US20160375147A1 - Maytansinoid derivatives, conjugates thereof, and methods of use - Google Patents

Maytansinoid derivatives, conjugates thereof, and methods of use Download PDF

Info

Publication number
US20160375147A1
US20160375147A1 US15/081,759 US201615081759A US2016375147A1 US 20160375147 A1 US20160375147 A1 US 20160375147A1 US 201615081759 A US201615081759 A US 201615081759A US 2016375147 A1 US2016375147 A1 US 2016375147A1
Authority
US
United States
Prior art keywords
compound
integer
formula
methyl
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/081,759
Inventor
Thomas Nittoli
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=55650789&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20160375147(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US15/081,759 priority Critical patent/US20160375147A1/en
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NITTOLI, THOMAS
Publication of US20160375147A1 publication Critical patent/US20160375147A1/en
Priority to US16/712,941 priority patent/US20200385402A1/en
Priority to US17/528,144 priority patent/US20220259225A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • A61K47/48384
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • A61K47/48561
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • 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
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Definitions

  • the present disclosure concerns maytansinoid derivatives, conjugates thereof, and methods of treating or preventing proliferative diseases with the same.
  • Proliferative diseases for example cancer
  • Current treatments of proliferative diseases include surgery, radiation, chemotherapy, hormone-based therapy and/or immunotherapy.
  • a number of these treatments, particularly chemotherapy utilize anti-proliferative drugs that limit the spread of the abnormal cells.
  • these drugs are typically indiscriminate in their ability to kill cells, affecting both normal and abnormal cells.
  • ADCs Antibody drug conjugates
  • ADCs are compounds composed of an antibody that is linked, via a chemical linker, to a cytotoxic agent. Such compounds leverage the antibody's binding specificity for its target to deliver a cytotoxic agent to an abnormal cell.
  • A is arylene or heteroarylene
  • L is a linker
  • BA is a binding agent
  • k is an integer from 1 to 30. Also provided herein are stereoisomers of compounds of Formula (I).
  • A is arylene or heteroarylene.
  • A is arylene or heteroarylene and L is a linker.
  • stereoisomers of compounds of Formula PT1 are also provided herein.
  • FIG. 1 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-4-aminobenzamido-citrulline-valine-caprolyl-6-maleimidyl.
  • FIG. 2 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 3 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 4 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 5 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 6 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(4-amino-2-fluoro)benzamido-Cit-Val-Cap-Mal.
  • FIG. 7 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(4-amino-2-trifluoromethyl)benzamido-Cit-Val-Cap-Mal.
  • FIG. 8 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(4-amino-2-methoxy)benzamido-Cit-Val-Cap-Mal.
  • FIG. 9 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-4-aminobenzamide.
  • FIG. 10 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-fluoro-4-amino)benzamide
  • FIG. 11 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-trifluoromethyl-4-amino)benzamide.
  • FIG. 12 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-methoxy-4-amino)benzamide.
  • FIG. 13 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-trifluoromethyl-4-amino)benzamide.
  • FIG. 14 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-chloro-4-amino-5-fluoro)benzamide.
  • FIG. 15 depicts a general synthetic sequence for preparing compounds of Formula (II) wherein substituent R is defined herein and below.
  • FIG. 16 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2,5-difluoro-4-amino)benzamide.
  • FIG. 17 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-fluoro-4-amino)benzamide
  • FIG. 18 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-chloro-4-amino)benzamide.
  • FIG. 19 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(5-amino-8-carboxyquinoline)carboxamide.
  • FIG. 20 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-bromo-4-amino)benzamide.
  • FIG. 21 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-methoxy-4-amino)benzamide.
  • FIG. 22 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-methyl-4-amino)benzamide.
  • FIG. 23 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-methyl-4-amino)benzamide.
  • FIG. 24 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(8-amino-5-carboxyquinoline)carboxamide.
  • FIG. 25 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-methoxy-4-amino)benzamido-Cit-Val-Cap-Mal.
  • FIG. 26 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-fluoro-4-amino)benzamido-Cit-Val-Cap-6-amine.
  • FIG. 27 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-methoxy-5-amino)benzamide.
  • FIG. 28 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino-4-methoxy)benzamide.
  • FIG. 29 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino-5-fluoro)benzamide.
  • FIG. 30 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-fluoro-5-amino)benzamide.
  • FIG. 31 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino)benzamide.
  • FIG. 32 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino-4-fluoro)benzamide.
  • FIG. 33 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-4-aminobenzamide-adipic-NHS.
  • FIG. 34 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-4-aminobenzamide-Cap-Mal.
  • FIG. 35 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-methylsulfonyl-4-amino)benzamide.
  • FIG. 36 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-hydroxy-4-amino)benzamide.
  • FIG. 37 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-amino)benzamide.
  • FIG. 38 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(4-methoxy-2-amino)benzamide.
  • FIG. 39 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-morpholino-4-amino)benzamide.
  • FIG. 40 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-acetamido-4-amino)benzamide.
  • FIG. 41 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-4-aminobenzamide-Cit-Val-cap-diBromomethylacryl.
  • FIG. 42 depicts the deconvoluted mass spectroscopy (MS) spectrum of the antibody drug conjugate, PRLR-Q-63 conjugate from EXAMPLE 43.
  • FIG. 43 depicts the deconvoluted MS spectrum of the Isotype Control-Q-63 conjugate from EXAMPLE 43.
  • FIG. 44 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 45.
  • FIG. 45 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 45.
  • FIG. 46 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 45.
  • FIG. 47 depicts the plot of % Cell Viability vs. Log 10 [M] of certain compounds tested in EXAMPLE 45.
  • alkyl refers to a monovalent and saturated hydrocarbon radical moiety. Alkyl is optionally substituted and can be linear, branched, or cyclic, i.e., cycloalkyl. Alkyl includes, but is not limited to, those having 1-20 carbon atoms, i.e., C 1-20 alkyl; 1-12 carbon atoms, i.e., C 1-12 alkyl; 1-8 carbon atoms, i.e., C 1-8 alkyl; 1-6 carbon atoms, i.e., C 1-6 alkyl; and 1-3 carbon atoms, i.e., C 1-3 alkyl.
  • alkyl moieties include, but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, i-butyl, a pentyl moiety, a hexyl moiety, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • haloalkyl refers to alkyl, as defined above, wherein the alkyl includes at least one substituent selected from a halogen, e.g., F, Cl, Br, or I.
  • alkenyl refers to a monovalent hydrocarbon radical moiety containing at least two carbon atoms and one or more non-aromatic carbon-carbon double bonds. Alkenyl is optionally substituted and can be linear, branched, or cyclic. Alkenyl includes, but is not limited to, those having 2-20 carbon atoms, i.e., C 2-20 alkenyl; 2-12 carbon atoms, i.e., C 2-12 alkenyl; 2-8 carbon atoms, i.e., C 2-8 alkenyl; 2-6 carbon atoms, i.e., C 2-6 alkenyl; and 2-4 carbon atoms, i.e., C 2-4 alkenyl. Examples of alkenyl moieties include, but are not limited to vinyl, propenyl, butenyl, and cyclohexenyl.
  • alkynyl refers to a monovalent hydrocarbon radical moiety containing at least two carbon atoms and one or more carbon-carbon triple bonds. Alkynyl is optionally substituted and can be linear, branched, or cyclic.
  • Alkynyl includes, but is not limited to, those having 2-20 carbon atoms, i.e., C 2-20 alkynyl; 2-12 carbon atoms, i.e., C 2-12 alkynyl; 2-8 carbon atoms, i.e., C 2-8 alkynyl; 2-6 carbon atoms, i.e., C 2-6 alkynyl; and 2-4 carbon atoms, i.e., C 2-4 alkynyl.
  • alkynyl moieties include, but are not limited to ethynyl, propynyl, and butynyl.
  • alkoxy refers to a monovalent and saturated hydrocarbon radical moiety wherein the hydrocarbon includes a single bond to an oxygen atom and wherein the radical is localized on the oxygen atom, e.g., CH 3 CH 2 —O for ethoxy.
  • Alkoxy substituents bond to the compound which they substitute through this oxygen atom of the alkoxy substituent.
  • Alkoxy is optionally substituted and can be linear, branched, or cyclic, i.e., cycloalkoxy.
  • Alkoxy includes, but is not limited to, those having 1-20 carbon atoms, i.e., C 1-20 alkoxy; 1-12 carbon atoms, i.e., C 1-12 alkoxy; 1-8 carbon atoms, i.e., C 1-8 alkoxy; 1-6 carbon atoms, i.e., C 1-6 alkoxy; and 1-3 carbon atoms, i.e., C 1-3 alkoxy.
  • alkoxy moieties include, but are not limited to methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, i-butoxy, a pentoxy moiety, a hexoxy moiety, cyclopropoxy, cyclobutoxy, cyclopentoxy, and cyclohexoxy.
  • haloalkoxy refers to alkoxy, as defined above, wherein the alkoxy includes at least one substituent selected from a halogen, e.g., F, Cl, Br, or I.
  • aryl refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms are carbon atoms.
  • Aryl is optionally substituted and can be monocyclic or polycyclic, e.g., bicyclic or tricyclic.
  • aryl moieties include, but are not limited to those having 6 to 20 ring carbon atoms, i.e., C 6-20 aryl; 6 to 15 ring carbon atoms, i.e., C 6-15 aryl, and 6 to 10 ring carbon atoms, i.e., C 6-10 aryl.
  • Examples of aryl moieties include, but are limited to phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, and pyrenyl.
  • arylene refers to a divalent moiety of an aromatic compound wherein the ring atoms are only carbon atoms.
  • Arylene is optionally substituted and can be monocyclic or polycyclic, e.g., bicyclic or tricyclic.
  • Examples of aryl moieties include, but are not limited to those having 6 to 20 ring carbon atoms, i.e., C 6-20 arylene; 6 to 15 ring carbon atoms, i.e., C 6-15 arylene, and 6 to 10 ring carbon atoms, i.e., C 6-10 arylene.
  • alkaryl refers to an aryl that is substituted with at least one alkyl. Alkaryl is optionally substituted.
  • heteroalkyl refers to an alkyl in which one or more carbon atoms are replaced by heteroatoms.
  • heteroalkenyl refers to an alkenyl in which one or more carbon atoms are replaced by heteroatoms.
  • heteroalkynyl refers to an alkenyl in which one or more carbon atoms are replaced by heteroatoms.
  • Suitable heteroatoms include, but are not limited to, nitrogen, oxygen, and sulfur atoms. Heteroalkyl is optionally substituted.
  • heteroalkyl moieties include, but are not limited to, aminoalkyl, sulfonylalkyl, sulfinylalkyl.
  • heteroalkyl moieties also include, but are not limited to, methylamino, methylsulfonyl, and methylsulfinyl.
  • heteroaryl refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms contain carbon atoms and at least one oxygen, sulfur, nitrogen, or phosphorus atom.
  • heteroaryl moieties include, but are not limited to those having 5 to 20 ring atoms; 5 to 15 ring atoms; and 5 to 10 ring atoms. Heteroaryl is optionally substituted.
  • heteroarylene refers to an arylene in which one or more ring atoms of the aromatic ring are replaced with an oxygen, sulfur, nitrogen, or phosphorus atom. Heteroarylene is optionally substituted.
  • heterocycloalkyl refers to a cycloalkyl in which one or more carbon atoms are replaced by heteroatoms. Suitable heteroatoms include, but are not limited to, nitrogen, oxygen, and sulfur atoms. Heterocycloalkyl is optionally substituted. Examples of heterocycloalkyl moieties include, but are not limited to, morpholinyl, piperidinyl, tetrahydropyranyl, pyrrolidinyl, imidazolidinyl, oxazolidinyl, thiazolidinyl, dioxolanyl, dithiolanyl, oxanyl, or thianyl.
  • optionally substituted when used to describe a radical moiety, e.g., optionally substituted alkyl, means that such moiety is optionally bonded to one or more substituents.
  • substituents include, but are not limited to halo, cyano, nitro, haloalkyl, azido, epoxy, optionally substituted heteroaryl, optionally substituted heterocycloalkyl,
  • R A , R B , and R C are, independently at each occurrence, a hydrogen atom, alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, heteroalkyl, heteroaryl, or heterocycloalkyl, or R A and R B , together with the atoms to which they are bonded, form a saturated or unsaturated carbocyclic ring, wherein the ring is optionally substituted and wherein one or more ring atoms is optionally replaced with a heteroatom.
  • R A , R B , and R C are not hydrogen atoms.
  • R A is methyl.
  • R A is methylamino, methylsulfonyl, and methylsulfinyl. In some examples, R A is methylamino.
  • R A is methylamino, methylsulfonyl, and methylsulfinyl. In some examples, R A is methylamino.
  • the substituents on the optionally substituted heteroaryl, optionally substituted heterocycloalkyl, or optionally substituted saturated or unsaturated carbocyclic ring, if they are substituted, are not substituted with substituents which are further optionally substituted with additional substituents.
  • the substituent bonded to the group is unsubstituted unless otherwise specified.
  • binding agent refers to any molecule capable of binding with specificity to a given binding partner.
  • linker refers to a divalent moiety that covalently links the binding agent to the maytansinoid derivatives described herein.
  • amide synthesis conditions refers to reaction conditions suitable to effect the formation of an amide, e.g., by the reaction of a carboxylic acid, activated carboxylic acid, or acyl halide with an amine.
  • amide synthesis conditions refers to reaction conditions suitable to effect the formation of an amide bond between a carboxylic acid and an amine.
  • the carboxylic acid is first converted to an activated carboxylic acid before the activated carboxylic acid reacts with an amine to form an amide.
  • Suitable conditions to effect the formation of an amide include, but are not limited to, those utilizing reagents to effect the reaction between a carboxylic acid an amine, including, but not limited to, dicyclohexylcarbodiimide (DCC), diisopropylcarbodiimide (DIC), (benzotriazol-1-yloxy)tris(dimethylamino)phosphonium hexafluorophosphate (BOP), (benzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyBOP), (7-azabenzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyAOP), bromotripyrrolidinophosphonium hexafluorophosphate (PyBrOP), O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexaflu
  • a carboxylic acid is first converted to an activated carboxylic ester before reacting with an amine to form an amide bond.
  • the carboxylic acid is reacted with a reagent.
  • the reagent activates the carboxylic acid by deprotonating the carboxylic acid and then forming a product complex with the deprotonated carboxylic acid as a result of nucleophilic attack by the deprotonated carboxylic acid onto the protonated reagent.
  • this activated ester is more susceptible subsequently to nucleophilic attack by an amine than the carboxylic acid is before it is converted. This results in amide bond formation.
  • the carboxylic acid is described as activated.
  • Exemplary reagents include DCC and DIC.
  • terapéuticaally effective amount refers to an amount (of a compound) that is sufficient to provide a therapeutic benefit to a patient in the treatment or management of a disease or disorder, or to delay or minimize one or more symptoms associated with the disease or disorder.
  • Certain groups, moieties, substituents, and atoms are depicted with a wiggly line that intersects a bond or bonds to indicate the atom through which the groups, moieties, substituents, atoms are bonded.
  • cyclic group e.g., aromatic, heteroaromatic, fused ring, and saturated or unsaturated cycloalkyl or heterocycloalkyl
  • substituents bonded to a cyclic group are meant to indicate, unless specified otherwise, that the cyclic group may be substituted with that substituent at any ring position in the cyclic group or on any ring in the fused ring group, according to techniques set forth herein or which are known in the field to which the instant disclosure pertains.
  • subscript q is an integer from 0 to 4 and in which the positions of substituent R 1 are described generically, includes the following groups in which the positions of substituent R 1 are described specifically:
  • the substituent R 1 may be bonded to any ring position in the cyclic group or on any ring in the fused ring group which is not occupied by one of these substituents other than R 1 .
  • the following non-limiting representative illustrations indicate that the cyclic group can be substituted with the indicated substituent at any ring position or on either ring in the fused ring group:
  • A is arylene or heteroarylene
  • L is a linker
  • BA is a binding agent
  • k is an integer from 1 to 30.
  • A is arylene. In some embodiments, A is heteroarylene. In some embodiments, the arylene or heteroarylene is substituted with one or more electron withdrawing groups and/or one or more electron donating groups.
  • A is a divalent radical of benzene, of pyridine, of naphthalene, or of quinolone, which are optionally substituted.
  • A is a divalent radical of benzene which is optionally substituted with a member selected from the group consisting of amino, amido, alkyl, halo, haloalkyl, alkoxy, and haloalkoxy.
  • A is:
  • A is:
  • A is:
  • R 1 is C 1-6 alkyl or C 1-6 alkoxy. In some of these embodiments, R 1 is methyl, ethyl, methoxy, or ethoxy. In some of these embodiments, R 1 is methoxy.
  • R 1 is, independently, alkyl or halo. In some embodiments, R 1 is, independently, C 1-6 alkyl, C 1-6 haloalkyl, or halo. In some embodiments, R 1 is, independently, halo. In some embodiments, R 1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R A is methyl.
  • R 1 is hydroxyl.
  • R 1 is N-methylformamide.
  • R 1 is morpholinyl.
  • R 1 is, independently, alkyl, alkoxy, or halo. In some embodiments, R 1 is, independently, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl, C 1-6 haloalkoxy, or halo. In some embodiments, R 1 is, independently, halo. In some embodiments, R 1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • A is:
  • A is:
  • A is:
  • A is:
  • n 0, 1 or 2.
  • A is:
  • n 0, 1 or 2.
  • A is:
  • n is 0 or 1; and R 1 is alkoxy, halo, or haloalkyl.
  • A is:
  • n is 0 or 1; and R 1 is alkoxy, halo, or haloalkyl. In some examples, R 1 is alkoxy.
  • A is:
  • n is 0 or 1; and R 1 is C 1-6 alkoxy, halo, or C 1-6 haloalkyl.
  • A is:
  • n is 0 or 1; and R 1 is C 1-6 alkoxy, halo, or C 1-6 haloalkyl. In some examples, R 1 is C 1-6 alkoxy.
  • A is:
  • n 0 or 1
  • R 1 is C 1-6 alkoxy, halo, or C 1-6 haloalkyl
  • L is
  • b is an integer from 2 to 8.
  • A is:
  • n 0 or 1
  • R 1 is C 1-6 alkoxy, halo, or C 1-6 haloalkyl
  • L is
  • b is an integer from 2 to 8.
  • A is:
  • n 0, 1, 2, 3, or 4.
  • A is:
  • n 0, 1, 2, 3, or 4.
  • A is:
  • R 1 is C 1-6 alkyl, halo, or C 1-6 haloalkyl
  • n 0, 1 or 2.
  • A is:
  • A is:
  • R 1 is C 1-6 alkyl, C 1-6 alkoxy, halo, C 1-6 haloalkyl, or C 1-6 haloalkoxy;
  • n 0, 1, 2, 3, or 4.
  • A is:
  • R 1 is C 1-6 alkyl, C 1-6 alkoxy, halo, C 1-6 haloalkyl, or C 1-6 haloalkoxy;
  • n 0, 1, 2, 3, or 4.
  • A is:
  • R 1 is C 1-6 alkyl, C 1-6 alkoxy, halo, or C 1-6 haloalkyl. In certain of these embodiments, R 1 is methoxy or methyl. In some specific embodiments, R 1 is methoxy.
  • A is:
  • A is:
  • R 1 is halo or trifluoromethyl
  • n 0, 1 or 2.
  • A is:
  • A is:
  • X is a hydrogen atom, halo, or trifluoromethyl.
  • A is:
  • X is a hydrogen atom, halo, or trifluoromethyl
  • A is:
  • R 1 is the bond to the carbonyl.
  • R 1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl.
  • R 1 is trifluoromethyl.
  • R 1 is methoxy.
  • R 1 is fluoro.
  • R 1 is hydrogen.
  • A is:
  • R 1 is the bond to the carbonyl.
  • R 1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl.
  • R 1 is trifluoromethyl.
  • R 1 is methoxy.
  • R 1 is fluoro.
  • R 1 is hydrogen.
  • R 1 is sulfonyl. In some embodiments, R 1 is N-methylformamide. In some embodiments, R 1 is hydroxyl. In some embodiments, R 1 is morpholinyl.
  • A is:
  • R 1 is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, aryl, heteroalkyl, halo, haloalkyl, or haloalkoxy;
  • R 1 is the bond to the carbonyl.
  • R 1 is alkyl or alkoxy.
  • R 1 is propylamino, difluoro-methoxy, phenyl, 2-fluorophenyl.
  • R 1 is trifluoromethyl.
  • R 1 is methoxy.
  • R 1 is fluoro.
  • R 1 is hydrogen.
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • X is F, Cl, Br, CN, methoxy, dimethylamino or cyclopropyl
  • A is:
  • X is F, Cl, Br, CN, methoxy, dimethylamino, 1-methyl-ethyl-thio or cyclopropyl;
  • A is:
  • each R 1 is independently, at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkyl, or haloalkoxy;
  • R 1 is the bond to the carbonyl.
  • R 1 is hydrogen, fluoro, trifluoromethyl, or methoxy.
  • R 1 is fluoro, chloro, bromo, or iodo.
  • A is:
  • A is:
  • each R 1 is independently, at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkyl, or haloalkoxy,
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • n 0, 1 2, or 3.
  • A is:
  • R 1 is C 1-6 alkyl, C 1-6 alkoxy, halo, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 1-6 heteroalkyl;
  • n 0, 1 2, 3 or 4.
  • A is:
  • A is:
  • A is:
  • X is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, heteroalkyl.
  • X is fluoro, chloro, bromo, iodo, dimethylamino, methylamino, methoxy, ethoxy, or trifluoromethyl.
  • A is:
  • X is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, heteroalkyl.
  • X is fluoro, chloro, bromo, iodo, dimethylamino, methylamino, methoxy, ethoxy, trifluoromethyl or methoxy;
  • linker portion of the conjugates described herein is a divalent moiety that covalently links the binding agent to the maytansinoid derivatives described herein.
  • Suitable linkers include those that release the maytansinoid portion in the presence of an enzyme or at a particular pH range or value.
  • the linker comprises an enzyme-cleavable moiety.
  • Illustrative enzyme-cleavable moieties include, but are not limited to, peptide bonds, ester linkages, hydrazones, and disulfide linkages.
  • the linker comprises a cathepsin-cleavable linker.
  • the linker comprises a non-cleavable moiety. In some embodiments, the non-cleavable linker is
  • the non-cleavable linker is
  • Suitable linkers also include, but are not limited to, those that are chemically bonded to two cysteine residues of a single binding agent, e.g., antibody. Such linkers can serve to mimic the antibody's disulfide bonds that are disrupted as a result of the conjugation process.
  • the linker comprises one or more amino acids. Suitable amino acids include natural, non-natural, standard, non-standard, proteinogenic, non-proteinogenic, and L -, or D - ⁇ -amino acids.
  • the linker comprises alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, or citrulline, or derivative thereof.
  • the linker comprises valine and citrulline.
  • the linker is:
  • SP is a spacer
  • AA 1 is an amino acid
  • AA 2 is an amino acid.
  • the spacer is a divalent moiety that connects the AA 1 -AA 2 moiety to the binding agent (BA).
  • Suitable spacers include, but are not limited to, those comprising alkylene or polyethylene glycol.
  • the ends of the spacers, i.e., the portion of the spacer directly bonded to the binding agent or AA 1 can be moieties derived from reactive moieties that are used for purposes of coupling the naked antibody or AA 1 to the spacer during the chemical synthesis of the conjugate.
  • suitable spacers include, but are not limited to, a primary amine-terminated alkylene or a primary amine-terminated polyethylene glycol.
  • the primary amine-terminating end of the spacer can be directly bonded to a deglycosylated antibody or aglycosylated antibody in the presence of transglutaminase.
  • the spacer comprises an alkylene. In some embodiments, the spacer comprises a C 5-7 alkylene. In some embodiments, the spacer is:
  • b is an integer from 2 to 8.
  • the spacer comprises a primary amine-terminated alkylene. In some embodiments, the spacer comprises a NH 2 —C 5-7 alkylene. In some embodiments, the spacer is:
  • b is an integer from 2 to 8.
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • R N is a hydrogen atom or alkyl
  • R M is alkyl
  • bonds to cysteines of a binding agent are bonds to cysteines of a binding agent
  • b is an integer from 2 to 8.
  • the spacer is:
  • b is an integer from 2 to 8.
  • the spacer is:
  • g is an integer from 2 to 20. In some embodiments, g is 2-8. In some embodiments, g is 2, 4, 6, or 8.
  • the spacer is
  • n is an integer from 4 to 10. In some embodiments, n is 4, 5, 6, 7, 8, 9, or 10.
  • the spacer is:
  • the spacer is
  • n is an integer from 4 to 10. In some embodiments, n is 4, 5, 6, 7, 8, 9, or 10.
  • the spacer is
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • X is N or O
  • R N and R M are each, independently, hydrogen or alkyl
  • b is an integer from 1 to 8.
  • AA 1 -AA 2 is: valine-citrulline, citrulline-valine, lysine-phenylalanine, phenylalanine-lysine, valine-asparagine, asparagine-valine, threonine-asparagine, asparagine-threonine, serine-asparagine, asparagine-serine, phenylalanine-asparagine, asparagine-phenylalanine, leucine-asparagine, asparagine-leucine, isoleucine-asparagine, asparagine-isoleucine, glycine-asparagine, asparagine-glycine, glutamic acid-asparagine, asparagine-glutamic acid, citrulline-asparagine, asparagine-citrulline, alanine-asparagine, or asparagine-alanine.
  • AA 1 -AA 2 is: valine-citrulline or citrulline-valine. In some embodiments, AA 1 -AA 2 is: valine-citrulline.
  • the linker is:
  • SP is a spacer
  • R AA1 is an amino acid side chain
  • R AA2 is an amino acid side chain.
  • amino acid side chain refers the monovalent non-hydrogen substituent bonded to the ⁇ -carbon of an ⁇ -amino acid, including natural and non-natural amino acids.
  • exemplary amino acid side chains include, but are not limited to, the ⁇ -carbon substituent of alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, and citrulline.
  • the linker is:
  • SP is a spacer
  • the linker is:
  • b is an integer from 2 to 8.
  • the linker is:
  • b is an integer from 2 to 8.
  • BA is an antibody and the linker is:
  • b is an integer from 2 to 8.
  • BA is an antibody and the linker is:
  • b is an integer from 2 to 8.
  • BA is an antibody and the linker is:
  • R N is a hydrogen atom or alkyl
  • R M is alkyl
  • b is an integer from 2 to 8.
  • the linker is:
  • b is an integer from 2 to 8.
  • the linker is:
  • g is an integer from 2 to 20. In some embodiments, g is 2 to 8. In some embodiments, g is 2, 4, 6, or 8.
  • the linker is:
  • the linker is:
  • the linker is:
  • the linker is:
  • the linker is:
  • the linker is:
  • the linker is:
  • the linker is:
  • Suitable binding agents include, but are not limited to, antibodies, lymphokines, hormones, growth factors, viral receptors, interleukins, or any other cell binding or peptide binding molecules or substances.
  • the binding agent is an antibody.
  • the antibody is a monoclonal antibody, polyclonal antibody, antibody fragment (Fab, Fab′, and F(ab)2, minibody, diabody, tribody, and the like), or bispecific antibody.
  • Antibodies herein can be humanized using methods described in U.S. Pat. No. 6,596,541 and US Publication No. 2012/0096572, each incorporated by reference in their entirety.
  • the binding agent binds to an antigen binding partner that is a polypeptide and may be a transmembrane molecule (e.g., receptor) or a growth factor that might be glycosylated or phosphorylated.
  • an antigen binding partner that is a polypeptide and may be a transmembrane molecule (e.g., receptor) or a growth factor that might be glycosylated or phosphorylated.
  • antigens include, but are not limited to, molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor vmc, factor IX, tissue factor (TF), and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophag
  • antigens also include, but are not limited to, BCMA, SLAMF7, B7H4, GPNMB, UPK3A, and LGR5.
  • the antigens include prolactin receptor (PRLR) or prostate-specific membrane antigen (PSMA).
  • PRLR prolactin receptor
  • PSMA prostate-specific membrane antigen
  • Binding agents also include, but are not limited to, ankyrin repeat proteins, interferons, lymphokines such as IL-2 or IL-3, hormones like insulin and glucocorticoids, growth factors such as EGF, transferrin and fibronectin type III.
  • the binding agents interact with or bind to tumor antigens, including antigens specific for a type of tumor or antigens that are shared, overexpressed or modified on a particular type of tumor.
  • tumor antigens include, but are not limited to: alpha-actinin-4 with lung cancer, ARTC1 with melanoma, BCR-ABL fusion protein with chronic myeloid leukemia, B-RAF, CLPP or Cdc27 with melanoma, CASP-8 with squamous cell carcinoma, and hsp70-2 with renal cell carcinoma as well as the following shared tumor-specific antigens, for example: BAGE-1, GAGE, GnTV, KK-LC-1, MAGE-A2, NA88-A, TRP2-INT2.
  • the binding agent is an antibody. In some embodiments, the binding agent is a monoclonal antibody. In some embodiments, the binding agent is a polyclonal antibody. In some embodiments, the antibody is an anti-PSMA, anti-MUC16, or anti-EGFRvIII, or anti-STEAP-2 antibody.
  • the linkers can be bonded to the binding agent, e.g., antibody or antigen-binding molecule, through an attachment at a particular amino acid within the antibody or antigen-binding molecule.
  • exemplary amino acid attachments that can be used in the context of this aspect of the disclosure include, e.g., lysine (see, e.g., U.S. Pat. No. 5,208,020; US 2010/0129314; Hollander et al., Bioconjugate Chem., 2008, 19:358-361; WO 2005/089808; U.S. Pat. No.
  • cysteine see, e.g., US 2007/0258987; WO 2013/055993; WO 2013/055990; WO 2013/053873; WO 2013/053872; WO 2011/130598; US 2013/0101546; and U.S. Pat. No. 7,750,116
  • selenocysteine see, e.g., WO 2008/122039; and Hofer et al., Proc. Natl. Acad. Sci., USA, 2008, 105:12451-12456
  • formyl glycine see, e.g., Carrico et al., Nat. Chem.
  • Linkers can be conjugated via glutamine via transglutaminase-based chemo-enzymatic conjugation (see, e.g., Dennler et al., Bioconjugate Chem. 2014, 25, 569-578).
  • Linkers can also be conjugated to an antigen-binding protein via attachment to carbohydrates (see, e.g., US 2008/0305497, WO 2014/065661, and Ryan et al., Food & Agriculture Immunol., 2001, 13:127-130) and disulfide linkers (see, e.g., WO 2013/085925, WO 2010/010324, WO 2011/018611, WO 2014/197854, and Shaunak et al., Nat. Chem. Biol., 2006, 2:312-313).
  • the binding agent is an antibody, and the antibody is bonded to the linker through a lysine residue. In some embodiments, the antibody is bonded to the linker through a cysteine residue.
  • A is:
  • A is:
  • R 1 independently at each occurrence, is C 1-6 alkyl, C 1-6 haloalkyl, or halo;
  • A is:
  • A is:
  • R 1 independently at each occurrence, is C 1-6 alkyl, C 1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4.
  • n is an integer from 0 to 3;
  • p is an integer from 0 to 6;
  • q is an integer from 0 to 5;
  • A is:
  • A is:
  • R 1 independently at each occurrence, is C 1-6 alkyl, C 1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4.
  • n is an integer from 0 to 3;
  • p is an integer from 0 to 6;
  • q is an integer from 0 to 5;
  • SP is a spacer
  • R AA1 is an amino acid side chain
  • R AA2 is an amino acid side chain.
  • A is:
  • SP is a spacer
  • R AA1 is an amino acid side chain
  • R AA2 is an amino acid side chain.
  • A is:
  • R 1 independently at each occurrence, is C 1-6 alkyl, C 1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4.
  • n is an integer from 0 to 3;
  • p is an integer from 0 to 6;
  • q is an integer from 0 to 5;
  • A is:
  • A is:
  • R 1 independently at each occurrence, is C 1-6 alkyl, C 1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4.
  • n is an integer from 0 to 3;
  • p is an integer from 0 to 6;
  • q is an integer from 0 to 5;
  • A is:
  • A is:
  • R 1 independently at each occurrence, is C 1-6 alkyl, C 1-6 haloalkyl, or halo;
  • n, m, p, and q are 0, 1, or 2;
  • b is an integer from 2 to 8.
  • A is:
  • b is an integer from 2 to 8.
  • A is:
  • A is:
  • A is:
  • b is an integer from 2 to 8.
  • A is:
  • A is:
  • A is:
  • A is:
  • A is:
  • R 1 is the bond to the carbonyl.
  • R 1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl.
  • R 1 is trifluoromethyl.
  • R 1 is methoxy.
  • R 1 is fluoro.
  • R 1 is hydrogen.
  • A is:
  • R 1 is the bond to the carbonyl.
  • R 1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl.
  • R 1 is trifluoromethyl.
  • R 1 is methoxy.
  • R 1 is fluoro.
  • R 1 is hydrogen.
  • A is:
  • BA is an antibody
  • A is:
  • R 1 is, independently at each occurrence, is halo
  • n 0, 1, or 2;
  • A is:
  • A is:
  • R AA2 is an amino acid side chain.
  • A is:
  • b is an integer from 2 to 8.
  • A is:
  • A is:
  • R 1 independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • q is an integer from 0 to 5;
  • A is:
  • the compound of Formula I is:
  • A is arylene or heteroarylene
  • L 1 and L 2 are linkers
  • BA is a binding agent
  • k is an integer from 0 to 30
  • t is an integer from 0 to 8.
  • L 1 is a linker which binds to the BA through a lysine residue.
  • the subscript, k represents the number of linkers, L 1 , bonded to the BA through lysine residues on the BA.
  • L 2 is a linker which binds to the BA through a cysteine residue.
  • the subscript, t represents the number of linkers, L 2 , bonded to the BA through cysteine residues on the BA.
  • t when the linker, L 2 , is a monodentate linker, t is an integer from 0 to 8. In some embodiments, when the linker, L 2 , is a bidentate linker, t is an integer from 0 to 4. In some of these examples, the sum of k+t is equal to 1-8.
  • the compound of Formula I is:
  • L 1 and L 2 are linkers
  • BA is a binding agent.
  • L 1 is a linker which binds to the BA through a lysine residue.
  • L 2 is a linker which binds to the BA through a cysteine residue.
  • the compound of Formula I is:
  • L 1 and L 2 are linkers
  • BA is a binding agent.
  • L 1 is a linker which binds to the BA through a lysine residue.
  • L 2 is a linker which binds to the BA through a cysteine residue.
  • A is:
  • the linker is:
  • SP is a spacer
  • AA 1 is an amino acid
  • AA 2 is an amino acid.
  • the spacer is a divalent moiety that connects the AA 1 -AA 2 moiety to the binding agent (BA).
  • Suitable spacers include, but are not limited to, those comprising alkylene or polyethylene glycol.
  • the ends of the spacers, i.e., the portion of the spacer directly bonded to the binding agent or AA 1 can be moieties derived from reactive moieties that are used for purposes of coupling the antibody or AA 1 to the spacer during the chemical synthesis of the conjugate.
  • the spacer comprises an alkylene. In some embodiments, the spacer comprises a C 5-7 alkylene. In some embodiments, the spacer is:
  • b is an integer from 2 to 8.
  • the spacer is:
  • the spacer is:
  • R N is a hydrogen atom or alkyl
  • R M is alkyl
  • bonds to cysteines of a binding agent are bonds to cysteines of a binding agent
  • b is an integer from 2 to 8.
  • the spacer is:
  • b is an integer from 2 to 8.
  • the spacer is:
  • g is an integer from 2 to 20. In some embodiments, g is 2-8. In some embodiments, g is 2, 4, 6, or 8.
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • the spacer is:
  • X is N or O
  • R N and R M are each, independently, hydrogen or alkyl
  • b is an integer from 1 to 8.
  • AA 1 -AA 2 is: valine-citrulline, citrulline-valine, lysine-phenylalanine, phenylalanine-lysine, valine-asparagine, asparagine-valine, threonine-asparagine, asparagine-threonine, serine-asparagine, asparagine-serine, phenylalanine-asparagine, asparagine-phenylalanine, leucine-asparagine, asparagine-leucine, isoleucine-asparagine, asparagine-isoleucine, glycine-asparagine, asparagine-glycine, glutamic acid-asparagine, asparagine-glutamic acid, citrulline-asparagine, asparagine-citrulline, alanine-asparagine, or asparagine-alanine.
  • AA 1 -AA 2 is: valine-citrulline or citrulline-valine. In some embodiments, AA 1 -AA 2 is: valine-citrulline.
  • the compound of Formula I is:
  • R N and R M are each, independently, hydrogen or aryl
  • b is an integer from 1 to 8
  • A is aryl or heteroaryl
  • t is an integer from 1-8.
  • the compound of Formula I is:
  • the compound of Formula I is:
  • k is an integer from 1 to 30. In some embodiment, k is an integer from 1 to 8. In some embodiment, k is an integer from 1 to 6. In some embodiments, k is an integer from 1 to 4. In some embodiments, k is an integer from 1 to 3. In some embodiments, the drug-antibody ratio (DAR) of the conjugate is from 1.0 to 3.0.
  • DAR drug-antibody ratio
  • A is arylene or heteroarylene.
  • these compounds represent the payload portion of the conjugates described herein and are released, e.g., by enzyme proteolysis, following internalization of the conjugate into a cell.
  • the methods provided herein include methods of treating a proliferative disease, e.g., cancer, comprising administering to a patient a therapeutically effective amount of a conjugate, e.g., antibody-drug conjugate that releases a compound of Formula (II) following internalization of said conjugate into a cell in said patient.
  • these compounds represent the metabolic product of the conjugates described herein, e.g., enzyme proteolysis product. In some embodiments, these compounds represent the catabolic product of the conjugates described herein. In some embodiments, these compounds represent the cellular product of the conjugates described herein.
  • A is a divalent radical of benzene, of pyridine, of naphthalene, or of quinolone, which are optionally substituted.
  • A is arylene
  • A is:
  • the compound of Formula (II) is a compound of the Formula (IIA):
  • R 1 and n are as defined herein.
  • the compound of Formula (II) is a compound of the Formula (IIB):
  • R 1 and q are as defined herein.
  • the compound of Formula (II) is a compound of the Formula (IIB2):

Abstract

Provided herein are maytansinoid derivatives, conjugates thereof, and methods of treating or preventing proliferative diseases with the same.

Description

  • This application claims priority to, and the benefit of, U.S. Provisional Patent Application No. 62/139,044, entitled MAYTANSINOID DERIVATIVES, CONJUGATES THEREOF AND METHODS OF TREATING PROLIFERATIVE DISEASES USING THE SAME, which was filed Mar. 27, 2015, and also claims priority to, and the benefit of U.S. Provisional Patent Application No. 62/252,239, entitled MAYTANSINOID DERIVATIVES, CONJUGATES THEREOF AND METHODS OF TREATING PROLIFERATIVE DISEASES USING THE SAME, which was filed Nov. 6, 2015. The contents of each of these provisional patent applications are herein incorporated by reference in their entirety for all purposes.
  • FIELD
  • The present disclosure concerns maytansinoid derivatives, conjugates thereof, and methods of treating or preventing proliferative diseases with the same.
  • BACKGROUND
  • Proliferative diseases, for example cancer, are characterized by the uncontrolled growth of abnormal cells. Current treatments of proliferative diseases include surgery, radiation, chemotherapy, hormone-based therapy and/or immunotherapy. A number of these treatments, particularly chemotherapy, utilize anti-proliferative drugs that limit the spread of the abnormal cells. However, these drugs are typically indiscriminate in their ability to kill cells, affecting both normal and abnormal cells. To address this problem, various approaches to targeted drug delivery have been explored, including the use of conjugates of tumor-targeted probes (such as antibodies or growth factors) with toxins, to selectively target abnormal cells. Antibody drug conjugates (ADCs) are compounds composed of an antibody that is linked, via a chemical linker, to a cytotoxic agent. Such compounds leverage the antibody's binding specificity for its target to deliver a cytotoxic agent to an abnormal cell. Thus, there is a need for anti-proliferative compounds and their conjugates.
  • SUMMARY
  • Provided herein are compounds of Formula (I):
  • Figure US20160375147A1-20161229-C00001
  • or a pharmaceutically acceptable salt thereof,
    wherein:
  • A is arylene or heteroarylene;
  • L is a linker;
  • BA is a binding agent; and
  • k is an integer from 1 to 30. Also provided herein are stereoisomers of compounds of Formula (I).
  • Provided herein are also compounds of Formula (II):
  • Figure US20160375147A1-20161229-C00002
  • or a pharmaceutically acceptable salt thereof, wherein A is arylene or heteroarylene. Also provided herein are stereoisomers of compounds of Formula (II).
  • Provided herein are also compounds of Formula PP5:
  • Figure US20160375147A1-20161229-C00003
  • or a salt thereof, wherein A is arylene or heteroarylene. Also provided herein are stereoisomers of compounds of Formula PP5.
  • Provided herein are also compounds of Formula PT1:
  • Figure US20160375147A1-20161229-C00004
  • or a salt thereof, wherein A is arylene or heteroarylene and L is a linker. Also provided herein are stereoisomers of compounds of Formula PT1.
  • Furthermore, provided herein are methods of treating proliferative diseases comprising administering the compounds described herein.
  • Furthermore, provided herein are methods of treating proliferative diseases comprising administering the conjugates described herein.
  • Furthermore, provided herein are of methods of preparing compounds of Formula (I) comprising reacting a deglycosylated antibody or aglycosylated antibody with a compound of Formula (PT1) in the presence of transglutaminase.
  • BRIEF DESCRIPTIONS OF THE DRAWINGS
  • FIG. 1 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-4-aminobenzamido-citrulline-valine-caprolyl-6-maleimidyl.
  • FIG. 2 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 3 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 4 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 5 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 41.
  • FIG. 6 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(4-amino-2-fluoro)benzamido-Cit-Val-Cap-Mal.
  • FIG. 7 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(4-amino-2-trifluoromethyl)benzamido-Cit-Val-Cap-Mal.
  • FIG. 8 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(4-amino-2-methoxy)benzamido-Cit-Val-Cap-Mal.
  • FIG. 9 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-4-aminobenzamide.
  • FIG. 10 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-fluoro-4-amino)benzamide
  • FIG. 11 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-trifluoromethyl-4-amino)benzamide.
  • FIG. 12 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-methoxy-4-amino)benzamide.
  • FIG. 13 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-trifluoromethyl-4-amino)benzamide.
  • FIG. 14 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-chloro-4-amino-5-fluoro)benzamide.
  • FIG. 15 depicts a general synthetic sequence for preparing compounds of Formula (II) wherein substituent R is defined herein and below.
  • FIG. 16 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2,5-difluoro-4-amino)benzamide.
  • FIG. 17 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-fluoro-4-amino)benzamide
  • FIG. 18 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-chloro-4-amino)benzamide.
  • FIG. 19 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(5-amino-8-carboxyquinoline)carboxamide.
  • FIG. 20 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-bromo-4-amino)benzamide.
  • FIG. 21 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-methoxy-4-amino)benzamide.
  • FIG. 22 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-methyl-4-amino)benzamide.
  • FIG. 23 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-methyl-4-amino)benzamide.
  • FIG. 24 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(8-amino-5-carboxyquinoline)carboxamide.
  • FIG. 25 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(3-methoxy-4-amino)benzamido-Cit-Val-Cap-Mal.
  • FIG. 26 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-(2-fluoro-4-amino)benzamido-Cit-Val-Cap-6-amine.
  • FIG. 27 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-methoxy-5-amino)benzamide.
  • FIG. 28 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino-4-methoxy)benzamide.
  • FIG. 29 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino-5-fluoro)benzamide.
  • FIG. 30 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-fluoro-5-amino)benzamide.
  • FIG. 31 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino)benzamide.
  • FIG. 32 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-amino-4-fluoro)benzamide.
  • FIG. 33 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-4-aminobenzamide-adipic-NHS.
  • FIG. 34 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-4-aminobenzamide-Cap-Mal.
  • FIG. 35 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-methylsulfonyl-4-amino)benzamide.
  • FIG. 36 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-hydroxy-4-amino)benzamide.
  • FIG. 37 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(2-amino)benzamide.
  • FIG. 38 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(4-methoxy-2-amino)benzamide.
  • FIG. 39 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-morpholino-4-amino)benzamide.
  • FIG. 40 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-(3-acetamido-4-amino)benzamide.
  • FIG. 41 depicts a synthetic sequence for preparing maytansin-N-methyl-L-alanine-N-4-aminobenzamide-Cit-Val-cap-diBromomethylacryl.
  • FIG. 42 depicts the deconvoluted mass spectroscopy (MS) spectrum of the antibody drug conjugate, PRLR-Q-63 conjugate from EXAMPLE 43.
  • FIG. 43 depicts the deconvoluted MS spectrum of the Isotype Control-Q-63 conjugate from EXAMPLE 43.
  • FIG. 44 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 45.
  • FIG. 45 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 45.
  • FIG. 46 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 45.
  • FIG. 47 depicts the plot of % Cell Viability vs. Log10 [M] of certain compounds tested in EXAMPLE 45.
  • DETAILED DESCRIPTION A. Definitions
  • As used herein, “alkyl” refers to a monovalent and saturated hydrocarbon radical moiety. Alkyl is optionally substituted and can be linear, branched, or cyclic, i.e., cycloalkyl. Alkyl includes, but is not limited to, those having 1-20 carbon atoms, i.e., C1-20 alkyl; 1-12 carbon atoms, i.e., C1-12 alkyl; 1-8 carbon atoms, i.e., C1-8 alkyl; 1-6 carbon atoms, i.e., C1-6 alkyl; and 1-3 carbon atoms, i.e., C1-3 alkyl. Examples of alkyl moieties include, but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, i-butyl, a pentyl moiety, a hexyl moiety, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • As used herein, “haloalkyl” refers to alkyl, as defined above, wherein the alkyl includes at least one substituent selected from a halogen, e.g., F, Cl, Br, or I.
  • As used herein, “alkenyl” refers to a monovalent hydrocarbon radical moiety containing at least two carbon atoms and one or more non-aromatic carbon-carbon double bonds. Alkenyl is optionally substituted and can be linear, branched, or cyclic. Alkenyl includes, but is not limited to, those having 2-20 carbon atoms, i.e., C2-20 alkenyl; 2-12 carbon atoms, i.e., C2-12 alkenyl; 2-8 carbon atoms, i.e., C2-8 alkenyl; 2-6 carbon atoms, i.e., C2-6 alkenyl; and 2-4 carbon atoms, i.e., C2-4 alkenyl. Examples of alkenyl moieties include, but are not limited to vinyl, propenyl, butenyl, and cyclohexenyl.
  • As used herein, “alkynyl” refers to a monovalent hydrocarbon radical moiety containing at least two carbon atoms and one or more carbon-carbon triple bonds. Alkynyl is optionally substituted and can be linear, branched, or cyclic. Alkynyl includes, but is not limited to, those having 2-20 carbon atoms, i.e., C2-20 alkynyl; 2-12 carbon atoms, i.e., C2-12 alkynyl; 2-8 carbon atoms, i.e., C2-8 alkynyl; 2-6 carbon atoms, i.e., C2-6 alkynyl; and 2-4 carbon atoms, i.e., C2-4 alkynyl. Examples of alkynyl moieties include, but are not limited to ethynyl, propynyl, and butynyl.
  • As used herein, “alkoxy” refers to a monovalent and saturated hydrocarbon radical moiety wherein the hydrocarbon includes a single bond to an oxygen atom and wherein the radical is localized on the oxygen atom, e.g., CH3CH2—O for ethoxy. Alkoxy substituents bond to the compound which they substitute through this oxygen atom of the alkoxy substituent. Alkoxy is optionally substituted and can be linear, branched, or cyclic, i.e., cycloalkoxy. Alkoxy includes, but is not limited to, those having 1-20 carbon atoms, i.e., C1-20 alkoxy; 1-12 carbon atoms, i.e., C1-12 alkoxy; 1-8 carbon atoms, i.e., C1-8 alkoxy; 1-6 carbon atoms, i.e., C1-6 alkoxy; and 1-3 carbon atoms, i.e., C1-3 alkoxy. Examples of alkoxy moieties include, but are not limited to methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, i-butoxy, a pentoxy moiety, a hexoxy moiety, cyclopropoxy, cyclobutoxy, cyclopentoxy, and cyclohexoxy.
  • As used herein, “haloalkoxy” refers to alkoxy, as defined above, wherein the alkoxy includes at least one substituent selected from a halogen, e.g., F, Cl, Br, or I.
  • As used herein, “aryl” refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms are carbon atoms. Aryl is optionally substituted and can be monocyclic or polycyclic, e.g., bicyclic or tricyclic. Examples of aryl moieties include, but are not limited to those having 6 to 20 ring carbon atoms, i.e., C6-20 aryl; 6 to 15 ring carbon atoms, i.e., C6-15 aryl, and 6 to 10 ring carbon atoms, i.e., C6-10 aryl. Examples of aryl moieties include, but are limited to phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, and pyrenyl.
  • As used herein, “arylene” refers to a divalent moiety of an aromatic compound wherein the ring atoms are only carbon atoms. Arylene is optionally substituted and can be monocyclic or polycyclic, e.g., bicyclic or tricyclic. Examples of aryl moieties include, but are not limited to those having 6 to 20 ring carbon atoms, i.e., C6-20 arylene; 6 to 15 ring carbon atoms, i.e., C6-15 arylene, and 6 to 10 ring carbon atoms, i.e., C6-10 arylene.
  • As used herein, “alkaryl” refers to an aryl that is substituted with at least one alkyl. Alkaryl is optionally substituted.
  • As used herein, “heteroalkyl” refers to an alkyl in which one or more carbon atoms are replaced by heteroatoms. As used herein, “heteroalkenyl” refers to an alkenyl in which one or more carbon atoms are replaced by heteroatoms. As used herein, “heteroalkynyl” refers to an alkenyl in which one or more carbon atoms are replaced by heteroatoms. Suitable heteroatoms include, but are not limited to, nitrogen, oxygen, and sulfur atoms. Heteroalkyl is optionally substituted. Examples of heteroalkyl moieties include, but are not limited to, aminoalkyl, sulfonylalkyl, sulfinylalkyl. Examples of heteroalkyl moieties also include, but are not limited to, methylamino, methylsulfonyl, and methylsulfinyl.
  • As used herein, “heteroaryl” refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms contain carbon atoms and at least one oxygen, sulfur, nitrogen, or phosphorus atom. Examples of heteroaryl moieties include, but are not limited to those having 5 to 20 ring atoms; 5 to 15 ring atoms; and 5 to 10 ring atoms. Heteroaryl is optionally substituted.
  • As used herein, “heteroarylene” refers to an arylene in which one or more ring atoms of the aromatic ring are replaced with an oxygen, sulfur, nitrogen, or phosphorus atom. Heteroarylene is optionally substituted.
  • As used herein, “heterocycloalkyl” refers to a cycloalkyl in which one or more carbon atoms are replaced by heteroatoms. Suitable heteroatoms include, but are not limited to, nitrogen, oxygen, and sulfur atoms. Heterocycloalkyl is optionally substituted. Examples of heterocycloalkyl moieties include, but are not limited to, morpholinyl, piperidinyl, tetrahydropyranyl, pyrrolidinyl, imidazolidinyl, oxazolidinyl, thiazolidinyl, dioxolanyl, dithiolanyl, oxanyl, or thianyl.
  • As used herein, “optionally substituted,” when used to describe a radical moiety, e.g., optionally substituted alkyl, means that such moiety is optionally bonded to one or more substituents. Examples of such substituents include, but are not limited to halo, cyano, nitro, haloalkyl, azido, epoxy, optionally substituted heteroaryl, optionally substituted heterocycloalkyl,
  • Figure US20160375147A1-20161229-C00005
  • wherein RA, RB, and RC are, independently at each occurrence, a hydrogen atom, alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, heteroalkyl, heteroaryl, or heterocycloalkyl, or RA and RB, together with the atoms to which they are bonded, form a saturated or unsaturated carbocyclic ring, wherein the ring is optionally substituted and wherein one or more ring atoms is optionally replaced with a heteroatom. In some embodiments, RA, RB, and RC are not hydrogen atoms. In some examples, RA is methyl. In some examples, RA is methylamino, methylsulfonyl, and methylsulfinyl. In some examples, RA is methylamino. In certain embodiments, when a radical moiety is optionally substituted with an optionally substituted heteroaryl, optionally substituted heterocycloalkyl, or optionally substituted saturated or unsaturated carbocyclic ring, the substituents on the optionally substituted heteroaryl, optionally substituted heterocycloalkyl, or optionally substituted saturated or unsaturated carbocyclic ring, if they are substituted, are not substituted with substituents which are further optionally substituted with additional substituents. In some embodiments, when a group described herein is optionally substituted, the substituent bonded to the group is unsubstituted unless otherwise specified.
  • As used herein, “binding agent” refers to any molecule capable of binding with specificity to a given binding partner.
  • As used herein, “linker” refers to a divalent moiety that covalently links the binding agent to the maytansinoid derivatives described herein.
  • As used herein, “amide synthesis conditions” refers to reaction conditions suitable to effect the formation of an amide, e.g., by the reaction of a carboxylic acid, activated carboxylic acid, or acyl halide with an amine. In some examples, amide synthesis conditions refers to reaction conditions suitable to effect the formation of an amide bond between a carboxylic acid and an amine. In some of these examples, the carboxylic acid is first converted to an activated carboxylic acid before the activated carboxylic acid reacts with an amine to form an amide. Suitable conditions to effect the formation of an amide include, but are not limited to, those utilizing reagents to effect the reaction between a carboxylic acid an amine, including, but not limited to, dicyclohexylcarbodiimide (DCC), diisopropylcarbodiimide (DIC), (benzotriazol-1-yloxy)tris(dimethylamino)phosphonium hexafluorophosphate (BOP), (benzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyBOP), (7-azabenzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyAOP), bromotripyrrolidinophosphonium hexafluorophosphate (PyBrOP), O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HBTU), O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU), 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (HATU), 2-Ethoxy-1-ethoxycarbonyl-1,2-dihydroquinoline (EEDQ), 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), 2-Chloro-1,3-dimethylimidazolidinium hexafluorophosphate (CIP), 2-chloro-4,6-dimethoxy-1,3,5-triazine (CDMT), and carbonyldiimidazole (CDI). In some examples, a carboxylic acid is first converted to an activated carboxylic ester before reacting with an amine to form an amide bond. In certain embodiments, the carboxylic acid is reacted with a reagent. The reagent activates the carboxylic acid by deprotonating the carboxylic acid and then forming a product complex with the deprotonated carboxylic acid as a result of nucleophilic attack by the deprotonated carboxylic acid onto the protonated reagent. For certain carboxylic acids, this activated ester is more susceptible subsequently to nucleophilic attack by an amine than the carboxylic acid is before it is converted. This results in amide bond formation. As such, the carboxylic acid is described as activated. Exemplary reagents include DCC and DIC.
  • As used herein, “therapeutically effective amount” refers to an amount (of a compound) that is sufficient to provide a therapeutic benefit to a patient in the treatment or management of a disease or disorder, or to delay or minimize one or more symptoms associated with the disease or disorder.
  • Certain groups, moieties, substituents, and atoms are depicted with a wiggly line that intersects a bond or bonds to indicate the atom through which the groups, moieties, substituents, atoms are bonded. For example, a phenyl group that is substituted with a propyl group depicted as:
  • Figure US20160375147A1-20161229-C00006
  • has the following structure:
  • Figure US20160375147A1-20161229-C00007
  • As used herein, illustrations showing substituents bonded to a cyclic group (e.g., aromatic, heteroaromatic, fused ring, and saturated or unsaturated cycloalkyl or heterocycloalkyl) through a bond between ring atoms are meant to indicate, unless specified otherwise, that the cyclic group may be substituted with that substituent at any ring position in the cyclic group or on any ring in the fused ring group, according to techniques set forth herein or which are known in the field to which the instant disclosure pertains. For example, the group,
  • Figure US20160375147A1-20161229-C00008
  • herein subscript q is an integer from 0 to 4 and in which the positions of substituent R1 are described generically, includes the following groups in which the positions of substituent R1 are described specifically:
  • Figure US20160375147A1-20161229-C00009
    Figure US20160375147A1-20161229-C00010
  • In addition and for example, the group,
  • Figure US20160375147A1-20161229-C00011
  • in which the positions of substituents other than R1 which are bonded to the cyclic group through a bond between ring atoms are described generically, includes the following groups in which the positions of these substituents other than R1 are described specifically:
  • Figure US20160375147A1-20161229-C00012
    Figure US20160375147A1-20161229-C00013
  • Also, for example, the group,
  • Figure US20160375147A1-20161229-C00014
  • in which the positions of substituents other than R1 which are bonded to the cyclic group through a bond between ring atoms are described generically, includes the following groups in which the positions of these substituents other than R1 are described specifically:
  • Figure US20160375147A1-20161229-C00015
    Figure US20160375147A1-20161229-C00016
  • each of these structures in which the positions of the substituents other than R1 are described specifically, the substituent R1 may be bonded to any ring position in the cyclic group or on any ring in the fused ring group which is not occupied by one of these substituents other than R1. The following non-limiting representative illustrations indicate that the cyclic group can be substituted with the indicated substituent at any ring position or on either ring in the fused ring group:
  • Figure US20160375147A1-20161229-C00017
  • B. Conjugates
  • Provided herein are compounds of Formula (I):
  • Figure US20160375147A1-20161229-C00018
  • or a pharmaceutically acceptable salt thereof,
    wherein:
  • A is arylene or heteroarylene;
  • L is a linker;
  • BA is a binding agent; and
  • k is an integer from 1 to 30.
  • 1. “A” Moieties
  • In some embodiments, A is arylene. In some embodiments, A is heteroarylene. In some embodiments, the arylene or heteroarylene is substituted with one or more electron withdrawing groups and/or one or more electron donating groups.
  • In some embodiments, A is a divalent radical of benzene, of pyridine, of naphthalene, or of quinolone, which are optionally substituted.
  • In some embodiments, A is a divalent radical of benzene which is optionally substituted with a member selected from the group consisting of amino, amido, alkyl, halo, haloalkyl, alkoxy, and haloalkoxy.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00019
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, halo, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00020
      • or azido,
        • wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and q is an integer from 0 to 5.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00021
  • wherein:
      • R1 is, independently at each occurrence, halo, haloalkyl, haloalkoxy, hydroxyl, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, aryl, alkaryl, aralkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00022
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and q is an integer from 0 to 5.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00023
  • wherein:
      • R1 is, independently at each occurrence, halo, haloalkyl, hydroxyl, alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00024
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, R1 is C1-6 alkyl or C1-6 alkoxy. In some of these embodiments, R1 is methyl, ethyl, methoxy, or ethoxy. In some of these embodiments, R1 is methoxy.
  • In some embodiments, R1 is, independently, alkyl or halo. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 haloalkyl, or halo. In some embodiments, R1 is, independently, halo. In some embodiments, R1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, R1 is
  • Figure US20160375147A1-20161229-C00025
  • In some embodiments, R1 is
  • Figure US20160375147A1-20161229-C00026
  • wherein RA is methyl. In some embodiments, R1 is hydroxyl. In some embodiments, R1 is N-methylformamide. In some embodiments, R1 is morpholinyl.
  • In some embodiments, R1 is, independently, alkyl, alkoxy, or halo. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 alkoxy, C1-6 haloalkyl, C1-6 haloalkoxy, or halo. In some embodiments, R1 is, independently, halo. In some embodiments, R1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00027
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00028
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00029
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00030
  • wherein n is 0, 1 or 2.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00031
  • wherein n is 0, 1 or 2.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00032
  • wherein n is 0 or 1; and R1 is alkoxy, halo, or haloalkyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00033
  • wherein n is 0 or 1; and R1 is alkoxy, halo, or haloalkyl. In some examples, R1 is alkoxy.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00034
  • wherein n is 0 or 1; and R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00035
  • wherein n is 0 or 1; and R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl. In some examples, R1 is C1-6 alkoxy.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00036
  • wherein n is 0 or 1; R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl; and L is
  • Figure US20160375147A1-20161229-C00037
  • wherein b is an integer from 2 to 8 and
  • Figure US20160375147A1-20161229-C00038
  • is a bond to the binding agent.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00039
  • wherein n is 0 or 1; R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl; and L is
  • Figure US20160375147A1-20161229-C00040
  • wherein b is an integer from 2 to 8 and
  • Figure US20160375147A1-20161229-C00041
  • is a bond to the binding agent.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00042
  • wherein n is 0, 1, 2, 3, or 4.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00043
  • wherein n is 0, 1, 2, 3, or 4.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00044
  • wherein:
  • R1 is C1-6 alkyl, halo, or C1-6 haloalkyl; and
  • n is 0, 1 or 2.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00045
  • wherein:
      • R1 is C1-6 alkyl, halo, or C1-6 haloalkyl; and
      • n is 0, 1 or 2.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00046
  • wherein:
  • R1 is C1-6 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, or C1-6 haloalkoxy; and
  • n is 0, 1, 2, 3, or 4.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00047
  • wherein:
  • R1 is C1-6 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, or C1-6 haloalkoxy; and
  • n is 0, 1, 2, 3, or 4.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00048
  • wherein R1 is C1-6 alkyl, C1-6 alkoxy, halo, or C1-6 haloalkyl. In certain of these embodiments, R1 is methoxy or methyl. In some specific embodiments, R1 is methoxy.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00049
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00050
  • wherein:
  • R1 is halo or trifluoromethyl; and
  • n is 0, 1 or 2.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00051
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00052
  • wherein:
  • X is a hydrogen atom, halo, or trifluoromethyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00053
  • wherein:
  • X is a hydrogen atom, halo, or trifluoromethyl;
  • Figure US20160375147A1-20161229-C00054
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00055
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00056
  • wherein:
      • R1 is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, aryl, heteroalkyl, halo, haloalkyl, haloalkoxy or hydroxyl;
  • Figure US20160375147A1-20161229-C00057
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00058
  • is the bond to the carbonyl. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen. In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00059
  • wherein:
      • R1 is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, aryl, heteroalkyl, halo, haloalkyl, haloalkoxy, or hydroxyl;
  • Figure US20160375147A1-20161229-C00060
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00061
  • is the bond to the carbonyl. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, R1 is sulfonyl. In some embodiments, R1 is N-methylformamide. In some embodiments, R1 is hydroxyl. In some embodiments, R1 is morpholinyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00062
  • wherein:
  • R1 is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, aryl, heteroalkyl, halo, haloalkyl, or haloalkoxy;
  • Figure US20160375147A1-20161229-C00063
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00064
  • is the bond to the carbonyl. In some embodiments, R1 is alkyl or alkoxy. In some specific embodiments, R1 is propylamino, difluoro-methoxy, phenyl, 2-fluorophenyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00065
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00066
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00067
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00068
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00069
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00070
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00071
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00072
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00073
  • wherein X is F, Cl, Br, CN, methoxy, dimethylamino or cyclopropyl;
  • Figure US20160375147A1-20161229-C00074
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00075
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00076
  • wherein X is F, Cl, Br, CN, methoxy, dimethylamino, 1-methyl-ethyl-thio or cyclopropyl;
  • Figure US20160375147A1-20161229-C00077
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00078
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00079
  • wherein each R1 is independently, at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkyl, or haloalkoxy;
  • Figure US20160375147A1-20161229-C00080
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00081
  • is the bond to the carbonyl. In some embodiments, R1 is hydrogen, fluoro, trifluoromethyl, or methoxy. In some embodiments, R1 is fluoro, chloro, bromo, or iodo.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00082
  • wherein
  • Figure US20160375147A1-20161229-C00083
  • wherein is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00084
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00085
  • wherein each R1 is independently, at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkyl, or haloalkoxy,
  • Figure US20160375147A1-20161229-C00086
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00087
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00088
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00089
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00090
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00091
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00092
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00093
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00094
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00095
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00096
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00097
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00098
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00099
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00100
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00101
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00102
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00103
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00104
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00105
  • wherein:
  • Figure US20160375147A1-20161229-C00106
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00107
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00108
  • wherein:
  • Figure US20160375147A1-20161229-C00109
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00110
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00111
  • wherein:
  • Figure US20160375147A1-20161229-C00112
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00113
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00114
  • wherein:
  • Figure US20160375147A1-20161229-C00115
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00116
  • is the bond to the carbonyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00117
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00118
  • wherein n is 0, 1 2, or 3.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00119
  • wherein:
  • R1 is C1-6 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, C1-6 haloalkoxy, C1-6 heteroalkyl; and
  • n is 0, 1 2, 3 or 4.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00120
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00121
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00122
  • wherein:
  • X is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, heteroalkyl. In some embodiments, X is fluoro, chloro, bromo, iodo, dimethylamino, methylamino, methoxy, ethoxy, or trifluoromethyl.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00123
  • wherein:
  • X is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, heteroalkyl. In some embodiments, X is fluoro, chloro, bromo, iodo, dimethylamino, methylamino, methoxy, ethoxy, trifluoromethyl or methoxy;
  • Figure US20160375147A1-20161229-C00124
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00125
  • is the bond to the carbonyl.
  • 2. Linkers
  • The linker portion of the conjugates described herein is a divalent moiety that covalently links the binding agent to the maytansinoid derivatives described herein. Suitable linkers include those that release the maytansinoid portion in the presence of an enzyme or at a particular pH range or value.
  • In some embodiments, the linker comprises an enzyme-cleavable moiety. Illustrative enzyme-cleavable moieties include, but are not limited to, peptide bonds, ester linkages, hydrazones, and disulfide linkages. In some embodiments, the linker comprises a cathepsin-cleavable linker.
  • In some embodiments, the linker comprises a non-cleavable moiety. In some embodiments, the non-cleavable linker is
  • Figure US20160375147A1-20161229-C00126
  • or a residue thereof. In some embodiments, the non-cleavable linker is
  • Figure US20160375147A1-20161229-C00127
  • or a residue thereof.
  • Suitable linkers also include, but are not limited to, those that are chemically bonded to two cysteine residues of a single binding agent, e.g., antibody. Such linkers can serve to mimic the antibody's disulfide bonds that are disrupted as a result of the conjugation process.
  • In some embodiments, the linker comprises one or more amino acids. Suitable amino acids include natural, non-natural, standard, non-standard, proteinogenic, non-proteinogenic, and L-, or D-α-amino acids. In some embodiments, the linker comprises alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, or citrulline, or derivative thereof.
  • In some embodiments, the linker comprises valine and citrulline.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00128
  • wherein:
  • SP is a spacer;
  • Figure US20160375147A1-20161229-C00129
  • is one or more bonds to the binding agent;
  • AA1 is an amino acid; and
  • AA2 is an amino acid.
  • The spacer is a divalent moiety that connects the AA1-AA2 moiety to the binding agent (BA). Suitable spacers include, but are not limited to, those comprising alkylene or polyethylene glycol. The ends of the spacers, i.e., the portion of the spacer directly bonded to the binding agent or AA1, can be moieties derived from reactive moieties that are used for purposes of coupling the naked antibody or AA1 to the spacer during the chemical synthesis of the conjugate.
  • In some examples, suitable spacers include, but are not limited to, a primary amine-terminated alkylene or a primary amine-terminated polyethylene glycol. The primary amine-terminating end of the spacer can be directly bonded to a deglycosylated antibody or aglycosylated antibody in the presence of transglutaminase.
  • In some embodiments, the spacer comprises an alkylene. In some embodiments, the spacer comprises a C5-7 alkylene. In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00130
  • wherein:
  • Figure US20160375147A1-20161229-C00131
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer comprises a primary amine-terminated alkylene. In some embodiments, the spacer comprises a NH2—C5-7 alkylene. In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00132
  • wherein:
  • Figure US20160375147A1-20161229-C00133
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00134
  • wherein:
  • Figure US20160375147A1-20161229-C00135
  • is a bond to the binding agent.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00136
  • wherein:
  • Figure US20160375147A1-20161229-C00137
  • is a bond to the binding agent.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00138
  • wherein:
  • RN is a hydrogen atom or alkyl;
  • RM is alkyl;
  • the two bonds represented by
  • Figure US20160375147A1-20161229-C00139
  • are bonds to cysteines of a binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00140
  • wherein:
  • Figure US20160375147A1-20161229-C00141
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00142
  • wherein:
  • Figure US20160375147A1-20161229-C00143
  • is a bond to the binding agent; and
  • g is an integer from 2 to 20. In some embodiments, g is 2-8. In some embodiments, g is 2, 4, 6, or 8.
  • In some embodiments, the spacer is
  • Figure US20160375147A1-20161229-C00144
  • wherein n is an integer from 4 to 10. In some embodiments, n is 4, 5, 6, 7, 8, 9, or 10.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00145
  • In some embodiments, the spacer is
  • Figure US20160375147A1-20161229-C00146
  • wherein n is an integer from 4 to 10. In some embodiments, n is 4, 5, 6, 7, 8, 9, or 10.
  • In some embodiments, the spacer is
  • Figure US20160375147A1-20161229-C00147
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00148
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00149
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00150
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00151
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00152
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00153
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00154
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00155
  • wherein
  • Figure US20160375147A1-20161229-C00156
  • is a bond to the binding agent;
    X is N or O; RN and RM are each, independently, hydrogen or alkyl; and b is an integer from 1 to 8.
  • In some embodiments, AA1-AA2 is: valine-citrulline, citrulline-valine, lysine-phenylalanine, phenylalanine-lysine, valine-asparagine, asparagine-valine, threonine-asparagine, asparagine-threonine, serine-asparagine, asparagine-serine, phenylalanine-asparagine, asparagine-phenylalanine, leucine-asparagine, asparagine-leucine, isoleucine-asparagine, asparagine-isoleucine, glycine-asparagine, asparagine-glycine, glutamic acid-asparagine, asparagine-glutamic acid, citrulline-asparagine, asparagine-citrulline, alanine-asparagine, or asparagine-alanine.
  • In some embodiments, AA1-AA2 is: valine-citrulline or citrulline-valine. In some embodiments, AA1-AA2 is: valine-citrulline.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00157
  • wherein:
  • SP is a spacer;
  • Figure US20160375147A1-20161229-C00158
  • is one or more bonds to the binding agent;
  • RAA1 is an amino acid side chain; and
  • RAA2 is an amino acid side chain.
  • As used herein, “amino acid side chain” refers the monovalent non-hydrogen substituent bonded to the α-carbon of an α-amino acid, including natural and non-natural amino acids. Exemplary amino acid side chains include, but are not limited to, the α-carbon substituent of alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, and citrulline.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00159
  • wherein:
  • SP is a spacer; and
  • Figure US20160375147A1-20161229-C00160
  • is one or more bonds to the binding agent.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00161
  • wherein:
  • Figure US20160375147A1-20161229-C00162
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00163
  • wherein:
  • Figure US20160375147A1-20161229-C00164
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, BA is an antibody and the linker is:
  • Figure US20160375147A1-20161229-C00165
  • wherein:
  • the two bonds represented by
  • Figure US20160375147A1-20161229-C00166
  • are bonds to cysteines of the antibody; and
  • b is an integer from 2 to 8.
  • In some embodiments, BA is an antibody and the linker is:
  • Figure US20160375147A1-20161229-C00167
  • wherein:
    the two bonds represented by
  • Figure US20160375147A1-20161229-C00168
  • are bonds to cysteines of the antibody; and
  • b is an integer from 2 to 8.
  • In some embodiments, BA is an antibody and the linker is:
  • Figure US20160375147A1-20161229-C00169
  • wherein:
  • RN is a hydrogen atom or alkyl;
  • RM is alkyl;
  • the two bonds represented by
  • Figure US20160375147A1-20161229-C00170
  • are bonds to cysteines of the antibody; and
  • b is an integer from 2 to 8.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00171
  • wherein:
  • Figure US20160375147A1-20161229-C00172
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00173
  • wherein:
  • Figure US20160375147A1-20161229-C00174
  • is a bond to the binding agent; and
  • g is an integer from 2 to 20. In some embodiments, g is 2 to 8. In some embodiments, g is 2, 4, 6, or 8.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00175
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00176
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00177
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00178
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00179
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00180
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00181
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00182
  • 3. Binding Agents
  • Suitable binding agents include, but are not limited to, antibodies, lymphokines, hormones, growth factors, viral receptors, interleukins, or any other cell binding or peptide binding molecules or substances.
  • In some embodiments, the binding agent is an antibody. In some embodiments, the antibody is a monoclonal antibody, polyclonal antibody, antibody fragment (Fab, Fab′, and F(ab)2, minibody, diabody, tribody, and the like), or bispecific antibody. Antibodies herein can be humanized using methods described in U.S. Pat. No. 6,596,541 and US Publication No. 2012/0096572, each incorporated by reference in their entirety.
  • Where the binding agent is an antibody, it binds to an antigen binding partner that is a polypeptide and may be a transmembrane molecule (e.g., receptor) or a growth factor that might be glycosylated or phosphorylated. Exemplary antigens include, but are not limited to, molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor vmc, factor IX, tissue factor (TF), and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophage inflammatory protein (MIP-I-alpha); a serum albumin, such as human serum albumin; Muellerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; a microbial protein, such as betalactamase; DNase; 19E; a cytotoxic T-lymphocyte associated antigen (CTLA), such as CTLA-4; inhibin; activin; vascular endothelial growth factor (VEGF); receptors for hormones or growth factors; protein A or D; rheumatoid factors; a neurotrophic factor such as bone-derived neurotrophic factor (BDNF), neurotrophin-3, -4, -5, or -6 (NT-3, NT4, NT-5, or NT-6), or a nerve growth factor such as NGF-13; platelet-derived growth factor (PDGF); fibroblast growth factor such as aFGF and bFGF; fibroblast growth factor receptor 2 (FGFR2), epidermal growth factor (EGF); transforming growth factor (TGF) such as TGF-alpha and TGF-beta, including TGF-β1, TGF-β2, TGF-β3, TGF-β4, or TGF-β5; insulin-like growth factor-1 and -II (IGF-1 and IGF-II); des(I-3)-IGF-1 (brain IGF-1), insulin-like growth factor binding proteins, EpCAM, GD3, FLT3, PSMA, PSCA, MUC1, MUC16, STEAP, CEA, TENB2, EphA receptors, EphB receptors, folate receptor, FOLRI, mesothelin, cripto, alphavbeta6, integrins, VEGF, VEGFR, EGFR, transferrin receptor, IRTA1, IRTA2, IRTA3, IRTA4, IRTA5; CD proteins such as CD2, CD3, CD4, CD5, CD6, CD8, CD11, CD14, CD19, CD20, CD21, CD22, CD25, CD26, CD28, CD30, CD33, CD36, CD37, CD38, CD40, CD44, CD52, CD55, CD56, CD59, CD70, CD79, CD80. CD81, CD103, CD105, CD134, CD137, CD138, CD152, or an antibody which binds to one or more tumor-associated antigens or cell-surface receptors disclosed in US Publication No. 2008/0171040 or US Publication No. 2008/0305044 and incorporated in their entirety by reference; erythropoietin; osteoinductive factors; immunotoxins; a bone morphogenetic protein (BMP); an interferon, such as interferon-alpha, -beta, and -gamma; colony stimulating factors (CSFs), e.g., M-CSF, GM-CSF, and G-CSF; interleukins (ILs), e.g., IL-1 to IL-10; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor; viral antigen such as, for example, a portion of the HIV envelope; transport proteins; homing receptors; addressins; regulatory proteins; integrins, such as CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4 and VCAM; a tumor associated antigen such as AFP, ALK, B7H4, BAGE proteins, β-catenin, brc-abl, BRCA1, BORIS, CA9 (carbonic anhydrase IX), caspase-8, CD20, CD40, CD123, CDK4, CEA, CLEC12A, c-kit, cMET, CTLA4, cyclin-B1, CYP1B1, EGFR, EGFRvIII, endoglin, Epcam, EphA2, ErbB2/Her2, ErbB3/Her3, ErbB4/Her4, ETV6-AML, Fra-1, FOLR1, GAGE proteins (e.g., GAGE-1, -2), GD2, GD3, GloboH, glypican-3, GM3, gp100, Her2, HLA/B-raf, HLA/EBNA1, HLA/k-ras, HLA/MAGE-A3, hTERT, IGF1R, LGR5, LMP2, MAGE proteins (e.g., MAGE-1, -2, -3, -4, -6, and -12), MART-1, mesothelin, ML-IAP, Muc1, Muc16 (CA-125), MUM1, NA17, NGEP, NY-BR1, NY-BR62, NY-BR85, NY-ESO1, OX40, p15, p53, PAP, PAX3, PAX5, PCTA-1, PDGFR-α, PDGFR-β, PDGF-A, PDGF-B, PDGF-C, PDGF-D, PLAC1, PRLR, PRAME, PSCA, PSGR, PSMA (FOLH1), RAGE proteins, Ras, RGS5, Rho, SART-1, SART-3, Steap-1, Steap-2, STn, survivin, TAG-72, TGF-β, TMPRSS2, Tn, TNFRSF17, TRP-1, TRP-2, tyrosinase, and uroplakin-3, and fragments of any of the above-listed polypeptides.
  • Exemplary antigens also include, but are not limited to, BCMA, SLAMF7, B7H4, GPNMB, UPK3A, and LGR5.
  • In some embodiments, the antigens include prolactin receptor (PRLR) or prostate-specific membrane antigen (PSMA).
  • Binding agents also include, but are not limited to, ankyrin repeat proteins, interferons, lymphokines such as IL-2 or IL-3, hormones like insulin and glucocorticoids, growth factors such as EGF, transferrin and fibronectin type III.
  • In some embodiments, the binding agents interact with or bind to tumor antigens, including antigens specific for a type of tumor or antigens that are shared, overexpressed or modified on a particular type of tumor. Examples include, but are not limited to: alpha-actinin-4 with lung cancer, ARTC1 with melanoma, BCR-ABL fusion protein with chronic myeloid leukemia, B-RAF, CLPP or Cdc27 with melanoma, CASP-8 with squamous cell carcinoma, and hsp70-2 with renal cell carcinoma as well as the following shared tumor-specific antigens, for example: BAGE-1, GAGE, GnTV, KK-LC-1, MAGE-A2, NA88-A, TRP2-INT2.
  • In some embodiments, the binding agent is an antibody. In some embodiments, the binding agent is a monoclonal antibody. In some embodiments, the binding agent is a polyclonal antibody. In some embodiments, the antibody is an anti-PSMA, anti-MUC16, or anti-EGFRvIII, or anti-STEAP-2 antibody.
  • The linkers can be bonded to the binding agent, e.g., antibody or antigen-binding molecule, through an attachment at a particular amino acid within the antibody or antigen-binding molecule. Exemplary amino acid attachments that can be used in the context of this aspect of the disclosure include, e.g., lysine (see, e.g., U.S. Pat. No. 5,208,020; US 2010/0129314; Hollander et al., Bioconjugate Chem., 2008, 19:358-361; WO 2005/089808; U.S. Pat. No. 5,714,586; US 2013/0101546; and US 2012/0585592), cysteine (see, e.g., US 2007/0258987; WO 2013/055993; WO 2013/055990; WO 2013/053873; WO 2013/053872; WO 2011/130598; US 2013/0101546; and U.S. Pat. No. 7,750,116), selenocysteine (see, e.g., WO 2008/122039; and Hofer et al., Proc. Natl. Acad. Sci., USA, 2008, 105:12451-12456), formyl glycine (see, e.g., Carrico et al., Nat. Chem. Biol., 2007, 3:321-322; Agarwal et al., Proc. Natl. Acad. Sci., USA, 2013, 110:46-51, and Rabuka et al., Nat. Protocols, 2012, 10:1052-1067), non-natural amino acids (see, e.g., WO 2013/068874, and WO 2012/166559), and acidic amino acids (see, e.g., WO 2012/05982). Linkers can be conjugated via glutamine via transglutaminase-based chemo-enzymatic conjugation (see, e.g., Dennler et al., Bioconjugate Chem. 2014, 25, 569-578). Linkers can also be conjugated to an antigen-binding protein via attachment to carbohydrates (see, e.g., US 2008/0305497, WO 2014/065661, and Ryan et al., Food & Agriculture Immunol., 2001, 13:127-130) and disulfide linkers (see, e.g., WO 2013/085925, WO 2010/010324, WO 2011/018611, WO 2014/197854, and Shaunak et al., Nat. Chem. Biol., 2006, 2:312-313).
  • In some embodiments, the binding agent is an antibody, and the antibody is bonded to the linker through a lysine residue. In some embodiments, the antibody is bonded to the linker through a cysteine residue.
  • 4. Illustrative Embodiments
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00183
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, halo, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00184
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00185
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00186
  • is one or more bonds to the binding agent;
      • AA1 is an amino acid; and
      • AA2 is an amino acid.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00187
  • wherein:
  • R1, independently at each occurrence, is C1-6 alkyl, C1-6 haloalkyl, or halo;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00188
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00189
  • is one or more bonds to the binding agent;
      • AA1 is an amino acid; and
      • AA2 is an amino acid.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00190
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, haloalkoxy, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00191
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00192
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00193
  • is one or more bonds to the binding agent;
      • AA1 is an amino acid; and AA2 is an amino acid.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00194
  • wherein:
  • R1, independently at each occurrence, is C1-6 alkyl, C1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4;
  • m is an integer from 0 to 3;
  • p is an integer from 0 to 6; and
  • q is an integer from 0 to 5; and
  • L is:
  • Figure US20160375147A1-20161229-C00195
  • wherein:
      • SP is:
  • Figure US20160375147A1-20161229-C00196
        • wherein:
  • Figure US20160375147A1-20161229-C00197
  • is a bond to the binding agent; and
      •  b is an integer from 2 to 8; and
      • AA1 is an amino acid; and
      • AA2 is an amino acid.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00198
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00199
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00200
  • wherein:
      • SP is:
  • Figure US20160375147A1-20161229-C00201
        • wherein:
  • Figure US20160375147A1-20161229-C00202
  • is a bond to the binding agent; and
      •  b is an integer from 2 to 8; and
      • AA1 is an amino acid; and
      • AA2 is an amino acid.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00203
  • wherein:
  • R1, independently at each occurrence, is C1-6 alkyl, C1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4;
  • m is an integer from 0 to 3;
  • p is an integer from 0 to 6; and
  • q is an integer from 0 to 5; and
  • L is:
  • Figure US20160375147A1-20161229-C00204
  • wherein:
  • SP is a spacer;
  • Figure US20160375147A1-20161229-C00205
  • is one or more bonds to the binding agent;
  • RAA1 is an amino acid side chain; and
  • RAA2 is an amino acid side chain.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00206
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00207
  • or azido,
      •  wherein RA, is alkyl or heteroalkyl n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00208
  • wherein:
  • SP is a spacer;
  • Figure US20160375147A1-20161229-C00209
  • is one or more bonds to the binding agent;
  • RAA1 is an amino acid side chain; and
  • RAA2 is an amino acid side chain.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00210
  • wherein:
  • R1, independently at each occurrence, is C1-6 alkyl, C1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4;
  • m is an integer from 0 to 3;
  • p is an integer from 0 to 6; and
  • q is an integer from 0 to 5; and
  • L is:
  • Figure US20160375147A1-20161229-C00211
  • wherein:
      • SP is a spacer; and
  • Figure US20160375147A1-20161229-C00212
  • is the one or more bonds to the binding agent.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00213
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00214
  • or azido,
      •  wherein RA is alkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00215
  • wherein:
      • SP is a spacer; and
  • Figure US20160375147A1-20161229-C00216
  • is the one or more bonds to the binding agent.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00217
  • wherein:
  • R1, independently at each occurrence, is C1-6 alkyl, C1-6 haloalkyl, or halo;
  • n is an integer from 0 to 4;
  • m is an integer from 0 to 3;
  • p is an integer from 0 to 6; and
  • q is an integer from 0 to 5; and
  • L is:
  • Figure US20160375147A1-20161229-C00218
  • wherein:
      • SP is:
  • Figure US20160375147A1-20161229-C00219
        • wherein:
  • Figure US20160375147A1-20161229-C00220
  • is a bond to the binding agent; and
      •  b is an integer from 2 to 8.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00221
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00222
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00223
  • wherein:
      • SP is:
  • Figure US20160375147A1-20161229-C00224
        • wherein:
  • Figure US20160375147A1-20161229-C00225
  • is a bond to the binding agent; and
      •  b is an integer from 2 to 8.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00226
  • wherein:
  • R1, independently at each occurrence, is C1-6 alkyl, C1-6 haloalkyl, or halo; and
  • n, m, p, and q are 0, 1, or 2; and
  • L is
  • Figure US20160375147A1-20161229-C00227
  • wherein:
  • Figure US20160375147A1-20161229-C00228
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00229
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00230
  • or azido,
      •  wherein RA is alkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
    L is
  • Figure US20160375147A1-20161229-C00231
  • wherein:
  • Figure US20160375147A1-20161229-C00232
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00233
  • wherein
      • R1 is, independently at each occurrence, is C1-6 haloalkyl, or halo; and
      • n is 0, 1, or 2; and
    L is:
  • Figure US20160375147A1-20161229-C00234
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00235
  • is the one or more bonds to the binding agent;
      • RAA1 is an amino acid side chain; and
      • RAA2 is an amino acid side chain.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00236
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00237
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • wherein n is an integer from 0 to 4;
    L is:
  • Figure US20160375147A1-20161229-C00238
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00239
  • is the one or more bonds to the binding agent;
      •  RAA1 is an amino acid side chain; and
      •  RAA2 is an amino acid side chain.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00240
  • wherein
      • R1 is, independently at each occurrence, is halo; and
      • n is 0, 1, or 2; and
    L is:
  • Figure US20160375147A1-20161229-C00241
  • wherein:
  • Figure US20160375147A1-20161229-C00242
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00243
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00244
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
    L is:
  • Figure US20160375147A1-20161229-C00245
  • wherein:
  • Figure US20160375147A1-20161229-C00246
  • is a bond to the binding agent;
      •  wherein n is an integer from 0 to 4; and
      •  b is an integer from 2 to 8.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00247
  • wherein
      • R1 is, independently at each occurrence, is halo; and
      • n is 0, 1, or 2; and
    L is:
  • Figure US20160375147A1-20161229-C00248
  • wherein
  • Figure US20160375147A1-20161229-C00249
  • is a bond to the binding agent.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00250
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00251
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • wherein n is an integer from 0 to 4;
    L is:
  • Figure US20160375147A1-20161229-C00252
  • wherein
  • Figure US20160375147A1-20161229-C00253
  • is a bond to the binding agent.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00254
  • and
  • L is
  • Figure US20160375147A1-20161229-C00255
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00256
  • wherein:
      • R1 is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, aryl, heteroalkyl, halo, haloalkoxy, haloalkyl, or haloalkoxy;
  • Figure US20160375147A1-20161229-C00257
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00258
  • is the bond to the carbonyl. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00259
  • wherein:
      • R1 is, independently at each occurrence, a hydrogen atom, alkyl, alkoxy, aryl, heteroalkyl, halo, haloalkyl, haloalkoxy;
  • Figure US20160375147A1-20161229-C00260
  • is the bond to the nitrogen atom; and
  • Figure US20160375147A1-20161229-C00261
  • is the bond to the carbonyl. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00262
  • and
  • L is
  • Figure US20160375147A1-20161229-C00263
  • In some embodiments,
  • BA is an antibody,
  • A is:
  • Figure US20160375147A1-20161229-C00264
  • wherein
  • R1 is, independently at each occurrence, is halo; and
  • n is 0, 1, or 2; and
  • L is:
  • Figure US20160375147A1-20161229-C00265
  • wherein
  • Figure US20160375147A1-20161229-C00266
  • is a bond to the binding agent.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00267
  • wherein
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00268
  • or azido; and
      • q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00269
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00270
  • is the one or more bonds to the binding agent;
      • RAA1 is an amino acid side chain; and
      • RAA2 is an amino acid side chain.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00271
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00272
  • or azido;
      • wherein q is an integer from 0 to 5;
    L is:
  • Figure US20160375147A1-20161229-C00273
  • wherein:
      • SP is a spacer;
  • Figure US20160375147A1-20161229-C00274
  • is the one or more bonds to the binding agent;
      • RAA1 is an amino acid side chain; and
  • RAA2 is an amino acid side chain.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00275
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00276
  • or azido;
      • wherein q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00277
  • wherein:
  • Figure US20160375147A1-20161229-C00278
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00279
  • wherein:
      • R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00280
  • or azido; and
      •  q is an integer from 0 to 5; and
    L is:
  • Figure US20160375147A1-20161229-C00281
  • wherein
  • Figure US20160375147A1-20161229-C00282
  • is a bond to the binding agent.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00283
  • wherein:
    R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00284
  • or azido;
  • wherein q is an integer from 0 to 5;
  • L is:
  • Figure US20160375147A1-20161229-C00285
  • wherein
  • Figure US20160375147A1-20161229-C00286
  • is a bond to the binding agent.
  • In some embodiments,
  • A is:
  • Figure US20160375147A1-20161229-C00287
  • and
  • L is
  • Figure US20160375147A1-20161229-C00288
  • In some embodiments, the compound of Formula I is:
  • Figure US20160375147A1-20161229-C00289
  • wherein A is arylene or heteroarylene, L1 and L2 are linkers, BA is a binding agent, k is an integer from 0 to 30, and t is an integer from 0 to 8. In some of these embodiments, L1 is a linker which binds to the BA through a lysine residue. In some of these embodiments, the subscript, k, represents the number of linkers, L1, bonded to the BA through lysine residues on the BA. In some of these embodiments, L2 is a linker which binds to the BA through a cysteine residue. In some of these embodiments, the subscript, t, represents the number of linkers, L2, bonded to the BA through cysteine residues on the BA. In some embodiments, when the linker, L2, is a monodentate linker, t is an integer from 0 to 8. In some embodiments, when the linker, L2, is a bidentate linker, t is an integer from 0 to 4. In some of these examples, the sum of k+t is equal to 1-8.
  • In some embodiments, the compound of Formula I is:
  • Figure US20160375147A1-20161229-C00290
  • wherein A is arylene or heteroarylene, L1 and L2 are linkers, and BA is a binding agent. In some of these embodiments, L1 is a linker which binds to the BA through a lysine residue. In some of these embodiments, L2 is a linker which binds to the BA through a cysteine residue.
  • In some embodiments, the compound of Formula I is:
  • Figure US20160375147A1-20161229-C00291
  • wherein A is arylene or heteroarylene, L1 and L2 are linkers, and BA is a binding agent. In some of these embodiments, L1 is a linker which binds to the BA through a lysine residue. In some of these embodiments, L2 is a linker which binds to the BA through a cysteine residue.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00292
  • wherein:
      • R1 is, independently at each occurrence, halo, haloalkyl, haloalkoxy, hydroxyl, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, aryl, alkaryl, aralkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00293
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, the linker is:
  • Figure US20160375147A1-20161229-C00294
  • wherein:
  • SP is a spacer;
  • Figure US20160375147A1-20161229-C00295
  • is one or more bonds to the binding agent;
  • AA1 is an amino acid; and
  • AA2 is an amino acid.
  • The spacer is a divalent moiety that connects the AA1-AA2 moiety to the binding agent (BA). Suitable spacers include, but are not limited to, those comprising alkylene or polyethylene glycol. The ends of the spacers, i.e., the portion of the spacer directly bonded to the binding agent or AA1, can be moieties derived from reactive moieties that are used for purposes of coupling the antibody or AA1 to the spacer during the chemical synthesis of the conjugate.
  • In some embodiments, the spacer comprises an alkylene. In some embodiments, the spacer comprises a C5-7 alkylene. In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00296
  • wherein:
  • Figure US20160375147A1-20161229-C00297
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00298
  • wherein:
  • Figure US20160375147A1-20161229-C00299
  • is a bond to the binding agent.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00300
  • wherein:
  • RN is a hydrogen atom or alkyl;
  • RM is alkyl;
  • the two bonds represented by
  • Figure US20160375147A1-20161229-C00301
  • are bonds to cysteines of a binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00302
  • wherein:
  • Figure US20160375147A1-20161229-C00303
  • is a bond to the binding agent; and
  • b is an integer from 2 to 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00304
  • wherein:
  • Figure US20160375147A1-20161229-C00305
  • is a bond to the binding agent; and
  • g is an integer from 2 to 20. In some embodiments, g is 2-8. In some embodiments, g is 2, 4, 6, or 8.
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00306
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00307
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00308
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00309
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00310
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00311
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00312
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00313
  • In some embodiments, the spacer is:
  • Figure US20160375147A1-20161229-C00314
  • wherein
  • Figure US20160375147A1-20161229-C00315
  • is a bond to the binding agent;
    X is N or O; RN and RM are each, independently, hydrogen or alkyl; and b is an integer from 1 to 8.
  • In some embodiments, AA1-AA2 is: valine-citrulline, citrulline-valine, lysine-phenylalanine, phenylalanine-lysine, valine-asparagine, asparagine-valine, threonine-asparagine, asparagine-threonine, serine-asparagine, asparagine-serine, phenylalanine-asparagine, asparagine-phenylalanine, leucine-asparagine, asparagine-leucine, isoleucine-asparagine, asparagine-isoleucine, glycine-asparagine, asparagine-glycine, glutamic acid-asparagine, asparagine-glutamic acid, citrulline-asparagine, asparagine-citrulline, alanine-asparagine, or asparagine-alanine.
  • In some embodiments, AA1-AA2 is: valine-citrulline or citrulline-valine. In some embodiments, AA1-AA2 is: valine-citrulline.
  • In some embodiments, the compound of Formula I is:
  • Figure US20160375147A1-20161229-C00316
  • wherein X is N or O,
  • RN and RM are each, independently, hydrogen or aryl,
  • b is an integer from 1 to 8,
  • A is aryl or heteroaryl, and
  • t is an integer from 1-8.
  • In some embodiments, the compound of Formula I is:
  • Figure US20160375147A1-20161229-C00317
    Figure US20160375147A1-20161229-C00318
    Figure US20160375147A1-20161229-C00319
    Figure US20160375147A1-20161229-C00320
    Figure US20160375147A1-20161229-C00321
    Figure US20160375147A1-20161229-C00322
    Figure US20160375147A1-20161229-C00323
    Figure US20160375147A1-20161229-C00324
    Figure US20160375147A1-20161229-C00325
    Figure US20160375147A1-20161229-C00326
    Figure US20160375147A1-20161229-C00327
    Figure US20160375147A1-20161229-C00328
    Figure US20160375147A1-20161229-C00329
    Figure US20160375147A1-20161229-C00330
    Figure US20160375147A1-20161229-C00331
    Figure US20160375147A1-20161229-C00332
    Figure US20160375147A1-20161229-C00333
    Figure US20160375147A1-20161229-C00334
    Figure US20160375147A1-20161229-C00335
    Figure US20160375147A1-20161229-C00336
    Figure US20160375147A1-20161229-C00337
    Figure US20160375147A1-20161229-C00338
    Figure US20160375147A1-20161229-C00339
    Figure US20160375147A1-20161229-C00340
    Figure US20160375147A1-20161229-C00341
    Figure US20160375147A1-20161229-C00342
    Figure US20160375147A1-20161229-C00343
    Figure US20160375147A1-20161229-C00344
    Figure US20160375147A1-20161229-C00345
    Figure US20160375147A1-20161229-C00346
    Figure US20160375147A1-20161229-C00347
    Figure US20160375147A1-20161229-C00348
    Figure US20160375147A1-20161229-C00349
    Figure US20160375147A1-20161229-C00350
    Figure US20160375147A1-20161229-C00351
    Figure US20160375147A1-20161229-C00352
    Figure US20160375147A1-20161229-C00353
  • wherein:
      • Ab is an antibody;
      • S is a bond to a cysteine of the antibody;
      • N is a bond to a lysine of the antibody;
      • k is an integer from 1 to 30; and
      • t is an integer from 1 to 8. In some examples, k is an integer from 1 to 8. In some examples, t is an integer from 1 to 4. In some examples, when S is a bond to a cysteine of the antibody, up to 8 conjugates set forth herein may be bonded to the antibody. In some examples, when N is a bond to a lysine of the antibody, up to 30 conjugates set forth herein may be bonded to the antibody.
  • In some embodiments, the compound of Formula I is:
  • Figure US20160375147A1-20161229-C00354
    Figure US20160375147A1-20161229-C00355
  • In some embodiment, k is an integer from 1 to 30. In some embodiment, k is an integer from 1 to 8. In some embodiment, k is an integer from 1 to 6. In some embodiments, k is an integer from 1 to 4. In some embodiments, k is an integer from 1 to 3. In some embodiments, the drug-antibody ratio (DAR) of the conjugate is from 1.0 to 3.0.
  • C. Maytansinoid Derivatives
  • Provided herein are compounds of Formula (II):
  • Figure US20160375147A1-20161229-C00356
  • or a pharmaceutically acceptable salt thereof,
    wherein A is arylene or heteroarylene.
  • In certain embodiments, these compounds represent the payload portion of the conjugates described herein and are released, e.g., by enzyme proteolysis, following internalization of the conjugate into a cell. The methods provided herein include methods of treating a proliferative disease, e.g., cancer, comprising administering to a patient a therapeutically effective amount of a conjugate, e.g., antibody-drug conjugate that releases a compound of Formula (II) following internalization of said conjugate into a cell in said patient.
  • In some embodiments, these compounds represent the metabolic product of the conjugates described herein, e.g., enzyme proteolysis product. In some embodiments, these compounds represent the catabolic product of the conjugates described herein. In some embodiments, these compounds represent the cellular product of the conjugates described herein.
  • In some embodiments, A is a divalent radical of benzene, of pyridine, of naphthalene, or of quinolone, which are optionally substituted.
  • In some embodiments, A is arylene.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00357
  • wherein
      • R1 is, independently at each occurrence, alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, halo, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00358
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIA):
  • Figure US20160375147A1-20161229-C00359
  • wherein R1 and n are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIB):
  • Figure US20160375147A1-20161229-C00360
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIB2):
  • Figure US20160375147A1-20161229-C00361
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIB3):
  • Figure US20160375147A1-20161229-C00362
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIC):
  • Figure US20160375147A1-20161229-C00363
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IID):
  • Figure US20160375147A1-20161229-C00364
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIE):
  • Figure US20160375147A1-20161229-C00365
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIF):
  • Figure US20160375147A1-20161229-C00366
  • wherein R1 and q are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIG):
  • Figure US20160375147A1-20161229-C00367
  • wherein R1 and q are as defined herein.
  • In some embodiments, R1 is, independently, alkyl or halo. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 haloalkyl, or halo. In some embodiments, R1 is, independently, C1-6 haloalkyl or halo. In some embodiments, R1 is, independently, halo. In some embodiments, R1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, R1 is, independently, alkyl, alkoxy, heteroalkyl, halo, haloalkyl, or haloalkoxy. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 alkoxy, C1-6 haloalkyl, C1-6 haloalkoxy, or halo. In some embodiments, R1 is, independently, C1-6 alkyl or C1-6 alkoxy. In some embodiments, R1 is, independently, alkoxy. In some embodiments, R1 is, independently, methoxy, ethoxy, propoxy. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0. In some embodiments, the compound of Formula (II) is a compound of Formula (IIA):
  • Figure US20160375147A1-20161229-C00368
  • wherein:
  • R1 is, independently at each occurrence, halo or trifluoromethyl; and
  • n is 0, 1, or 2.
  • In some embodiments, the compound of Formula (II) is a compound of Formula (IIB):
  • Figure US20160375147A1-20161229-C00369
  • wherein:
  • R1 is, independently at each occurrence, halo or trifluoromethyl; and
  • q is 0, 1, or 2.
  • In some embodiments, the compound of Formula (II) is:
  • Figure US20160375147A1-20161229-C00370
    Figure US20160375147A1-20161229-C00371
  • In some embodiments, the compound of Formula (II) is a compound selected from
  • Figure US20160375147A1-20161229-C00372
    Figure US20160375147A1-20161229-C00373
  • In some embodiments, the compound of Formula (II) is:
  • Figure US20160375147A1-20161229-C00374
  • In certain embodiments, these compounds represent the payload portion of the conjugates described herein and are released, e.g., by enzyme proteolysis, following internalization of the conjugate into a cell. The methods provided herein include methods of treating a proliferative disease, e.g., cancer, comprising administering to a patient a therapeutically effective amount of a conjugate, e.g., antibody-drug conjugate that releases a compound of Formula (II) following internalization of said conjugate into a cell in said patient.
  • In some embodiments, these compounds represent the metabolic product of the conjugates described herein, e.g., enzyme proteolysis product.
  • In some embodiments, A is a divalent radical of benzene, of pyridine, of naphthalene, or of quinolone, which are optionally substituted.
  • In some embodiments, A is arylene.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00375
  • wherein:
      • R1 is, independently at each occurrence, alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, halo, haloalkoxy, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00376
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIA):
  • Figure US20160375147A1-20161229-C00377
  • wherein R1 is, independently at each occurrence, methoxy, halo or trifluoromethyl; and
    n is 0, 1, or 2.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIB):
  • Figure US20160375147A1-20161229-C00378
  • wherein R1 is, independently at each occurrence, methoxy, halo or trifluoromethyl; and q is 0, 1, or 2.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIB2):
  • Figure US20160375147A1-20161229-C00379
  • wherein R1 is, independently at each occurrence, methoxy, halo or trifluoromethyl; and q is 0, 1, or 2.
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIB3):
  • Figure US20160375147A1-20161229-C00380
  • wherein R1 is, independently at each occurrence, methoxy, halo or trifluoromethyl; and q is 0, 1, or 2. In some embodiments, R1 is, independently, alkyl or halo. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 haloalkyl, or halo. In some embodiments, R1 is, independently, C1-6 haloalkyl or halo. In some embodiments, R1 is, independently, halo. In some embodiments, R1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, R1 is, independently, alkyl, alkoxy, heteroalkyl, halo, haloalkyl, or haloalkoxy. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 alkoxy, C1-6 haloalkyl, C1-6 haloalkoxy, or halo. In some embodiments, R1 is, independently, C1-6 alkyl or C1-6 alkoxy. In some embodiments, R1 is, independently, alkoxy. In some embodiments, R1 is, independently, methoxy, ethoxy, propoxy. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, the compound of Formula (II) is:
  • Figure US20160375147A1-20161229-C00381
    Figure US20160375147A1-20161229-C00382
  • In some embodiments, the compound of Formula (II) is a compound of the Formula (IIH):
  • Figure US20160375147A1-20161229-C00383
  • wherein R1 and n are as defined herein.
  • In some embodiments, the compound of Formula (II) is a compound selected from
  • Figure US20160375147A1-20161229-C00384
    Figure US20160375147A1-20161229-C00385
  • In some embodiments, the compound of Formula (II) is a compound selected from
  • Figure US20160375147A1-20161229-C00386
    Figure US20160375147A1-20161229-C00387
  • D. Preparation of Compounds
  • Compounds of Formula I can be synthesized by coupling compounds of Formula P1 with a binding agent, e.g., antibody under standard conjugation conditions (see, e.g, Doronina et al., Nature Biotechnology 2003, 21, 7, 778, which is incorporated herein by reference). When the binding agent is an antibody, the antibody can be coupled to a compound of Formula P1 via one or more cysteine or lysine residues of the antibody. Compounds of Formula P1 can be coupled to cysteine residues, for example, by subjecting the antibody to a reducing agent, e.g., dithiotheritol, to cleave the disulfide bonds of the antibody, purifying the reduced antibody, e.g., by gel filtration, and subsequently reacting the antibody with a compound of formula P1 containing a reactive moiety, e.g., a maleimido group. Suitable solvents include, but are not limited to water, DMA, DMF, and DMSO. Compounds of formula P1 containing a reactive moiety, e.g., activated ester or acid halide group, can be coupled to lysine residues. Suitable solvents include, but are not limited to water, DMA, DMF, and DMSO. The compounds of Formula I can be purified using known protein techniques, including, for example, size exclusion chromatography, dialysis, and ultrafiltration/diafiltration.
  • Figure US20160375147A1-20161229-C00388
  • wherein RL is a reactive linker, A is arylene or heteroarylene, L is a linker, and BA is a binding agent.
  • In some embodiments, the compound of formula P1 includes A, wherein A is:
  • Figure US20160375147A1-20161229-C00389
  • wherein n is 0 or 1; and R1 is alkoxy, halo, or haloalkyl.
  • In some embodiments, the compound of formula P1 includes A, wherein A is:
  • Figure US20160375147A1-20161229-C00390
  • wherein n is 0 or 1; and R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl.
  • In some embodiments, the compound of formula P1 includes A, wherein A is:
  • Figure US20160375147A1-20161229-C00391
  • wherein n is 0 or 1; R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl; and RL is
  • Figure US20160375147A1-20161229-C00392
  • wherein b is an integer from 2 to 8 and
  • Figure US20160375147A1-20161229-C00393
  • is a bond to the binding agent.
  • The reactive linker is a moiety comprising a portion in its structure that is capable of reacting with the binding agent (e.g., reacting with an antibody at its cysteine or lysine residues) to form the compound of Formula I. Following conjugation to the binding agent, the reactive linker becomes the linker (L) moiety of the compound of Formula I. Illustrative reactive linkers include, but are not limited to, those that comprise haloacetyl, isothiocyanate, or maleimide portions that are capable of reacting with the binding agent. Reactive portions also include moieties having the following structure:
  • Figure US20160375147A1-20161229-C00394
  • wherein X is —O— or —NH— and LG is a leaving group, e.g., Br.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00395
  • wherein:
  • SPR is a reactive spacer;
  • AA1 is an amino acid; and
  • AA2 is an amino acid.
  • The reactive spacer is a moiety that contains the above-described reactive linker portion that is capable of reacting with the binding agent and connects this portion to AA1. Suitable spacers include, but are not limited to, those comprising alkylene or polyethylene glycol connecting the AA1 to the portion capable of reacting with binding agent (e.g., haloacetyl, isothiocyanate, or maleimide).
  • In some embodiments, the reactive spacer comprises a non-cleavable moiety selected from
  • Figure US20160375147A1-20161229-C00396
  • wherein
  • Figure US20160375147A1-20161229-C00397
  • represents one or more bonds to the maytansinoid derivative; and wherein n is an integer from 4 to 10. In some examples, n is 4, 5, 6, 7, 8, 9, or 10. In some embodiments, the reactive spacer is
  • Figure US20160375147A1-20161229-C00398
  • In some embodiments, the reactive spacer is
  • Figure US20160375147A1-20161229-C00399
  • In some embodiments, the reactive spacer is:
  • Figure US20160375147A1-20161229-C00400
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive spacer is:
  • Figure US20160375147A1-20161229-C00401
  • In some embodiments, the spacer is
  • Figure US20160375147A1-20161229-C00402
  • In some embodiments, the spacer is
  • Figure US20160375147A1-20161229-C00403
  • wherein g is an integer from 1 to 24.
  • In some embodiments, the reactive spacer is:
  • Figure US20160375147A1-20161229-C00404
  • wherein b is an integer from 2 to 8 and g is an integer from 2 to 20.
  • In some embodiments, the reactive spacer is:
  • Figure US20160375147A1-20161229-C00405
  • In some embodiments, AA1-AA2 is: valine-citrulline, citrulline-valine, lysine-phenylalanine, phenylalanine-lysine, valine-asparagine, asparagine-valine, threonine-asparagine, asparagine-threonine, serine-asparagine, asparagine-serine, phenylalanine-asparagine, asparagine-phenylalanine, leucine-asparagine, asparagine-leucine, isoleucine-asparagine, asparagine-isoleucine, glycine-asparagine, asparagine-glycine, glutamic acid-asparagine, asparagine-glutamic acid, citrulline-asparagine, asparagine-citrulline, alanine-asparagine, or asparagine-alanine.
  • In some embodiments, AA1-AA2 is: valine-citrulline or citrulline-valine. In some embodiments, AA1-AA2 is: valine-citrulline.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00406
  • wherein:
  • SPR is a reactive spacer;
  • RAA1 is an amino acid side chain; and
  • RAA2 is an amino acid side chain.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00407
  • wherein:
  • SP is a reactive spacer.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00408
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00409
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00410
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00411
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00412
  • wherein b is an integer from 2 to 8, RN is a hydrogen atom or alkyl, and RM is alkyl.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00413
  • wherein b is an integer form 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00414
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00415
  • wherein b is an integer from 2 to 8; RN is a hydrogen atom or alkyl; and RM is alkyl.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00416
  • wherein b is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00417
  • wherein g is an integer from 2 to 8.
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00418
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00419
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00420
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00421
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00422
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00423
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00424
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00425
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00426
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00427
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00428
  • In some embodiments, the reactive linker is:
  • Figure US20160375147A1-20161229-C00429
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1A:
  • Figure US20160375147A1-20161229-C00430
  • wherein:
  • A is:
  • Figure US20160375147A1-20161229-C00431
      • wherein:
        • R1 is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00432
  • or azido, wherein RA is alkyl or heteroalkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5;
  • SPR is a reactive spacer;
  • AA1 is an amino acid; and
  • AA2 is an amino acid.
  • In some embodiments, the compound of Formula P1A is a compound which includes A wherein A is:
  • Figure US20160375147A1-20161229-C00433
  • wherein n is 0 or 1; and R1 is alkoxy, halo, or haloalkyl. In some examples, R1 is methylsulfonyl, N-methylformamide, hydroxyl, or morpholinyl.
  • In some embodiments, the compound of Formula P1A is a compound which includes A wherein A is:
  • Figure US20160375147A1-20161229-C00434
  • wherein n is 0 or 1; and R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl.
  • In some embodiments, the compound of Formula P1A is a compound which includes A wherein A is:
  • Figure US20160375147A1-20161229-C00435
  • wherein q is an integer from 0 to 5; and R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl.
  • In some embodiments, the compound of Formula P1A is a compound which includes A wherein A is:
  • Figure US20160375147A1-20161229-C00436
  • wherein q is an integer from 0 to 5; and R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl.
  • In some embodiments, the compound of Formula P1A is a compound which includes A wherein A is:
  • Figure US20160375147A1-20161229-C00437
  • wherein n is 0 or 1; and R1 is alkoxy, halo, or haloalkyl. In some examples, R1 is methylsulfonyl, N-methylformamide, hydroxyl, or morpholinyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1B:
  • Figure US20160375147A1-20161229-C00438
  • wherein
  • A is:
  • Figure US20160375147A1-20161229-C00439
      • wherein:
        • R1 is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkoxy, haloalkyl, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00440
  • or azido, wherein RA is alkyl or heteroalkyl;
      •  n is an integer from 0 to 4;
      •  m is an integer from 0 to 3;
      •  p is an integer from 0 to 6; and
      •  q is an integer from 0 to 5; and
  • SPR is a reactive spacer.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1C:
  • Figure US20160375147A1-20161229-C00441
  • wherein:
  • SPR is a reactive spacer;
  • AA1 is an amino acid;
  • AA2 is an amino acid;
  • R1 is, independently at each occurrence, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and
  • n is 0, 1, or 2.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1D:
  • Figure US20160375147A1-20161229-C00442
  • wherein:
  • SPR is a reactive spacer;
  • AA1 is an amino acid;
  • AA2 is an amino acid;
  • R1 is, independently at each occurrence, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and
  • n is 0, 1, or 2. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1D, wherein R1 is alkoxy, halo, or haloalkyl. In some embodiments, R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1D, wherein R1 is C1-6 alkoxy, halo, or C1-6 haloalkyl; and SPR-AA1-AA2 is
  • Figure US20160375147A1-20161229-C00443
  • wherein b is an integer from 2 to 8 and
  • Figure US20160375147A1-20161229-C00444
  • is a bond to the binding agent. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments b is 2. In some embodiments b is 3. In some embodiments b is 4. In some embodiments b is 5. In some embodiments b is 6. In some embodiments b is 7. In some embodiments b is 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1E:
  • Figure US20160375147A1-20161229-C00445
  • wherein:
  • SPR is a reactive spacer;
  • RAA1 is an amino acid side chain;
  • RAA2 is an amino acid side chain;
  • R1 is, independently at each occurrence, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and
  • n is 0, 1, or 2.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1F:
  • Figure US20160375147A1-20161229-C00446
  • wherein:
  • SPR is a reactive spacer;
  • R1 is, independently at each occurrence, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and
  • n is 0, 1, or 2.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1G:
  • Figure US20160375147A1-20161229-C00447
  • wherein:
  • SPR is a reactive spacer; and
  • R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1H:
  • Figure US20160375147A1-20161229-C00448
  • wherein:
  • R1 is hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl; and
  • b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1I:
  • Figure US20160375147A1-20161229-C00449
  • wherein:
  • R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl; and
  • b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1J:
  • Figure US20160375147A1-20161229-C00450
  • wherein:
  • R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl; and
  • b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1K:
  • Figure US20160375147A1-20161229-C00451
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1L:
  • Figure US20160375147A1-20161229-C00452
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1M:
  • Figure US20160375147A1-20161229-C00453
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1N:
  • Figure US20160375147A1-20161229-C00454
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1O:
  • Figure US20160375147A1-20161229-C00455
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1P:
  • Figure US20160375147A1-20161229-C00456
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1Q:
  • Figure US20160375147A1-20161229-C00457
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1R:
  • Figure US20160375147A1-20161229-C00458
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1S:
  • Figure US20160375147A1-20161229-C00459
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1T:
  • Figure US20160375147A1-20161229-C00460
  • wherein RN is a hydrogen atom or alkyl, RM is alkyl, R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1U:
  • Figure US20160375147A1-20161229-C00461
  • wherein RN is a hydrogen atom or alkyl, RM is alkyl, R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1V:
  • Figure US20160375147A1-20161229-C00462
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1W:
  • Figure US20160375147A1-20161229-C00463
  • wherein R1 is a hydrogen atom, alkyl, alkoxy, halo, haloalkoxy, haloalkyl, or trifluoromethyl, g is an integer from 2 to 20; and b is an integer from 2 to 8.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1X:
  • Figure US20160375147A1-20161229-C00464
  • wherein:
    R1 is alkyl, alkenyl, alkynyl, alkoxy, heteroalkyl, halo, haloalkyl, or haloalkoxy; and b is an integer from 2 to 8. In some embodiments, R1 is methyl, ethyl, methoxy, or ethoxy. In some of these embodiments, R1 is methoxy. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1Y:
  • Figure US20160375147A1-20161229-C00465
  • wherein R1 is alkyl, alkenyl, alkynyl, alkoxy, heteroalkyl, halo, haloalkyl, haloalkoxy. In some embodiments, R1 is methyl, ethyl, methoxy, or ethoxy. In some of these embodiments, R1 is methoxy. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, the compound of Formula P1 is a compound of Formula P1Z:
  • Figure US20160375147A1-20161229-C00466
  • wherein Rc is selected from alkyl or haloalkyl and wherein the alkyl or haloalkyl is linear, branched, or cyclic.
  • In some embodiments, the compound of Formula P1 is a compound having one of the following structures:
  • Figure US20160375147A1-20161229-C00467
    Figure US20160375147A1-20161229-C00468
    Figure US20160375147A1-20161229-C00469
    Figure US20160375147A1-20161229-C00470
    Figure US20160375147A1-20161229-C00471
    Figure US20160375147A1-20161229-C00472
    Figure US20160375147A1-20161229-C00473
    Figure US20160375147A1-20161229-C00474
    Figure US20160375147A1-20161229-C00475
    Figure US20160375147A1-20161229-C00476
    Figure US20160375147A1-20161229-C00477
    Figure US20160375147A1-20161229-C00478
    Figure US20160375147A1-20161229-C00479
    Figure US20160375147A1-20161229-C00480
    Figure US20160375147A1-20161229-C00481
    Figure US20160375147A1-20161229-C00482
    Figure US20160375147A1-20161229-C00483
    Figure US20160375147A1-20161229-C00484
    Figure US20160375147A1-20161229-C00485
    Figure US20160375147A1-20161229-C00486
    Figure US20160375147A1-20161229-C00487
    Figure US20160375147A1-20161229-C00488
    Figure US20160375147A1-20161229-C00489
    Figure US20160375147A1-20161229-C00490
    Figure US20160375147A1-20161229-C00491
    Figure US20160375147A1-20161229-C00492
    Figure US20160375147A1-20161229-C00493
    Figure US20160375147A1-20161229-C00494
    Figure US20160375147A1-20161229-C00495
    Figure US20160375147A1-20161229-C00496
    Figure US20160375147A1-20161229-C00497
    Figure US20160375147A1-20161229-C00498
    Figure US20160375147A1-20161229-C00499
    Figure US20160375147A1-20161229-C00500
    Figure US20160375147A1-20161229-C00501
    Figure US20160375147A1-20161229-C00502
    Figure US20160375147A1-20161229-C00503
    Figure US20160375147A1-20161229-C00504
    Figure US20160375147A1-20161229-C00505
    Figure US20160375147A1-20161229-C00506
    Figure US20160375147A1-20161229-C00507
    Figure US20160375147A1-20161229-C00508
    Figure US20160375147A1-20161229-C00509
    Figure US20160375147A1-20161229-C00510
    Figure US20160375147A1-20161229-C00511
    Figure US20160375147A1-20161229-C00512
    Figure US20160375147A1-20161229-C00513
    Figure US20160375147A1-20161229-C00514
    Figure US20160375147A1-20161229-C00515
    Figure US20160375147A1-20161229-C00516
    Figure US20160375147A1-20161229-C00517
    Figure US20160375147A1-20161229-C00518
    Figure US20160375147A1-20161229-C00519
    Figure US20160375147A1-20161229-C00520
    Figure US20160375147A1-20161229-C00521
  • In some embodiments, the compound of Formula P1 is a compound having one of the following structures:
  • Figure US20160375147A1-20161229-C00522
    Figure US20160375147A1-20161229-C00523
  • Compounds of Formula P1 can by synthesized by reacting compounds of Formula P2 with the compound of Formula P3 under amide synthesis conditions. Suitable amide synthesis conditions include, but are not limited to, contacting the compound of Formula P2 in the presence of a carboxylic acid activating agent and base. Suitable activating agents include, but are not limited to EDC, HATU, HBTU, DCC, BOP, and EEDQ. Suitable bases include, but are not limited to DIEA, DBU, Tributylamine, and 2,6-Lutidine.
  • The compound of Formula P2 can be synthesized directly from maytansinol and alanine using known techniques (see, e.g., U.S. Pat. No. 4,308,269, which is incorporated herein by reference).
  • Figure US20160375147A1-20161229-C00524
  • Compounds of Formula I can be synthesized by coupling compounds of Formula PP3:
  • Figure US20160375147A1-20161229-C00525
  • with compounds of Formula PP4 under amide synthesis conditions:
  • Figure US20160375147A1-20161229-C00526
  • wherein:
  • BA is a binding agent;
  • SP is a spacer;
  • RAA1 is an amino acid side chain;
  • RAA2 is an amino acid side chain;
  • A is arylene or heteroarylene; and
  • k is an integer from 1 to 10.
  • Compounds of Formula PP3 can be synthesized by contacting compounds of Formula PP5 with a suitable reducing agent:
  • Figure US20160375147A1-20161229-C00527
  • wherein A is arylene or heteroarylene.
  • In some embodiments, the suitable reducing agent includes a metal, a metal foil, a metal powder, a metal amalgam, or metal filings. In certain embodiments, the metal is selected from zinc, iron, aluminum, palladium, or Raney nickel.
  • For example, in some embodiments, the following reducing agent conditions are employed. With respect to the amount of compound PP5, for example, in some of the methods herein about twenty (20) equivalents of zinc dust and forty (40) equivalents of acetic acid were combined. In some examples, the reducing reaction was conducted at room temperature for about from 1 to 20 hours. In some of these examples, the aforementioned acetic acid is substituted with another suitable mild acid or proton donor. Examples of suitable mild acids or proton donors include, but are not limited to formic acid, pTsOH, and NH4Cl. In some of these examples, the reducing metal is substituted with a suitable reducing agent selected from iron, aluminum, palladium, or Raney nickel. In some of these examples, suitable solvents includes those solvents having 10-50% water (by volume) in a miscible organic solvent. Example miscible organic solvents include, but are not limited to THF, Dioxane, and diethyl ether. In some examples, the reducing reactions set forth herein are conducted at reaction temperatures which range from 0 to 50° C. In some examples, the reducing reactions set forth herein are conducted at reaction times which range from 1 to 40 hours.
  • Suitable acids include, but are not limited to, acetic acid.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00528
  • wherein:
      • R1 is, independently at each occurrence, alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, halo, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00529
      • or azido,
        • wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, the compound of Formula PP5 is a compound of the Formula PP5A:
  • Figure US20160375147A1-20161229-C00530
  • wherein R1 and n are as defined herein.
  • In some embodiments, R1 is, independently, alkyl, alkoxy, heteroalkyl, halo, haloalkyl, or haloalkoxy. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 alkoxy, C1-6 haloalkyl, C1-6 haloalkoxy, or halo. In some embodiments, R1 is, independently, C1-6 alkyl or C1-6 alkoxy. In some embodiments, R1 is, independently, alkoxy. In some embodiments, R1 is, independently, methoxy, ethoxy, propoxy. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, the compound of Formula PP5 is a compound of the Formula PP5A:
  • Figure US20160375147A1-20161229-C00531
  • wherein:
  • R1 is, independently at each occurrence, halo or trifluoromethyl; and
  • n is 0, 1, or 2.
  • In some embodiments, the compound of Formula PP5 is a compound of the Formula PP5A2:
  • Figure US20160375147A1-20161229-C00532
  • wherein:
  • R1 is, independently at each occurrence, halo or trifluoromethyl; and
  • q is an integer from 0 to 5
  • In some embodiments, the compound of Formula PP5 is a compound of the Formula PP5A3:
  • Figure US20160375147A1-20161229-C00533
  • wherein:
  • R1 is, independently at each occurrence, halo or trifluoromethyl; and
  • q is an integer from 0 to 5. In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, the compound of Formula PP5 is a compound of the Formula PP5A4:
  • Figure US20160375147A1-20161229-C00534
  • wherein R1 and n are as defined herein.
  • Compounds of Formula PP5 can be synthesized by contacting compounds of Formula P2 with compounds of Formula PP6 under amide synthesis conditions:
  • Figure US20160375147A1-20161229-C00535
  • Suitable compounds of Formula PP6 include, but are not limited to, 3-nitro-benzoic acid, 3-chloro-5-nitro-benzoic acid, 3-fluoro-5-nitro-benzoic acid, 3-nitro-1-naphthalenecarboxylic acid, 2-fluoro-5-nitro-benzoic acid, 3-(dimethylamino)-5-nitro-benzoic acid, 3-ethoxy-5-nitro-benzoic acid, 2-methoxy-5-nitro-benzoic acid, 4-methoxy-3-nitro-benzoic acid, 2,6-difluoro-3-nitro-benzoic acid, 2-chloro-6-fluoro-3-nitro-benzoic acid, 6-chloro-2-fluoro-3-nitro-benzoic acid, 2-chloro-4-fluoro-5-nitro-benzoic acid, 4-chloro-2-fluoro-5-nitro-benzoic acid, 2-ethoxy-5-nitro-benzoic acid, 2-(methylamino)-3-nitro-benzoic acid, 6-nitro-8-quinolinecarboxlic acid, 4-(dimethylamino)-3-nitro-benzoic acid hydrochloride (1:1), 2-methyl-nitro-benzoic acid, 3-methyl-4-nitro-benzoic acid, 4-nitro-1-naphthalenecarboxylic acid, 4-nitro-1-naphthalenecarboxylic acid, 2,6-dimethyl-4-nitro-benzoic acid, 3-fluoro-4-nitro-benzoic acid, 3-chloro-4-nitro-benzoic acid, 3-bromo-4-nitro-benzoic acid, 3-cyano-4-nitro-benzoic acid, 3-cyclopropyl-4-nitro-benzoic acid, 3-methoxy-4-nitro-benzoic acid, 2-methoxy-4-nitro-benzoic acid, 5-chloro-2-methyl-4-nitro-benzoic acid, 8-nitro-5-isoquinolinecarboxylic acid, 5-nitro-8-quinolinecarboxylic acid, 8-nitro-5-quinolinecarboxylic acid, 2,5-difluoro-4-nitro-benzoic acid, 2-(dimethylamino)-4-nitro-benzoic acid, 2-chloro-5-fluoro-4-nitro-benzoic acid, 3-(dimethylamino)-4-nitro-benzoic acid, 2-[(1-methylethyl)thio]-4-nitro-benzoic acid, 4-nitro-3-(trifluoromethyl)-benzoic acid, 4-nitro-2-(trifluoromethyl)-benzoic acid, 3,5-dimethoxy-4-nitro-benzoic acid, 4-nitro-2-(propylamino)-benzoic acid, 3-(difluoromethoxy)-4-nitro-benzoic acid, 2-(2-fluoro-phenyl)-4-nitro-benzoic acid, 4-nitro-2-(4-pyridinyl)-benzoic acid, 4-nitro-3-(4-pyridinyl)-benzoic acid, or 4-nitro-2-(1-pyrrolidinyl)-benzoic acid.
  • Suitable compounds of Formula PP6 include compounds having any one of the following formula:
  • Figure US20160375147A1-20161229-C00536
  • wherein R1 is, independently at each occurrence, C1-6 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, or C1-6 haloalkoxy, wherein n is 0, 1, 2, 3, or 4. In certain of these embodiments, R1 is methoxy or methyl. In some specific embodiments, R1 is methoxy, fluoro, or trifluoromethyl. In certain embodiments, n is 1 or 2. In some of these embodiments, n is 1. In some embodiments, R1 is fluoro, chloro, bromo, or iodo.
  • In some embodiments, R1 is 1-methylethyl-thiol, phenyl, 2-fluorophenyl, pyridinyl, 4-pyridinyl, pyrrolidinyl, or 1-pyrrolidinyl. In some embodiments, R1 is trifluoromethyl. In some embodiments, R1 is methoxy. In some embodiments, R1 is fluoro. In some embodiments, R1 is hydrogen.
  • In some embodiments, provided herein are compounds of Formula PP5:
  • Figure US20160375147A1-20161229-C00537
  • wherein A is arylene or heteroarylene.
  • In some embodiments, the compound of Formula PP5 is a compound selected from
  • Figure US20160375147A1-20161229-C00538
    Figure US20160375147A1-20161229-C00539
  • In some embodiments, the compound of Formula PP5 is:
  • Figure US20160375147A1-20161229-C00540
  • Compounds of Formula III:
  • Figure US20160375147A1-20161229-C00541
  • can be synthesized by contacting compounds of Formula PP1:
  • Figure US20160375147A1-20161229-C00542
  • with a binding agent under conjugation conditions,
    wherein:
  • BA is a binding agent;
  • SP is a spacer;
  • SPR is a reactive spacer;
  • RAA1 is an amino acid side chain;
  • RAA2 is an amino acid side chain;
  • A is arylene or heteroarylene; and
  • k is an integer from 1 to 30.
  • Compounds of Formula PP1 can be prepared by contacting a compound of Formula PP2 with the compound of Formula P2:
  • Figure US20160375147A1-20161229-C00543
  • wherein:
  • SPR is a reactive linker;
  • RAA1 is an amino acid side chain;
  • RAA1 is an amino acid side chain; and
  • A is arylene or heteroarylene.
  • Compounds of Formula PP1 can be prepared by contacting a compound of Formula PP3 with the compound of Formula PP7:
  • Figure US20160375147A1-20161229-C00544
  • wherein:
  • SPR is a reactive linker;
  • RAA1 is an amino acid side chain;
  • RAA1 is an amino acid side chain; and
  • A is arylene or heteroarylene.
  • Compounds of Formula PP2 can be prepared by contacting a compound of Formula PP8 with a bifunctional spacer:
  • Figure US20160375147A1-20161229-C00545
  • Compounds of Formula PP7 can be prepared by contacting a compound of Formula PP9 with a bifunctional spacer:
  • Figure US20160375147A1-20161229-C00546
  • Bifunctional spacers are compounds that react with the compound of Formula PP3 to append the SPR moiety present in the compounds of Formula PP2. Illustrative bifunctional spacers include, but are not limited to:
  • Figure US20160375147A1-20161229-C00547
  • Compounds of Formula PP8 can be prepared by contacting a compound of Formula PP10 with a compound of Formula PP11, following by removal of the protecting group:
  • Figure US20160375147A1-20161229-C00548
  • wherein PG is an amine protecting group and Y is a moiety that renders the carbonyl to which it is attached electrophilic. Compound of Formula PP10 can be prepared by coupling its corresponding amino acids using standard amino acid coupling techniques, including, for example, active ester formation using HATU, BOP/HOBt, or EDC/N-hydroxysuccinamide in the presence of DIEA, DBU, or tributylamine.
  • Bifunctional spacers are compounds that react with the compound of Formula PP9 to append the SPR moiety present in the compounds of Formula PP7. Illustrative bifunctional spacers include, but are not limited to:
  • Figure US20160375147A1-20161229-C00549
  • Antibody drug conjugate compounds of Formula (I) can also be prepared by reacting a suitable antibody, e.g., deglycosylated antibody or aglycosylated antibody with a compound of Formula (PT1) in the presence of transglutaminase:
  • Figure US20160375147A1-20161229-C00550
  • wherein:
  • A is arylene or heteroarylene; and
  • L is a linker.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00551
  • wherein:
      • R1 is, independently at each occurrence, halo, haloalkyl, haloalkoxy, hydroxyl, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, aryl, alkaryl, aralkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00552
  • or azido,
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, A is:
  • Figure US20160375147A1-20161229-C00553
  • wherein:
      • R1 is, independently at each occurrence, halo, haloalkyl, hydroxyl, alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, heteroaryl, heteroalkyl, heterocycloalkyl, cyano, nitro,
  • Figure US20160375147A1-20161229-C00554
  • or azido
      •  wherein RA is alkyl or heteroalkyl;
      • n is an integer from 0 to 4;
      • m is an integer from 0 to 3;
      • p is an integer from 0 to 6; and
      • q is an integer from 0 to 5.
  • In some embodiments, R1 is, independently, alkyl or halo. In some embodiments, R1 is, independently, C1-6 alkyl, C1-6 haloalkyl, or halo. In some embodiments, R1 is, independently, halo. In some embodiments, R1 is, independently, fluoro, chloro, bromo, iodo, or trifluoromethyl. In some embodiments, n, m, p, or q is 0, 1 or 2. In some embodiments, n, m, p, or q is 0 or 1. In some embodiments, n, m, p, or q is 0.
  • In some embodiments, R1 is
  • Figure US20160375147A1-20161229-C00555
  • In some embodiments, R1 is
  • Figure US20160375147A1-20161229-C00556
  • wherein RA is methyl. In some embodiments, R1 is hydroxyl. In some embodiments, R1 is N-methylformamide. In some embodiments, R1 is morpholinyl.
  • In some embodiments, the linker comprises one or more amino acids. Suitable amino acids include natural, non-natural, standard, non-standard, proteinogenic, non-proteinogenic, and L-, or D-α-amino acids. In some embodiments, the linker comprises alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, or citrulline, or derivative thereof.
  • In some embodiments, the linker comprises valine and citrulline.
  • In some embodiments, the linker is:

  • ├AA1-AA2
  • wherein:
  • one
  • Figure US20160375147A1-20161229-C00557
  • one or more bonds to the payload;
  • the other
  • Figure US20160375147A1-20161229-C00558
  • is one or more bonds to the —NH2 of PT1;
  • AA1 is an amino acid; and
  • AA2 is an amino acid.
  • The linker may further comprise a divalent moiety that connects the AA1-AA2 moiety to the —NH2 of PT1. Suitable divalent moieties include, but are not limited to, those comprising alkylene or polyethylene glycol. The divalent moiety may comprise one or more reactive groups to facilitate bonding to the rest of the compound, or one or more residues of such reactive groups.
  • PT1 includes a primary amine-terminated alkylene or a primary amine-terminated polyethylene glycol. The primary amine-terminating moiety can be directly bonded to a deglycosylated antibody or aglycosylated antibody in the presence of transglutaminase.
  • In some embodiments, the compound comprises a primary amine-terminated alkylene. In some embodiments, the compound comprises a NH2—C5-7 alkylene. In some embodiments, the compound comprises:
  • Figure US20160375147A1-20161229-C00559
  • wherein:
  • Figure US20160375147A1-20161229-C00560
  • is a bond to the payload; and
  • b is an integer from 2 to 8.
  • In some embodiments, the compound comprises:
  • Figure US20160375147A1-20161229-C00561
  • wherein:
  • Figure US20160375147A1-20161229-C00562
  • is a bond to the payload.
  • In some embodiments, the compound of PT1 is
  • Figure US20160375147A1-20161229-C00563
  • In some embodiments, the compound of Formula (I) is prepared by contacting a binding agent with PT1 in the presence of transglutaminase under conditions suitable for a transglutamination reaction. In some embodiments, the transglutaminase reaction is at a pH between about 7 and about 8 for at least 4 hr. In some examples, the pH is 7.2, 7.3, 7.4, 7.5, 7.6, 7.8, or 8.
  • In some embodiments, the compound of Formula (I) is prepared by a transglutaminase reaction wherein the concentration of the compound of Formula (PT1) is at a concentration of at least 30 molar equivalents compared to the deglycosylated antibody or aglycosylated antibody. In some embodiments, the compound of Formula (I) is prepared by a transglutaminase reaction wherein the concentration of the compound of Formula (PT1) is at a concentration of 30 to 150 molar equivalents compared to the deglycosylated antibody or aglycosylated antibody.
  • In some embodiments, the compound of Formula (I) is prepared by a transglutaminase reaction wherein the concentration of the compound of Formula (PT1) is 1 to 30 U per milligram of deglycosylated antibody or aglycosylated antibody.
  • In some embodiments, the antibody is deglycosylated with peptide N-glycosidase F (PNGaseF) prior to the transglutaminase reaction.
  • In some embodiments, the antibody is aglycosylated. An aglycosylated antibody can be prepared by mutagenesis techniques to remove one or more amino acid sequences that are necessary for glycosylation of the antibody. In certain embodiments the antibody comprises a heavy chain with a mutation that substitutes another amino acid for N180. In certain embodiments, the aglycosylated antibody comprises one or more N180Q heavy chain polypeptides.
  • In some embodiments, the compound of Formula (I) is prepared by a transglutaminase reaction which is conducted in one or more solvent(s) selected from the group consisting of water, buffered water, saline water, buffered saline water, and an organic.
  • In some embodiments, the compound of Formula (I) is prepared by a transglutaminase reaction which is conducted in water buffered with phosphate, HEPES, or MOPS.
  • In some embodiments, the compound of Formula (I) is prepared by a transglutaminase reaction which includes reacting the glutaminyl-modified antibody with a reactive spacer compound to form an antibody-spacer conjugate; and then reacting the antibody-spacer conjugate with a reactive payload compound to form an antibody-spacer-payload conjugate.
  • In some embodiments, provided herein is a glutaminyl-modified antibody produced by a method set forth herein.
  • In some embodiments, provided herein is a pharmaceutical composition comprising a glutaminyl-modified antibody produced by a method set forth herein.
  • In some embodiments, provided herein is a method of treating a condition in a subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of the antibody or antibody-drug-conjugate provided herein.
  • In some embodiments, provided herein is an antibody or antibody-drug-conjugate described herein for therapy.
  • In some embodiments, provided herein is an antibody or antibody-drug-conjugate described herein for the treatment of cancer.
  • E. Methods of Use and Pharmaceutical Compositions
  • The present disclosure includes methods of treating or preventing diseases, conditions, or disorders e.g., proliferative diseases such as cancer, comprising administering a therapeutically effective amount or one or more of the compounds disclosed herein, e.g., one or more of the compounds of Formula (I) or (II). Diseases, disorders, and/or conditions include, but are not limited to, those associated with the antigens listed herein. In some embodiments, the antigen is PSMA, MUC16, or EGFRvIII.
  • The compounds disclosed herein can be used for treating primary and/or metastatic tumors arising in the brain and meninges, oropharynx, lung and bronchial tree, gastrointestinal tract, male and female reproductive tract, muscle, bone, skin and appendages, connective tissue, spleen, immune system, blood forming cells and bone marrow, liver and urinary tract, and special sensory organs such as the eye. In certain embodiments, the compounds provided herein are used to treat one or more of the following cancers: renal cell carcinoma, pancreatic carcinoma, head and neck cancer, prostate cancer, malignant gliomas, osteosarcoma, colorectal cancer, gastric cancer (e.g., gastric cancer with MET amplification), malignant mesothelioma, multiple myeloma, ovarian cancer, small cell lung cancer, non-small cell lung cancer, synovial sarcoma, thyroid cancer, breast cancer, or melanoma. In some embodiments, the cancer is breast cancer.
  • The compounds described herein can be administered alone or together with one or more additional therapeutic agents. The one or more additional therapeutic agents can be administered just prior to, concurrent with, or shortly after the administration of the compounds described herein. The present disclosure also includes pharmaceutical compositions comprising any of the compounds described herein in combination with one or more additional therapeutic agents, and methods of treatment comprising administering such combinations to subjects in need thereof.
  • Suitable additional therapeutic agents include, but are not limited to: an EGFR antagonist (e.g., an anti-EGFR antibody [e.g., cetuximab or panitumumab] or small molecule inhibitor of EGFR [e.g., gefitinib or erlotinib]), an antagonist of another EGFR family member such as Her2/ErbB2, ErbB3 or ErbB4 (e.g., anti-ErbB2 [e.g., trastuzumab or T-DM1 {KADCYLA®}], anti-ErbB3 or anti-ErbB4 antibody or small molecule inhibitor of ErbB2, ErbB3 or ErbB4 activity), an antagonist of EGFRvIII (e.g., an antibody that specifically binds EGFRvIII), a cMET antagonist (e.g., an anti-cMET antibody), an IGF1R antagonist (e.g., an anti-IGF1R antibody), a B-raf inhibitor (e.g., vemurafenib, sorafenib, GDC-0879, PLX-4720), a PDGFR-α inhibitor (e.g., an anti-PDGFR-α antibody), a PDGFR-β inhibitor (e.g., an anti-PDGFR-β antibody or small molecule kinase inhibitor such as, e.g., imatinib mesylate or sunitinib malate), a PDGF ligand inhibitor (e.g., anti-PDGF-A, -B, -C, or -D antibody, aptamer, siRNA, etc.), a VEGF antagonist (e.g., a VEGF-Trap such as aflibercept, see, e.g., U.S. Pat. No. 7,087,411 (also referred to herein as a “VEGF-inhibiting fusion protein”), anti-VEGF antibody (e.g., bevacizumab), a small molecule kinase inhibitor of VEGF receptor (e.g., sunitinib, sorafenib or pazopanib)), a DLL4 antagonist (e.g., an anti-DLL4 antibody disclosed in US 2009/0142354 such as REGN421), an Ang2 antagonist (e.g., an anti-Ang2 antibody disclosed in US 2011/0027286 such as H1H685P), a FOLH1 antagonist (e.g., an anti-FOLH1 antibody), a STEAP1 or STEAP2 antagonist (e.g., an anti-STEAP1 antibody or an anti-STEAP2 antibody), a TMPRSS2 antagonist (e.g., an anti-TMPRSS2 antibody), a MSLN antagonist (e.g., an anti-MSLN antibody), a CA9 antagonist (e.g., an anti-CA9 antibody), a uroplakin antagonist (e.g., an anti-uroplakin [e.g., anti-UPK3A] antibody), a MUC16 antagonist (e.g., an anti-MUC16 antibody), a Tn antigen antagonist (e.g., an anti-Tn antibody), a CLEC12A antagonist (e.g., an anti-CLEC12A antibody), a TNFRSF17 antagonist (e.g., an anti-TNFRSF17 antibody), a LGR5 antagonist (e.g., an anti-LGR5 antibody), a monovalent CD20 antagonist (e.g., a monovalent anti-CD20 antibody such as rituximab), etc. Other agents that may be beneficially administered in combination with compounds of the disclosure include, e.g., tamoxifen, aromatase inhibitors, and cytokine inhibitors, including small-molecule cytokine inhibitors and antibodies that bind to cytokines such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-9, IL-11, IL-12, IL-13, IL-17, IL-18, or to their respective receptors.
  • Suitable therapeutic agents also include, but are not limited to chemotherapeutic agents, including alkylating agents such as thiotepa and cyclosphosphamide (Cytoxan™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK™; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxanes, e.g. paclitaxel (Taxol™, Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel (Taxotere™; Aventis Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • The compounds described herein can also be administered and/or co-formulated in combination with antivirals, antibiotics, analgesics, corticosteroids, steroids, oxygen, antioxidants, COX inhibitors, cardioprotectants, metal chelators, IFN-gamma, and/or NSAIDs.
  • In some embodiments of the methods described herein, multiple doses of a compound described herein (or a pharmaceutical composition comprising a combination of an compound described herein and any of the additional therapeutic agents mentioned herein) may be administered to a subject over a defined time course. The methods according to this aspect of the disclosure comprise sequentially administering to a subject multiple doses of a compound described herein. As used herein, “sequentially administering” means that each dose of the compound is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months). The present disclosure includes methods which comprise sequentially administering to the patient a single initial dose of a compound described herein, followed by one or more secondary doses of the compound, and optionally followed by one or more tertiary doses of the compound.
  • The terms “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration of the compounds described herein. Thus, the “initial dose” is the dose which is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”); the “secondary doses” are the doses which are administered after the initial dose; and the “tertiary doses” are the doses which are administered after the secondary doses. The initial, secondary, and tertiary doses can all contain the same amount the compound described herein, but generally can differ from one another in terms of frequency of administration. In certain embodiments, the amount of the compound contained in the initial, secondary and/or tertiary doses varies from one another (e.g., adjusted up or down as appropriate) during the course of treatment. In certain embodiments, two or more (e.g., 2, 3, 4, or 5) doses are administered at the beginning of the treatment regimen as “loading doses” followed by subsequent doses that are administered on a less frequent basis (e.g., “maintenance doses”).
  • In certain exemplary embodiments of the present disclosure, each secondary and/or tertiary dose is administered 1 to 26 (e.g., 1, 1½, 2, 2½, 3, 3½, 4, 4½, 5, 5½, 6, 6½, 7, 7½, 8, 8½, 9, 9½, 10, 10½, 11, 11½, 12, 12½, 13, 13½, 14, 14½, 15, 15½, 16, 16½, 17, 17½, 18, 18½, 19, 19½, 20, 20½, 21, 21½, 22, 22½, 23, 23½, 24, 24½, 25, 25½, 26, 26½, or more) weeks after the immediately preceding dose. The phrase “the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose the compound which is administered to a patient prior to the administration of the very next dose in the sequence with no intervening doses.
  • The methods according to this aspect of the disclosure may comprise administering to a patient any number of secondary and/or tertiary doses of the compound. For example, in certain embodiments, only a single secondary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient. Likewise, in certain embodiments, only a single tertiary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient. The administration regimen may be carried out indefinitely over the lifetime of a particular subject, or until such treatment is no longer therapeutically needed or advantageous.
  • In embodiments involving multiple secondary doses, each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 2 weeks or 1 to 2 months after the immediately preceding dose. Similarly, in embodiments involving multiple tertiary doses, each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 2 to 12 weeks after the immediately preceding dose. In certain embodiments of the disclosure, the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the treatment regimen. The frequency of administration may also be adjusted during the course of treatment by a physician depending on the needs of the individual patient following clinical examination.
  • The present disclosure includes administration regimens in which 2 to 6 loading doses are administered to a patient at a first frequency (e.g., once a week, once every two weeks, once every three weeks, once a month, once every two months, etc.), followed by administration of two or more maintenance doses to the patient on a less frequent basis. For example, according to this aspect of the disclosure, if the loading doses are administered at a frequency of once a month, then the maintenance doses may be administered to the patient once every six weeks, once every two months, once every three months, etc.
  • The present disclosure includes pharmaceutical compositions of the compounds and/or conjugates described herein, e.g., the compounds of Formula (I) and (II), e.g., compositions comprising a compound described herein, a salt, stereoisomer, polymorph thereof, and a pharmaceutically acceptable carrier, diluent, and/or excipient. Examples of suitable carriers, diluents and excipients include, but are not limited to: buffers for maintenance of proper composition pH (e.g., citrate buffers, succinate buffers, acetate buffers, phosphate buffers, lactate buffers, oxalate buffers and the like), carrier proteins (e.g., human serum albumin), saline, polyols (e.g., trehalose, sucrose, xylitol, sorbitol, and the like), surfactants (e.g., polysorbate 20, polysorbate 80, polyoxolate, and the like), antimicrobials, and antioxidants.
  • F. Examples
  • Proton NMR spectra were acquired on Varian Inova 300 or 500 MHz instruments, while mass spectra were collected on an Agilent 1100 or 1200 series LC/MSD with electrospray ionization source and either single-quad or ion trap analyzer. Certain linker payloads in enzymatic assays were analyzed by a Waters Xevo TQ-S mass spectrometer. All starting materials and solvents were purchased commercially and used without purification, unless otherwise noted.
  • Example 1
  • Compound 10 was synthesized from Compound 1 as described below and as depicted in FIG. 1.
  • Maytansin-N-Methyl-L-Alanine-4-Aminobenzamido-Cit-Val-Cap-Mal (10)
  • Step A:
  • To a round-bottom flask was weighed Boc-L-valine (1.03 g, 4.74 mmol), N-hydroxysuccinimide (1.22 g, 10.6 mmol), and EDC (1.60 g, 8.35 mmol). The reagents were dissolved in dry DCM (30 mL), the flask sealed via rubber septum, purged with argon, and the reaction stirred at ambient temperature. After 3 days no Boc-valine remained by TLC (after staining with ninhydrin), so the reaction was washed with water and sat. aq. NaHCO3, the aqueous layer extracted with DCM, combined organic layers washed with brine, dried over Na2SO4, and filtered. The filtrate was then evaporated and dried in vacuo giving Boc-L-valine-succinate as a white solid (1.52 g, 100%). 1H-NMR (300 MHz, CDCl3): δ 4.98 (br d, 1H), 4.58 (dd, 1H), 2.82 (m, 4H), 2.27 (m, 1H), 1.44 (s, 9H), 1.03 (dd, 6H).
  • Boc-L-Valine-L-Citrulline (3):
  • Boc-L-valine-succinate (1) of the preceding step (1.50 g, 4.77 mmol) was dissolved in acetonitrile (MeCN, 15 mL), treated with a solution of L-citrulline (2, 1.07 g, 6.11 mmol) in water (9 mL) and sat. aq. NaHCO3 (6 mL), the flask sealed with a vented septum, and the reaction stirred at ambient temperature. The reaction was incomplete after 18 h, so additional sat. aq. NaHCO3 (3 mL) was added to bring the pH up to ca. 7 and the reaction stirred another 36 h. The reaction was partially concentrated in vacuo to remove MeCN and washed once with ethyl acetate (EtOAc) to remove any nonpolar impurities. The aqueous layer was then acidified to pH 3 with 10% v/v HCl, saturated with NaCl, and extracted 4 times with 9:1 EtOAc/isopropanol. The combined organic extracts were washed with brine, dried over Na2SO4, and filtered. The filtrate was then evaporated and dried in vacuo giving the title compound as a white solid (1.56 g, 87%). MS (ESI, pos.): calc'd for C16H30N4O6, 374.2. found 375.2 (M+H), 397.2 (M+Na).
  • Boc-L-Valine-L-Citrulline-p-Aminobenzoic Acid t-Butyl Ester (5):
  • Step B:
  • The product of the preceding step (3, 152 mg, 0.406 mmol), tert-butyl-4-aminobenzoate (4, 150 mg, 0.776 mmol), and 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (HATU, 488 mg, 1.28 mmol) were weighed into a round-bottom flask and dissolved in anhydrous N,N-dimethylformamide (DMF, 3 mL). N,N-Diisopropylethylamine (DIEA, 0.25 mL, 1.44 mmol) was added to the reaction, the flask sealed via rubber septum, purged with argon, and the reaction stirred at ambient temperature. After 18 h the reaction was purified directly on a 100 g C18 RediSep Gold column via ISCO system (gradient elution: 20-80% MeCN in water, 0.05% acetic acid in both, over 20 min). The product-containing fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized overnight giving an impure white solid (115 mg). This was dissolved in DCM and repurified on a 12 g silica gel RediSep column via ISCO (gradient elution: 0-10% methanol in DCM over 12 min), and the slower-running product fractions evaporated and dried in vacuo giving the title compound as a pale yellow solid (65 mg, 29%). MS (ESI, pos.): calc'd for C27H43N5O7, 549.3. found 450.3 (M-Boc+H), 572.3 (M+Na), 1099.5 (2M+H), 1121.5 (2M+Na).
  • L-Valine-L-Citrulline-p-Aminobenzoic Acid (6):
  • Step C:
  • The title compound was prepared using the method of Mehta et al. (Tet. Lett. 1992, 33, 5441-5444). The product of the preceding step (5, 61 mg, 0.111 mmol) was dissolved in dry DCM (3 mL) in a round-bottom flask, and treated with trifluoroacetic acid (TFA, 110 uL, 1.44 mmol) and triethylsilane [TES (Et3SiH), 50 uL, 0.313 mmol]. The flask was sealed via septum, purged with argon, and stirred at ambient temperature for 18 h. The reaction was incomplete by LCMS, so additional TFA (90 uL) and TES (25 uL) were added and the reaction stirred another 6 h. The reaction was still incomplete so it was capped and stored at −20° C. for 3 d. After warming to ambient temperature and stirring another 24 h it was concentrated in vacuo to an oil, triturated twice with diethyl ether, and dried under high vacuum giving the title compound as an off-white solid (55 mg, 98%). MS (ESI, pos.): calc'd for C18H27N5O5, 393.2. found 394.0 (M+H), 787.2 (2M+H). 1H-NMR (500 MHz, DMSO-d6) showed a mixture of amide rotamers: δ 10.52 (s, 0.5H), 10.46 (s, 0.5H), 8.83 (d, 0.5H), 8.71 (d, 0.5H), 8.06 (br s, 3H), 7.89 (m, 2H), 7.72 (m, 2H), 6.03 (m, 1H), 4.55 (m, 1H), 3.05 (m, 1H), 2.97 (m, 1H), 2.10 (m, 1H), 1.73 (m, 1H), 1.63 (m, 1H), 1.5-1.3 (m, 2H), 0.95 (m, 6H).
  • 6-(Maleimidyl-Caprolyl)-L-Valine-L-Citrulline-p-Aminobenzoic Acid (8):
  • Step D:
  • The product of the preceding step (6, 55 mg, 0.108 mmol) was dissolved in water (3 mL), treated with sat. aq. NaHCO3, then with a solution of 6-maleimidyl-caproic acid succinate ester (56 mg, 0.182 mmol) in MeCN (3 mL). The flask was capped under argon and the reaction stirred at ambient temperature for 22 h. The reaction was complete by LCMS, so it was partially concentrated in vacuo and purified directly on a 30 g C18 Aq RediSep Gold column via ISCO (gradient elution: 20-80% MeCN in water, 0.05% acetic acid in both, over 12 min). The major product fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized overnight giving an impure pale yellow solid (92 mg). This was found to be impure by LCMS so it was dissolved in MeCN/water and repurified on a 100 g C18 Aq Gold column (gradient elution: 0-50% MeCN in water, 0.05% acetic acid in both, over 20 min). The cleanest product fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized giving the title compound as a white solid (34 mg, 53%). MS (ESI, pos.): calc'd for C28H38N6O8, 586.3. found 587.3 (M+H), 609.3 (M+Na). 1H-NMR (500 MHz, DMSO-d6) showed a mixture of amide rotamers: δ 10.26 (s, 0.6H), 10.11 (s, 0.4H), 8.43 (d, 0.4H), 8.13 (d, 0.6H), 7.93-7.70 (m, 4H), 6.99 (m, 2H), 5.97 (m, 1H), 5.41 (m, 2H), 4.38 (m, 1H), 4.21-4.12 (m, 1H), 3.36 (m, 2H), 3.02 (m, 1H), 2.95 (m, 1H), 2.17 (m, 1H), 2.12 (m, 1H), 1.95 (m, 1H), 1.78-1.58 (m, 2H), 1.48 (m, 6H), 1.36 (m, 1H), 1.18 (m, 2H), 0.85 (m, 6H).
  • Maytansin-N-Methyl-L-Alanine-4-Aminobenzamide-Citrulline-Valine-Caprolyl-6-Maleimidyl (10):
  • Step E:
  • The product of the preceding step (8, 33 mg, 0.056 mmol), HATU (33 mg, 0.087 mmol), and maytansin-N-methyl-L-alanine (9, prepared as a gold solid from maytansinol using the methods described in U.S. Patent Application 2007/0037972 A1, 25 mg, 0.038 mmol), were weighed into a round-bottom flask, dissolved in anhydrous DMF (2 mL), and treated with DIEA (20 uL, 0.115 mmol). The flask was sealed via rubber septum, purged with argon, and the reaction stirred at ambient temperature for 20 h. The reaction was diluted with water (1 mL) and purified directly on a 50 g C18 Aq RediSep Gold column via ISCO (gradient elution: 20-80% MeCN in water, 0.05% acetic acid in both, over 12 min). The product fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized overnight giving the title compound as a white solid (8 mg, 17%). MS (ESI, pos.): calc'd for C60H80N9O16Cl, 1217.5. found 1218.6 (M+H), 1200.7 (M−H2O+H), 1240.7 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 9.25 (s, 1H), 7.68-7.61 (m, 2H), 7.33 (d, 2H), 6.91 (s, 1H), 6.84 (s, 1H), 6.75 (d, 1H), 6.67 (s, 2H), 6.45 (dd, 1H), 6.27 (br s, 1H), 6.21 (d, 1H), 5.74 (dd, 1H), 5.44 (m, 1H), 4.98 (m, 1H), 4.88 (d, 1H), 4.77 (t, 1H), 4.53 (br s, 1H), 4.33-4.25 (m, 2), 4.00 (s, 3H), 3.65 (d, 1H), 3.48 (m, 4H), 3.56 (s, 3H), 3.20 (m, 1H), 3.11 (d, 1H), 3.05 (m, 3H), 2.88 (s, 3H), 2.69 (t, 1H), 2.26-2.19 (m, 3H), 2.10 (m, 2H), 1.94 (m, 1H), 1.70-1.55 (m, 6H), 1.66 (s, 3H), 1.46 (d, 3H), 1.33-1.26 (m, 7H), 0.96 (m, 6H), 0.85 (s, 3H).
  • Example 2
  • Compound 15 was synthesized as described below and as depicted in FIG. 6.
  • Maytansin-N-Methyl-L-Alanine-(4-Amino-2-Fluoro)Benzamido-Cit-Val-Cap-Mal (15)
  • Boc-L-Valine-L-Citrulline-(4-Amino-2-Fluoro)Benzoic Acid t-Butyl Ester (12).
  • Step A:
  • Following the procedure of Wipf & Heimgartner (Helv. Chim. Acta, 1998, 71, 140-154), Boc-L-valine-L-citrulline (3, 155 mg, 0.414 mmol) and dicyclohexylcarbodiimide (DCC, 95 mg, 0.460 mmol) were dissolved in dry dichloromethane (DCM, 3 mL), cooled to 0° C., and stirred for 5 min. (+)-Camphor-10-sulfonic acid (CSA, 15 mg, 0.065 mmol) and tert-butyl-4-amino-2-fluorobenzoate (99 mg, 0.469 mmol) were then added dry and the reaction allowed to slowly warm to ambient temperature while stirring for 3 d. LCMS analysis showed a large new peak with m/z 566 (ESI, neg.). The reaction was diluted with DCM and washed with 10% v/v HCl, water, and saturated NaHCO3. The aqueous layers were each extracted once with DCM, and the combined organic layers washed with brine, dried over Na2SO4, and filtered. The filtrate was then evaporated and dried in vacuo giving a pale gold solid which was purified on a 24 g RediSep Gold column via ISCO (gradient elution: Ethyl Acetate—5:5:1 EtOAc/DCM/methanol over 12 min). The cleanest product fractions were combined, concentrated in vacuo, and dried under high vacuum giving the title compound as a white solid (95 mg, 40%). MS (ESI, pos.): calc'd for C27H42N5O7F, 567.3. found 568.3 (M+H), 590.4 (M+Na).
  • L-Valine-L-Citrulline-(4-Amino-2-Fluoro)Benzoic Acid Trifluoroacetate Salt (13).
  • Step B:
  • The title compound was prepared from the product of the preceding step (12, 94 mg, 0.166), using Step C, Example 1, to give an off-white solid (112 mg). MS (ESI, pos.): calc'd for C18H26N5O5F, 411.2. found 412.2 (M+H), 395.2 (M−H2O+H).
  • 6-(Maleimido)-Caproamidyl-L-Valine-L-Citrulline-(4-Amino-2-Fluoro)Benzoic Acid (14):
  • Step C:
  • The title compound was prepared from the product of the preceding step (13, 106 mg, 0.166 mmol), using Step D, Example 1, to give a white solid (92 mg) that was only 70% pure by LCMS but used without further purification. MS (ESI, pos.): calc'd for C28H37N6O8F, 604.3. found 605.2 (M+H), 627.2 (M+Na).
  • Maytansin-N-Methyl-L-Alanine-(4-Amino-2-Fluoro)Benzamido-Cit-Val-Cap-Mal (15):
  • Step D:
  • The title compound was prepared from the product of the preceding step (14, 50 mg, 0.077 mmol) and maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol), using Step E, Example 1, to give a white solid (18 mg) that was only 55% pure by LCMS. Purifying twice by HPLC using a Phenomenex Gemini C18 5u, 30×150 mm column (20-80%, then 40-60%, MeCN in water, 0.1% HOAc both phases, over 20 min, 30 mL/min) gave the title compound as a white solid (3 mg, 3%). MS (ESI, pos.): calc'd for C60H79N9O16ClF, 1235.5. found 1236.5 (M+H), 1258.5 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 9.40 (s, 1H), 7.64 (d, 1H, J=12 Hz), 7.42 (s, 1H), 7.14 (t, 1H, J=8 Hz), 6.90 (s, 1H), 6.86 (s, 1H), 6.77 (m, 1H), 6.68 (s, 2H), 6.46 (dd, 1H, J=15 Hz, 11 Hz), 6.25 (br s, 1H), 6.19 (br m, 1H), 5.75 (dd, 1H, J=15 Hz, 9 Hz), 5.48 (br m, 1H), 4.88 (d, 1H, J=12 Hz), 4.76 (m, 1H), 4.29 (t, 1H, J=11 Hz), 4.23 (t, 1H, J=7 Hz), 4.01 (s, 2H), 3.99 (m, 1H), 3.70 (m, 1H), 3.53-3.47 (m, 4H), 3.36 (s, 3H), 3.20 (m, 1H), 3.13 (d, 1H, J=12 Hz), 3.03 (m, 3H), 2.81 (s, 2H), 2.67 (dd, 1H, J=15 Hz, 12 Hz), 2.25 (t, 1H, J=7 Hz), 2.21 (m, 2H), 2.10 (m, 1H), 1.70-1.64 (m, 2H), 1.67 (s, 3H), 1.46-1.41 (m, 6H), 1.33-1.25 (m, 10H), 0.99-0.95 (m, 6H), 0.89-0.80 (m, 1H), 0.85 (s, 3H).
  • Example 3
  • Compound 20 was synthesized as described below and as depicted in FIG. 7.
  • Maytansin-N-Methyl-L-Alanine-(4-Amino-2-Trifluoromethyl)Benzamido-Cit-Val-Cap-Mal (20)
  • Boc-L-Valine-L-Citrulline-(4-Amino-2-Trifluoromethyl)Benzoic Acid t-Butyl Ester (17):
  • Step A:
  • The title compound was prepared from Boc-L-valine-L-citrulline (3, 175 mg, 0.467 mmol) and tert-butyl-4-amino-2-trifluoromethylbenzoate (150 mg, 0.574 mmol), using the method of Wipf & Heimgartner (Helv. Chim. Acta, 1998, 71, 140-154) to give a white solid (77 mg, 27%). MS (ESI, neg.): calc'd for C28H42N5O7F3, 617.3. found 616.4 (M−H).
  • L-Valine-L-Citrulline-(4-Amino-2-Trifluoromethyl)Benzoic Acid Trifluoroacetate Salt (18):
  • Step B:
  • The title compound was prepared from the product of the preceding step (17, 67 mg, 0.108), using Step C, Example 1, to give an off-white solid (77 mg). MS (ESI, pos.): calc'd for C19H26N5O5F3, 461.2. found 462.3 (M+H), 445.2 (M−H2O+H).
  • 6-(Maleimido)-Caproamidyl-L-Valine-L-Citrulline-(4-Amino-2-Trifluoromethyl)Benzoic Acid (19):
  • Step C:
  • The title compound was prepared from the product of the preceding step (18, 75 mg, 0.108 mmol), using Step D, Example 1, to give a white solid (47 mg, 66%). MS (ESI, pos.): calc'd for C29H37N6O8F3, 654.3. found 655.3 (M+H).
  • Maytansin-N-Methyl-L-Alanine-(4-Amino-2-Trifluoromethyl)Benzamido-Cit-Val-Cap-Mal (20):
  • Step D:
  • The title compound was prepared from the product of the preceding step (19, 34 mg, 0.052 mmol) and maytansin-N-methyl-L-alanine (9, 34 mg, 0.052 mmol), using Step E, Example 1, to give a white solid (11 mg, 16%) after a second ISCO purification (100 g C18 Aq Gold column, 30-70% MeCN in water, 0.05% HOAc both, over 15 min, 50 mL/min). MS (ESI, pos.): calc'd for C61H79N9O16ClF3, 1285.5. found 1287.4 (M+H), 1268.4 (M−H2O+H), 1308.4 (M+Na). 1H-NMR (500 MHz, DMSO-d6): δ 10.4 (s, 1H), 8.16 (d, 1H, J=7 Hz), 8.13 (s, 1H), 7.80 (d, 1H, J=8 Hz), 7.25 (s, 1H), 7.12 (m, 1H), 6.99 (s, 2H), 6.93 (s, 1H), 6.84 (s, 1H), 6.63-6.55 (m, 2H), 6.01 (s, 1H), 5.95 (m, 1H), 5.58 (dd, 1H, J=15 Hz, 9 Hz), 5.38 (m, 3H), 4.64 (dd, 1H, J=12 Hz, 3 Hz), 4.30 (m, 1H), 4.17-4.08 (m, 2H), 3.96 (s, 2H), 3.93 (m, 1H), 3.53 (d, 1H, J=9 Hz), 3.40 (br m, 1H), 3.36 (m, 1H), 3.27 (s, 3H), 3.25 (m, 1H), 3.04 (s, 3H), 3.03-2.92 (m, 2H), 2.84-2.70 (m, 2H), 2.53 (m, 2H), 2.20-2.09 (m, 3H), 1.94 (m, 1H), 1.70 (m, 1H), 1.64 (s, 3H), 1.60-1.53 (m, 3H), 1.51-1.44 (m, 6H), 1.40 (m, 1H), 1.37-1.32 (m, 3H), 1.30-1.24 (m, 2H), 1.20-1.12 (m, 5H), 0.89 (m, 1H), 0.86 (s, 3H), 0.84-0.80 (m, 6H).
  • Example 4
  • Compound 25 was synthesized as described below and as depicted in FIG. 8.
  • Maytansin-N-Methyl-L-Alanine-(4-Amino-2-Methoxy)Benzamido-Cit-Val-Cap-Mal (25)
  • Boc-L-Valine-L-Citrulline-(4-Amino-2-Methoxy)Benzoic Acid t-Butyl Ester (22):
  • Step A:
  • The title compound was prepared from Boc-L-valine-L-citrulline (3, 143 mg, 0.382 mmol) and tert-butyl-4-amino-2-methoxybenzoate (109 mg, 0.488 mmol), using the method of Wipf & Heimgartner (Helv. Chim. Acta, 1998, 71, 140-154) to give a white solid (92 mg, 42%). MS (ESI, neg.): calc'd for C28H45N5O8, 579.3. found 580.3 (M−H), 602.3 (M+Na).
  • L-Valine-L-Citrulline-(4-Amino-2-Methoxy)Benzoic Acid Trifluoroacetate Salt (23):
  • Step B:
  • The title compound was prepared from the product of the preceding step (22, 90 mg, 0.155), using Step C, Example 1, to give a pale solid (99 mg) that was triturated twice with DCM, dissolved in MeCN and THF, filtered, and the solvent evaporated in vacuo to give the title compound as an off-white solid (79 mg, 95%). MS (ESI, pos.): calc'd for C19H29N5O6, 423.2. found 424.2 (M+H), 407.2 (M−H2O+H), 446.2 (M+Na).
  • 6-(Maleimido)-Caproamidyl-L-Valine-L-Citrulline-(4-Amino-2-Methoxy)Benzoic Acid (24).
  • Step C:
  • The title compound was prepared from the product of the preceding step (23, 76 mg, 0.141 mmol), using Step D, Example 1, to give a white solid (50 mg, 57%). MS (ESI, pos.): calc'd for C29H40N6O9, 616.3. found 617.2 (M+H).
  • Maytansin-N-Methyl-L-Alanine-(4-Amino-2-Methoxy)Benzamido-Cit-Val-Cap-Mal (25).
  • Step D:
  • The title compound was prepared from the product of the preceding step (24, 49 mg, 0.079 mmol) and maytansin-N-methyl-L-alanine (9, 34 mg, 0.052 mmol), using Step E, Example 1, to give a white solid (34 mg, 34%). MS (ESI, pos.): calc'd for C61H82N9O17Cl, 1247.6. found 1248.5 (M+H), 1230.5 (M−H2O+H), 1270.5 (M+Na). 1H-NMR (500 MHz, DMSO-d6): δ 9.26 (br s, 1H), 7.49 (br m, 1H), 6.99 (m, 1H), 6.95 (s, 1H), 6.87 (s, 1H), 6.85 (m, 1H), 6.69 (s, 2H), 6.45 (dd, 1H, J=15 Hz, 11 Hz), 6.29 (br s, 1H), 5.73 (dd, 1H, J=16 Hz, 10 Hz), 4.84 (d, 1H, J=12 Hz), 4.70 (br m, 1H), 4.30 (t, 1H, J=12 Hz), 4.01 (s, 3H), 3.74 (br m, 4H), 3.55-3.47 (m, 5H), 3.35 (s, 3H), 3.14 (m, 2H), 3.02 (m, 4H), 2.83 (br s, 3H), 2.71 (s, 3H), 2.65 (m, 1H), 2.60 (t, 1H, J=8 Hz), 2.24-2.18 (m, 3H), 2.09 (m, 1H), 1.77 (pentet, 1H, J=8 Hz), 1.70-1.61 (m, 6H), 1.67 (s, 3H), 1.51-1.40 (m, 6H), 1.30-1.26 (m, 5H), 0.94 (m, 6H), 0.85 (s, 3H).
  • Example 5
  • Compound 27 was synthesized from Compound 26 as described below and as depicted in FIG. 9.
  • Maytansin-N-Methyl-L-Alanine-4-Aminobenzamide (27) Step A: Maytansin-N-Methyl-L-Alanine-(4-Nitro)Benzamide:
  • To a dry, round-bottom flask was weighed maytansin-N-methyl-L-alanine (9, 96 mg, 0.15 mmol), 4-nitrobenzoic acid (26) (42 mg, 0.25 mmol), and HATU (0.12 g, 0.31 mmol). The reagents were dissolved in anhydrous DMF (3.0 mL), treated with DIEA (0.10 mL, 0.57 mmol), and the flask purged with argon and sealed with a rubber septum. The reaction was stirred at ambient temperature for 3 d, after which LCMS showed complete conversion of Maytan-NMA, so it was diluted with a few mL of water and purified directly on a 100 g C18 Aq Gold column (gradient elution: 20-80% MeCN in water, 0.05% acetic acid in both, over 15 min). The cleanest product fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized overnight giving the title compound as a yellow solid (64 mg, 54%). MS (ESI, pos.): calc'd for C39H47N4O12Cl, 798.3. found 798.4 (M+H).
  • Step B: Maytan-NMA-(4-Amino)Benzamide (27):
  • The product of the preceding step (63 mg, 0.079 mmol) and zinc dust (<10 um, 98+% pure, 108 mg, 1.65 mmol) were dissolved/suspended in a mixture of THF (4 mL) and water (1 mL). Acetic acid (0.180 mL, 3.14 mmol) was added to the mixture, the flask sealed with a rubber septum, and the reaction stirred at ambient temperature for 1 h. LCMS of the crude mixture showed complete conversion, so the reaction was filtered over Celite, washed off with MeCN, and the filtrate concentrated in vacuo. The crude product was purified directly on a 50 g C18 Aq Gold column (gradient elution: 20-80% MeCN in water, 0.05% acetic acid in both, over 12 min). The cleanest product fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized overnight giving 46 mg of white solid that was only 88% pure by LCMS. This was dissolved in 1:1 MeCN/water (3 mL) and repurified by HPLC using a Phenomenex Gemini C18 5u, 30×150 mm column in two injections (40-80% and 30-70% MeCN in water, 0.05% HOAc both phases, over 20 min, 30 mL/min), and the cleanest fraction were concentrated, frozen, and lyophilized as above giving the title compound as a white solid (31 mg, 48%). MS (ESI, pos.): calc'd for C41H53N4O12Cl, 768.3. found 751.2 (M−H2O+H), 769.2 (M+H). 1H-NMR (500 MHz, CDCl3): δ 7.24 (d, 2H, J=9 Hz), 6.93 (s, 1H), 6.82 (s, 1H), 6.76 (d, 1H, J=12 Hz), 6.57 (d, 2H, J=9 Hz), 6.45 (dd, 1H, J=16 Hz, 12 Hz), 6.23 (s, 1H), 5.74 (dd, 1H, J=16 Hz, 9 Hz), 5.43 (br m, 1H), 4.87 (dd, 1H, J=12 Hz, 3 Hz), 4.32 (m, 1H), 3.99 (s, 3H), 3.85 (s, 2H), 3.65 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=9 Hz), 3.47 (br s, 1H), 3.36 (s, 3H), 3.10 (d, 1H, J=13 Hz), 3.07 (s, 3H), 3.04 (d, 1H, J=9 Hz), 2.93 (s, 3H), 2.67 (m, 1H), 2.20 (dd, 1H, J=14 Hz, 3 Hz), 1.67 (m, 1H), 1.66 (s, 3H), 1.51-1.47 (m, 2H), 1.44 (d, 3H, J=7 Hz), 1.31 (d, 3H, J=7 Hz), 1.27 (m, 1H), 0.84 (s, 3H).
  • Example 6
  • Compound 29 was synthesized from Compound 28 as described below and as depicted in FIG. 10.
  • Maytansin-N-Methyl-L-Alanine-(2-Fluoro-4-Amino)Benzamide (29) Step A: Maytansin-N-Methyl-L-Alanine-(2-Fluoro-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 40 mg, 0.056 mmol) and 2-fluoro-4-nitrobenzoic acid (28) (26 mg, 0.140 mmol), using Step A of Example 5, to give a light yellow solid (16 mg, 35%). MS (ESI, pos.): calc'd for C39H46N4O12ClF, 816.3. found 817.2 (M+H), 839.2 (M+Na).
  • Step B: Maytan-NMA-(2-Fluoro-4-Amino)Benzamide (29).
  • The title compound was prepared from the product of the preceding step (15 mg, 0.018 mmol), using Step B of Example 5, to give a white solid (8 mg, 50%). MS (ESI, pos.): calc'd for C39H48N4O10ClF, 786.3. found 769.2 (M−H2O+H), 787.2 (M+H), 809.3 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.05-6.99 (m, 2H), 6.92 (s, 1H), 6.85 (s, 1H), 6.81 (d, 1H, J=11 Hz), 6.47 (dd, 1H, J=15 Hz, 11 Hz), 6.36-6.29 (m, 2H), 6.22 (s, 1H), 5.73 (dd, 1H, J=16 Hz, 9 Hz), 5.48 (m, 1H), 4.87 (dd, 1H, J=12 Hz, 3 Hz), 4.30 (m, 1H), 4.00 (s, 3H), 3.93 (s, 2H), 3.73 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=9 Hz), 3.41 (br m, 1H), 3.36 (s, 3H), 3.13 (d, 1H, J=12 Hz), 3.04 (s, 3H), 3.02 (m, 1H), 2.83 (s, 3H), 2.66 (dd, 1H, J=15 Hz, 13 Hz), 2.19 (dd, 1H, J=15 Hz, 3 Hz), 1.67 (s, 3H), 1.63 (m, 1H), 1.51-1.45 (m, 2H), 1.43 (d, 3H, J=7 Hz), 1.30 (d, 3H, J=7 Hz), 1.27 (m, 1H), 0.84 (s, 3H).
  • Example 7
  • Compound 31 was synthesized as described below and as depicted in FIG. 11.
  • Maytansin-N-Methyl-L-Alanine-(2-Trifluoromethyl-4-Amino)Benzamide (31) Step A: Maytansin-N-Methyl-L-Alanine-(2-Trifluoromethyl-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 68 mg, 0.105 mmol) and 2-trifluoromethyl-4-nitrobenzoic acid (30) (37 mg, 0.157 mmol), using Step A of Example 5, to give a pale yellow solid (82 mg, 90%). MS (ESI, pos.): calc'd for C40H46N4O12ClF3, 866.3. found 867.1 (M+H), 889.1 (M+Na).
  • Step B: Maytan-NMA-(2-Trifluoromethyl-4-Amino)Benzamide (31):
  • The title compound was prepared from the product of the preceding step (79 mg, 0.091 mmol), using Step B of Example 5, to give a white solid (29 mg, 35%). MS (ESI, pos.): calc'd for C40H48N4O10ClF3, 836.3. found 818.8 (M−H2O+H), 836.8 (M+H), 858.0 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.00-6.91 (m, 3H), 6.85 (d, 1H, J=3 Hz), 6.75 (br d, 1H, J=18 Hz), 6.63 (d, 1H, J=11 Hz), 6.45 (dd, 1H, J=26 Hz, 19 Hz), 6.23 (s, 1H), 5.73 (dd, 1H, J=26 Hz, 15 Hz), 4.88 (dd, 1H, J=20 Hz, 5 Hz), 4.31 (m, 1H), 4.01 (s, 3H), 3.96 (m, 1H), 3.66 (d, 1H, J=22 Hz), 3.52 (d, 1H, J=15 Hz), 3.37 (s, 3H), 3.14 (s, 3H), 3.11 (m, 1H), 3.03 (d, 1H, J=16 Hz), 2.72 (m, 1H), 2.66 (s, 3H), 2.23 (dd, 1H, J=24 Hz, 5 Hz), 1.66 (s, 3H), 1.51-1.45 (m, 2H), 1.43 (d, 3H, J=12 Hz), 1.31 (d, 3H, J=11 Hz), 1.27 (m, 1H), 0.87 (s, 3H).
  • Example 8
  • Compound 33 was synthesized as described below and as depicted in FIG. 12.
  • Maytansin-N-Methyl-L-Alanine-(2-Methoxy-4-Amino)Benzamide (33) Step A: Maytansin-N-Methyl-L-Alanine-(2-Methoxy-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 68 mg, 0.105 mmol) and 2-methoxy-4-nitrobenzoic acid (32) (32 mg, 0.162 mmol), using Step A of Example 5, to give a pale yellow solid (78 mg, 90%). MS (ESI, pos.): calc'd for C40H49N4O13Cl, 828.3. found 811.1 (M−H2O+H), 829.2 (M+H), 851.2 (M+Na).
  • Step B: Maytan-NMA-(2-Methoxy-4-Amino)Benzamide (33):
  • The title compound was prepared from the product of the preceding step (75 mg, 0.090 mmol), using Step B of Example 5, to give a white solid (62 mg, 79%). MS (ESI, pos.): calc'd for C40H51N4O11Cl, 798.3. found 781.2 (M−H2O+H), 799.2 (M+H), 821.2 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.03 (d, 1H, J=14 Hz), 6.99 (s, 1H), 6.94 (d, 1H, J=12 Hz), 6.86 (s, 1H), 6.81 (d, 1H, J=8 Hz), 6.46 (dd, 1H, J=16 Hz, 11 Hz), 6.24 (s, 1H), 6.17 (s, 1H), 6.11 (d, 1H, J=8 Hz), 5.73 (dd, 1H, J=15 Hz, 9 Hz), 5.54 (m, 1H), 4.81 (m, 1H), 4.31 (t, 1H, J=11 Hz), 4.00 (s, 3H), 3.81 (d, 1H, J=13 Hz), 3.79 (m, 1H), 3.52 (d, 1H, J=9 Hz), 3.36 (s, 3H), 3.12 (d, 1H, J=13 Hz), 3.04 (m, 1H), 3.02 (s, 3H), 2.72 (s, 3H), 2.64 (t, 1H, J=12 Hz), 2.18 (dd, 1H, J=14 Hz, 3 Hz), 1.66 (s, 3H), 1.63 (m, 2H), 1.51-1.45 (m, 2H), 1.41 (d, 3H, J=7 Hz), 1.30 (d, 3H, J=7 Hz), 1.26 (m, 1H), 0.84 (s, 3H).
  • Example 9
  • Compound 35 was synthesized as described below and as depicted in FIG. 13.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Trifluoromethyl-4-Amino)Benzamide (35) Step A: Maytansin-N-Methyl-L-Alanine-(3-Trifluoromethyl-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 46 mg, 0.071 mmol) and 3-trifluoromethyl-4-nitrobenzoic acid (34) (25 mg, 0.106 mmol), using the method from Step A of Example 5, to give a light yellow solid (37 mg, 61%). MS (ESI, pos.): calc'd for C40H46N4O12ClF3, 866.3. found 849.2 (M−H2O+H), 867.2 (M+H), 889.2 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(3-Trifluoromethyl-4-Amino)Benzamide (35).
  • The title compound was prepared from the product of the preceding step (36 mg, 0.042 mmol), using the method from Step B of Example 5, to give a white solid (17 mg, 46%). MS (ESI, pos.): calc'd for C40H48N4O10ClF3, 836.3. found 819.2 (M−H2O+H), 837.2 (M+H), 859.2 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.54 (s, 1H), 7.37 (d, 1H, J=8 Hz), 6.91 (br s, 1H), 6.83 (s, 1H), 6.70 (d, 1H, J=11 Hz), 6.66 (d, 1H, J=8 Hz), 6.45 (dd, 1H, J=15 Hz, 11 Hz), 6.28 (s, 1H), 5.73 (dd, 1H, J=16 Hz, 9 Hz), 5.44 (m, 1H), 4.88 (dd, 1H, J=12 Hz, 3 Hz), 4.40 (s, 2H), 4.30 (t, 1H, J=11 Hz), 3.99 (s, 3H), 3.63 (d, 1H, J=13 Hz), 3.52 (d, 1H, J=9 Hz), 3.36 (s, 3H), 3.12 (d, 1H, J=13 Hz), 3.03 (m, 4H), 2.92 (s, 3H), 2.69 (m, 1H), 2.21 (dd, 1H, J=14 Hz, 3 Hz), 1.65 (m, 4H), 1.52-1.44 (m, 4H), 1.31 (d, 3H, J=6 Hz), 1.26 (m, 2H), 0.85 (s, 3H).
  • Example 10
  • Compound 37 was synthesized as described below and as depicted in FIG. 14.
  • Maytansin-N-Methyl-L-Alanine-N-(2-Chloro-4-Amino-5-Fluoro)Benzamide (37)
  • Step A: Maytansin-N-Methyl-L-Alanine-(2-Chloro-4-Nitro-5-Fluoro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 46 mg, 0.071 mmol) and 3-trifluoromethyl-4-nitrobenzoic acid (36.26 mg, 0.118 mmol), using the method from Step A of Example 5, to give a white solid (33 mg, 55%). MS (ESI, pos.): calc'd for C39H45N4O12Cl2F, 850.2. found 833.1 (M−H2O+H), 851.1 (M+H), 873.1 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(2-Chloro-4-Amino-5-Fluoro)Benzamide (37):
  • The title compound was prepared from the product of the preceding step (36 mg, 0.042 mmol), using the method from Step B of Example 5, to give a white solid (17 mg, 46%). MS (ESI, pos.): calc'd for C39H47N4O10Cl2F, 820.3. found 803.2 (M−H2O+H), 821.2 (M+H), 843.2 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 6.88 (d, 2H, J=13 Hz), 6.83 (d, 1H, J=11 Hz), 6.77 (d, 1H, J=8 Hz), 6.72 (d, 1H, J=10 Hz), 6.46 (dd, 1H, J=15 Hz, 11 Hz), 6.25 (s, 1H), 5.71 (dd, 1H, J=16 Hz, 10 Hz), 5.52 (m, 1H), 4.85 (dd, 1H, J=12 Hz, 3 Hz), 4.30 (t, 1H, J=11 Hz), 4.01 (s, 3H), 3.92 (s, 2H), 3.73 (d, 1H, J=13 Hz), 3.52 (d, 1H, J=9 Hz), 3.37 (s, 3H), 3.15 (d, 1H, J=13 Hz), 3.10 (s, 3H), 3.03 (d, 1H, J=10 Hz), 2.75 (s, 3H), 2.68 (dd, 1H, J=14 Hz, 12 Hz), 2.22 (dd, 1H, J=15 Hz, 3 Hz), 1.67 (s, 3H), 1.61 (m, 1H), 1.50-1.43 (m, 5H), 1.31 (d, 3H, J=6 Hz), 1.27-1.24 (m, 1H), 0.86 (s, 3H).
  • Example 11
  • Compound 39 was synthesized as described below and as depicted in FIG. 16.
  • Maytansin-N-Methyl-L-Alanine-N-(2,5-Difluoro-4-Amino)Benzamide (39) Step A: Maytansin-N-Methyl-L-Alanine-(2,5-Difluoro-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 48 mg, 0.074 mmol) and 2,5-difluoro-4-nitrobenzoic acid (38, 27 mg, 0.133 mmol), using the method from Step A of Example 5, to give a yellow solid (37 mg, 60%). MS (ESI, pos.): calc'd for C39H45N4O12ClF2, 834.3. found 817.2 (M−H2O+H), 835.2 (M+H), 857.2 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(2,5-Difluoro-4-Amino)Benzamide (39).
  • The title compound was prepared from the product of the preceding step (36 mg, 0.043 mmol), using the method from Step B of Example 5, to give a white solid (22 mg, 59%). MS (ESI, pos.): calc'd for C39H47N4O10ClF2, 804.3. found 787.3 (M−H2O+H), 805.3 (M+H), 827.3 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 6.89-6.84 (m, 3H), 6.77 (d, 1H, J=11 Hz), 6.49-6.41 (m, 2H), 6.24 (s, 1H), 5.72 (dd, 1H, J=16 Hz, 9 Hz), 5.47 (q, 1H, J=7 Hz), 4.87 (dd, 1H, J=12 Hz, 3 Hz), 4.30 (td, 1H, J=12 Hz, 2 Hz), 4.00 (m, 5H), 3.71 (d, 1H, J=13 Hz), 3.52 (d, 1H, J=9 Hz), 3.36 (s, 3H), 3.33 (br s, 1H), 3.13 (d, 1H, J=13 Hz), 3.06 (s, 3H), 3.02 (d, 1H, J=10 Hz), 2.83 (d, 3H, J=2 Hz), 2.66 (dd, 1H, J=15 Hz, 12 Hz), 2.18 (dd, 1H, J=14 Hz, 3 Hz), 1.67 (s, 3H), 1.63 (d, 1H, J=14 Hz), 1.51-1.45 (m, 1H), 1.43 (d, 3H, J=7 Hz), 1.31 (d, 3H, J=6 Hz), 1.27 (m, 1H), 0.84 (s, 3H).
  • Example 12
  • Compound 41 was synthesized as described below and as depicted in FIG. 17.
  • Maytansin-N-Methyl-L-Alanine-(3-Fluoro-4-Amino)Benzamide (41) Step A: Maytansin-N-Methyl-L-Alanine-(3-Fluoro-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 47 mg, 0.072 mmol) and 3-fluoro-4-nitrobenzoic acid (40) (24 mg, 0.130 mmol), using the method from Step A of Example 5, to give a yellow solid (40 mg, 68%). MS (ESI, pos.): calc'd for C39H46N4O12ClF, 816.3. found 799.2 (M−H2O+H), 817.2 (M+H), 839.2 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Fluoro-4-Amino)Benzamide (41):
  • The title compound was prepared from the product of the preceding step (39 mg, 0.048 mmol), using the method from Step B of Example 5, to give a white solid (24 mg, 60%). MS (ESI, pos.): calc'd for C39H48N4O10ClF, 786.3. found 769.2 (M−H2O+H), 787.2 (M+H), 809.2 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.11-7.04 (m, 2H), 6.89 (s, 1H), 6.83 (d, 1H, J=2 Hz), 6.72 (d, 1H, J=11 Hz), 6.68 (t, 1H, J=8 Hz), 6.45 (dd, 1H, J=15 Hz, 11 Hz), 6.25 (s, 1H), 5.72 (dd, 1H, J=15 Hz, 9 Hz), 5.43 (m, 1H), 4.86 (dd, 1H, J=12 Hz, 3 Hz), 4.31 (t, 1H, J=11 Hz), 3.99 (s, 3H), 3.92 (m, 2H), 3.62 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=9 Hz), 3.40 (bs s, 1H), 3.36 (s, 3H), 3.12 (m, 1H), 3.08 (s, 3H), 3.04 (d, 1H, J=10 Hz), 2.92 (s, 3H), 2.68 (t, 1H, J=13 Hz), 2.20 (dd, 1H, J=15 Hz, 3 Hz), 1.66 (s, 3H), 1.63 (m, 1H), 1.49-1.46 (m, 1H), 1.44 (d, 3H, J=7 Hz), 1.31 (d, 3H, J=7 Hz), 1.27 (m, 1H), 0.84 (s, 3H).
  • Example 13
  • Compound 43 was synthesized as described below and as depicted in FIG. 18.
  • Maytansin-N-Methyl-L-Alanine-(3-Chloro-4-Amino)Benzamide (43) Step A: Maytansin-N-Methyl-L-Alanine-(3-Chloro-4-Amino)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 45 mg, 0.069 mmol) and 3-chloro-4-nitrobenzoic acid (42) (26 mg, 0.129 mmol), using the method from Step A of Example 5, to give a yellow solid (36 mg, 62%). MS (ESI, pos.): calc'd for C39H46N4O12Cl2, 832.2. found 815.2 (M−H2O+H), 833.2 (M+H), 855.2 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Chloro-4-Amino)Benzamide (43).
  • The title compound was prepared from the product of the preceding step (35 mg, 0.042 mmol), using the method from Step B of Example 5, to give a white solid (24 mg, 67%). MS (ESI, pos.): calc'd for C39H48N4O10Cl2, 802.3. found 785.2 (M−H2O+H), 803.2 (M+H), 825.1 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.35 (s, 1H), 7.17 (d, 1H, J=8 Hz), 6.90 (s, 1H), 6.83 (d, 1H, J=2 Hz), 6.68 (m, 2H), 6.44 (dd, 1H, J=15 Hz, 11 Hz), 6.30 (s, 1H), 5.73 (dd, 1H, J=15 Hz, 9 Hz), 5.42 (m, 1H), 4.87 (dd, 1H, J=12 Hz, 3 Hz), 4.32-4.27 (m, 3H), 3.99 (s, 3H), 3.60 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=10 Hz), 3.44 (bs s, 1H), 3.36 (s, 3H), 3.10 (m, 4H), 3.03 (d, 1H, J=10 Hz), 2.92 (s, 3H), 2.69 (m, 1H), 2.21 (dd, 1H, J=15 Hz, 3 Hz), 1.65 (s, 3H), 1.63 (m, 1H), 1.52-1.45 (m, 1H), 1.43 (d, 3H, J=7 Hz), 1.31 (d, 3H, J=7 Hz), 1.27 (m, 1H), 0.83 (s, 3H).
  • Example 14
  • Compound 45 was synthesized as described below and as depicted in FIG. 19.
  • Maytansin-N-Methyl-L-Alanine-(5-Amino-8-Carboxyquinoline)Carboxamide (45) Step A: Maytansin-N-Methyl-L-Alanine-(5-Nitro-8-Carboxyquinoline)Carboxamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 45 mg, 0.069 mmol) and 5-nitro-8-carboxyquinoline (44) (24 mg, 0.110 mmol), using the method from Step A of Example 5, to give a pale yellow solid (26 mg, 44%). MS (ESI, pos.): calc'd for C42H48N5O12Cl, 849.3. found 832.2 (M−H2O+H), 850.2 (M+H), 872.2 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(5-Amino-8-Carboxyquinoline)Carboxamide (45):
  • The title compound was prepared from the product of the preceding step (25 mg, 0.029 mmol), using the method from Step B of Example 5, to give a pale yellow solid (8 mg, 31%). MS (ESI, pos.): calc'd for C42H50N5O10Cl, 819.3. found 802.3 (M−H2O+H), 820.3 (M+H). 1H-NMR (500 MHz, DMSO-d6): δ 8.80 (br s, 1H), 8.54 (d, 1H, J=10 Hz), 7.39 (br s, 1H), 7.28 (s, 1H), 7.05-7.00 (br m, 2H), 6.90 (s, 1H), 6.84 (m, 1H), 6.62 (dd, 1H, J=15 Hz, 11 Hz), 6.52 (br m, 1H), 6.22 (s, 2H), 5.98 (s, 1H), 5.62 (br m, 2H), 4.56 (br m, 1H), 4.11 (m, 1H), 3.98 (s, 3H), 3.64 (d, 1H, J=13 Hz), 3.53 (d, 1H, J=9 Hz), 3.27 (s, 3H), 2.92-2.88 (m, 3H), 2.81 (d, 1H, J=10 Hz), 2.42 (br s, 2H), 2.07-2.04 (m, 1H), 1.75 (m, 2H), 1.66 (s, 3H), 1.55-1.45 (m, 3H), 1.42 (d, 3H, J=7 Hz), 1.32 (d, 1H, J=14 Hz), 1.24 (s, 1H), 1.14 (d, 3H, J=7 Hz), 0.84 (s, 3H).
  • Example 15
  • Compound 47 was synthesized as described below and as depicted in FIG. 20.
  • Maytansin-N-Methyl-L-Alanine-(3-Bromo-4-Amino)Benzamide (47) Step A: Maytansin-N-Methyl-L-Alanine-(3-Bromo-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 49 mg, 0.075 mmol) and 3-bromo-4-nitrobenzoic acid (46) (30 mg, 0.122 mmol), using the method from Step A of Example 5, to give a yellow solid (38 mg, 58%). MS (ESI, pos.): calc'd for C39H46N4O12BrCl, 876.2/878.2. found 861.1 (M−H2O+H), 879.1 (M+H), 901.1 (M+Na) for the most abundant isotopes.
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Bromo-4-Amino)Benzamide (47):
  • The title compound was prepared from the product of the preceding step (37 mg, 0.042 mmol), using the method from Step B of Example 5, to give a white solid (28 mg, 74%). MS (ESI, pos.): calc'd for C39H48N4O10BrCl, 846.2/848.2. found 831.1 (M−H2O+H), 849.1 (M+H), 871.2 (M+Na) for the most abundant isotopes. 1H-NMR (500 MHz, CDCl3): δ 7.51 (s, 1H), 7.20 (d, 1H, J=8 Hz), 6.90 (s, 1H), 6.83 (s, 1H), 6.68 (m, 2H), 6.44 (dd, 1H, J=15 Hz, 11 Hz), 6.33 (s, 1H), 5.74 (dd, 1H, J=15 Hz, 10 Hz), 5.42 (m, 1H), 4.88 (dd, 1H, J=12 Hz, 3 Hz), 4.31 (m, 3H), 3.99 (s, 3H), 3.60 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=9 Hz), 3.46 (br s, 1H), 3.36 (s, 3H), 3.10 (s, 3H), 3.09 (m, 1H), 3.03 (d, 1H, J=10 Hz), 2.70 (t, 1H, J=13 Hz), 2.21 (dd, 1H, J=15 Hz, 3 Hz), 1.65 (s, 3H), 1.51-1.45 (m, 2H), 1.43 (d, 3H, J=7 Hz), 1.30 (d, 3H, J=6 Hz), 1.27 (m, 1H), 0.85 (s, 3H).
  • Example 16
  • Compound 49 was synthesized as described below and as depicted in FIG. 21.
  • Maytansin-N-Methyl-L-Alanine-(3-Methoxy-4-Amino)Benzamide (49) Step A: Maytansin-N-Methyl-L-Alanine-(3-Methoxy-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 49 mg, 0.075 mmol) and 3-methoxy-4-nitrobenzoic acid (48) (23 mg, 0.117 mmol), using the method from Step A of Example 5, to give a yellow solid (34 mg, 55%). MS (ESI, pos.): calc'd for C40H49N4O13Cl, 828.3. found 811.2 (M−H2O+H), 829.3 (M+H), 851.3 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Methoxy-4-Amino)Benzamide (49).
  • The title compound was prepared from the product of the preceding step (33 mg, 0.040 mmol), using the method from Step B of Example 5, to give a white solid (25 mg, 74%). MS (ESI, pos.): calc'd for C40H51N4O11Cl, 798.3. found 781.2 (M−H2O+H), 799.2 (M+H). 1H-NMR (500 MHz, CDCl3): δ 6.94 (s, 1H), 6.92 (s, 1H), 6.85 (d, 1H, J=8 Hz), 6.81 (s, 1H), 6.74 (br d, 1H, J=10 Hz), 6.54 (d, 1H, J=10 Hz), 6.44 (dd, 1H, J=15 Hz, 11 Hz), 6.33 (s, 1H), 5.76 (m, 1H), 5.43 (br s, 1H), 4.87 (dd, 1H, J=12 Hz, 3 Hz), 4.31 (t, 1H, J=11 Hz), 3.98 (s, 3H), 3.70 (s, 3H), 3.64 (br d, 1H, J=13 Hz), 3.36 (s, 3H), 3.11-3.02 (m, 5H), 2.94 (s, 3H), 2.68 (m, 1H), 2.20 (dd, 1H, J=14 Hz, 3 Hz), 1.67 (m, 1H), 1.65 (s, 3H), 1.49 (dd, 1H, J=9 Hz, 7 Hz), 1.44 (d, 3H, J=7 Hz), 1.30 (d, 3H, J=7 Hz), 1.27 (m, 1H), 0.85 (s, 3H).
  • Example 17
  • Compound 51 was synthesized as described below and as depicted in FIG. 22.
  • Maytansin-N-Methyl-L-Alanine-(2-Methyl-4-Amino)Benzamide (51) Step A: Maytansin-N-Methyl-L-Alanine-(2-Methyl-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 49 mg, 0.075 mmol) and 2-methyl-4-nitrobenzoic acid (50) (24 mg, 0.132 mmol), using the method from Step A of Example 5, to give a pale yellow solid (32 mg, 52%). MS (ESI, pos.): calc'd for C40H49N4O12Cl, 812.3. found 795.3 (M−H2O+H), 813.3 (M+H), 835.3 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(2-Methyl-4-Amino)Benzamide (51):
  • The title compound was prepared from the product of the preceding step (30 mg, 0.037 mmol), using the method from Step B of Example 5, to give a white solid (17 mg, 55%). MS (ESI, pos.): calc'd for C40H51N4O10Cl, 782.3. found 765.2 (M−H2O+H), 783.2 (M+H). 1H-NMR (500 MHz, CDCl3): δ 6.93 (s, 1H), 6.85 (s, 1H), 6.81 (d, 1H, J=8 Hz), 6.77 (d, 1H, J=11 Hz), 6.49 (s, 1H), 6.45 (dd, 1H, J=15 Hz, 11 Hz), 6.34 (d, 1H, J=7 Hz), 6.28 (s, 1H), 5.74 (dd, 1H, J=15 Hz, 9 Hz), 5.38 (m, 1H), 4.90 (m, 1H), 4.32 (t, 1H, J=11 Hz), 4.00 (s, 3H), 3.69 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=9 Hz), 3.36 (s, 3H), 3.15 (m, 1H), 3.12 (s, 3H), 3.01 (d, 1H, J=10 Hz), 2.73 (s, 3H), 2.69 (m, 1H), 2.22 (m, 1H), 2.18 (s, 3H), 1.68 (m, 1H), 1.66 (s, 3H), 1.51 (m, 1H), 1.47 (d, 3H, J=7 Hz), 1.30 (d, 3H, J=6 Hz), 1.28 (m, 1H), 0.87 (s, 3H).
  • Example 18
  • Compound 53 was synthesized as described below and as depicted in FIG. 23.
  • Maytansin-N-Methyl-L-Alanine-(3-Methyl-4-Amino)Benzamide (53) Step A: Maytansin-N-Methyl-L-Alanine-(3-Methyl-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 49 mg, 0.075 mmol) and 3-methyl-4-nitrobenzoic acid (52) (26 mg, 0.143 mmol), using the method from Step A of Example 5, to give a yellow solid (34 mg, 56%). MS (ESI, pos.): calc'd for C40H49N4O12Cl, 812.3. found 795.2 (M−H2O+H), 813.2 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Methyl-4-Amino)Benzamide (53):
  • The title compound was prepared from the product of the preceding step (33 mg, 0.041 mmol), using the method from Step B of Example 5, to give a white solid (24 mg, 71%). MS (ESI, pos.): calc'd for C40H51N4O10Cl, 782.3. found 765.2 (M−H2O+H), 783.2 (M+H). 1H-NMR (500 MHz, CDCl3): δ 7.15 (s, 1H), 7.09 (d, 1H, J=8 Hz), 6.94 (s, 1H), 6.82 (s, 1H), 6.72 (br d, 1H, J=10 Hz), 6.54 (d, 1H, J=8 Hz), 6.44 (dd, 1H, J=15 Hz, 12 Hz), 6.33 (s, 1H), 5.75 (dd, 1H, J=14 Hz, 9 Hz), 5.42 (m, 1H), 4.87 (dd, 1H, J=12 Hz, 10 Hz), 4.31 (t, 1H, J=11 Hz), 3.98 (s, 3H), 3.63 (br d, 1H, J=13 Hz), 3.50 (d, 1H, J=9 Hz), 3.36 (s, 3H), 3.10 (m, 1H), 3.07 (s, 3H), 3.03 (d, 1H, J=10 Hz), 2.91 (s, 3H), 2.68 (t, 1H, J=13 Hz), 2.19 (dd, 1H, J=14 Hz, 3 Hz), 2.06 (s, 3H), 1.66 (m, 1H), 1.65 (s, 3H), 1.48 (dd, 1H, J=11 Hz, 7 Hz), 1.43 (d, 3H, J=7 Hz), 1.29 (d, 3H, J=7 Hz), 1.27 (m, 1H), 0.84 (s, 3H).
  • Example 19
  • Compound 55 was synthesized as described below and as depicted in FIG. 24.
  • Maytansin-N-Methyl-L-Alanine-(8-Amino-5-Carboxyquinoline)Carboxamide (55) Step A: Maytansin-N-Methyl-L-Alanine-(8-Nitro-5-Carboxyquinoline)Carboxamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 47 mg, 0.072 mmol) and 8-nitro-5-carboxyquinoline (54) (35 mg, 0.160 mmol), using the method from Step A of Example 5, to give a pale yellow solid (30 mg, 49%). MS (ESI, pos.): calc'd for C42H48N5O12Cl, 849.3. found 832.6 (M−H2O+H), 850.7 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-(8-Amino-5-Carboxyquinoline)Carboxamide (55):
  • The title compound was prepared from the product of the preceding step (29 mg, 0.034 mmol), using the method from Step B of Example 5, to give a pale yellow solid (18 mg, 60%). MS (ESI, pos.): calc'd for C42H50N5O10Cl, 819.3. found 802.0 (M−H2O+H), 820.0 (M+H). 1H-NMR (500 MHz, CDCl3): δ 8.77 (dd, 1H, J=4 Hz, 2 Hz), 8.18 (d, 1H, J=8 Hz), 7.40 (dd, 1H, J=9 Hz, 4 Hz), 7.23 (m, 1H), 6.99 (s, 1H), 6.87 (s, 1H), 6.73 (d, 1H, J=8 Hz), 6.68 (d, 1H, J=11 Hz), 6.47 (dd, 1H, J=15 Hz, 11 Hz), 6.26 (s, 1H), 5.77 (dd, 1H, J=15 Hz, 9 Hz), 5.32 (br m, 1H), 5.21 (br s, 2H), 4.99 (d, 1H, J=9 Hz), 4.34 (t, 1H, J=11 Hz), 4.01 (s, 3H), 3.78 (br m, 1H), 3.67 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=10 Hz), 3.37 (s, 3H), 3.16 (d, 1H, J=13 Hz), 3.12 (s, 3H), 3.00 (d, 1H, J=10 Hz), 2.79 (s, 3H), 2.73 (m, 1H), 2.24 (dd, 1H, J=14 Hz, 3 Hz), 1.76 (d, 1H, J=14 Hz), 1.68 (s, 3H), 1.61 (d, 3H, J=7 Hz), 1.33 (m, 1H), 1.30 (d, 3H, J=7 Hz), 1.25 (s, 1H), 0.90 (s, 3H).
  • Example 20
  • Compound 60 was synthesized as described below and as depicted in FIG. 25.
  • Maytansin-N-Methyl-L-Alanine-(3-Methoxy-4-Amino)Benzamido-Cit-Val-Cap-Mal (60)
  • Step A: Boc-L-Valine-L-Citrulline-(3-Methoxy-4-Amino)Benzoic Acid t-Butyl Ester (57):
  • The title compound was prepared from Boc-L-valine-L-citrulline (3, 100 mg, 0.267 mmol) and 4-amino-3-methoxybenzoic acid tert-butyl ester (56, 61 mg, 0.273 mmol), using the method from Step A of Example 2, to give a white solid (74 mg, 48%). MS (ESI, pos.): calc'd for C28H45N5O8, 579.3. found 580.4 (M+H), 602.6 (M+Na).
  • Step B: L-valine-L-citrulline-(3-methoxy-4-amino)benzoic acid (58):
  • The title compound was prepared from the product of the preceding step (57, 72 mg, 0.124 mmol), using the method from Step C of Example 1, to give an off-white solid (68 mg, 100%). MS (ESI, pos.): calc'd for C19H29N5O6, 423.2. found 424.4 (M+H), 847.4 (2M+H).
  • Step C: 6-(Maleimidyl-Caprolyl)-L-Valine-L-Citrulline-(3-Methoxy-4-Amino)Benzoic Acid (59):
  • The title compound was prepared from the product of the preceding step (58, 67 mg, 0.124 mmol), using the method from Step D of Example 1, to give a white solid (45 mg, 59%). MS (ESI, pos.): calc'd for C29H40N6O9, 616.3. found 617.5 (M+H), 639.6 (M+Na).
  • Step D: Maytansin-N-Methyl-L-Alanine-(3-Methoxy-4-Amino)Benzamido-Cit-Val-Cap-Mal (60).
  • The title compound was prepared from the product of the preceding step (59, 44 mg, 0.071 mmol) and maytansin-N-methyl-L-alanine (9, 49 mg, 0.075 mmol), using the method from Step E of Example 1, to give a white solid (14 mg, 16%). MS (ESI, pos.): calc'd for C61H82N9O17Cl, 1247.6. found 1231.1 (M−H2O+H), 1249.1 (M+H), 1271.1 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 8.48 (s, 1H), 8.24 (d, 1H, J=8 Hz), 7.11 (d, 1H, J=8 Hz), 6.96-6.93 (m, 3H), 6.83 (s, 1H), 6.72-6.68 (m, 3H), 6.45 (dd, 1H, J=16 Hz, 11 Hz), 6.25 (s, 1H), 6.18 (d, 1H, J=9 Hz), 5.77 (dd, 1H, J=15 Hz, 10 Hz), 5.44 (m, 1H), 5.03 (br s, 1H), 4.90 (d, 1H, J=10 Hz), 4.62 (m, 1H), 4.54 (br s, 2H), 4.33-4.28 (m, 2H), 3.99 (s, 3H), 3.75 (s, 3H), 3.61 (d, 1H, J=13 Hz), 3.52-3.48 (m, 3H), 3.36 (s, 3H), 3.24 (m, 2H), 3.11 (d, 1H, J=13 Hz), 3.07 (s, 3H), 3.03 (d, 1H, J=10 Hz), 2.90 (s, 3H), 2.70 (m, 1H), 2.26-2.20 (m, 3H), 2.12 (m, 1H), 2.00 (m, 1H), 1.78 (m, 1H), 1.63-1.57 (m, 6H), 1.46 (d, 2H, J=7 Hz), 1.33-1.25 (m, 6H), 0.95 (m, 6H), 0.86 (s, 3H).
  • Example 21
  • Compound 63 was synthesized as described below and as depicted in FIG. 26.
  • Maytansin-N-Methyl-L-Alanine-(2-Fluoro-4-Amino)Benzamido-Cit-Val-Cap-6-Amine (63) Step A: Boc-6-Aminohexanoic Acid Succinate Ester:
  • The title compound was prepared from Boc-6-aminohexanoic acid (64, 502 mg, 2.17 mmol), using the method from Step A of Example 1, to give a white solid (712 mg, 99%). MS (ESI, pos.): calc'd for C15H24N2O6, 328.2. found 351.2 (M+Na).
  • Step B: Boc-(6-Amino-Caprolyl)-L-Valine-L-Citrulline (62):
  • The title compound was prepared from the product of the preceding steps (710 mg, 2.16 mmol) and L-valine-L-citrulline TFA salt (970 mg, 2.51 mmol), using the method from Step D of Example 1, to give a pale gold solid (720 mg, 69%). MS (ESI, pos.): calc'd for C22H41N5O7, 487.3. found 488.3 (M+H), (M+H).
  • Step C: Maytansin-N-Methyl-L-Alanine-(2-Fluoro-4-Amino)Benzamido-Cit-Val-Cap-6-Boc-Amine:
  • The title compound was prepared from the product of the preceding step (62, 25 mg, 0.051 mmol) and Maytansin-N-methyl-L-alanine-(2-fluoro-4-amino)benzamide (29, 35 mg, 0.041 mmol), using the method from Step A of Example 2, to give a white solid (17 mg, 33%). MS (ESI, pos.): calc'd for C61H87N9O16ClF, 1255.6. found 1238.5 (M−H2O+H), 1256.6 (M+H), 1278.6 (M+Na).
  • Step D: Maytansin-N-Methyl-L-Alanine-(2-Fluoro-4-Amino)Benzamido-Cit-Val-Cap-6-Amine (63):
  • The product of the preceding step (16 mg, 0.013 mmol) was dissolved in acetonitrile (MeCN, 3 mL) and water (1 mL), treated with trifluoroacetic acid (TFA, 1.0 mL, 13.0 mmol), the flask sealed with a rubber septum, purged with argon, and the reaction stirred at ambient temperature. After 24 h, the reaction was partially concentrated in vacuo at ambient temperature, diluted with water (ca. 1 mL), and purified twice on C18 Aq RediSep Gold columns via ISCO system (20-80% MeCN in water, 0.1% TFA both phases). The purest fractions by LCMS were combined, partially concentrated in vacuo at ambient temperature, frozen at −78° C., and lyophilized to give the title compound as a white solid (9 mg, 56%). MS (ESI, pos.): calc'd for C56H79N9O14ClF, 1155.5. found 1156.6 (M+H), 1178.6 (M+Na). 1H-NMR (500 MHz, CD3OD): δ 8.72 (d, 1H, J=12 Hz), 8.39 (d, 1H, J=13 Hz), 8.14 (d, 1H, J=10 Hz), 7.84 (dd, 1H, J=21 Hz, 3 Hz), 7.75 (dd, 1H, J=21 Hz, 3 Hz), 7.60 (dd, 1H, J=15 Hz, 3 Hz), 7.33 (dd, 1H, J=14 Hz, 3 Hz), 7.25 (m, 1H), 7.21 (s, 1H), 6.97 (s, 1H), 6.75 (m, 1H), 6.72 (s, 1H), 6.67 (m, 1H), 5.78-5.60 (m, 3H), 4.77 (m, 1H), 4.45 (m, 2H), 4.26 (m, 1H), 4.20 (d, 1H, J=13 Hz), 4.05-4.00 (m, 2H), 4.03 (s, 3H), 3.69 (dd, 1H, J=21 Hz, 5 Hz), 3.64 (d, 1H, J=16 Hz), 3.41 (s, 3H), 3.30-3.26 (m, 2H), 3.23-3.09 (m, 2H), 3.05 (m, 3H), 2.97 (dd, 1H, J=16 Hz, 5 Hz), 2.92 (d, 1H, J=13 Hz), 2.86 (m, 3H), 2.82 (d, 1H, J=13 Hz), 2.75 (m, 1H), 2.36-2.29 (m, 2H), 2.25-2.20 (m, 1H), 2.08-2.02 (m, 2H), 1.76 (s, 3H), 1.73-1.50 (m, 10H), 1.45 (m, 2H), 1.36-1.32 (m, 2H), 1.28 (d, 3H, J=11 Hz), 1.05 (d, 3H, J=11 Hz), 1.03-0.98 (m, 4H), 0.93 (s, 3H).
  • Example 22
  • Compound 65 was synthesized as described below and as depicted in FIG. 27.
  • Maytansin-N-Methyl-L-Alanine-N-(2-Methoxy-5-Amino)Benzamide (65) Step A: Maytansin-N-Methyl-L-Alanine-(2-Methoxy-5-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg 0.077 mmol) and 2-methoxy-5-nitrobenzoic acid (64) (25 mg, 0.127 mmol), using the method from Step A of Example 5, to give a white solid (51 mg, 80%). MS (ESI, pos.): calc'd for C40H49ClN4O13, 829.3. found 812.0 (M−H2O+H), 830.0 (M+H), 852.0 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(2-Methoxy-5-Amino)Benzamide (65).
  • The title compound was prepared from the product of the preceding step (50 mg, 0.060 mmol), using the method from Step B of Example 5, to give a white solid (19 mg, 40%). MS (ESI, pos.): calc'd for C40H51ClN4O11, 798.3. found 781.3 (M−H2O), 799.3 (M+H), 822.3 (M+Na).
  • Example 23
  • Compound 67 was synthesized as described below and as depicted in FIG. 28.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Amino-4-Methoxy)Benzamide (67) Step A: Maytansin-N-Methyl-L-Alanine-(3-Nitro-4-Methoxy)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg 0.077 mmol) and 3-nitro-4-methoxybenzoic acid (66) (25 mg, 0.127 mmol), using the method from Step A of Example 5, to give a white solid (46 mg, 72%). MS (ESI, pos.): calc'd for C40H49ClN4O13, 829.3. found 812.0 (M−H2O+H), 830.0 (M+H), 852.0 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(3-Amino-4-Methoxy)Benzamide (67):
  • The title compound was prepared from the product of the preceding step (45 mg, 0.054 mmol), using the method from Step B of Example 5, to give a white solid (23 mg, 53%). MS (ESI, pos.): calc'd for C40H51ClN4O11, 798.3. found 781.3 (M−H2O), 799.3 (M+H), 822.3 (M+Na).
  • Example 24
  • Compound 69 was synthesized as described below and as depicted in FIG. 29.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Amino-5-Fluoro)Benzamide (69) Step A: Maytansin-N-Methyl-L-Alanine-(3-Nitro-5-Fluoro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol) and 3-nitro-5-fluorobenzoic acid (68) (24 mg, 0.127 mmol), using the method from Step A of Example 5, to give a yellow solid (37 mg, 59%). MS (ESI, pos.): calc'd for C39H46ClFN4O12, 816.3. found 817.2 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(3-Amino-5-Fluoro)Benzamide (69):
  • The title compound was prepared from the product of the preceding step (37 mg, 0.045 mmol), using the method from Step B of Example 5, to give a white solid (9.1 mg, 24%). MS (ESI, pos.): calc'd for C39H48ClFN4O10, 786.3. found 769.3 (M−H2O), 787.3 (M+H).
  • Example 25
  • Compound 71 was synthesized as described below and as depicted in FIG. 30.
  • Maytansin-N-Methyl-L-Alanine-N-(2-Fluoro-5-Amino)Benzamide (71) Step A: Maytansin-N-Methyl-L-Alanine-(2-Fluoro-5-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol) and 2-fluoro-5-nitrobenzoic acid (70) (24 mg, 0.127 mmol), using the method from Step A of Example 5, to give a yellow solid (37 mg, 59%). MS (ESI, pos.): calc'd for C39H46ClFN4O12, 816.3. found 799.3 (M−H2O), 817.2 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(2-Fluoro-5-Amino)Benzamide (71).
  • The title compound was prepared from the product of the preceding step (37 mg, 0.045 mmol), using the method from Step B of Example 5, to give a white solid (2.2 mg, 6%). MS (ESI, pos.): calc'd for C39H48ClFN4O10, 786.3. found 769.3 (M−H2O), 787.3 (M+H).
  • Example 26
  • Compound 73 was synthesized as described below and as depicted in FIG. 31.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Amino)Benzamide (73) Step A: Maytansin-N-Methyl-L-Alanine-(3-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol) and 3-nitrobenzoic acid (72) (21 mg, 0.127 mmol), using the method from Step A of Example 5, to give a white solid (34 mg, 56%). MS (ESI, pos.): calc'd for C39H47ClN4O12, 798.3. found 781.2 (M−H2O), 799.3 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(3-Amino)Benzamide (73):
  • The title compound was prepared from the product of the preceding step (34 mg, 0.043 mmol), using the method from Step B of Example 5, to give a white solid (9.3 mg, 27%). MS (ESI, pos.): calc'd for C39H49ClN4O10, 768.3. found 751.2 (M−H2O), 769.2 (M+H).
  • Example 27
  • Compound 75 was synthesized as described below and as depicted in FIG. 32.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Amino-4-Fluoro)Benzamide (75) Step A: Maytansin-N-Methyl-L-Alanine-(3-Nitro-4-Fluoro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol) and 3-nitro-4-fluorobenzoic acid (74) (24 mg, 0.127 mmol), using the method from Step A of Example 5, to give a white solid (34 mg, 54%). MS (ESI, pos.): calc'd for C39H46ClFN4O12, 816.3. found 799.3 (M−H2O), 817.2 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(3-Amino-4-Fluoro)Benzamide (75).
  • The title compound was prepared from the product of the preceding step (34 mg, 0.042 mmol), using the method from Step B of Example 5, to give a white solid (12 mg, 37%). MS (ESI, pos.): calc'd for C39H48ClFN4O10, 786.3. found 769.3 (M−H2O), 787.3 (M+H).
  • Example 28
  • Compound 77 was synthesized as described below and as depicted in FIG. 33.
  • Maytansin-N-Methyl-L-Alanine-N-4-Aminobenzamide-Adipic-NHS (77) Step A: Maytansin-N-Methyl-L-Alanine-N-(4-Amino)Benzamide-Adipic Acid (76):
  • The title compound was prepared from 27 (20 mg, 0.026 mmol) and adipic anhydride (17 mg, 0.133 mmol) were weighed into a round-bottom flask and dissolved in pyridine (1.5 mL). The flask was sealed via rubber septum, purged with nitrogen, and the reaction was stirred at ambient temperature. After 4 h the reaction was purified directly on a 30 g C18 RediSep Gold Aq column via ISCO system (gradient elution: 20-90% MeCN in water, 0.05% acetic acid in both, over 20 min). The product-containing fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized to give an off-white solid (16 mg, 67%). MS (ESI, pos.): calc'd for C45H57ClN4O13, 896.4. found 879.4 (M−H2O), 897.4 (M+H).
  • Step B: Maytansin-N-Methyl-L-Alanine-N-(4-Amino)Benzamide-Adipic-NHS (77):
  • The title compound was prepared from the product of the preceding step (76, 16 mg, 0.017 mmol), using the method from Step A of Example 1, to give a white solid (10 mg, 58%). MS (ESI, pos.): calc'd for C49H60ClN5O15, 993.5. found 976.0 (M−H2O), 994.0 (M+H). 1H-NMR (500 MHz, CDCl3): δ 7.92 (s, 1H), 7.57 (d, 2H, J=8 Hz), 7.36 (d, 2H, J=8 Hz), 6.93 (s, 1H), 6.86 (s, 1H), 6.75 (d, 1H, J=12 Hz), 6.47 (dd, 1H, J=15 Hz, 11 Hz), 6.29 (s, 1H), 5.74 (dd, 1H, J=16 Hz, 9 Hz), 5.47 (m, 1H), 4.90 (dd, 1H, J=12 Hz, 3 Hz), 4.32 (t, 1H, J=11 Hz), 4.02 (s, 4H), 3.63 (d, 1H, J=13 Hz), 3.62 (br s, 1H), 3.53 (d, 1H, J=9 Hz), 3.38 (s, 3H), 3.13 (d, 1H, J=13 Hz), 2 (s, 3H), 2.80 (d, 2H, J=10 Hz), 2.73 (s, 3H), 2.60 (m, 1H), 2.27 (t, 2H, J=10 Hz), 2.11 (br s, 1H), 2.07 (s, 2H), 1.62 (s, 3H), 1.58-1.52 (m, 2H), 1.51-1.46 (m, 2H), 1.34-1.29 (m, 3H), 1.25-1.20 (m, 2H), 1.13 (d, 3H, J=7 Hz), 0.82 (s, 3H).
  • Example 29
  • Compound 78 was synthesized as described below and as depicted in FIG. 34.
  • Maytansin-N-Methyl-L-Alanine-4-Aminobenzamide-Cap-Mal (78)
  • The title compound was prepared from 27 (15 mg, 0.019 mmol) and 6-maleimidohexanoic acid (6 mg, 0.029 mmol), using the method from Step A of Example 5, to give an off-white solid (9.8 mg, 52%). MS (ESI, pos.): calc'd for C49H60ClN5O13, 962.5. found 944.8 (M−H2O), 962.8 (M+H). 1H-NMR (500 MHz, DMSO-d6): δ 10.01 (s, 1H), 7.56 (d, 2H, J=9 Hz), 7.29 (d, 2H, J=9 Hz), 7.21 (s, 1H), 6.99 (s, 2H), 6.91 (s, 1H), 6.83 (s, 1H), 6.62-6.56 (m, 2H), 5.97 (s, 1H), 5.61 (dd, 1H, J=15 Hz, 10 Hz), 5.44 (m, 1H), 4.59 (d, 1H, J=12 Hz), 4.10 (t, 1H, J=12 Hz), 3.94 (s, 3H), 3.51 (d, 1H, J=9 Hz), 3.40-3.36 (m, 2H), 3.26 (s, 3H), 3.23-3.21 (m, 2H), 2.94 (s, 3H), 2.80 (d, 1H, J=10 Hz), 2.73 (s, 3H), 2.64-2.57 (m, 1H), 2.27 (t, 2H, J=10 Hz), 2.11-2.09 (m, 1H), 2.07 (s, 2H), 1.62 (s, 3H), 1.57-1.52 (m, 2H), 1.51-1.46 (m, 2H), 1.34-1.29 (m, 3H), 1.25-1.19 (m, 2H), 1.13 (d, 3H, J=7 Hz), 0.82 (s, 3H).
  • Example 30
  • Compound 80 was synthesized as described below and as depicted in FIG. 35.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Methylsulfonyl-4-Amino)Benzamide (80) Step A: Maytansin-N-Methyl-L-Alanine-(3-Methylsulfonyl-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 40 mg, 0.062 mmol) and 3-methylsulfonyl-4-nitrobenzoic acid (79) (25 mg, 0.102 mmol), using the method from Step A of Example 5, to give a yellow solid (37 mg, 69%). MS (ESI, pos.): calc'd for C50H49N4O14ClS, 876.3. found 857.6 (M−H2O+H), 875.6 (M+H), 897.6 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Methylsulfonyl-4-Amino)Benzamide (80):
  • The title compound was prepared from the product of the preceding step (36 mg, 0.041 mmol), using the method from Step B of Example 5, to give a white solid (28 mg, 76%). MS (ESI, pos.): calc'd for C40H51N4O12ClS, 846.3. found 829.1 (M−H2O+H), 847.1 (M+H), 869.1 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.85 (s, 1H), 7.44 (d, 1H, J=9 Hz), 6.90 (s, 1H), 6.84 (s, 1H), 6.71 (d, 1H, J=9 Hz), 6.66 (d, 1H, J=11 Hz), 6.45 (dd, 1H, J=15 Hz, 11 Hz), 6.23 (s, 1H), 5.72 (dd, 1H, J=15 Hz, 10 Hz), 5.32-5.25 (m, 2H), 4.89 (dd, 1H, J=12 Hz, 3 Hz), 4.31 (t, 1H, J=11 Hz), 3.99 (s, 3H), 3.65 (d, 1H, J=13 Hz), 3.51 (d, 1H, J=9 Hz), 3.36 (s, 3H), 3.14 (d, 1H, J=13 Hz), 3.05 (s, 3H), 3.01 (m, 1H), 2.99 (s, 3H), 2.95 (s, 3H), 2.68 (t, 1H, J=14 Hz), 2.20 (dd, 1H, J=15 Hz, 3 Hz), 1.70 (m, 1H), 1.67 (s, 3H), 1.55 (s, 3H), 1.52 (m, 1H), 1.47 (d, 1H, J=7 Hz), 1.31 (d, 1H, J=7 Hz), 1.28 (m, 1H), 0.85 (s, 3H).
  • Example 31
  • Compound 82 was synthesized as described below and as depicted in FIG. 36.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Hydroxy-4-Amino)Benzamide (82) Step A: Maytansin-N-Methyl-L-Alanine-(3-Hydroxy-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 47 mg, 0.072 mmol) and 3-hydroxy-4-nitrobenzoic acid (81) (20 mg, 0.109 mmol), using the method from Step A of Example 5, to give a yellow solid (29 mg, 49%). MS (ESI, pos.): calc'd for C39H47N4O13Cl, 814.3. found 796.8 (M−H2O+H), 814.8 (M+H), 836.8 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Hydroxy-4-Amino)Benzamide (82):
  • The title compound was prepared from the product of the preceding step (28 mg, 0.034 mmol), using the method from Step B of Example 5, to give a pale yellow solid (20 mg, 69%). MS (ESI, pos.): calc'd for C40H51N4O12ClS, 784.3. found 767.7 (M−H2O+H), 785.7 (M+H). 1H-NMR (500 MHz, CDCl3): δ 9.19 (s, 1H), 7.17 (s, 1H), 6.88 (s, 1H), 6.79 (br s, 2H), 6.63-6.56 (m, 3H), 6.47 (m, 1H), 5.93 (s, 1H), 4.93 (s, 2H), 4.57 (d, 1H, J=11 Hz), 4.09 (t, 1H, J=12 Hz), 3.93 (s, 3H), 3.50 (d, 1H, J=9 Hz), 3.26 (s, 3H), 3.18 (br m, 1H), 2.99 (s, 3H), 2.79 (d, 1H, J=10 Hz), 2.76 (s, 3H), 2.06 (dd, 1H, J=14 Hz, 2 Hz), 1.60 (s, 3H), 1.48-1.46 (m, 2H), 1.33-1.27 (m, 4H), 1.13 (d, 3H, J=6 Hz), 0.80 (s, 3H).
  • Example 32
  • Compound 84 was synthesized as described below and as depicted in FIG. 37.
  • Maytansin-N-Methyl-L-Alanine-N-(2-Amino)Benzamide (84) Step A: Maytansin-N-Methyl-L-Alanine-(2-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 30 mg, 0.046 mmol) and 2-nitrobenzoic acid (83) (15 mg, 0.092 mmol), using the method from Step A of Example 5, to give a yellow solid (26 mg, 71%). MS (ESI, pos.): calc'd for C39H47N4O12Cl, 798.3. found 799.3 (M+H), 821.3 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(2-Amino)Benzamide (84):
  • The title compound was prepared from the product of the preceding step (24 mg, 0.038 mmol), using the method from Step B of Example 5, to give a white solid (13 mg, 54%). MS (ESI, pos.): calc'd for C39H49N4O10Cl, 768.3. found 769.0 (M+H). 1H-NMR (500 MHz, CDCl3): δ 7.16 (m, 1H), 7.08 (d, 1H, J=6 Hz), 6.92 (s, 1H), 6.84 (d, 1H, J=2 Hz), 6.71 (d, 1H, J=8 Hz), 6.63 (t, 1H, J=7 Hz), 6.57 (m, 1H), 6.47 (dd, 1H, J=15 Hz, 11 Hz), 6.27 (s, 1H), 5.76 (m, 1H), 5.20 (br s, 1H), 4.99 (m, 1H), 4.39-4.32 (m, 3H), 4.01 (s, 3H), 3.74 (br s, 1H), 3.59 (d, 1H, J=13 Hz), 3.52 (d, 1H, J=9 Hz), 3.39 (s, 3H), 3.20 (s, 3H), 3.15 (d, 1H, J=12 Hz), 2.99 (d, 1H, J=12 Hz), 2.94 (s, 3H), 2.73 (t, 1H, J=14 Hz), 2.26 (m, 1H), 1.75 (d, 1H, J=12 Hz), 1.69 (s, 3H), 1.55 (d, 3H, J=7 Hz), 1.50 (m, 1H), 1.35-1.30 (m, 4H), 0.90 (s, 3H).
  • Example 33
  • Compound 86 was synthesized as described below and as depicted in FIG. 38.
  • Maytansin-N-Methyl-L-Alanine-N-(4-Methoxy-2-Amino)Benzamide (86) Step A: Maytansin-N-Methyl-L-Alanine-(4-Methoxy-2-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 30 mg, 0.046 mmol) and 4-methoxy-2-nitrobenzoic acid (85) (18 mg, 0.092 mmol), using the method from Step A of Example 5, to give a yellow solid (18 mg, 47%). MS (ESI, pos.): calc'd for C40H49N4O13Cl, 828.3. found 829.4 (M+H), 851.3 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(4-Methoxy-2-Amino)Benzamide (86):
  • The title compound was prepared from the product of the preceding step (18 mg, 0.022 mmol), using the method from Step B of Example 5, to give a white solid (15 mg, 84%). MS (ESI, pos.): calc'd for C40H51N4O11Cl, 798.3. found 799.1 (M+H). 1H-NMR (500 MHz, CDCl3): δ 6.94 (s, 1H), 6.85 (d, 1H, J=2 Hz), 6.79 (dd, 1H, J=9 Hz, 3 Hz), 6.67 (d, 1H, J=9 Hz), 6.65 (m, 1H), 6.60 (d, 1H, J=11 Hz), 6.48 (dd, 1H, J=15 Hz, 12 Hz), 6.27 (s, 1H), 5.76 (m, 1H), 5.22 (br s, 1H), 4.98 (m, 1H), 4.36 (t, 1H, J=11 Hz), 4.01 (s, 3H), 3.93 (br s, 1H), 3.72 (br s, 1H), 3.65 (m, 1H), 3.61 (s, 3H), 3.52 (d, 1H, J=9 Hz), 3.39 (s, 3H), 3.18 (s, 3H), 3.16 (m, 1H), 2.99 (d, 1H, J=10 Hz), 2.94 (s, 3H), 2.72 (t, 1H, J=13 Hz), 2.26 (dd, 1H, J=15 Hz, 3 Hz), 1.74 (d, 1H, J=14 Hz), 1.70 (s, 3H), 1.55 (d, 3H, J=7 Hz), 1.52-1.48 (m, 1H), 1.35-1.30 (m, 4H), 0.90 (s, 3H).
  • Example 34
  • Compound 88 was synthesized as described below and as depicted in FIG. 39.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Morpholino-4-Amino)Benzamide (88) Step A: Maytansin-N-Methyl-L-Alanine-(3-Morpholino-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 30 mg, 0.046 mmol) and 3-morpholino-4-nitrobenzoic acid (87) (23 mg, 0.092 mmol), using the method from Step A of Example 5, to give a yellow solid (28 mg, 70%). MS (ESI, pos.): calc'd for C43H54N5O13Cl, 883.3. found 884.5 (M+H), 906.3 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Morpholino-4-Amino)Benzamide (88):
  • The title compound was prepared from the product of the preceding step (28 mg, 0.032 mmol), using the method from Step B of Example 5, to give an off-white solid (12 mg, 52%). MS (ESI, pos.): calc'd for C43H56N5O11Cl, 853.4. found 853.9 (M+H), 875.9 (M+Na). 1H-NMR (500 MHz, CDCl3): δ 7.12 (s, 1H), 7.02 (d, 1H, J=8 Hz), 6.98 (s, 1H), 6.86 (d, 1H, J=2 Hz), 6.81 (d, 1H, J=11 Hz), 6.61 (d, 1H, J=8 Hz), 6.48 (dd, 1H, J=16 Hz, 12 Hz), 6.29 (s, 1H), 5.77 (dd, 1H, J=15 Hz, 9 Hz), 5.48 (m, 1H), 4.89 (dd, 1H, J=12 Hz, 3 Hz), 4.34 (t, 1H, J=12 Hz), 4.19 (br m, 1H), 4.01 (s, 3H), 3.80-3.75 (m, 5H), 3.53 (m, 2H), 3.39 (s, 3H), 3.15 (d, 1H, J=13 Hz), 3.07 (d, 1H, J=10 Hz), 3.03 (s, 3H), 2.96 (s, 3H), 2.73 (m, 5H), 2.22 (dd, 1H, J=14 Hz, 3 Hz), 1.71 (m, 1H), 1.69 (s, 3H), 1.54-1.49 (m, 1H), 1.47 (d, 3H, J=8 Hz), 1.33 (d, 3H, J=7 Hz), 1.30 (m, 1H), 0.88 (s, 3H).
  • Example 35
  • Compound 90 was synthesized as described below and as depicted in FIG. 40.
  • Maytansin-N-Methyl-L-Alanine-N-(3-Acetamido-4-Amino)Benzamide (90) Step A: Maytansin-N-Methyl-L-Alanine-(3-Acetamido-4-Nitro)Benzamide:
  • The title compound was prepared from maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol) and 3-acetamido-4-nitrobenzoic acid (89) (29 mg, 0.129 mmol), using the method from Step A of Example 5, to give a fluffy pale yellow solid (36 mg, 54%). MS (ESI, pos.): calc'd for C41H50N5O13Cl, 855.3. found 839.1 (M−H2O), 857.1 (M+H), 879.1 (M+Na).
  • Step B: Maytansin-N-Methyl-L-Alanine-(3-Acetamido-4-Amino)Benzamide (90):
  • The title compound was prepared from the product of the preceding step (35 mg, 0.042 mmol), using the method from Step B of Example 5, to give a white solid (19 mg, 57%). MS (ESI, pos.): calc'd for C41H52N5O11Cl, 825.3. found 808.4 (M−H2O), 826.4 (M+H). 1H-NMR (500 MHz, DMSO-d6): δ 9.05 (s, 1H), 7.30 (s, 1H), 7.17 (s, 1H), 6.98 (d, 1H, J=9 Hz), 6.91 (s, 1H), 6.79 (s, 1H), 6.58 (m, 3H), 5.97 (s, 1H), 5.64 (br s, 1H), 5.34 (m, 3H), 4.60 (d, 1H, J=12 Hz), 4.10 (t, 1H, J=12 Hz), 3.93 (s, 3H), 3.50 (d, 1H, J=9 Hz), 3.33 (s, 3H), 3.29 (m, 1H), 3.26 (s, 3H), 3.16 (d, 1H, J=12 Hz), 2.97 (s, 3H), 2.79 (m, 4H), 2.07 (d, 1H, J=13 Hz), 1.95 (s, 2H), 1.59 (s, 3H), 1.51-1.42 (m, 2H), 1.29 (m, 3H), 1.13 (d, 3H, J=6 Hz), 0.81 (s, 3H).
  • Example 36
  • Compound 100 was synthesized as described below and as depicted in FIG. 41.
  • Maytansin-N-Methyl-L-Alanine-N-4-Aminobenzamide-Cit-Val-Cap-diBromomethylacryl (100) Step A: 3-Bromo-2-Bromomethyl-Propionyl Chloride (92):
  • To a 10 mL round bottom flask equipped with a magnetic stirrer and nitrogen inlet was charged 3-romo-2-bromomethyl-propionic acid (91, 1.0 g; 4.1 mmol) and thionyl chloride (3.0 mL). This solution was heated to reflux for 3 hours and concentrated to 0.90 g (84% yield) as a brown oil. 1H-NMR (300 MHz, CDCl3): δ 3.85-3.75 (m, 4H), 3.60 (pentet, 1H, J=9 Hz).
  • Step B: 6-Amino-Hexanoic Acid Tert-Butyl Ester (94):
  • To a 50 mL round bottom flask equipped with a magnetic stirrer and nitrogen inlet was charged 6-aminohexanoic acid (93, 2.0 g; 15 mmol) and thionyl chloride (5.0 mL; 69 mmol; 4.5 equiv.). This solution was stirred at or below 30° C. for 2 hours and concentrated in vacuo to dryness. To the tan semi-solid a slurry of sodium bicarbonate (2.6 g; 30 mmol; 2.0 equiv.) in t-BuOH (5.0 mL; 87 mmol; 5.7 equiv.) was added and the slurry was stirred at ambient temperature for another 2 h. The butanol was removed in vacuo at 40° C. The thick white slurry was diluted with ethyl acetate and washed with 4 portions of 1 N NaOH, 3 portions of H2O, 1 portion of brine. The organics were dried over Na2SO4, filtered and concentrated to afford 2.2 g (77% yield) as a colorless oil. MS (ESI, pos.): calc'd for C10H21NO2, 187.3. found 188.4 (M+H), 1H-NMR (300 MHz, CDCl3): δ 2.68-2.64 (m, 2H), 2.21-2.16 (m, 2H), 1.62-1.52 (m, 2H), 1.48-1.38 (m, 9H), 1.36-1.20 (m, 2H), 1.09 (m, 2H).
  • Step C: 6-(3-Bromo-2-Bromomethyl-Propionylamino)-Hexanoic Acid Tert-Butyl Ester (95):
  • To a 50 mL round bottom flask equipped with a magnetic stirrer and nitrogen inlet was charged 6-aminohexanoic acid tert-butyl ester (94, 0.50 g; 2.7 mmol) and dimethylaminopyridine (0.03 g; 0.27 mmol; 0.10 equiv.) in DCM (5.0 mL). This solution was chilled to 0° C. via an ice bath. 3-Bromo-2-bromomethyl-propionyl chloride (92, 0.90 g; 3.4 mmol; 1.2 equiv.) was dissolved in DCM (5 mL) and slowly added to the reaction mixture at 0° C. Stir and slowly warm to ambient temperature overnight. Dilute reaction mixture with ethyl acetate, wash the organic mixture with H2O, 5% NaHCO3 and brine. The organics were dried over Na2SO4, filtered, concentrated and purified on a silica gel column eluting with 0-100% ethyl acetate in hexanes to afford 0.49 g (42% yield) as a clear yellow oil. 1H-NMR (300 MHz, CDCl3): δ 5.92 (br s, 1H), 3.64-3.58 (m, 2H), 3.54-3.48 (m, 2H), 3.36-3.29 (m, 2H), 2.89-2.83 (m, 1H), 2.24-2.20 (m, 2H), 1.65-1.51 (m, 4H), 1.44-1.35 (m, 11H).
  • Step D: 6-(3-Bromo-2-Bromomethyl-Propionylamino)-Hexanoic Acid (96):
  • To a 50 mL round bottom flask equipped with a magnetic stirrer and nitrogen inlet was charged 6-(3-Bromo-2-bromomethyl-propionylamino)-hexanoic acid tert-butyl ester (95, 0.26 g; 0.62 mmol) and trifluoroacetic acid (0.70 mL; 9.3 mmol; 15 equiv.) in DCM (10 mL). This solution was stirred at ambient temperature overnight, concentrated to dryness, dissolved in acetonitrile and H2O (1.0 mL each), frozen and lyophilized to afford 0.22 g (100%) as a solid. MS (ESI, pos.): calc'd for C10H17Br2NO3, 359.0 found 358.0, 360.0, 362.0 (M+H), 380.0, 382.0, 384.0 (M+Na), 356.0, 358.0, 360.0 (M−H). 1H-NMR (300 MHz, CDCl3): δ 11.97 (s, 1H), 8.20-8.16 (m, 1H), 3.58-3.56 (d, 4H), 3.11-3.04 (m, 2H), 3.02-2.97 (m, 1H), 2.21-2.16 (m, 2H), 1.54-1.37 (m, 4H), 1.33-1.29 (m, 2H).
  • Step E: Boc-Val-Cit-4-Aminobenzoic Acid (97):
  • The title compound was prepared from the product of Example 1, Step D (6, 100 mg, 0.254 mmol) and di-tert-butyl dicarbonate (61 mg, 0.279 mmol), which were weighed into a round-bottom flask and treated with 1 M NaOH (2 mL) in THF (3 mL) and H2O (1.5 mL). The flask was sealed via rubber septum and the reaction was stirred at ambient temperature overnight, concentrated in vacuo and neutralized to pH 7 with dropwise addition of 1 M HCl (2 mL). The aqueous the reaction with ethyl acetate, dried the organic layer over Na2SO4, filter and concentrated to give an off-white solid (93 mg, 74%). MS (ESI, pos.): calc'd for C23H35N5O7, 493.5. found 394.2 (M+1-Boc), 494.2 (M+H), 516.2 (M+Na).
  • Step F: Maytansin-N-Methyl-L-Alanine-N-4-Aminobenzamide-Cit-Val-Boc (98):
  • The title compound was prepared from the product of the preceding step Boc-Val-Cit-4-aminobenzoic acid (97, 76 mg, 0.154 mmol) and maytansin-N-methyl-L-alanine (9, 50 mg, 0.077 mmol) using the method from Step A of Example 5, to give a white solid (22 mg, 26%). MS (ESI, pos.): calc'd for C55H77ClN8O15, 1124.5. found 1107.2 (M−H2O), 1125.3 (M+H), 1147.2 (M+Na).
  • Step G: Maytansin-N-Methyl-L-Alanine-N-4-Aminobenzamide-Cit-Val (99):
  • The title compound was prepared from the product of the preceding step (98, 20 mg, 0.018 mmol) weighed into a round-bottom flask dissolved in ACN (3 mL) and H2O (1 mL) and treated with TFA (1 mL). The flask was sealed via rubber septum, purged with nitrogen, and the reaction was stirred at ambient temperature. After 48 h the reaction was purified directly on a 30 g C18 RediSep Gold Aq column via ISCO system (gradient elution: 10-65% MeCN in water, 0.05% acetic acid in both, over 20 min). The product-containing fractions were combined, partially concentrated in vacuo, frozen on dry ice, and lyophilized to give an off-white solid (8 mg, 46%). MS (ESI, pos.): calc'd for C50H69ClN8O13, 1024.5. found 1007.2 (M−H2O), 1026.2 (M+H).
  • Step H: Maytansin-N-Methyl-L-Alanine-N-4-Aminobenzamide-Cit-Val-Capryl-Bis(BrMe)Acrylamide (100):
  • The title compound was prepared from the product of the preceding step (99, 8 mg, 0.008 mmol) and 6-(3-Bromo-2-bromomethyl-propionylamino)-hexanoic acid (96), using the method from Step A of Example 5, to give a white solid (8 mg, 75%). MS (ESI, pos.): calc'd for C60H84Br2ClN9O15, 1363.4. found 1363.1 (M+H). 1H-NMR (500 MHz, CDCl3): δ 9.32 (s, 1H), 8.72 (m, 1H), 8.37 (m, 1H), 7.65 (m, 2H), 7.45 (m, 1H), 7.34 (m, 1H), 7.32 (m, 2H), 7.22-7.18 (m, 2H), 6.89-6.84 (m, 2H), 6.70-6.60 (m, 2H), 6.44 (m, 1H), 6.32 (br s, 1H), 6.12 (s, 1H), 5.76-5.71 (m, 1H), 5.65 (s, 1H), 5.37-5.29 (m, 3H), 4.86-4.70 (m, 3H), 4.52 (br s, 1H), 4.31-4.25 (m, 1H), 4.12 (m, 1H), 3.99 (s, 3H), 3.65-3.58 (m, 1H), 3.51 (m, 1H), 3.34-3.21 (m, 8H), 3.13-3.02 (m, 5H), 2.88 (s, 3H), 2.68 (m, 1H), 2.28-2.17 (m, 3H), 2.11-1.83 (m, 2H), 1.80-1.70 (m, 2H), 1.66 (s, 3H), 1.57-1.49 (m, 4H), 1.47-1.43 (m, 3H), 1.30-1.26 (m, 7H), 1.00-0.91 (m, 6H), 0.85 (s, 3H).
  • Example 37 Conjugate Preparation and Characterization
  • Five antibodies were conjugated to the linker-payload compounds of the disclosure using the procedures below. The four targeting antibodies used in these experiments were: (1) a PSMA antibody having the heavy and light chain variable domains of clone AB-PG1-XG1-006 as set forth in WO2007002222A2, (2) anti-MUC16 antibody having variable regions derived from clone 3A5 from WO2007001851, and (3) two PRLR antibodies having the heavy and light chain variable domains of clone H1H6765P and H1H6958N2 as set forth in WO2015026907A1. All the monoclonal antibodies were expressed in CHO cells and purified by Protein A. A non-binding isotype control derived from an antigen having no relation to oncology was also used.
  • Example 38 Conjugation Method for Compound 10 Conjugation Method for Maleimides
  • The antibody (1-10 mg/ml) in 50 mM HEPES, 150 mM NaCl, pH 7.5, was treated with 1 mM dithiothreitol at 37° C. for 30 min. After gel filtration (G-25, pH 4.5 sodium acetate), the maleimido linker payload derivative 10, 15, 20, 25, 60, and 78 (1.2 equivalents/SH group) in DMSO (10 mg/ml) was added to the reduced antibody and the mixture adjusted to pH 7.0 with 1M HEPES (pH 7.4). After 1 h the reaction was quenched with excess N-ethyl maleimide. The conjugates were purified by size exclusion chromatography and sterile filtered. Protein concentrations and payload to antibody ratios were determined by UV spectral analysis. Size exclusion HPLC established that all conjugates used were >95% monomeric, and RP-HPLC established that there was <0.5% unconjugated linker payload. All conjugated antibodies were analyzed by UV for linker payload loading values according to Hamblett et al, Cancer Res 2004 10 7063. Yields and payload to antibody ratios are reported in Table 1.
  • Example 39 Conjugation Method for Active Esters
  • The antibodies (1-10 mg/ml) in 50 mM HEPES, 150 mM NaCl, pH 8.0, and 15% (v/v) DMA were conjugated with a 6 fold excess of compound 77 for 1-2 hours at ambient temperature. The conjugates were purified by size exclusion chromatography and sterile filtered. Protein and linker payload concentrations were determined by UV spectral analysis. Size-exclusion HPLC established that all conjugates used were >95% monomeric, and RP-HPLC established that there was <0.5% unconjugated linker payload. Yields are reported in Table 1 based on protein. All conjugated antibodies were analyzed by UV for linker payload loading values according to Hamblett et al, Cancer Res 2004 10 7063. Yields and payload to antibody ratios are reported in Table 1.
  • TABLE 1
    Compound ε252 nm (cm−1 M−1) ε280 nm (cm−1 M−1)
    10 45990 20600
    15 68900 26500
    20 65000 33000
    25 64550 25550
    60 32000 8600
    63 53500 22300
    77 44500 17166
    78 47600 15600
    Antibody ε252 nM (cm−1 M−1) ε280 nm (cm−1 M−1)
    PSMA 77652 224320
    MUC16 85888 247360
    PRLR 80673 220420
    PRLR-Q 82000 195400
    Isotype Control 75113 218360
    Isotype Control-Q 68741 203757
    Antibody Conjugate Payload:Antibody (UV) Yield %
    PSMA-10 2.4 70
    MUC16-10 1.5 50
    Isotype Control-10 2.4 75
    PSMA-25 2.5 65
    MUC16-25 2.0 40
    Isotype Control-25 2.5 65
    PSMA-60 3.9 60
    MUC16-60 1.5 40
    PRLR-60 3.8 70
    Isotype Control-60 3.1 70
    PRLR-Q-63 2.9 (3.3 ESI-MS) 60
    Isotype Control-Q- 3.2 (3.1 ESI-MS) 60
    63
    PSMA-77 4.0 55
    MUC16-77 2.8 40
    PRLR-77 4.3 70
    Isotype Control-77 3.9 65
    PSMA-78 NA NA
    MUC16-78 2.0 40
    PRLR-78 2.8 50
    Isotype Control-78 4.0 70
  • Example 40 In Vitro Linker-Payload Cell-Free Enzymatic Assays Cathepsin B Incubation
  • In vitro cell-free enzymatic assay procedure was adopted from Dubowchik, et al. Bioconjugate Chem. 2002 13 855. The linker payload 10 was set at 100 μg/mL final in 25 mM sodium acetate buffer, 1 mM EDTA, pH 5.0 and pre-incubated at 37° C. Cathepsin B (Sigma #C8571) was activated at room temperature for 15 minutes with 1 equivalent of 30 mM DTT, 15 mM EDTA to 2 equivalents of cathepsin B stock. The activated cathepsin B solution was added to the substrate solutions at a 1:20 molar ratio (purified H2O, instead of activated cathepsin B was added for the control sample.) Samples were incubated at 37° C. overnight and the resulting samples are detected by LC-MS through Q1 Scan.
  • LC-MS Detection
  • Samples are centrifuged at 12,000 g for 5 min. Supernatant was recovered and analyzed by liquid chromatography-mass spectrometry (Thermo Quantiva) by combined infusion of 0.3 ml/min of 30:70 mobile phase B:A (Mobile Phase A: 0.1% FA in H2O; Mobile Phase B: 0.1% FA in Acetonitrile) at 20 μl/min from supernatant. MS1 is set at an appropriate range for detection of molecular ion of either linker payload or payload. The supernatant contained the predicted payload, p-amino-benzamide maytansinoid (27), with a mass of 791.27 M+Na (calc'd monoisotopic mass for C39H49ClN4O10, 768.31) and the control samples without cathepsin B contained 10 with a mass of 1240.50 M+Na (calc'd monoisotopic mass for C60H80ClN9O16, 1217.54). No predicted payload molecular ion was detected in the control samples.
  • The results of this Example are significant in part because cathepsin B proteolysis of 10 should only occur after internalization of the ADC in the cell where the enzyme exists. Off target effects should be reduced since the antibody delivers the cytotoxic payload directly to targeted cells.
  • Example 41 In Vitro Cytotoxicity Assays
  • In this Example, the ability of various antibody-drug conjugates or their associated payloads to kill antigen-expressing tumor cells in vitro was assessed.
  • Ovcar3 (Muc16+) or C4-2 (PSMA+) cells were seeded in 96 well plates at 3000 (C42) cells per well in complete growth media and grown overnight. For cell viability curves, serially diluted conjugates or payloads were added to the cells at final concentrations ranging from 300 nM to 5 pM and incubated for 3 days. To measure viability, cells were incubated with CCK8 (Dojindo) for the final 1-3 hours and the absorbance at 450 nm (OD450) was determined on a Victor (Perkin Elmer). Background OD450 levels determined from digitonin (40 nM) treated cells were subtracted from all wells and viability is expressed as a percentage of the untreated controls. IC50 values were determined from a four-parameter logistic equation over a 10-point response curve (GraphPad Prism). All conjugate curves and IC50 values are corrected for payload equivalents.
  • In C4-2 cells (prostate cancer line), natively expressing PSMA at 271 fold above isotype control binding, the maytansinoid conjugates PSMA-10 and PSMA-25 possessed IC50 values of 0.11 and 0.59 nM, respectively (FIGS. 2-5). The payloads 27 and 33 alone had IC50 values of 0.20 and 0.55 nM, respectively. The naked PSMA antibody and isotype control were devoid of any anti-proliferation activity at the concentrations assayed.
  • In Ovcar-3 cells (ovarian cancer line), natively expressing MUC16 at 320 fold above isotype control binding, the maytansinoid conjugates MUC16-10 and MUC16-25 possessed IC50 value of 0.74 and 0.63 nM, respectively (FIGS. 2-5). The payloads 27 and 33 alone had IC50 values of 0.06 and 0.11 nM, respectively. The naked MUC16 antibody and isotype control were devoid of any anti-proliferation activity at the concentrations assayed.
  • Table 2 lists the anti-proliferating ability of the payloads only in both Ovcar3 (Muc16+) or C4-2 (PSMA+) cells. All compounds possess sub-nanomolar activities with compounds 35 and 37 at or near single digit picomolar IC50s.
  • TABLE 2
    C4-2 Ovcar3
    IC50 IC50
    Compound # (nM) % kill (nM) % kill
    29 0.27 83 0.13 93
    33 0.45 86 0.20 93
    31 0.10 82 0.04 93
    27 0.20 84 0.09 92
    35 0.004 84 <0.01 93
    37 0.01 86 <0.01 94
  • Example 42 Antibody Expression
  • Assay/Experiment Type: Cloning, expression and purification of antibodies modified to contain site-specific conjugation motifs
  • This Example provides the generation of antibodies with amino acids sequences that allow site-specific conjugation by transglutaminase reactions.
  • To generate antibodies, mutagenesis was performed on a plasmid encoding the CH1, CH2, and CH3 domains of human IgG1 (amino acids 1 through 330 of UniprotKB accession no. P01857) to generate an N to Q mutation at position 180 using a QuikChange Lightning Multi Site-Directed Mutagenesis Kit (Agilent, #210516). Two antibody variable region heavy (VH) fragments, one encoding the VH of an anti-human PRLR antibody, H1H6958N2 (international patent application WO 2015026907 A1) and another encoding the VH of the isotype control antibody recognizing an exogenous antigen, were selected. Primers were designed (idtdna.com) to amplify the VH regions of these two antibodies using Kapa HiFi DNA polymerase (Kapa Biosciences; # KK2102). While PCR amplification was proceeding, a plasmid containing the human IgG1 N180Q mutation was digested with LguI enzyme (Fermentas, # FD1934) at 37° C. for 30 minutes. Once the amplification was complete, the digested human IgG1 plasmid and both PCR products were run out on a 1% agarose gel containing SYBR Safe stain (Life Technologies, # S33102). Bands of the appropriate size were identified and excised from the gel using a clean razor blade. All excised products were purified using a Gel Extraction kit (Qiagen, #28704). An In-Fusion cloning reaction (Clontech, #638911) was then performed using a ratio of 3:1 of digested human IgG1 vector to VH PCR product and then incubated for 15 minutes at 50° C. After incubation, each reaction was transformed into Mix and Go competent cells (Zymo, # T3007), incubated on ice for 5 minutes, and competent cells were plated on LB+Carbenicillin plates (Teknova, VWR, #101320-126), which were incubated overnight at 37° C.
  • The following day, single colonies were inoculated from the plate into LB broth containing 100 μg/mL Carbenicillin and grown overnight shaking in a table top incubator at 37° C. Cells were then pelleted by centrifugation and miniprepped on a Hamilton Starlet robot using a PureLink HiPure Plasmid miniprep kit (Thermo Fisher, #K210003). Purified DNA was sequenced and results were analyzed using Sequencher software (GeneCodes). A clone from each ligation reaction was selected for re-transformation into Mix and Go competent cells, incubated on ice for 5 minutes and competent cells were plated on LB plates containing Carbenicillin, which were incubated overnight at 37° C.
  • The following day, a single colony was picked from each plate and grown in LB broth containing 100 μg/mL Carbenicillin for 3 to 4 hours. The sample was then diluted into LB broth containing 100 μg/ml Carbenicillin and transferred to a 37° C. shaking incubator to grow overnight. A maxiprep DNA extraction was then performed on both plasmids and the full DNA open reading frames were sequenced. Once the sequences were confirmed, the heavy chain DNA along with previously cloned affiliated light chain DNA was stably transfected into a CHO cell line to produce each antibody.
  • The H1H6958N2 (international patent application WO 2015026907 A1) containing the N180Q Fc mutation is referred to as PRLR-Q, and the isotype control antibody recognizing an exogenous antigen also containing the N180Q Fc mutation is referred to as ISOTYPE CONTROL-Q.
  • Example 43 Bacterial Transglutaminase Conjugation
  • PRLR-Q (MW 145438 Da) and Isotype Control-Q (MW 144602 Da) antibodies were conjugated at 1-10 mg/mL in PBS pH 7.4. Linker payload 63 was added in a 10 to 25-fold molar excess over antibody and the enzymatic reaction was initiated by addition of 1-5 units of bacterial transglutaminase (Zedira, T1001) per mg antibody and incubated with shaking at 37° C. for 4-16 hours. The conjugates were purified by size exclusion chromatography and sterile filtered. Protein and linker payload concentrations were determined by UV spectral analysis. Size-exclusion HPLC established that all conjugates used were >95% monomeric. Yields are reported in Table 1 based on protein. All conjugated antibodies were analyzed by UV for linker payload loading values according to Hamblett et al, Cancer Res 2004 10 7063. In addition, the conjugates were analyzed by ESI-MS for linker payload loadings using a Waters Synapt G2-Si QTOF mass spectrometry coupled with Acquity UPLC. The chromatographic separation was achieved on a C4 column (Waters protein BEH C4, 50 mm×1 mm, 1.7 μm) in a 25 minute gradient (minute:percentage of mobile phase B; 0:20%, 1:20%, 18:40%, 18.1:90, 20:95%, 20.8:95%, 20.9:20% 25:20%). The mobile phase A was 0.1% formic acid in water and mobile phase B was 0.1% formic acid in acetonitrile. The flow rate was set at 100 μl/min. The detector TOF scan was set from m/z 700-5000 for 25 minutes with major parameters as listed (Capillary voltage 3.2 kV; Sampling Cone 150; Source Offset at 80; Source temperatures 120° C.; Desolvation temperature 500° C.; Trap collision Energy 30; Transfer Collision Energy Off; Gas controls OFF; Resolving Quadrupole: LM resolution at 4.7). The combined spectra were deconvoluted with MaxEnt function within MassLynx software. The resulting molecular ions which when weighted according to intensities corresponded to the loadings listed in Table 3. The actual mass spec spectra are listed in FIGS. 42 and 43.
  • TABLE 3
    The summary of intensity-weighted average linker-payload loadings in
    PRLR-Q and Isotype Control-Q conjugates for compound 63.
    PRLR-Q-63 Isotype Control-Q-63
    Intensity Intensity
    Molecular Corresponding weighted Corresponding weighted
    Ion MW linker payload Relative average Molecular linker payload Relative average
    (Da) loading intensity loading Ion MW (DA) loading intensity loading
    3.3 146575 1 1648307 3.1
    146864 2 1424490 147719 2 9007543
    148010 3 4716164 148868 3 20406614
    149151 4 4658294 150008 4 18048784
  • Example 44
  • Equilibrium dissociation constants (KD values) for human PRLR binding to purified anti-PRLR antibodies that were either unmodified H1H6958N2, PRLR-Q, and PRLR-Q-63 were determined using a real-time surface plasmon resonance biosensor using a Biacore 3000 instrument. The Biacore sensor surface was first derivatized by amine coupling with a monoclonal mouse anti-human Fc antibody (GE, # BR-1008-39) to capture anti-PRLR monoclonal antibodies. All binding studies were performed in 0.01M HEPES pH 7.4, 0.15M NaCl, 3 mM EDTA, and 0.05% v/v Surfactant Tween-20 (HBS-ET running buffer) at 25° C. and 37° C. Different concentrations of human PRLR extracellular domain expressed with a C-terminal myc-myc-hexahistidine tag (hPRLR-MMH; SEQ ID NO: 401 as described in patent application WO 2015026907 A1), in HBS-ET running buffer (ranging from 40 nM to 3.33 nM) were injected over the anti-PRLR antibody captured surface for 4 minutes at a flow rate of 50 μL/minute and their dissociation in HBS-ET running buffer was monitored for 8 minutes. Kinetic association rate constant (ka) and dissociation rate constant (kd) were determined by fitting the real-time sensorgrams to a 1:1 binding model using Scrubber 2.0c curve fitting software. Binding dissociation equilibrium constants (KD) and dissociative half-lives (t½) were calculated from the kinetic rate constants as:
  • K D ( M ) = kd ka , and t 1 2 ( min ) = ln ( 2 ) 60 * kd
  • Binding kinetic parameters for hPRLR-MMH binding to different anti-PRLR antibodies at 25° C. are shown in Table 4. At 25° C., hPRLR-MMH bound to the parental antibody H1H6958N2 with a KD value of 1.09 nM. Human PRLR-MMH bound to the PRLR-Q with a KD value of 850 pM and to PRLR-Q-63 with a KD value of 1.50 nM.
  • TABLE 4
    Binding Kinetics parameters of anti-PRLR antibodies binding to hPRLR-MMH at
    25° C.
    100 nM
    mAb hPRLR-
    Capture MMH Bound ka kd KD t 1/2
    Antibody Level (RU) (RU) (1/Ms) (1/s) (M) (min)
    H1H6958N2 141.3 ± 0.8 38 4.17E+05 4.54E−04 1.09E−09 25
    PRLR-Q 148.5 ± 0.6 46 4.48E+05 3.79E−04 8.50E−10 30
    PRLR-Q-63 153.7 ± 0.7 45 3.80E+05 5.69E−04 1.50E−09 20
  • Example 45
  • The present example provides cytotoxicity assays for conjugates provided herein. To evaluate the ability of anti-PRLR antibodies conjugated with 63, 60, 77, and 78 to kill a PRLR expressing cell line, an in vitro cytotoxicity assay using a T47D ductal carcinoma line (ATCC, # HTB-133), which was previously determined to express >27,000 copies of human PRLR at its cell surface, was utilized.
  • For the assay, T47D cells were seeded onto white 96 well plates at 2,000 cells/well in media containing DMEM supplemented with 10% FBS, NEAA, and penicillin/streptomycin (complete media). They were grown overnight at 37° C. in 5% CO2. To determine cell viability curves, the following day antibody drug conjugates, unconjugated antibodies, or free payloads were added to the cells at final serial dilutions ranging from 100 nM to 0.01 nM in complete medium and then incubated for an additional 5 days. Luciferase activity was detected after the addition of CellTiter-Glo™ reagent (Promega, G7571) to each well, which contains reagents to lyse the remaining viable cells to release their ATP, ATPase inhibitors to prevent degradation of the ATP, as well as luciferin and luciferase to catalyse the luminescent reaction. Viable cells prior to addition of CellTiter-Glo will be the only source of ATP since the dead cells in culture will not synthesis ATP and any of their released ATP will be destroyed via endogenous ATPases. Relative light units (RLUs) were measured on a Victor luminometer (PerkinElmer) and the results were determined using a four-parameter logistic equation over a 10-point response curve (GraphPad Prism). All measured values and calculated IC50 values were corrected for payload equivalents. All IC50 values are expressed in nM concentration and percent kill is reported for the highest concentration tested. The results are summarized in FIGS. 45 through 48.
  • As shown in FIG. 44, the anti-PRLR antibody site-specifically conjugated with 63 (PRLR-Q-63) demonstrated cytotoxicity of T47D cells with an IC50 of 1.1 nM and a maximum percent killing of 55%. The free payload, 29, demonstrated cytotoxicity of T47D cells with an IC50 of 0.07 nM and a maximum percent killing of 67%. An isotype control antibody conjugated with 63 (ISOTYPE CONTROL-Q-63) did not demonstrate any killing of T47D cells, and the unconjugated anti-PRLR antibody (PRLR-Q) did not demonstrate any killing of T47D cells.
  • As shown in FIG. 45, the anti-PRLR antibody conjugated with 60 (PRLR-60) demonstrated cytotoxicity of T47D cells with an IC50 of 0.6 nM and a maximum percent killing of 60%. The free payload, 49, demonstrated cytotoxicity of T47D cells with an IC50 of 0.03 nM and a maximum percent killing of 69%. An isotype control antibody conjugated with 60 (ISOTYPE CONTROL-60) did not demonstrate any killing of T47D cells, and the unconjugated anti-PRLR antibody (PRLR) did not demonstrate any killing of T47D cells.
  • As shown in FIG. 46, the anti-PRLR antibody conjugated with 77 (PRLR-77) demonstrated cytotoxicity of T47D cells with an IC50 of 0.4 nM and a maximum percent killing of 69%. The free payload, 27, demonstrated cytotoxicity of T47D cells with an IC50 of 0.1 nM and a maximum percent killing of 67%. An isotype control antibody conjugated with 77 (ISOTYPE CONTROL-77) did not demonstrate any killing of T47D cells, and the unconjugated anti-PRLR antibody (PRLR) did not demonstrate any killing of T47D cells.
  • As shown in FIG. 47, the anti-PRLR antibody conjugated with 78 (PRLR-78) demonstrated cytotoxicity of T47D cells with an IC50 of 0.9 nM and a maximum percent killing of 61%. The free payload, 27, demonstrated cytotoxicity of T47D cells with an IC50 of 0.1 nM and a maximum percent killing of 67%. An isotype control antibody conjugated with 78 (ISOTYPE CONTROL-78) did not demonstrate any killing of T47D cells, and the unconjugated anti-PRLR antibody (PRLR) did not demonstrate any killing of T47D cells.
  • Table 5 lists the anti-proliferating ability of the payloads only in Ovcar3 (Muc16+), C4-2 (PSMA+), and T47D (PRLR+) cells.
  • TABLE 5
    T47D
    Compound C4-2 Ovcar3 IC50
    # IC50 (nM) % kill IC50 (nM) % kill (nM) % kill
    29 0.27 83 0.13 93 0.07 67
    33 0.45 86 0.20 93
    31 0.10 82 0.04 93
    27 0.20 84 0.09 92
    35 0.004 84 <0.01 93
    37 0.01 86 <0.01 94
    39 0.02 70
    41 0.03 72
    43 0.01 71
    45 3.73 72
    47 0.01 72
    49 0.06 70
    51 0.49 72
    53 0.2 72
    55 0.18 75
    65 1.13 72
    67 0.19 71
    69 0.39 73
    71 0.73 68
    73 0.57 73
    75
    86 0.73 90
    84 0.07 88
    90 >100 86
    80 1.3 91
    88 0.05 89
    82 19.95 93
  • The embodiments and examples described above are intended to be merely illustrative and non-limiting. Those skilled in the art will recognize or will be able to ascertain using no more than routine experimentation, numerous equivalents of specific compounds, materials and procedures. All such equivalents are considered to be within the scope and are encompassed by the appended claims.

Claims (22)

1-62. (canceled)
63. A compound of Formula (I):
Figure US20160375147A1-20161229-C00564
or a pharmaceutically acceptable salt thereof,
wherein:
A is arylene or heteroarylene;
L is a linker;
BA is a binding agent; and
k is an integer from 1 to 30.
64. The compound of claim 63, wherein
BA is an antibody or antigen binding fragment thereof; and
k is an integer from 1-6.
65. The compound of claim 64, wherein
Figure US20160375147A1-20161229-C00565
R1, independently at each occurrence, is alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, aralkyl, halo, haloalkyl, haloalkoxy, heteroaryl, heterocycloalkyl, hydroxyl, cyano, nitro,
Figure US20160375147A1-20161229-C00566
or azido,
RA is alkyl or heteroalkyl;
n is an integer from 0 to 4;
m is an integer from 0 to 3;
p is an integer from 0 to 6; and
q is an integer from 0 to 5.
66. The compound of claim 65, wherein
A is:
Figure US20160375147A1-20161229-C00567
R1 is halo, haloalkyl, alkoxy, alkyl,
Figure US20160375147A1-20161229-C00568
—OH, heterocycloalkyl, or
Figure US20160375147A1-20161229-C00569
and RA is alkyl.
67. The compound of claim 66, wherein A is
Figure US20160375147A1-20161229-C00570
68. The compound of claim 66, wherein A is:
Figure US20160375147A1-20161229-C00571
Figure US20160375147A1-20161229-C00572
is the bond linking A to the N atom, and
Figure US20160375147A1-20161229-C00573
is the bond linking A to the carbonyl.
69. The compound of claim 64, wherein
L is
Figure US20160375147A1-20161229-C00574
SP is a spacer;
Figure US20160375147A1-20161229-C00575
is one or more bonds to the binding agent;
RAA1 is an amino acid side chain; and
RAA2 is an amino acid side chain.
70. The compound of claim 69, wherein RAA1 is
Figure US20160375147A1-20161229-C00576
and RAA2 is
Figure US20160375147A1-20161229-C00577
71. The compound of claim 69, wherein
SP is
Figure US20160375147A1-20161229-C00578
is a bond to the antibody or antigen binding fragment thereof,
Figure US20160375147A1-20161229-C00579
is a bond to a cysteine on the antibody or antigen binding fragment thereof;
b is an integer from 2 to 8.
RN is a hydrogen atom or alkyl; and
RM is alkyl.
72. The compound of claim 64, wherein
L is
Figure US20160375147A1-20161229-C00580
wherein:
Figure US20160375147A1-20161229-C00581
is a bond to the antibody or antigen binding fragment thereof; and
Figure US20160375147A1-20161229-C00582
is a bond to a cysteine on the antibody or antigen binding fragment thereof.
73. The compound of claim 64, wherein the compound is
Figure US20160375147A1-20161229-C00583
Figure US20160375147A1-20161229-C00584
wherein t is an integer from 1 to 6;
Ab is an antibody or antigen binding fragment thereof;
S is a bond to a cysteine on said antibody or fragment thereof; and
N is a bond to a lysine on said antibody or fragment thereof.
74. The compound of claim 64, wherein the antibody, or antigen binding fragment thereof, binds PSMA, MUC16, or PRLR.
75. A pharmaceutical composition comprising the compound of claim 63 and a pharmaceutically acceptable diluent, excipient, or carrier.
76. A method for killing tumor cells comprising contacting the tumor cells with a compound of claim 63 or a pharmaceutical composition of claim 75.
77. A method for treating cancer in a patient comprising administering to a patient having said cancer a compound of claim 63 or a pharmaceutical composition of claim 75.
78. A compound of Formula II:
Figure US20160375147A1-20161229-C00585
or a pharmaceutically acceptable salt thereof,
wherein A is arylene or heteroarylene.
79. A pharmaceutical composition comprising the compound of claim 78 and a pharmaceutically acceptable diluent, excipient, or carrier.
80. A method for killing tumor cells comprising contacting the tumor cells with a compound of claim 78 or a pharmaceutical composition of claim 79.
81. A method for treating cancer in a patient having the cancer comprising administering a compound of claim 78 or a pharmaceutical composition of claim 79.
82. A compound of Formula P1:
Figure US20160375147A1-20161229-C00586
wherein A is arylene or heteroarylene and RL is a reactive linker.
83. A process for preparing a compound of Formula I:
Figure US20160375147A1-20161229-C00587
or a pharmaceutically acceptable salt thereof,
wherein:
A is arylene or heteroarylene;
L is a linker;
BA is a binding agent; and
k is an integer from 1 to 30
comprising contacting a compound of Formula P1
Figure US20160375147A1-20161229-C00588
wherein A is arylene or heteroarylene and RL is a reactive linker
with a binding agent.
US15/081,759 2015-03-27 2016-03-25 Maytansinoid derivatives, conjugates thereof, and methods of use Abandoned US20160375147A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/081,759 US20160375147A1 (en) 2015-03-27 2016-03-25 Maytansinoid derivatives, conjugates thereof, and methods of use
US16/712,941 US20200385402A1 (en) 2015-03-27 2019-12-12 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use
US17/528,144 US20220259225A1 (en) 2015-03-27 2021-11-16 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201562139044P 2015-03-27 2015-03-27
US201562252239P 2015-11-06 2015-11-06
US15/081,759 US20160375147A1 (en) 2015-03-27 2016-03-25 Maytansinoid derivatives, conjugates thereof, and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/712,941 Continuation US20200385402A1 (en) 2015-03-27 2019-12-12 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use

Publications (1)

Publication Number Publication Date
US20160375147A1 true US20160375147A1 (en) 2016-12-29

Family

ID=55650789

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/081,759 Abandoned US20160375147A1 (en) 2015-03-27 2016-03-25 Maytansinoid derivatives, conjugates thereof, and methods of use
US16/712,941 Abandoned US20200385402A1 (en) 2015-03-27 2019-12-12 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use
US17/528,144 Pending US20220259225A1 (en) 2015-03-27 2021-11-16 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/712,941 Abandoned US20200385402A1 (en) 2015-03-27 2019-12-12 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use
US17/528,144 Pending US20220259225A1 (en) 2015-03-27 2021-11-16 Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use

Country Status (17)

Country Link
US (3) US20160375147A1 (en)
EP (1) EP3273998B1 (en)
JP (2) JP6948950B2 (en)
KR (1) KR20170131587A (en)
CN (2) CN115645543A (en)
AU (1) AU2016243527B2 (en)
BR (1) BR112017020149A8 (en)
CA (1) CA2978340A1 (en)
CL (1) CL2017002422A1 (en)
CO (1) CO2017010890A2 (en)
EA (1) EA034950B1 (en)
IL (1) IL254267B (en)
MX (2) MX2017012380A (en)
PH (1) PH12017501780A1 (en)
SG (1) SG11201707148PA (en)
WO (1) WO2016160615A1 (en)
ZA (1) ZA201706040B (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018093866A1 (en) 2016-11-16 2018-05-24 Regeneron Pharmaceuticals, Inc. Anti-met antibodies, bispecific antigen binding molecules that bind met, and methods of use thereof
WO2018213077A1 (en) 2017-05-18 2018-11-22 Regeneron Pharmaceuticals, Inc. Cyclodextrin protein drug conjugates
WO2019094395A2 (en) 2017-11-07 2019-05-16 Regeneron Pharmaceuticals, Inc. Hydrophilic linkers for antibody drug conjugates
WO2019212965A1 (en) 2018-04-30 2019-11-07 Regeneron Pharmaceuticals, Inc. Antibodies, and bispecific antigen-binding molecules that bind her2 and/or aplp2, conjugates, and uses thereof
WO2019222663A1 (en) 2018-05-17 2019-11-21 Regeneron Pharmaceuticals, Inc. Anti-cd63 antibodies, conjugates, and uses thereof
WO2020172475A1 (en) 2019-02-21 2020-08-27 Regeneron Pharmaceuticals, Inc. Methods of treating ocular cancer using anti-met antibodies and bispecific antigen binding molecules that bind met
WO2021055895A1 (en) 2019-09-19 2021-03-25 Regeneron Pharmaceuticals, Inc. Anti-ptcra antibody-drug conjugates and uses thereof
WO2021097220A1 (en) 2019-11-15 2021-05-20 Seagen Inc. Methods of treating her2 positive breast cancer with tucatinib in combination with an anti-her2 antibody-drug conjugate
WO2021174113A1 (en) 2020-02-28 2021-09-02 Regeneron Pharmaceuticals, Inc. Bispecific antigen binding molecules that bind her2, and methods of use thereof
US11129903B2 (en) 2015-07-06 2021-09-28 Regeneron Pharmaceuticals, Inc. Multispecific antigen-binding molecules and uses thereof
WO2021211984A1 (en) 2020-04-16 2021-10-21 Regeneron Pharmaceuticals, Inc. Diels-alder conjugation methods
WO2022087243A1 (en) 2020-10-22 2022-04-28 Regeneron Pharmaceuticals, Inc. Anti-fgfr2 antibodies and methods of use thereof
WO2022103724A1 (en) 2020-11-10 2022-05-19 Regeneron Pharmaceuticals, Inc. Selenium antibody conjugates
US11345715B2 (en) 2013-03-15 2022-05-31 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US11352446B2 (en) 2016-04-28 2022-06-07 Regeneron Pharmaceuticals, Inc. Methods of making multispecific antigen-binding molecules
US11446389B2 (en) 2016-01-25 2022-09-20 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US11596635B2 (en) 2013-08-26 2023-03-07 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US11896682B2 (en) 2019-09-16 2024-02-13 Regeneron Pharmaceuticals, Inc. Radiolabeled MET binding proteins for immuno-PET imaging and methods of use thereof

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201707148PA (en) * 2015-03-27 2017-10-30 Regeneron Pharma Maytansinoid derivatives, conjugates thereof, and methods of use
DK3515487T3 (en) 2016-09-23 2023-08-28 Regeneron Pharma Bispecific anti-MUC16-CD3 antibodies and anti-MUC16 drug conjugates
EA201990781A9 (en) 2016-09-23 2019-11-27 ANTI-STEAP2 ANTIBODIES, CONJUGATES ANTIBODY MEDICINES AND BISSPECIFIC ANTIGEN BINDING MOLECULES THAT BIND STEAP2 AND CD3, AND THEIR APPLICATION
AU2017366873A1 (en) 2016-11-29 2019-06-13 Regeneron Pharmaceuticals, Inc. Methods of treating PRLR positive breast cancer
KR20200117988A (en) * 2017-11-30 2020-10-14 센추리온 바이오파마 코포레이션 Maytansinoid drug delivery system
WO2020057543A1 (en) * 2018-09-21 2020-03-26 中国人民解放军军事科学院军事医学研究院 Aryl nitro-based linker, linker-containing antibody-drug conjugate, and use of linker
CN113292574B (en) * 2020-02-21 2022-05-03 四川大学 Chiral polycyclic tropane compounds, preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4260608A (en) * 1978-11-14 1981-04-07 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and methods of use thereof
US20130029900A1 (en) * 2011-06-21 2013-01-31 Immunogen, Inc. Novel maytansinoid derivatives with peptide linker and conjugates thereof

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS55164686A (en) 1979-06-11 1980-12-22 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US7087411B2 (en) 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US20070258987A1 (en) 2000-11-28 2007-11-08 Seattle Genetics, Inc. Recombinant Anti-Cd30 Antibodies and Uses Thereof
AR048098A1 (en) 2004-03-15 2006-03-29 Wyeth Corp CALIQUEAMYCIN CONJUGATES
BRPI0510883B8 (en) 2004-06-01 2021-05-25 Genentech Inc drug-antibody conjugate compound, pharmaceutical composition, method of manufacturing a drug-antibody conjugate compound, and uses of a formulation, a drug-antibody conjugate and a chemotherapeutic agent, and a combination
CA2587589A1 (en) 2004-11-29 2006-06-22 Seattle Genetics, Inc. Engineered antibodies and immunoconjugates
US7989595B2 (en) 2005-06-20 2011-08-02 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
DK1912677T3 (en) 2005-06-20 2014-01-13 Psma Dev Company L L C PSMA antibody-drug conjugates
AU2006280146B2 (en) 2005-08-09 2012-06-28 Immunogen, Inc. Method of acylating maytansinol with chiral amino acids
MX2008008138A (en) 2005-12-22 2008-11-28 Baxter Int Improved monocyte activation test better able to detect non-endotoxin pyrogenic contaminants in medical products.
US7750116B1 (en) 2006-02-18 2010-07-06 Seattle Genetics, Inc. Antibody drug conjugate metabolites
NO347649B1 (en) 2006-12-14 2024-02-12 Regeneron Pharma Human antibody or antibody fragment that specifically binds human delta-like ligand 4 (hDII4), nucleic acid molecule that codes for such and vector and host-vector systems, as well as method for production, composition and use.
WO2008122039A2 (en) 2007-04-02 2008-10-09 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Selenocysteine mediated hybrid antibody molecules
CA2687178C (en) 2007-05-23 2014-02-04 Ventana Medical Systems, Inc. Polymeric carriers for immunohistochemistry and in situ hybridization
WO2009134976A1 (en) 2008-04-30 2009-11-05 Immunogen, Inc Potent conjugates and hydrophilic linkers
EP2326349B1 (en) 2008-07-21 2015-02-25 Polytherics Limited Novel reagents and method for conjugating biological molecules
JO3182B1 (en) 2009-07-29 2018-03-08 Regeneron Pharma High Affinity Human Antibodies to Human Angiopoietin-2
EP2464972A1 (en) 2009-08-10 2012-06-20 UCL Business PLC Reversible covalent linkage of functional molecules
TWI540136B (en) 2010-04-15 2016-07-01 梅迪繆思有限公司 Pyrrolobenzodiazepines and conjugates thereof
WO2012005982A2 (en) 2010-07-06 2012-01-12 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Reporter for rna polymerase ii termination
JP6146913B2 (en) 2010-08-02 2017-06-14 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Mice producing a binding protein containing a VL domain
ES2898854T3 (en) 2011-05-27 2022-03-09 Ambrx Inc Compositions Containing, Methods Involving, and Uses of Unnatural Amino Acid Linked Dolastatin Derivatives
US8815226B2 (en) 2011-06-10 2014-08-26 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
EP2751111B1 (en) 2011-10-14 2017-04-26 MedImmune Limited Asymmetrical bis-(5H-Pyrrolo[2,1-c][1,4]benzodiazepin-5-one) derivatives for the treatment of proliferative or autoimmune diseases
JP6399930B2 (en) 2011-10-14 2018-10-03 シアトル ジェネティクス,インコーポレーテッド Pyrrolobenzodiazepine and target conjugates
JP6393617B2 (en) 2011-10-14 2018-09-19 シアトル ジェネティクス,インコーポレーテッド Pyrrolobenzodiazepine and target conjugates
WO2013053872A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Synthesis method and intermediates useful in the preparation of pyrrolobenzodiazepines
WO2013068874A1 (en) 2011-11-11 2013-05-16 Pfizer Inc. Antibody-drug conjugates
CA2857398A1 (en) 2011-12-05 2013-06-13 Igenica Biotherapeutics, Inc. Antibody-drug conjugates and related compounds, compositions, and methods
CN110201186A (en) 2012-10-23 2019-09-06 西纳福克斯股份有限公司 Modified antibody, antibody-conjugate and preparation method thereof
CA2891280C (en) * 2012-11-24 2018-03-20 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
EP3964237A1 (en) * 2013-03-15 2022-03-09 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US11229711B2 (en) 2013-06-06 2022-01-25 Magenta Therapeutics, Inc. Linkers for antibody-drug conjugates and related compounds, compositions, and methods of use
TWI641620B (en) 2013-08-21 2018-11-21 再生元醫藥公司 Anti-prlr antibodies and uses thereof
MX2016002149A (en) * 2013-08-26 2016-10-28 Regeneron Pharma Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses.
SG11201707148PA (en) * 2015-03-27 2017-10-30 Regeneron Pharma Maytansinoid derivatives, conjugates thereof, and methods of use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4260608A (en) * 1978-11-14 1981-04-07 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and methods of use thereof
US20130029900A1 (en) * 2011-06-21 2013-01-31 Immunogen, Inc. Novel maytansinoid derivatives with peptide linker and conjugates thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Doronina, Svetlana O. et al, "Development of potent monoclonal antibody aurostatin conjugates for cancer therapy." Nat. Biotech. (2003) 21(7) p778-785 *
Dubowchik, Gene M. et al, "Cathepsin b-labile dipeptide linkers for lysosomal release of doxorubicin from internalizing immunoconjugates: model studies of enzymatic drug release and antigen specific in vitro anticancer activity." Bioconjugate Chem. (2002) 13 p855-869 *
Wolf, Philipp, "Anti-psma antibody-drug conjugates and immunotoxins." Chapter 15 of Antibody-drug conjugates and immunotoxins (2012) Gail Phillips (ed) ISBN 978-1-4614-5456-4 *

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11345715B2 (en) 2013-03-15 2022-05-31 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US11596635B2 (en) 2013-08-26 2023-03-07 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US11129903B2 (en) 2015-07-06 2021-09-28 Regeneron Pharmaceuticals, Inc. Multispecific antigen-binding molecules and uses thereof
US11191844B2 (en) 2015-07-06 2021-12-07 Regeneran Pharmaceuticals, Inc. Multispecific antigen-binding molecules and uses thereof
US11446389B2 (en) 2016-01-25 2022-09-20 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US11352446B2 (en) 2016-04-28 2022-06-07 Regeneron Pharmaceuticals, Inc. Methods of making multispecific antigen-binding molecules
US11142578B2 (en) 2016-11-16 2021-10-12 Regeneron Pharmaceuticals, Inc. Anti-MET antibodies, bispecific antigen binding molecules that bind MET, and methods of use thereof
WO2018093866A1 (en) 2016-11-16 2018-05-24 Regeneron Pharmaceuticals, Inc. Anti-met antibodies, bispecific antigen binding molecules that bind met, and methods of use thereof
WO2018213077A1 (en) 2017-05-18 2018-11-22 Regeneron Pharmaceuticals, Inc. Cyclodextrin protein drug conjugates
WO2019094395A2 (en) 2017-11-07 2019-05-16 Regeneron Pharmaceuticals, Inc. Hydrophilic linkers for antibody drug conjugates
WO2019212965A1 (en) 2018-04-30 2019-11-07 Regeneron Pharmaceuticals, Inc. Antibodies, and bispecific antigen-binding molecules that bind her2 and/or aplp2, conjugates, and uses thereof
WO2019222663A1 (en) 2018-05-17 2019-11-21 Regeneron Pharmaceuticals, Inc. Anti-cd63 antibodies, conjugates, and uses thereof
WO2020172475A1 (en) 2019-02-21 2020-08-27 Regeneron Pharmaceuticals, Inc. Methods of treating ocular cancer using anti-met antibodies and bispecific antigen binding molecules that bind met
US11896682B2 (en) 2019-09-16 2024-02-13 Regeneron Pharmaceuticals, Inc. Radiolabeled MET binding proteins for immuno-PET imaging and methods of use thereof
US11814428B2 (en) 2019-09-19 2023-11-14 Regeneron Pharmaceuticals, Inc. Anti-PTCRA antibody-drug conjugates and uses thereof
WO2021055895A1 (en) 2019-09-19 2021-03-25 Regeneron Pharmaceuticals, Inc. Anti-ptcra antibody-drug conjugates and uses thereof
WO2021097220A1 (en) 2019-11-15 2021-05-20 Seagen Inc. Methods of treating her2 positive breast cancer with tucatinib in combination with an anti-her2 antibody-drug conjugate
WO2021174113A1 (en) 2020-02-28 2021-09-02 Regeneron Pharmaceuticals, Inc. Bispecific antigen binding molecules that bind her2, and methods of use thereof
US11958910B2 (en) 2020-02-28 2024-04-16 Regeneron Pharmaceuticals, Inc. Bispecific antigen binding molecules that bind HER2, and methods of use thereof
WO2021211984A1 (en) 2020-04-16 2021-10-21 Regeneron Pharmaceuticals, Inc. Diels-alder conjugation methods
WO2022087243A1 (en) 2020-10-22 2022-04-28 Regeneron Pharmaceuticals, Inc. Anti-fgfr2 antibodies and methods of use thereof
US11866502B2 (en) 2020-10-22 2024-01-09 Regeneron Pharmaceuticals, Inc. Anti-FGFR2 antibodies and methods of use thereof
WO2022103724A1 (en) 2020-11-10 2022-05-19 Regeneron Pharmaceuticals, Inc. Selenium antibody conjugates

Also Published As

Publication number Publication date
CN107995912A (en) 2018-05-04
WO2016160615A1 (en) 2016-10-06
US20200385402A1 (en) 2020-12-10
BR112017020149A8 (en) 2023-05-02
CO2017010890A2 (en) 2020-04-13
IL254267B (en) 2021-03-25
EP3273998B1 (en) 2019-09-04
CN115645543A (en) 2023-01-31
MX2021008101A (en) 2021-08-05
JP6948950B2 (en) 2021-10-13
JP2018516851A (en) 2018-06-28
AU2016243527B2 (en) 2021-04-29
MX2017012380A (en) 2018-04-11
EA201700464A1 (en) 2018-11-30
IL254267A (en) 2018-06-28
EP3273998A1 (en) 2018-01-31
CA2978340A1 (en) 2016-10-06
BR112017020149A2 (en) 2018-09-11
KR20170131587A (en) 2017-11-29
CL2017002422A1 (en) 2018-03-16
ZA201706040B (en) 2020-05-27
SG11201707148PA (en) 2017-10-30
PH12017501780A1 (en) 2018-04-30
US20220259225A1 (en) 2022-08-18
AU2016243527A1 (en) 2017-09-21
EA034950B1 (en) 2020-04-09
JP2021113191A (en) 2021-08-05

Similar Documents

Publication Publication Date Title
US20220259225A1 (en) Maytansinoid Derivatives, Conjugates Thereof, and Methods of Use
US11446389B2 (en) Maytansinoid derivatives, conjugates thereof, and methods of use
US11352348B2 (en) Splicing modulator antibody-drug conjugates and methods of use
US11491237B2 (en) Cyclodextrin protein drug conjugates
CN110621673A (en) Double-stranded linked cytotoxic drug conjugates
US11717576B2 (en) Antibody-immune agonist conjugate and applications thereof
CN113423430A (en) Amanitoxin conjugates containing branched linkers
TW202345904A (en) Ligand-drug-conjugates with improved pharmacokinetic and drug release properties

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NITTOLI, THOMAS;REEL/FRAME:038870/0339

Effective date: 20160506

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION