1 ⁇ 20; Q, T, m, n, Ri, R 2 , R3, R4, R5 , R6 and M are described the same previously in formula (I).
  • the drug can be any of many small molecule 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).
  • PBD pyrrolobenzodiazepine
  • indolinobenzodiazepines imidazobenzothiadiazepines
  • oxazolidinobenzodiazepines oxazolidinobenzodiazepines
  • the cell-binding agent can be first modified with the hydrophilic linkers of the present invention to introduce reactive disulfide groups, maleimido, haloacetyl, azide, 1-yne, ketone, or hydrazide groups.
  • 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 containing 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 cell-binding agent and a drug containing a free thiol group.
  • Synthesis of conjugates bearing an acid labile hydrazone link 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).
  • the drug can be modified with the hydrophilic linkers of the present invention to give a modified drug of formula (IV) bearing functionality capable of reacting with a cell binding agent.
  • a thiol-containing drug can be reacted with the hydrophilic linker of formula (I) bearing a maleimdo or a haloacetyl or an ethylsulfonyl substituent at neutral pH in aqueous buffer to give a drug connected to the hydrophilic linker via a thioether link.
  • a thiol-containing drug can undergo disulfide exchange with a hydrophilic linker bearing a pyrdiyldithio moiety to give a modified drug attached via a disulfide bond to the hydrophilic cross linker.
  • a drug bearing a hydroxyl group or a thiol group can be reacted with a hydrophilic linker bearing a halogen of this invention, in the presence of a mild base, to give a modified drug bearing an ether or thiol ether link.
  • a hydroxyl group containing drug can be condensed with a hydrophilic cross linker of formula (I) bearing a carboxyl group, in the presence of a dehydrating agent, such as EDC or dicyclohexylcarbodimide, to give an ester link.
  • a dehydrating agent such as EDC or dicyclohexylcarbodimide
  • An amino group containing drug can similarly undergo condensation with a carboxyl group on the hydrophilic linker of formula (I) to give an amide bond.
  • 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 exchange or by dialysis.
  • standard biochemical means such as gel filtration on a Sephadex G25 or Sephacryl S300 column, adsorption chromatography, and ion exchange or by dialysis.
  • the cell-binding agent-drug conjugates can be purified by chromatography such as by HPLC, medium pressure column chromatography or ion exchange chromatography.
  • the cell-binding agent modified by reaction with linkers of the present invention are:
  • substituents are as described above for the hydrophilic linkers and the cell-binding agent drug conjugates.
  • Z is a disulfide substituent, a maleimido, haloacetyl group, or an N-hydroxysuccinimide ester
  • Cb linked with Ri is through thioether, amide, or disulfide bond.
  • the modified cell-binding agent can be prepared via a reaction of the cell-binding agent with the hydrophilic linkers by methods known in the art for other cross-linkers (U.S. Patent Nos. 5,846,545, 5,585,499, 5,475,092, 5,414,064, 5,208,020, and 4,563,304; Carlsson, J. et al. Biochem. J. (1978) 173, 723-737(1978); Goff, D. A.,
  • the reaction between the cell-binding agent and the cross-linker can be conducted in aqueous solution.
  • the cross-linking reagent is dissolved in aqueous buffer, optionally containing a small amount (typically ⁇ 10% by volume) of 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 dimethylsulf oxide (DMSO) at a high concentration, for example 1-100 mM, and then an appropriate aliquot is added to the buffered aqueous solution of the cell-binding agent.
  • An appropriate aliquot is an amount of solution that introduces 1-10 cross-linking groups per cell-binding agent, preferably 1-5 groups, and the volume to be added should not exceed 10 %, preferably 5 %, and most preferably 0-3 % of the volume of the cell-binding agent solution.
  • the aqueous solutions for the cell-binding agents are buffered between 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, NaCl and KC1.
  • the reaction is incubated at a temperature of from 4 °C to 40 °C, preferably at ambient temperature.
  • the progress of the reaction can be monitored by measuring the increase in the absorption at 320 nm or another appropriate wavelength.
  • isolation of the modified cell- binding agent can be performed in a routine way, using for example gel filtration chromatography, or adsorptive chromatography.
  • Figure 20 shows the results from the modification of the cell-binding agent, the her2 antibody, with a chydrophilic cross-linker of the present invention.
  • the time course of linker/antibody (L/A) incorporation is shown, for example, along with the drugs/antibody (D/A) linked.
  • the hydrophilic cross-linkers described herein have diverse functional groups that can react with any cell-binding agent that possesses a suitable substituent.
  • cell-binding agents bearing an amino or hydroxyl substituent can react with cross linkers bearing an N-hydroxysuccinimide (NHS) ester
  • cell-binding agents bearing a thiol substituent can react with cross linkers bearing a maleimido or haloacetyl group
  • cell- binding agents bearing a carbonyl substituent can react with cross linkers bearing a hydrazide or a hydroxylamine.
  • cytotoxic drugs modified by reaction with cross-linkers of the present invention are (IV)
  • Y is a disulfide substituent, a maleimido, haloacetyl group, or an N-hydroxysuccinimide ester.
  • the modified drugs can be prepared by reacting the drug with the cross linkers of the present invention to give a modified drug of formula (IV) bearing a functionality capable of reacting with a cell binding agent.
  • a thiol-containing drug can be reacted with the cross linker of formula (I) bearing a maleimdo substituent at neutral pH in aqueous buffer to give a drug connected to the hydrophilic linker via thioether linkage.
  • a thiol-containing drug can undergo disulfide exchange with a hydrophilic linker bearing a pyrdiyldithio moiety to give a modified drug attached via a disulfide bond to the hydrophilic cross linker.
  • a drug bearing a hydroxyl group can be reacted with a cross linker bearing a halogen, in the presence of a mild base, to give a modified drug bearing an ether link.
  • a hydroxyl group containing drug can be condensed with a cross linker of formula (I) bearing a carboxyl group, in the presence of a dehydrating agent, such as EDC or dicyclohexylcarbodimide (DCC), to give an ester link.
  • a drug bearing a thiol group can be reacted with a cross linker bearing a malimido or a vinylsulfonyl, or a haloacetyl group, to give a modified drug bearing thioether link.
  • An amino group containing drug can similarly undergo condensation with a carboxyl group on the hydrophilic cross linker of formula (I) to give an amide bond.
  • the modified drug can be purified by standard methods such as column chromatography over silica gel or alumina, crystallization, preparatory thin layer chromatography, ion exchange chromatography or HPLC.
  • 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 , F Vj [Parham, J.
  • Immunol. 131, 2895-2902 (1983)] fragments produced by a Fab expression library, anti- idiotypic (anti-Id) antibodies, CDR's, 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.
  • interferons such as type I, II, III
  • peptides such as lymphokines such as IL-2, IL-3, IL-4, IL-6, GM-CSF, interferon-gamma (IFN- ⁇ ); hormones such as insulin, TRH (thyrotropin releasing hormones), MSH (melanocyte-stimulating hormone), steroid hormones, such as androgens and estrogens, melanocyte-stimulating hormone
  • MSH growth factors and colony-stimulating factors
  • EGF epidermal growth factors
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • TGF transforming growth factors
  • IGF-I, IGF-II insulin and insulin like growth factors
  • GEF vaccinia growth factors
  • FGFs fibroblast growth factors
  • platelet-derived growth factors 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; apo
  • PSMA prostate-specific membrane antigen
  • TKI small molecular tyrosine kinase inhibitors
  • nanoparticles Liong, et al, ACS Nano, 2008, 19, 1309-12; Medarova, 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).
  • monoclonal antibodies are preferred as a cell-surface binding agent if an appropriate one is available.
  • antibodies may be murine, human, humanized, chimeric, or derived from other species.
  • Particularly monoclonal antibodies 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 antibody 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 maintained 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.
  • DMEM Dulbecco's minimal essential medium
  • antibody-producing cell lines can also be created by techniques other than fusion, such as direct transformation of B lymphocytes with oncogenic DNA, or transfection with an oncovirus, such as Epstein-Barr virus (EBV, also called human herpesvirus 4 (HHV-4)) or Kaposi's sarcoma-associated herpesvirus (KSHV).
  • EBV Epstein-Barr virus
  • HHV-4 human herpesvirus 4
  • KSHV Kaposi's sarcoma-associated herpesvirus
  • 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).
  • the anti-receptor peptide or a peptide analog is used either alone or conjugated to an immunogenic carrier, as the immunogen for producing anti-receptor peptide monoclonal antibodies.
  • 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.
  • 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.
  • 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.
  • mice examples include the Xenomouse. (Abgenix, Inc.), the HuMAb-Mouse
  • Antibodies immunospecific for a malignant cell antigen can also be obtained commercially 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 antibodies 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.
  • 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 techniques as for phage display antibodies (Szardenings, J Recept Signal Transduct Res.
  • 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, as long as such attachment permits the peptide or protein to retain its antigen binding specificity.
  • antibodies used for conjugation of drugs via the hydrophilic linkers of this prevention for treating cancer, autoimmune disease, and infectious disease include, but are not limited to, 3F8 (anti-GD2), Abagovomab (anti CA-125), Abciximab (anti CD41 (integrin alpha-lib), 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-CCLl l (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), Brentuxima
  • Clenoliximab (anti-CD4), Clivatuzumab (anti-MUCl), Conatumumab (anti-TR AIL-R2) , CR6261 (anti-Influenza A hemagglutinin), Dacetuzumab (anti-CD40), Daclizumab (Zenapax, anti-CD25 (a chain of IL-2 receptor)), Daratumumab (anti-CD38 (cyclic ADP ribose hydrolase), Denosumab (Prolia, anti-RANKL), Detumomab (anti-B -lymphoma cell), Dorlimomab, Dorlixizumab, Ecromeximab (anti-GD3 ganglioside), Eculizumab (Soliris, anti-C5), Edobacomab (anti-endo toxin), Edrecolomab (Panorex, MA 7-1A, anti-
  • EpCAM EpCAM
  • Efalizumab Raptiva, anti-LFA-1 (CDl la), 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 ⁇ ⁇ 3), Exbivirumab ( anti-hepatitis B surface antigen), Fanolesomab (NeutroSpec, anti-CD15), Faralimomab (anti-interferon receptor), Farletuzumab (anti-folate receptor 1), Felvizumab (
  • Girentuximab anti-carbonic anhydrase 9
  • Glembatumumab CR011, anti-GPNMB
  • Golimumab (Simponi, anti-TNF-a), Gomiliximab (anti-CD23 (IgE receptor)), Ibalizumab (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 receptor)),
  • Inotuzumab (anti-CD22), Ipilimumab (anti-CD152), Iratumumab (anti- CD30
  • TNFRSF8 Keliximab
  • CEA-Cide Labetuzumab
  • Lebrikizumab Lebrikizumab
  • 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- CD23 (IgE receptor
  • Mapatumumab anti-TRAIL-Rl
  • Maslimomab anti- T-cell receptor
  • Matuzumab anti-EGFR
  • Mepolizumab Bosatria, anti-IL-5
  • Metelimumab anti-TGF beta 1
  • Milatuzumab anti-CD74
  • Minretumomab anti-TAG
  • Ticilimumab (Tremelimumab, (anti-CTLA-4), Tigatuzumab (anti-TRAIL-R2), TNX-650 (anti-IL-13), Tocilizumab (Atlizumab, Actemra, RoActemra, (anti-IL-6 receptor), Toralizumab (anti-CD 154 (CD40L)), 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), Ustekinumab (Stelara, anti-IL-12, IL-23), Vapaliximab (anti-AOC3 (VAP-1)),
  • Vedolizumab (anti-integrin ⁇ 4 ⁇ ), Veltuzumab (anti-CD20), Vepalimomab (anti-AOC3 (VAP-1), Visilizumab (Nuvion, anti-CD3), Vitaxin (anti-vascular integrin avb3),
  • Volociximab anti-integrin ⁇ ⁇
  • Votumumab HuaSPECT, anti-tumor antigen
  • CTAA16.88 Zalutumumab (HuMax-EGFr, (anti-EGFR), Zanolimumab (HuMax-CD4, anti-CD4), Ziralimumab (anti-CD147 (basigin)), Zolimomab (anti-CD5), Etanercept
  • CYT- 356 Oncoltad®, for prostate cancers
  • HNK20 OraVax Inc. for respiratory syncytial virus
  • ImmuRAIT from Immunomedics for NHL
  • Lym-1 anti-HLA-DRlO, Peregrine Pharm.
  • MAK-195F anti-TNF (tumor necrosis factor; TNFA, TNF-alpha; TNFSF2), from Abbott / Knoll for Sepsis toxic shock
  • MEDI-500 [T10B9, anti-CD3, TRo$ (T cell receptor alpha/beta), complex, from Medlmmune Inc for Graft-versus-host disease]
  • RING SCAN anti-TAG 72 (tumour associated glycoprotein 72), from Neoprobe Corp.
  • LymphoCide Immunomedics, NJ
  • Smart ID10 Protein Design Labs
  • Oncolym Techniclone Inc, CA
  • Allomune BioTransplant, CA
  • anti-VEGF Geneentech, CA
  • CEAcide Immunomedics, NJ
  • IMC-1C11 ImClone Systems, NJ
  • Cetuximab ImClone, NJ
  • antibodies as binding 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
  • CD2 Hodgkin's disease, NHL lymphoma, multiple myeloma
  • CD3 epsilon T cell lymphoma, lung, breast, gastric, ovarian cancers, autoimmune diseases, malignant ascites
  • CD19 B cell malignancies
  • CD20 non-Hodgkin's lymphoma
  • CD22 leukemia, lymphoma, multiple myeloma, SLE
  • CD30 Hodgkin's lymphoma
  • CD33 leukemia, autoimmune diseases
  • CD38 multiple myeloma
  • CD40 lymphoma, multiple myeloma, leukemia (CLL)
  • CD51 Metalstatic melanoma, sarcoma
  • CD52 leukemia
  • CEACAM5 carcinoembryonic antigen; CEA, CD66e
  • CEA CD66e
  • DLL4 A-like-4
  • EGFR Epidermal Growth Factor Receptor, various cancers
  • CTLA4 melanoma
  • CXCR4 CD 184, Heme-oncology, solid tumors
  • Endoglin CD 105, solid tumors
  • EPCAM epidermal 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 farnesol receptor
  • GD2 ganglioside cancers
  • G-28 a cell surface antigen glyvolipid, melanoma
  • GD3 idiotype cancers
  • Heat shock proteins cancers
  • HER1 lung, stomach cancers
  • HER2 breast2
  • HLA-DR10 HLA-DRB
  • HLA-DRB HLA-DRB
  • human chorionic gonadotropin carcinoma
  • IGF1R insulin-like growth factor 1 receptor, solid tumors, blood cancers
  • IL-6R interleukin 6 receptor, multiple myeloma, RA, Castleman's disease, IL6 dependent tumors
  • Integrins ⁇ 3, ⁇ 5 ⁇ 1, ⁇ 6 ⁇ 4, ⁇ 11 ⁇ 3, ⁇ 5 ⁇ 5, ⁇ 5, 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
  • CEA colonrectal
  • gplOO melanoma
  • MARTI melanoma
  • the cell-binding -drug conjugates via the hydrophilic likers of this invention are used for the treatment of cancers.
  • the 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 (
  • the cell-binding-drug conjugates via the hydrophilic likers of this invention are used in accordance with the compositions and methods for the treatment 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,
  • 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 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 inflammatory 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, Complement component 2 deficiency, Cranial arteritis, CREST syndrome, Crohns Disease (a type of idiopathic inflammatory bowel diseases), Cushing's Syndrome, Cutaneous leukocytoclastic angiitis, Dego'
  • Parsonnage-Turner syndrome Pars planitis, Pemphigus, Pemphigus vulgaris, Pernicious anaemia, Perivenous encephalomyelitis, POEMS syndrome, Polyarteritis nodosa,
  • Polymyalgia rheumatica Polymyositis, Primary biliary cirrhosis, Primary sclerosing cholangitis, Progressive inflammatory neuropathy, Psoriasis, Psoriatic Arthritis, Pyoderma gangrenosum, Pure red cell aplasia, Rasmussen's encephalitis, Raynaud phenomenon, Relapsing polychondritis, Reiter's syndrome, 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
  • a binding molecule used for the conjugate via the hydrophilic linkers of this invention for the treatment or prevention of an autoimmune disease includes, 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-U.sub.lRNP; 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
  • the binding molecule for the conjugate in the present invention can bind to both a receptor or a receptor complex expressed on an activated 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, CD37, CD38, CD56, CD70, CD79, CD90, CD 125, CD152/CTLA-4, PD-1, or ICOS), a TNF receptor superfamily member (e.g.
  • useful binding ligands that are immunospecific for a viral or a microbial antigen are humanized or human monoclonal antibodies.
  • viral antigen includes, but is not limited to, any viral peptide, polypeptide protein (e.g. HIV gpl20, HIV nef, RSV F glycoprotein, influenza virus neuramimidase, 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.
  • polypeptide protein e.g. HIV gpl20, HIV nef, RSV F glycoprotein
  • influenza virus neuramimidase influenza virus hemagglutinin
  • HTLV tax herpes simplex virus glycoprotein
  • herpes simplex virus glycoprotein e.g. gB, gC, gD, and gE
  • microbial antigen includes, but is not limited to, any microbial peptide, polypeptide, protein, saccharide, polysaccharide, or lipid molecule (e.g., a bacterial, fungi, pathogenic protozoa, or yeast polypeptide including, e.g., LPS and capsular polysaccharide 5/8) that is capable of eliciting an immune response.
  • microbial antigen includes, but is not limited to, any microbial peptide, polypeptide, protein, saccharide, polysaccharide, or lipid molecule (e.g., a bacterial, fungi, pathogenic protozoa, or yeast polypeptide including, e.g., LPS and capsular polysaccharide 5/8) that is capable of eliciting an immune response.
  • 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 IgG.sub.l antibody for the treatment of cytomegalovirus; and anti-LPS antibodies.
  • the cell binding molecules-drug conjugates via the hydrophilic linkers of this invention can be used in the treatment of infectious diseases.
  • infectious diseases include, but are not limited to, Acinetobacter infections, Actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (Acquired immune deficiency syndrome), Amebiasis, Anaplasmosis, Anthrax, Arcanobacterium haemolyticum infection, Argentine hemorrhagic fever, Ascariasis, Aspergillosis, Astrovirus infection, Babesiosis, Bacillus cereus infection, Bacterial pneumonia, Bacterial vaginosis, Bacteroides infection,
  • Metagonimiasis Microsporidiosis, Molluscum contagiosum, Mumps, Murine typhus
  • Tinea cruris Jock itch
  • Tinea manuum Tinea manuum
  • Tinea nigra Tinea pedis
  • Tinea unguium Onychomycosis
  • Tinea versicolor Tinea 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 pseudotuberculosis infection, Yersiniosis, Yellow fever, Zygomycosis.
  • the cell binding molecules which are more proffered to be antibodies 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, Bacillus 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,
  • Enterovirus 71 Sin Nombre virus, Helicobacter 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
  • Orthomyxoviridae 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 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,
  • 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 wasneckii, 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 (Entomophthoramycosis), Pseudomonas aeruginosa, Campylobacter (Vibrio) fetus, Aeromonas hydrophila, Edwardsiella tarda, Yersinia pestis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Salmonella typhimurium, Treponema per pneumonia, Treponema carateneum, Borrelia vincentii, Borrelia burgdorferi, Leptospira icterohemorrhagiae, P
  • Trypanosoma rhodesiense Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Pneumocystis pneumonia, Plasmodium vivax, Plasmodium falciparum, Plasmodium malaria); or Helminiths (Schistosoma japonicum, Schistosoma mansoni, Schistosoma haematobium, and hookworms).
  • 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, Picornaviridae, 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 (Nas
  • Central nervous system virus [such as, JCV (Progressive multifocal leukoencephalopathy), MeV (Subacute sclerosing panencephalitis), LCV (Lymphocytic choriomeningitis), Arbovirus encephalitis, Orthomyxoviridae (probable) (Encephalitis lethargica), RV (Rabies), Chandipura virus, Herpesviral meningitis, Ramsay Hunt syndrome type II;
  • Poliovirus Poliovirus (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 mononucleosis), 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 (Cytomegal
  • the present invention also concerns pharmaceutical compositions comprising the conjugate via the hydrophilic linkers of the invention together with a pharmaceutically acceptable carrier for treatment of cancer and autoimmune disorders.
  • the method for treatment of cancer and autoimmune disorders can be practiced in vitro, in vivo, or ex vivo.
  • 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.
  • Examples 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 malignant cells.
  • HSC hematopoietic stem cells
  • the bone marrow cells After incubation the bone marrow cells are washed with medium containing serum and returned to the patient by i.v. 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.
  • the conjugate via the linkers of the invention will be supplied as solutions or as a lyophilized solid that can be redissolved in sterile water for injection.
  • suitable protocols of conjugate administration are as follows.
  • Conjugates are given weekly for 8 weeks as an i.v. bolus.
  • Bolus doses are given in 50 to
  • human serum albumin e.g. 0.5 to 1 mL of a concentrated solution of human serum albumin, 100 mg/mL
  • Dosages will be about 50 ⁇ g to 20 mg/kg of body weight per week, i.v. (range of 10 ⁇ g to 200 mg/kg per injection). 8 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
  • 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 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 compound 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 administered 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.
  • typical total daily dose ranges are from 0.01 to 100 mg/kg of body weight.
  • unit doses for humans range from 1 mg to 3000 mg per day.
  • the unit dose range is from 1 to 500 mg administered one to four times a day, and even more preferably from 10 mg to 500 mg, once a day.
  • 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 intranasally, 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 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 WO05058367A2, 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,
  • PBD pyrrolobenzodiazepine
  • Nitrosoureas (carmustine, lomustine, chlorozotocin, fotemustine, nimustine, ranimustine); Alkylsulphonates: (busulfan, treosulfan, improsulfan and piposulfan); Triazenes:
  • 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
  • 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).
  • 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:
  • Hormonal therapies such as ⁇ Receptor antagonists: [Anti-estrogen: (megestrol, raloxifene, tamoxifen); LHRH agonists: (goserelin, leuprolide acetate); Anti- androgens: (bicalutamide, flutamide, calusterone, dromostanolone propionate, 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, Inter
  • 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 . ⁇ , ⁇ , l and ⁇ , see, e.g., J. Med. Chem. , 39 (11), 2103-2117 (1996), Angew Chem Intl. Ed. Engl.
  • 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).
  • 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.
  • An anti-autoimmune disease agent includes, but is not limited to, 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.
  • An anti-infectious disease agent includes, but is not limited to, a).
  • Aminoglycosides amikacin, astromicin, gentamicin (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
  • Glycopeptides bleomycin, vancomycin (oritavancin, telavancin), teicoplanin (dalbavancin), ramoplanin; g).
  • Glycylcyclines e. g. tigecycline; g).
  • ⁇ - 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; 1).
  • Monobactams aztreonam, tigemonam; m).
  • Oxazolidinones linezolid; n).
  • Penicillins amoxicillin, ampicillin (pivampiciUin, 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
  • 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, chlortetracycline, clomocycline, demeclocycline, lymecycline, meclocycline, metacycline, minocycline, oxytetracycline, penimepicycline, rolitetracycline, tetracycline, glycylcyclines (e.g. tigecycline); u).
  • antibiotics include 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,
  • Bacitracin Bactoprenol inhibitors
  • DADAL/AR inhibitors cycloserine
  • dictyostatin discodermolide
  • eleutherobin epothilone
  • ethambutol etoposide
  • faropenem fusidic acid
  • furazolidone
  • quinupristin/dalfopristin quinupristin/dalfopristin, rifampicin (rifampin), tazobactam tinidazole, uvaricin;
  • 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, becon; d).
  • Neuraminidase inhibitors oseltamivir, zanamivir, peramivir; e). Nucleosides &
  • nucleotides abacavir, aciclovir, adefovir, amdoxovir, apricitabine, brivudine, cidofovir, clevudine, dexelvucitabine, didanosine (ddl), elvucitabine, emtricitabine (FTC), entecavir, famciclovir, fluorouracil (5-FU), 3'-fluoro-substituted 2', 3 '-dideoxynucleoside analogues (e.g.
  • M-thymidine and M-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 etravirine, rilpivirine, delavirdine, docosanol, emivirine, efavirenz, foscarnet (phosphonoformic acid), imiquimod, interferon alfa, loviride, lodenosine, methisazone, nev
  • 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 are 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 hydrophilic linker of the present invention also include radioisotopes. Examples of radioisotopes (radionuclides) are 3 H, U C, 14 C, 18 F,
  • Radioisotope 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 hydrophilic 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, DTP A and TETA (Macrocyclics, Dallas, Tex.).
  • indolinobenzodiazepines imidazobenzothiadiazepines, or oxazolidinobenzodiazepines
  • calicheamicins and the enediyne antibiotics, actinomycin, azaserines, bleomycins, epirubicin, tamoxifen, idarubicin, dolastatins/auristatins (e.g. monomethyl auristatin E,
  • MMAE MMAE
  • MMAF auristatin PYE
  • auristatin TP Auristatins 2-AQ, 6-AQ, EB (AEB), and EFP (AEFP)
  • duocarmycins thiotepa
  • vincristine hemiasterlins
  • esperamicins and their analogues and derivatives 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. 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.
  • tubulysins for conjugation of cell binding molecules are described in the patent application of
  • Maytansinoids that are preferred to be used in the present invention including maytansinol and maytansinol analogues are described in U.S. Patent Nos. 4,256,746, 4,361,650 and 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, 8,163,888 .
  • Taxanes which includes Paclitaxel (Taxol), a cytotoxic natural product, and docetaxel (Taxotere), a semi-synthetic derivative, and their analogs which are preferred for conjugation via the hydrophilic linkers of the present patent are exampled in:. K C.
  • CC-1065 analogues and doucarmycin analogs are also preferred to be used for a conjugate with the hydrophilic linkers of the present patent.
  • the examples of the CC-1065 analogues and doucarmycin analogs as well as their synthesis are described in:
  • Daunorubicin/Doxorubicin Analogues are also preferred for conjugation via the hydrophilic linkers of the present patent.
  • the preferred structures and their synthesis are exampled 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.
  • Auristatins and dolastatins are preferred in conjugation via the hydrophilic 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
  • AE auristain E
  • AEB auristatin EFP
  • MMAE monomethyl auristatin E
  • MMAF Monomethylauristatin
  • AFP Auristatin F phenylene diamine
  • AFP phenylalanine variant of MMAE
  • 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;
  • Analogues and derivatives of the cytotoxic drugs/agents described in the present patent can be conjugated via a hydrophilic linker of the present patent.
  • 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.
  • the drugs/cytotoxic agents of the present invention include analogues and derivatives of the compounds described herein.
  • hexamethydisilazane (20.0 mL, 96 mmol) was heated at 120 °C for 1 h under argon. After the mixture was cooled to 0 °C, ethyl acrylate (10.4 mL, 96 mmol) was carefully added dropwise, and the resulting mixture was stirred at 50 °C for 2 h. Then the mixture was cooled to room temperature, dibromoethane (40.0 mL) was added, and the mixture was stirred for 5 h at 120 °C. The formed trimethylbromosilane and excess dibromoethane were removed under vacuum.
  • Example 9 Ethyl 3-((2-((2-(3, 6-endoxo-A- tetrahydrophthalido)ethyl)(ethoxy)phosphoryl)ethyl)(ethoxy)phosphoryl)propanoate, or ethyl 3-((2-((2-((3aR,4R,7S)-l,3-dioxo-3a,4,7,7a-tetrahydro-lH-4,7-epoxyisoindol-2(3H)- yl)ethyl)(ethoxy)phosphoryl)ethyl)(ethoxy)phosphoryl)propanoate (24)
  • Example 10 3-((2-((2-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l- yl)eth droxy)phosphoryl)propanoic acid (25)
  • Compound 24 (2.60 g, 5.14 mmol)
  • toluene (20 ml) and HC1 (8N,10 ml) was heated at 120 ⁇ 140°C for 8 h.
  • 5 x 10 ml of water was gradually added to keep the reaction volume around 40 ml.
  • Methyl 3-((2-bromoethyl)thio)propanoate 52 (4.50 g, 19.91 mmol) in acetic acid (40 ml), H 2 0 2 (30%, 20 ml) and KMn0 4 (l.OOg, 6.33 mmol) was stirred overnight. The mixture was concentrated, diluted with EtOAc (100 ml) and 1 M NaH 2 P0 4 (150 ml), separated, the aqueous layer was extracted with EtOAc (2 x 100 ml).
  • Example 21 2,5-dioxopyrrolidin-l-yl 3-((3-((3-(pyridin-2- yldi e (69).
  • Compound 68 (2.50 g, 6.08 mmol) in DMA (50 ml) was added NHS (0.80 g, 6.96 mmol) and EDC (3.00 g, 15.62 mmol). The mixture was stirred under Ar overnight, evaporated and purified on S1O 2 chromatography eluted with EtOAc/DCM (1 :10 to 1:5), pooled the fractions and evaporated to afford the title compound (2.74 g, 86% yield).
  • ESI MS m/z+ 547.10 (M+Na).
  • Example 22 methyl 3-((3-mercaptopropyl)thio)propanoate (71).
  • Methyl 3-bromopropanoate 51 (10.010 g, 60.24 mmol) in DMA (80 ml) was added ethane- 1,2-dithiol (40.0 g, 425.4 mmol) and DIPEA (150 ml). The mixture was stirred at 45 °C under Ar for 8 h, evaporated and purified on S1O 2 chromatography eluted with 1 : 10:0.01% EtOAc/DCM/HOAc, pooled the fractions, and evaporated to afford the title compound 71 (8.56 g, 79% yield). ESI MS m/z+ 203.10 (M+Na).
  • Example 23 methyl 3-((2-((3-bromobutyl)thio)ethyl)thio)propanoate (82).
  • the antiHer2 antibody is modified with phosphinate linker at 8 mg/mL antibody, a 10 fold molar excess of phosphinate linker ( ⁇ 30mM stock solution in DMA).
  • the reaction is carried out in 100 mM NaH 2 P0 4 , pH7.4 buffer with DMA (5% v/v) for 15, 30, 60, 120, and 240 minutes at 25 °C.
  • the modified antiHer2 was purified by G25 column with 50 mM NaH 2 P0 4 , 50 mM NaCl, and 2 mM EDTA, pH6.5 to remove the excess phosphinate linker.
  • Example 29 Conjugate synthesis.
  • a hydrophilic linker containing thiopyridine (SPP) linker was dissolved in DMA at a concentration of approximately 10 mM.
  • An antibody was dialyzed into buffer A (50 mM NaH 2 P0 4 , 50 mM NaCl, 2 mM EDTA, pH 6.5).
  • buffer A 50 mM NaH 2 P0 4 , 50 mM NaCl, 2 mM EDTA, pH 6.5.
  • the linker reaction was at 8 mg/ml, and 4-6 equivalents of linker were added while stirring in the presence of 5% (v/v) DMA. The reaction was allowed to proceed at ambient temperature for 90 minutes.
  • the targeted cells e.g. Ramos cells, 20,000 cells
  • the targeted cells were cultured in the presence of various concentrations of unconjugated antibody or the antibody conjugate for 96 hours after which cell viability was measured by propidium iodide exclusion and analyzed by flow cytometry using a Becton Dickinson FACSort (Becton Dickinson, Franklin Lakes, NJ). Red fluorescent intensity (emission at 617 nm in the FL2 channel) of the cells excited at 488 nm was measured.
  • the regions for viable cells were also set using both the forward light scatter and right-angle light scatter properties of the cells.
  • the loss of viability was determined by the loss of cells from within the gated region defining viable cells. The average number of viable cells per 6 replicate cultures was calculated.
  • the survival fraction was plotted versus conjugate concentration to determine the / 50 value (50% cell killing concentration) of the conjugate.
  • Antczak, C et al. Bioconjug Chem 2006, 17, 1551.

Abstract

Cell binding agent-drug conjugates comprising hydrophilic linkers, and methods of using such linkers and conjugates are provided.

Description

HYDROPHILIC LINKERS AND THEIR USES FOR CONJUGATION OF DRUGS TO
CELL BINDING MOLECULES
FIELD OF THE INVENTION
5 The present invention relates to the preparation of novel hydrophilic linkers used for the conjugation of a drug, 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 comprising either modification of drugs with these hydrophilic linkers first, followed by reaction with cell-binding agents; or modification of cell-binding agents 10 with these hydrophilic linkers first, followed by reaction with drugs.
BACKGROUND OF THE INVENTION
Nowadays the development on targeted cancer therapy requires not only to supplement the conventional chemotherapy and radiotherapy while sparing healthy cells, greatly reducing or eliminating the often unpalatable side effects, but also aim to
15 overcome drug resistance (Tiirk, D, and Szakacs, G., Curr Opin Drug Discov Devel.
2009, 12, 246; Zhao, R. et al, J. Med. Chem. 2011, 56, 5404; Yauch, R. L, Settleman, J. Curr Opin Genet Dev. 2012, 22, 45). There are several systemic deliveries for targeted treatment of tumor that have been studied during the past three decades: Heat-activated targeted drug delivery; Tissue-selective drug delivery for cancer using carrier-mediated
20 transport systems; Tumor-activated prodrug therapy for targeted delivery of
chemotherapy; Pressure-induced filtration of drug across vessels to tumor; Promoting selective permeation of the anticancer agent into the tumor; Two-step targeting using a bispecific antibody; Site-specific delivery and light-activation of anticancer proteins.
Many special formulations and carriers have been studied for target delivery of
25 anticancer drugs, such as Albumin-based drug carriers; Carbohydrate-enhanced
chemotherapy; Proteins and peptides based drug carriers; Fatty acids as targeting
vectors linked to active drugs; Microsphere carriers; Monoclonal antibodies as carriers;
Vitamins, e.g. folates as carriers; Nanoparticle carriers,; Liposome carriers, e. g.
pegylated liposomes (enclosed in a polyethylene glycol bilayer); Polyethylene glycol
30 (PEG) carriers; Single-chain antigen-binding molecule carriers; Polymeric micelle
carriers; Lipoprotein-based drug carriers; Dendrimers; etc. An ideal drug delivery vehicle must be non-toxic, biocompatible, non-immunogenic, and biodegradable (Scott, R; Crabbe, D; et al (2008) Expert Opin. Drug Deli. 5, 459) and avoid recognition by the host's defense mechanisms (Saltzman, W.; Torchilin, V. (2008). "Drug delivery systems" Access Science. McGraw-Hill Co.). The link between the delivery vehicles, in particular, antibodies and the cell-killing agent plays a critical role in the development of targeted drug delivery systems, as the nature of the linker significantly affects the potency, selectivity and the pharmacokinetics of the resulting conjugates (Zhao, R.;
Wilhelm, S. et al, (2011) J. Med. Chem. 36, 5404; Doronina, S.; Mendelsohn, B.; et al, (2006) Bioconjug Chem, 17, 114; Hamann, P.; Hinman, L; et al. (2005) Bioconjug
Chem. 16, 346). Four types of linkers had been used for preparation of cell binding agent-drug conjugates that have entered the clinic: (a) acid-labile linkers, exploiting the acidic endosomal and lysosomal intracellular microenvironment; (b) linkers cleavable by lysosomal proteases; (c) chemically stable thioether linkers that release a lysyl adduct after proteolytic degradation of the antibody inside the cell; and (d) disulfide - containing linkers, which are cleaved upon exposure to an intracellular thiol (Zhao, R.; Wilhelm, S. et al, 2011 J. Med. Chem. 36, 5404).
Conjugates of cell-binding agents with drugs or modified chemical compounds via different types of linkers have been described (U.S. Patent Nos. 4,680,338, 5,122,368, 5,141,648, 5,208,020, 5,416,064; 5,475,092, 5,543,390, 5,563,250 5,585,499, 5,880,270, 6,214,345, 6,436,931, 6,372,738, 6,340,701, 6,989,452, 7,129,261, 7,375,078, 7,498,302, 7,507,420, 7,691,962, 7,910,594, 7,968,586, 7,989,434, 7,994,135, 7,999,083, 8,153,768, 8,236,319, Zhao, R.; et al, (2011) J. Med. Chem. 36, 5404; Doronina, S.; et al, (2006) Bioconjug Chem, 17, 114; Hamann, P.; et al. (2005) Bioconjug Chem. 16, 346). Typically, in these conjugates, the cell-binding agents are first modified with a bifunctional agent such as SPDP (N-succinimidyl 3-(2-pyridyldithio) propionate), or SMCC (succinimidyl-4-(N- maleimidomethyl)cyclohexane-l-carboxylate), or SPDB (N-succinimidyl 4-(2- pyridyldithio)butanoate), to introduce an active disulfide or a maleimido moiety. Reaction with a thiol-containing cytotoxic drug provides a conjugate in which the cell-binding agent, such as a monoclonal antibody, and drug are linked via disulfide bonds or thioether bonds.
However, the use of the cell binding molecule-drug conjugates, such as antibody- drug conjugates (ADCs), in developing therapies for a wide variety of cancers has been limited both by the availability of specific targeting agents (carriers) as well as the conjugation methodologies which result in the formation of protein aggregates when the amount of the drugs that are conjugated to the carrier (i.e., the drug loading) is increased. Normally the tendency for cytotoxic drug conjugates to aggregate is especially problematic when the conjugation reactions are performed with the hydrophobic linkers. Since higher drug loading increases the inherent potency of the conjugate, it is desirable to have as much drug loaded on the carrier as is consistent with retaining the affinity of the carrier protein. The presence of aggregated protein, which may be nonspecifically toxic and immunogenic, and therefore must be removed for therapeutic applications, makes the scale -up process for the production of these conjugates more difficult and decreases the yield of the products.
Consequently, there is a critical need to improve methods for conjugating drugs/cytotoxic drugs to carriers (cell binding molecules) that minimize the amount of aggregation and thereby allow for as high a drug loading as possible through application of a hydrophilic cross-linker.
SUMMARY OF THE INVENTION
The present invention provides hydrophilic linkers containing phosphinate, sulfonyl, and/or sulfoxide groups to link drugs to a cell-binding agent (e.g., an antibody). The preferred formula of the cell binding molecule - hydrophilic linker- drug conjugates can be represented as: Cb-(-L-Drug)n, wherein Cb is a cell-binding agent, L is a hydrophilic linker, Drug is a drug molecule, and n is an integer from 1 to 20. The advantages in applying the hydrophilic linker in the cell molecule-drug conjugate are: a), reducing the aggregation of the conjugates in water based media; b). enabling higher drug-per-cell binding molecule -ratio conjugate, resulting in higher potency; c). being retained inside the target cell after the drug-linker released from the conjugates, which can combat permeability-glycoprotein (Pgp)-expressing multidrug resistant (MDR) cells.
In one aspect of the present invention, the hydrophilic linker is represented by formula (I) wherein Y can react with a cell-binding agent and Z can react with a cytotoxic drug:
Figure imgf000004_0001
Wherein:
Y represents a functional group that enables reaction with a cell-binding agent; Q and T are either -P(=0)(OM)-, or -S(02)-, or -S(O)-;
m and n are integer from 0 to 5, but not 0 at the same time; In addition, when m = 1, n = 0, Q is not -P(=0)(OM)-; or when n = 1, m = 0, T is not -P(=0)(OM)-;
Z represents a functional group that enables linkage of 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;
Ri, P2, P3, PM, P5 and P6, are the same or different and are 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 (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
Additionally Ri, R2, R3 and R4 are respectively a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl group, the conjugated drug and among themselves (Ri, R2 , R3 and R4). The atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming 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.
M is H, or Na, or K, or N+RiR2R3 or a pharmaceutical salt. Ri, R2 and R3 are described above.
In another aspect, this invention provides a cell-binding agent-drug conjugate of formula (II), in which the cell-binding agent, Cb, and the drug, Drug, have reacted at the two end
Figure imgf000005_0001
(Π)
wherein: Cb represents a cell-binding agent;
Drug represents the drug linked to the cell-binding agent via the hydrophilic linker by a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
q is 1 ~ 20; Q, T, m, n, Ri, R2, R3, R4, R5 , R6 and M are described the same previously in formula (I).
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 hydrophilic linker, which s :
Figure imgf000006_0001
(III)
Wherein the substituents are as defined above.
In an even further aspect, the present invention provides a modified drug of formula (IV), in which the drug, Drug, has reacted with the hydrophilic linker, which still has Y, a group c :
Figure imgf000006_0002
(IV)
Wherein the substituents are as defined above.
The present invention further relates to a method of making a cell-binding molecule-drug conjugate of formula (II), wherein the drug is linked to a cell-binding agent via the hydrophilic linker.
The present invention also relates to a method of making a modified cell-binding molecule of formula (III), wherein the cell-binding molecule is reacted with the hydrophilic linker.
The present invention also relates to a method of making a modified drug of formula (IV), wherein the drug is reacted with the hydrophilic linker.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the synthesis of diphosphinate-containing cross-linking reagents that contain a pyridyldisulfide group and a reactive carboxylic acid ester, and the linker used for the conjugation of an antibody. Ammonium phosphinate are first converted into bis(trimethylsilyl) phosphonite, followed by Michael addition with an acrylate and then substitution reaction with excess amount of 1,2-dibromo ethane to form (2-bromoethyl)(3- ethoxy-3-oxopropyl)phosphinic acid (4). The bromoethyl phosphinic acid moiety (5) was then substituted with bis(trimethylsilyl) phosphonite (2) and 1 ,2-dibromo ethane to generate (2-bromoethyl)(2-(ethoxy(3-ethoxy-3-oxopropyl)phosphoryl)ethyl)phosphinic acid (8), whose bromide group was then replaced by potassium ethyl xanthogenate, followed by basic hydrolysis and a substitution reaction with an excess of 2,2'- dithiobispyridine, as well as condensation reaction of the acid 10 with N-hydroxysuccimide (NHS) in an acid medium using the carbodiimide coupling agent EDC to give the diphosphinate linker (11). The linker 11 can be used for the conjugation of drugs to a cell binding molecule (such as antibody).
Figure 2 shows the synthesis of a triphosphinate-containing linker 18 that contains a pyridyldisulfide group and a reactive carboxylic acid ester. The linker can be used for the conjugation of a drug to cell binding molecule via a disulfide bond.
Figures 3 shows the synthesis of tetraphosphinate-containing cross linkers 23 that contain a reactive carboxylic acid ester and a pyridyldisulfide group, enabling linkage of a drug to a cell binding molecule via a disulfide bond.
Figure 4 shows the synthesis of diphosphinate-containing cross-linker 26 that contain a maleimido group and a reactive carboxylic acid ester enabling linkage of a drug to a cell binding molecule via a thioether bond
Figures 5 shows the synthesis of monophosphinate, diphosphinate, triphosphinate and tetraphosphinate-containing linkers that contain a pyridyldisulfide group and a reactive carboxylic acid ester via substitution of ethyl 2-bromoacetate with bis(trimethylsilyl) phosphonite (2).
Figure 6 shows the synthesis of linking reagents that contain a vinylsulfonyl (55), a monosulfonyl (61) and disulfonyl (69) groups.
Figures 7 shows the synthesis of mono-, and di-sulfonyl linkers
Figure 8 shows the synthesis of mono-sulfonyl linkers containing polyethylene glycols. Figure 9 shows the synthesis of di-sulfonyl linkers containing polyethylene glycols. The linker can be used for the conjugation of an antibody or a protein via a disulfide bond.
Figure 10 shows the synthesis of a hydrophilic linker containing a phosphinic acid and a sulfonyl group.
Figure 11 shows the synthesis of a hydrophilic linker containing contain a phosphinate, a sulfonyl and a ketone groups that can be used for the conjugation of a drug to an antibody or a protein via a hydrazone bond.
Figure 12 shows the synthesis of hydrophilic linkers that contain a disulfonyl, polyethylene glycols, a much hindered pyridyldisulfide and a reactive carboxylic acid ester groups via substitution of l,3-dibromo-3-methylbutane. The linker is used for the conjugation of drugs to an antibody or a protein via a hinder disulfide bond.
Figure 13 shows the synthesis of hydrophilic linkers containing a phosphinate and a sulfonyl groups that are used for the conjugation of drugs to a cell binding molecule either via a triazole or via a thiolether bond.
Figures 14, shows synthesis of hydrophilic linkers that contain a disulfonyl, phosphinate, a pyridyldisulfide group, a polyethylene glycol (PEG) chain and a reactive carboxylic acid ester. The linkers are used for the conjugation of a cell binding molecule via cell thioether bond
Figures 15 shows synthesis of hydrophilic linkers that contain a disulfonyl, phosphinate, an alkoxylamino and a maleimido substituents, enabling ketone or aldehyde- containing drug to link to an antibody via a thioether and an alkoxime bond..
Figure 16 shows the synthesis of disulfonyl-containing linkers that contains an alkoxylamino and a maleimido substituent, enabling ketone or aldehyde- containing drug to link to an antibody via a thioether and an alkoxime bond.
Figure 17 shows the synthesis of sulfoxide-containing and disulf oxide-containing linkers that are used for the conjugation of drugs to a cell binding molecule via a disulfide bond.
Figures 18 show the synthesis of a disulfonyl-containing linker (221) that used for conjugation of amine-containing cytotoxic drugs to an antibody via the Val-Cit-PABC linkage. Figure 19 (19-a~19-z) shows the antibody-drug conjugate (ADC) structures of the typical cytotoxic agents (the analogs of tubulysins, calicheamicins, maytansinoids, auristatins, doxorubicin, daunorubicin, CC-1065, pyrrolobenzodiazepine dimmers) via the hydrophilic linkers of this patent.
Figure 20 shows the use of a hydrophilic linker (86) in modifying a cell-binding agent (antiHer2 antibody) and following by production of a cell-binding agent-drug (TZ03) conjugate containing the hydrophilic linker.
Figure 21 shows 5 days in vito assays of the cytotoxicity of the antiCD22-TZ041 (tubulysin analog) conjugate with different drug load ratios via a hydrophilic linker (86). DETAILED DESCRIPTION OF THE INVENTION DEFINITIONS
"Alkyl" means an aliphatic hydrocarbon group which may be straight or branched having 1 to 8 carbon atoms in the chain or cyclic. "Branched" means that one or much lower alkyl groups such as methyl, ethyl or propyl are attached to a linear alkyl chain. Exemplary alkyl groups include methyl, ethyl, n-propyl, /-propyl, «-butyl, Z-butyl, «-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-methylhexyl, 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 d-Cs alkyl group can be unsubstituted or substituted with one or more groups including, but not limited to, -Ci~C8 alkyl, -0-(Ci~C8 alkyl), - aryl, -C(0)R', -OC(0)R', -C(0)OR', -C(0)NH2, -C(0)NHR', -C(0)N(R')2-NHC(0)R', - S(0)2R', -S(0)R', -OH, -halogen (F, CI, Br or I), -N3, -NH2, -NH(R'), -N(R') 2 and -CN; where each R' is independently selected from -Ci~Cs alkyl and aryl.
A "C3~Cs carbocycle" means a 3-, 4-, 5-, 6-, 7- or 8-membered saturated or unsaturated non-aromatic carbocyclic ring. Representative C3~Cs 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~Cs carbocycle group can be unsubstituted or substituted with one or more groups including, but not limited to, -Ci~C8 alkyl, -0-( Ci~C8 alkyl), -aryl, -C(0)R', -OC(0)R', -C(0)OR', - C(0)NH2, -C(0)NHR', -C(0)N(R') 2-NHC(0)R', -S(0) 2R', -S(0)R', -OH, -halogen, -N3, - NH2, -NH(R'), -N(R') 2 and -CN; where each R' is independently selected from -Ci~C8 alkyl and aryl.
A "Cs-Cs carbocyclo" refers to a C3~Cs carbocycle group defined above wherein one of hydrogen atoms on the carbocycle is replaced with a bond.
"Heterocycle" refers to an aromatic or non-aromatic C3~Ci4 carbocycle in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group of O, N, S Se, and P. Preferable heteroatoms are oxygen, nitrogen and sulphur. Suitable heterocyclics are also disclosed in The Handbook of Chemistry and Physics, 76th Edition, CRC Press, Inc., 1995-1996, p. 2-25 to 2-26, the disclosure of which is hereby incorporated by reference.
Preferred non aromatic heterocyclic include, but are not limited to pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxiranyl, tetrahydrofuranyl, dioxolanyl, tetrahydro-pyranyl, dioxanyl, dioxolanyl, piperidyl, piperazinyl, morpholinyl, pyranyl, imidazolinyl, pyrrolinyl, pyrazolinyl, thiazolidinyl, tetrahydrothiopyranyl, dithianyl, thiomorpholinyl, dihydro- pyranyl, tetrahydropyranyl, dihydropyranyl, tetrahydro-pyridyl, dihydropyridyl, tetrahydropyrinidinyl, dihydrothiopyranyl, azepanyl, 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.
"Halogen atom" refers to fluorine, chlorine, bromine or iodine atom; preferably bromine and chlorine atom.
"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 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 combinations.
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 pharmaceutically 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, glucoronic, 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.
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 hydrophilic cross-linkers. Examples of some suitable cross-linkers and their synthesis are shown in Figures 1 to 18.
THE HYDROPHILIC LINKERS
The synthetic routes to produce hydrophilic cross linkers as well as the preparation of the conjugates of drugs to a cell binding molecules of the present invention are shown in Figures 1-19. The hydrophilic cross linkers possess three elements: a) substituents that are either phosphinate, or sulfonyl or sulfoxide groups, or mixed of these groups, b) a group, such as a N-hydroxysuccimimide ester, maleimido group, haloacetyl group, and hydrazide, capable of reaction with a cell-binding agent, and c) a group, such as but not limited to, a disulfide, maleimide, haloacetyl, aldehyde, ketone, azide, amine, alkoxylamino and hydrazide, capable of reaction with a drug. The hydrophilic substituents can be introduced by methods described herein. For example of the phosphinate substituents, they can be introduced by first treating a commercially available ammonium phosphinate with an acrylate via Michael addition and followed by substitution of excess amount of dibromo alkane to a phosphinate group. For example of the sulfonyl and sulfoxide substituents, they can be introduced by first generation of thioether components, followed by oxidization of these components. More detail synthesis of the hydrophilic linkers and their uses for the preparation of cell binding ligand-drug conjugates of this invention are disclosed in the figures 1-19.
mpounds of the formula (I) below:
Figure imgf000012_0001
(I)
wherein:
Y represents a functional group that enables reaction with a cell-binding agent; Q and T are either -P(=0)(OM)-, or -S(02)-, or -S(O)-;
m and n are integer from 0 to 5, but not 0 at the same time; In addition, when m = 1 , n = 0, Q is not -P(=0)(OM)-; or when n = 1 , m = 0, T is not -P(=0)(OM)-;
Z represents a functional group that enables linkage of 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;
Ri, P2, P3, P4, P5 and P6, are the same or different and are 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 (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
In another embodiment, Ri, R2, R3, and R4 can be respectively a chain of atoms selected from C, Ν, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl or sulfoxide group, the conjugated drug and themselves (Ri, R2, R3 and R4). The atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming 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.
M is H, or Na, or K, or N+RiR2R3 or a pharmaceutical salt. Ri, R2 and R3 are described above.
Examples of the functional group, Y, that enables reaction with a cell-binding agent include amine reacting agents such as but not limited to N-hydroxysuccinmide esters, p- nitrophenyl esters, dinitrophenyl esters, pentafluorophenyl esters; thiol reactive agents such as but not limited to pyridyldisulfides, nitropyridyldisulfides, maleimides, haloacetates and carboxylic acid chlorides.
Examples of the functional group, Z, which enables linkage of a cytotoxic drug, include groups that enable linkage via an either disulfide, or thioether, thioester, peptide, hydrazone, ester, carbamate, carbanate, alkoxime or amide bond. Such functional groups include, but are not limited to, thiol, disulfide, amino, carboxy, aldehydes, maleimido, haloacetyl, hydrazines, and hydroxy.
In preferred embodiments, Ri, R2, R3, and R4, are linear alkyl having from 1-6 carbon atoms, or polyethyleneoxy unit of formula (OCH2CH2)p, p = 1-100,
The synthesis of 2-dithio-pyridyl containing cross-linkers of formulae (I) is shown, for example, in Figures 1, 2, 3, 5, 6, 7, 8, 9, 10, 12, 13, 14, and 17. The synthesis of maleimido-containing cross linkers of the formula (I) is shown, for example, in Figures 4, 8, 9, 10,12, 14, and 18. The synthesis of thioether-containing cross linkers of the formula (I) is shown, for example, in Figures 6, 8, 10, and 13, The synthesis of polyethylene glycol- containing hydrophilic cross linkers of formula (I) is shown, for example, in Figures 8, 9, and 14. The synthesis of azide-containing hydrophilic cross linkers of formula (I) for
Huisgen 1,3-dipolar cycloaddition of azides to alkynes is shown, for example, in Figures 13. The synthesis of hydrophilic cross linkers of formula (I) bearing a hydrazide or ketone moieties enabling linkage via acid-labile bonds is shown, for example, in Figures 11, 16and 18. The synthesis of hydrophilic cross linkers of formula (I) bearing an alkoxylamino moiety enabling linkage via alkoxime bonds is shown, for example, in Figure 15 andl6. The synthesis of dipeptide -containing cross linkers of the formula (I) is shown, for example, in Figure 18.
CELL-BINDING AGENT-DRUG CONJUGATES
The conjugates of the present invention can be represented by the following formula, Cb-(-L-Drug)n, wherein Cb is a cell-binding agent, L is a hydrophilic linker, Drug is a drug molecule, and n is an integer from 1 to 20.
The hydrophilic 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 (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.
containing linkers are:
Figure imgf000014_0001
-maleimidocaproyl containing)
Figure imgf000014_0002
(MP, maleimidopropanoyl containing)
Figure imgf000015_0001
-aminobenzyloxycarbonyl containing)
Figure imgf000015_0002
-(N-maleimidomethyl)cyclohexane-l carboxylate)
Figure imgf000015_0003
((4-acetyl)aminobenzoate containing)
Figure imgf000015_0004
(4-thio-2-hydroxysulfonyl-butyrate, 2-sulfo-SPDB)
Figure imgf000015_0005
wherein:
Cb represents a cell-binding agent;
Drug represents the drug linked to the cell-binding agent via the hydrophilic linkers of this invention by a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, heterocyclic ring, amine, imine, alkoxime or amide bond; q is 1 ~ 20; Q, T, m, n, Ri, R2, R3, R4, R5 , R6 and M are described the same previously in formula (I).
As described in more detail below, the drug can be any of many small molecule 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 hydrophilic linkers of the present invention to introduce reactive disulfide groups, maleimido, haloacetyl, azide, 1-yne, ketone, or hydrazide groups. 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 containing 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 cell-binding agent and a drug containing a free thiol group. Synthesis of conjugates bearing an acid labile hydrazone link 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).
Alternatively, the drug can be modified with the hydrophilic linkers of the present invention to give a modified drug of formula (IV) bearing functionality capable of reacting with a cell binding agent. For example a thiol-containing drug can be reacted with the hydrophilic linker of formula (I) bearing a maleimdo or a haloacetyl or an ethylsulfonyl substituent at neutral pH in aqueous buffer to give a drug connected to the hydrophilic linker via a thioether link. A thiol-containing drug can undergo disulfide exchange with a hydrophilic linker bearing a pyrdiyldithio moiety to give a modified drug attached via a disulfide bond to the hydrophilic cross linker. A drug bearing a hydroxyl group or a thiol group can be reacted with a hydrophilic linker bearing a halogen of this invention, in the presence of a mild base, to give a modified drug bearing an ether or thiol ether link. A hydroxyl group containing drug can be condensed with a hydrophilic cross linker of formula (I) bearing a carboxyl group, in the presence of a dehydrating agent, such as EDC or dicyclohexylcarbodimide, to give an ester link. An amino group containing drug can similarly undergo condensation with a carboxyl group on the hydrophilic linker of formula (I) to give an amide bond.
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 exchange or by dialysis. In some cases (e.g. folic acid, melanocyte stimulating hormone, EGF etc) the cell-binding agent-drug conjugates can be purified by chromatography such as by HPLC, medium pressure column chromatography or ion exchange chromatography.
MODIFIED CELL-BINDING AGENTS
The cell-binding agent modified by reaction with linkers of the present invention are
Figure imgf000017_0001
wherein the substituents are as described above for the hydrophilic linkers and the cell-binding agent drug conjugates.
In preferred embodiments, Z is a disulfide substituent, a maleimido, haloacetyl group, or an N-hydroxysuccinimide ester, and Cb linked with Ri is through thioether, amide, or disulfide bond. The modified cell-binding agent can be prepared via a reaction of the cell-binding agent with the hydrophilic linkers by methods known in the art for other cross-linkers (U.S. Patent Nos. 5,846,545, 5,585,499, 5,475,092, 5,414,064, 5,208,020, and 4,563,304; Carlsson, J. et al. Biochem. J. (1978) 173, 723-737(1978); Goff, D. A.,
BioConjugate Chem. (1990), 1, 381-386; L. Delprino et al. J. Pharm. Sci. (1993), 82, 506- 512; S. Arpicco et al., Bioconjugate Chem \991), 8, 327-337).
Advantageously, because the phosphinate groups and sulfonyl groups on the hydrophilic linkers are soluble in water or require only a small percentage of organic solvent to maintain solubility in aqueous solution, the reaction between the cell-binding agent and the cross-linker can be conducted in aqueous solution. The cross-linking reagent is dissolved in aqueous buffer, optionally containing a small amount (typically <10% by volume) of 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 dimethylsulf oxide (DMSO) at a high concentration, for example 1-100 mM, and then an appropriate aliquot is added to the buffered aqueous solution of the cell-binding agent. An appropriate aliquot is an amount of solution that introduces 1-10 cross-linking groups per cell-binding agent, preferably 1-5 groups, and the volume to be added should not exceed 10 %, preferably 5 %, and most preferably 0-3 % of the volume of the cell-binding agent solution. The aqueous solutions for the cell-binding agents are buffered between 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, NaCl and KC1. After the addition the reaction is incubated at a temperature of from 4 °C to 40 °C, preferably at ambient temperature. The progress of the reaction can be monitored by measuring the increase in the absorption at 320 nm or another 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 chromatography.
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. Figure 20 shows the results from the modification of the cell-binding agent, the her2 antibody, with a chydrophilic cross-linker of the present invention. The time course of linker/antibody (L/A) incorporation is shown, for example, along with the drugs/antibody (D/A) linked. The hydrophilic cross-linkers described herein have diverse functional groups that can react with any cell-binding agent that possesses a suitable substituent. For example cell-binding agents bearing an amino or hydroxyl substituent can react with cross linkers bearing an N-hydroxysuccinimide (NHS) ester, cell-binding agents bearing a thiol substituent can react with cross linkers bearing a maleimido or haloacetyl group. Additionally, cell- binding agents bearing a carbonyl substituent can react with cross linkers bearing a hydrazide or a hydroxylamine. One skilled in the art can readily determine which linker to use based on the known reactivity of the available functional group on the cell-binding agent. MODIFIED CYTOTOXIC DRUGS
The cytotoxic drugs modified by reaction with cross-linkers of the present invention are
Figure imgf000019_0001
(IV)
wherein the substituents are as defined above.
In preferred embodiments, Y is a disulfide substituent, a maleimido, haloacetyl group, or an N-hydroxysuccinimide ester.
The modified drugs can be prepared by reacting the drug with the cross linkers of the present invention to give a modified drug of formula (IV) bearing a functionality capable of reacting with a cell binding agent. For example a thiol-containing drug can be reacted with the cross linker of formula (I) bearing a maleimdo substituent at neutral pH in aqueous buffer to give a drug connected to the hydrophilic linker via thioether linkage. A thiol-containing drug can undergo disulfide exchange with a hydrophilic linker bearing a pyrdiyldithio moiety to give a modified drug attached via a disulfide bond to the hydrophilic cross linker. A drug bearing a hydroxyl group can be reacted with a cross linker bearing a halogen, in the presence of a mild base, to give a modified drug bearing an ether link. A hydroxyl group containing drug can be condensed with a cross linker of formula (I) bearing a carboxyl group, in the presence of a dehydrating agent, such as EDC or dicyclohexylcarbodimide (DCC), to give an ester link. A drug bearing a thiol group can be reacted with a cross linker bearing a malimido or a vinylsulfonyl, or a haloacetyl group, to give a modified drug bearing thioether link. An amino group containing drug can similarly undergo condensation with a carboxyl group on the hydrophilic cross linker of formula (I) to give an amide bond. The modified drug can be purified by standard methods such as column chromatography over silica gel or alumina, 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, FVj [Parham, J.
Immunol. 131, 2895-2902 (1983)], fragments produced by a Fab expression library, anti- idiotypic (anti-Id) antibodies, CDR's, 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-6, GM-CSF, interferon-gamma (IFN-γ); hormones such as insulin, TRH (thyrotropin releasing hormones), MSH (melanocyte-stimulating hormone), steroid hormones, such as androgens and estrogens, melanocyte-stimulating hormone
(MSH); growth factors and colony-stimulating factors such as epidermal growth factors (EGF), granulocyte-macrophage colony-stimulating factor (GM-CSF), transforming growth factors (TGF), such as TGFa, TGF , 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; 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 membrane antigen (PSMA) 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); 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, 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 monoclonal antibodies are preferred as a cell-surface binding agent if an appropriate one is available. And antibodies may be murine, human, humanized, chimeric, or derived from other species.
Production of antibodies used in the present invention involves in vivo or in vitro procedures 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 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 antibody 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 maintained 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, 20 mm glutamine, 20% fetal calf serum 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 transfection 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, the anti-receptor peptide or a peptide analog is used either alone or conjugated to an immunogenic carrier, as the immunogen for producing anti-receptor peptide monoclonal antibodies.
There are also a number of other well-known techniques for making monoclonal antibodies 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, Inc.), 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 human 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.
Antibodies immunospecific for a malignant cell antigen can also be obtained commercially 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 antibodies 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 techniques 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, 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 hydrophilic linkers of this prevention for treating cancer, autoimmune disease, and infectious disease include, but are not limited to, 3F8 (anti-GD2), Abagovomab (anti CA-125), Abciximab (anti CD41 (integrin alpha-lib), 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-CCLl l (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 (cACIO, anti-CD30 TNFRSF8), 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 (Erbitux, IMC-C225, anti-EGFR), Citatuzumab bogatox (anti-EpCAM), Cixutumumab (anti-IGF-1),
Clenoliximab (anti-CD4), Clivatuzumab (anti-MUCl), Conatumumab (anti-TR AIL-R2) , CR6261 (anti-Influenza A hemagglutinin), Dacetuzumab (anti-CD40), Daclizumab (Zenapax, anti-CD25 (a chain of IL-2 receptor)), Daratumumab (anti-CD38 (cyclic ADP ribose hydrolase), Denosumab (Prolia, anti-RANKL), Detumomab (anti-B -lymphoma cell), Dorlimomab, Dorlixizumab, Ecromeximab (anti-GD3 ganglioside), Eculizumab (Soliris, anti-C5), Edobacomab (anti-endo toxin), Edrecolomab (Panorex, MA 7-1A, anti-
EpCAM), Efalizumab (Raptiva, anti-LFA-1 (CDl la), 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 νβ3), 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-γ), Foravirumab (anti-rabies virus glycoprotein), Fresolimumab (anti-TGF-β), Galiximab (anti-CD80), Gantenerumab (anti- beta amyloid), Gavilimomab (anti-CD 147 (basigin)), Gemtuzumab (anti-CD33),
Girentuximab (anti-carbonic anhydrase 9), Glembatumumab (CR011, anti-GPNMB),
Golimumab (Simponi, anti-TNF-a), Gomiliximab (anti-CD23 (IgE receptor)), Ibalizumab (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 receptor)),
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- CD23 (IgE receptor), Mapatumumab (anti-TRAIL-Rl), 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 a4), Nebacumab (anti-endo toxin), Necitumumab (anti-EGFR), Nerelimomab (anti-TNF-a), Nimotuzumab (Theracim, Theraloc, anti-EGFR), Nofetumomab, Ocrelizumab (anti-CD20), Odulimomab (Afolimomab, anti-LFA-1 (CD 11a)), 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), Panitumumab (Vectibix, ABX-EGF, anti-EGFR), Panobacumab (anti- Pseudomonas aeruginosa), Pascolizumab (anti-IL-4), Pemtumomab (Theragyn, anti-MUCl), Pertuzumab (Omnitarg, 2C4, anti-HER2/neu), Pexelizumab (anti-C5), Pintumomab (anti- adenocarcinoma antigen), Priliximab (anti-CD4), Pritumumab (anti-vimentin), PRO 140 (anti-CCR5), Racotumomab (1E10, anti-(N-glycolylneuraminic acid (NeuGc, NGNA)- 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
(LeukArrest, anti-CDl l, CD 18), Ruplizumab (Antova, anti-CD 154 (CD40L)), 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-1 -phosphate), Sontuzumab (anti-episialin), Stamulumab (anti-myostatin), Sulesomab (LeukoScan, (anti- NCA-90 (granulocyte antigen), Tacatuzumab (anti-alpha-fetoprotein), Tadocizumab (anti- integrin αιπ,β3), 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 (anti-IL-13), Tocilizumab (Atlizumab, Actemra, RoActemra, (anti-IL-6 receptor), Toralizumab (anti-CD 154 (CD40L)), 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), Ustekinumab (Stelara, anti-IL-12, IL-23), Vapaliximab (anti-AOC3 (VAP-1)),
Vedolizumab, (anti-integrin α4βγ), Veltuzumab (anti-CD20), Vepalimomab (anti-AOC3 (VAP-1), Visilizumab (Nuvion, anti-CD3), Vitaxin (anti-vascular integrin avb3),
Volociximab (anti-integrin δβι), 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
(Enbrel®), Alefacept (Amevive®), Abatacept (Orencia®), Rilonacept (Arcalyst), 14F7 [anti-IRP-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), 225.28S [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-DRlO, 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, TRo$ (T cell receptor alpha/beta), complex, from Medlmmune 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 adhesion molecule), anti-TACSTDl (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 binding 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
(carcinomas), CA242 (colorectal), placental alkaline phosphatase (carcinomas), prostate specific antigen (prostate), prostatic acid phosphatase (prostate), epidermal growth factor (carcinomas), CD2 (Hodgkin's disease, NHL lymphoma, multiple myeloma), CD3 epsilon (T cell lymphoma, lung, breast, gastric, ovarian cancers, autoimmune diseases, malignant ascites), CD19 (B cell malignancies), CD20 (non-Hodgkin's lymphoma), CD22 (leukemia, lymphoma, multiple myeloma, SLE), CD30 (Hodgkin's lymphoma), CD33 (leukemia, autoimmune 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 (carcinomas), 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 (A-like-4), EGFR (Epidermal Growth Factor Receptor, various cancers), CTLA4 (melanoma), CXCR4 (CD 184, Heme-oncology, solid tumors), Endoglin (CD 105, 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), HER1 (lung, stomach cancers), HER2 (breast, lung and ovarian cancers), HLA-DR10 (NHL), HLA-DRB (NHL, B cell leukemia), human chorionic gonadotropin (carcinoma), IGF1R (insulin-like growth factor 1 receptor, solid tumors, blood cancers), IL-2 receptor
(interleukin 2 receptor, T-cell leukemia and lymphomas), IL-6R (interleukin 6 receptor, multiple myeloma, RA, Castleman's disease, IL6 dependent tumors), Integrins (ανβ3, α5β1, α6β4, α11β3, α5β5, ανβ5, 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 (colorectal), gplOO (melanoma), MARTI (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 glycoprotein 72, AML, gastric, colorectal, ovarian cancers), T cell transmembrane protein (cancers), Tie (CD202b), TNFRSF10B (tumor necrosis factor receptor superfamily member 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, lymphoma, 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). Examples of these antigens that antibodies against are: Many other Cluster of Differentiations (CD4, CD5, CD6, CD7, CD8, CD9, CD10, CDl la, CDl lb, CDl lc, CD12w, CD14, CD15, CD16, CDwl7, CD18, CD21, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD31, CD32, CD34, CD35, CD36, CD37, CD41, CD42, CD43, CD44, CD45, CD46, CD47, CD48, CD49b, CD49c, CD53, CD54, CD55, CD58, CD59, CD61, CD62E, CD62L, CD62P, CD63, CD68, CD69, CD71, CD72, CD79, CD81, CD82, CD83, CD86, CD87, CD88, CD89, CD90, CD91, CD95, CD96, CD100, CD103, CD105, CD106, CD109, CD117, CD120, CD127, CD133, CD134, CD135, CD138, CD141, CD142, CD143, CD144, CD147, CD151, CD152, CD154, CD156, CD158, CD163, CD166, .CD168, CD184, CDwl86, CD195, CD202 (a, b), CD209, CD235a, CD271, CD303, CD304), Annexin Al, Nucleolin, Endoglin (CD105), ROB04, Amino-peptidase N, -like-4 (DLL4), VEGFR-2 (CD309), CXCR4 9CD184), Tie2, B7-H3, WT1, MUC1, LMP2, HPV E6 E7, EGFRvIII, HER-2/neu, Idiotype, MAGE A3, p53 nonmutant, NY-ESO-1, GD2, CEA,
MelanA/MARTl, Ras mutant, gplOO, p53 mutant, Proteinase3 (PR1), bcr-abl, Tyrosinase, Survivin, hTERT, Sarcoma translocation breakpoints, EphA2, PAP, ML-IAP, AFP, EpCAM, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, ALK, Androgen receptor, Cyclin B l, Polysialic acid, MYCN, RhoC, TRP-2, GD3, Fucosyl GMl , Mesothelin, PSCA, MAGE Al, sLe(a), CYPIB I, PLACl, GM3, BORIS, Tn, GloboH, ETV6-AML, NY-BR-1, RGS5, SART3, STn, Carbonic anhydrase IX, PAX5, OY-TES1, Sperm protein 17, LCK, HMWMAA, AKAP-4, SSX2, XAGE 1, B7H3, Legumain, Tie 2, Page4, VEGFR2, MAD- CT-1, FAP, PDGFR-β, MAD-CT-2, Fos-related antigen 1.
In another specific embodiment, the cell-binding -drug conjugates via the hydrophilic likers of this invention are used for the treatment of cancers. The 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,
Gestational Trophoblastic Tumor, Head and Neck Cancer, Hypopharyngeal Cancer, Islet Cell Carcinoma, Kidney Cancer (renal cell cancer), Laryngeal Cancer, Leukemia (Acute Lymphoblastic, Acute Myeloid, Chronic Lymphocytic, Chronic Myelogenous, Hairy Cell), Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer (Non-Small Cell, Small Cell, Lymphoma (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, Myeloproliferative 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 hydrophilic likers of this invention are used in accordance with the compositions and methods for the treatment 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 Spondylitis, 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 inflammatory 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, Complement 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, Dressler'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), Hypogammaglobulinemia, 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, Paroxysmal nocturnal hemoglobinuria, Parry Romberg syndrome,
Parsonnage-Turner syndrome, Pars planitis, Pemphigus, Pemphigus vulgaris, Pernicious anaemia, Perivenous encephalomyelitis, POEMS syndrome, Polyarteritis nodosa,
Polymyalgia rheumatica, Polymyositis, Primary biliary cirrhosis, Primary sclerosing cholangitis, Progressive inflammatory neuropathy, Psoriasis, Psoriatic Arthritis, Pyoderma gangrenosum, Pure red cell aplasia, Rasmussen's encephalitis, Raynaud phenomenon, Relapsing polychondritis, Reiter's syndrome, 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 hydrophilic linkers of this invention for the treatment or prevention of an autoimmune disease includes, 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-U.sub.lRNP; 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 antibody; Anti-p62 antibody; Anti-splOO antibody; Anti-Mitochondrial(M2) antibody; Rheumatoid 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 present invention, can bind to both a receptor or a receptor complex expressed on an activated 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, CD37, CD38, CD56, CD70, CD79, CD90, CD 125, CD152/CTLA-4, PD-1, or ICOS), a TNF receptor superfamily member (e.g. CD27, CD40, CD95/Fas, CD134/OX40, CD137/4-1BB, INF-R1, TNFR-2, RANK, TACI, BCMA, osteoprotegerin, Apo2/TRAIL-Rl, 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 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, polypeptide protein (e.g. HIV gpl20, HIV nef, RSV F glycoprotein, influenza virus neuramimidase, 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 bacterial, fungi, pathogenic protozoa, or yeast polypeptide 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 IgG.sub.l antibody for the treatment of cytomegalovirus; and anti-LPS antibodies.
The cell binding molecules-drug conjugates via the hydrophilic 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, 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 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 infection, Cholera, Chromoblastomycosis, Clonorchiasis, Clostridium difficile infection, Coccidioidomycosis, Colorado tick fever, Common cold (Acute viral rhinopharyngitis; Acute coryza), Creutzfeldt- Jakob disease, Crimean-Congo hemorrhagic fever, Cryptococcosis, Cryptosporidiosis, Cutaneous larva migrans, Cyclosporiasis, Cysticercosis, Cytomegalovirus infection, Dengue fever, Dientamoebiasis, Diphtheria,
Diphyllobothriasis, Dracunculiasis, Ebola hemorrhagic fever, Echinococcosis, Ehrlichiosis, Enterobiasis (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, Gnathostomiasis, 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, Hemolytic -uremic syndrome, Hemorrhagic fever with renal syndrome, Hepatitis A, Hepatitis B, Hepatitis C, Hepatitis D, Hepatitis E, Herpes simplex, Histoplasmosis, Hookworm infection, 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 pneumonia, 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, Rickettsialpox, Rift Valley fever, Rocky mountain spotted fever, Rotavirus infection, Rubella, Salmonellosis, 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 (Ringworm 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 pseudotuberculosis infection, Yersiniosis, Yellow fever, Zygomycosis.
The cell binding molecules, which are more proffered to be antibodies 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, Bacillus 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, Cryptococcus 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 B 19, Human herpesvirus 6 and Human herpesvirus 7, Fasciolopsis buski, Fasciola hepatica and Fasciola gigantica, FFI prion, Filarioidea superfamily, Clostridium 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, Helicobacter 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
metapneumo virus, Ehrlichia chaffeensis, Human papillomavirus, Human parainfluenza viruses, Hymenolepis nana and Hymenolepis diminuta, Epstein-Barr Virus,
Orthomyxoviridae 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,
Pneumocystis jirovecii, Poliovirus, Prevotella genus, Naegleria fowleri, JC virus,
Chlamydophila psittaci, Coxiella burnetii, Rabies virus, Streptobacillus moniliformis and Spirillum minus, Respiratory syncytial virus, Rhino sporidium seeberi, Rhinovirus, Rickettsia genus, Rickettsia akari, Rift Valley fever virus, Rickettsia rickettsii, Rotavirus, Rubella virus, Salmonella genus, SARS coronavirus, Sarcoptes scabiei, Schistosoma genus, Shigella genus, Varicella zoster virus, Variola major or Variola minor, Sporothrix schenckii, Staphylococcus 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 (Entomophthoramycosis), 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, Borrelia burgdorferi, Leptospira icterohemorrhagiae, Pneumocystis carinii, Brucella abortus, Brucella suis, Brucella melitensis, Mycoplasma spp., Rickettsia prowazeki, Rickettsia tsutsugumushi, Clamydia spp.; pathogenic fungi (Aspergillus fumigatus, Candida albicans, Histoplasma capsulatum); protozoa (Entomoeba histolytica, Trichomonas tenas, Trichomonas hominis, Tryoanosoma gambiense,
Trypanosoma rhodesiense, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Pneumocystis pneumonia, Plasmodium 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, Picornaviridae, 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)] ;
Central nervous system virus: [such as, JCV (Progressive multifocal leukoencephalopathy), MeV (Subacute sclerosing panencephalitis), LCV (Lymphocytic choriomeningitis), Arbovirus encephalitis, Orthomyxoviridae (probable) (Encephalitis lethargica), RV (Rabies), Chandipura virus, Herpesviral meningitis, Ramsay Hunt syndrome type II;
Poliovirus (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 mononucleosis), 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, Coronavirus; HBV (Hepatitis B virus), CBV, HAV (Hepatitis A virus), HCV (Hepatitis C virus), HDV (Hepatitis D virus), HEV (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 hydrophilic linkers of the invention together with a pharmaceutically acceptable carrier for treatment of cancer and autoimmune disorders. The method for treatment of cancer and autoimmune 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. Examples 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 malignant cells. For instance, clinical ex vivo treatment to remove tumour cells or lymphoid 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 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 incubation (=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. 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 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 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 50 μg to 20 mg/kg of body weight per week, i.v. (range of 10 μ g to 200 mg/kg per injection). 8 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 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 compound 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 administered 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 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. Preferably the unit dose range is from 1 to 500 mg administered one to four times a day, and even more preferably from 10 mg to 500 mg, once a day. 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 intranasally, 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 WO05058367A2, 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 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 including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide 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, 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). 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: (goserelin, leuprolide acetate); Anti- androgens: (bicalutamide, flutamide, calusterone, dromostanolone propionate, 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 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 .γΐ, δΐ, l and βΐ, 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 l-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 (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, 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) . An anti-infectious disease agent includes, but is not limited to, a).
Aminoglycosides: amikacin, astromicin, gentamicin (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). Glycopeptides: bleomycin, vancomycin (oritavancin, telavancin), teicoplanin (dalbavancin), ramoplanin; g). Glycylcyclines: e. g. tigecycline; g). β- 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; 1). Monobactams: aztreonam, tigemonam; m). Oxazolidinones: linezolid; n). Penicillins: amoxicillin, ampicillin (pivampiciUin, 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, 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).
Neuraminidase inhibitors: oseltamivir, zanamivir, peramivir; e). Nucleosides &
nucleotides: abacavir, aciclovir, adefovir, amdoxovir, apricitabine, brivudine, cidofovir, clevudine, dexelvucitabine, didanosine (ddl), 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. M-thymidine and M-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 hydrophilic linker of the present invention also include radioisotopes. Examples of radioisotopes (radionuclides) are 3H, UC, 14C, 18F,
32^ P, 35 Sc, 64^ Cu, 68^ Ga, 86v Y, 99^ lc, 111T In, 123T 124T 125T 131T 133v
1, 1, 1, Xe, 177T Lu, 211 A. t. , or 213D Bi- 1, .
Radioisotope 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 hydrophilic 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, DTP A and TETA (Macrocyclics, Dallas, Tex.).
6) . The pharmaceutically acceptable salts, acids or derivatives of any of the above drugs.
Preferred cytotoxic agents that conjugated to a cell-binding molecule via a hydrophilic 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 oxazolidinobenzodiazepines), 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, esperamicins, and their analogues and derivatives 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. 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, 4362^1369. Pando, O.; 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, 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., 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 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; WO2004005269. Stanton, M.; et al, U.S. Pat. Appl. Publ. 20040249130. Hoefle, G.; et al, Ger. Offen. DE 10254439 ; DE 10241152; DE 10008089. Leung, D.; et al, PCT 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 described in the patent application of
PCT/IB2012/053554
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.
Maytansinoids that are preferred to be used in the present invention including maytansinol and maytansinol analogues are described in U.S. Patent Nos. 4,256,746, 4,361,650 and 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, 8,163,888 .
Taxanes, which includes Paclitaxel (Taxol), a cytotoxic natural product, and docetaxel (Taxotere), a semi-synthetic derivative, and their analogs which are preferred for conjugation via the hydrophilic 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. ScL , 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; 7,667,054.
CC-1065 analogues and doucarmycin analogs are also preferred to be used for a conjugate with the hydrophilic linkers of the present patent. The examples of the CC-1065 analogues 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, 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, 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,
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, 8,012,978. Daunorubicin/Doxorubicin Analogues are also preferred for conjugation via the hydrophilic linkers of the present patent. The preferred structures and their synthesis are exampled 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.l 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.
Auristatins and dolastatins are preferred in conjugation via the hydrophilic 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.
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;
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;
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.
Analogues and derivatives of the cytotoxic drugs/agents described in the present patent can be conjugated via a hydrophilic linker of 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), unless otherwise specified. Cell culture reagents were obtained 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. NMR spectra were recorded on Varian Mercury 300 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. Low resolution mass spectral data were acquired on a Waters Micromass ZMD mass spec with Waters 2795 HPLC separations module and the 2996 photodiode array detector.
Example 1: (2-Bromoethyl)(3-ethoxy-3-oxopropyl)phosphinic acid (or ethyl 3- [2- Bromoethyl(hydroxy)phosphinyl]propanoate) (4)
O O OH
A mixture of ammonium hypophosphite (8.00 g, 96 mmol) and
hexamethydisilazane (20.0 mL, 96 mmol) was heated at 120 °C for 1 h under argon. After the mixture was cooled to 0 °C, ethyl acrylate (10.4 mL, 96 mmol) was carefully added dropwise, and the resulting mixture was stirred at 50 °C for 2 h. Then the mixture was cooled to room temperature, dibromoethane (40.0 mL) was added, and the mixture was stirred for 5 h at 120 °C. The formed trimethylbromosilane and excess dibromoethane were removed under vacuum. Then 100 mL of aqueous ethanol (1 : 1) were added dropwise to the residue and refluxed for 0.5 h. Then the solvent was removed under vacuum and extracted with ethyl acetate. The organic layer was dried over magnesium sulfate and the solvent was removed under vacuum to give the title compound 4 (10.85 g, 41 % yield). ιΐί NMR (300 MHz, CD3OD): δ 1.26 (t, J = 7.1 Hz, 3H), 2.07 (m, 2H), 2.42 (m, 2H), 2.62 (m, 2H), 3.59 (m, 2H), 4.15 (q, J = 7.1 Hz, 2H). 31P NMR (100 MHz, CD3OD): δ 49.5; ESI MS m/z- C7H13Br04P (M-H), cacld. 271.98, found 271.97.
Example 2. Ethyl 3-[2-Bromoethyl(ethoxy)phosphinyl]propanoate (5) and Ethyl 3- [Ethoxy( vinyl) phosphinyl]propanoate (6).
Figure imgf000052_0001
An amount of 10.84 g of 4 (20 mmol) was treated with 100.0 mL of triethyl orthoformate, and the mixture was refluxed with a Dean-Stark trap to remove ethanol and ethyl formate. Excess triethyl orthoformate was removed under vacuum to give 5 and 6 ([39.2:60.8 31P NMR ratio], 11.83 g). 6: *H NMR (300 MHz, CD3OD) δ 1.27 (m, 6H), 2.19 (m, 2H), 2.57 (m, 2H), 4.11 (m, 4H), 6.36 (m, 3H). 31P NMR (100 MHz, CD3OD): δ 44.9; 5: 31P NMR (100 MHz, CD3OD) δ 53.3; ESI MS m/z+, 5: 323.01 (M + Na), 6: 243.09 (M + Na). Example 3. (2-bromoethyl)(2-(ethoxy(3-ethoxy-3-oxopropyl)phosphoryl)ethyl)phosphinic acid (8)
Figure imgf000053_0001
A mixture of ammonium hypophosphite (8.00 g, 96 mmol) and hexamethy- disilazane (20.0 mL, 96 mmol) was heated at 120 °C for 1 h under argon. After the mixture was cooled to 0 °C, ethyl acrylate (10.4 mL, 96 mmol) was carefully added dropwise, and the resulting mixture was stirred at 50 °C for 2 h. Then the mixture of compound 5 and 6 (10.0 g, 38.4 mmol estimated from above ratio) was added and heated at 120 °C for 2 h under argon, followed by addition of 1,2-dibromoethane (40 ml) and the mixture was stirred for 5 h at 120 °C. After the solvent was removed under vacuum, 100 mL of aqueous ethanol (1: 1) were added dropwise to the residue and refluxed for 0.5 h. The mixture was concentrated and purified by S1O2 chromatographic column (1 :20 to 1 : 10 MeOH/CH2Cl2) to afford the title compound 8 (6.48 g,43%% yield). ESI MS m/z- 391.2 (M - H).
Example 4. (2-(ethoxy(3-ethoxy-3-oxopropyl)phosphoryl)ethyl)(2- ((ethoxycarbonothioyl)thio)ethyl)phosphinic acid (9).
Figure imgf000053_0002
Compound 8 (6.01 g, 15.30 mmol) in 150 ml of ethanol was added Potassium ethyl xanthogenate (3.00 g, 18.75 mmol).. After being stirred under Ar for 3 h, the mixture was acidified with 3 M H3PO4 to pH 3.0. The mixture was concentrated, and purified on a S1O2 column eluted with water/MeCN/HOAc (1: 10:0.01), pooled the fraction, added DMF (~5 ml), evaporated to dryness to afford the title compound 9
(5.38 g, 83% yield). ESI MS, m/z- 433.10 (M-H).
Example 5. 3-(Hydroxy(2-(hydroxy(2-(pyridin-2- yldisulfanyl)ethyl)phosphoryl)ethyl)phosphoryl)propanoic acid (10)
Figure imgf000053_0003
Compound 9 (5.00 g, 11.51 mmol) in 100 ml of methanol was added 50 ml of 3 M NaOH. After being stirred under Ar for 3 h, the mixture was neutralized with 3 M H3P04 to pH 7.2 under Ar. The mixture was added dropwise to the solution of 1 ,2-bis(pyridin-2- yl)disulfane (10.0 g, 45.45 mmol) in 200 ml of methanol. After being stirred for 4 h under Ar, the mixture was concentrated, diluted with EtOAc/Hexane (1 : 1), separated, and the organic layer was washed with pure water (3 x 25 ml) while the generated each of aqueous layer was washed with EtOAc/Hexane (1 :1, 35 ml). The aqueous layers were combined, acidified with HCl/HOAc to pH ~2, concentrated to ~ 10 ml, diluted with MeCN (60 ml), sonicated (or quickly stirred) for 1 h, filtered, washed the pellet with water/MeCN (1 : 10). The solution was then concentrated and purified on a S1O2 column eluted with
water/MeCN/HOAc (1 :8:0.01), pooled the fraction, added DMF (~5 ml), evaporated to dryness to afford the title compound 10 (3.62 g, 79% yield). ESI MS, m/z- 398.02 (M-H). Example 6. (3-((2,5-dioxopyrrolidin-l-yl)oxy)-3-oxopropyl)(2-(hydroxy(2-(pyridin-2- yldisu id (11).
Figure imgf000054_0001
Compound 10 (200 g, 5.01 mmol) in DMA (50 ml) was added 0.2 ml of HC1 (cone) and the mixture was evaporated to dryness. Then the compound redissolved in dry DMA (60 ml) was added, NHS (0.80 g, 6.95 mmol) and EDC (3.00 g, 15.62 mmol). The mixture was stirred under Ar overnight, evaporated and purified on short C-18 chromatography eluted with water/dioxane at 4°C. The fractions containing the product were pooled, freezed at -78°C, lyophilized to afford the title compound (1.26 g, 51% yield). MS m/z- 495.2 (M-H).
Examp -endoxo- -tetrahydrophthalhide
Figure imgf000054_0002
Maleimide (10.0 g, 103.0 mmol) in ethylether (350 ml) was added furan (11.0 ml, 151.2 mmol). The mixture was heated inside a 1 L of autoclave bomb at 100°C for 8 h. The bomb was cooled down to room temperature, and the inside solid was rinsed with methanol, concentrated and crystallized in ethyl acetate/hexane to afford 16.9 g (99%) of the title compound. lU NMR (DMF-d7, 300 MHz): 11.06 (s, 1H) (NH), 6.61 (m, 2H), 5.15 (m, 2H), 2.97 (m, 2H). 13C NMR 178.86, 137.72, 82.05, 49.93. MS m/z+ 188.4 (M + Na). Example 8. Ethyl 3-((2-((2-bromoethyl)(ethoxy)phosphoryl)ethyl)(ethoxy)phosphoryl) propanoate (13), Ethyl 3-(ethoxy(2-(ethoxy(vinyl)phosphoryl)ethyl)phosphoryl) propanoate (14).
Figure imgf000055_0001
Compound 8 (4.51 g, 11.47 mmol) was treated with 100.0 mL of triethyl orthoformate, and the mixture was refluxed with a Dean-Stark trap to remove ethanol and ethyl formate. Excess triethyl orthoformate was removed under vacuum to give the mixture of 13 and 14. ESI MS m/z+, 13: 443.10 (M + Na), 14: 363.20 (M + Na).
Example 9. Ethyl 3-((2-((2-(3, 6-endoxo-A- tetrahydrophthalido)ethyl)(ethoxy)phosphoryl)ethyl)(ethoxy)phosphoryl)propanoate, or ethyl 3-((2-((2-((3aR,4R,7S)-l,3-dioxo-3a,4,7,7a-tetrahydro-lH-4,7-epoxyisoindol-2(3H)- yl)ethyl)(ethoxy)phosphoryl)ethyl)(ethoxy)phosphoryl)propanoate (24)
Figure imgf000055_0002
3, 6-Endoxo- Δ-tetrahydrophthalhide (2.40 g, 14.55 mmol) in DMA (60 ml) was added K2C03 (4. 2 g, 30.39 mmol) and KI (0.40 g, 3.45 mmol). After stirring under Ar for 1 hr, the mixture of compound 13 and 14 (2.6 g, -6.80 mmol estimated) in DMA (10 ml) was added. The mixture was stirred under Ar overnight, evaporated, re-dissolved in EtOAc (100 ml), washed with water (2 x 50 ml) and 1.0 M NaH2P04 (2 x 50 ml), dried over Na2S04, filtered, evaporated and purified by Si02 chromatography eluted with
EtOAc/hexane (1 :10 ~ 1:5) to afford the title compound (2.64 g, 77% yield). ESI MS m/z+ 528.60 (M + Na).
Example 10. 3-((2-((2-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l- yl)eth droxy)phosphoryl)propanoic acid (25)
Figure imgf000055_0003
Compound 24 (2.60 g, 5.14 mmol), in the mixture of DMA (20 ml), toluene (20 ml) and HC1 (8N,10 ml) was heated at 120 ~ 140°C for 8 h. During the reaction time, 5 x 10 ml of water was gradually added to keep the reaction volume around 40 ml. The mixture was concentrated and purified by C-18 chromatography eluted with (1 : 10:0.01 to 1 :3:0.01) water/CH3CN/HOAc to afford the title compound (1.12 g, 62% yield). ESI MS m/z- 352.10 (M - H).
Example 11. (2-((2-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l- yl)ethyl)(hydroxy)phosphoryl)ethyl)(3-((2,5-dioxopyrrolidin-l-yl)oxy)-3- oxopropyl)phosphinic acid (26)
Figure imgf000056_0001
Compound 25 (1.10 g, 3.11 mmol) in DMA (50 ml) was added 0.1 ml of HC1 (cone) and the mixture was evaporated to dryness. Then the compound redissolved in dry DMA (40 ml) was added NHS (0.41 g, 3.56 mmol) and EDC (2.00 g, 10.42 mmol). The mixture was stirred under Ar overnight, evaporated and purified on S1O2 chromatography eluted with 1 : 1: 1% Acetone/DCM/HOAc, pooled the fractions, evaporated and solidified with EtOH/Tol/Hexane to afford the title compound (712 mg, 51% yield). ESI MS m/z- 449.10 (M-H).
Example 12. methyl 3-((2-bromoethyl)thio)propanoate (52)
Figure imgf000056_0002
A mixture of methyl 3-mercaptopropanoate (5.20 g, 51.6 mmol) and 1,2- dibromoethane (30 ml, 348.1 mmol) in DIPEA (100 ml) was stirred at 45°C for 8 h. The mixture was concentrated under vacuum and purified on S1O2 eluted with EtOAc/Hexane (1 : 12 to 1 :5) to give the title compound 52 (9.09 g, 78% yield). ESI MS m/z- 249.20 (M +
Na).
Example 13. methyl 3-((2-bromoethyl)sulfonyl)propanoate (53)
Figure imgf000057_0001
Methyl 3-((2-bromoethyl)thio)propanoate 52 (4.50 g, 19.91 mmol) in acetic acid (40 ml), H202 (30%, 20 ml) and KMn04 (l.OOg, 6.33 mmol) was stirred overnight. The mixture was concentrated, diluted with EtOAc (100 ml) and 1 M NaH2P04 (150 ml), separated, the aqueous layer was extracted with EtOAc (2 x 100 ml). The organic layers were combined, dried over Na2S04, filtered, concentrated and purified on Si02 column eluted with EtOAc/Hexane (1 :10 ~ 1:5) to afford the title compound (4.05g, 79% yield). ESI MS m/z+281.02 (M + Na).
Example 14. 3-(Vinylsulfonyl)propanoic acid (54)
Figure imgf000057_0002
The compound 53 (4.0 g, 15.50 mmol) in 50 ml of THF was added 1 M NaOH (50 ml). After stirred under Ar for 24 h, the mixture was adjusted pH = ~ 7 with 1 M HC1 at 4°C, concentrated, diluted with EtOAc (100 ml), separated. The aqueous layer was extracted with EtOAc (4 x 80 ml). The organic layers were combined, ,, dried over Na2S04, filtered, evaporated, and Si02 chromatographic purification (1 :15 to 1 :10 MeOH/CH2C12) to afford the title compound 54 (1.80 g, 71% yield). ESI MS m/z- 163.10 (M-H).
Examp -dioxopyrrolidin-l-yl 3-(vinylsulfonyl)propanoate (55)
Figure imgf000057_0003
Compound 54 (1.70 g, 10.36 mmol) in DMA (50 ml) was added NHS (1.75 g, 15.21 mmol) and EDC (5.00 g, 26.04 mmol). The mixture was stirred under Ar overnight, evaporated and purified on Si02 chromatography eluted with EtOAc/Hexane (1 : 10 ~ 1 :4), pooled the fractions and evaporated to afford the title compound (2.24g, 83% yield). ESI MS m/z+ 284.10 (M+Na). Example 16. methyl 3-((3-mercaptopropyl)thio)propanoate (64).
Figure imgf000058_0001
Compound 63 (10.010 g, 60.25 mmol) in DMA (40 ml) was added propane-1,3- dithiol (40.0 g, 370.3 mmol) and DIPEA (100 ml). The mixture was stirred at 45 °C under Ar for 8 h, evaporated and purified on Si02 chromatography eluted with 1: 10:0.01%
EtOAc/DCM/HOAc, pooled the fractions, and evaporated to afford the title compound (9.58 g, 82% yield). ESI MS m/z+ 217.2 (M+Na).
Example 17. Methyl 3-((3-((3-bromopropyl)thio)propyl)thio)propanoate (65)
Figure imgf000058_0002
Compound 64 (5.01 g, 25.76 mmol) in DMA (40 ml) was added 1,3- dibromopropane (30.0 g, 150.0 mmol) and DIPEA (100 ml). The mixture was stirred at 45 °C under Ar for 8 h, evaporated and purified on Si02 chromatography eluted with
EtOAc/hexane (1 :10 to 1:5), pooled the fractions, evaporated to afford the title compound 65 (6.87 g, 85% yield). ESI MS m/z+ 337.2 (M+Na).
Example 18. methyl 3-((3-((3-bromopropyl)sulfonyl)propyl)sulfonyl)propanoate (66)
O O O o o
Compound 65 (6.70 g, 21.33 mmol) was in acetic acid (40 ml) was added H202 (33%, 20 ml) and KMn04 (1.01 g, 6.33 mmol). The mixture was stirred at 40 °C overnight. The mixture was concentrated, diluted with EtOAc (100 ml) and 1 M NaH2P04 (100 ml), separated. The aqueous layer was extracted with EtOAc (2 x 80 ml). The organic layers were combined, dried over Na2S04, filtered, evaporated, purified by Si02 chromatography eluted with EtOAc/hexane (1: 10 ~ 1 :4) to afford the title compound (6.69 g, 83% yield). ESI MS m/z+ 401.10 (M + Na).
Example 19. methyl 3-((3-((3-(acetylthio)propyl)sulfonyl)propyl)sulfonyl)propanoate (67)
Figure imgf000059_0001
Compound 66 (6.60 g, 17.46 mmol) in 100 ml of THF was added thioacetic acid (3.0 ml, 41.97 mmol) and DIPEA (20 ml, 115.0 mmol). The mixture was stirred at 40°C overnight, evaporated, co-evaporated with acetic acid (5 ml)/toluene (200 ml), and purified with S1O2 chromatography eluted with EtOAc/Hexane ( 1 : 10 to 1 :4) to afford the title compound (5.22 g, 80% yield). ESI MS m/z+ 397.10 (M+Na).
Example 20. 3-((3-((3-(pyridin-2-yldisulfanyl)propyl)sulfonyl)propyl)sulfonyl)propanoic acid (6
Figure imgf000059_0002
Compound 67 (5.20 g, 13.90 mmol) in 100 ml of methanol was added 50 ml of 3 M NaOH. After being stirred under Ar for 3 h, the mixture was neutralized with 3 M H3PO4 to pH 7.2 under Ar. The mixture was added dropwise to the solution of l,2-bis(5-pyridin-2- yl)disulfane (15.0 g, 68.18 mmol) in 200 ml of methanol. After being stirred for 24 h under Ar, the mixture was concentrated, diluted with EtOAc/Hexane (1 : 1), separated, and the organic layer was washed with pure water (3 x 25 ml) while the generated each of aqueous layer was washed with EtOAc/Hexane (1 :1, 35 ml). The aqueous layers were combined, acidified with HCl/HOAc to pH 3 ~ 4, concentrated to ~ 10 ml, diluted with MeCN (60 ml), sonicated (or quickly stirred) for 1 h, filtered, washed the pellet with water/MeCN (1 : 10). The solution was then concentrated and purified on a S1O2 column eluted with CH3 OH/CH2CI2/HO Ac (1 : 10:0.01), pooled the fraction, evaporated to dryness to afford the title compound 68 (5.93 g, 82% yield). ESI MS, m/z- 426.10 (M-H).
Example 21. 2,5-dioxopyrrolidin-l-yl 3-((3-((3-(pyridin-2- yldi e (69).
Figure imgf000059_0003
Compound 68 (2.50 g, 6.08 mmol) in DMA (50 ml) was added NHS (0.80 g, 6.96 mmol) and EDC (3.00 g, 15.62 mmol). The mixture was stirred under Ar overnight, evaporated and purified on S1O2 chromatography eluted with EtOAc/DCM (1 :10 to 1:5), pooled the fractions and evaporated to afford the title compound (2.74 g, 86% yield). ESI MS m/z+ 547.10 (M+Na).
Example 22. methyl 3-((3-mercaptopropyl)thio)propanoate (71).
O
Methyl 3-bromopropanoate 51 (10.010 g, 60.24 mmol) in DMA (80 ml) was added ethane- 1,2-dithiol (40.0 g, 425.4 mmol) and DIPEA (150 ml). The mixture was stirred at 45 °C under Ar for 8 h, evaporated and purified on S1O2 chromatography eluted with 1 : 10:0.01% EtOAc/DCM/HOAc, pooled the fractions, and evaporated to afford the title compound 71 (8.56 g, 79% yield). ESI MS m/z+ 203.10 (M+Na).
Example 23. methyl 3-((2-((3-bromobutyl)thio)ethyl)thio)propanoate (82).
Figure imgf000060_0001
Compound 71 (8.51 g, 47.26 mmol) in DMA (40 ml) was added 1,3-dibromobutane (30.0 g, 140.25 mmol) and DIPEA (100 ml). The mixture was stirred at 45 °C under Ar for 8 h, evaporated and purified on S1O2 chromatography eluted with EtOAc/hexane (1: 10 to 1 :5), pooled the fractions, evaporated to afford the title compound 82 (12.16 g, 82% yield). ESI MS m/z+ 337.2 (M+Na).
Exa yl)sulfonyl)ethyl)sulfonyl)propanoate (83).
Figure imgf000060_0002
Compound 82 (6.00 g, 19.10 mmol) was in acetic acid (40 ml) was added H2O2 (33%, 20 ml) and KMn04 (1.01 g, 6.33 mmol). The mixture was stirred at 40 °C overnight. The mixture was concentrated, diluted with EtOAc (100 ml) and 1 M NaH2P04 (100 ml), separated. The aqueous layer was extracted with EtOAc (2 x 80 ml). The organic layers were combined, dried over Na2S04, filtered, evaporated, purified by S1O2 chromatography eluted with EtOAc/hexane (1: 10 ~ 1 :4) to afford the title compound 83 (6.01 g, 83% yield). ESI MS m/z+ 401.02 (M + Na). Examp io)butyl)sulfonyl)ethyl)sulfonyl)propanoate (84).
Figure imgf000061_0001
Compound 83 (6. 0 g, 15.87 mmol) in 100 ml of THF was added thioacetic acid (3.0 ml, 41.97 mmol) and DIPEA (20 ml, 115.0 mmol). The mixture was stirred at 50°C overnight, evaporated, co-evaporated with acetic acid (5 ml)/toluene (200 ml), and purified with S1O2 chromatography eluted with EtOAc/Hexane ( 1 : 10 to 1 :4) to afford the title compound 84 (4.27 g, 72% yield). ESI MS m/z+ 397.20 (M+Na).
Example 26. 3-((2-((3-(pyridin-2-yldisulfanyl)butyl)sulfonyl)ethyl)sulfonyl)propanoic acid
(85).
Figure imgf000061_0002
Compound 84 (2.10 g, 5.61 mmol) in 100 ml of methanol was added 50 ml of 2 M NaOH. After being stirred under Ar for 1 h, the mixture was neutralized with 3 M H3PO4 to pH 7.2 under Ar. The mixture was added dropwise to the solution of l,2-bis(5-pyridin-2- yl)disulfane (6.0 g, 27.27 mmol) in 100 ml of methanol. After being stirred for 15 h under Ar, the mixture was concentrated, diluted with EtOAc/Hexane (1 : 1), separated, and the organic layer was washed with pure water (3 x 25 ml) while the generated each of aqueous layer was washed with EtOAc/Hexane (1 :1, 35 ml). The aqueous layers were combined, acidified with HCl/HOAc to pH 3 ~ 4, concentrated to ~ 10 ml, diluted with MeCN (60 ml), sonicated for 1 h, filtered, washed the pellet with water/MeCN (1 :10). The solution was then concentrated and purified on a S1O2 column eluted with CH3OH/CH2CI2/HOAC (1 : 10:0.01), pooled the fraction, evaporated to dryness to afford the title compound 85 (1.98 g, 82% yield). ESI MS, m z- 426.10 (M-H).
Example 27. 2,5-dioxopyrrolidin-l-yl 3-((2-((3-(pyridin-2-yldisulfanyl)butyl)sulfonyl)- ethyl)
Figure imgf000061_0003
Compound 85 (1.00 g, 2.34 mmol) in DMA (50 ml) was added NHS (0.41 g, 3.56 mmol) and EDC (2.00 g, 10.42 mmol). The mixture was stirred under Ar overnight, evaporated and purified on Si02 chromatography eluted with EtOAc/DCM (1 : 10 to 1 :5), pooled the fractions and evaporated to afford the title compound 86 (993 mg, 81 % yield). ESI MS m/z+ 547.10 (M+Na).
Example 28. Modification of antibody with phosphinate linker
The antiHer2 antibody is modified with phosphinate linker at 8 mg/mL antibody, a 10 fold molar excess of phosphinate linker (~30mM stock solution in DMA). The reaction is carried out in 100 mM NaH2P04, pH7.4 buffer with DMA (5% v/v) for 15, 30, 60, 120, and 240 minutes at 25 °C. The modified antiHer2 was purified by G25 column with 50 mM NaH2P04, 50 mM NaCl, and 2 mM EDTA, pH6.5 to remove the excess phosphinate linker. Example 29: Conjugate synthesis.
A hydrophilic linker containing thiopyridine (SPP) linker was dissolved in DMA at a concentration of approximately 10 mM. An antibody was dialyzed into buffer A (50 mM NaH2P04, 50 mM NaCl, 2 mM EDTA, pH 6.5). For the linker reaction, the antibody was at 8 mg/ml, and 4-6 equivalents of linker were added while stirring in the presence of 5% (v/v) DMA. The reaction was allowed to proceed at ambient temperature for 90 minutes. Unreacted linker was removed from the antibody by Sephadex G25 gel filtration using a Sephadex G25 column equilibrated with Buffer A at pH 6.5 or 150 mM potassium phosphate buffer containing 100 mM NaCl, pH 7.4 as indicated. For the SPP containing linker, the extent of modification was assessed by release of pyridine-2-thione using 50 mM DTT and measuring the absorbance at 343 nm as described below (ε343 = 8080 M"1 cm"1 for free pyridine-2-thione). For the conjugation reaction, thiol-containing drug (such tubulysin TZ041) was dissolved in DMA (N, N-dimethylacetamide) at a concentration of approximately 10 mM. The drug (1 - 1.5-fold molar excess relative to the number of linker molecules per antibody as indicated) was slowly added with stirring to the antibody which was at a concentration of 2.5 mg/ml in buffer A (pH 6.5 or pH 7.4) in a final concentration of 3% (v/v) DMA. The reaction was allowed to proceed at ambient temperature for the indicated times. Drug-conjugated antibody was purified using a Sephadex G25 column equilibrated with buffer B (PBS (NaH2P04), pH 6.5). The extent of drug conjugation to antibody was assessed by measuring A254 and A2so of the conjugate.. Example 30. In vitro cytotoxicity evaluation of a tubulysin (ZT041) conjugates of antibodies with a disulfide linkers containing sulfonyl groups:
The targeted cells (e.g. Ramos cells, 20,000 cells) were cultured in the presence of various concentrations of unconjugated antibody or the antibody conjugate for 96 hours after which cell viability was measured by propidium iodide exclusion and analyzed by flow cytometry using a Becton Dickinson FACSort (Becton Dickinson, Franklin Lakes, NJ). Red fluorescent intensity (emission at 617 nm in the FL2 channel) of the cells excited at 488 nm was measured. The regions for viable cells were also set using both the forward light scatter and right-angle light scatter properties of the cells. The loss of viability was determined by the loss of cells from within the gated region defining viable cells. The average number of viable cells per 6 replicate cultures was calculated. The survival fraction was plotted versus conjugate concentration to determine the / 50 value (50% cell killing concentration) of the conjugate.
REFERENCE
U.S. Patent Nos. 4,680,338, 5,122,368, 5,141,648, 5,208,020, 5,416,064; 5,475,092,
5,543,390, 5,563,250 5,585,499, 5,880,270, 6,214,345, 6,436,931, 6,372,738, 6,340,701, 6,989,452, 7,129,261, 7,375,078, 7,498,302, 7,507,420, 7,691,962, 7,910,594, 7,968,586, 7,989,434, 7,994,135, 7,999,083, 8,153,768, 8,236,319.
US Patent Application Nos. 20120201809, 20120082617, 20120034295,
20110293513, 20110076722, 20110064753, 20110064752, 20110064666, 20100189727, 20100136652, 20100104589, 20100074840, 20100062008, 20090176253, 20090088390 Albrecht, H.; et al. J Immunol Methods 2006, 310, 100.
Alley, S. C; et al. Curr Opin Chem Biol 2010, 14, 529.
Anderson, D. C; et al. Bioconjug Chem 1993, 4, 10.
Antczak, C; et al. Bioconjug Chem 2006, 17, 1551.
Aoki, S.; et al. Bioorg Med Chem 2009, 17, 3405.
Austin, C. D.; et al. Proc Natl Acad Sci U S A 2005, 102, 17987.
Barbour, N. P.; et al. Pharm Res 1995, 12, 215.
Beeson, C; et al. Bioconjug Chem 2003, 14, 921.
Bickel, U.; et al. Bioconjug Chem 1995, 6, 211.
Chen, J.; et al. Expert Opin Drug Deliv 2005, 2, 873. DiJoseph, J. F.; et al. Blood 2004, 103, 1807.
Doronina, S. O.; et al. Bioconjugate Chem., 2006, 17, 114. Doronina, S. O.; et al. Bioconjug Chem 2008, 19, 1960. Ebner, A.; et al. Bioconjug Chem 2007, 18, 1176.
Erickson, H. K.; et al. Bioconjug Chem 2010, 21, 84. Garsky, V. M.; et al. J. Med. Chem., 2001, 4216.
Greenwald, R. B.; et al. J. Med. Chem., 1999, 42, 3657. Haenseler, E.; et al. Biochemistry, 1992, 31, 891.
Hamann, P. R.; et al. Bioconjugate Chem., 2002, 13, 40. Hamann, P. R.; et al. Bioconjugate Chem., 2005, 16, 354. Jeffrey, S. C; et al. J. Med. Chem., 2005, 48, 1344 Johnson, D. A.; et al. Cancer Res 1991, 51, 5774.
Jones, D. S.; et al. Bioconjug Chem 2001, 12, 1012.
Jones, D. S.; et al. Bioconjug Chem 1999, 10, 480.
Jones, D. S.; et al. Bioconjug Chem 1994, 5, 390.
Kashef, N.; et al. J Med Microbiol 2006, 55, 1441.
Kellogg, B. A.; et al. Bioconjug Chem 2011, 22, 111. Kelly, R. K.; et al. Eur J Cancer 2011, 47, 1736.
King, H. D.; et al. Bioconjug Chem 1999, 10, 279.
King, H. D.; et al, J. Med. Chem. , 2002, 45, 4336
Klussman, K.; et al. Bioconjug Chem 2004, 15, 765. Kovar, M.; et al. Bioconjugate Chem., 2002, 13, 206. Kratz, F., et al. J. Med. Chem. , 2002, 45, 5523.
Kumaresan, P. R.; et al. Bioconjug Chem 2008, i9, 1313. Kumaresan, P. R.; et al. Bioconjug Chem 2007, iS, 175. Lee, L. S.; et al. Bioconjug Chem 1999, 10, 973.
Li, L.; et al. Bioconjug Chem 2002, 13, 985.
Lipinski, T.; et al. Glycoconj J 2011, 28, 149.
Meyer-Losic, F.; et al. J. Med. Chem., 2006, 49, 6908 Mikolajczyk, S. D.; et al. Bioconjug Chem 1994, 5, 636. Miller, M. L.; et al. J. Med. Chem., 2004, 47, 4802 Mitchell, J. S.; et al. Bioconjug Chem 2007, 18, 268.
Moon, S.-J.; et al. J. Med. Chem. , 2008, 51, 6916
Ojima, I.; et al. J. Med. Chem., 2002, 45, 5620
Ruppert, C; et al. Bioconjug Chem 2002, 13, 804.
Safavy, A.; et al. Bioconjug Chem 2003, 14, 302.
Safavy, A.; et al. Bioconjug Chem 2004, 15, 1264.
Senter, P. et al. Photochem. Photobio., 1985, 42, 231.
Scott, C. F., Jr.; et al. J Natl Cancer Inst 1987, 79, 1163.
Sharkey, R. M.; et al. Mol Cancer Ther 2012, 11, 224.
Siiman, O.; et al. Bioconjugate Chem., 2000, 11, 549.
Skwarczynski, M.; et al. J. Med. Chem. , 2006, 49, 7253
Srinivasachar, K.; Neville, D. M., Jr. Biochemistry 1989, 28, 2501.
Studer, M.; et al. Bioconjug Chem 1992, 3, 424.
Sun, X.; et al. Bioconjug Chem 2011, 22, 728.
Suzawa, T.; et al. Bioorg Med Chem 2000, 8, 2175.
Tadayoni, B. M.; et al. Bioconjug Chem 1993, 4, 139.
ten Hoeve, W.; et al. Bioconjug Chem 1997, 8, 257.
Tsai, N. M.; et al. Biotechniques 2001, 30, 396.
Walker, M. A.; rt al. Bioorg Med Chem Lett 2004, 14, 4323.
Wilbur, D. S.; et al. Bioconjug Chem 2011, 22, 1089.
Widdison, W. C; et al. J. Med. Chem., 2006, 49, 4392.
Zhao, R. Y.; et al. J. Med. Chem., 2011, 54, 3606
Zhao, R. Y.; et al. J. Med. Chem., 2012, 55, 766

Claims

What is claimed is:
1. A hydrophilic linker of the formula (I)
Figure imgf000066_0001
(I)
Wherein:
Y represents a functional group that enables reaction with a cell-binding agent;
Q and T are either -P(=0)(OM)-, or -S(02)-, or -S(O)-;
m and n are integer from 0 to 5, but not 0 at the same time; In addition, when m = 1, n = 0, Q is not -P(=0)(OM)-; or when n = 1 , m = 0, T is not -P(=0)(OM)-;
Z represents a functional group that enables linkage of 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;
Pvi, P2, P3, PM, Re and Re, are the same or different and are 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 (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
Additionally Ri, R2, R3 and R4 are respectively a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl group, the conjugated drug and among themselves (Ri, R2, R3 and R4). The atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxy lamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
M is H, or Na, or K, or N+RiR2R3 or a pharmaceutical salt. Ri, R2 and R3 are described above.
2. -binding agent-drug conjugate of formula (II)
Figure imgf000066_0002
wherein:
Cb represents a cell-binding agent/molecule; Drug represents the drug linked to the cell-binding agent via the hydrophilic linker by a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
Q and T are either -P(=0)(OM)-, or -S(02)-, or -S(O)-;
m and n are integer from 0 to 5, but not 0 at the same time; In addition, when m = 1, n = 0, Q is not -P(=0)(OM)-; or when n = 1, m = 0, T is not -P(=0)(OM)-;
Ri, P2, P3, PM, Re and Re, are the same or different and are 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 (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
Additionally Ri, R2, R3 and R4 are respectively a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl group, the conjugated drug and among themselves (Ri, R2 , R3 and R4). The atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxy lamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
M is H, or Na, or K, or N+RiR2R3 or a pharmaceutical salt. Ri, R2 and R3 are described above.
3.
Figure imgf000067_0001
wherein:
Cb represents a cell-binding agent/molecule;
Z represents a functional group that enables reaction with a cytotoxic drug;
Q and T are either -P(=0)(OM)-, or -S(02)-, or -S(O)-;
m and n are integer from 0 to 5, but not 0 at the same time; In addition, when m = 1, n = 0, Q is not -P(=0)(OM)-; or when n = 1, m = 0, T is not -P(=0)(OM)-; Z represents a functional group that enables linkage of 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;
Ri, R2, R3, R4, R5 and Re, are the same or different and are 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 (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof.
Additionally Ri, R2, R3 and R4 are respectively a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl group, the conjugated drug and among themselves (Ri, R2, R3 and R4). The atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxy lamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
M is H, or Na, or K, or N+RiR2R3 or a pharmaceutical salt. Ri, R2 and R3 are described above.
4.
Figure imgf000068_0001
(IV)
Wherein:
Y represents a functional group that enables reaction with a cell-binding agent;
Q and T are either -P(=0)(OM)-, or -S(02)-, or -S(O)-;
m and n are integer from 0 to 5, but not 0 at the same time; In addition, when m = 1, n = 0, Q is not -P(=0)(OM)-; or when n = 1, m = 0, T is not -P(=0)(OM)-;
" Drug" represents the drug linked to the cell-binding agent via the hydrophilic linker by a disulfide, thioether, thioester, peptide, hydrazone, ether, ester, carbamate, carbonate, cycloheteroalkyane, heteroaromatic, alkoxime or amide bond;
Ri, R2, R3, R4, R5 and Re, are the same or different and are 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 (OCH2CH2)p, wherein p is an integer from 0 to about 1000, or combination thereof. Additionally Ri, R2, 3 and R4 are respectively a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand, the phosphinate or sulfonyl group, the conjugated drug and among themselves (Ri, R2, R3 and R4). The atoms used in forming the hydrophilic linker may be combined in all chemically relevant ways, such as forming alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxy lamines, urethanes, amino acids, peptides, acyloxylamines, hydroxamic acids, or combination thereof.
M is H, or Na, or K, or N+RiR2R3 or a pharmaceutical salt. Ri, R2 and R3 are described above.
5. The formula (II) and (IV) of claims 2 and 4, wherein the Drug is 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 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 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 including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide 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). 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 propionate, 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 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 .γΐ, δΐ, l and βΐ; 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 l-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; ellornithine (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;
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). Aminoglycosides: amikacin, astromicin, gentamicin (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). Glycopeptides: bleomycin, vancomycin (oritavancin, telavancin), teicoplanin (dalbavancin), ramoplanin; g). Glycylcyclines: e. g. tigecycline; g). β-Lactamase inhibitors: penam (sulbactam, tazobactam), clavam (clavulanic acid); i). Lincosamides: clindamycin, lincomycin; j). Lipopeptides: daptomycin, A54145, calcium-dependent antibiotics (CD A); 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; 1). Monobactams: aztreonam, tigemonam; m). Oxazolidinones: linezolid; n). Penicillins: amoxicillin, ampicillin (pivampicillin, hetacillin, bacampicillin, metampicillin, talampicillin), azidocillin, azlocillin, benzylpeniciUin, benzathine benzylpeniciUin, benzathine phenoxymethylpeniciUin, clometociUin, procaine benzylpeniciUin, carbeniciUin (carindaciUin), cloxacillin, dicloxacillin, epicillin, flucloxacillin, mecillinam (pivmecillinam), mezlocillin, meticillin, nafcillin, oxacillin, penamecillin, penicillin, pheneticillin, phenoxymethylpeniciUin, 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, 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). Neuraminidase inhibitors: oseltamivir, zanamivir, peramivir; e). Nucleosides & nucleotides: abacavir, aciclovir, adefovir, amdoxovir, apricitabine, brivudine, cidofovir, clevudine, dexelvucitabine, didanosine (ddl), 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. M-thymidine and M-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 radioisotopes selected from (radionuclides) 3H, nC, 14C, 18F, 32P, 35S, 64Cu, 68Ga, 86Y, "Tc, U 1ln, 123I, 124I, 125I, 131I, 133Xe, 177Lu, 211At, or 213Bi.
6) . The pharmaceutically acceptable salts, acids or derivatives of any of the above drugs.
6. The conjugate of claims 2 or 4, wherein "Drug" is preferred from 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), siRNA or a combination thereof, and pharmaceutically acceptable salts, acids or derivatives of any of the above.
7. The conjugate of claims 2 and 3 wherein the cell binding agent/molecule is selected from antibodies, proteins, vitamins (e.g. folates), peptides, polymeric micelles, liposomes, lipoprotein-based drug carriers, nano-particle drug carriers, dendrimers, and combination thereof.
8. The conjugate of claims 2 and 3, wherein the cell-binding agent binds to target cells which are selected from tumor cells, virus infected cells, microorganism infected cells, parasite infected cells, autoimmune cells, activated cells, myeloid cells, activated T-cells, B cells, or melanocytes; cells expressing the CD19, CD20, CD22, CD30, CD33, CD37, CD38, CD40, CD 51, CD52, CD56, CD66, CD70, CD74, CD79, CD80, CD98, CD125, CD221, CD227, CD262, CD309, CD326, CEACAM3, CEACAM5 (carcinoembryonic antigen), DLL4 (A-like-4), EGFR, CTLA4, CXCR4 (CD 184), Endoglin (CD 105), EPCAM (epithelial cell adhesion molecule), ERBB2 (Epidermal Growth Factor Receptor 2), FCGR1, FOLR (folate receptor,), GD2 ganglioside, G-28 (a cell surface antigen glyvolipid), GD3 idiotype, Heat shock proteins, HER1, HER2, HLA-DR10, HLA-DRB , human chorionic gonadotropin, IGF1R (insulin-like growth factor 1 receptor), IL-2 receptor (interleukin 2 receptor), IL-6R (interleukin 6 receptor), Integrins (ανβ3, α5β1, α6β4, α11β3, α5β5, ανβ5), MAGE-1, MAGE-2, MAGE-3, MAGE 4, anti-transferrin receptor, ρ97, MS4A1 (membrane-spanning 4-domains subfamily A member 1), MUC1 or MUC1-KLH, MUC16 (CA125), CEA, gplOO, MARTI, MPG, MS4A1
(membrane-spanning 4-domains subfamily A), Nucleolin, Neu oncogene product, P21, Paratope of anti-(N-glycolylneuraminic acid), PLAP-like testicular alkaline phosphatase, PSMA, PSA, ROB04, TAG 72 (tumor associated glycoprotein 72), T cell transmembrane protein, Tie (CD202b), TNFRSF10B (tumor necrosis factor receptor superfamily member 10B), TNFRSF13B (tumor necrosis factor receptor superfamily member 13B), TPBG
(trophoblast glycoprotein), TRAIL-R1 (Tumor necrosis apoprosis Inducing ligand Receptor 1), VCAM-1 (CD 106), VEGF, VEGF-A, VEGF-2 (CD309), CanAg, CALLA, and cells expressing insulin growth factor receptor, or epidermal growth factor receptor.
9. The conjugate of claim 2 and 3, wherein the cell-binding agent is preferred an antibody, a single chain antibody, an antibody fragment that binds to the target cell, a monoclonal antibody, 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 domain 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 humanized 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.
10. The conjugate of claim 8, wherein the tumor cells are selected from lymphoma cells, myeloma cells, renal cells, breast cancer cells, prostate cancer cells, ovarian cancer cells, colorectal cancer cells, gastric cancer cells, squamous cancer cells, small-cell lung cancer cells, testicular cancer cells, or any cells that grow and divide at an unregulated, quickened pace to cause cancers.
11. A pharmaceutical composition comprising a therapeutically effective amount of the immunoconjugate of claims 2, and a pharmaceutically acceptable salt, carrier, diluent, or excipient therefore, or a combination therefore, for the treatment or prevention of a cancer, or an autoimmune disease, or an infectious disease.
12. The immunoconjugate of claims 2 and 4, wherein the drug/cytotoxic agent are selected from a toxin, a chemotherapeutic agent, a drug moiety, an antibiotic, a radioactive isotope, and a nucleolytic enzyme.
13. The immunoconjugate of claim 2, the immunoconjugate having the formula Cb-(-L- Drug)n, wherein Cb is a cell-binding agent, L is the hydrophilic linker, Drug is a drug
molecule, and n is an integer from 1 to 20.
14. The immunoconjugate of claim 13, wherein the hydrophilic linker L may be composed of one or more linker components of 6-maleimidocaproyl (MC), maleimidopropanoyl (MP), valine - citrulline (val-cit), alanine-phenylalanine (ala-phe), p-aminobenzyloxycarbonyl (PAB), 4-thio- pentanoate (SPP), 4-(N-maleimidomethyl)cyclohexane-l-carboxylate (MCC), 4-thio-butyrate (SPDB), maleimidoethyl containing (ME), 4-thio-2-hydroxysulfonyl-butyrate (2-Sulfo-SPDB) and (4-acetyl)aminobenzoate (SIAB).
15. The immunoconjugate of claims 2 and 13, wherein Drug is preferably selected from tubulysins, maytansinoids, taxanoids (taxanes), CC-1065 analogs, daunorubicin and doxorubicin compounds, benzodiazepine dimers (e.g., dimers of pyrrolobenzodiazepine (PBD), tomaymycin, anthramycin, indolinobenzodiazepines, imidazobenzothiadiazepines, or
oxazolidinobenzodiazepines), 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, esperamicins, and their analogues and derivatives thereof.
16. The immunoconjugate of claims 2 and 13, having in vitro, in vivo or ex vivo cell killing activity.
17. The immunoconjugate of claim 13, wherein the linker comprises 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, or an alkoxime unit.
18. The immunoconjugate of claim 13, wherein the linker can be cleavable by a protease.
19. A pharmaceutical composition comprising the immunoconjugate of claim 2 or 13, and a pharmaceutically acceptable carrier.
20. A pharmaceutical composition comprising a therapeutically effective amount of the immunoconjugate of claim 2 or 13, administered concurrently with the other therapeutic agents such as the chemo therapeutic agent, the radiation therapy, immunotherapy agents, autoimmune disorder agents, anti-infectious agents or the other immunoconjugates for synergistically effective treatment or prevention of a cancer, or an autoimmune disease, or an infectious disease.
PCT/IB2012/056700 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules WO2014080251A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
PCT/IB2012/056700 WO2014080251A1 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
ES12888899T ES2701076T3 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for the conjugation of drugs to molecules that bind to cells
CN201280076481.3A CN105849086B (en) 2012-11-24 2012-11-24 Hydrophily chain junctor and its application on drug molecule and cell-binding molecules conjugation reaction
AU2012395148A AU2012395148B2 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
EP12888899.7A EP2922818B1 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
CA2891280A CA2891280C (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
JP2015543532A JP6133431B2 (en) 2012-11-24 2012-11-24 Use of hydrophilic conjugates and conjugation reactions between drug molecules and cell binding molecules
US14/432,073 US10131682B2 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to a cell binding molecules

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2012/056700 WO2014080251A1 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules

Publications (1)

Publication Number Publication Date
WO2014080251A1 true WO2014080251A1 (en) 2014-05-30

Family

ID=50775615

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2012/056700 WO2014080251A1 (en) 2012-11-24 2012-11-24 Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules

Country Status (8)

Country Link
US (1) US10131682B2 (en)
EP (1) EP2922818B1 (en)
JP (1) JP6133431B2 (en)
CN (1) CN105849086B (en)
AU (1) AU2012395148B2 (en)
CA (1) CA2891280C (en)
ES (1) ES2701076T3 (en)
WO (1) WO2014080251A1 (en)

Cited By (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015095953A1 (en) 2013-12-27 2015-07-02 The Centre For Drug Research And Development Sulfonamide-containing linkage systems for drug conjugates
WO2015127685A1 (en) 2014-02-28 2015-09-03 Hangzhou Dac Biotech Co., Ltd Charged linkers and their uses for conjugation
US20150284416A1 (en) * 2015-06-16 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Novel linkers for conjugation of cell-binding molecules
WO2015151081A2 (en) 2015-07-12 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Bridge linkers for conjugation of a cell-binding molecule
WO2015151078A3 (en) * 2015-06-15 2016-03-10 Suzhou M-Conj Biotech Co., Ltd Hydrophilic linkers for conjugation
RU2578604C1 (en) * 2014-12-22 2016-03-27 Федеральное государственное бюджетное научное учреждение "Научно-исследовательский институт по изысканию новых антибиотиков имени Г.Ф. Гаузе" Chimeric antibiotics based on azithromycin and glycopeptide antibiotics, having antibacterial activity and synthesis method thereof
WO2015151079A3 (en) * 2015-06-20 2016-05-06 Hangzhou Dac Biotech Co, Ltd Auristatin analogues and their conjugates with cell-binding molecules
WO2016077260A1 (en) 2014-11-10 2016-05-19 Bristol-Myers Squibb Company Tubulysin analogs and methods of making and use
WO2015151080A3 (en) * 2015-07-04 2016-06-02 Suzhou M-Conj Biotech Co., Ltd Specific conjugation of cell-binding molecules
WO2016090038A1 (en) * 2014-12-03 2016-06-09 Genentech, Inc. Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
WO2016090040A1 (en) * 2014-12-03 2016-06-09 Genentech, Inc. Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
WO2015155753A3 (en) * 2015-08-10 2016-06-30 Suzhou M-Conj Biotech Co., Ltd Novel linkers and their uses in specific conjugation of drugs to biological molecule
CN105769863A (en) * 2015-04-01 2016-07-20 重庆理工大学 Application of Tipranavir in anti-cancer drug and anti-cancer drug
WO2016115218A1 (en) * 2015-01-14 2016-07-21 The California Institute For Biomedical Research Antibody drug conjugates for the treatment of immune conditions
WO2016115201A1 (en) 2015-01-14 2016-07-21 Bristol-Myers Squibb Company Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using
WO2016147031A2 (en) 2015-03-19 2016-09-22 Hangzhou Dac Biotech Co., Ltd Novel hydrophilic linkers and ligand-drug conjugates thereof
WO2016160615A1 (en) * 2015-03-27 2016-10-06 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US9505747B2 (en) 2012-03-29 2016-11-29 Endocyte, Inc. Processes for preparing tubulysin derivatives and conjugates thereof
WO2016196591A1 (en) * 2015-06-01 2016-12-08 Indiana University Research & Technology Corporation Protein tyrosine phosphatases or shp2 inhibitors and uses thereof
US9527871B2 (en) 2015-01-14 2016-12-27 Bristol-Myers Squibb Company Benzodiazepine dimers, conjugates thereof, and methods of making and using
US9555139B2 (en) 2007-03-14 2017-01-31 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2017023667A1 (en) * 2015-07-31 2017-02-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Immunostimulatory nanocarrier
US9663499B2 (en) 2013-06-07 2017-05-30 The California Institute For Biomedical Research Small molecule inhibitors of fibrosis
US9662402B2 (en) 2012-10-16 2017-05-30 Endocyte, Inc. Drug delivery conjugates containing unnatural amino acids and methods for using
US9688694B2 (en) 2015-06-23 2017-06-27 Bristol-Myers Squibb Company Macrocyclic benzodiazepine dimers, conjugates thereof, preparation and uses
JP2017128553A (en) * 2016-01-22 2017-07-27 株式会社Ihi Anticancer agent and method for controlling anticancer agent
WO2017134547A1 (en) 2016-02-01 2017-08-10 Pfizer Inc. Tubulysin analogs and methods for their preparation
US9879086B2 (en) 2014-09-17 2018-01-30 Zymeworks Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
JP2018509424A (en) * 2015-03-13 2018-04-05 エンドサイト・インコーポレイテッドEndocyte, Inc. Conjugates for treating diseases
US9950076B2 (en) 2016-01-25 2018-04-24 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
EP3269393A4 (en) * 2015-04-27 2018-05-16 Institute of Basic Medical Sciences, Chinese Academi of Medical Sciences Hsp90 inhibition peptide conjugate and application thereof in treating tumor
US9974774B2 (en) 2013-07-26 2018-05-22 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
JP2018525346A (en) * 2015-06-29 2018-09-06 ウィリアム マーシュ ライス ユニバーシティWilliam Marsh Rice University Total synthesis of sisidimycin A and its analogs
US10080805B2 (en) 2012-02-24 2018-09-25 Purdue Research Foundation Cholecystokinin B receptor targeting for imaging and therapy
WO2018237335A1 (en) 2017-06-23 2018-12-27 VelosBio Inc. Ror1 antibody immunoconjugates
US10201614B2 (en) 2013-03-15 2019-02-12 Zymeworks Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US10233212B2 (en) 2015-11-03 2019-03-19 Industrial Technology Research Institute Compounds, linker-drugs and ligand-drug conjugates
WO2019106200A1 (en) 2017-11-30 2019-06-06 Vaxxilon Ag Vaccine against klebsiella pneumoniae
WO2019106201A1 (en) 2017-11-30 2019-06-06 Vaxxilon Ag Vaccine against klebsiella pneumoniae
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10336735B2 (en) 2014-12-10 2019-07-02 The Scripps Research Institute Small molecule inhibitors of fibrosis
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10487149B2 (en) 2016-04-01 2019-11-26 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US10500204B2 (en) 2007-06-25 2019-12-10 Endocyte, Inc. Vitamin receptor drug delivery conjugates for treating inflammation
WO2020006722A1 (en) * 2018-07-05 2020-01-09 Hangzhou Dac Biotech Co., Ltd Cross-linked pyrrolobenzodiazepine dimer (pbd) derivative and its conjugates
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
US10570151B2 (en) 2013-03-15 2020-02-25 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US10646585B2 (en) 2017-09-15 2020-05-12 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and ligand-drug conjugates thereof
US10647676B2 (en) 2004-07-23 2020-05-12 Endocyte, Inc. Bivalent linkers and conjugates thereof
WO2020104697A1 (en) 2018-11-22 2020-05-28 Vaxxilon Ag Stable vaccine against clostridium difficile
US10675355B2 (en) 2013-12-27 2020-06-09 Var2 Pharmaceuticals Aps VAR2CSA-drug conjugates
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10738086B2 (en) 2007-06-25 2020-08-11 Endocyte Inc. Conjugates containing hydrophilic spacer linkers
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US10881743B2 (en) 2017-12-06 2021-01-05 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US10913800B2 (en) 2018-12-21 2021-02-09 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
CN112584871A (en) * 2018-05-01 2021-03-30 希默赛生物技术公司 Branched sugar alcohol-based compounds and compositions and methods thereof
US10973920B2 (en) 2014-06-30 2021-04-13 Glykos Finland Oy Saccharide derivative of a toxic payload and antibody conjugates thereof
US10994020B2 (en) 2017-01-06 2021-05-04 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and methods of inducing exon skipping
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
JP2021073185A (en) * 2015-08-10 2021-05-13 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.Hangzhou Dac Biotech Co.,Ltd. Novel connected body and use thereof in specific conjugation between biomolecule and drug
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11110180B2 (en) 2017-10-04 2021-09-07 Avidity Biosciences Inc. Nucleic acid-polypeptide compositions and uses thereof
US11142767B2 (en) 2017-07-21 2021-10-12 The Governors Of The University Of Alberta Antisense oligonucleotides that bind to exon 51 of human dystrophin pre-mRNA
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
AU2016363013B2 (en) * 2015-12-04 2022-03-10 Seagen Inc. Conjugates of quaternized tubulysin compounds
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11446387B2 (en) 2020-03-27 2022-09-20 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
WO2022194264A1 (en) * 2021-03-19 2022-09-22 Shenzhen Enduring Biotech, Ltd. Pegylated t cell engager with dual specificities to cd3 and cd19
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11512289B2 (en) 2015-02-18 2022-11-29 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
US11525137B2 (en) 2020-03-19 2022-12-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11578090B2 (en) 2019-06-06 2023-02-14 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US11596635B2 (en) 2013-08-26 2023-03-07 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2023118961A1 (en) * 2021-12-21 2023-06-29 Intocell, Inc. Antibody drug conjugates comprising toxins with polar groups and uses thereof
WO2023122347A2 (en) 2021-12-23 2023-06-29 Mirecule, Inc. Compositions for delivery of polynucleotides
US11702473B2 (en) * 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11807628B2 (en) 2018-05-29 2023-11-07 Intocell, Inc. Benzodiazepine derivatives and uses thereof
US11826430B2 (en) 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11834458B2 (en) 2021-03-23 2023-12-05 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11857634B2 (en) 2018-04-20 2024-01-02 University of Pittsburgh—of the Commonwealth System of Higher Education Cationic amphiphilic polymers for codelivery of hydrophobic agents and nucleic acids
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
US11912779B2 (en) 2021-09-16 2024-02-27 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
US11952349B2 (en) 2019-11-13 2024-04-09 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9428845B1 (en) 2010-12-28 2016-08-30 Warp Drive Bio, Inc. Identifying new therapeutic agents
HUE048574T2 (en) 2012-07-12 2020-08-28 Hangzhou Dac Biotech Co Ltd Conjugates of cell binding molecules with cytotoxic agents
US11873281B2 (en) 2012-07-12 2024-01-16 Hangzhou Dac Biotech Co., Ltd. Conjugates of cell binding molecules with cytotoxic agents
WO2015038649A1 (en) 2013-09-10 2015-03-19 Synta Pharmaceuticals Corp. Targeted therapeutics
WO2015066053A2 (en) 2013-10-28 2015-05-07 Synta Pharmaceuticals Corp. Targeted therapeutics
WO2015116774A1 (en) 2014-01-29 2015-08-06 Synta Pharmaceuticals Corp. Targeted therapeutics
WO2015134464A2 (en) 2014-03-03 2015-09-11 Synta Pharmaceuticals Corp. Targeted therapeutics
WO2015138638A1 (en) 2014-03-11 2015-09-17 Theraly Pharmaceuticals, Inc. Long acting trail receptor agonists for treatment of autoimmune diseases
WO2015143004A1 (en) 2014-03-18 2015-09-24 Synta Pharmaceuticals Corp. Targeted therapeutics
EP4289950A3 (en) 2015-01-09 2024-01-24 Revolution Medicines, Inc. Macrocyclic compounds that participate in cooperative binding and medical uses thereof
US9989535B2 (en) 2015-10-01 2018-06-05 Warp Drive Bio, Inc. Methods and reagents for analyzing protein-protein interfaces
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
WO2017106627A1 (en) 2015-12-17 2017-06-22 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
WO2017151425A1 (en) * 2016-02-29 2017-09-08 Madrigal Pharmaceuticals, Inc. Hsp90 inhibitor drug conjugates
WO2017177148A1 (en) 2016-04-07 2017-10-12 The Johns Hopkins University Compositions and methods for treating pancreatitis and pain with death receptor agonists
AU2017249412B2 (en) 2016-04-12 2022-07-07 Ginkgo Bioworks, Inc. Compositions and methods for the production of compounds
CR20180594A (en) 2016-06-02 2019-07-29 Abbvie Inc GLUCOCORTICOID RECEPTOR AGONIST AND IMMUNOCONJUGATES THEREOF
US10517958B2 (en) 2016-10-04 2019-12-31 Zymeworks Inc. Compositions and methods for the treatment of platinum-drug resistant cancer
WO2018081592A2 (en) 2016-10-28 2018-05-03 Warp Drive Bio, Inc. Compositions and methods for the production of compounds
KR20220147720A (en) * 2016-11-14 2022-11-03 항저우 디에이씨 바이오테크 씨오, 엘티디 Conjugation linkers, cell binding molecule-drug conjugates containing the likers, methods of making and uses such conjugates with the linkers
CN111065416A (en) * 2017-06-21 2020-04-24 芬兰吉利科斯有限公司 Hydrophilic linkers and conjugates thereof
CN109106951A (en) * 2017-08-18 2019-01-01 四川百利药业有限责任公司 A kind of camptothecine-antibody coupling matter
CA3083985A1 (en) * 2017-11-30 2019-06-06 Centurion Biopharma Corporation Maytansinoid-based drug delivery systems
RU2020117698A (en) 2017-12-01 2022-01-04 Эббви Инк. GLUCOCORTICOID RECEPTOR AGONIST AND ITS IMMUNOCONJUGATES
WO2019109188A1 (en) * 2017-12-06 2019-06-13 Ontario Institute For Cancer Research (Oicr) Acyl hydrazone linkers, methods and uses thereof
CA3086366A1 (en) 2017-12-22 2019-06-27 Ontario Institute For Cancer Research (Oicr) Heterocyclic acyl hydrazone linkers, methods and uses thereof
MX2020006192A (en) * 2017-12-31 2020-08-20 Hangzhou Dac Biotech Co Ltd A conjugate of a tubulysin analog with branched linkers.
CN110643034B (en) * 2018-06-26 2022-05-06 湖南华腾医药有限公司 Multi-arm PEG (polyethylene glycol) oritavancin derivative and preparation thereof
CN108774315A (en) * 2018-06-27 2018-11-09 湖南华腾制药有限公司 PEGylated Norfloxacin and its antibacterial application
JP7335957B2 (en) * 2018-07-09 2023-08-30 シンシス セラピューティクス,インコーポレイテッド Antibody-ALK5 inhibitor conjugates and uses thereof
US20210308273A1 (en) 2018-08-02 2021-10-07 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
US11168141B2 (en) 2018-08-02 2021-11-09 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
KR20210081324A (en) 2018-08-02 2021-07-01 다인 세라퓨틱스, 인크. Muscle targeting complexes and their use for treating facioscapulohumeral muscular dystrophy
US11911484B2 (en) 2018-08-02 2024-02-27 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
CN110256313B (en) * 2019-05-15 2021-01-29 江苏省原子医学研究所 Photosensitizer prodrug compound and preparation method and application thereof
WO2020256721A1 (en) * 2019-06-19 2020-12-24 Synthis, Llc Antib0dy-alk5 inhibitor conjugates and their uses
WO2021091982A1 (en) 2019-11-04 2021-05-14 Revolution Medicines, Inc. Ras inhibitors
CN115873020A (en) 2019-11-04 2023-03-31 锐新医药公司 RAS inhibitors
CA3160142A1 (en) 2019-11-04 2021-05-14 Revolution Medicines, Inc. Ras inhibitors
CA3108168A1 (en) * 2020-02-05 2021-08-05 Yue Zhang Conjugates of cell-binding molecules with cytotoxic agents
CN114502200A (en) * 2020-04-15 2022-05-13 深圳康源久远生物技术有限公司 Antibody drug conjugates
US11767353B2 (en) 2020-06-05 2023-09-26 Theraly Fibrosis, Inc. Trail compositions with reduced immunogenicity
PE20231207A1 (en) 2020-09-15 2023-08-17 Revolution Medicines Inc INDOLIC DERIVATIVES AS RAS INHIBITORS IN CANCER TREATMENT
CN112316158B (en) * 2020-11-19 2021-09-21 四川大学 Method for closing antibacterial agent activity in collagen solution by using supermolecule encapsulating agent
US11969475B2 (en) 2021-07-09 2024-04-30 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
US11638761B2 (en) 2021-07-09 2023-05-02 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating Facioscapulohumeral muscular dystrophy
US11672872B2 (en) 2021-07-09 2023-06-13 Dyne Therapeutics, Inc. Anti-transferrin receptor antibody and uses thereof
US11771776B2 (en) 2021-07-09 2023-10-03 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
US11633498B2 (en) 2021-07-09 2023-04-25 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
WO2023137443A1 (en) 2022-01-14 2023-07-20 Regeneron Pharmaceuticals, Inc. Verrucarin a derivatives and antibody drug conjugates thereof
US11931421B2 (en) 2022-04-15 2024-03-19 Dyne Therapeutics, Inc. Muscle targeting complexes and formulations for treating myotonic dystrophy

Citations (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4000304A (en) 1974-05-06 1976-12-28 Mead Johnson & Company Diuretic antiturombogenic and antiarrhythmic processes using N-substituted indole dimers and pyrrolobenzodia-zepine rearrangement products thereof
US4427587A (en) 1982-11-10 1984-01-24 Bristol-Myers Company Total synthesis of antitumor antibiotics BBM-2040A and BBM-2040B
US4464467A (en) 1982-07-26 1984-08-07 Bristol-Myers Company Antitumor antibiotics produced by new streptomyces
US4508647A (en) 1982-07-26 1985-04-02 Bristol-Myers Company Antitumor antibiotics BBM-2040A and BBM-2040B
US4663453A (en) 1983-05-18 1987-05-05 Hoechst-Roussel Pharmaceuticals Inc. Benzo[b]pyrrolo[3,2,1-jk][1,4]benzodiazepines having dopamine receptor activity
US4683230A (en) 1985-02-25 1987-07-28 Bristol-Myers Company BMY-28121, a new antitumor antibiotic
US4723003A (en) 1983-05-18 1988-02-02 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines
US4723007A (en) 1983-05-18 1988-02-02 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines
US4761412A (en) 1983-05-18 1988-08-02 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines useful for the treatment of psychoses
US4764616A (en) 1983-05-18 1988-08-16 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines
US4935362A (en) 1985-02-25 1990-06-19 Bristol-Myers Company BMY-28121, a new antitumor antibiotic
US5880122A (en) 1996-11-01 1999-03-09 American Home Products Corporation 3-Carboxamide derivatives of 5H-pyrrolo 2,1-c! 1,4!-benzodiazepines
US6344451B1 (en) 1999-02-04 2002-02-05 American Home Products Pyrrolobenzodiazepine carboxyamide vasopressin agonists
US6562806B1 (en) 1998-08-27 2003-05-13 Spirogen Limited Pyrrolobenzodiazepines
US6608192B1 (en) 1998-08-27 2003-08-19 Spirogen Limited Collections of compounds
US6660856B2 (en) 2002-03-08 2003-12-09 Kaohsiung Medical University Synthesis of pyrrolo[2,1-c][1,4]benzodiazepine analogues
US6747144B1 (en) 1998-08-27 2004-06-08 Spirogen Limited Collections of compounds
US6800622B1 (en) 2003-03-25 2004-10-05 Council Of Scientific And Industrial Research Pyrene-linked pyrrolo[2,1-c][1,4]benzodiazepine hybrids useful as anti-cancer agents
US6884799B2 (en) 2003-03-31 2005-04-26 Council Of Scientific And Industrial Research Non-cross-linking pyrrolo[2,1-c][1,4]benzodiazepines and process thereof
US6909006B1 (en) 1999-08-27 2005-06-21 Spirogen Limited Cyclopropylindole derivatives
US6951853B1 (en) 2004-03-30 2005-10-04 Council Of Scientific And Industrial Research Process for preparing pyrrolo[2, 1-c] [1,4] benzodiazepine hybrids
US6977254B2 (en) 2001-04-12 2005-12-20 Wyeth Hydroxy cyclohexenyl phenyl carboxamides tocolytic oxytocin receptor antagonists
US6979684B1 (en) 2004-06-30 2005-12-27 Council Of Scientific And Industrial Research Pyrrolo[2,1-c][1,4]benzodiazepine-napthalimide conjugates linked through piperazine moiety and process for preparation thereof
US7015215B2 (en) 2003-03-31 2006-03-21 Council Of Scientific And Industrial Research Pyrrolo[2,1-c][1,4] benzodiazepines compounds and process thereof
US7022699B2 (en) 2001-04-12 2006-04-04 Wyeth Cyclohexenyl phenyl diazepines vasopressin and oxytocin receptor modulators
US7049311B1 (en) 1998-08-27 2006-05-23 Spirogen Limited Pyrrolbenzodiazepines
US7056913B2 (en) 2004-03-30 2006-06-06 Council Of Scientific And Industrial Research C8—linked pyrrolo[2,1-c][1,4]benzodiazepine-acridone/acridine hybrids
US7064120B2 (en) 2001-04-12 2006-06-20 Wyeth Tricyclic pyridyl carboxamides and derivatives thereof tocolytic oxytocin receptor antagonists
US7109193B2 (en) 2001-04-12 2006-09-19 Wyeth Tricyclic diazepines tocolytic oxytocin receptor antagonists
US7173026B2 (en) 2004-12-27 2007-02-06 Council Of Scientific And Industrial Research Pyrrolo [2,1-c][1,4]benzodiazepine-anthraquinone conjugates useful as antitumour agents
US7189710B2 (en) 2004-03-30 2007-03-13 Council Of Scientific And Industrial Research C2-fluoro pyrrolo [2,1−c][1,4]benzodiazepine dimers
US7202239B2 (en) 2001-04-12 2007-04-10 Wyeth Cyclohexylphenyl carboxamides tocolytic oxytocin receptor antagonists
US7312210B2 (en) 2005-11-10 2007-12-25 Council Of Scientific And Industrial Research Pyrrolo[2,1-c][1,4]benzodiazepine compounds and processes for the preparation thereof
US7326700B2 (en) 2001-04-12 2008-02-05 Wyeth Cyclohexenyl phenyl carboxamides tocolytic oxytocin receptor antagonists
WO2008034019A2 (en) * 2006-09-13 2008-03-20 Polymerix Corporation Active agents and their oligomers and polymers
WO2008070291A2 (en) * 2006-10-24 2008-06-12 Kereos, Inc. Improved linkers for anchoring targeting ligands
US7407951B2 (en) 2002-11-14 2008-08-05 Spirogen Limited Pyrrolobenzodiazepines
US7429658B2 (en) 2003-09-11 2008-09-30 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
WO2009002993A1 (en) * 2007-06-25 2008-12-31 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
US7511032B2 (en) 2003-10-22 2009-03-31 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Pyrrolobenzodiazepine derivatives, compositions comprising the same and methods related thereto
US7528128B2 (en) 2006-08-14 2009-05-05 Council Of Scientific & Industrial Research Pyrrolo[2,1-C][1,4]benzodiazepine hybrids and a process for the preparation thereof
US7528126B2 (en) 2004-03-09 2009-05-05 Spirogen Limited Pyrrolobenzodiazepines
US7557099B2 (en) 2004-03-01 2009-07-07 Spirogen Limited Pyrrolobenzodiazepines as key intermediates in the synthesis of dimeric cytotoxic pyrrolobenzodiazepines
US7608615B2 (en) 2006-08-14 2009-10-27 Council Of Scientific & Industrial Research Pyrrolo[2,1-C][1,4]benzodiazepine hybrids and a process for the preparation thereof
US7612062B2 (en) 2005-04-21 2009-11-03 Spirogen Limited Pyrrolobenzodiazepines
US7678787B2 (en) 2005-06-09 2010-03-16 Wyeth Pyrrolobenzodiazepine pyridine carboxamides and derivatives as follicle-stimulating hormone receptor antagonists
US7691848B2 (en) 2005-03-02 2010-04-06 Wyeth Pyrrolobenzodiazepine arylcarboxamides and derivatives thereof as follicle-stimulating hormone receptor antagonists
US7741319B2 (en) 2004-03-01 2010-06-22 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
US20100203007A1 (en) 2009-02-05 2010-08-12 Immunogen Inc. Novel benzodiazepine derivatives
US20100316656A1 (en) 2007-07-19 2010-12-16 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives and their therapeutic use
US8034808B2 (en) 2004-04-27 2011-10-11 Ipsen Pharma S.A.S. Therapeutic compositions containing at least one pyrrolobenzodiazepine derivative and fludarabine
US8153627B2 (en) 2008-03-05 2012-04-10 Council Of Scientific & Industrial Research Quinazoline linked pyrrolo[2,1-C][1, 4]benzodiazepine hybrids as potential anticancer agents and process for the preparation thereof
US8163736B2 (en) 2006-01-25 2012-04-24 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives

Family Cites Families (272)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3479408A (en) 1966-10-26 1969-11-18 I C I Organics Inc Beta,beta'-thiodiethyl sulfones
DE2657380C3 (en) 1976-12-17 1981-02-05 Bayer Ag, 5090 Leverkusen Halogenethyl sulfones, process for their preparation and their use for regulating plant growth
US4169888A (en) 1977-10-17 1979-10-02 The Upjohn Company Composition of matter and process
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
JPS55102583A (en) 1979-01-31 1980-08-05 Takeda Chem Ind Ltd 20-acyloxy-20-demethylmaytansinoid compound
JPS55162791A (en) 1979-06-05 1980-12-18 Takeda Chem Ind Ltd Antibiotic c-15003pnd and its preparation
JPS5645483A (en) 1979-09-19 1981-04-25 Takeda Chem Ind Ltd C-15003phm and its preparation
EP0028683A1 (en) 1979-09-21 1981-05-20 Takeda Chemical Industries, Ltd. Antibiotic C-15003 PHO and production thereof
JPS5645485A (en) 1979-09-21 1981-04-25 Takeda Chem Ind Ltd Production of c-15003pnd
US4491632A (en) 1979-10-22 1985-01-01 The Massachusetts General Hospital Process for producing antibodies to hepatitis virus and cell lines therefor
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4391904A (en) 1979-12-26 1983-07-05 Syva Company Test strip kits in immunoassays and compositions therein
DE3167442D1 (en) 1980-07-07 1985-01-10 Nat Res Dev Improvements in or relating to cell lines
US4341761A (en) 1980-07-25 1982-07-27 E. I. Du Pont De Nemours And Company Antibodies to immunogenic peptides and their use to purify human fibroblast interferon
WO1982001188A1 (en) 1980-10-08 1982-04-15 Takeda Chemical Industries Ltd 4,5-deoxymaytansinoide compounds and process for preparing same
US4450254A (en) 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
US4466917A (en) 1981-02-12 1984-08-21 New York University Malaria vaccine
US4563304A (en) 1981-02-27 1986-01-07 Pharmacia Fine Chemicals Ab Pyridine compounds modifying proteins, polypeptides or polysaccharides
US4493890A (en) 1981-03-23 1985-01-15 Miles Laboratories, Inc. Activated apoglucose oxidase and its use in specific binding assays
US4451570A (en) 1981-03-26 1984-05-29 The Regents Of The University Of California Immunoglobulin-secreting human hybridomas from a cultured human lymphoblastoid cell line
JPS57192389A (en) 1981-05-20 1982-11-26 Takeda Chem Ind Ltd Novel maytansinoid
US4414205A (en) 1981-08-28 1983-11-08 University Patents, Inc. Cell growth inhibitory substances
US4399121A (en) 1981-11-04 1983-08-16 Miles Laboratories, Inc. Iodothyronine immunogens and antibodies
US4427783A (en) 1981-12-14 1984-01-24 Hoffmann-La Roche Inc. Immunoassay of thymosin α1
US4671958A (en) 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4493795A (en) 1983-10-17 1985-01-15 Syntex (U.S.A.) Inc. Synthetic peptide sequences useful in biological and pharmaceutical applications and methods of manufacture
US4753894A (en) 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US5169774A (en) 1984-02-08 1992-12-08 Cetus Oncology Corporation Monoclonal anti-human breast cancer antibodies
US6054561A (en) 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
US4912227A (en) 1984-02-21 1990-03-27 The Upjohn Company 1,2,8,8A-tetrahydrocyclopropa(c)pyrrolo(3,2-e)-indol-4-(5H)-ones and related compounds
US4978757A (en) 1984-02-21 1990-12-18 The Upjohn Company 1,2,8,8a-tetrahydrocyclopropa (C) pyrrolo [3,2-e)]-indol-4(5H)-ones and related compounds
US4764368A (en) 1984-08-29 1988-08-16 Dana-Farber Cancer Institute, Inc. Acid-cleavable compound
US4970198A (en) 1985-10-17 1990-11-13 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
US4680338A (en) 1985-10-17 1987-07-14 Immunomedics, Inc. Bifunctional linker
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
JPH0684377B1 (en) 1986-04-17 1994-10-26 Kyowa Hakko Kogyo Kk
EP0247730A3 (en) 1986-04-28 1989-04-12 Antibody Technology Limited Antibodies, their preparation and use and products containing them
US5332837A (en) 1986-12-19 1994-07-26 The Upjohn Company CC-1065 analogs
US5108912A (en) 1987-01-30 1992-04-28 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
US4816444A (en) 1987-07-10 1989-03-28 Arizona Board Of Regents, Arizona State University Cell growth inhibitory substance
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US5773435A (en) 1987-08-04 1998-06-30 Bristol-Myers Squibb Company Prodrugs for β-lactamase and uses thereof
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US4952394A (en) 1987-11-23 1990-08-28 Bristol-Myers Company Drug-monoclonal antibody conjugates
US4994578A (en) 1987-11-27 1991-02-19 Meiji Seika Kaisha, Ltd. Certain anti-tumor duocarmycin antibiotics from streptomyces
US5563250A (en) 1987-12-02 1996-10-08 Neorx Corporation Cleavable conjugates for the delivery and release of agents in native form
US5141648A (en) 1987-12-02 1992-08-25 Neorx Corporation Methods for isolating compounds using cleavable linker bound matrices
FI102355B1 (en) 1988-02-11 1998-11-30 Bristol Myers Squibb Co A method for preparing anthracycline immunoconjugates having a linking spacer
EP0329184A3 (en) 1988-02-19 1990-05-23 Neorx Corporation Antimers and antimeric conjugation
US5147786A (en) 1988-04-22 1992-09-15 Monsanto Company Immunoassay for the detection of α-haloacetamides
US4943628A (en) 1988-06-13 1990-07-24 Ortho Pharmaceutical Corporation HIV peptide-inducted T cell stimulation
JP2642165B2 (en) 1988-07-22 1997-08-20 協和醗酵工業株式会社 Novel DC-89 compound and method for producing the same
US5084468A (en) 1988-08-11 1992-01-28 Kyowa Hakko Kogyo Co., Ltd. Dc-88a derivatives
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
JP2598116B2 (en) 1988-12-28 1997-04-09 協和醗酵工業株式会社 New substance DC113
US4978744A (en) 1989-01-27 1990-12-18 Arizona Board Of Regents Synthesis of dolastatin 10
US4879278A (en) 1989-05-16 1989-11-07 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
US5094848A (en) * 1989-06-30 1992-03-10 Neorx Corporation Cleavable diphosphate and amidated diphosphate linkers
US5187186A (en) 1989-07-03 1993-02-16 Kyowa Hakko Kogyo Co., Ltd. Pyrroloindole derivatives
JP2510335B2 (en) 1989-07-03 1996-06-26 協和醗酵工業株式会社 DC-88A derivative
US5286637A (en) 1989-08-07 1994-02-15 Debiopharm, S.A. Biologically active drug polymer derivatives and method for preparing same
US5208323A (en) 1989-08-10 1993-05-04 Universite Laval Coupling of an anti-tumor to an antibody using glutaraldehyde preactivated anti-tumor agent
US5495009A (en) 1989-10-24 1996-02-27 Gilead Sciences, Inc. Oligonucleotide analogs containing thioformacetal linkages
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5334528A (en) 1989-10-30 1994-08-02 The Regents Of The University Of California Monoclonal antibodies to cyclodiene insecticides and method for detecting the same
US5165923A (en) 1989-11-20 1992-11-24 Imperial Cancer Research Technology Methods and compositions for the treatment of hodgkin's disease
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
FR2656555B1 (en) 1989-12-29 1994-10-28 Serimer MECHANICAL SYSTEM FOR AUTOMATIC GUIDANCE OF ONE OR MORE TORCHES OF AN ARC WELDING UNIT.
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
IT1238675B (en) 1990-01-26 1993-09-01 Ist Naz Stud Cura Dei Tumori MONOCLONAL ANTIBODY ABLE TO RECOGNIZE A SPECIFIC EPITOPE OF ANTHRACYCLINIC GLYCOSIDES AND HYBRIDOME SECONDING SUCH ANTIBODY
EP0527189A1 (en) 1990-04-25 1993-02-17 PHARMACIA &amp; UPJOHN COMPANY Novel cc-1065 analogs
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
JPH05507080A (en) 1990-05-03 1993-10-14 スクリップス クリニック アンド リサーチ ファウンデーション Intermediates for the formation of calicheamycin and esperamycin oligosaccharides
US5767236A (en) 1990-05-09 1998-06-16 Biomeasure, Inc. Linear therapeutic peptides
US5137877B1 (en) 1990-05-14 1996-01-30 Bristol Myers Squibb Co Bifunctional linking compounds conjugates and methods for their production
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
DE69121334T2 (en) 1990-07-26 1997-03-20 Kyowa Hakko Kogyo Kk DC-89 derivatives as anti-tumor agents
GB9017024D0 (en) 1990-08-03 1990-09-19 Erba Carlo Spa New linker for bioactive agents
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ES2108048T3 (en) 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5543390A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US6797492B2 (en) 1991-05-17 2004-09-28 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
JP4124480B2 (en) 1991-06-14 2008-07-23 ジェネンテック・インコーポレーテッド Immunoglobulin variants
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
DE69230824T2 (en) 1991-08-09 2000-07-27 Teikoku Hormone Mfg Co Ltd NEW TETRAPEPTIDE DERIVATIVES
ES2227512T3 (en) 1991-12-02 2005-04-01 Medical Research Council PRODUCTION OF ANTIBODIES AGAINST SELF-ANTIGENS FROM REPERTORIES OF ANTIBODY SEGMENTS FIXED IN A PHOTO.
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
CA2076465C (en) 1992-03-25 2002-11-26 Ravi V. J. Chari Cell binding agent conjugates of analogues and derivatives of cc-1065
GB9314960D0 (en) 1992-07-23 1993-09-01 Zeneca Ltd Chemical compounds
JP3514490B2 (en) 1992-08-21 2004-03-31 杏林製薬株式会社 Trifluoromethylpyrroloindole carboxylate derivative and method for producing the same
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US6015562A (en) 1992-09-22 2000-01-18 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US5324483B1 (en) 1992-10-08 1996-09-24 Warner Lambert Co Apparatus for multiple simultaneous synthesis
DE4236237A1 (en) 1992-10-27 1994-04-28 Behringwerke Ag Prodrugs, their preparation and use as medicines
US6034065A (en) 1992-12-03 2000-03-07 Arizona Board Of Regents Elucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US6569834B1 (en) 1992-12-03 2003-05-27 George R. Pettit Elucidation and synthesis of antineoplastic tetrapeptide w-aminoalkyl-amides
US5410024A (en) 1993-01-21 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US5436149A (en) 1993-02-19 1995-07-25 Barnes; Wayne M. Thermostable DNA polymerase with enhanced thermostability and enhanced length and efficiency of primer extension
US5475011A (en) 1993-03-26 1995-12-12 The Research Foundation Of State University Of New York Anti-tumor compounds, pharmaceutical compositions, methods for preparation thereof and for treatment
DE4314091A1 (en) 1993-04-29 1994-11-03 Boehringer Mannheim Gmbh Immunological detection method for triazines
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
DE4325824A1 (en) 1993-07-31 1995-02-02 Basf Ag Process for the preparation of homopolymers of ethylene or copolymers of ethylene
EP0731106B1 (en) 1993-10-01 2004-11-17 Teikoku Hormone Mfg. Co., Ltd. Dolastatin derivatives
GB9320575D0 (en) 1993-10-06 1993-11-24 Amp Gmbh Coaxial connector having improved locking mechanism
US5922545A (en) 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
US5786377A (en) 1993-11-19 1998-07-28 Universidad De Santiago De Compostela Pyrrolo 3,2-E!indol derivatives, process for the preparation thereof and applications
ES2220927T3 (en) 1994-04-22 2004-12-16 Kyowa Hakko Kogyo Co., Ltd. DERIVATIVES OF DC-89.
JPH07309761A (en) 1994-05-20 1995-11-28 Kyowa Hakko Kogyo Co Ltd Method for stabilizing duocamycin derivative
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5521284A (en) 1994-08-01 1996-05-28 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides and esters
US5530097A (en) 1994-08-01 1996-06-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory peptide amides
SI0769967T1 (en) 1994-08-19 2008-06-30 Wallone Region Conjugates comprising an antitumour agent and their use
US5554725A (en) 1994-09-14 1996-09-10 Arizona Board Of Regents Acting On Behalf Of Arizona State University Synthesis of dolastatin 15
US6111166A (en) 1994-09-19 2000-08-29 Medarex, Incorporated Transgenic mice expressing human Fcα and β receptors
US5599902A (en) 1994-11-10 1997-02-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Cancer inhibitory peptides
CA2205872A1 (en) 1994-11-29 1996-06-06 Kyorin Pharmaceutical Co., Ltd. Acrylamide derivatives and process for production thereof
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
WO1996022384A1 (en) 1995-01-18 1996-07-25 Boehringer Mannheim Gmbh Anti-cd 30 antibodies preventing proteolytic cleavage and release of membrane-bound cd 30 antigen
KR100408909B1 (en) 1995-04-21 2004-04-29 데이꼬꾸 조끼 세이야꾸 가부시키가이샤 Novel peptide derivatives
WO1996035451A1 (en) 1995-05-10 1996-11-14 Kyowa Hakko Kogyo Co., Ltd. Novel toxin complex
US5686237A (en) 1995-06-05 1997-11-11 Al-Bayati; Mohammed A. S. Use of biomarkers in saliva to evaluate the toxicity of agents and the function of tissues in both biomedical and environmental applications
WO1996040662A2 (en) 1995-06-07 1996-12-19 Cellpro, Incorporated Aminooxy-containing linker compounds and their application in conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
GB9517001D0 (en) 1995-08-18 1995-10-18 Denny William Enediyne compounds
SE9503380D0 (en) 1995-09-29 1995-09-29 Pharmacia Ab Protein derivatives
EP0862553A4 (en) 1995-10-03 1999-02-03 Scripps Research Inst Cbi analogs of cc-1065 and the duocarmycins
DE69632989T2 (en) 1995-10-17 2005-08-25 Combichem, Inc., San Diego Template for the synthesis of combinatorial libraries in solution
WO1997023243A1 (en) 1995-12-22 1997-07-03 Bristol-Myers Squibb Company Branched hydrazone linkers
FR2742751B1 (en) 1995-12-22 1998-01-30 Rhone Poulenc Rorer Sa NOVEL TAXOIDS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US6143901A (en) 1996-07-31 2000-11-07 Genesoft, Inc. Complex formation between dsDNA and pyrrole imidazole polyamides
WO1997032850A1 (en) 1996-03-08 1997-09-12 The Scripps Research Institute Mcbi analogs of cc-1065 and the duocarmycins
US6309646B1 (en) * 1996-05-09 2001-10-30 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Protein-polysaccharide conjugate vaccines and other immunological reagents prepared using homobifunctional and heterobifunctional vinylsulfones, and processes for preparing the conjugates
WO1997045411A1 (en) 1996-05-31 1997-12-04 The Scripps Research Institute Analogs of cc-1065 and the duocarmycins
US5741892A (en) 1996-07-30 1998-04-21 Basf Aktiengesellschaft Pentapeptides as antitumor agents
US6130237A (en) 1996-09-12 2000-10-10 Cancer Research Campaign Technology Limited Condensed N-aclyindoles as antitumor agents
JPH1087666A (en) 1996-09-18 1998-04-07 Kyorin Pharmaceut Co Ltd Intermediate for producing duocarmycin sa and its derivative and production thereof
DE19638870B4 (en) 1996-09-23 2009-05-14 Helmholtz-Zentrum für Infektionsforschung GmbH Tubulysins, methods for their production and agents containing them
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
US6759509B1 (en) 1996-11-05 2004-07-06 Bristol-Myers Squibb Company Branched peptide linkers
US5811452A (en) 1997-01-08 1998-09-22 The Research Foundation Of State University Of New York Taxoid reversal agents for drug-resistance in cancer chemotherapy and pharmaceutical compositions thereof
US6239104B1 (en) 1997-02-25 2001-05-29 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
US5965537A (en) 1997-03-10 1999-10-12 Basf Aktiengesellschaft Dolastatin 15 derivatives with carbonyl and heterocyclic functionalities at the C-terminus
US6306393B1 (en) 1997-03-24 2001-10-23 Immunomedics, Inc. Immunotherapy of B-cell malignancies using anti-CD22 antibodies
WO1998043663A1 (en) 1997-03-28 1998-10-08 The Scripps Research Institute Sandramycin analogs
CA2289115A1 (en) 1997-05-07 1998-11-12 Bristol-Myers Squibb Company Recombinant antibody-enzyme fusion proteins
JP2002503228A (en) 1997-05-22 2002-01-29 ザ スクリップス リサーチ インスティテュート Duocarmycin and analogs of CC-1065
US6143721A (en) 1997-07-18 2000-11-07 Basf Aktiengesellschaft Dolastatin 15 derivatives
CA2306420A1 (en) 1997-10-14 1999-04-22 The Scripps Research Institute Iso-cbi and iso-ci analogs of cc-1065 and the duocarmycins
JP2001525382A (en) 1997-12-08 2001-12-11 ザ スクリップス リサーチ インスティテュート Synthesis of CC-1065 / Duocarmycin analogs
ATE284412T1 (en) 1998-01-09 2004-12-15 Univ Arizona State ANTI-CRYPTOCOCCUS PEPTIDES
US6162930A (en) 1998-03-06 2000-12-19 Baylor University Anti-mitotic agents which inhibit tubulin polymerization
US5981564A (en) 1998-07-01 1999-11-09 Universite Laval Water-soluble derivatives of paclitaxel, method for producing same and uses thereof
JP3045706B1 (en) 1998-09-14 2000-05-29 科学技術振興事業団 Compound for alkylating a specific base sequence of DNA and method for synthesizing the same
CA2372053C (en) 1999-04-28 2008-09-02 Board Of Regents, The University Of Texas System Compositions and methods for cancer treatment by selectively inhibiting vegf
US6323315B1 (en) 1999-09-10 2001-11-27 Basf Aktiengesellschaft Dolastatin peptides
US7303749B1 (en) 1999-10-01 2007-12-04 Immunogen Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
ATE349438T1 (en) 1999-11-24 2007-01-15 Immunogen Inc CYTOTOXIC ACTIVE INGREDIENTS CONTAINING TAXANES AND THEIR THERAPEUTIC USE
WO2001049698A1 (en) 1999-12-29 2001-07-12 Immunogen, Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
IT1317731B1 (en) 2000-01-18 2003-07-15 Indena Spa SEMISYNTHETIC TASSANI WITH ANTI-TUMOR AND ANTIANGIOGENETIC ACTIVITY.
DE10008089A1 (en) 2000-02-22 2001-10-31 Biotechnolog Forschung Gmbh Production of tubulysin compounds comprises multi-stage process including condensation of N-methylpipecolinoyl-isoleucine with substituted thiazole-4-carboxylic acid derivative
US7097840B2 (en) 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
JP2003529609A (en) 2000-03-16 2003-10-07 ジーンソフト インコーポレイティッド Charged compounds containing nucleic acid binding moieties and uses thereof
JP4061819B2 (en) 2000-05-12 2008-03-19 独立行政法人科学技術振興機構 Method for synthesizing interstrand crosslinker
CA2411545A1 (en) 2000-06-14 2002-01-03 Medarex, Inc. Tripeptide prodrug compounds
JP2004510702A (en) 2000-06-14 2004-04-08 メダレックス,インコーポレイティド Prodrug compounds having isoleucine
US6333410B1 (en) 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
US7897404B2 (en) 2000-09-29 2011-03-01 Roche Diagnostics Operations, Inc. Conjugates of defined stoichiometry
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US7090843B1 (en) 2000-11-28 2006-08-15 Seattle Genetics, Inc. Recombinant anti-CD30 antibodies and uses thereof
US20020169125A1 (en) 2001-03-21 2002-11-14 Cell Therapeutics, Inc. Recombinant production of polyanionic polymers and uses thereof
EP1243276A1 (en) 2001-03-23 2002-09-25 Franciscus Marinus Hendrikus De Groot Elongated and multiple spacers containing activatible prodrugs
WO2002087497A2 (en) 2001-04-26 2002-11-07 Board Of Regents, The University Of Texas System Therapeutic agent/ligand conjugate compositions, their methods of synthesis and use
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
KR20030033007A (en) 2001-05-31 2003-04-26 코울터 파머수티컬, 인코포레이티드 Cytotoxins, prodrugs, linkers and stabilizers useful therefor
US6762179B2 (en) 2001-05-31 2004-07-13 Vertex Pharmaceuticals Incorporated Thiazole compounds useful as inhibitors of protein kinase
CA2450316A1 (en) 2001-06-11 2002-12-19 Medarex, Inc. Cd10-activated prodrug compounds
US7098305B2 (en) 2001-09-06 2006-08-29 Ardana Bioscience Limited Sustained release of microcrystalline peptide suspensions
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
US6716821B2 (en) 2001-12-21 2004-04-06 Immunogen Inc. Cytotoxic agents bearing a reactive polyethylene glycol moiety, cytotoxic conjugates comprising polyethylene glycol linking groups, and methods of making and using the same
US8361464B2 (en) 2002-03-01 2013-01-29 Immunomedics, Inc. Anthracycline-Antibody Conjugates for Cancer Therapy
US6534660B1 (en) 2002-04-05 2003-03-18 Immunogen, Inc. CC-1065 analog synthesis
US6756397B2 (en) 2002-04-05 2004-06-29 Immunogen, Inc. Prodrugs of CC-1065 analogs
LT1507556T (en) 2002-05-02 2016-10-10 Wyeth Holdings Llc Calicheamicin derivative-carrier conjugates
US6596757B1 (en) 2002-05-14 2003-07-22 Immunogen Inc. Cytotoxic agents comprising polyethylene glycol-containing taxanes and their therapeutic use
KR20040106547A (en) 2002-05-15 2004-12-17 엔도사이트, 인코포레이티드 Vitamin-mitomycin conjugates
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US7767803B2 (en) 2002-06-18 2010-08-03 Archemix Corp. Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
DE10230872A1 (en) 2002-07-09 2004-01-22 Morphochem AG Aktiengesellschaft für kombinatorische Chemie Process for the preparation of substituted thiazol-2-ylmethyl esters
EP1521769B1 (en) 2002-07-09 2015-09-09 Dömling, Alexander Tubulysin conjugates
US7816377B2 (en) 2002-07-09 2010-10-19 R&D-Biopharmaceuticals Gmbh Tubulysin analogues
EP2357006B1 (en) 2002-07-31 2015-09-16 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
ATE499116T1 (en) 2002-08-16 2011-03-15 Immunogen Inc HIGH REACTIVITY AND SOLUBILITY CROSS-LINKERS AND THEIR USE IN THE PRODUCTION OF CONJUGATES FOR THE TARGETED DELIVERY OF SMALL MOLECULAR DRUGS
DE10241152A1 (en) 2002-09-05 2004-03-18 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Tubulysin biosynthesis genes
DE10254439A1 (en) 2002-11-21 2004-06-03 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Tubulysins, manufacturing processes and tubulysin agents
US7662387B2 (en) 2003-02-20 2010-02-16 Seattle Genetics Anti-cd70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
DE10310082A1 (en) 2003-03-07 2004-09-16 Ktb Tumorforschungsgesellschaft Mbh Protein-binding doxorubicin peptide derivatives
US7384636B2 (en) 2003-03-31 2008-06-10 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Polypeptide
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
US7902338B2 (en) 2003-07-31 2011-03-08 Immunomedics, Inc. Anti-CD19 antibodies
EP1648512A4 (en) 2003-07-31 2009-01-21 Immunomedics Inc Anti-cd19 antibodies
BR122018071968B8 (en) 2003-11-06 2021-07-27 Seattle Genetics Inc antibody-drug conjugate, pharmaceutical composition, article of manufacture and use of an antibody-drug conjugate
CN102895666B (en) 2003-12-16 2015-08-19 尼克塔治疗公司 The micromolecule of chemical modification
EP1718667B1 (en) 2004-02-23 2013-01-09 Genentech, Inc. Heterocyclic self-immolative linkers and conjugates
JP4942643B2 (en) 2004-03-02 2012-05-30 シアトル ジェネティックス, インコーポレイテッド Partially added antibodies and methods for conjugating them
AU2005245396A1 (en) 2004-05-14 2005-12-01 Immunogen, Inc. A facile method for synthesizing baccatin III compounds
US7691962B2 (en) 2004-05-19 2010-04-06 Medarex, Inc. Chemical linkers and conjugates thereof
DE102004030227A1 (en) 2004-06-23 2006-01-26 Dömling, Alexander, Dr. Treatment of diseases (e.g. proliferative diseases of tumor cells, ophthalmic diseases and skin diseases) associated with angiogenesis comprises administration of carbamide compounds
JP4193771B2 (en) 2004-07-27 2008-12-10 セイコーエプソン株式会社 Gradation voltage generation circuit and drive circuit
WO2006033913A2 (en) 2004-09-16 2006-03-30 Synta Pharmaceuticals Corp. Bis (thio-hydrazide amides) for treament of hyperplasia
EP2899277A1 (en) 2004-11-26 2015-07-29 Pieris AG Compound with affinity for the cytotoxic T lymphocyte-associated antigen (CTLA-4)
EP1669358A1 (en) 2004-12-07 2006-06-14 Aventis Pharma S.A. Cytotoxic agents comprising new taxanes
EP1688415A1 (en) 2004-12-07 2006-08-09 Aventis Pharma S.A. Cytotoxic agents comprising new C-2 modified taxanes
US7301019B2 (en) 2005-01-21 2007-11-27 Immunogen, Inc. Method for the preparation of maytansinoid esters
WO2006096754A2 (en) 2005-03-07 2006-09-14 Archemix Corp. Stabilized aptamers to psma and their use as prostate cancer therapeutics
EP2239573A3 (en) 2005-03-30 2011-02-08 Saladax Biomedical Inc. Doxorubicin derivatives and conjugates for doxorubicin immunoassay
ES2609919T3 (en) 2005-04-28 2017-04-25 Ventana Medical Systems, Inc. Enzymes conjugated to antibodies using a heterobifunctional PEG linker
PL1945647T3 (en) 2005-11-08 2012-04-30 Immunogen Inc Processes for preparation of maytansinol
CA2627190A1 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
CN101535244B (en) 2005-11-23 2014-07-23 文塔納医疗系统公司 Molecular conjugate
EP1832577A1 (en) 2006-03-07 2007-09-12 Sanofi-Aventis Improved prodrugs of CC-1065 analogs
US7949572B2 (en) 2006-06-27 2011-05-24 Google Inc. Distributed electronic commerce system with independent third party virtual shopping carts
GB0615211D0 (en) 2006-07-31 2006-09-06 Ge Healthcare Uk Ltd Asymmetric flouro-substituted polymethine dyes
TW201509433A (en) 2006-12-13 2015-03-16 Cerulean Pharma Inc Cyclodextrin-based polymers for therapeutics delivery
NZ599239A (en) 2007-03-14 2013-10-25 Endocyte Inc Binding ligand linked drug delivery conjugates of tubulysins
WO2008125116A2 (en) 2007-03-31 2008-10-23 Aarhus Universitet Upar inhibition
EP2148886B1 (en) 2007-05-10 2014-02-19 R & D Biopharmaceuticals Gmbh Tubulysine derivatives
EP2181101A2 (en) 2007-07-20 2010-05-05 Helmholtz-Zentrum für Infektionsforschung GmbH Tubulysin d analogues
WO2009134279A1 (en) 2007-07-20 2009-11-05 The Regents Of The University Of California Tubulysin d analogues
EP3388086B1 (en) 2007-08-17 2020-10-07 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
US9187521B2 (en) 2007-10-25 2015-11-17 Endocyte, Inc. Tubulysins and processes for preparing
EP2245054B1 (en) 2008-01-30 2018-11-28 Pieris Pharmaceuticals GmbH Muteins of tear lipocalin having affinity to human c-met receptor tyrosine kinase and methods for obtaining the same
WO2009131239A1 (en) 2008-04-25 2009-10-29 Kyowa Hakko Kirin Co Ltd Stable polyvalent antibody
EP2276506A4 (en) 2008-04-30 2014-05-07 Immunogen Inc Potent conjugates and hydrophilic linkers
KR20230003298A (en) 2008-04-30 2023-01-05 이뮤노젠 아이엔씨 Cross-linkers and their uses
US8852630B2 (en) 2008-05-13 2014-10-07 Yale University Chimeric small molecules for the recruitment of antibodies to cancer cells
CN102215844A (en) 2008-09-17 2011-10-12 恩多塞特公司 Folate receptor binding conjugates of antifolates
EP2174947A1 (en) 2008-09-25 2010-04-14 Universität des Saarlandes Bioactive pre-tubulysins and use thereof
JP2012511033A (en) 2008-12-08 2012-05-17 テゴファーム コーポレーション Masking ligand for reversible inhibition of multivalent compounds
PT3903829T (en) 2009-02-13 2023-06-02 Immunomedics Inc Immunoconjugates with an intracellularly-cleavable linkage
JP2011026294A (en) 2009-06-26 2011-02-10 Canon Inc Compound
IT1394860B1 (en) 2009-07-22 2012-07-20 Kemotech S R L PHARMACEUTICAL COMPOUNDS
US8394922B2 (en) 2009-08-03 2013-03-12 Medarex, Inc. Antiproliferative compounds, conjugates thereof, methods therefor, and uses thereof
AU2010292172A1 (en) 2009-09-09 2012-05-03 Centrose, Llc Extracellular targeted drug conjugates
AR082205A1 (en) 2010-07-12 2012-11-21 Covx Technologies Ireland Ltd CONJUGATES OF MULTIFUNCTIONAL ANTIBODIES
WO2012138749A1 (en) 2011-04-04 2012-10-11 Immunogen, Inc. Methods for decreasing ocular toxicity of antibody drug conjugates
US9156854B2 (en) * 2011-04-18 2015-10-13 Immunogen, Inc. Maytansinoid derivatives with sulfoxide linker
HUE048574T2 (en) 2012-07-12 2020-08-28 Hangzhou Dac Biotech Co Ltd Conjugates of cell binding molecules with cytotoxic agents

Patent Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4000304A (en) 1974-05-06 1976-12-28 Mead Johnson & Company Diuretic antiturombogenic and antiarrhythmic processes using N-substituted indole dimers and pyrrolobenzodia-zepine rearrangement products thereof
US4464467A (en) 1982-07-26 1984-08-07 Bristol-Myers Company Antitumor antibiotics produced by new streptomyces
US4508647A (en) 1982-07-26 1985-04-02 Bristol-Myers Company Antitumor antibiotics BBM-2040A and BBM-2040B
US4427587A (en) 1982-11-10 1984-01-24 Bristol-Myers Company Total synthesis of antitumor antibiotics BBM-2040A and BBM-2040B
US4764616A (en) 1983-05-18 1988-08-16 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines
US4663453A (en) 1983-05-18 1987-05-05 Hoechst-Roussel Pharmaceuticals Inc. Benzo[b]pyrrolo[3,2,1-jk][1,4]benzodiazepines having dopamine receptor activity
US4723003A (en) 1983-05-18 1988-02-02 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines
US4723007A (en) 1983-05-18 1988-02-02 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines
US4761412A (en) 1983-05-18 1988-08-02 Hoechst-Roussel Pharmaceuticals Inc. Benzopyrrolobenzodiazepines and quinobenzodiazepines useful for the treatment of psychoses
US4935362A (en) 1985-02-25 1990-06-19 Bristol-Myers Company BMY-28121, a new antitumor antibiotic
US4683230A (en) 1985-02-25 1987-07-28 Bristol-Myers Company BMY-28121, a new antitumor antibiotic
US5880122A (en) 1996-11-01 1999-03-09 American Home Products Corporation 3-Carboxamide derivatives of 5H-pyrrolo 2,1-c! 1,4!-benzodiazepines
US7049311B1 (en) 1998-08-27 2006-05-23 Spirogen Limited Pyrrolbenzodiazepines
US6562806B1 (en) 1998-08-27 2003-05-13 Spirogen Limited Pyrrolobenzodiazepines
US6608192B1 (en) 1998-08-27 2003-08-19 Spirogen Limited Collections of compounds
US20030195196A1 (en) 1998-08-27 2003-10-16 Spirogen Limited Pyrrolobenzodiazepines
US7704924B2 (en) 1998-08-27 2010-04-27 Spirogen Limited Library of compounds comprising pyrrolobenzodiazepine moieties
US6747144B1 (en) 1998-08-27 2004-06-08 Spirogen Limited Collections of compounds
US7265105B2 (en) 1998-08-27 2007-09-04 Spirogen Limited Pyrrolobenzodiazepines
US7067511B2 (en) 1998-08-27 2006-06-27 Spirogen Limited Pyrrolobenzodiazepines
US6344451B1 (en) 1999-02-04 2002-02-05 American Home Products Pyrrolobenzodiazepine carboxyamide vasopressin agonists
US6909006B1 (en) 1999-08-27 2005-06-21 Spirogen Limited Cyclopropylindole derivatives
US7064120B2 (en) 2001-04-12 2006-06-20 Wyeth Tricyclic pyridyl carboxamides and derivatives thereof tocolytic oxytocin receptor antagonists
US7109193B2 (en) 2001-04-12 2006-09-19 Wyeth Tricyclic diazepines tocolytic oxytocin receptor antagonists
US7202239B2 (en) 2001-04-12 2007-04-10 Wyeth Cyclohexylphenyl carboxamides tocolytic oxytocin receptor antagonists
US7022699B2 (en) 2001-04-12 2006-04-04 Wyeth Cyclohexenyl phenyl diazepines vasopressin and oxytocin receptor modulators
US6977254B2 (en) 2001-04-12 2005-12-20 Wyeth Hydroxy cyclohexenyl phenyl carboxamides tocolytic oxytocin receptor antagonists
US7326700B2 (en) 2001-04-12 2008-02-05 Wyeth Cyclohexenyl phenyl carboxamides tocolytic oxytocin receptor antagonists
US6660856B2 (en) 2002-03-08 2003-12-09 Kaohsiung Medical University Synthesis of pyrrolo[2,1-c][1,4]benzodiazepine analogues
US7407951B2 (en) 2002-11-14 2008-08-05 Spirogen Limited Pyrrolobenzodiazepines
US6800622B1 (en) 2003-03-25 2004-10-05 Council Of Scientific And Industrial Research Pyrene-linked pyrrolo[2,1-c][1,4]benzodiazepine hybrids useful as anti-cancer agents
US6884799B2 (en) 2003-03-31 2005-04-26 Council Of Scientific And Industrial Research Non-cross-linking pyrrolo[2,1-c][1,4]benzodiazepines and process thereof
US7015215B2 (en) 2003-03-31 2006-03-21 Council Of Scientific And Industrial Research Pyrrolo[2,1-c][1,4] benzodiazepines compounds and process thereof
US7429658B2 (en) 2003-09-11 2008-09-30 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
US7511032B2 (en) 2003-10-22 2009-03-31 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Pyrrolobenzodiazepine derivatives, compositions comprising the same and methods related thereto
US7834005B2 (en) 2003-10-22 2010-11-16 The United States Of America As Represented By The Department Of Health And Human Services Pyrrolobenzodiazepine derivatives, compositions comprising the same and methods related thereto
US7741319B2 (en) 2004-03-01 2010-06-22 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
US7557099B2 (en) 2004-03-01 2009-07-07 Spirogen Limited Pyrrolobenzodiazepines as key intermediates in the synthesis of dimeric cytotoxic pyrrolobenzodiazepines
US7528126B2 (en) 2004-03-09 2009-05-05 Spirogen Limited Pyrrolobenzodiazepines
US7189710B2 (en) 2004-03-30 2007-03-13 Council Of Scientific And Industrial Research C2-fluoro pyrrolo [2,1−c][1,4]benzodiazepine dimers
US6951853B1 (en) 2004-03-30 2005-10-04 Council Of Scientific And Industrial Research Process for preparing pyrrolo[2, 1-c] [1,4] benzodiazepine hybrids
US7056913B2 (en) 2004-03-30 2006-06-06 Council Of Scientific And Industrial Research C8—linked pyrrolo[2,1-c][1,4]benzodiazepine-acridone/acridine hybrids
US8034808B2 (en) 2004-04-27 2011-10-11 Ipsen Pharma S.A.S. Therapeutic compositions containing at least one pyrrolobenzodiazepine derivative and fludarabine
US6979684B1 (en) 2004-06-30 2005-12-27 Council Of Scientific And Industrial Research Pyrrolo[2,1-c][1,4]benzodiazepine-napthalimide conjugates linked through piperazine moiety and process for preparation thereof
US7173026B2 (en) 2004-12-27 2007-02-06 Council Of Scientific And Industrial Research Pyrrolo [2,1-c][1,4]benzodiazepine-anthraquinone conjugates useful as antitumour agents
US7691848B2 (en) 2005-03-02 2010-04-06 Wyeth Pyrrolobenzodiazepine arylcarboxamides and derivatives thereof as follicle-stimulating hormone receptor antagonists
US7612062B2 (en) 2005-04-21 2009-11-03 Spirogen Limited Pyrrolobenzodiazepines
US7678787B2 (en) 2005-06-09 2010-03-16 Wyeth Pyrrolobenzodiazepine pyridine carboxamides and derivatives as follicle-stimulating hormone receptor antagonists
US7312210B2 (en) 2005-11-10 2007-12-25 Council Of Scientific And Industrial Research Pyrrolo[2,1-c][1,4]benzodiazepine compounds and processes for the preparation thereof
US8163736B2 (en) 2006-01-25 2012-04-24 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives
US7608615B2 (en) 2006-08-14 2009-10-27 Council Of Scientific & Industrial Research Pyrrolo[2,1-C][1,4]benzodiazepine hybrids and a process for the preparation thereof
US7528128B2 (en) 2006-08-14 2009-05-05 Council Of Scientific & Industrial Research Pyrrolo[2,1-C][1,4]benzodiazepine hybrids and a process for the preparation thereof
WO2008034019A2 (en) * 2006-09-13 2008-03-20 Polymerix Corporation Active agents and their oligomers and polymers
WO2008070291A2 (en) * 2006-10-24 2008-06-12 Kereos, Inc. Improved linkers for anchoring targeting ligands
WO2009002993A1 (en) * 2007-06-25 2008-12-31 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
US20100316656A1 (en) 2007-07-19 2010-12-16 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives and their therapeutic use
US8153627B2 (en) 2008-03-05 2012-04-10 Council Of Scientific & Industrial Research Quinazoline linked pyrrolo[2,1-C][1, 4]benzodiazepine hybrids as potential anticancer agents and process for the preparation thereof
US20100203007A1 (en) 2009-02-05 2010-08-12 Immunogen Inc. Novel benzodiazepine derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2922818A4 *

Cited By (193)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10647676B2 (en) 2004-07-23 2020-05-12 Endocyte, Inc. Bivalent linkers and conjugates thereof
US9555139B2 (en) 2007-03-14 2017-01-31 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
US10500204B2 (en) 2007-06-25 2019-12-10 Endocyte, Inc. Vitamin receptor drug delivery conjugates for treating inflammation
US10738086B2 (en) 2007-06-25 2020-08-11 Endocyte Inc. Conjugates containing hydrophilic spacer linkers
US11344623B2 (en) 2012-02-24 2022-05-31 Purdue Research Foundation Cholecystokinin B receptor targeting for imaging and therapy
US10080805B2 (en) 2012-02-24 2018-09-25 Purdue Research Foundation Cholecystokinin B receptor targeting for imaging and therapy
US10765756B2 (en) 2012-02-24 2020-09-08 Purdue Research Foundation Cholecystokinin B receptor targeting for imaging and therapy
US9505747B2 (en) 2012-03-29 2016-11-29 Endocyte, Inc. Processes for preparing tubulysin derivatives and conjugates thereof
US11701430B2 (en) 2012-10-12 2023-07-18 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10646584B2 (en) 2012-10-12 2020-05-12 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10994023B2 (en) 2012-10-12 2021-05-04 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9662402B2 (en) 2012-10-16 2017-05-30 Endocyte, Inc. Drug delivery conjugates containing unnatural amino acids and methods for using
US11345715B2 (en) 2013-03-15 2022-05-31 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US10201614B2 (en) 2013-03-15 2019-02-12 Zymeworks Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US10570151B2 (en) 2013-03-15 2020-02-25 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US11617777B2 (en) 2013-03-15 2023-04-04 Zymeworks Bc Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US9981956B2 (en) 2013-06-07 2018-05-29 The Scripps Research Institute Small molecule inhibitors of fibrosis
US9663499B2 (en) 2013-06-07 2017-05-30 The California Institute For Biomedical Research Small molecule inhibitors of fibrosis
US9974774B2 (en) 2013-07-26 2018-05-22 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US9993460B2 (en) 2013-07-26 2018-06-12 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US10548876B2 (en) 2013-07-26 2020-02-04 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US10500192B2 (en) 2013-07-26 2019-12-10 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11135201B2 (en) 2013-07-26 2021-10-05 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11147800B2 (en) 2013-07-26 2021-10-19 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11596635B2 (en) 2013-08-26 2023-03-07 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
WO2015095953A1 (en) 2013-12-27 2015-07-02 The Centre For Drug Research And Development Sulfonamide-containing linkage systems for drug conjugates
US11560422B2 (en) 2013-12-27 2023-01-24 Zymeworks Inc. Sulfonamide-containing linkage systems for drug conjugates
US10675355B2 (en) 2013-12-27 2020-06-09 Var2 Pharmaceuticals Aps VAR2CSA-drug conjugates
US20170152274A1 (en) * 2014-02-28 2017-06-01 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
EP3122757A4 (en) * 2014-02-28 2018-02-07 Hangzhou Dac Biotech Co., Ltd Charged linkers and their uses for conjugation
US10464955B2 (en) 2014-02-28 2019-11-05 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
US20190127399A1 (en) * 2014-02-28 2019-05-02 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
US20190127400A1 (en) * 2014-02-28 2019-05-02 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
US20190127401A1 (en) * 2014-02-28 2019-05-02 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
US10696699B2 (en) 2014-02-28 2020-06-30 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
US10696700B2 (en) 2014-02-28 2020-06-30 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
WO2015127685A1 (en) 2014-02-28 2015-09-03 Hangzhou Dac Biotech Co., Ltd Charged linkers and their uses for conjugation
US10683314B2 (en) 2014-02-28 2020-06-16 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
US10973920B2 (en) 2014-06-30 2021-04-13 Glykos Finland Oy Saccharide derivative of a toxic payload and antibody conjugates thereof
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10450378B2 (en) 2014-09-17 2019-10-22 Zymeworks Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US10414822B2 (en) 2014-09-17 2019-09-17 Zymeworks Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US9879086B2 (en) 2014-09-17 2018-01-30 Zymeworks Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US11591405B2 (en) 2014-09-17 2023-02-28 Zymeworks Bc Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
WO2016077260A1 (en) 2014-11-10 2016-05-19 Bristol-Myers Squibb Company Tubulysin analogs and methods of making and use
US10077287B2 (en) 2014-11-10 2018-09-18 Bristol-Myers Squibb Company Tubulysin analogs and methods of making and use
WO2016090038A1 (en) * 2014-12-03 2016-06-09 Genentech, Inc. Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
RU2731055C2 (en) * 2014-12-03 2020-08-28 Дженентек, Инк. Conjugates of antibodies to staphylococcus aureus with rifamycin and use thereof
US20180021450A1 (en) * 2014-12-03 2018-01-25 Genentech, Inc. Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
CN107249642A (en) * 2014-12-03 2017-10-13 豪夫迈·罗氏有限公司 Anti-Staphylococcus aureus antibody rifamycin conjugate and application thereof
WO2016090040A1 (en) * 2014-12-03 2016-06-09 Genentech, Inc. Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
US20180125995A1 (en) * 2014-12-03 2018-05-10 Genentech, Inc. Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
US10336735B2 (en) 2014-12-10 2019-07-02 The Scripps Research Institute Small molecule inhibitors of fibrosis
RU2578604C1 (en) * 2014-12-22 2016-03-27 Федеральное государственное бюджетное научное учреждение "Научно-исследовательский институт по изысканию новых антибиотиков имени Г.Ф. Гаузе" Chimeric antibiotics based on azithromycin and glycopeptide antibiotics, having antibacterial activity and synthesis method thereof
US10112975B2 (en) 2015-01-14 2018-10-30 Briston-Myers Squibb Company Benzodiazepine dimers, conjugates thereof, and methods of making and using
WO2016115218A1 (en) * 2015-01-14 2016-07-21 The California Institute For Biomedical Research Antibody drug conjugates for the treatment of immune conditions
WO2016115201A1 (en) 2015-01-14 2016-07-21 Bristol-Myers Squibb Company Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using
US9676775B2 (en) 2015-01-14 2017-06-13 Bristol-Myers Squibb Company Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using
US9527871B2 (en) 2015-01-14 2016-12-27 Bristol-Myers Squibb Company Benzodiazepine dimers, conjugates thereof, and methods of making and using
US9822112B2 (en) 2015-01-14 2017-11-21 Bristol-Meyers Squibb Company Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using
US9822144B2 (en) 2015-01-14 2017-11-21 Bristol-Myers Squibb Company Benzodiazepine dimers, conjugates thereof, and methods of making and using
US9676794B2 (en) 2015-01-14 2017-06-13 Bristol-Myers Squibb Company Benzodiazepine dimers, conjugates thereof, and methods of making and using
US9526801B2 (en) 2015-01-14 2016-12-27 Bristol-Myers Squibb Company Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11717539B2 (en) 2015-02-18 2023-08-08 Enlivex Therapeutics RDO Ltd. Combination immune therapy and cytokine control therapy for cancer treatment
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11512289B2 (en) 2015-02-18 2022-11-29 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
JP2018509424A (en) * 2015-03-13 2018-04-05 エンドサイト・インコーポレイテッドEndocyte, Inc. Conjugates for treating diseases
AU2020200566B2 (en) * 2015-03-19 2020-05-21 Hangzhou Dac Biotech Co., Ltd Novel Hydrophilic Linkers and Ligand-Drug Conjugates Thereof
CN107614488A (en) * 2015-03-19 2018-01-19 杭州多禧生物科技有限公司 Novel hydrophilic connector and its application on ligand drug conjugation conjugate
WO2016147031A2 (en) 2015-03-19 2016-09-22 Hangzhou Dac Biotech Co., Ltd Novel hydrophilic linkers and ligand-drug conjugates thereof
US10456479B2 (en) 2015-03-19 2019-10-29 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and ligand-drug conjugates thereof
EP3271329A4 (en) * 2015-03-19 2019-05-01 Hangzhou Dac Biotech Co., Ltd Novel hydrophilic linkers and ligand-drug conjugates thereof
AU2019203316B2 (en) * 2015-03-19 2020-05-14 Hangzhou Dac Biotech Co., Ltd Novel Hydrophilic Linkers and Ligand-Drug Conjugates Thereof
WO2016147031A3 (en) * 2015-03-19 2016-11-10 Hangzhou Dac Biotech Co., Ltd Novel hydrophilic linkers and ligand-drug conjugates thereof
EP3912982A1 (en) * 2015-03-19 2021-11-24 Hangzhou Dac Biotech Co., Ltd Novel hydrophilic linkers and ligand-drug conjugates thereof
JP2018516851A (en) * 2015-03-27 2018-06-28 レゲネロン ファーマシューティカルス,インコーポレーテッド Maytansinoid derivatives, conjugates thereof, and methods of use
WO2016160615A1 (en) * 2015-03-27 2016-10-06 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
AU2016243527B2 (en) * 2015-03-27 2021-04-29 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
CN107995912A (en) * 2015-03-27 2018-05-04 里珍纳龙药品有限公司 CHROMATOGRAPHIC FRACTIONATION AND MASS derivative, its conjugate and application method
EA034950B1 (en) * 2015-03-27 2020-04-09 Регенерон Фармасьютикалз, Инк. Maytansinoid derivatives, conjugates thereof, and methods of use
JP2021113191A (en) * 2015-03-27 2021-08-05 レゲネロン ファーマシューティカルス,インコーポレーテッド Maytansinoid derivatives, conjugates thereof, and methods of use
CN105769863A (en) * 2015-04-01 2016-07-20 重庆理工大学 Application of Tipranavir in anti-cancer drug and anti-cancer drug
US11702473B2 (en) * 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11883429B2 (en) 2015-04-21 2024-01-30 Enlivex Therapeutics Rdo Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
EP3269393A4 (en) * 2015-04-27 2018-05-16 Institute of Basic Medical Sciences, Chinese Academi of Medical Sciences Hsp90 inhibition peptide conjugate and application thereof in treating tumor
US10918731B2 (en) 2015-04-27 2021-02-16 Institute Of Basic Medical Sciences, Chinese Academy Of Medical Sciences HSP90 inhibitory peptide conjugate and application thereof in treating tumor
US10532977B2 (en) 2015-06-01 2020-01-14 Indiana University Research And Technology Corporation Small molecule inhibitors of protein tyrosine phosphatases and uses thereof
WO2016196591A1 (en) * 2015-06-01 2016-12-08 Indiana University Research & Technology Corporation Protein tyrosine phosphatases or shp2 inhibitors and uses thereof
US10494332B2 (en) 2015-06-01 2019-12-03 Indiana University Research And Technology Corporation Protein tyrosine phosphatases or SHP2 inhibitors and uses thereof
CN108026123A (en) * 2015-06-15 2018-05-11 杭州多禧生物科技有限公司 Hydrophilic chain junctor for coupling
EP3307749A4 (en) * 2015-06-15 2019-06-19 Hangzhou Dac Biotech Co., Ltd Hydrophilic linkers for conjugation
CN108026123B (en) * 2015-06-15 2021-02-05 杭州多禧生物科技有限公司 Hydrophilic linkers for coupling
WO2015151078A3 (en) * 2015-06-15 2016-03-10 Suzhou M-Conj Biotech Co., Ltd Hydrophilic linkers for conjugation
CN112125929A (en) * 2015-06-15 2020-12-25 杭州多禧生物科技有限公司 Hydrophilic linkers for coupling
AU2021201765B2 (en) * 2015-06-15 2022-12-08 Hangzhou Dac Biotech Co., Ltd Hydrophilic Linkers for Conjugate
US10975112B2 (en) * 2015-06-16 2021-04-13 Hangzhou Dac Biotech Co., Ltd. Linkers for conjugation of cell-binding molecules
US20150284416A1 (en) * 2015-06-16 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Novel linkers for conjugation of cell-binding molecules
CN107921144A (en) * 2015-06-20 2018-04-17 杭州多禧生物科技有限公司 The auspicious statin analog of Australia and its conjugation conjugate with cell-binding molecules
CN107921144B (en) * 2015-06-20 2023-11-28 杭州多禧生物科技有限公司 Aureostatin analogs and conjugate conjugates thereof with cell-binding molecules
WO2015151079A3 (en) * 2015-06-20 2016-05-06 Hangzhou Dac Biotech Co, Ltd Auristatin analogues and their conjugates with cell-binding molecules
US9688694B2 (en) 2015-06-23 2017-06-27 Bristol-Myers Squibb Company Macrocyclic benzodiazepine dimers, conjugates thereof, preparation and uses
US9902740B2 (en) 2015-06-23 2018-02-27 Bristol-Myers Squibb Company Macrocyclic benzodiazepine dimers, conjugates thereof, preparation and uses
JP2018525346A (en) * 2015-06-29 2018-09-06 ウィリアム マーシュ ライス ユニバーシティWilliam Marsh Rice University Total synthesis of sisidimycin A and its analogs
WO2015151080A3 (en) * 2015-07-04 2016-06-02 Suzhou M-Conj Biotech Co., Ltd Specific conjugation of cell-binding molecules
WO2015151081A3 (en) * 2015-07-12 2016-06-02 Suzhou M-Conj Biotech Co., Ltd Bridge linkers for conjugation of cell-binding molecules
WO2015151081A2 (en) 2015-07-12 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Bridge linkers for conjugation of a cell-binding molecule
CN108449940A (en) * 2015-07-12 2018-08-24 苏州美康加生物科技有限公司 With the bridging junctor of the conjugation coupling of cell-binding molecules
CN108449940B (en) * 2015-07-12 2021-06-08 杭州多禧生物科技有限公司 Conjugated bridge linkers to cell binding molecules
WO2017023667A1 (en) * 2015-07-31 2017-02-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Immunostimulatory nanocarrier
JP2021073185A (en) * 2015-08-10 2021-05-13 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.Hangzhou Dac Biotech Co.,Ltd. Novel connected body and use thereof in specific conjugation between biomolecule and drug
WO2015155753A3 (en) * 2015-08-10 2016-06-30 Suzhou M-Conj Biotech Co., Ltd Novel linkers and their uses in specific conjugation of drugs to biological molecule
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
US10683327B2 (en) 2015-11-03 2020-06-16 Industrial Technology Research Institute Compounds, linker-drugs and ligand-drug conjugates
US10233212B2 (en) 2015-11-03 2019-03-19 Industrial Technology Research Institute Compounds, linker-drugs and ligand-drug conjugates
US10618935B2 (en) 2015-11-03 2020-04-14 Industrial Technology Research Institute Antibody-drug conjugate (ADC) and method for forming the same
AU2016363013B2 (en) * 2015-12-04 2022-03-10 Seagen Inc. Conjugates of quaternized tubulysin compounds
JP2017128553A (en) * 2016-01-22 2017-07-27 株式会社Ihi Anticancer agent and method for controlling anticancer agent
US10463749B2 (en) 2016-01-25 2019-11-05 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US9950076B2 (en) 2016-01-25 2018-04-24 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US11446389B2 (en) 2016-01-25 2022-09-20 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
WO2017134547A1 (en) 2016-02-01 2017-08-10 Pfizer Inc. Tubulysin analogs and methods for their preparation
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US10548986B2 (en) 2016-03-02 2020-02-04 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10550188B2 (en) 2016-04-01 2020-02-04 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US10800848B2 (en) 2016-04-01 2020-10-13 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US10487149B2 (en) 2016-04-01 2019-11-26 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US10787519B2 (en) 2016-04-01 2020-09-29 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11179472B2 (en) 2017-01-06 2021-11-23 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and methods of inducing exon skipping
US10994020B2 (en) 2017-01-06 2021-05-04 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and methods of inducing exon skipping
US11400163B2 (en) 2017-01-06 2022-08-02 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and methods of inducing exon skipping
US11311627B1 (en) 2017-01-06 2022-04-26 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and methods of inducing exon skipping
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11813335B2 (en) 2017-02-08 2023-11-14 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2018237335A1 (en) 2017-06-23 2018-12-27 VelosBio Inc. Ror1 antibody immunoconjugates
US10335496B2 (en) 2017-06-23 2019-07-02 VelosBio Inc. ROR1 antibody immunoconjugates
US11891603B2 (en) 2017-07-21 2024-02-06 The Governors Of The University Of Alberta Antisense oligonucleotides that bind to exon 51 of human dystrophin pre-mRNA
US11142767B2 (en) 2017-07-21 2021-10-12 The Governors Of The University Of Alberta Antisense oligonucleotides that bind to exon 51 of human dystrophin pre-mRNA
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10646585B2 (en) 2017-09-15 2020-05-12 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and ligand-drug conjugates thereof
US11364302B1 (en) 2017-10-04 2022-06-21 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US11110180B2 (en) 2017-10-04 2021-09-07 Avidity Biosciences Inc. Nucleic acid-polypeptide compositions and uses thereof
US11891413B2 (en) 2017-11-30 2024-02-06 Idorsia Pharmaceuticals Ltd Vaccine against Klebsiella pneumoniae
WO2019106200A1 (en) 2017-11-30 2019-06-06 Vaxxilon Ag Vaccine against klebsiella pneumoniae
US11479574B2 (en) 2017-11-30 2022-10-25 Idorsia Pharmaceuticals Ltd Vaccine against Klebsiella pneumoniae
WO2019106201A1 (en) 2017-11-30 2019-06-06 Vaxxilon Ag Vaccine against klebsiella pneumoniae
US11576980B2 (en) 2017-12-06 2023-02-14 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11583591B2 (en) 2017-12-06 2023-02-21 Avidity Biosciences Llc Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11497814B2 (en) 2017-12-06 2022-11-15 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11554176B2 (en) 2017-12-06 2023-01-17 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11872287B2 (en) 2017-12-06 2024-01-16 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US10881743B2 (en) 2017-12-06 2021-01-05 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11246941B2 (en) 2017-12-06 2022-02-15 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11253607B2 (en) 2017-12-06 2022-02-22 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11712478B2 (en) 2017-12-06 2023-08-01 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
US11857634B2 (en) 2018-04-20 2024-01-02 University of Pittsburgh—of the Commonwealth System of Higher Education Cationic amphiphilic polymers for codelivery of hydrophobic agents and nucleic acids
CN112584871A (en) * 2018-05-01 2021-03-30 希默赛生物技术公司 Branched sugar alcohol-based compounds and compositions and methods thereof
US11807628B2 (en) 2018-05-29 2023-11-07 Intocell, Inc. Benzodiazepine derivatives and uses thereof
WO2020006722A1 (en) * 2018-07-05 2020-01-09 Hangzhou Dac Biotech Co., Ltd Cross-linked pyrrolobenzodiazepine dimer (pbd) derivative and its conjugates
AU2018430758B2 (en) * 2018-07-05 2022-01-27 Hangzhou Dac Biotech Co., Ltd Cross-linked pyrrolobenzodiazepine dimer (PBD) derivative and its conjugates
WO2020104697A1 (en) 2018-11-22 2020-05-28 Vaxxilon Ag Stable vaccine against clostridium difficile
US11834510B2 (en) 2018-12-21 2023-12-05 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
US10913800B2 (en) 2018-12-21 2021-02-09 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
US11028179B2 (en) 2018-12-21 2021-06-08 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
US11826430B2 (en) 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11578090B2 (en) 2019-06-06 2023-02-14 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US11952349B2 (en) 2019-11-13 2024-04-09 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11555190B2 (en) 2020-03-19 2023-01-17 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11525137B2 (en) 2020-03-19 2022-12-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11446387B2 (en) 2020-03-27 2022-09-20 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
US11707532B2 (en) 2020-03-27 2023-07-25 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
WO2022194264A1 (en) * 2021-03-19 2022-09-22 Shenzhen Enduring Biotech, Ltd. Pegylated t cell engager with dual specificities to cd3 and cd19
US11834458B2 (en) 2021-03-23 2023-12-05 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11912779B2 (en) 2021-09-16 2024-02-27 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
WO2023118961A1 (en) * 2021-12-21 2023-06-29 Intocell, Inc. Antibody drug conjugates comprising toxins with polar groups and uses thereof
WO2023122347A2 (en) 2021-12-23 2023-06-29 Mirecule, Inc. Compositions for delivery of polynucleotides
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle

Also Published As

Publication number Publication date
CN105849086B (en) 2018-07-31
ES2701076T3 (en) 2019-02-20
AU2012395148B2 (en) 2016-10-27
US20150250896A1 (en) 2015-09-10
CN105849086A (en) 2016-08-10
US10131682B2 (en) 2018-11-20
AU2012395148A1 (en) 2015-05-21
EP2922818B1 (en) 2018-09-05
CA2891280A1 (en) 2014-05-30
JP6133431B2 (en) 2017-05-24
EP2922818A4 (en) 2016-11-09
CA2891280C (en) 2018-03-20
EP2922818A1 (en) 2015-09-30
JP2016501859A (en) 2016-01-21

Similar Documents

Publication Publication Date Title
AU2021201765B2 (en) Hydrophilic Linkers for Conjugate
US10696700B2 (en) Charged linkers and their uses for conjugation
AU2012395148B2 (en) Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
US10328157B2 (en) Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
US10975112B2 (en) Linkers for conjugation of cell-binding molecules
AU2016228256B2 (en) Conjugates of Cell Binding Molecules with Cytotoxic Agents
WO2015151081A2 (en) Bridge linkers for conjugation of a cell-binding molecule

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12888899

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14432073

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2012888899

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2891280

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015543532

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2012395148

Country of ref document: AU

Date of ref document: 20121124

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE