WO2014134457A2 - Conjugates comprising cell-binding agents and cytotoxic agents - Google Patents

Conjugates comprising cell-binding agents and cytotoxic agents Download PDF

Info

Publication number
WO2014134457A2
WO2014134457A2 PCT/US2014/019434 US2014019434W WO2014134457A2 WO 2014134457 A2 WO2014134457 A2 WO 2014134457A2 US 2014019434 W US2014019434 W US 2014019434W WO 2014134457 A2 WO2014134457 A2 WO 2014134457A2
Authority
WO
WIPO (PCT)
Prior art keywords
ala
optionally substituted
methyl
val
independently
Prior art date
Application number
PCT/US2014/019434
Other languages
French (fr)
Other versions
WO2014134457A3 (en
Inventor
Wayne C. Widdison
Original Assignee
Immunogen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunogen, Inc. filed Critical Immunogen, Inc.
Publication of WO2014134457A2 publication Critical patent/WO2014134457A2/en
Publication of WO2014134457A3 publication Critical patent/WO2014134457A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • 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

Definitions

  • ADC Antibody-drug conjugates
  • cell binding agent-drug conjugates are emerging as a powerful class of anti-tumor agents with efficacy across a range of cancers.
  • Cell binding agent-drug conjugates are commonly composed of three distinct elements: a cell-binding agent (e.g., an antibody); a linker; and a cytotoxic moiety.
  • the linker component is an important element in developing targeted anti-cancer agents that possess an optimal therapeutic window, i.e., therapeutic activity at a low, non-toxic dose.
  • a first embodiment of the invention features a conjugate represented by the following formula, or a pharmaceutically acceptable salt thereof:
  • CBA is a cell binding agent
  • DM is a drug moiety represented by the following formula:
  • P is -(NR m -CH 2 CH 2 ) s -, in which s is an integer between 1 and 5 and R m is H, or alkyl optionally substituted with a charged substituent or an
  • s i is the site covalently linked to the CBA
  • s2 is the site covalently linked to the group X ⁇ in formula ( ⁇ ) or the group (CR 3 R 4 ) n in Formula (I), JCBI' in s1 e — I s2
  • Formula (V) is s2 R a Q9 N-NR e — s2 s1
  • -N-N R e -C( 0)— ⁇ s2 s1
  • N-NR e — Ar s2
  • Ar is an optionally substituted arylene or an optionally substituted heteroarylene
  • R a , R b , R c , R e , Ri to R 6 , and R 9 are independently H or an optionally substituted alkyl
  • Cy is the non- alkyne residue of an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne (see below in the second embodiment)
  • R201, R202 and R203 each are independently H or an optionally substituted alkyl (preferably, R201, R202 and R203 are all H;
  • R 2 oi is CH 3 and R202 and R203 are both H) ;
  • Z 3 is pyridyl or ? wherein R 2 o 4 and R205 are each independently optionally substituted alkyl (preferably, R2o 4 and R205 are both methyl or ethyl) and Cy' is the non-alkene residue of an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene (see below in the second embodiment); and R 30 i is H or optionally substituted alkyl (preferably R 0 i is H); m and n, for each occurrence, are independently an integer between 0 and 10; and w is an integer between 1 and 20.
  • X ⁇ and Z 2 are each independently independently optionally substituted alkyl (preferably, R2o 4 and R205 are both methyl or ethyl) and Cy' is the non-alkene residue of an optionally substituted strained cycloalkene or an optionally substituted strained hetero
  • a second embodiment of the invention features a cytotoxic compound represented by the following formula, or a salt e.g. , a pharmaceutically acceptable salt) thereof:
  • X' is a halogen
  • c is an optionally substituted cycloalkyne or an optionally
  • substituted heterocycloalkyne an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene, and the remainder of the variables are as defined in the first embodiment.
  • an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne that can readily react with an azide to form a triazole through copper free click chemistry or can readily react with a tetrazine (see, for example, J. Am. Chem. Soc. (2012) 134:9199-9208; WO 2011/136645; US 2009/0068738, Lang K. et al, J. Am. Chem. Soc. (2012) 134: 10317- 10320, the entire teaching of these references are incorporated herein b its entirety). More preferably,
  • a third embodiment of the invention features a linker compound represented by the following formula, or a salt (e.g. , a pharmaceutically acceptable salt) thereof:
  • Ar is phenylene
  • p or p' is an integer from 2 to 20, more preferably, 2 to 14, even more preferably, 2 to 8. Most preferably, p or p' is 2, 4, 6 or 8.
  • R 9 is H.
  • composition comprising a conjugate represented by Formula ( ⁇ ), (I) or (V), a cytotoxic compound represented by Formula ( ⁇ ), (III) or (VI), or a pharmaceutical acceptable salt thereof.
  • the composition may also include a carrier (e.g. , a pharmaceutically acceptable carrier).
  • the composition can further include a second therapeutic agent.
  • the present invention also includes a method of inhibiting abnormal cell growth or treating a proliferative disorder, a destructive bone disorder, an autoimmune disorder, a graft versus host disease, a transplant rejection, an immune deficiency, an inflammatory diseases, an infectious disease, a viral disease, a fibrotic disease, a neurodegenerative disorder, pancreatitis, or a kidney disease in a mammal (e.g. , human), comprising administering to said mammal a therapeutically effective amount of a conjugate represented by Formula ( ⁇ ), (I) or (V), a cytotoxic compound represented by Formula ( ⁇ ), (III) or (VI), or a pharmaceutical acceptable salt thereof.
  • a conjugate represented by Formula ( ⁇ ), (I) or (V) a cytotoxic compound represented by Formula ( ⁇ ), (III) or (VI)
  • the method described above further comprises
  • Figures 1-8 depict synthetic schemes for preparing representative linker compounds or cytotoxic compounds of the present invention.
  • Alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twenty carbon atoms. "Monovalent” means that alkyl has one point of attachment to the remainder of the molecule. Examples of alkyl groups include, but are not limited to, methyl, ethyl, 1 -propyl, 2-propyl, 1 -butyl, 2-methyl-l -propyl, - CH 2 CH(CH 3 )2, 2-butyl, 2-methyl-2-propyl, 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3- methyl-2-butyl, 3-methyl-l -butyl, 2-methyl-l -butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2-methyl-3-penty
  • Alkylene refers to a saturated linear or branched-chain divalent hydrocarbon radical of one to twenty carbon atoms, examples of which include, but are not limited to, those having the same core structures of the alkyl groups as exemplified above. "Divalent” means that the alkylene has two points of attachment to the remainder of the molecule. Preferably, the alkylene group has one to ten carbon atoms. More preferably, the alkylene group has one to four carbon atoms.
  • an integer “between” x and y includes integers x and y unless otherwise specified to the contrary.
  • an integer between 1 and 5" can be 1, 2, 3, 4, or 5.
  • cyclic alkyl and “cycloalkyl” can be used interchangeably. They refer to a monovalent saturated carbocyclic ring radical. "Monovalent” means that cycloalkyl has one point of attachment to the remainder of the molecule. Preferably, the cycloalkyl is 3 to 7 membered monocyclic ring radical.
  • Bicyclic cycloalkyl having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6], or [6,6] system, and bicyclic cycloalkyl having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane.
  • Bicyclic cycloalkyl also include a spiro cycloalkyl with rings connected through just one atom.
  • Examples of monocyclic cycloalkyl groups include, but are not limited to,
  • Cycloalkylene refers to a divalent saturated carbocyclic ring radical having 3 to 12 carbon atoms as a monocyclic ring, or 7 to 12 carbon atoms as a bicyclic ring.
  • cycloalkylene has two points of attachment to the remainder of the molecule.
  • the cycloalkylene is a 3- to 7-membered monocyclic.
  • cyclic alkylene groups include, but not limited to, those having the same core structures of the cylcolakyl groups as exemplified above. More preferably, the cycloalkylene group is cyclohexylene.
  • Cycloalkyne refers carbocyclic ring having one or more triple bonds. It can be monocyclic, bicyclic or tricyclic; bicyclic and tricyclic can be bridged or fused.
  • the carbocyclic ring optionally contains one or more double bonds and/or is optionally fused with one or more aromatic (e.g., phenyl ring) or heteroaromatic rings.
  • cycloalkyne examples include, but are not limited to, those described in J. Am. Chem. Soc. (2012) 134:9199-9208; WO 2011/136645, US 2009/0068738, Lang K. et al. J. Am. Chem. Soc.
  • cycloalkyne is cyclooctyne.
  • Heterocycloalkyne refers to a heterocyclic ring having one or more triple bonds. Examples of heterocycloalkyne include, but are not limited to,
  • DIBAC dibenzoazacyclooctyne
  • BARAC biarylazacyclooctynone
  • thiacyclooctyne thiabenzocyclooctyne
  • thiacycloheptyne tetramethylthiacycloheptyne
  • relaxed cycloalkene refers to carbocyclic ring having one or more double bonds, in which at least one of the double bonds forced by structural constraints has bond angles other than the typical 120° angle of non-strained alkenes.
  • the strained cycloalkenes are more reactive than non-strained alkenes. Examples of strained cycloalkene include, but are not limited to, cyclooctene, norbornene and other cycloalkenes described in Debets, M. F. et al, Acc. Chem. Res., (2011) 44(9):805-815.
  • “Strained heterocycloalkene” as used herein refers to a heterocyclic ring having one or more double bonds, in which at least one of the double bonds forced by structural constraints has bond angles other than the typical 120° angle of non-strained heterocycloalkenes.
  • the strained heterocycloalkenes are more reactive than non-strained heterocycloalkenes.
  • aryl group means an aromatic hydrocarbon ring system having six to fourteen carbon ring atoms.
  • aryl may be used interchangeably with the terms “aryl ring” “aromatic ring,” “aryl group” and “aromatic group”.
  • An aryl group typically has six to fourteen ring atoms.
  • Aryl group also includes an aromatic hydrocarbon ring system fused to a non-aromatic carbocyclic ring system, such as a cycloalkyl group.
  • Examples includes phenyl, naphthyl, anthracenyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, fluorenyl, indanyl, indenyl and the like.
  • An aryl group is monovalent, i.e., has one point of attachment to the remainder of the molecule.
  • a "substituted aryl group” is substituted at any one or more substitutable ring atom, which is a ring carbon atom bonded to a hydrogen.
  • “Arylene” as used herein refers to a divalent aryl group, i.e., an aryl group having two points of attachment to the remainder of the molecule.
  • Divalent means that the arylene has two points of attachment to the remainder of the molecule. Both aryl and arylene groups are sometime represented herein by "Ar.”
  • Arylene is preferably phenylene.
  • Heteroaryl used interchangeably with “heteroaromatic,” “heteroaryl ring,”
  • heteroaryl group refers to aromatic ring systems having five to fourteen ring atoms selected from carbon and at least one
  • heteroatoms typically 1 to 4, more typically 1 or 2 heteroatoms (e.g. , oxygen, nitrogen or sulfur).
  • Heteroaryl includes monocyclic rings and polycyclic rings (e.g. , bicyclic) in which a monocyclic heteroaromatic ring is fused to one or more other aromatic or heteroaromatic rings.
  • “5-14 membered heteroaryl” includes monocyclic, bicyclic or tricyclic ring systems. Heteroaryls are monovalent, meaning that there is one point of attachment to the remainder of the molecule.
  • “Monocyclic 5-6 membered heteroaryl” means a monocyclic aromatic ring system having five or six ring atoms selected from carbon and at least one (typically 1 to 3, more typically 1 or 2) heteroatoms (e.g. , oxygen, nitrogen or sulfur).
  • monocyclic 5-6 membered heteroaryl groups include furanyl (e.g. , 2-furanyl, 3-furanyl), imidazolyl (e.g. , N- imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl), isoxazolyl (e.g.
  • 3-isoxazolyl 4- isoxazolyl, 5-isoxazolyl), oxadiazolyl (e.g. , 2-oxadiazolyl, 5-oxadiazolyl), oxazolyl (e.g. , 2- oxazolyl, 4-oxazolyl, 5-oxazolyl), pyrazolyl (e.g. , 3-pyrazolyl, 4-pyrazolyl), pyrrolyl (e.g. , 1- pyrrolyl, 2-pyrrolyl, 3-pyrrolyl), pyridyl (e.g. , 2-pyridyl, 3-pyridyl, 4-pyridyl), pyrimidinyl
  • thiazolyl e.g. , 2-thiazolyl, 4-thiazolyl, 5-thiazolyl
  • isothiazolyl triazolyl (e.g. , 2-triazolyl, 5-triazolyl), tetrazolyl (e.g. , tetrazolyl), and thienyl (e.g. , 2-thienyl, 3-thienyl).
  • polycyclic aromatic heteroaryl groups examples include carbazolyl, benzimidazolyl, benzothienyl, benzofuranyl, isobenzofuranyl, indolyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, indazolyl, isoindolyl, acridinyl, or benzisoxazolyl.
  • a "substituted heteroaryl group” is substituted at any one or more substitutable ring atom, which is a ring carbon or ring nitrogen atom bonded to a hydrogen.
  • Heteroarylene refers to a divalent heteroaryl, i.e., a heteroaryl with two points of attachment to the remainder of the molecule.
  • heterocycle refers to a saturated or unsaturated non-aromatic 3-12 membered ring radical optionally containing one or more double bonds. It can be
  • heterocycle contains 1 to 4 heteroatoms, which may be the same or different, selected from N, O or S.
  • the heterocycle optionally contains one or more double bonds and/or is optionally fused with one or more aromatic (e.g. , phenyl ring) or heteroaromatic rings.
  • 3-7 membered monocyclic heterocycle means a radical having from 3-7 atoms (including 1-3 heteroatoms) arranged in a monocyclic ring.
  • heterocycle is intended to include all the possible isomeric forms.
  • a heterocycle may be a monocycle having 3 to 7 ring members (e.g.
  • heterocyclic rings include, but are not limited to, aziridinyl, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, isoindolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,
  • Spiro moieties are also included within the scope of this definition.
  • the heterocycle, heteroaryl, or heteroarylene groups may be carbon (carbon-linked) or nitrogen (nitrogen-linked) attached where such is possible.
  • carbon bonded heterocycle, heteroaryl or heroarylene groups are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position
  • nitrogen bonded heterocycle, heteroaryl, or heteroarylene groups are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, IH-indazole, position 2 of a isoindole or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or O-carboline.
  • heteroatoms present in heteroaryl, heteroarylene, or heterocyclcyl can include the oxidized forms such as NO, SO, and S0 2 .
  • Halogen refers to F, CI, Br or I.
  • a group is described as being “optionally substituted,” the group may be either (1) not substituted, or (2) substituted. If a carbon of a group is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogen atoms on the carbon (to the extent there are any) may separately and/or together be replaced with an independently selected optional substituent.
  • Suitable substituents for an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, heterocycle, alkylene, alkenylene, alkynylene, cycloalkene, heterocycloalkene, cycloalkyne, heterocycloalkyne, cycloalkylene arylene, and heterarylene are those which do not significantly adversely affect the biological activity of the conjugate.
  • the substituent for the optionally substituted alkyl, alkylene, cycloalkylene, arylene, and heteroarylene described above is selected from the group consisting of halogen, -CN, - NR 10 iRio2, -CF 3 , -OR 100 , aryl, heteroaryl, heterocyclyl, -SR 101 , -SOR 101 , -SO 2 R 101 , and -S0 3 M.
  • the suitable substituent is selected from the group consisting of - halogen, -OH, -N0 2 , -CN, Ci_ 4 alkyl, -OR100, NRioiRio 2 , -NRioiCORi 02 , -SR100, -SO2R101, - S0 2 NR 10 iRio 2 , -CORioi, -OCOR 10 i, and -OCONR 10 iRio 2 , wherein R 100 , R101, and R 102 are each independently -H or C 1-4 alkyl.
  • alkenyl refers to aliphatic linear or branched-chain monovalent hydrocarbon radical of two to twenty carbon atoms with at least one carbon-carbon double bond, wherein the alkenyl radical includes radicals having "cis” and “trans” orientations, or by an alternative nomenclature, "E” and “Z” orientations.
  • the alkenyl has two to ten carbon atoms, also referred to as "C 2-1 o alkenyl.” More preferably, the alkyl has two to four carbon atoms, also referred to as “C 2 _ 4 alkenyl.”
  • Alkynyl refers to aliphatic linear or branched-chain monovalent hydrocarbon radical of two to twenty carbon atoms with at least one carbon-carbon triple bond. "Monovalent” means that alkynyl has one point of attachment to the remainder of the molecule. Examples include, but are not limited to ethynyl, propynyl, 1-butynyl, 2-butynyl,
  • the alkynyl has two to ten carbon atoms, also referred to as "C 2 -io alkynyl.” More preferably, the alkynyl has two to four carbon atoms, also referred to as "C 2 _ 4 alkynyl.”
  • ionizable group refers to a functional group that can be converted to a charged group by protonation with an acid or deprotonation with a base.
  • ionizable groups include -S0 3 H, -Z'-S0 3 H, -OP0 3 H 2 , -Z'-OP0 3 H 2 , -P0 3 H 2 , -Z'-P0 3 H 2 , - C0 2 H, -Z'C0 2 H, -NR11R12, or -Z'-NR u Ri 2 , R11 and R 12 , for each occurrence, are independently H or an optionally substituted alkyl; and Z' includes an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene. In certain embodiments, Z' is alkylene.
  • charged substituent refers to a substituent that is either positively or negatively charged.
  • the charge in such a substituent is not removable by treatment with a base or an acid and thus permanent.
  • the charged substituents include, but not limited to, -N + R 13 R 14 R 15 and -Z'-N + R 13 R 14 R 15 , in which R 1 to R 15 are each independently an optionally substituted alkyl; and Z' includes an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene. In certain embodiments, Z' is alkylene.
  • Charged substituents may contain a counterion.
  • the counterion is negative and can be represented by "X " , e.g. , as -N + R 13 R 14 Ri 5 X ⁇ and -Z'-
  • anions include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, bromide, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, pamoate, pantothenate,
  • the counter ions for the positively charged substituents are chloride, bromide, sulfate, and phosphate.
  • the counter ions for the negatively charged substituents include, but are not limited to, an alkali metal ion (e.g. , sodium and potassium), an alkaline earth metal ion (e.g. , calcium and magnesium), aluminum ion, ammonium, protonated trialkyl amines (e.g. , trimethylamine and triethylamine), a tetraalkyl ammonium (e.g. , tetra methyl ammonium, and tetrabutyl ammonium), and a protonated heteroaromatic group (e.g. , pyridine, pyrimidine, triazines, tetrazines).
  • an alkali metal ion e.g. , sodium and potassium
  • an alkaline earth metal ion e.g. , calcium and magnesium
  • aluminum ion ion
  • ammonium e.g. , ammonium, protonated trialkyl amines (e.g.
  • the counter ions for the negatively charged substituents are sodium, potassium, lithium, protonated triethyl amine, protonated pyridiene. Most preferably, the counter ions for the negatively charged substituents are sodium and potassium. The counter ions for both the negatively and positively charged substituents may be removed or replaced in subsequent purification steps.
  • amino acid as used herein, including the residue represented by variable "XX" described above, refers to naturally occurring amino acids, unnatural amino acids, amino acid analogs, or amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids refers to those twenty L-amino acids encoded by the universal genetic codes and appearing in proteins or peptides, as well as selenocysteine and pyrrolysine that are incorporated into proteins by distinctive biosynthetic mechanisms. They include Histidine, Alanine, Isoleucine, Arginine, Leucine, Asparagine, Lysine, Aspartic acid, Methionine, Cysteine, Phenylalanine, Glutamic acid, Threonine, Glutamine, Tryptophan, Glycine, Valine, Proline, Serine, Tyrosine,
  • Naturally occurring amino acids also refers to those produced by the body, but are not encoded by the universal genetic codes, such as ⁇ - Alanine, Ornithine, and citrulline.
  • the term “Naturally occurring amino acids” further includes those naturally occurring L-amino acids that are later modified (e.g. , via post- translational modification by enzymes) in the body, such as hydroxyproline, ⁇ - carboxyglutamate, and O-phospho serine.
  • unnatural amino acids as used herein is intended to include the "D" stereochemical form of the naturally occurring amino acids described above. It is further understood that the term “unnatural amino acids” includes homologues of the natural L- amino acids or their D isomers, and synthetically modified forms of the natural L-amino acids or their D isomers.
  • the synthetically modified forms include, but are not limited to, amino acids having side chains shortened or lengthened by up to two carbon atoms, amino acids comprising optionally substituted aryl groups, and amino acids comprised halogenated groups, preferably halogenated alkyl and aryl groups and also N substituted amino acids e.g.
  • N-methyl-Histidine N-methyl-Alanine, N-methyl-Isoleucine, N-methyl-Arginine, N-methyl-
  • amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group.
  • amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but functions in a manner similar to a naturally occurring amino acid.
  • Peptide as used herein, including the peptide represented by variable "P” when the variable is [XX] 2 _ 10 , refers to short polymers of amino acid monomers linked by peptide bonds, the covalent chemical bonds formed between two amino acid monomers when the carboxyl group of N-terminal monomer reacts with the amino group of the C-terminal monomer.
  • the preferred length of the peptide is two to ten amino acids as described above.
  • any one of the peptides described herein can be connected to the rest of the molecules in either direction.
  • AA1, AA2, and AA3 each represent an amino acid (naturally-occurring, or unnatural)
  • AA1-AA2-AA3 in either direction refers to the tripeptide N-AA1-AA2-AA3-C), and the tripeptide N-AA3-AA2-AA1-C).
  • variable P is an amino acid represented by [XX] ⁇ or a peptide represented by [XX] 2-1 o, in which each XX is the residue of an independently selected amino acid selected from the group consisting of: Histidine, Alanine, Isoleucine, Arginine, Leucine,
  • Threonine Glutamine, Tryptophan, Glycine, Valine, Proline, Serine, Tyrosine,
  • N-methyl-Aspartic acid N-methyl-Methionine, N-methyl-Cysteine, N-methyl-Phenylalanine,
  • N-methyl-Glutamic acid N-methyl-Threonine, N-methyl-Glutamine, N-methyl-Tryptophan, N-methyl-Glycine, N-methyl- Valine, N-methyl-Proline, N-methyl-Serine, N-methyl- Tyrosine, N-methyl-selenocysteine, N-methyl-pyrrolysine, hydroxyproline, ⁇ - carboxyglutamate, selinocysteine, O-phosphoserine, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, citrulline, Ornithine, cysteine sulfonic acid, cysteine sulfinic acid, 3-aminoalanine, 3-dimethylaminoalanine, 2-amino-4-(dimethylamino)butanoic acid, 2,4-diaminobutanoic acid, 2-amino-6-(dimethylamino)
  • the peptide represented by variable "P" is cleavable by a protease.
  • the protease is a protease expressed in tumor tissue.
  • the protease is a lysosomal protease.
  • protease cleavable by a protease refers to peptides containing a cleavage recognition sequence of a protease.
  • a cleavage recognition sequence for a protease is an amino acid sequence recognized by the protease during proteolytic cleavage.
  • Many protease cleavage sites are known in the art, and these and other cleavage sites can be included in the linker moiety. See, e.g. , Matayoshi et al. Science 247:954 (1990); Dunn et al. Meth. Enzymol. 241 :254 (1994); Seidah et al. Meth. Enzymol.
  • the peptide sequence is chosen based on its ability to be cleaved by a tumor- associated protease, e.g. , a protease that is found on the surface of a cancerous cell or extracellularly in the vicinity of tumor cells.
  • a tumor- associated protease e.g. , a protease that is found on the surface of a cancerous cell or extracellularly in the vicinity of tumor cells.
  • proteases include thimet oligopeptidase (TOP), CD10 (neprilysin), a matrix metalloprotease (such as MMP2 or MMP9), a type II transmebrane serine protease (such as Hepsin, testisin, TMPRSS4 or matriptase/MT-SPl), legumain and enzymes described in the following reference: Current Topics in Developmental Biology: Cell Surface Proteases, vol. 54 Zucker S . 2003, Boston, MA. The ability of a peptide to be cleaved by tumor-associated protease can be tested using in vitro protease cleavage assays known in the art.
  • the peptide sequence is chosen based on its ability to be cleaved by a lysosomal protease, which include cathepsins B, C, D, H, L and S, and furin.
  • a lysosomal protease which include cathepsins B, C, D, H, L and S, and furin.
  • the peptide sequence is capable of being cleaved by an appropriate isolated protease in vitro, which can be tested using in vitro protease cleavage assays known in the art.
  • the peptide is selected from the group consisting of Val-Cit, Val-Lys, Phe-Lys, Lys-Lys, Ala-Lys, Phe-Cit, Leu-Cit, Lle-Cit, Trp, Cit, Phe-Ala, Phe-N 9 - tosyl-Arg, Phe-N 9 -nitro-Arg, Phe-Phe-Lys, D-Phe-Phe-Lys, Gly-Phe-Lys, Leu-Ala-Leu, Ile- Ala-Leu, Val-Ala-Val, Ala-Leu- Ala-Leu, ⁇ -Ala-Leu- Ala-Leu, Gly-Phe-Leu-Gly, Val-Arg, Arg-Val, Arg-Arg, Val-D-Cit, Val-D-Lys, Val-D-Arg, D- Val-Cit, D-Val-Lys, D-Val, D-Val-Ly
  • the peptide is selected from the group consisting of Gly-Gly-Gly, Ala- Ala- Ala, D- Ala- Ala- Ala, Ala-D- Ala- Ala, and Ala-Val-Ala.
  • any of the peptide sequences herein above may be in either direction, as defined above.
  • An amino acid or peptide can be attached to a linker/spacer or a cell binding agent through the terminal amine or terminal carboxylic acid of the amino acid or peptide.
  • the amino acid can also be attached to a linker/spacer or a cell-binding agent through a side chain reactive group, including, but not restricted to, the thiol group of cysteine, the epsilon amine of lysine, and the side chain hydroxyls of serine or threonine.
  • a blocking group is an atom or a chemical moiety that protects an amino acid or a peptide (e.g., the N-terminus) from undesired reactions and can be used during the synthesis of a drug-ligand conjugate. It should remain attached to the amino acid or peptide (e.g., to the N-terminus) throughout the synthesis, and may be removed after completion of synthesis of the drug conjugate by chemical or other conditions that selectively achieve its removal.
  • the blocking groups suitable for N-terminus protection are well known in the art of peptide chemistry. Exemplary blocking groups include, but are not limited to, methyl esters, tert-butyl esters, 9- fluorenylmethyl carbamate (Fmoc) and carbobenzoxy (Cbz).
  • blocking group or “protecting group” refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound, a derivative thereof, or a conjugate thereof.
  • an "amine-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound.
  • Such groups are well known in the art (see for example P. Wuts and T. Greene, 2007, Protective Groups in Organic Synthesis, Chapter 7, J.
  • carbamates such as methyl and ethyl carbamate, FMOC, substituted ethyl carbamates, carbamates cleaved by 1,6- ⁇ - elimination (also termed "self immolative"), ureas, amides, peptides, alkyl and aryl derivatives.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t- butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethylenoxycarbonyl (Fmoc).
  • reactive ester refers to an ester group having a leaving group that is readily displaced by an amine group.
  • examples of a reactive ester include, but are not limited to, N-hydroxysuccinimide ester, N-hydroxysulfosuccinimide ester, nitrophenyl ⁇ e.g., 2 or 4-nitrophenyl) ester, dinitrophenyl ⁇ e.g., 2,4-dinitrophenyl) ester, sulfo- tetrafluorophenyl ⁇ e.g., 4-sulfo-2,3,5,6-tetrafluorophenyl)ester and pentafluorophenyl ester.
  • DARPin and "(designed) ankyrin repeat protein” are used interchangeably to refer to certain genetically engineered antibody mimetic proteins typically exhibiting preferential (sometimes specific) target binding.
  • the target may be protein, carbohydrate, or other chemical entities, and the binding affinity can be quite high.
  • the DARPins may be derived from natural ankyrin repeat-containing proteins, and preferably consist of at least three, usually four or five ankyrin repeat motifs (typically about 33 residues in each ankyrin repeat motif) of these proteins.
  • a DARPin contains about four- or five-repeats, and may have a molecular mass of about 14 or 18 kDa, respectively.
  • Libraries of DARPins with randomized potential target interaction residues with diversities of over 10 12 variants can be generated at the DNA level, for use in selecting
  • DARPins that bind desired targets ⁇ e.g., acting as receptor agonists or antagonists, inverse agonists, enzyme inhibitors, or simple target protein binders) with picomolar affinity and specificity, using a variety of technologies such as ribosome display or signal recognition particle (SRP) phage display.
  • SRP signal recognition particle
  • AVIBODY cell binding agents includes a family of proteins as cell binding agents that specifically bind desired targets. As is well known, antibodies bind such desired targets through "Target Binding Regions” (TBRs) or Fv domains. AVIBODYTM CBA typically contains two, three, or four TBRs more commonly known as Dia-, Tria- and Tetra-bodies.
  • TBRs / Fv domains are linked together by fusing the Fv V-domains together in a "head to tail” orientation, forming stable, specific, and highly customizable multimeric antibody-like proteins as AVIBODYTM CBA. See, for example, U.S. Publication Nos. 20080152586 and 20120171115 for details, the entire teachings of which are incorporated herein by reference.
  • cell binding agent refers to a compound that can bind a cell (e.g., on a cell-surface ligand) or bind a ligand associated with or proximate to the cell, either in a specific or non-specific manner. In certain embodiments, binding to the cell or a ligand on or near the cell is specific.
  • the cell-binding agent may be of any kind presently known, or that become known and includes peptides and non-pep tides.
  • the cell-binding agents are proteins or polypeptides, or compounds comprising proteins or polypeptides, including both antibody and non-antibody proteins or polypeptides.
  • the cell-binding agents e.g., proteins or polypeptides
  • the cell-binding agents comprise one or more Cys residues.
  • the side chain -SH group of the Cys residues may be intact, or may be in a disulfide bond that can be reduced.
  • reduction of the disulfide bond(s) does not significantly negatively impact the cell-binding function of the proteins or polypeptides (e.g., in the case of antibody or antigen-binding portion thereof, reduction of the disulfide bonds does not substantially increase the dissociation of light chains / heavy chains).
  • the cell-binding agents e.g., proteins or polypeptides
  • the cell-binding agents comprise one or more amino acids that are modified to contain a reactive functional group that can react with the linkers (e.g., Formula (IV)) or cytotoxic compounds
  • the reactive functional group can be introduced into the cell-binding agent through any chemical or enzymatic method known in the art. See, for example, Davis L.K. et al,. J. Am. Chem. Soc. (2012) 134: 10317- 10320; Boeggeman E. et al,. Bioconjug Chem.
  • Cell-binding agent can also peptides derived from phage display (see, for example,
  • one or more reactive functional groups that are capable of reacting with the linkers (Formula (IV)) or the cytotoxic compounds (Formula (III)) of the present invention can be introduced into the cell-binding agent by any methods known in the art.
  • a terminal amine group on the cell-binding agent can be converted to a carbonyl group through transamination reaction (see, for example, US 2010/0099649; Angew.
  • the cell-binding agent can be engineered to include one or more free cysteine residues (i.e., cysteine residues having a free -SH group that can react with the linkers or the cytotoxic compounds of the present invention) according to any methods known in the art (see, for example, US 7,521,541).
  • thiol groups can be generated by controlled reduction of interchain disulfides of antibodies, followed by treatment with a cytotoxic agent bearing a maleimido group, as described in US patents 7,659,241, 8,309,300; 7,855,275; 7,723,485 and 7,521,541.
  • conjugates having 2 For example, partial or complete reduction of interchain disulfides followed by conjugation with a cytotoxic agent bearing a maleimido group can yield a conjugate with 2, 3, 4, 5, 6, 7 or 8 cytotoxic agent molecules covalently linked to each antibody molecule.
  • conjugates having 2 For example, partial or complete reduction of interchain disulfides followed by conjugation with a cytotoxic agent bearing a maleimido group can yield a conjugate with 2, 3, 4, 5, 6, 7 or 8 cytotoxic agent molecules covalently linked to each antibody molecule.
  • conjugates having 2 having 2
  • cytotoxic agent molecules covalently attached to each antibody molecule can be prepared by complete reduction of interchain disulfide of the antibody followed by partial re-oxidation and then conjugation with cytotoxic agent.
  • these conjugates can be prepared by partial or complete reduction of interchain disulfides of the antibody followed by conjugation with cytotoxic agent and then partial re- oxidation.
  • Thiol groups can also be introduced into the cell-binding agent (e.g. , antibodies) by reaction with a crosslinking agent such as 2-iminothiolane (see for example Goff and Carroll, Bioconjugate Chem.
  • cytotoxic agent bearing a maleimido group e.g. , compounds of Formula (III)
  • cytotoxic agent bearing a maleimido group e.g. , compounds of Formula (III)
  • All these methods for introducing reactive functional groups are applicable for cell-binding agents that are not antibodies, which, for example, include centyrin, Darpin, Avibody, adnectin or antibody fragment, such as minibodies, diabodies, tribodies, tetrabodies, nanobodies, probodies, domain bodies or unibodies.
  • one or more reactive functional groups e.g. , a cysteine having a free thiol group
  • a cysteine having a free thiol group can be introduced according to methods described in US 2010/0255056, US 2010/0216708 and US 2011/0274623.
  • the cell-binding agent is a Darpin and it can be prepared according to methods described in US Publication Nos. 2004/0132028, 2009/0082274, 2011/0118146, and 2011/0224100, WO 02/20565 and WO 06/083275.
  • Darpin comprises one or more cysteine residues at specific positions that do not interfere with antigen binding.
  • cysteine residue can react with the linkers (e.g. , Formula (IV)) or the cytotoxic compounds (e.g. , Formula (III)) of the present invention.
  • Avibodies having one or more cysteine residues can be prepared according to methods described in US 2008/0139791 and US 2012/0171115.
  • the Cys side chain -SH groups or the other reactive functional groups described above may be covalently linked to the linkers (e.g. , linkers of Formula (IV)), which are in turn linked to the cytotoxic compounds, thus conjugating the cell-binding agents to the cytotoxic compounds to yield the conjugates of the invention (e.g. , conjugates of Formula (I)).
  • the Cys side chain -SH groups or the reactive functional groups may be covalently linked to the cytotoxic compounds of the invention having linkers bound thereto (e.g. , cytotoxic compounds of Formula (III)).
  • Each protein-based cell-binding agents may contain multiple Cys side chain -SH groups and/or the reactive functional groups available for linking the compounds of the invention.
  • Examples of the cell binding agents include an antibody, a single chain antibody, an antibody fragment that specifically binds to the target cell, a monoclonal antibody, a single chain monoclonal antibody, a monoclonal antibody fragment that specifically binds to a target cell, a chimeric antibody, a chimeric antibody fragment that specifically binds to the target cell, a bispecific antibody, a domain antibody, a domain antibody fragment that specifically binds to the target cell, an interferon, a lymphokine (e.g., IL-2, IL-3, IL-4, and IL-6), a hormone (e.g., insulin, thyrotropin releasing hormone, melanocyte-stimulating hormone, and a steroid hormone (e.g., androgen and estrogen)), a vitamin (e.g., folate), a growth factor (e.g., EGF, TGF-alpha, FGF, VEGF), a colony stimulating factor, a nutrient- transport a
  • centyrin a protein scaffold based on a consensus sequence of fibronectin type III (FN3) repeats; see U.S. Patent Publication Nos.
  • an Ankyrin Repeat Protein e.g., a designed ankyrin repeat protein, known as DARPin; see U.S. Patent Publication Nos. 2004/0132028, 2009/0082274, 2011/0118146, and 2011/0224100, incorporated herein by reference, and also see C. Zahnd et al. (2010) Cancer Res., 70: 1595- 1605; Zahnd et al, J. Biol. Chem.(2006) 281(46):35167-35175; and Binz, H.K., Amstutz, P. & Pluckthun, A., Nature Biotechnology (2005) 23: 1257-1268, incorporated herein by reference), an ankyrin-like repeats protein or synthetic peptide (see e.g., U.S. Patent
  • WO 2007/062466 incorporated herein by reference
  • an Adnectin a fibronectin domain scaffold protein; see US Patent Publication Nos. 2007/0082365; 2008/0139791, incorporated herein by reference
  • Avibody including diabodies, triabodies, and tetrabodies; see U.S. Publication Nos. 2008/0152586 and 2012/0171115
  • other cell-binding molecules or substances see U.S. Publication Nos. 2008/0152586 and 2012/0171115
  • the cell-binding agent is an antibody, a single chain antibody, an antibody fragment that specifically binds to the target cell, a monoclonal antibody, a single chain monoclonal antibody, a monoclonal antibody fragment that specifically binds to a target cell, a chimeric antibody, a chimeric antibody fragment that specifically binds to the target cell, a domain antibody, a domain antibody fragment that specifically binds to the target cell, a lymphokine, a hormone, a vitamin, a growth factor, a colony stimulating factor, or a nutrient-transport molecule.
  • the cell-binding agent is a monoclonal antibody, a single chain monoclonal antibody, or a monoclonal antibody fragment that specifically binds to a target cell.
  • the cell-binding agent is a bispecific antibody, an ankyrin repeat protein, a Centyrin, or an Avibody.
  • Antibody fragment refers to Fab, Fab', and F(ab') 2 , Fv, minibodies, diabodies, tribodies, tetrabodies, nanobodies, probodies, domain bodies, unibodies, and the like (Parham, J. Immunol. 131:2895-2902 (1983); Spring et al. J. Immunol. 113:470-478 (1974); Nisonoff et al. Arch. Biochem. Biophys.
  • the cell-binding agent is a minibody, a diabody, a tribody, a tetrabody, a nanobody, a probody, a domain body, or an unibody.
  • Monoclonal antibody techniques allow for the production of extremely specific cell- binding agents in the form of specific monoclonal antibodies.
  • Particularly well known in the art are techniques for creating monoclonal antibodies produced by immunizing mice, rats, hamsters or any other mammal with the antigen of interest such as the intact target cell, antigens isolated from the target cell, whole virus, attenuated whole virus, and viral proteins such as viral coat proteins.
  • Sensitized human cells can also be used.
  • Another method of creating monoclonal antibodies is the use of phage libraries of scFv (single chain variable region), specifically human scFv (see e.g., Griffiths et al., U.S. Patent Nos.
  • the appropriate cell-binding agent is a matter of choice that depends upon the particular cell population that is to be targeted, but in general human monoclonal antibodies are preferred if an appropriate one is available.
  • the monoclonal antibody MY9 is a murine IgGi antibody that binds specifically to the CD33 Antigen (J.D. Griffin et al., Leukemia Res., 8:521 (1984)) and can be used if the target cells express CD33 as in the disease of acute myelogenous leukemia (AML).
  • the cell-binding agent is a resurfaced antibody, a resurfaced single chain antibody, or a resurfaced antibody fragment.
  • the cell-binding agent is a humanized antibody, a humanized single chain antibody, or a humanized antibody fragment.
  • the humanized antibody is huMy9-6 or another related antibody, which is described in U.S. Pat.
  • the humanized antibody is an anti-folate receptor antibody described in U.S. Provisional Application Nos. 61/307,797,
  • the cell-binding agent is an antigen-binding portion of a monoclonal antibody, sharing sequences critical for antigen-binding with an antibody disclosed herein, such as huMy9-6 or its related antibodies described in U.S. Pat. Nos.
  • derivative antibodies may have substantially the same or identical (1) light chain and/or heavy chain CDR3 regions; (2) light chain and/or heavy chain CDR1, CDR2, and CDR3 regions; or (3) light chain and/or heavy chain regions, compared to an antibody described herein. Sequences within these regions may contain conservative amino acid substitutions, including substitutions within the CDR regions. Preferably, there is no more than 1, 2, 3, 4, or 5 conservative substitutions. In an alternative, the derivative antibodies have a light chain region and/or a heavy chain region that is at least about 90%, 95%, 99% or 100% identical to an antibody described herein.
  • These derivative antibodies may have substantially the same binding specificity and/or affinity to the target antigen compared to an antibody described herein.
  • the K d and/or R values of the derivative antibodies are within 10-fold (either higher or lower), 5- fold (either higher or lower), 3-fold (either higher or lower), or 2-fold (either higher or lower) of an antibody described herein.
  • These derivative antibodies may be fully human antibodies, or humanized antibodies, or chimeric antibodies.
  • the derivative antibodies may be produced according to any art-recognized methods.
  • Specific exemplary antigens or ligands include renin; a growth hormone (e.g. , human growth hormone and bovine growth hormone); a growth hormone releasing factor; a parathyroid hormone; a thyroid stimulating hormone; a lipoprotein; alpha- 1- antitrypsin; insulin A-chain; insulin B-chain; proinsulin; a follicle stimulating hormone; calcitonin; a luteinizing hormone; glucagon; a clotting factor (e.g. , factor vmc, factor IX, tissue factor, and von Willebrands factor); an anti-clotting factor (e.g.
  • a growth hormone e.g. , human growth hormone and bovine growth hormone
  • a growth hormone releasing factor e.g., a parathyroid hormone; a thyroid stimulating hormone; a lipoprotein; alpha- 1- antitrypsin; insulin A-chain; insulin B-chain; proinsulin; a follicle stimulating hormone; calcitonin;
  • plasminogen activator e.g. , a urokinase, a human urine or tissue-type plasminogen activator
  • bombesin e.g. , bombesin
  • a thrombin e.g. , hemopoietic growth factor; tumor necrosis factor- alpha and -beta; an enkephalinase
  • RANTES i.e. , the regulated on activation normally
  • T-cell expressed and secreted human macrophage inflammatory protein- 1 -alpha; a serum albumin (human serum albumin); Muellerian-inhibiting substance; relaxin A-chain; relaxin
  • B-chain prorelaxin; a mouse gonadotropin-associated peptide; a microbial protein (beta- lactamase); DNase; IgE; a cytotoxic T-lymphocyte associated antigen (e.g. , CTLA-4); inhibin; activin; a vascular endothelial growth factor; protein A or D; a rheumatoid factor; a neurotrophic factor(e.g. , bone-derived neurotrophic factor, neurotrophin-3, -4, -5, or -6), a nerve growth factor (e.g. , NGF- ⁇ ); a platelet-derived growth factor; a fibroblast growth factor (e.g.
  • fibroblast growth factor receptor 2 an epidermal growth factor; a transforming growth factor (e.g. , TGF-alpha, TGF- ⁇ , TGF- 2, TGF- 3, TGF- 4, and TGF- ⁇ 5); insulin-like growth factor-I and -II; des(l-3)-IGF-I (brain IGF-I); an insulin-like growth factor binding protein; melanotransferrin; EpCAM; GD3; FLT3; PSMA; PSCA; MUC 1 ; MUC 16; STEAP; CEA; TENB2; an EphA receptor; an EphB receptor; a folate receptor; FOLR1 ; mesothelin; cripto; an alpha v beta 6 ; integrins; VEGF; VEGFR; EGFR; transferrin receptor; IRTAl ; IRTA2; IRTA3; IRTA4; IRTA5; CD proteins (e.g.
  • CD81, CD103, CD105, CD134, CD137, CD138, and CD152 one or more tumor-associated antigens or cell-surface receptors (see US Publication No. 20080171040 or US Publication No. 20080305044, incorporated in their entirety by reference); erythropoietin; an
  • osteoinductive factor an immunotoxin; a bone morphogenetic protein; an interferon (e.g. , interferon-alpha, -beta, and -gamma); a colony stimulating factor (e.g. , M-CSF, GM-CSF, and G-CSF); interleukins (e.g. , IL- 1 to IL- 10); a superoxide dismutase; a T-cell receptor; a surface membrane protein; a decay accelerating factor; a viral antigen s(e.g. , a portion of the HIV envelope); a transport protein, a homing receptor; an addressin; a regulatory protein; an integrin (e.g.
  • a tumor associated antigen e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. , HER2, HER3 and HER4 receptor
  • endoglin e.g. ,
  • GM-CSF a ligand / growth factor which binds to myeloid cells
  • IL-2 which binds to activated T-cells can be used for prevention of transplant graft rejection, for therapy and prevention of graft-versus-host disease, and for treatment of acute T-cell leukemia.
  • MSH which binds to melanocytes, can be used for the treatment of melanoma, as can antibodies directed towards melanomas.
  • Folic acid can be used to target the folate receptor expressed on ovarian and other tumors.
  • Epidermal growth factor can be used to target squamous cancers, such as lung and head and neck.
  • Somatostatin can be used to target neuroblastomas and other tumor types.
  • Estrogen or estrogen analogues
  • Androgen or androgen analogues
  • testes can be used to target testes.
  • salt refers to organic or inorganic salts of a compound of the invention.
  • a salt is a pharmaceutically acceptable salt.
  • Other non- pharmaceutically acceptable salts are also included in the present invention.
  • the salts include salts, formed by reacting a compound of the invention, which comprises a basic group, with an inorganic acid or organic acid (such as a carboxylic acid), and salts, formed by reacting a compound of the invention, which comprises an acidic group, with an inorganic base or organic base (such as an amine).
  • Exemplary salts include those pharmaceutically acceptable salts described immediately below.
  • Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate "mesylate,” ethanesulfonate, benzenesulfonate, p-toluenesulfonate, pamoate (i.e.
  • a pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion.
  • the counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • a pharmaceutically acceptable salt may have more than one charged atom in its structure.
  • a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion.
  • an inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid and the like
  • an organic acid such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like
  • desired salts may be prepared by any suitable method, for example, treatment of the free acid with an inorganic, such as an alkali metal hydroxide or alkaline earth metal hydroxide, organic base, such as an amine (primary, secondary or tertiary), or the like.
  • an inorganic such as an alkali metal hydroxide or alkaline earth metal hydroxide
  • organic base such as an amine (primary, secondary or tertiary), or the like.
  • suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • amino acids such as glycine and arginine
  • ammonia such as glycine and arginine
  • primary, secondary, and tertiary amines such as piperidine, morpholine and piperazine
  • inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • abnormal cell growth and “proliferative disorder” are used interchangeably in this application.
  • abnormal cell growth refers to cell growth that is independent of normal regulatory
  • tumor cells tumors
  • mutated tyrosine kinase or overexpression of a receptor tyrosine kinase tumor cells
  • benign and malignant cells of other proliferative diseases in which aberrant tyrosine kinase activation occurs tumor cells that proliferate by expressing a mutated tyrosine kinase or overexpression of a receptor tyrosine kinase
  • benign and malignant cells of other proliferative diseases in which aberrant tyrosine kinase activation occurs any tumors that proliferate by receptor tyrosine kinases
  • any tumors that proliferate by aberrant serine/threonine kinase activation and (5) benign and malignant cells of other proliferative diseases in which aberrant serine/threonine kinase activation occurs.
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells, and/or benign or pre-cancerous cells.
  • cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include skin cancer (e.g. , melanoma), Merkel cell carcinoma, squamous cell cancer (e.g. , epithelial squamous cell cancer), lung cancer (e.g.
  • small-cell lung cancer non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung
  • cancer of the peritoneum hepatocellular cancer
  • gastric or stomach cancer e.g. , gastrointestinal cancer
  • pancreatic cancer cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, testicular cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, acute leukemia, head and neck cancer, brain cancer (e.g.
  • glioblastoma and neuroblastoma cancers of lymphatic organs and hematological malignancy including Leukemia (Acute lymphoblastic leukemia (ALL), Acute myelogenous leukemia (AML), Chronic lymphocytic leukemia (CLL), Chronic myelogenous leukemia (CML), Acute monocytic leukemia (AMOL), Hairy cell leukemia (HCL), T-cell prolymphocytic leukemia (T-PLL), Large granular lymphocytic leukemia, Adult T-cell leukemia), Lymphoma (small lymphocytic lymphoma (SLL), Hodgkin's lymphomas
  • ALL Acute lymphoblastic leukemia
  • AML Acute myelogenous leukemia
  • CLL Chronic lymphocytic leukemia
  • CML Chronic myelogenous leukemia
  • Acute monocytic leukemia AMOL
  • HCL Hairy cell leukemia
  • Nodular sclerosis Mixed cellularity, Lymphocyte-rich, Lymphocyte depleted or not depleted, and Nodular lymphocyte-predominant Hodgkin lymphoma
  • Non-Hodgkin's lymphomas all subtypes
  • Chronic lymphocytic leukemia/Small lymphocytic lymphoma B- cell prolymphocytic leukemia
  • Lymphoplasmacytic lymphoma such as Waldenstrom macro globulinemia
  • Splenic marginal zone lymphoma Plasma cell neoplasms (Plasma cell myeloma, Plasmacytoma, Monoclonal immunoglobulin deposition diseases, Heavy chain diseases), Extranodal marginal zone B cell lymphoma (MALT lymphoma), Nodal marginal zone B cell lymphoma (NMZL), Follicular lymphoma, Mantle cell lymphoma, Diffuse large
  • B cell lymphoma Mediastinal (thymic) large B cell lymphoma, Intravascular large B cell lymphoma, Primary effusion lymphoma, Burkitt lymphoma/leukemia, T cell prolymphocytic leukemia, T cell large granular lymphocytic leukemia, Aggressive NK cell leukemia, Adult T cell leukemia/lymphoma, Extranodal NK/T cell lymphoma (nasal type), Enteropathy-type T cell lymphoma, Hepatosplenic T cell lymphoma, Blastic NK cell lymphoma, Mycosis fungoides / Sezary syndrome, Primary cutaneous CD30-positive T cell lymphoproliferative disorders, Primary cutaneous anaplastic large cell lymphoma, Lymphomatoid papulosis,
  • Angioimmunoblastic T cell lymphoma Peripheral T cell lymphoma (unspecified), Anaplastic large cell lymphoma), multiple myeloma (plasma cell myeloma or Kahler's disease).
  • therapeutic agent encompasses both a biological agent such as an antibody, a peptide, a protein, an enzyme or a chemotherapeutic agent.
  • chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include Erlotinib (TARCEVA®,
  • Imatinib mesylate GLEEVEC®, Novartis
  • PTK787/ZK 222584 Novartis
  • AstraZeneca AG1478, AG1571 (SU 5271; Sugen), alkylating agents such as thiotepa and
  • CYTOXAN® cyclosphosphamide alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
  • ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine;
  • acetogenins especially bullatacin and bullatacinone
  • a camptothecin including the synthetic analog topotecan
  • bryostatin callystatin
  • CC-1065 including its adozelesin, carzelesin and bizelesin synthetic analogs
  • cryptophycins particularly cryptophycin 1 and cryptophycin 8
  • dolastatin duocarmycin (including the synthetic analogs, KW-2189 and CB1-TM1);
  • pancratistatin a sarcodictyin
  • spongistatin nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide,
  • calicheamicin omegall Angew Chem. Intl. Ed. Engl. (1994) 33: 183-186
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6- diazo-5-oxo-L-norleucine, ADRIAMYCIN ® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubic
  • methotrexate, pteropterin, trimetrexate purine analogs such as fludarabine, 6- mercaptopurine, thiamniprine, 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 aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK ® polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin;
  • vindesine dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g.
  • TAXOL ® paclitaxel; Bristol-Myers Squibb Oncology, Princeton, N.J.
  • ABRAXANE® Cremophor-free
  • albumin-engineered nanoparticle formulations of paclitaxel American Pharmaceutical Partners, Schaumberg, 111.
  • TAXOTERE ® doxetaxel; Rhone-Poulenc Rorer, Antony, France
  • chloranmbucil GEMZAR ® (gemcitabine); 6-thioguanine; mercaptopurine;
  • methotrexate platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE ® (vinorelbine); novantrone;
  • teniposide edatrexate; daunomycin; aminopterin; capecitabine (XELODA ® ); ibandronate; CPT-11 ; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.
  • chemotherapeutic agent also included in the definition of "chemotherapeutic agent” are: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen
  • aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE ®
  • anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog);
  • protein kinase inhibitors e.g., IL-12 kinase inhibitors
  • lipid kinase inhibitors e.g., IL-12 IL-12
  • antisense oligonucleotides particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Ralf and H-Ras
  • ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME ® ) and HER2 expression inhibitors
  • vaccines such as gene therapy vaccines, for example, ALLOVECTIN ® , LEUVECTIN ® , and VAXID ® ;
  • PROLEUKIN® rIL-2 a topoisomerase 1 inhibitor such as LURTOTECAN ® ; ABARELIX® rmRH; (ix) anti- angiogenic agents such as bevacizumab (AVASTIN ® , Genentech); and (x) pharmaceutically acceptable salts, acids and derivatives of any of the above.
  • Other anti- angiogenic agents include MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix - metalloproteinase 9) inhibitors, COX-II (cyclooxygenase II) inhibitors, and VEGF receptor tyrosine kinase inhibitors.
  • VEGF receptor tyrosine kinase inhibitors include 4-(4-bromo-2-fluoroanilino)-6-methoxy-7-(l-methylpiperidin-4- ylmethoxy)quinazoline (ZD6474; Example 2 within WO 01/32651), 4-(4-fluoro-2- methylindol-5-yloxy)-6-methoxy-7-(3-pyrrolidin- l-ylpropoxy)-quinazoline (AZD2171 ;
  • Example 240 within WO 00/47212 vatalanib (PTK787; WO 98/35985) and SU11248 (sunitinib; WO 01/60814), and compounds such as those disclosed in PCT Publication Nos. WO 97/22596, WO 97/30035, WO 97/32856, and WO 98/13354).
  • chemotherapeutic agents that can be used in combination with the present compounds include inhibitors of PI3K (phosphoinositide-3 kinase), such as those reported in Yaguchi et al. (2006) J. Nat. Cancer Inst. 98(8):545-556; U.S. Pat. No. 7,173,029;
  • PI3K phosphoinositide-3 kinase
  • PI3K inhibitors include SF-1126 (PI3K inhibitor, Semafore Pharmaceuticals), BEZ-235 (PI3K inhibitor, Novartis), XL- 147 (PI3K inhibitor, Exelixis, Inc.).
  • Chemotherapeutic agents may also include any of the generic drugs or biosimilars of the brand-name drugs referenced herein, or improvements thereof, including improved formulations, delivery means (sustained release, bioadhesive coating, targeted delivery etc. ), and dosage forms.
  • viral infection refers to the invasion of a host organism's bodily tissues by disease-causing viruses.
  • examples of the viral infections include CMV infection, HIV infection and AIDS.
  • parasite infection refers to the invasion of a host organism's bodily tissues by disease-causing parasites.
  • examples of the parasite infections include giardiasis, amoebiasis, and schistosomiasis.
  • composition indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • terapéuticaally effective amount means that amount of active compound or conjugate that elicits the desired biological response in a subject. Such response includes alleviation of the symptoms of the disease or disorder being treated, prevention, inhibition or a delay in the recurrence of symptom of the disease or of the disease itself, an increase in the longevity of the subject compared with the absence of the treatment, or prevention, inhibition or delay in the progression of symptom of the disease or of the disease itself. Determination of the effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Toxicity and therapeutic efficacy of compound I can be determined by standard pharmaceutical procedures in cell cultures and in experimental animals.
  • the effective amount of compound or conjugate of the present invention or other therapeutic agent to be administered to a subject will depend on the stage, category and status of the multiple myeloma and characteristics of the subject, such as general health, age, sex, body weight and drug tolerance.
  • the effective amount of compound or conjugate of the present invention or other therapeutic agent to be administered will also depend on administration route and dosage form. Dosage amount and interval can be adjusted individually to provide plasma levels of the active compound that are sufficient to maintain desired therapeutic effects.
  • the present invention provides a conjugate represented by Formula ( ⁇ ) or (I) as described in the first embodiment and its alternatively embodiments above.
  • the ionizable group is -SO 3 H, - Z'-S0 3 H, -OP0 3 H 2 , -Z'-OP0 3 H 2 , -P0 3 H 2 , -Z'-P0 3 H 2 , -C0 2 H, -Z'C0 2 H, -NRnRi 2 , or -Z - NRnRi 2 , and the charged group is -N ⁇ R ⁇ RisX " , or -Z'-N ' ⁇ R RwRisX " ; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R 12 , for each occurrence, are independently H or an optionally substituted alkyl; R 1 to R15 are each independently an optionally substituted alkyl; and X " is a pharmaceutically acceptable anion.
  • this first specific embodiment is an optionally substituted alkylene, an optionally substituted cycl
  • variable Z' is alkylene and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first specific embodiment.
  • R m is H, -CH 2 CH 2 S0 3 H, or a pharmaceutically acceptable salt of -CH 2 CH 2 S0 3 H, and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments.
  • the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • JCB' is and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • s1 ⁇ N-NR e --i s2
  • J C B' in Formula ( ⁇ ) or (I) is * 3 ⁇ 4 ,
  • R a , R b , R c , and R e are each H; and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, or eighth specific embodiment.
  • the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth specific embodiment.
  • R 3 , R4, R5 and R 6 are all H, m and n are each independently an integer from 1 to 5 (e.g., 1, 2, 3, 4 or 5); and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth or eleventh specific embodiment,
  • DM is a drug moiety represented by Formula (II) as described in the first embodiment above or by Formula (IIA) described in the tenth specific embodiment.
  • w is preferably an integer between 1 and 10, between 1 and 6, or alternatively, between 1 and 4; and the remainder of the variables in Formula ( ⁇ ) or (I) are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth,, ninth, tenth, eleventh,twelfth, thirteenth or fourteenth specific embodiment.
  • the present invention also provides a conjugate represented by Formula (V) as described in the first embodiment and its alternatively embodiments above.
  • variables is as defined in the first embodiment or each of its alternative embodiments or its first or second specific embodiment described above.
  • R4 and R 2 are both H; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second or third specific embodiment.
  • R a , R b , R c , and R e are each H; and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third or fourth specific embodiment.
  • DM is represented by formula (IIA) described above; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth or fifth specific embodiment.
  • w is preferably an integer between 1 and 10, between 1 and 6, or alternatively, between 1 and 4; and the remainder of the variables in Formula (V) are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth fifth or sixth specific embodiment.
  • the present invention also provides a cytotoxic compound represented by Formula ( ⁇ ) or (III) as described in the second embodiment and its alternatively embodiments above.
  • the ionizable group is -SO 3 H, -Z'-S0 3 H, -OP0 3 H 2 , -Z'-OP0 3 H 2 , -P0 3 H 2 , -Z'-P0 3 H 2 , -C0 2 H, -Z'C0 2 H, -NRnRi 2 , or -Z - NRnRi 2 , and the charged group is -N + R 13 R 14 Ri 5 X ⁇ , or -Z'-N ' ⁇ R RwRisX " ; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R 12 , for each occurrence, are independently H or an optionally substituted alkyl; R 1 to R15 are each independently an optionally substituted alkyl; and X " is an anion (e.g.
  • variable Z' is alkylene and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first specific embodiment.
  • R m is H, -CH 2 CH 2 S0 3 H, or a pharmaceutically acceptable salt of -CH 2 CH 2 S0 3 H, and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments.
  • the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • J CB is maleimide and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • J CB is NH 2 -NR e - or NH 2 -0-; and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • R a , R b , R c , and R e are each H; and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, or eighth specific embodiment.
  • DM is represented by the formula (IIA) as described above; and the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth specific embodiment.
  • R 3 , R 4 , R 5 and R 6 are all H, m and n are each independently an integer from 1 to 5 (e.g., 1, 2, 3, 4 or 5); the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth or eleventh specific embodiment,
  • the cytotoxic compound in Formula ( ⁇ ) or (III), is represented by any one of the following formulae, or a salt (e.g. , a
  • DM is a drug moiety represented by Formula (II) as described in the second embodiment above or Formula (IIA) as described above.
  • the present invention also provides a cytotoxic compound represented by Formula (VI) or (III) as described in the second embodiment or each of its alternatively embodiments above.
  • R and R 2 are both H; and the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments or its first, second or third specific embodiment.
  • R a , R b , R c , and R e are each H; and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third or fourth specific embodiment.
  • DM is represented by formula (IIA) described above; and the remainder of the variables are as defined in the second embodiment or each its alternative embodiments or its first, second, third, fourth or fifith specific embodiment.
  • the present invention further provides a linker compound represented by Formula (IV) or (IV) as described in the third embodiment and its alternatively embodiments above.
  • Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene;
  • Rn and R 12 for each occurrence, are independently H or an optionally substituted alkyl;
  • R 13 to R15 are each independently an optionally substituted alkyl; and
  • X " is an anion (e.g. , a pharmaceutically
  • variable Z' is alkylene and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first specific embodiment.
  • R m is H, -CH 2 CH 2 S0 3 H, or a pharmaceutically acceptable salt of -CH 2 CH 2 S0 3 H, and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments.
  • the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • JCB is maleimide and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • JCB is NH 2 -NR E - or NH 2 - 0-; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
  • R A , R B , R C , and R E are each H; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, or eighth specific embodiment.
  • R 3 , R 4 , R 5 and R 6 are all H, m and n are each independently an integer from 1 to 5 (e.g., 1 , 2, 3, 4 or 5); the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth ,ninth, tenth or eleventh specific embodiment,
  • the linker compound is represented by any one of the following formulae, or a salt ⁇ e.g. , a pharmaceutically acceptable salt) thereof:
  • the present invention further provides a linker compound represented by Formula (VII) as described in the third embodiment and its alternatively embodiments above.
  • R and R 2 are both H; and the remainder of the variables is as defined in the third embodiment or each of its alternative embodiments or its first, second or third specific embodiment.
  • R a , R b , R c , and R e are each H; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third or fourth specific embodiment.
  • conjugates of the present invention can be prepared according to any known method in the art. See, for example, WO 2009/134977, U.S. Patent Nos. 7,811,572,
  • the conjugates of the present invention can be prepared by reacting a cell-binding agent with a drug-linker compound (e.g., compounds of formula ( ⁇ ) or (III)) having a reactive moiety capable of forming a covalent bond with the cell-binding agent to form a cell-binding agent- cytotoxic agent conjugate.
  • a drug-linker compound e.g., compounds of formula ( ⁇ ) or (III)
  • the conjugate can then be purified.
  • the drug-linker compound can be generated in situ and used to react with the antibody without purification.
  • the drug-linker compound can be generated and purified before conjugating to the cell-binding agent.
  • the conjugates of the present invention can be prepared by: a) reacting a cell-binding agent with a linker compound (e.g., compounds of formula IV) to form a modified cell-binding agent having the linkers covalently bound thereto; b) optionally purifying the modified cell-binding agent; c) conjugating a cytotoxic agent to the modified cell-binding agent to form the cell-binding agent-cytotoxic compound conjugate of the present invention; and d) purifying the cell-binding agent-cytotoxic compound conjugate.
  • a linker compound e.g., compounds of formula IV
  • the conjugate of the present invention can be prepared by mixing together a cell-binding agent, a cytotoxic agent and a linker compound.
  • the cell-binding agent is contacted with a cytotoxic agent first to form a mixture comprising the cell-binding agent and the cytotoxic agent, followed by contacting the mixture with a linker compound (e.g., compounds of formula (IV)) to form the cell-binding agent-cytotoxic compound conjugate.
  • a linker compound e.g., compounds of formula (IV)
  • conjugates of the present invention using tangential flow filtration (TFF), non-adsorptive
  • TFF systems Any suitable TFF systems may be utilized for purification, including a Pellicon type system (Millipore, Billerica, MA), a Sartocon Cassette system (Sartorius AG, Edgewood,
  • Any suitable adsorptive chromatography resin may be utilized for purification.
  • Preferred adsorptive chromatography resins include hydroxyapatite chromatography, hydrophobic charge induction chromatography (HCIC), hydrophobic interaction
  • hydroxyapatite resins include ceramic hydroxyapatite (CHT).
  • HCIC resin MEP Hypercel resin
  • HIC resins include Butyl-Sepharose,
  • Suitable ion exchange resins include SP-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethacrylate resins, terpolymer graft copoethyl-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-N-(2-aminoe
  • Sepharose, CM-Sepharose, and Q-Sepharose resins All from GE Healthcare, Piscataway,
  • Unosphere S resin Bio-Rad Laboratories, Hercules, CA.
  • suitable mixed mode ion exchangers include Bakerbond ABx resin (JT Baker, Phillipsburg NJ).
  • IMAC resins examples include Chelating Sepharose resin (GE Healthcare,
  • suitable dye ligand resins include Blue Sepharose resin (GE Healthcare, Piscataway, NJ) and Affi-gel Blue resin (Bio-Rad Laboratories, Hercules, CA).
  • suitable affinity resins include Protein A Sepharose resin (e.g. , MabSelect, GE Healthcare, Piscataway, NJ), His-Tag metal affinity resins, anti-FLAG affinity resins, and lectin affinity resins, e.g. Lentil Lectin Sepharose resin (GE Healthcare, Piscataway, NJ), where the antibody bears appropriate lectin binding sites.
  • suitable reversed phase resins include C4, C8, and C18 resins (Grace Vydac, Hesperia, CA).
  • any suitable non-adsorptive chromatography resin may be utilized for purification.
  • size-exclusion chromatography can be used for purifying the conjugates of the invention.
  • suitable non-adsorptive chromatography resins include, but are not limited to, SEPHADEXTM G- 10, G-25, G-50, G- 100, SEPHACRYLTM resins (e.g. , S-200 and S-300), SUPERDEXTM resins (e.g. , SUPERDEXTM 75 and SUPERDEXTM 200), BIO-GEL® resins (e.g. , P-6, P-10, P-30, P-60, and P- 100), and others known to those of ordinary skill in the art.
  • the conjugate when the cell-binding agent is an epitope-tagged Avibody, the conjugate can be purified using hydroxyl apatite chromatography, size-exclusion
  • affinity chromatography preferably affinity chromatography, more preferably His-tag metal affinity chromatography and anti-FLAG M2 affinity chromatography (see, for example,
  • the conjugate when the cell-binding agent is a centyrin, can be purified using protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, size-exclusion chromatography, tangential flow filtration, affinity chromatography, hydroxylapatite chromatography and lectin
  • the conjugate can be purified using HPLC.
  • the conjugate can be purified by using affinity chromatography, more preferably His-tag metal affinity chromatography. See, for example, US 2010/0255056, US 2010/0216708 and US 2011/0274623.
  • the conjugate when the cell-binding agent is a DARPin, can be purified by affinity chromatography, size exclusion chromatography, hydroxylapatite chromatography, tangential flow fitration, preferably affinity chromatography, more preferably His-Tag affinity chromatography. See, for example, U.S. Patent Publication Nos. 2004/0132028, 2009/0082274, 2011/0118146, and 2011/0224100, WO 02/20565 and WO 06/083275.
  • the number of cytotoxic compound molecule bound per cell-binding agent (e.g. , antibody) molecule can be determined spectroscopically by measuring the ratio of the absorbance at 280 nm and 252 nm. An average of about 0.5- about 20 cytotoxic compound molecule bound per cell-binding agent (e.g. , antibody) molecule.
  • the average number of linked cytotoxic compound per cell-binding agent in the conjugate is about 0.5 to about 10, about 0.5 to 2 (e.g., 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, or 2.1), about 2 to about 8 (e.g. , 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9,
  • the cytotoxic compounds and cell-binding agent-drug conjugates of the invention can be evaluated for their ability to suppress proliferation of various cancer cell lines in vitro.
  • the in vitro cytotoxicity assays can be conducted using methods known in the art (e.g. , Widdison, W. C. et al. "Semisynthetic maytansine analogues for the targeted treatment of cancer," J. Med. Chem. (2006) 49(14):4392-408).
  • cells to be evaluated can be exposed to the compounds or conjugates for 1-5 days and the surviving fractions of cells measured in direct assays by known methods. IC 50 values can then be calculated from the results of the assays. COMPOSITIONS AND METHODS OF USE
  • the present invention includes a composition (e.g. , a pharmaceutical composition) comprising conjugates (e.g. , conjugates of Formula ( ⁇ ) or (I)) or cytotoxic compounds (e.g. , compounds of Formula ( ⁇ ) or (III)) described herein, and a carrier (a pharmaceutically acceptable carrier).
  • the present invention also includes a composition (e.g. , a pharmaceutical composition) comprising the conjugate of Formula ( ⁇ ) or (I) or the cytotoxic compound of Formula ( ⁇ ) or (III), and a carrier (a pharmaceutically acceptable carrier), and further comprising a second therapeutic agent.
  • the present compositions are useful for inhibiting abnormal cell growth or treating a proliferative disorder in a mammal (e.g. , human).
  • compositions are also useful for treating an autoimmune disorder, a destructive bone disorder, a graft versus host disease, a transplant rejection, an immune deficiency, an inflammatory disease, an infectious disease, a viral disease, a fibrotic disease, a
  • neurodegenerative disorder a pancreatitis or kidney disease in a mammal (e.g. , human).
  • the present invention includes a method of inhibiting abnormal cell growth or treating a proliferative disorder in a mammal (e.g. , human) comprising administering to said mammal a therapeutically effective amount of conjugates (e.g. , conjugates of formula ( ⁇ ) or (I)) or cytotoxic compounds (e.g. , compounds of formula ( ⁇ ) or (III)) described herein, or a composition thereof, alone or in combination with a second therapeutic agent.
  • conjugates e.g. , conjugates of formula ( ⁇ ) or (I)
  • cytotoxic compounds e.g. , compounds of formula ( ⁇ ) or (III)
  • the proliferative disorder is cancer in general; alternatively, the proliferative disorder is cancer selected from the group consisting of breast cancer, colon cancer, brain cancer, prostate cancer, kidney cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, colorectal cancer, gastric cancer, squamous cancer, small-cell lung cancer, nonsmall-cell lung cancer, testicular cancer, Merkel cell carcinoma, glioblastoma, neuroblastoma, a cancer of a lymphatic organ, and a hematological malignancy.
  • the present invention provides a method for inducing cell death in selected cell populations comprising contacting target cells or tissue containing target cells with an effective amount of the conjugates of the present invention.
  • the target cells are cells to which the cell-binding agent of the conjugates can bind.
  • chemotherapeutic drugs that are therapeutically effective will depend on the particular cancer being treated, the extent of the disease and other factors familiar to the physician of skill in the art and can be determined by the physician.
  • the contents of the PDR are expressly incorporated herein in its entirety by reference.
  • One of skill in the art can review the PDR, using one or more of the following parameters, to determine dosing regimen and dosages of the chemotherapeutic agents and conjugates that can be used in accordance with the teachings of this invention. These parameters include: Comprehensive index; Manufacturer; Products (by company's or trademarked drug name); Category index; Generic/chemical index (non- trademark common drug names); Color images of medications; Product information, consistent with FDA labeling; Chemical information; Function/action; Indications &
  • the present invention also provides methods of treating a non-cancerous condition comprising administering to a subject in need of treatment an effective amount of any of the conjugates described above.
  • the conditions include, but not limited to, an autoimmune disorder (e.g. , systemic lupus, rheumatoid arthritis, and multiple sclerosis), a graft versus host disease, a transplant rejection (e.g. , a renal transplant rejection, a liver transplant rejection, a lung transplant rejection, a cardiac transplant rejection, and a bone marrow transplant rejection), an immune deficiency, an inflammatory diseases (i.e. , myositis and pancreatitis), a destructive bone disorder, an infectious disease (e.g.
  • the condition selected from the group consisting of cancer, rheumatoid arthritis, multiple sclerosis, graft versus host disease, transplant rejection, lupus, myositis, infectious disease, and immune deficiency.
  • Examples of in vitro uses include treatments of autologous bone marrow prior to their transplant into the same patient in order to kill diseased or malignant cells: treatments of bone marrow prior to their transplantation in order to kill competent T cells and prevent graft- versus-host-disease (GVHD); treatments of cell cultures in order to kill all cells except for desired variants that do not express the target antigen; or to kill variants that express undesired antigen.
  • treatments of autologous bone marrow prior to their transplant into the same patient in order to kill diseased or malignant cells treatments of bone marrow prior to their transplantation in order to kill competent T cells and prevent graft- versus-host-disease (GVHD); treatments of cell cultures in order to kill all cells except for desired variants that do not express the target antigen; or to kill variants that express undesired antigen.
  • GVHD graft- versus-host-disease
  • Examples of clinical ex vivo use are to remove tumor cells or lymphoid cells from bone marrow prior to autologous transplantation in cancer treatment or in treatment of autoimmune disease, or to remove T cells and other lymphoid cells from autologous or allogenic bone marrow or tissue prior to transplant in order to prevent GVHD.
  • Treatment can be carried out as follows. Bone marrow is harvested from the patient or other individual and then incubated in medium containing serum to which is added the cytotoxic agent of the invention, concentrations range from about 10 ⁇ to 1 pM, for about 30 minutes to about 48 hours at about 37°C.
  • the exact conditions of concentration and time of incubation i.e., the dose, are readily determined by one of ordinary skill in the art.
  • the bone marrow cells are washed with medium containing serum and returned to the patient intravenously according to known methods.
  • the treated marrow cells are stored frozen in liquid nitrogen using standard medical equipment.
  • the cytotoxic compounds or conjugates of the invention will be supplied as a solution or a lyophilized powder that are tested for sterility and for endotoxin levels.
  • suitable protocols of conjugate administration are as follows.
  • Conjugates are given weekly for 4 weeks as an intravenous bolus each week.
  • Bolus doses are given in 50 to 1000 ml of normal saline to which 5 to 10 ml of human serum albumin can be added. Dosages will be 10 ⁇ g to 2000 mg per administration, intravenously (range of 100 ng to 20 mg/kg per day). After four weeks of treatment, the patient can continue to receive treatment on a weekly basis. Specific clinical protocols with regard to route of
  • administration can be determined by one of ordinary skill in the art as the clinical situation warrants.
  • Suitable pharmaceutically acceptable carriers, diluents, and excipients are well known and can be determined by those of ordinary skill in the art as the clinical situation warrants.
  • suitable carriers, diluents and/or excipients include: (1) Dulbecco's phosphate buffered saline, pH about 7.4, containing or not containing about 1 mg/ml to 25 mg/ml human serum albumin, (2) 0.9% saline (0.9% w/v NaCl), and (3) 5% (w/v) dextrose; and may also contain an antioxidant such as tryptamine and a stabilizing agent such as Tween 20.
  • cytotoxic agents [193]
  • each of the cytotoxic agents described herein can be modified in such a manner that the resulting compound still retains the specificity and/or activity of the starting compound.
  • the skilled artisan will also understand that many of these compounds can be used in place of the cytotoxic agents described herein.
  • the cytotoxic agents of the present invention include analogues and derivatives of the compounds described herein.
  • Z-Gly-Gly-Gly-Gly-OH (compound 1, 1.3 g, 4.0 mmol), tert-butyl-3-aminopropionate (compound 2, 0.583 g, 4.0 mmol), hydroxybenzotriazole (0.651 g, 4.25 mmol) and N-(3- dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (0.81 g, 4.23 mmol) were weighed into a 50 mL flask then dissolved in dimethylformamide (20 mL) and magnetic stirring under a nitrogen atmosphere.
  • reaction mixture was purified in four equal portions by reverse phase HPLC using a 5.0 cm x 25 cm C18 column.
  • the column was eluted at 100 mL/min with deionized water containing 0.3 % formic acid and 5% acetonitrile for 10 min followed by a 13 min linear gradient from 5% acetonitrile to 33 % acetonitrile.
  • Product fractions (R t 21 min) were combined and solvent was removed by rotary evaporation under vacuum to give 832 mg (62 %) of compound 5.
  • Reaction of commercially available Boc-Gly-Gly-Gly-OH (compound 8) with N- hydroxyxuccinimide and EDC coupling agent affords compound 9.
  • Reaction of compound 9 with l-(2-aminoethyl)-maleimide HCl in the presence of a base such as diisopropyl ethyl amine (DIPEA) followed by Boc deprotection with HCl in methoxymethyl ether gives compound 10.
  • Reaction of compound 10 with glutamic anhydride gives compound 11.
  • Reaction of compound 11 with DM' using EDC as coupling agent will give the desired product compound 12.
  • compounds having different peptide moieties other than Gly-Gly-Gly in compound 12 can be prepared using different Boc protected peptides or Boc protected amino acids.
  • Other cyclic anhydrides or a mono t-Butyl ester protected diacid can be used in place of glutaric anhydride and the t-Bu ester if present can be deprotected with trifluoroacetic acid to give other spacers between the DM' and peptide moieties.
  • Compounds of the present invention having a maleimide group and a tetrapeptide moiety can be prepared according to the synthetic scheme shown above.
  • the Z (benzyloxy-carbonyl) protected tripeptide (compound 13) can be prepared by standard techniques (see, for example, Lloyd- Williams, P., Albericio, F., and Giralt, E. (1997) Chemical Approaches to the Synthesis of Peptides and Proteins (Boca Raton: CRC Press); and Benoiton, N. L. (2006) Chemistry of Peptide Synthesis, (Boca Raton: Taylor & Francis)).
  • Compound 13 can then react with beta-alanine t-butyl ester to yield compound 14.
  • Greene's protective groups in organic synthesis 4th ed. (Hoboken, N.J.: Wiley- Interscience)
  • Other photocleavable protecting groups known to one skilled in the art can also be used for protecting the side chain carboxylic acid group.
  • Amino acid side chains bearing a hydroxyl group can be protected, typically as esters such as acetyl or trifluoroacetyl esters.
  • Amino acid side chains bearing an amine group can be protected, typically as a carbamate such as a methyl or ethyl carbamate. Deprotection of the above-mentioned protecting group can be carried out by standard methods known in the art. (Greene)
  • Compounds of the present invention having a maleimide group and a tripeptide moiety can be prepared according to the synthetic scheme shown above.
  • the Boc protected peptide (compound 19) can be prepared by standard techniques (see, for example, Lloyd- Williams, P., Albericio, F., and Giralt, E. (1997) Chemical Approaches to the Synthesis of Peptides and Proteins (Boca Raton: CRC Press); and Benoiton, N. L. (2006) Chemistry of Peptide Synthesis, (Boca Raton: Taylor & Francis)).
  • Compound 19 can then be converted to an activated ester (compound 20) by reaction with N-hydroxysuccinimide and EDC.
  • Compound 20 can then react with l-(2-aminoethyl)-maleimide HC1 followed by deprotection with HC1 to yield compound 21.
  • Compound 21 can then react with glutaric anhydride to yield compound 22, which can be coupled to DM' using EDC to form compound 23.
  • the amino acid side chains bear a carboxylic acid
  • it can be protected, typically as esters such as methyl or ethyl esters.
  • the carboxylic acid can be protected as the 2-(trimethylsilyl)ethanol ester or other photocleavable esters known in the art (Wuts, P. G. M., and Greene, T. W. (2007) Greene's protective groups in organic synthesis, 4th ed. (Hoboken, N.J.: Wiley- Interscience)), which can be removed by Bu 4 NF or treatment with light of appropriate wavelength.
  • photocleavable protecting groups known to one skilled in the art (Greene's protecting groups in organic synthesis fourth ed.) can also be used for protecting the side chain carboxylic acid group.
  • Amino acid side chains bearing a hydroxyl group can be protected, typically as esters such as acetyl or trifluoroacetyl esters.
  • Amino acid side chains bearing an amine group can be protected, typically as a carbamate such as a methyl or ethyl carbamate. Deprotection of the above-mentioned protecting group can be carried out by standard methods known in the art. (Greene)
  • Boc protected Ala-Ala-Ala-OH (compound 25) can be activated by reaction with N- hydroxysuccinimde and EDC coupling agent followed by reaction with hydrazine.
  • the resulting hydrazide (compound 26) can be FMoc protected by reaction with FMoc-Cl and the Boc protecting group can be removed by reaction with dilute HC1.
  • the resulting compound 27 can react with l,4-Dioxane-2,6-dione in the presence of DIPEA to give compound 28.
  • DM' can be coupled to compound 28 using EDC and the FMoc protecting group can be removed by reaction with morpholine to give desired compound 29.
  • the scheme can be generalized by replacing Boc-Ala-Ala-Ala-OH (compound 25) with a Boc protected amino acid or peptide and the glutaric anhydride can also be replaced by a different cyclic anhydride or with a mono protected diacid to give other spacers between the DM' and peptide moieties.
  • Boc-Ala-Ala-Ala-OH compound 25
  • glutaric anhydride can also be replaced by a different cyclic anhydride or with a mono protected diacid to give other spacers between the DM' and peptide moieties.
  • Boc protected H-Gly-Gly-Gly-OH (compound 8) can be protected by reaction with a 9-Fluorenemethanol DIC coupling agent and dimethylaminopyridine (DMAP).
  • DMAP dimethylaminopyridine
  • the resulting compound 30 can be Boc deprotected with trifluoroacetic acid to give compound 31, which can the react with l,4-Dioxane-2,6-dione followed by protection reaction with
  • compound 32 trimethylsilylethanol in the presence of DIC and DMAP to yield compound 32.
  • Compound 32 can then be treated with morpholine to deprotect the 9-Fluorenemethane ester to give compound 33.
  • DM' can then be coupled to compound 33 with EDC coupling agent and HOBT to afford compound 34.
  • the trimethyl silylethane ester of compound 34 can be removed by treatment with tetrabutyl ammonium fluoride, followed by reaction with N- hydroxy succinimide to activate the carboxylic acid group and then with hydrazine to give desired compound 35.
  • the method can be generalized by replacing Boc-Gly-Gly-Gly-OH with a Boc- protected amino acid or peptide of choice.
  • the l,4-Dioxane-2,6-dione can also be replaced by a different cyclic anhydride or with a mono trimethylsilylethane protected diacid to give other compounds of the present invention having other spacers between the DM' and the peptide moieties.
  • Boc protected Ala-Ala-Ala-OH (compound 25) can be activated by reaction with N- hydroxysuccinimide (NHS) and EDC, followed by reaction with 1,2-diaminoethane to give compound 36.
  • Compound 36 can be treated with FMoc-aminoxyacetic acid in the presence EDC coupling agent followed by Boc deprotection with dilute HCI.
  • the resulting compound 37 can react with l,4-Dioxane-2,6-dione to give compound 38.
  • a cell-binding agent bearing one or more thiols will be reacted with a maytansinoid bearing a maleimide moiety.
  • Low molecular weight impurities can be removed from the conjugate by, for example, diafiltration, tangential flow filtration, size exclusion
  • thiols remain un-modified they can be left unreacted or they can optionally be allowed to form internal disulfide bonds with other free thiols in the cell-binding agent or they can optionally be capped by a thiol-reactive quenching agent, such as N-ethyl maleimide or 2- bromo acetamide.
  • a thiol-reactive quenching agent such as N-ethyl maleimide or 2- bromo acetamide.
  • a cell binding agent bearing one or more aldehydes or ketones will be reacted with a maytansinoid bearing a hydrazide moiety. Any unreacted aldehydes or ketones can be left unreacted or can optionally be reacted with a separate reagent such as the hydrazide of acetic acid. Low molecular weight impurities can optionally be removed from the conjugate by, for example, diafiltration, tangential flow filtration, size exclusion chromatography or by other chromatographic techniques known to one skilled in the art.
  • the conjugate can optionally be reduced with a reducing agent such as but not limited to sodium cyanoborohydride or sodium triacetoxy borohydride.
  • a cell binding agent bearing one or more aldehydes or ketones will be reacted with a maytansinoid bearing an alkoxyamine moiety. Any unreacted aldehydes or ketones can be left unreacted or can optionally be reacted with a separate reagent such as the hydrazide of acetic acid. Low molecular weight impurities can optionally be removed from the conjugate by, for example, diafiltration, tangential flow filtration, size exclusion chromatography or by other chromatographic techniques known to one skilled in the art. The conjugate can optionally be reduced with a reducing agent such as but not limited to sodium
  • Boc-Gly 3 -OH (25b, 6 g, 20.7 mmol) was dissolved in DMF (40 mL) to which was added N-hydroxysuccinimide (2.4 g, 20.7 mmol) and EDC (4 g, 20.7 mmol) in a flask containing a magnetic stir bar.
  • the reaction was magnetically stirred for 1 hour then slowly poured into a magnteically stirred solution of ethylene diamine (8 g, 133 mmol) in DMF (20 mL) with vigorous magnetic stirring at room temperature. Diethyl ether (200 mL) was added to precipitate a solid. The flask was then vortexed and placed in a sonication bath for 15 min.
  • Fmoc aminoxyacetic acid (187 mg, 0.60 mmol) was dissolved in anhydrous DMF (1 mL) to which was added TBTU (243 mg, 0.76 mmol) and DIPEA (95 ⁇ , 0.54 mmol) and magnetically stirred for 3 min.
  • a solution of Boc-Gly 3 NH-(CH 2 ) 2 -NH 2 200 mg, 0.54 mmol) in anhydrous DMF (1 mL) was then added with magnetic stirring.
  • the desired product was isolated by semi-preparative CI 8 HPLC eluting with deionized water containing 0.1% formic acid and a linear gradient of acetonitrile 5 - 95% over 25 min. Product containing fractions were combined and concentrated in vacuo to give 40 mg (0.062 mmol, 51.2% yield) of the desired product (38b) as a white residue. MS (m + H+) found: 643.4, calcd. 643.2; (m-1) found: 641.2, calculated: 641.2.
  • H-Gly 3 -beta-Ala-0-tBu 150 mg, 0.47 mmol
  • 5-azidopentanoic acid 88 mg, 0.62 mmol
  • HOBT 145 mg, 0.95 mmol
  • EDC 270 mg, 1.4 mmol
  • reaction was magnetically stirred for 2 hours then purified on the 27 mm x 220 mm load and lock C18 column 40 mL/min with 220 nmdetection eluting with deionized water containing 0.2% formic acid using 5% acetonitrile for the first 5 min then a linear ramp of 5% to 95% acetonitrile from 5 min to 32 min. Detection 220 nm and Mass spec. Fractions containing pure desired product were combined, frozen and lyophilized to give 70 mg (33% yield) of desired product.
  • FMoc-Gly 3 - MayNMA (37.7mg, 0.036mmol) was dissolved in 20% morpholine in DMSO (5 mL) and magnetically stirred for 1 hour.
  • the reaction was purified using a Combiflash Rf 200i using CI 8 high performance gold 30 g column at 35 mL/min. Eluting with deionized water containing 0.1% formic acid with an acetonitrile gradient 2-95% over 25 min. Fractions containing desired product were combined, frozen and lyophilized to desired product 20 mg ( 75 % yield).
  • FMoc-aminoxy-Gly 3 - MayNMA (2 mg, 18 ⁇ ) was treated with a solution of 20% morpholine in DMSO (1 mL) with magnetic stirring at room temperature for 2 hours.
  • the reaction was purified by semi-preparative CI 8 HPLC using a XB-C18 21.2x150mm, 5 ⁇ column with a flow rate of 21.2 mL/min. Eluting with deionized water containing 0.1% formic acid and 5% acetonitrile for 5 min then a linear gradient of acetonitrile 5% - 95% from 5 min - 25 min.
  • Chloroform-d ⁇ 0.80 (s, 3H), 1.18 - 1.27 (m, 2H), 1.27 - 1.33 (m, 6H), 1.38 - 1.54 (m, 2H),

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention provides linker compounds and cytotoxic compounds that are useful for forming a CBA-drug conjugate and conjugates so formed. Such conjugates and/or cytotoxic compounds may be effective for treating a range of diseases, such as cancer, with a relatively high activity at a relatively low, non-toxic dose.

Description

CONJUGATES COMPRISING CELL-BINDING AGENTS AND CYTOTOXIC
AGENTS
REFERENCE TO RELATED APPLICATION
[01] This application claims the benefit of the filing date, under 35 U.S.C. § 119(e), of U.S. Provisional Application No. 61/770,796, filed on February 28, 2013, the entire contents of which are incorporated herein by reference.
BACKGROUND OF THE INVENTION
[02] Antibody-drug conjugates (ADC) and cell binding agent-drug conjugates are emerging as a powerful class of anti-tumor agents with efficacy across a range of cancers. Cell binding agent-drug conjugates (such as ADCs) are commonly composed of three distinct elements: a cell-binding agent (e.g., an antibody); a linker; and a cytotoxic moiety. The linker component is an important element in developing targeted anti-cancer agents that possess an optimal therapeutic window, i.e., therapeutic activity at a low, non-toxic dose.
[03] Therefore, there is a need for targeted therapies such as ADCs and other cell bind agent-drug conjugates having a new class of linker components.
SUMMARY OF THE INVENTION
[04] A first embodiment of the invention features a conjugate represented by the following formula, or a pharmaceutically acceptable salt thereof:
),
Figure imgf000002_0001
[05] In Formula (Γ), (I) or (V) above, CBA is a cell binding agent; DM is a drug moiety represented by the following formula:
Figure imgf000003_0001
in which R, R' and R", for each occurrence, are independently H or an optionally substituted alkyl; one of Ei and E2 is -C(=0)-, and the other is -NR9-; or one of Ei and E2 is -C(=0)- or - NR9-, and the other is absent; P is [XX] O, in which each XX is a residue of an
independently selected amino acid, or P is -(NRm-CH2CH2)s-, in which s is an integer between 1 and 5 and Rm is H, or alkyl optionally substituted with a charged substituent or an
s1 |-CRbRc-C(=0)— I s2 ionizable group; JCB' in Formula (Γ ) or (I) is
Figure imgf000003_0002
Figure imgf000003_0003
-N-NRe— I s2 s1 ¾=N-NRe-C(=0)— \ s2 N-N Re-C(=0) s1 =N-NRe— Ar s2 s1 j-N-NRe~Ar \ s2 s1 =N-0— \ s2 s1 f-N-O— I s2
Figure imgf000003_0004
Figure imgf000004_0001
, in which s i is the site covalently linked to the CBA, s2 is the site covalently linked to the group X\ in formula (Γ ) or the group (CR3R4)n in Formula (I), JCBI' in s1
Figure imgf000004_0002
e— I s2
Formula (V) is s2 Ra Q9 N-NRe— s2 s1 |— N-NR s1 =N-NRe-C(=0)— \ s2 s1 |-N-N Re-C(=0)— \ s2 s1 |=N-NRe— Ar s2
Figure imgf000004_0003
Figure imgf000005_0001
Figure imgf000005_0002
, in which s 1 is the site covalently linked to the CBA, s2 is the site covalently linked to the group X\ Ar is an optionally substituted arylene or an optionally substituted heteroarylene; Ra, Rb, Rc, Re, Ri to R6, and R9, for each occurrence, are independently H or an optionally substituted alkyl; Cy is the non- alkyne residue of an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne (see below in the second embodiment); R201, R202 and R203 each are independently H or an optionally substituted alkyl (preferably, R201, R202 and R203 are all H;
or R2oi is CH3 and R202 and R203 are both H) ; Z3 is pyridyl or
Figure imgf000005_0003
? wherein R2o4 and R205 are each independently optionally substituted alkyl (preferably, R2o4 and R205 are both methyl or ethyl) and Cy' is the non-alkene residue of an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene (see below in the second embodiment); and R30i is H or optionally substituted alkyl (preferably R 0i is H); m and n, for each occurrence, are independently an integer between 0 and 10; and w is an integer between 1 and 20. In addition, for formula (Γ) or (V), X\ and Z2 are each independently
absent, -S02NR9-, -NR9S02-, -C(=0)-NR9, -NR9-C(=0)-, -C(=0)-0-, -0-C(=0)-, -CH2-0-, - 0-CH2-, -(CH2CH20)p- ,-(OCH2CH2)p-, -NR9-C(=0)-CH2-, or -CH2-C(=0)-NR9-, wherein p and p' are independently an integer from 1 to 1000. Alternatively, for or formula (Γ) or (V), Zi and Z2 are each independently absent, -S02NR9-, -NR9S02-, -C(=0)-NR9, - NR9-C(=0)-, -C(=0)-0-, -0-C(=0)-, -CH2-0-, _-0-CH2-, -(CH2CH20)p-, or -(OCH2CH2)p'-, wherein p and p' are independently an integer from 1 to 1000.
[06] In one embodiment, for formula (Γ) or (I), one of Έ\ and E2 is -C(=0)-, and the other is -NR9-; and the remainder of the variables are as described above.
[07] A second embodiment of the invention features a cytotoxic compound represented by the following formula, or a salt e.g. , a pharmaceutically acceptable salt) thereof:
Figure imgf000006_0001
[08] In Formula (ΙΙΓ) or (III) above, JCB is maleimide (
Figure imgf000006_0002
X'- CRbRc-C(=0)-NRe-, Ra-C(=0)-, Ra-C(=0)-Ar-, NH2-NR6-, NH2-NRe-Ar-, NH2-NRe-C(=0)-
Figure imgf000006_0003
, in which X' is a halogen; c is an optionally substituted cycloalkyne or an optionally
substituted heterocycloalkyne,
Figure imgf000007_0001
an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene, and the remainder of the variables are as defined in the first embodiment.
[09] In Formula (VI), JCBi is Ra-C(=0)-, Ra-C(=0)-Ar-, NH2-NRe-, NH2-NRe-Ar-, NH2-
Figure imgf000007_0002
and the remainder of the variables are as defined for Formula (Γ), (I), or (V) above.
[10] Preferably, for Formula (Γ), (I) or (VI) above,
Figure imgf000007_0003
an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne that can readily react with an azide to form a triazole through copper free click chemistry or can readily react with a tetrazine (see, for example, J. Am. Chem. Soc. (2012) 134:9199-9208; WO 2011/136645; US 2009/0068738, Lang K. et al, J. Am. Chem. Soc. (2012) 134: 10317- 10320, the entire teaching of these references are incorporated herein b its entirety). More preferably,
Figure imgf000007_0004
is cycloocytyne. In another preferred embodiment,
Figure imgf000007_0005
or
Figure imgf000007_0006
[11] A third embodiment of the invention features a linker compound represented by the following formula, or a salt (e.g. , a pharmaceutically acceptable salt) thereof:
R3 4 R5 R6 o (IV),
Figure imgf000008_0001
JcBi-Z! Z2^^ jD
R1 R2 O (VII).
[12] In Formula (IV), (IV) or (VII), JD is -OH or halogen; or -C(=0)-JD is a reactive ester; JCB or JCBI is as defined in the second embodiment above; and the remainder of the variables are as defined in the first embodiment above.
[13] In an alternative embodiment, in Formulae (Γ), (I), (ΙΙΓ), (III), (IV), (IV), (V), (VI) or (VII), Ar is phenylene.
[14] In another alternative embodiment, in Formulae (Γ), (I), (ΙΙΓ), (III), (IV), (IV), (V),
(VI) or (VII), m and n, for each occurrence, are independently an integer between 2 and 5. In yet another alternative embodiment, in Formulae (Γ), (I), (ΙΙΓ), (III), (IV), (IV), (V), (VI) or
(VII) , Ar is phenylene, and m and n, for each occurrence, are independently an integer between 2 and 5. The reminder of variables in each of the alternative embodiments above are as defined in the first, second, or third embodiment. In another alternative, in Formulae (Γ), (ΙΙΓ), (IV), (V), (VI) or (VII), one of X\ and Z2 is absent. In another alternative, in Formulae (Γ), (ΙΙΓ), (IV), (V), (VI) or (VII), one of Zi and Z2 is absent, and the other
is -CH2-O- or -O-CH2-. In yet another alternative, one of X\ and Z2 is absent, and the other is -(CH2CH20)p- or -(OCH2CH2)p'-. Preferably, p or p' is an integer from 2 to 20, more preferably, 2 to 14, even more preferably, 2 to 8. Most preferably, p or p' is 2, 4, 6 or 8. In yet another alternative, in Formulae (Γ), (ΙΙΓ), (IV), (V), (VI) or (VII), Z2 is absent and Zx is -SO2NR9-, -NR9SO2-, -C(=0)NR9- or -NR9C(=0)-. In yet another alternative, in Formulae (Γ), (ΙΙΓ), (IV), (V), (VI) or (VII), Z2 is -0-CH2- and Zi is -CH2-C(=0)-NR9-. Preferably, R9 is H.
[15] Also within the scope of this invention is a composition (e.g. , a pharmaceutical composition) comprising a conjugate represented by Formula (Γ), (I) or (V), a cytotoxic compound represented by Formula (ΙΙΓ), (III) or (VI), or a pharmaceutical acceptable salt thereof. The composition may also include a carrier (e.g. , a pharmaceutically acceptable carrier). The composition can further include a second therapeutic agent.
[16] The present invention also includes a method of inhibiting abnormal cell growth or treating a proliferative disorder, a destructive bone disorder, an autoimmune disorder, a graft versus host disease, a transplant rejection, an immune deficiency, an inflammatory diseases, an infectious disease, a viral disease, a fibrotic disease, a neurodegenerative disorder, pancreatitis, or a kidney disease in a mammal (e.g. , human), comprising administering to said mammal a therapeutically effective amount of a conjugate represented by Formula (Γ), (I) or (V), a cytotoxic compound represented by Formula (ΙΙΓ), (III) or (VI), or a pharmaceutical acceptable salt thereof.
[17] In a specific embodiment, the method described above further comprises
administering to said mammal sequentially or consecutively a second chemotherapeutic agent.
BRIEF DESCRIPTION OF THE DRAWINGS
Figures 1-8 depict synthetic schemes for preparing representative linker compounds or cytotoxic compounds of the present invention.
DETAILED DESCRIPTION OF THE INVENTION
[18] Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulae. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents which may be included within the scope of the present invention as defined by the claims. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention.
[19] It should be understood that any of the embodiments described herein, including those described under different aspects of the invention (e.g. , compounds, conjugates,
compositions, methods of making and using) and different parts of the specification
(including embodiments described only in the Examples) can be combined with one or more other embodiments of the invention, unless explicitly disclaimed or improper. Combination of embodiments are not limited to those specific combinations claimed via the multiple dependent claims. DEFINITIONS
[20] "Alkyl" as used herein refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twenty carbon atoms. "Monovalent" means that alkyl has one point of attachment to the remainder of the molecule. Examples of alkyl groups include, but are not limited to, methyl, ethyl, 1 -propyl, 2-propyl, 1 -butyl, 2-methyl-l -propyl, - CH2CH(CH3)2, 2-butyl, 2-methyl-2-propyl, 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3- methyl-2-butyl, 3-methyl-l -butyl, 2-methyl-l -butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3- dimethyl-2-butyl, 3,3-dimethyl-2-butyl, 1-heptyl, 1-octyl, and the like. Preferably, the alkyl group has one to ten carbon atoms. More preferably, the alkyl group has one to four carbon atoms.
[21] "Alkylene" as used herein refers to a saturated linear or branched-chain divalent hydrocarbon radical of one to twenty carbon atoms, examples of which include, but are not limited to, those having the same core structures of the alkyl groups as exemplified above. "Divalent" means that the alkylene has two points of attachment to the remainder of the molecule. Preferably, the alkylene group has one to ten carbon atoms. More preferably, the alkylene group has one to four carbon atoms.
[22] As used herein, an integer "between" x and y includes integers x and y unless otherwise specified to the contrary. For example, "an integer between 1 and 5" can be 1, 2, 3, 4, or 5.
[23] The terms "cyclic alkyl" and "cycloalkyl" can be used interchangeably. They refer to a monovalent saturated carbocyclic ring radical. "Monovalent" means that cycloalkyl has one point of attachment to the remainder of the molecule. Preferably, the cycloalkyl is 3 to 7 membered monocyclic ring radical. Bicyclic cycloalkyl having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6], or [6,6] system, and bicyclic cycloalkyl having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane.
Bicyclic cycloalkyl also include a spiro cycloalkyl with rings connected through just one atom. Examples of monocyclic cycloalkyl groups include, but are not limited to,
cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. More preferably, the cycloalkyl is cyclohexyl. [24] "Cycloalkylene" as used herein refers to a divalent saturated carbocyclic ring radical having 3 to 12 carbon atoms as a monocyclic ring, or 7 to 12 carbon atoms as a bicyclic ring. "Divalent" means that the cycloalkylene has two points of attachment to the remainder of the molecule. Preferably, the cycloalkylene is a 3- to 7-membered monocyclic. Examples of cyclic alkylene groups include, but not limited to, those having the same core structures of the cylcolakyl groups as exemplified above. More preferably, the cycloalkylene group is cyclohexylene.
[25] "Cycloalkyne" as used herein refers carbocyclic ring having one or more triple bonds. It can be monocyclic, bicyclic or tricyclic; bicyclic and tricyclic can be bridged or fused. The carbocyclic ring optionally contains one or more double bonds and/or is optionally fused with one or more aromatic (e.g., phenyl ring) or heteroaromatic rings.
Examples of cycloalkyne include, but are not limited to, those described in J. Am. Chem. Soc. (2012) 134:9199-9208; WO 2011/136645, US 2009/0068738, Lang K. et al. J. Am. Chem. Soc. (2012) 134: 10317-10320, for example, cyclooctyne, monofluorocyclooctyne, difluorooctyne, DIFO, DIF02, DIFO3, bicylo[6.1.0]non-4-yne, benzocyclooctyne, difluorobenzocyclooctyne, dibenzocyclooctyne, DIBO, and those described in Debets, M. F. et al, Acc. Chem. Res. (2011) 44(9):805-815; and Gold B. et al, J. Am. Chem. Soc. (2013) 135(4): 1558-1569. Preferably, cycloalkyne is cyclooctyne.
[26] "Heterocycloalkyne" as used herein refers to a heterocyclic ring having one or more triple bonds. Examples of heterocycloalkyne include, but are not limited to,
dibenzoazacyclooctyne (DIBAC), biarylazacyclooctynone (BARAC), thiacyclooctyne, thiabenzocyclooctyne, thiacycloheptyne and tetramethylthiacycloheptyne.
[27] "Strained cycloalkene" as used herein refers to carbocyclic ring having one or more double bonds, in which at least one of the double bonds forced by structural constraints has bond angles other than the typical 120° angle of non-strained alkenes. The strained cycloalkenes are more reactive than non-strained alkenes. Examples of strained cycloalkene include, but are not limited to, cyclooctene, norbornene and other cycloalkenes described in Debets, M. F. et al, Acc. Chem. Res., (2011) 44(9):805-815.
[28] "Strained heterocycloalkene" as used herein refers to a heterocyclic ring having one or more double bonds, in which at least one of the double bonds forced by structural constraints has bond angles other than the typical 120° angle of non-strained heterocycloalkenes. The strained heterocycloalkenes are more reactive than non-strained heterocycloalkenes.
[29] The term "aryl group" means an aromatic hydrocarbon ring system having six to fourteen carbon ring atoms. The term "aryl" may be used interchangeably with the terms "aryl ring" "aromatic ring," "aryl group" and "aromatic group". An aryl group typically has six to fourteen ring atoms. "Aryl group" also includes an aromatic hydrocarbon ring system fused to a non-aromatic carbocyclic ring system, such as a cycloalkyl group. Examples includes phenyl, naphthyl, anthracenyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, fluorenyl, indanyl, indenyl and the like. An aryl group is monovalent, i.e., has one point of attachment to the remainder of the molecule. A "substituted aryl group" is substituted at any one or more substitutable ring atom, which is a ring carbon atom bonded to a hydrogen. " [30] "Arylene" as used herein refers to a divalent aryl group, i.e., an aryl group having two points of attachment to the remainder of the molecule. "Divalent" means that the arylene has two points of attachment to the remainder of the molecule. Both aryl and arylene groups are sometime represented herein by "Ar." Arylene is preferably phenylene.
[31] Heteroaryl (used interchangeably with "heteroaromatic," "heteroaryl ring,"
"heteroaryl group," "heteroaromatic ring," and "heteroaromatic group") refers to aromatic ring systems having five to fourteen ring atoms selected from carbon and at least one
(typically 1 to 4, more typically 1 or 2) heteroatoms (e.g. , oxygen, nitrogen or sulfur).
"Heteroaryl" includes monocyclic rings and polycyclic rings (e.g. , bicyclic) in which a monocyclic heteroaromatic ring is fused to one or more other aromatic or heteroaromatic rings. As such, "5-14 membered heteroaryl" includes monocyclic, bicyclic or tricyclic ring systems. Heteroaryls are monovalent, meaning that there is one point of attachment to the remainder of the molecule.
[32] "Monocyclic 5-6 membered heteroaryl" means a monocyclic aromatic ring system having five or six ring atoms selected from carbon and at least one (typically 1 to 3, more typically 1 or 2) heteroatoms (e.g. , oxygen, nitrogen or sulfur). Examples of monocyclic 5-6 membered heteroaryl groups include furanyl (e.g. , 2-furanyl, 3-furanyl), imidazolyl (e.g. , N- imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl), isoxazolyl (e.g. , 3-isoxazolyl, 4- isoxazolyl, 5-isoxazolyl), oxadiazolyl (e.g. , 2-oxadiazolyl, 5-oxadiazolyl), oxazolyl (e.g. , 2- oxazolyl, 4-oxazolyl, 5-oxazolyl), pyrazolyl (e.g. , 3-pyrazolyl, 4-pyrazolyl), pyrrolyl (e.g. , 1- pyrrolyl, 2-pyrrolyl, 3-pyrrolyl), pyridyl (e.g. , 2-pyridyl, 3-pyridyl, 4-pyridyl), pyrimidinyl
(e.g. , 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl), pyridazinyl (e.g. , 3-pyridazinyl), thiazolyl (e.g. , 2-thiazolyl, 4-thiazolyl, 5-thiazolyl), isothiazolyl, triazolyl (e.g. , 2-triazolyl, 5-triazolyl), tetrazolyl (e.g. , tetrazolyl), and thienyl (e.g. , 2-thienyl, 3-thienyl). Examples of polycyclic aromatic heteroaryl groups include carbazolyl, benzimidazolyl, benzothienyl, benzofuranyl, isobenzofuranyl, indolyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, indazolyl, isoindolyl, acridinyl, or benzisoxazolyl. A "substituted heteroaryl group" is substituted at any one or more substitutable ring atom, which is a ring carbon or ring nitrogen atom bonded to a hydrogen.
[33] "Heteroarylene" as used herein refers to a divalent heteroaryl, i.e., a heteroaryl with two points of attachment to the remainder of the molecule.
[34] The terms "heterocycle," "heterocyclyl," heterocyclic and "heterocyclic ring" are used interchangeably herein and refer to a saturated or unsaturated non-aromatic 3-12 membered ring radical optionally containing one or more double bonds. It can be
monocyclic, bicyclic, tricyclic, or fused. The heterocycle contains 1 to 4 heteroatoms, which may be the same or different, selected from N, O or S. The heterocycle optionally contains one or more double bonds and/or is optionally fused with one or more aromatic (e.g. , phenyl ring) or heteroaromatic rings. "3-7 membered monocyclic heterocycle" means a radical having from 3-7 atoms (including 1-3 heteroatoms) arranged in a monocyclic ring. The term "heterocycle" is intended to include all the possible isomeric forms. A heterocycle may be a monocycle having 3 to 7 ring members (e.g. , 2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (e.g. , 4 to 9 carbon atoms and 1 to 6 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. Heterocycles are described in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W. A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; "The Chemistry of Heterocyclic Compounds, A series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and /. Am. Chem. Soc. (1960) 82:5566.
[35] Examples of heterocyclic rings include, but are not limited to, aziridinyl, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, isoindolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyco[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, and azabicyclo[2.2.2]hexanyl. Spiro moieties are also included within the scope of this definition. Examples of a heterocyclic group wherein ring atoms are substituted with oxo (=0) moieties are pyrimidinonyl and 1,1-dioxo-thiomorpholinyl.
[36] The heterocycle, heteroaryl, or heteroarylene groups may be carbon (carbon-linked) or nitrogen (nitrogen-linked) attached where such is possible. By way of example and not limitation, carbon bonded heterocycle, heteroaryl or heroarylene groups are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
[37] By way of example and not limitation, nitrogen bonded heterocycle, heteroaryl, or heteroarylene groups are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, IH-indazole, position 2 of a isoindole or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or O-carboline.
[38] The heteroatoms present in heteroaryl, heteroarylene, or heterocyclcyl can include the oxidized forms such as NO, SO, and S02.
[39] "Halogen" refers to F, CI, Br or I.
[40] If a group is described as being "optionally substituted," the group may be either (1) not substituted, or (2) substituted. If a carbon of a group is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogen atoms on the carbon (to the extent there are any) may separately and/or together be replaced with an independently selected optional substituent.
[41] Suitable substituents for an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, heterocycle, alkylene, alkenylene, alkynylene, cycloalkene, heterocycloalkene, cycloalkyne, heterocycloalkyne, cycloalkylene arylene, and heterarylene are those which do not significantly adversely affect the biological activity of the conjugate. Unless otherwise specified, exemplary substituents for these groups include linear, branched or cyclic alkyl, alkenyl or alkynyl having from 1 to 10 carbon atoms, aryl, heteroaryl, heterocyclyl, halogen, guanidinium [-NH(C=NH)NH2], -ORioo, NRi0iRi02, -N02, -NR101COR102, -SR100, a sulfoxide represented by -SORioi, a sulfone represented by -S02Rioi, a sulfonate -SO3M, a sulfate - OSO3M, a sulfonamide represented by -S02NR1oiRio2, cyano, an azido, -CORioi, -OCOR101, -OCONR1oiRio2 and a polyethylene glycol unit (-OCH2CH2)nR1oi wherein M is H or a cation (such as Na+ or K+); R101, Rio2 and R103 are each independently selected from H, linear, branched or cyclic alkyl, alkenyl or alkynyl having from 1 to 10 carbon atoms, a polyethylene glycol unit (-OCH2CH2)n-Rio4, wherein n is an integer from 1 to 24, an aryl having from 6 to 10 carbon atoms, a heterocyclic ring having from 3 to 10 carbon atoms and a heteroaryl having 5 to 10 carbon atoms; and R104 is H or a linear or branched alkyl having 1 to 4 carbon atoms, wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclcyl in the groups represented by R10o, R101, R102, Rio3 and Rio4 are optionally substituted with one or more (e.g., 2, 3, 4, 5, 6 or more) substituents independently selected from halogen, -OH, -CN, - N02, and unsubstituted linear or branched alkyl having 1 to 4 carbon atoms. Preferably, the substituent for the optionally substituted alkyl, alkylene, cycloalkylene, arylene, and heteroarylene described above is selected from the group consisting of halogen, -CN, - NR10iRio2, -CF3, -OR100, aryl, heteroaryl, heterocyclyl, -SR101, -SOR101, -SO2R101, and -S03M. Alternatively, the suitable substituent is selected from the group consisting of - halogen, -OH, -N02, -CN, Ci_4 alkyl, -OR100, NRioiRio2, -NRioiCORi02, -SR100, -SO2R101, - S02NR10iRio2, -CORioi, -OCOR10i, and -OCONR10iRio2, wherein R100, R101, and R102 are each independently -H or C1-4 alkyl.
[42] "Alkenyl" as used herein refers to aliphatic linear or branched-chain monovalent hydrocarbon radical of two to twenty carbon atoms with at least one carbon-carbon double bond, wherein the alkenyl radical includes radicals having "cis" and "trans" orientations, or by an alternative nomenclature, "E" and "Z" orientations. "Monovalent" means that alkenyl has one point of attachment to the remainder of the molecule. Examples include, but are not limited to, ethylenyl or vinyl (-CH=CH2), allyl (-CH2CH=CH2), and the like. Preferably, the alkenyl has two to ten carbon atoms, also referred to as "C2-1o alkenyl." More preferably, the alkyl has two to four carbon atoms, also referred to as "C2_4 alkenyl."
[43] "Alkynyl" as used herein refers to aliphatic linear or branched-chain monovalent hydrocarbon radical of two to twenty carbon atoms with at least one carbon-carbon triple bond. "Monovalent" means that alkynyl has one point of attachment to the remainder of the molecule. Examples include, but are not limited to ethynyl, propynyl, 1-butynyl, 2-butynyl,
1-pentynyl, 2-pentynyl, 3-pentynyl, hexynyl, and the like. Preferably, the alkynyl has two to ten carbon atoms, also referred to as "C2-io alkynyl." More preferably, the alkynyl has two to four carbon atoms, also referred to as "C2_4 alkynyl."
[44] The term "ionizable group" refers to a functional group that can be converted to a charged group by protonation with an acid or deprotonation with a base. Examples of the ionizable groups include -S03H, -Z'-S03H, -OP03H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, - C02H, -Z'C02H, -NR11R12, or -Z'-NRuRi2, R11 and R12, for each occurrence, are independently H or an optionally substituted alkyl; and Z' includes an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene. In certain embodiments, Z' is alkylene.
[45] The term "charged substituent" refers to a substituent that is either positively or negatively charged. The charge in such a substituent is not removable by treatment with a base or an acid and thus permanent. Examples of the charged substituents include, but not limited to, -N+R13R14R15 and -Z'-N+R13R14R15, in which R1 to R15 are each independently an optionally substituted alkyl; and Z' includes an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene. In certain embodiments, Z' is alkylene.
[46] Charged substituents may contain a counterion. For positive charged substituents, the counterion is negative and can be represented by "X" , e.g. , as -N+R13R14Ri5X~ and -Z'-
N+R13R14Ri5X". The counter ions for the positively charged substituents are anions
(preferably pharmaceutically acceptable anions), which include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, bromide, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, pamoate, pantothenate,
phosphate/diphospate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate, and triethiodide. Preferably, the counter ions for the positively charged substituents are chloride, bromide, sulfate, and phosphate.
[47] The counter ions for the negatively charged substituents include, but are not limited to, an alkali metal ion (e.g. , sodium and potassium), an alkaline earth metal ion (e.g. , calcium and magnesium), aluminum ion, ammonium, protonated trialkyl amines (e.g. , trimethylamine and triethylamine), a tetraalkyl ammonium (e.g. , tetra methyl ammonium, and tetrabutyl ammonium), and a protonated heteroaromatic group (e.g. , pyridine, pyrimidine, triazines, tetrazines). Preferably, the counter ions for the negatively charged substituents are sodium, potassium, lithium, protonated triethyl amine, protonated pyridiene. Most preferably, the counter ions for the negatively charged substituents are sodium and potassium. The counter ions for both the negatively and positively charged substituents may be removed or replaced in subsequent purification steps.
[48] "Amino acid" as used herein, including the residue represented by variable "XX" described above, refers to naturally occurring amino acids, unnatural amino acids, amino acid analogs, or amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
[49] The term "Naturally occurring amino acids" as used herein refers to those twenty L-amino acids encoded by the universal genetic codes and appearing in proteins or peptides, as well as selenocysteine and pyrrolysine that are incorporated into proteins by distinctive biosynthetic mechanisms. They include Histidine, Alanine, Isoleucine, Arginine, Leucine, Asparagine, Lysine, Aspartic acid, Methionine, Cysteine, Phenylalanine, Glutamic acid, Threonine, Glutamine, Tryptophan, Glycine, Valine, Proline, Serine, Tyrosine,
selenocysteine and pyrrolysine. The term "Naturally occurring amino acids" also refers to those produced by the body, but are not encoded by the universal genetic codes, such as β- Alanine, Ornithine, and citrulline. The term "Naturally occurring amino acids" further includes those naturally occurring L-amino acids that are later modified (e.g. , via post- translational modification by enzymes) in the body, such as hydroxyproline, γ- carboxyglutamate, and O-phospho serine.
[50] The term "unnatural amino acids" as used herein is intended to include the "D" stereochemical form of the naturally occurring amino acids described above. It is further understood that the term "unnatural amino acids" includes homologues of the natural L- amino acids or their D isomers, and synthetically modified forms of the natural L-amino acids or their D isomers. The synthetically modified forms include, but are not limited to, amino acids having side chains shortened or lengthened by up to two carbon atoms, amino acids comprising optionally substituted aryl groups, and amino acids comprised halogenated groups, preferably halogenated alkyl and aryl groups and also N substituted amino acids e.g.
N-methyl-Histidine, N-methyl-Alanine, N-methyl-Isoleucine, N-methyl-Arginine, N-methyl-
Leucine, N-methyl-Asparagine, N-methyl-Lysine, N-methyl-Aspartic acid, N-methyl-
Methionine, N-methyl-Cysteine, N-methyl-Phenylalanine, N-methyl-Glutamic acid, N- methyl-Threonine, N-methyl-Glutamine, N-methyl-Tryptophan, N-methyl-Glycine, N- methyl- Valine, N-methyl-Proline, N-methyl-Serine, N-methyl-Tyrosine, N-methyl- selenocysteine, and N-methyl-pyrrolysine, each including an L or D isomer.
[51] The term "amino acid analogs" as used herein refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group. They include 3- aminoalanine, 3-dimethylaminoalanine, 2-amino-4-(dimethylamino)butanoic acid, 2,4- diaminobutanoic acid, 2-amino-6-(dimethylamino)hexanoic acid, 2-amino-5- (dimethylamino)pentanoic acid, homoserine, norleucine, cysteine sulfonic acid, cysteine sulfinic acid, methionine sulfoxide, and methionine methyl sulfonium. Such analogs may have modified R groups {e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid analogs also include D isomers of the above-referenced L-isomers.
[52] The term "amino acid mimetics" as used herein refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but functions in a manner similar to a naturally occurring amino acid.
[53] "Peptide" as used herein, including the peptide represented by variable "P" when the variable is [XX]2_10, refers to short polymers of amino acid monomers linked by peptide bonds, the covalent chemical bonds formed between two amino acid monomers when the carboxyl group of N-terminal monomer reacts with the amino group of the C-terminal monomer. The preferred length of the peptide is two to ten amino acids as described above.
[54] Any one of the peptides described herein can be connected to the rest of the molecules in either direction. For example, when AA1, AA2, and AA3 each represent an amino acid (naturally-occurring, or unnatural), "AA1-AA2-AA3 in either direction" refers to the tripeptide N-AA1-AA2-AA3-C), and the tripeptide N-AA3-AA2-AA1-C).
[55] In certain embodiments, variable P is an amino acid represented by [XX] \ or a peptide represented by [XX]2-1o, in which each XX is the residue of an independently selected amino acid selected from the group consisting of: Histidine, Alanine, Isoleucine, Arginine, Leucine,
Asparagine, Lysine, Aspartic acid, Methionine, Cysteine, Phenylalanine, Glutamic acid,
Threonine, Glutamine, Tryptophan, Glycine, Valine, Proline, Serine, Tyrosine,
selenocysteine, and pyrrolysine, N-methyl-Histidine, N-methyl-Alanine, N-methyl-
Isoleucine, N-methyl-Arginine, N-methyl-Leucine, N-methyl-Asparagine, N-methyl-Lysine,
N-methyl-Aspartic acid, N-methyl-Methionine, N-methyl-Cysteine, N-methyl-Phenylalanine,
N-methyl-Glutamic acid, N-methyl-Threonine, N-methyl-Glutamine, N-methyl-Tryptophan, N-methyl-Glycine, N-methyl- Valine, N-methyl-Proline, N-methyl-Serine, N-methyl- Tyrosine, N-methyl-selenocysteine, N-methyl-pyrrolysine, hydroxyproline, γ- carboxyglutamate, selinocysteine, O-phosphoserine, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, citrulline, Ornithine, cysteine sulfonic acid, cysteine sulfinic acid, 3-aminoalanine, 3-dimethylaminoalanine, 2-amino-4-(dimethylamino)butanoic acid, 2,4-diaminobutanoic acid, 2-amino-6-(dimethylamino)hexanoic acid, 2-amino-5- (dimethylamino)pentanoic acid, and β-alanine, each independently as an L or D isomer. In another alternative, each XX is the residue of an independently selected amino acid selected from glycine or alanine, each independently as an L or D isomer.
[56] In certain embodiments, the peptide represented by variable "P" is cleavable by a protease. In one embodiment, the protease is a protease expressed in tumor tissue. In another embodiment, the protease is a lysosomal protease.
[57] The term "peptide cleavable by a protease" as used herein refers to peptides containing a cleavage recognition sequence of a protease. A cleavage recognition sequence for a protease is an amino acid sequence recognized by the protease during proteolytic cleavage. Many protease cleavage sites are known in the art, and these and other cleavage sites can be included in the linker moiety. See, e.g. , Matayoshi et al. Science 247:954 (1990); Dunn et al. Meth. Enzymol. 241 :254 (1994); Seidah et al. Meth. Enzymol. 244: 175 (1994); Thornberry, Meth. Enzymol. 244:615 (1994); Weber et al. Meth. Enzymol. 244:595 (1994); Smith et al. Meth. Enzymol. 244:412 (1994); Bouvier et al. Meth. Enzymol. 248:614 (1995), Hardy et al. in AMYLOID PROTEIN PRECURSOR IN DEVELOPMENT, AGING, AND ALZHEIMER 'S DISEASE, Ed. Masters et al. pp. 190-198 (1994).
[58] In one embodiment, the peptide sequence is chosen based on its ability to be cleaved by a tumor- associated protease, e.g. , a protease that is found on the surface of a cancerous cell or extracellularly in the vicinity of tumor cells. The examples of such proteases include thimet oligopeptidase (TOP), CD10 (neprilysin), a matrix metalloprotease (such as MMP2 or MMP9), a type II transmebrane serine protease (such as Hepsin, testisin, TMPRSS4 or matriptase/MT-SPl), legumain and enzymes described in the following reference: Current Topics in Developmental Biology: Cell Surface Proteases, vol. 54 Zucker S . 2003, Boston, MA. The ability of a peptide to be cleaved by tumor-associated protease can be tested using in vitro protease cleavage assays known in the art.
[59] In another embodiment, the peptide sequence is chosen based on its ability to be cleaved by a lysosomal protease, which include cathepsins B, C, D, H, L and S, and furin. Preferably, the peptide sequence is capable of being cleaved by an appropriate isolated protease in vitro, which can be tested using in vitro protease cleavage assays known in the art.
[60] In certain embodiments, the peptide is selected from the group consisting of Val-Cit, Val-Lys, Phe-Lys, Lys-Lys, Ala-Lys, Phe-Cit, Leu-Cit, Lle-Cit, Trp, Cit, Phe-Ala, Phe-N9- tosyl-Arg, Phe-N9-nitro-Arg, Phe-Phe-Lys, D-Phe-Phe-Lys, Gly-Phe-Lys, Leu-Ala-Leu, Ile- Ala-Leu, Val-Ala-Val, Ala-Leu- Ala-Leu, β-Ala-Leu- Ala-Leu, Gly-Phe-Leu-Gly, Val-Arg, Arg-Val, Arg-Arg, Val-D-Cit, Val-D-Lys, Val-D-Arg, D- Val-Cit, D-Val-Lys, D-Val-Arg, D- Val-D-Cit, D- Val-D-Lys, D- Val-D-Arg, D-Arg-D-Arg, Ala- Ala, Ala-D-Ala, D- Ala- Ala, and D-Ala-D-Ala, Gly-Gly-Gly, Ala- Ala- Ala, D- Ala- Ala- Ala, Ala-D- Ala-Ala, Ala-Ala-D-Ala, Ala- Val-Cit, and Ala-Val-Ala. In another alternative, the peptide is selected from the group consisting of Gly-Gly-Gly, Ala- Ala- Ala, D- Ala- Ala- Ala, Ala-D- Ala- Ala, and Ala-Val-Ala. In a related embodiment, any of the peptide sequences herein above may be in either direction, as defined above.
[61] An amino acid or peptide can be attached to a linker/spacer or a cell binding agent through the terminal amine or terminal carboxylic acid of the amino acid or peptide. The amino acid can also be attached to a linker/spacer or a cell-binding agent through a side chain reactive group, including, but not restricted to, the thiol group of cysteine, the epsilon amine of lysine, and the side chain hydroxyls of serine or threonine.
[62] Amino acids and peptides may be protected by blocking groups. A blocking group is an atom or a chemical moiety that protects an amino acid or a peptide (e.g., the N-terminus) from undesired reactions and can be used during the synthesis of a drug-ligand conjugate. It should remain attached to the amino acid or peptide (e.g., to the N-terminus) throughout the synthesis, and may be removed after completion of synthesis of the drug conjugate by chemical or other conditions that selectively achieve its removal. The blocking groups suitable for N-terminus protection are well known in the art of peptide chemistry. Exemplary blocking groups include, but are not limited to, methyl esters, tert-butyl esters, 9- fluorenylmethyl carbamate (Fmoc) and carbobenzoxy (Cbz).
[63] The term "blocking group" or "protecting group" refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound, a derivative thereof, or a conjugate thereof. For example, an "amine-protecting group" is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Such groups are well known in the art (see for example P. Wuts and T. Greene, 2007, Protective Groups in Organic Synthesis, Chapter 7, J. Wiley & Sons, NJ) and exemplified by carbamates such as methyl and ethyl carbamate, FMOC, substituted ethyl carbamates, carbamates cleaved by 1,6-β- elimination (also termed "self immolative"), ureas, amides, peptides, alkyl and aryl derivatives. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t- butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethylenoxycarbonyl (Fmoc). For a general description of protecting groups and their use, see P. G.M. Wuts & T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 2007.
[64] The term "reactive ester" as used herein refers to an ester group having a leaving group that is readily displaced by an amine group. Examples of a reactive ester, include, but are not limited to, N-hydroxysuccinimide ester, N-hydroxysulfosuccinimide ester, nitrophenyl {e.g., 2 or 4-nitrophenyl) ester, dinitrophenyl {e.g., 2,4-dinitrophenyl) ester, sulfo- tetrafluorophenyl {e.g., 4-sulfo-2,3,5,6-tetrafluorophenyl)ester and pentafluorophenyl ester.
[65] As used herein, the terms "DARPin" and "(designed) ankyrin repeat protein" are used interchangeably to refer to certain genetically engineered antibody mimetic proteins typically exhibiting preferential (sometimes specific) target binding. The target may be protein, carbohydrate, or other chemical entities, and the binding affinity can be quite high. The DARPins may be derived from natural ankyrin repeat-containing proteins, and preferably consist of at least three, usually four or five ankyrin repeat motifs (typically about 33 residues in each ankyrin repeat motif) of these proteins. In certain embodiments, a DARPin contains about four- or five-repeats, and may have a molecular mass of about 14 or 18 kDa, respectively. Libraries of DARPins with randomized potential target interaction residues with diversities of over 10 12 variants can be generated at the DNA level, for use in selecting
DARPins that bind desired targets {e.g., acting as receptor agonists or antagonists, inverse agonists, enzyme inhibitors, or simple target protein binders) with picomolar affinity and specificity, using a variety of technologies such as ribosome display or signal recognition particle (SRP) phage display. See, for example, U.S. Patent Publication Nos. 20040132028,
20090082274, 20110118146, and 20110224100, WO 02/20565 and WO 06/083275(the entire teachings of which are incorporated herein by reference), and also see C. Zahnd et al. 2010,
Cancer Res., 70; 1595-1605; Zahnd et al, 2006, /. Biol. Chem., 281, 46, 35167-35175; and
Binz, H.K., Amstutz, P. & Pluckthun, A. 2005, Nature Biotechnology, 23, 1257-1268 (all incorporated herein by reference). Also see U.S. Patent Publication No. 2007/0238667; U.S.
Patent No. 7,101,675; WO 2007/147213; and WO 2007/062466 (the entire teachings of which are incorporated herein by reference), for the related ankyrin-like repeats protein or synthetic peptide.As used herein, "AVIBODY cell binding agents (or "AVIBODY CBA" in short) includes a family of proteins as cell binding agents that specifically bind desired targets. As is well known, antibodies bind such desired targets through "Target Binding Regions" (TBRs) or Fv domains. AVIBODY™ CBA typically contains two, three, or four TBRs more commonly known as Dia-, Tria- and Tetra-bodies. These TBRs / Fv domains are linked together by fusing the Fv V-domains together in a "head to tail" orientation, forming stable, specific, and highly customizable multimeric antibody-like proteins as AVIBODY™ CBA. See, for example, U.S. Publication Nos. 20080152586 and 20120171115 for details, the entire teachings of which are incorporated herein by reference.
[66] The term "cell binding agent" as used herein refers to a compound that can bind a cell (e.g., on a cell-surface ligand) or bind a ligand associated with or proximate to the cell, either in a specific or non-specific manner. In certain embodiments, binding to the cell or a ligand on or near the cell is specific. The cell-binding agent may be of any kind presently known, or that become known and includes peptides and non-pep tides.
[67] In certain embodiments, the cell-binding agents are proteins or polypeptides, or compounds comprising proteins or polypeptides, including both antibody and non-antibody proteins or polypeptides. Preferably, the cell-binding agents (e.g., proteins or polypeptides) comprise one or more Cys residues. The side chain -SH group of the Cys residues may be intact, or may be in a disulfide bond that can be reduced. Preferably, reduction of the disulfide bond(s) does not significantly negatively impact the cell-binding function of the proteins or polypeptides (e.g., in the case of antibody or antigen-binding portion thereof, reduction of the disulfide bonds does not substantially increase the dissociation of light chains / heavy chains). Alternatively or in addition, the cell-binding agents (e.g., proteins or polypeptides) comprise one or more amino acids that are modified to contain a reactive functional group that can react with the linkers (e.g., Formula (IV)) or cytotoxic compounds
(e.g., Formula (III)) of the invention. For example, the reactive functional group
is -SH, -C(=0)-, -NHNH2, -N3, -alkyne, tetrazine, strained trans-cycloalkene or strained trans-heterocycloalkene, dithioester, or diene (see, for example, Vu Hong et al. Bioconjugate
Chem. (2010) 21(10): 1912-1916; Glassner, M. et al, J. Am. Chem. Soc. (2012) 134:7274-
7277; Hansell, C. F. et al., J. Am. Chem. Soc. (2011) 133: 13828-13831; Neal K. Devaraj,
Synlett (2012) 23(15):2147-2152; Chenoweth, K. et al., Organic & Biomolecular Chemistry
(2009) 7(24):5255; Jewett, J.C. et al., Journal of the American Chemical Society (2010)
132(11):3688-3690; Seitchik J.L. et al., J. Am. Chem. Soc. (2012) 134(6):2898-2901; and Sletten E.M. et al, Angew Chem. Int. Ed. Engl. (2009) 48(38):6974-6998). The reactive functional group can be introduced into the cell-binding agent through any chemical or enzymatic method known in the art. See, for example, Davis L.K. et al,. J. Am. Chem. Soc. (2012) 134: 10317- 10320; Boeggeman E. et al,. Bioconjug Chem. (2009 Jun) 20(6): 1228-36; Stan, AC et al, Cancer Res. (1999 Jan 1) 59(1): 115-21 ; Mahal, L. K. et al, Science (1997) 276: 1125-1128; Saxon, E. and Bertozzi, C. R., Science (2000) 287:2007-2010; Hang, H. C. et al, Proc. Natl. Acad. Sci. USA (2003) 100: 14846-14851 ; Vocadlo, D. J. et al . Proc. Natl. Acad. Sci. USA (2003) 100:9116-9121 ; Prescher, J. A. et al, Nature (2004) 430:873-877; Dube, D. H. et al, (2006) Proc. Natl. Acad. Sci. USA 103:4819-4824; Jeger S, et al. Angew Chem Int Ed Engl.( 2010 Dec 17) 49(51):9995-7; and Lang K. et al. J. Am. Chem. Soc.
(2012) 134: 10317- 10320.
[68] Cell-binding agent can also peptides derived from phage display (see, for example,
Wang et al. Proc. Natl. Acad. Sci. USA (2011) 108(17):6909-6914) or peptide library techniques (see, for example, Dane et al, Mol. Cancer. Ther. (2009) 8(5): 1312- 1318).
[69] In one embodiment, one or more reactive functional groups that are capable of reacting with the linkers (Formula (IV)) or the cytotoxic compounds (Formula (III)) of the present invention can be introduced into the cell-binding agent by any methods known in the art. For example, a terminal amine group on the cell-binding agent can be converted to a carbonyl group through transamination reaction (see, for example, US 2010/0099649; Angew.
Chem. Int. Ed. (2006) 45(32):5307; Chem. Biol. (2001) 2(4):247; /. Am. Chem. Soc. (2008)
130(35): 11762.) Alternatively, the cell-binding agent can be engineered to include one or more free cysteine residues (i.e., cysteine residues having a free -SH group that can react with the linkers or the cytotoxic compounds of the present invention) according to any methods known in the art (see, for example, US 7,521,541). In another alternative, thiol groups (-SH) can be generated by controlled reduction of interchain disulfides of antibodies, followed by treatment with a cytotoxic agent bearing a maleimido group, as described in US patents 7,659,241, 8,309,300; 7,855,275; 7,723,485 and 7,521,541. For example, partial or complete reduction of interchain disulfides followed by conjugation with a cytotoxic agent bearing a maleimido group can yield a conjugate with 2, 3, 4, 5, 6, 7 or 8 cytotoxic agent molecules covalently linked to each antibody molecule. Alternatively, conjugates having 2,
3, 4, 5, 6, particularly 4 or 6, cytotoxic agent molecules covalently attached to each antibody molecule can be prepared by complete reduction of interchain disulfide of the antibody followed by partial re-oxidation and then conjugation with cytotoxic agent. In another alternative, these conjugates can be prepared by partial or complete reduction of interchain disulfides of the antibody followed by conjugation with cytotoxic agent and then partial re- oxidation. Thiol groups can also be introduced into the cell-binding agent (e.g. , antibodies) by reaction with a crosslinking agent such as 2-iminothiolane (see for example Goff and Carroll, Bioconjugate Chem. , (1990) 1(6):381— 386) followed by reaction with a cytotoxic agent bearing a maleimido group (e.g. , compounds of Formula (III)) to provide a conjugate. All these methods for introducing reactive functional groups are applicable for cell-binding agents that are not antibodies, which, for example, include centyrin, Darpin, Avibody, adnectin or antibody fragment, such as minibodies, diabodies, tribodies, tetrabodies, nanobodies, probodies, domain bodies or unibodies.
[70] In one embodiment, when the cell-binding agent is a centyrin, one or more reactive functional groups (e.g. , a cysteine having a free thiol group) can be introduced according to methods described in US 2010/0255056, US 2010/0216708 and US 2011/0274623.
[71] In another embodiment, the cell-binding agent is a Darpin and it can be prepared according to methods described in US Publication Nos. 2004/0132028, 2009/0082274, 2011/0118146, and 2011/0224100, WO 02/20565 and WO 06/083275. Preferably, Darpin comprises one or more cysteine residues at specific positions that do not interfere with antigen binding. Such cysteine residue can react with the linkers (e.g. , Formula (IV)) or the cytotoxic compounds (e.g. , Formula (III)) of the present invention.
[72] In yet another embodiment, Avibodies having one or more cysteine residues can be prepared according to methods described in US 2008/0139791 and US 2012/0171115.
[73] The Cys side chain -SH groups or the other reactive functional groups described above may be covalently linked to the linkers (e.g. , linkers of Formula (IV)), which are in turn linked to the cytotoxic compounds, thus conjugating the cell-binding agents to the cytotoxic compounds to yield the conjugates of the invention (e.g. , conjugates of Formula (I)). Alternatively, the Cys side chain -SH groups or the reactive functional groups may be covalently linked to the cytotoxic compounds of the invention having linkers bound thereto (e.g. , cytotoxic compounds of Formula (III)). Each protein-based cell-binding agents may contain multiple Cys side chain -SH groups and/or the reactive functional groups available for linking the compounds of the invention.
[74] Examples of the cell binding agents include an antibody, a single chain antibody, an antibody fragment that specifically binds to the target cell, a monoclonal antibody, a single chain monoclonal antibody, a monoclonal antibody fragment that specifically binds to a target cell, a chimeric antibody, a chimeric antibody fragment that specifically binds to the target cell, a bispecific antibody, a domain antibody, a domain antibody fragment that specifically binds to the target cell, an interferon, a lymphokine (e.g., IL-2, IL-3, IL-4, and IL-6), a hormone (e.g., insulin, thyrotropin releasing hormone, melanocyte-stimulating hormone, and a steroid hormone (e.g., androgen and estrogen)), a vitamin (e.g., folate), a growth factor (e.g., EGF, TGF-alpha, FGF, VEGF), a colony stimulating factor, a nutrient- transport molecule (e.g., transferrin; see O'Keefe et al., J. Biol. Chem. (1985) 260:932-937, incorporated herein by reference), a centyrin (a protein scaffold based on a consensus sequence of fibronectin type III (FN3) repeats; see U.S. Patent Publication Nos.
2010/0255056, 2010/0216708 and 2011/0274623 incorporated herein by reference), an Ankyrin Repeat Protein (e.g., a designed ankyrin repeat protein, known as DARPin; see U.S. Patent Publication Nos. 2004/0132028, 2009/0082274, 2011/0118146, and 2011/0224100, incorporated herein by reference, and also see C. Zahnd et al. (2010) Cancer Res., 70: 1595- 1605; Zahnd et al, J. Biol. Chem.(2006) 281(46):35167-35175; and Binz, H.K., Amstutz, P. & Pluckthun, A., Nature Biotechnology (2005) 23: 1257-1268, incorporated herein by reference), an ankyrin-like repeats protein or synthetic peptide (see e.g., U.S. Patent
Publication No. 2007/0238667; U.S. Patent No. 7,101,675; WO 2007/147213; and
WO 2007/062466, incorporated herein by reference), an Adnectin (a fibronectin domain scaffold protein; see US Patent Publication Nos. 2007/0082365; 2008/0139791, incorporated herein by reference), Avibody (including diabodies, triabodies, and tetrabodies; see U.S. Publication Nos. 2008/0152586 and 2012/0171115), and other cell-binding molecules or substances.
[75] In certain embodiments, the cell-binding agent is an antibody, a single chain antibody, an antibody fragment that specifically binds to the target cell, a monoclonal antibody, a single chain monoclonal antibody, a monoclonal antibody fragment that specifically binds to a target cell, a chimeric antibody, a chimeric antibody fragment that specifically binds to the target cell, a domain antibody, a domain antibody fragment that specifically binds to the target cell, a lymphokine, a hormone, a vitamin, a growth factor, a colony stimulating factor, or a nutrient-transport molecule. Alternatively, the cell-binding agent is a monoclonal antibody, a single chain monoclonal antibody, or a monoclonal antibody fragment that specifically binds to a target cell.
[76] In certain embodiments, the cell-binding agent is a bispecific antibody, an ankyrin repeat protein, a Centyrin, or an Avibody. [77] "Antibody fragment" refers to Fab, Fab', and F(ab')2, Fv, minibodies, diabodies, tribodies, tetrabodies, nanobodies, probodies, domain bodies, unibodies, and the like (Parham, J. Immunol. 131:2895-2902 (1983); Spring et al. J. Immunol. 113:470-478 (1974); Nisonoff et al. Arch. Biochem. Biophys. 89:230-244 (1960); Kim et al., Mol, Cancer Ther., 7:2486-2497 (2008), Carter, Nature Revs., 6:343-357 (2006); R. Kontermann & S. Dubel, 2001 Antibody Engineering, Springer- Verlag, Heidelberg-New York).
[78] In certain embodiments, the cell-binding agent is a minibody, a diabody, a tribody, a tetrabody, a nanobody, a probody, a domain body, or an unibody.
[79] Monoclonal antibody techniques allow for the production of extremely specific cell- binding agents in the form of specific monoclonal antibodies. Particularly well known in the art are techniques for creating monoclonal antibodies produced by immunizing mice, rats, hamsters or any other mammal with the antigen of interest such as the intact target cell, antigens isolated from the target cell, whole virus, attenuated whole virus, and viral proteins such as viral coat proteins. Sensitized human cells can also be used. Another method of creating monoclonal antibodies is the use of phage libraries of scFv (single chain variable region), specifically human scFv (see e.g., Griffiths et al., U.S. Patent Nos. 5,885,793 and 5,969,108; McCafferty et al., WO 92/01047; Liming et al., WO 99/06587). In addition, resurfaced antibodies disclosed in U.S. Patent No. 5,639,641 may also be used, as may chimeric antibodies and humanized antibodies.
[80] Selection of the appropriate cell-binding agent is a matter of choice that depends upon the particular cell population that is to be targeted, but in general human monoclonal antibodies are preferred if an appropriate one is available. For example, the monoclonal antibody MY9 is a murine IgGi antibody that binds specifically to the CD33 Antigen (J.D. Griffin et al., Leukemia Res., 8:521 (1984)) and can be used if the target cells express CD33 as in the disease of acute myelogenous leukemia (AML).
[81] In one embodiment, the cell-binding agent is a resurfaced antibody, a resurfaced single chain antibody, or a resurfaced antibody fragment.
[82] In another embodiment, the cell-binding agent is a humanized antibody, a humanized single chain antibody, or a humanized antibody fragment. In a specific embodiment, the humanized antibody is huMy9-6 or another related antibody, which is described in U.S. Pat.
Nos. 7,342,110 and 7,557,189. In another specific embodiment, the humanized antibody is an anti-folate receptor antibody described in U.S. Provisional Application Nos. 61/307,797,
61/346,595, and 61/413,172 and U.S. Application No. 13/033,723 (published as US 2012/0009181 Al). The teachings of all these applications are incorporated herein by reference in its entirety.
[83] In certain embodiments, the cell-binding agent is an antigen-binding portion of a monoclonal antibody, sharing sequences critical for antigen-binding with an antibody disclosed herein, such as huMy9-6 or its related antibodies described in U.S. Pat. Nos.
7,342,110 and 7,557, 189, incorporated herein by reference. These derivative antibodies may have substantially the same or identical (1) light chain and/or heavy chain CDR3 regions; (2) light chain and/or heavy chain CDR1, CDR2, and CDR3 regions; or (3) light chain and/or heavy chain regions, compared to an antibody described herein. Sequences within these regions may contain conservative amino acid substitutions, including substitutions within the CDR regions. Preferably, there is no more than 1, 2, 3, 4, or 5 conservative substitutions. In an alternative, the derivative antibodies have a light chain region and/or a heavy chain region that is at least about 90%, 95%, 99% or 100% identical to an antibody described herein. These derivative antibodies may have substantially the same binding specificity and/or affinity to the target antigen compared to an antibody described herein. Preferably, the Kd and/or R values of the derivative antibodies are within 10-fold (either higher or lower), 5- fold (either higher or lower), 3-fold (either higher or lower), or 2-fold (either higher or lower) of an antibody described herein. These derivative antibodies may be fully human antibodies, or humanized antibodies, or chimeric antibodies. The derivative antibodies may be produced according to any art-recognized methods.
[84] Specific exemplary antigens or ligands include renin; a growth hormone (e.g. , human growth hormone and bovine growth hormone); a growth hormone releasing factor; a parathyroid hormone; a thyroid stimulating hormone; a lipoprotein; alpha- 1- antitrypsin; insulin A-chain; insulin B-chain; proinsulin; a follicle stimulating hormone; calcitonin; a luteinizing hormone; glucagon; a clotting factor (e.g. , factor vmc, factor IX, tissue factor, and von Willebrands factor); an anti-clotting factor (e.g. , Protein C); an atrial natriuretic factor; a lung surfactant; a plasminogen activator (e.g. , a urokinase, a human urine or tissue-type plasminogen activator); bombesin; a thrombin; hemopoietic growth factor; tumor necrosis factor- alpha and -beta; an enkephalinase; RANTES (i.e. , the regulated on activation normally
T-cell expressed and secreted); human macrophage inflammatory protein- 1 -alpha; a serum albumin (human serum albumin); Muellerian-inhibiting substance; relaxin A-chain; relaxin
B-chain; prorelaxin; a mouse gonadotropin-associated peptide; a microbial protein (beta- lactamase); DNase; IgE; a cytotoxic T-lymphocyte associated antigen (e.g. , CTLA-4); inhibin; activin; a vascular endothelial growth factor; protein A or D; a rheumatoid factor; a neurotrophic factor(e.g. , bone-derived neurotrophic factor, neurotrophin-3, -4, -5, or -6), a nerve growth factor (e.g. , NGF-β); a platelet-derived growth factor; a fibroblast growth factor (e.g. , aFGF and bFGF); fibroblast growth factor receptor 2; an epidermal growth factor; a transforming growth factor (e.g. , TGF-alpha, TGF-βΙ, TGF- 2, TGF- 3, TGF- 4, and TGF- β5); insulin-like growth factor-I and -II; des(l-3)-IGF-I (brain IGF-I); an insulin-like growth factor binding protein; melanotransferrin; EpCAM; GD3; FLT3; PSMA; PSCA; MUC 1 ; MUC 16; STEAP; CEA; TENB2; an EphA receptor; an EphB receptor; a folate receptor; FOLR1 ; mesothelin; cripto; an alphavbeta6; integrins; VEGF; VEGFR; EGFR; transferrin receptor; IRTAl ; IRTA2; IRTA3; IRTA4; IRTA5; CD proteins (e.g. , 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, and CD152), one or more tumor-associated antigens or cell-surface receptors (see US Publication No. 20080171040 or US Publication No. 20080305044, incorporated in their entirety by reference); erythropoietin; an
osteoinductive factor; an immunotoxin; a bone morphogenetic protein; an interferon (e.g. , interferon-alpha, -beta, and -gamma); a colony stimulating factor (e.g. , M-CSF, GM-CSF, and G-CSF); interleukins (e.g. , IL- 1 to IL- 10); a superoxide dismutase; a T-cell receptor; a surface membrane protein; a decay accelerating factor; a viral antigen s(e.g. , a portion of the HIV envelope); a transport protein, a homing receptor; an addressin; a regulatory protein; an integrin (e.g. , CDl la, CDl lb, CDl lc, CD18, an ICAM, VLA-4, and VCAM;) a tumor associated antigen (e.g. , HER2, HER3 and HER4 receptor); endoglin; c-Met; c-kit; 1GF1R; PSGR; NGEP; PSMA; PSCA; TMEFF2; LGR5; B7H4; and fragments of any of the above- listed polypeptides.
[85] For example, GM-CSF, a ligand / growth factor which binds to myeloid cells can be used as a cell-binding agent to diseased cells from acute myelogenous leukemia. IL-2 which binds to activated T-cells can be used for prevention of transplant graft rejection, for therapy and prevention of graft-versus-host disease, and for treatment of acute T-cell leukemia.
MSH, which binds to melanocytes, can be used for the treatment of melanoma, as can antibodies directed towards melanomas. Folic acid can be used to target the folate receptor expressed on ovarian and other tumors. Epidermal growth factor can be used to target squamous cancers, such as lung and head and neck. Somatostatin can be used to target neuroblastomas and other tumor types. Estrogen (or estrogen analogues) can be used to target breast cancer. Androgen (or androgen analogues) can be used to target testes.
[86] The term "salt" as used herein refers to organic or inorganic salts of a compound of the invention. Preferably, a salt is a pharmaceutically acceptable salt. Other non- pharmaceutically acceptable salts are also included in the present invention. The salts include salts, formed by reacting a compound of the invention, which comprises a basic group, with an inorganic acid or organic acid (such as a carboxylic acid), and salts, formed by reacting a compound of the invention, which comprises an acidic group, with an inorganic base or organic base (such as an amine). Exemplary salts include those pharmaceutically acceptable salts described immediately below.
[87] The term "pharmaceutically acceptable salt" as used herein refers to
pharmaceutically acceptable organic or inorganic salts of a compound of the invention.
Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate "mesylate," ethanesulfonate, benzenesulfonate, p-toluenesulfonate, pamoate (i.e. , l,l'-methylene-bis-(2-hydroxy-3-naphthoate)) salts, alkali metal (e.g. , sodium and potassium) salts, alkaline earth metal (e.g. , magnesium) salts, and ammonium salts. A pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion. The counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound. Furthermore, a pharmaceutically acceptable salt may have more than one charged atom in its structure.
Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion.
[88] If the compound of the invention contains one or more basic moieties, desired salts
(e.g. , pharmaceutically acceptable salts) may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
[89] If the compound of the invention contains one or more acidic moieties, desired salts (e.g. , pharmaceutically acceptable salts) may be prepared by any suitable method, for example, treatment of the free acid with an inorganic, such as an alkali metal hydroxide or alkaline earth metal hydroxide, organic base, such as an amine (primary, secondary or tertiary), or the like. Illustrative examples of suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
[90] The terms "abnormal cell growth" and "proliferative disorder" are used interchangeably in this application. "Abnormal cell growth," as used herein, unless otherwise indicated, refers to cell growth that is independent of normal regulatory
mechanisms (e.g. , loss of contact inhibition). This includes, for example, the abnormal growth of: (1) tumor cells (tumors) that proliferate by expressing a mutated tyrosine kinase or overexpression of a receptor tyrosine kinase; (2) benign and malignant cells of other proliferative diseases in which aberrant tyrosine kinase activation occurs; (3) any tumors that proliferate by receptor tyrosine kinases; (4) any tumors that proliferate by aberrant serine/threonine kinase activation; and (5) benign and malignant cells of other proliferative diseases in which aberrant serine/threonine kinase activation occurs.
[91] The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancerous cells, and/or benign or pre-cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include skin cancer (e.g. , melanoma), Merkel cell carcinoma, squamous cell cancer (e.g. , epithelial squamous cell cancer), lung cancer (e.g. , small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung), cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer (e.g. , gastrointestinal cancer), pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, testicular cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, acute leukemia, head and neck cancer, brain cancer (e.g. , glioblastoma and neuroblastoma), cancers of lymphatic organs and hematological malignancy including Leukemia (Acute lymphoblastic leukemia (ALL), Acute myelogenous leukemia (AML), Chronic lymphocytic leukemia (CLL), Chronic myelogenous leukemia (CML), Acute monocytic leukemia (AMOL), Hairy cell leukemia (HCL), T-cell prolymphocytic leukemia (T-PLL), Large granular lymphocytic leukemia, Adult T-cell leukemia), Lymphoma (small lymphocytic lymphoma (SLL), Hodgkin's lymphomas
(Nodular sclerosis, Mixed cellularity, Lymphocyte-rich, Lymphocyte depleted or not depleted, and Nodular lymphocyte-predominant Hodgkin lymphoma), Non-Hodgkin's lymphomas (all subtypes), Chronic lymphocytic leukemia/Small lymphocytic lymphoma, B- cell prolymphocytic leukemia, Lymphoplasmacytic lymphoma (such as Waldenstrom macro globulinemia), Splenic marginal zone lymphoma, Plasma cell neoplasms (Plasma cell myeloma, Plasmacytoma, Monoclonal immunoglobulin deposition diseases, Heavy chain diseases), Extranodal marginal zone B cell lymphoma (MALT lymphoma), Nodal marginal zone B cell lymphoma (NMZL), Follicular lymphoma, Mantle cell lymphoma, Diffuse large
B cell lymphoma, Mediastinal (thymic) large B cell lymphoma, Intravascular large B cell lymphoma, Primary effusion lymphoma, Burkitt lymphoma/leukemia, T cell prolymphocytic leukemia, T cell large granular lymphocytic leukemia, Aggressive NK cell leukemia, Adult T cell leukemia/lymphoma, Extranodal NK/T cell lymphoma (nasal type), Enteropathy-type T cell lymphoma, Hepatosplenic T cell lymphoma, Blastic NK cell lymphoma, Mycosis fungoides / Sezary syndrome, Primary cutaneous CD30-positive T cell lymphoproliferative disorders, Primary cutaneous anaplastic large cell lymphoma, Lymphomatoid papulosis,
Angioimmunoblastic T cell lymphoma, Peripheral T cell lymphoma (unspecified), Anaplastic large cell lymphoma), multiple myeloma (plasma cell myeloma or Kahler's disease).
[92] The term "therapeutic agent" encompasses both a biological agent such as an antibody, a peptide, a protein, an enzyme or a chemotherapeutic agent.
[93] The term "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include Erlotinib (TARCEVA®,
Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant
(FASLODEX®, AstraZeneca), Sutent (SU11248, Pfizer), Letrozole (FEMARA®, Novartis),
Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin
(Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), Lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline),
Lonafarnib (SCH 66336), Sorafenib (BAY43-9006, Bayer Labs), and Gefitinib (IRESSA®,
AstraZeneca), AG1478, AG1571 (SU 5271; Sugen), alkylating agents such as thiotepa and
CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine;
acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, KW-2189 and CB1-TM1);
eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide,
mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g. , calicheamicin, especially calicheamicin gammall and
calicheamicin omegall (Angew Chem. Intl. Ed. Engl. (1994) 33: 183-186); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6- diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esonibicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin,
methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamniprine, 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; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin;
sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2' ,2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan;
vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g. , TAXOL® (paclitaxel; Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANE® (Cremophor-free), albumin-engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, 111.), and TAXOTERE® (doxetaxel; Rhone-Poulenc Rorer, Antony, France); chloranmbucil; GEMZAR® (gemcitabine); 6-thioguanine; mercaptopurine;
methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® (vinorelbine); novantrone;
teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA®); ibandronate; CPT-11 ; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.
[94] Also included in the definition of "chemotherapeutic agent" are: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen
(including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE®
(megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR®
(vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca);
(iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); (iv) protein kinase inhibitors; (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Ralf and H-Ras; (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®;
PROLEUKIN® rIL-2; a topoisomerase 1 inhibitor such as LURTOTECAN®; ABARELIX® rmRH; (ix) anti- angiogenic agents such as bevacizumab (AVASTIN®, Genentech); and (x) pharmaceutically acceptable salts, acids and derivatives of any of the above. Other anti- angiogenic agents include MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix - metalloproteinase 9) inhibitors, COX-II (cyclooxygenase II) inhibitors, and VEGF receptor tyrosine kinase inhibitors. Examples of such useful matrix metalloproteinase inhibitors that can be used in combination with the present compounds/compositions are described in WO 96/33172, WO 96/27583, EP 818442, EP 1004578, WO 98/07697, WO 98/03516, WO 98/34918, WO 98/34915, WO 98/33768, WO 98/30566, EP 606,046, EP 931,788, WO 90/05719, WO 99/52910, WO 99/52889, WO 99/29667, WO 99/07675, EP 945864, U.S. Pat. No. 5,863,949, U.S. Pat. No. 5,861,510, and EP 780,386, all of which are incorporated herein in their entireties by reference. Examples of VEGF receptor tyrosine kinase inhibitors include 4-(4-bromo-2-fluoroanilino)-6-methoxy-7-(l-methylpiperidin-4- ylmethoxy)quinazoline (ZD6474; Example 2 within WO 01/32651), 4-(4-fluoro-2- methylindol-5-yloxy)-6-methoxy-7-(3-pyrrolidin- l-ylpropoxy)-quinazoline (AZD2171 ;
Example 240 within WO 00/47212), vatalanib (PTK787; WO 98/35985) and SU11248 (sunitinib; WO 01/60814), and compounds such as those disclosed in PCT Publication Nos. WO 97/22596, WO 97/30035, WO 97/32856, and WO 98/13354).
[95] Other examples of chemotherapeutic agents that can be used in combination with the present compounds include inhibitors of PI3K (phosphoinositide-3 kinase), such as those reported in Yaguchi et al. (2006) J. Nat. Cancer Inst. 98(8):545-556; U.S. Pat. No. 7,173,029;
U.S. Pat. No. 7,037,915; U.S. Pat. No. 6,608,056; U.S. Pat. No. 6,608,053; U.S. Pat. No.
6,838,457; U.S. Pat. No. 6,770,641; U.S. Pat. No. 6,653,320; U.S. Pat. No. 6,403,588;
WO 2006/046031; WO 2006/046035; WO 2006/046040; WO 2007/042806; WO
2007/042810; WO 2004/017950; US 2004/092561; WO 2004/007491; WO 2004/006916;
WO 2003/037886; US 2003/149074; WO 2003/035618; WO 2003/034997; US 2003/158212;
EP 1417976; US 2004/053946; JP 2001247477; JP 08175990; JP 08176070; U.S. Pat. No.
6,703,414; and WO 97/15658, all of which are incorporated herein in their entireties by reference. Specific examples of such PI3K inhibitors include SF-1126 (PI3K inhibitor, Semafore Pharmaceuticals), BEZ-235 (PI3K inhibitor, Novartis), XL- 147 (PI3K inhibitor, Exelixis, Inc.).
[96] Chemotherapeutic agents may also include any of the generic drugs or biosimilars of the brand-name drugs referenced herein, or improvements thereof, including improved formulations, delivery means (sustained release, bioadhesive coating, targeted delivery etc. ), and dosage forms.
[97] The term "viral infection" refers to the invasion of a host organism's bodily tissues by disease-causing viruses. Examples of the viral infections include CMV infection, HIV infection and AIDS.
[98] The term "parasite infection" refers to the invasion of a host organism's bodily tissues by disease-causing parasites. Examples of the parasite infections include giardiasis, amoebiasis, and schistosomiasis.
[99] The phrase "pharmaceutically acceptable" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
[100] The term "therapeutically effective amount" means that amount of active compound or conjugate that elicits the desired biological response in a subject. Such response includes alleviation of the symptoms of the disease or disorder being treated, prevention, inhibition or a delay in the recurrence of symptom of the disease or of the disease itself, an increase in the longevity of the subject compared with the absence of the treatment, or prevention, inhibition or delay in the progression of symptom of the disease or of the disease itself. Determination of the effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Toxicity and therapeutic efficacy of compound I can be determined by standard pharmaceutical procedures in cell cultures and in experimental animals. The effective amount of compound or conjugate of the present invention or other therapeutic agent to be administered to a subject will depend on the stage, category and status of the multiple myeloma and characteristics of the subject, such as general health, age, sex, body weight and drug tolerance. The effective amount of compound or conjugate of the present invention or other therapeutic agent to be administered will also depend on administration route and dosage form. Dosage amount and interval can be adjusted individually to provide plasma levels of the active compound that are sufficient to maintain desired therapeutic effects. CELL BINDING AGENT-DRUG MOIETY CONJUGATES
[101] The present invention provides a conjugate represented by Formula (Γ) or (I) as described in the first embodiment and its alternatively embodiments above.
[102] In a first specific embodiment, in Formula (Γ) or (I), the ionizable group is -SO3H, - Z'-S03H, -OP03H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, -C02H, -Z'C02H, -NRnRi2, or -Z - NRnRi2, and the charged group is -N^ R^RisX", or -Z'-N'^R RwRisX"; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R12, for each occurrence, are independently H or an optionally substituted alkyl; R1 to R15 are each independently an optionally substituted alkyl; and X" is a pharmaceutically acceptable anion. In this first specific embodiment, the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments.
[103] In a second specific embodiment, within the ionizable group or the charged substituent described in the preceding paragraph immediately above, variable Z' is alkylene and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first specific embodiment.
[104] In a third specific embodiment, in Formula (Γ) or (I), Rmis H, -CH2CH2S03H, or a pharmaceutically acceptable salt of -CH2CH2S03H, and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments.
[105] In a fourth specific embodiment, in Formula (Γ) or (I), P is [XX]2^, and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[106] In a fifth specific embodiment, in Formula (Γ) or (I), P is [XX] 2, and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[107] In a sixth specific embodiment, in Formula (I)' or (I), P is [XX]3, and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
Figure imgf000037_0001

Figure imgf000038_0001
Figure imgf000038_0002
. In this seventh specific embodiment, the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[109] In an eighth specific embodiment, in Formula (Γ ) or (I), JCB' is
Figure imgf000038_0003
and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment. s1 §=N-NRe--i s2
[110] In a ninth specific embodiment, JCB' in Formula (Γ) or (I) is * ¾ ,
Figure imgf000039_0001
s 1 -N-NRe— s2 s1 s2 s1 |— N— O— | s2
¾ ¾ , , or ¾ ? ; and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[I l l] In a tenth specific embodiment, in Formula (Γ) or (I), Ra, Rb, Rc, and Re are each H; and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, or eighth specific embodiment.
[112] In a eleventh specific embodiment, in Formula (Γ) or (I), DM is represented by the following formula:
Figure imgf000039_0002
In this tenth specific embodiment, the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth specific embodiment.
[113] In a twelfth specific embodiment, in Formula (I) or (Γ), R3, R4, R5 and R6 are all H, m and n are each independently an integer from 1 to 5 (e.g., 1, 2, 3, 4 or 5); and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth or eleventh specific embodiment,
[114] In a thirteenth specific embodiment, in Formula (I) or (Γ), Z2 is -0-CH2- and X\ is - CH2-C(=0)-NH-; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh or twelfth specific embodiment.
[115] In a fourteenth specific embodiment, in Formula (I), the conjugate is represented by any one of the following formulae, or a pharmaceutically acceptable salt thereof:
Figure imgf000040_0001
in which DM is a drug moiety represented by Formula (II) as described in the first embodiment above or by Formula (IIA) described in the tenth specific embodiment.
[116] In Formula (Γ), (I), (IA), (IB), (IC) or (ID), w is preferably an integer between 1 and 10, between 1 and 6, or alternatively, between 1 and 4; and the remainder of the variables in Formula (Γ) or (I) are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth,, ninth, tenth, eleventh,twelfth, thirteenth or fourteenth specific embodiment.
[117] The present invention also provides a conjugate represented by Formula (V) as described in the first embodiment and its alternatively embodiments above.
[118] In a first specific embodiment, in Formula (V), one of X\ and Z2 is absent; and the remainder of the variables is as defined in the first embodiment.
[119] In a second specific embodiment, in Formula (V), both X\ and Z2 are absent; and the remainder of the variables is as defined in the first embodiment.
Figure imgf000041_0001
40
Figure imgf000042_0001
Figure imgf000042_0002
the variables is as defined in the first embodiment or each of its alternative embodiments or its first or second specific embodiment described above.
[121] In a fourth specific embodiment, in Formula (V), R4 and R2 are both H; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second or third specific embodiment.
[122] In a fifth specific embodiment, in Formula (V), Ra, Rb, Rc, and Re are each H; and the remainder of the variables are as defined in the first embodiment or each of its alternative embodiments or its first, second, third or fourth specific embodiment.
[123] In a sixth specific embodiment, in Formula (V), DM is represented by formula (IIA) described above; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth or fifth specific embodiment.
[124] In Formula (V), w is preferably an integer between 1 and 10, between 1 and 6, or alternatively, between 1 and 4; and the remainder of the variables in Formula (V) are as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth fifth or sixth specific embodiment. CYTOTOXIC COMPOUNDS
[125] The present invention also provides a cytotoxic compound represented by Formula (ΙΙΓ) or (III) as described in the second embodiment and its alternatively embodiments above.
[126] In a first specific embodiment, in Formula (ΙΙΓ) or (III),the ionizable group is -SO3H, -Z'-S03H, -OP03H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, -C02H, -Z'C02H, -NRnRi2, or -Z - NRnRi2, and the charged group is -N+R13R14Ri5X~, or -Z'-N'^R RwRisX"; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R12, for each occurrence, are independently H or an optionally substituted alkyl; R1 to R15 are each independently an optionally substituted alkyl; and X" is an anion (e.g. , a pharmaceutically acceptable anion). In this first specific embodiment, the remainder of the variables are as defined in the second embodiment or each of its alternative
embodiments.
[127] In a second specific embodiment, within the ionizable group or the charged substituent described in the preceding paragraph immediately above, variable Z' is alkylene and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first specific embodiment.
[128] In a third specific embodiment, in Formula (ΙΙΓ) or (III), Rm is H, -CH2CH2S03H, or a pharmaceutically acceptable salt of -CH2CH2S03H, and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments.
[129] In a fourth specific embodiment, in Formula (ΙΙΓ) or (III), P is [XX]2_4, and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[130] In a fifth specific embodiment, in Formula (ΙΙΓ) or (III), P is [XX]2, and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[131] In a sixth specific embodiment, in Formula (ΙΙΓ) or (III), P is [XX] 3, and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[132] In a seventh specific embodiment, in Formula (ΙΙΓ) or (III), JCB is maleimide, X'- CRbRc-C(=0)-, X'-CRbRc-C(=0)-NRe-, Ra-C(=0)-, Ra-C(=0)-phenylene-, NH2-NR6-, N -NRe-phenylene-, ΝΗ2-0-, -N3, -C≡CH,
Figure imgf000044_0001
Figure imgf000044_0002
seventh specific embodiment, the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[133] In an eighth specific embodiment, in Formula (ΙΙΓ) or (III), JCB is maleimide and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[134] In a ninth specific embodiment, in Formula (ΙΙΓ) or (III), JCB is NH2-NRe- or NH2-0-; and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[135] In a tenth specific embodiment, in Formula (ΙΙΓ) or (III), Ra, Rb, Rc, and Re are each H; and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, or eighth specific embodiment.
[136] In a eleventh specific embodiment, in Formula (ΙΙΓ) or (III), DM is represented by the formula (IIA) as described above; and the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth specific embodiment.
[137] In a twelfth specific embodiment, in Formula (III) or (ΙΙΓ), R3, R4, R5 and R6 are all H, m and n are each independently an integer from 1 to 5 (e.g., 1, 2, 3, 4 or 5); the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth or eleventh specific embodiment,
[138] In a thirteenth specific embodiment, in Formula (III) or (ΙΙΓ), Z2 is -0-CH2- and X\ is -CH2-C(=0)-NH-; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh or twelfth specific embodiment.
[139] In a fourteenth specific embodiment, in Formula (ΙΙΓ) or (III), the cytotoxic compound is represented by any one of the following formulae, or a salt (e.g. , a
pharmaceutically acceptable salt) thereof:
Figure imgf000045_0001
in which DM is a drug moiety represented by Formula (II) as described in the second embodiment above or Formula (IIA) as described above.
[140] The present invention also provides a cytotoxic compound represented by Formula (VI) or (III) as described in the second embodiment or each of its alternatively embodiments above.
[141] In a first specific embodiment, in Formula (VI), one of X\ and Z2 is absent; and the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments above.
[142] In a second specific embodiment, in Formula (VI), both X\ and Z2 are absent; and the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments above. [143] In a third specific embodiment, in Formula (VI), JCBi is Ra-C(=0)-, Ra-C(= -
phenylene-, NH2-NRe-, NH2-NRe-phenylene-, ΝΗ2-0-, -N3, -C≡CH,
Figure imgf000046_0001
Figure imgf000046_0002
. In this third specific embodiment, the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments or its first or second specific embodiment described above.
[144] In a fourth specific embodiment, in Formula (VI), R and R2 are both H; and the remainder of the variables is as defined in the second embodiment or each of its alternative embodiments or its first, second or third specific embodiment.
[145] In a fifth specific embodiment, in Formula (VI), Ra, Rb, Rc, and Re are each H; and the remainder of the variables are as defined in the second embodiment or each of its alternative embodiments or its first, second, third or fourth specific embodiment.
[146] In a sixth specific embodiment, in Formula (VI), DM is represented by formula (IIA) described above; and the remainder of the variables are as defined in the second embodiment or each its alternative embodiments or its first, second, third, fourth or fifith specific embodiment.
LINKER COMPOUNDS
[147] The present invention further provides a linker compound represented by Formula (IV) or (IV) as described in the third embodiment and its alternatively embodiments above.
[148] In a first specific embodiment, in Formula (IV) or (IV), the ionizable group -S03H, - Z'-S03H, -OP03H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, -C02H, -Z'C02H, -NRnRi2, or -Z - NRnRi2, and the charged group is -N^^RMR^X", or -Z'-N'^R RwRisX"; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R12, for each occurrence, are independently H or an optionally substituted alkyl; R13 to R15 are each independently an optionally substituted alkyl; and X" is an anion (e.g. , a pharmaceutically acceptable anion). In this first specific embodiment, the remainder of the variables are as defined in the third embodiment or each of its alternative
embodiments.
[149] In a second specific embodiment, within the ionizable group or the charged substituent described in the preceding paragraph immediately above, variable Z' is alkylene and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first specific embodiment.
[150] In a third specific embodiment, in Formula (IV) or (IV), Rm is H, -CH2CH2S03H, or a pharmaceutically acceptable salt of -CH2CH2S03H, and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments.
[151] In a fourth specific embodiment, in Formula (IV) or (IV), P is [XX]2_4, and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[152] In a fifth specific embodiment, in Formula (IV) or (IV), P is [XX]2, and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[153] In a sixth specific embodiment, in Formula (IV) or (IV), P is [XX]3, and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, or third specific embodiment.
[154] In a seventh specific embodiment, in Formula (IV) or (IV), JCB is maleimide, X'- CRbRc-C(=0)-, X'-CRbRc-C(=0)-NRe-, Ra-C(=0)-, Ra-C(=0)-phenylene-, NH2-NR6-, NH2-NRe-phenylene-, or NH2-0-. In this seventh specific embodiment, the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[155] In an eighth specific embodiment, in Formula (IV) or (IV), JCB is maleimide and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment. [ 156] In a nineth specific embodiment, in Formula (IV) or (IV), JCB is NH2-NRE- or NH2- 0-; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, or sixth specific embodiment.
[ 157] In a tenth specific embodiment, in Formula (IV) or (IV), RA, RB, RC, and RE are each H; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, or eighth specific embodiment.
[ 158] In a eleventh specific embodiment, in formula (IV) or (IV), JD is -OH or -CI; or - C(=0)-JD is N-hydroxysuccinimide ester; and the remainder of the variables is as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth, ninth or tenth specific embodiment.
[ 159] In a twelfth specific embodiment, in Formula (IV) or (IV), R3, R4, R5 and R6 are all H, m and n are each independently an integer from 1 to 5 (e.g., 1 , 2, 3, 4 or 5); the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth ,ninth, tenth or eleventh specific embodiment,
[ 160] In a thirteenth specific embodiment, in Formula (IV) or (IV), Z2 is -0-CH2- and X\ is -CH2-C(=0)-NH-; and the remainder of the variables is as defined in the first embodiment or each of its alternative embodiments or its first, second, third, fourth, fifth, sixth, seventh, eighth ,ninth, tenth, eleventh or twelfth specific embodiment.
[ 161] In a fourteenth specific embodiment, in Formula (IV), the linker compound is represented by any one of the following formulae, or a salt {e.g. , a pharmaceutically acceptable salt) thereof:
Figure imgf000048_0001
Figure imgf000049_0001
in which JD is defined as in the third embodiment; alternatively, JD is -OH or -CI; or -C(=0)- JD is N-hydroxysuccinimide ester.
[162] The present invention further provides a linker compound represented by Formula (VII) as described in the third embodiment and its alternatively embodiments above.
[163] In a first specific embodiment, in Formula (VII), one of X\ and Z2 is absent; and the remainder of the variables is as defined in the third embodiment or each of its alternative embodiments above.
[164] In a second specific embodiment, in Formula (VII), both X\ and Z2 are absent; and the remainder of the variables is as defined in the third embodiment or each of its alternative embodiments above.
[165] In a third specific embodiment, in Formula (VII), JCBi is Ra- =0)-, Ra-C(=0)-
henylene-, NH2-NR6-, NH2-NRe-phenylene-, ΝΗ2-0-, -N3, -C≡CH,
Figure imgf000049_0002
Figure imgf000049_0003
. In this third specific embodiment, the remainder of the variables is as defined in the third embodiment or each of its alternative embodiments or its first or second specific embodiment described above. [166] In a fourth specific embodiment, in Formula (VII), R and R2 are both H; and the remainder of the variables is as defined in the third embodiment or each of its alternative embodiments or its first, second or third specific embodiment.
[167] In a fifth specific embodiment, in Formula (VII), Ra, Rb, Rc, and Re are each H; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third or fourth specific embodiment.
[168] In a sixth specific embodiment, in Formula (VII), JD is -OH or -CI; or -C(=0)-JD is N-hydroxysuccinimide ester; and the remainder of the variables are as defined in the third embodiment or each of its alternative embodiments or its first, second, third, fourth or fifth specific embodiment.
PRODUCTION OF CELL-BINDING AGENT-DRUG CONJUGATES
[169] The conjugates of the present invention can be prepared according to any known method in the art. See, for example, WO 2009/134977, U.S. Patent Nos. 7,811,572,
6,441,163, 7,368,565, 8,163,888, U.S Publication Nos. 2006/0182750, 2011/0003969, 2012/0253021 and Widdison, W. C. et ah, "Semisynthetic maytansine analogues for the targeted treatment of cancer," /. Med. Chem. (2006) 49(14):4392-4408. In one embodiment, the conjugates of the present invention can be prepared by reacting a cell-binding agent with a drug-linker compound (e.g., compounds of formula (ΙΙΓ) or (III)) having a reactive moiety capable of forming a covalent bond with the cell-binding agent to form a cell-binding agent- cytotoxic agent conjugate. The conjugate can then be purified. The drug-linker compound can be generated in situ and used to react with the antibody without purification.
Alternatively, the drug-linker compound can be generated and purified before conjugating to the cell-binding agent. .
[170] In another embodiment, the conjugates of the present invention can be prepared by: a) reacting a cell-binding agent with a linker compound (e.g., compounds of formula IV) to form a modified cell-binding agent having the linkers covalently bound thereto; b) optionally purifying the modified cell-binding agent; c) conjugating a cytotoxic agent to the modified cell-binding agent to form the cell-binding agent-cytotoxic compound conjugate of the present invention; and d) purifying the cell-binding agent-cytotoxic compound conjugate.
[171] In another embodiment, the conjugate of the present invention can be prepared by mixing together a cell-binding agent, a cytotoxic agent and a linker compound. Preferably, the cell-binding agent is contacted with a cytotoxic agent first to form a mixture comprising the cell-binding agent and the cytotoxic agent, followed by contacting the mixture with a linker compound (e.g., compounds of formula (IV)) to form the cell-binding agent-cytotoxic compound conjugate. The conjugate can then be purified.
[172] Any purification methods known in the art can be used to purify the conjugates of the present invention (see, for example, Bioconjugate Techniques, 2nd Edition By Greg T.
Hermanson, Published by Academic Press, Inc., 2008). In one embodiment, the conjugates of the present invention using tangential flow filtration (TFF), non-adsorptive
chromatography, adsorptive chromatography, adsorptive filtration, selective precipitation, high performance liquid chromatography (HPLC), dialysis or any other suitable purification process, as well as combinations thereof.
[173] Any suitable TFF systems may be utilized for purification, including a Pellicon type system (Millipore, Billerica, MA), a Sartocon Cassette system (Sartorius AG, Edgewood,
NY), and a Centrasette type system (Pall Corp., East Hills, NY).
[174] Any suitable adsorptive chromatography resin may be utilized for purification.
Preferred adsorptive chromatography resins include hydroxyapatite chromatography, hydrophobic charge induction chromatography (HCIC), hydrophobic interaction
chromatography (HIC), ion exchange chromatography, mixed mode ion exchange
chromatography, immobilized metal affinity chromatography (IMAC), dye ligand
chromatography, affinity chromatography, reversed phase chromatography, and combinations thereof. Examples of suitable hydroxyapatite resins include ceramic hydroxyapatite (CHT
Type I and Type II, Bio-Rad Laboratories, Hercules, CA), HA Ultrogel hydroxyapatite (Pall
Corp., East Hills, NY), and ceramic fluoroapatite (CFT Type I and Type II, Bio-Rad
Laboratories, Hercules, CA). An example of a suitable HCIC resin is MEP Hypercel resin
(Pall Corp., East Hills, NY). Examples of suitable HIC resins include Butyl-Sepharose,
Hexyl-Sepaharose, Phenyl-Sepharose, and Octyl Sepharose resins (all from GE Healthcare,
Piscataway, NJ), as well as Macro-prep Methyl and Macro-Prep t-Butyl resins (Biorad
Laboratories, Hercules, CA). Examples of suitable ion exchange resins include SP-
Sepharose, CM-Sepharose, and Q-Sepharose resins (all from GE Healthcare, Piscataway,
NJ), and Unosphere S resin (Bio-Rad Laboratories, Hercules, CA). Examples of suitable mixed mode ion exchangers include Bakerbond ABx resin (JT Baker, Phillipsburg NJ).
Examples of suitable IMAC resins include Chelating Sepharose resin (GE Healthcare,
Piscataway, NJ) and Profinity IMAC resin (Bio-Rad Laboratories, Hercules, CA). Examples of suitable dye ligand resins include Blue Sepharose resin (GE Healthcare, Piscataway, NJ) and Affi-gel Blue resin (Bio-Rad Laboratories, Hercules, CA). Examples of suitable affinity resins include Protein A Sepharose resin (e.g. , MabSelect, GE Healthcare, Piscataway, NJ), His-Tag metal affinity resins, anti-FLAG affinity resins, and lectin affinity resins, e.g. Lentil Lectin Sepharose resin (GE Healthcare, Piscataway, NJ), where the antibody bears appropriate lectin binding sites. Examples of suitable reversed phase resins include C4, C8, and C18 resins (Grace Vydac, Hesperia, CA).
[175] Any suitable non-adsorptive chromatography resin may be utilized for purification. For example, size-exclusion chromatography can be used for purifying the conjugates of the invention. Examples of suitable non-adsorptive chromatography resins include, but are not limited to, SEPHADEX™ G- 10, G-25, G-50, G- 100, SEPHACRYL™ resins (e.g. , S-200 and S-300), SUPERDEX™ resins (e.g. , SUPERDEX™ 75 and SUPERDEX™ 200), BIO-GEL® resins (e.g. , P-6, P-10, P-30, P-60, and P- 100), and others known to those of ordinary skill in the art.
[176] In one embodiment, when the cell-binding agent is an epitope-tagged Avibody, the conjugate can be purified using hydroxyl apatite chromatography, size-exclusion
chromatography, tangential flow filtration, gel electrophoresis, dialysis, and affinity chromatography, preferably affinity chromatography, more preferably His-tag metal affinity chromatography and anti-FLAG M2 affinity chromatography (see, for example,
US 2008/0152586 and US 2012/0171115).
[177] In another embodiment, when the cell-binding agent is a centyrin, the conjugate can be purified using protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, size-exclusion chromatography, tangential flow filtration, affinity chromatography, hydroxylapatite chromatography and lectin
chromatography. Alternatively, the conjugate can be purified using HPLC. Preferably, the conjugate can be purified by using affinity chromatography, more preferably His-tag metal affinity chromatography. See, for example, US 2010/0255056, US 2010/0216708 and US 2011/0274623.
[178] In another embodiment, when the cell-binding agent is a DARPin, the conjugate can be purified by affinity chromatography, size exclusion chromatography, hydroxylapatite chromatography, tangential flow fitration, preferably affinity chromatography, more preferably His-Tag affinity chromatography. See, for example, U.S. Patent Publication Nos. 2004/0132028, 2009/0082274, 2011/0118146, and 2011/0224100, WO 02/20565 and WO 06/083275.
[179] The number of cytotoxic compound molecule bound per cell-binding agent (e.g. , antibody) molecule can be determined spectroscopically by measuring the ratio of the absorbance at 280 nm and 252 nm. An average of about 0.5- about 20 cytotoxic
compounds/antibody molecule(s) can be linked by the methods described herein. In one embodiment, the average number of linked cytotoxic compound per cell-binding agent in the conjugate (i.e., average w value) is about 0.5 to about 10, about 0.5 to 2 (e.g., 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, or 2.1), about 2 to about 8 (e.g. , 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9,
4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0,
6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, or 8.1), about 2.5 to about 7, about 3 to about 5, about 2.5 to about 5.0 (e.g. , about 2.5, about 2.6, about 2.7, about 2.8, about 2.9, about 3.0, about 3.1, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, about 5.0), about 2.5 to about 4.0, about 3.0 to about 4.0, about 3.2 to about 4.2, or about 4.5 to 5.5 (e.g. , about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, or about 5.5).
IN VITRO EVALUATION OF CYTOTOXICITY
[180] The cytotoxic compounds and cell-binding agent-drug conjugates of the invention can be evaluated for their ability to suppress proliferation of various cancer cell lines in vitro. The in vitro cytotoxicity assays can be conducted using methods known in the art (e.g. , Widdison, W. C. et al. "Semisynthetic maytansine analogues for the targeted treatment of cancer," J. Med. Chem. (2006) 49(14):4392-408). For example, cells to be evaluated can be exposed to the compounds or conjugates for 1-5 days and the surviving fractions of cells measured in direct assays by known methods. IC50 values can then be calculated from the results of the assays. COMPOSITIONS AND METHODS OF USE
[181] The present invention includes a composition (e.g. , a pharmaceutical composition) comprising conjugates (e.g. , conjugates of Formula (Γ) or (I)) or cytotoxic compounds (e.g. , compounds of Formula (ΙΙΓ) or (III)) described herein, and a carrier (a pharmaceutically acceptable carrier). The present invention also includes a composition (e.g. , a pharmaceutical composition) comprising the conjugate of Formula (Γ) or (I) or the cytotoxic compound of Formula (ΙΙΓ) or (III), and a carrier (a pharmaceutically acceptable carrier), and further comprising a second therapeutic agent. The present compositions are useful for inhibiting abnormal cell growth or treating a proliferative disorder in a mammal (e.g. , human). The present compositions are also useful for treating an autoimmune disorder, a destructive bone disorder, a graft versus host disease, a transplant rejection, an immune deficiency, an inflammatory disease, an infectious disease, a viral disease, a fibrotic disease, a
neurodegenerative disorder, a pancreatitis or kidney disease in a mammal (e.g. , human).
[182] The present invention includes a method of inhibiting abnormal cell growth or treating a proliferative disorder in a mammal (e.g. , human) comprising administering to said mammal a therapeutically effective amount of conjugates (e.g. , conjugates of formula (Γ) or (I)) or cytotoxic compounds (e.g. , compounds of formula (ΙΙΓ) or (III)) described herein, or a composition thereof, alone or in combination with a second therapeutic agent. In one embodiment, the proliferative disorder is cancer in general; alternatively, the proliferative disorder is cancer selected from the group consisting of breast cancer, colon cancer, brain cancer, prostate cancer, kidney cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, colorectal cancer, gastric cancer, squamous cancer, small-cell lung cancer, nonsmall-cell lung cancer, testicular cancer, Merkel cell carcinoma, glioblastoma, neuroblastoma, a cancer of a lymphatic organ, and a hematological malignancy.
[183] Similarly, the present invention provides a method for inducing cell death in selected cell populations comprising contacting target cells or tissue containing target cells with an effective amount of the conjugates of the present invention. The target cells are cells to which the cell-binding agent of the conjugates can bind.
[184] If desired, other active agents, such as other anti-tumor agents, may be administered along with the conjugate.
[185] Cancer therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the Physician's Desk Reference (PDR). The PDR discloses dosages of the agents that have been used in treatment of various cancers. The dosing regimen and dosages of these aforementioned
chemotherapeutic drugs that are therapeutically effective will depend on the particular cancer being treated, the extent of the disease and other factors familiar to the physician of skill in the art and can be determined by the physician. The contents of the PDR are expressly incorporated herein in its entirety by reference. One of skill in the art can review the PDR, using one or more of the following parameters, to determine dosing regimen and dosages of the chemotherapeutic agents and conjugates that can be used in accordance with the teachings of this invention. These parameters include: Comprehensive index; Manufacturer; Products (by company's or trademarked drug name); Category index; Generic/chemical index (non- trademark common drug names); Color images of medications; Product information, consistent with FDA labeling; Chemical information; Function/action; Indications &
Contraindications; Trial research, side effects, warnings.
[186] The present invention also provides methods of treating a non-cancerous condition comprising administering to a subject in need of treatment an effective amount of any of the conjugates described above. Examples of the conditions include, but not limited to, an autoimmune disorder (e.g. , systemic lupus, rheumatoid arthritis, and multiple sclerosis), a graft versus host disease, a transplant rejection (e.g. , a renal transplant rejection, a liver transplant rejection, a lung transplant rejection, a cardiac transplant rejection, and a bone marrow transplant rejection), an immune deficiency, an inflammatory diseases (i.e. , myositis and pancreatitis), a destructive bone disorder, an infectious disease (e.g. , viral infections and parasite infections), a viral disease, a fibrotic disease, a neurodegenerative disorder, or a kidney disease. In one embodiment, the condition selected from the group consisting of cancer, rheumatoid arthritis, multiple sclerosis, graft versus host disease, transplant rejection, lupus, myositis, infectious disease, and immune deficiency.
[187] Examples of in vitro uses include treatments of autologous bone marrow prior to their transplant into the same patient in order to kill diseased or malignant cells: treatments of bone marrow prior to their transplantation in order to kill competent T cells and prevent graft- versus-host-disease (GVHD); treatments of cell cultures in order to kill all cells except for desired variants that do not express the target antigen; or to kill variants that express undesired antigen.
[188] The conditions of non-clinical in vitro use are readily determined by one of ordinary skill in the art. [189] Examples of clinical ex vivo use are to remove tumor cells or lymphoid cells from bone marrow prior to autologous transplantation in cancer treatment or in treatment of autoimmune disease, or to remove T cells and other lymphoid cells from autologous or allogenic bone marrow or tissue prior to transplant in order to prevent GVHD. Treatment can be carried out as follows. Bone marrow is harvested from the patient or other individual and then incubated in medium containing serum to which is added the cytotoxic agent of the invention, concentrations range from about 10 μΜ to 1 pM, for about 30 minutes to about 48 hours at about 37°C. The exact conditions of concentration and time of incubation, i.e., the dose, are readily determined by one of ordinary skill in the art. After incubation the bone marrow cells are washed with medium containing serum and returned to the patient intravenously according to known methods. In circumstances where the patient receives other treatment such as a course of ablative chemotherapy or total-body irradiation between the time of harvest of the marrow and reinfusion of the treated cells, the treated marrow cells are stored frozen in liquid nitrogen using standard medical equipment.
[190] For clinical in vivo use, the cytotoxic compounds or conjugates of the invention will be supplied as a solution or a lyophilized powder that are tested for sterility and for endotoxin levels. Examples of suitable protocols of conjugate administration are as follows.
Conjugates are given weekly for 4 weeks as an intravenous bolus each week. Bolus doses are given in 50 to 1000 ml of normal saline to which 5 to 10 ml of human serum albumin can be added. Dosages will be 10 μg to 2000 mg per administration, intravenously (range of 100 ng to 20 mg/kg per day). After four weeks of treatment, the patient can continue to receive treatment on a weekly basis. Specific clinical protocols with regard to route of
administration, excipients, diluents, dosages, times, etc., can be determined by one of ordinary skill in the art as the clinical situation warrants.
[191] Suitable pharmaceutically acceptable carriers, diluents, and excipients are well known and can be determined by those of ordinary skill in the art as the clinical situation warrants. Examples of suitable carriers, diluents and/or excipients include: (1) Dulbecco's phosphate buffered saline, pH about 7.4, containing or not containing about 1 mg/ml to 25 mg/ml human serum albumin, (2) 0.9% saline (0.9% w/v NaCl), and (3) 5% (w/v) dextrose; and may also contain an antioxidant such as tryptamine and a stabilizing agent such as Tween 20.
[192] The method for inducing cell death in selected cell populations can be practiced in vitro, in vivo, or ex vivo. ANALOGUES AND DERIVATIVES
[193] One skilled in the art of cytotoxic agents will readily understand that each of the cytotoxic agents described herein can be modified in such a manner that the resulting compound still retains the specificity and/or activity of the starting compound. The skilled artisan will also understand that many of these compounds can be used in place of the cytotoxic agents described herein. Thus, the cytotoxic agents of the present invention include analogues and derivatives of the compounds described herein.
[194] All references cited herein and in the examples that follow are expressly incorporated by reference in their entireties.
Example 1.
Figure imgf000057_0001
MayNMA-Gly3-mal
Z-Gly-Gly-Gly-P-Ala-OtBu (compound 3)
Figure imgf000057_0002
Z-Gly-Gly-Gly-OH (compound 1, 1.3 g, 4.0 mmol), tert-butyl-3-aminopropionate (compound 2, 0.583 g, 4.0 mmol), hydroxybenzotriazole (0.651 g, 4.25 mmol) and N-(3- dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (0.81 g, 4.23 mmol) were weighed into a 50 mL flask then dissolved in dimethylformamide (20 mL) and magnetic stirring under a nitrogen atmosphere. After 3 h the reaction solvent volume was reduced under vacuum to approximately (10 mL) and crude product was precipitated by trituration with anhydrous diethyl ether (45 mL). Product was precipitated by centrifugation and supernatant was decanted. The pellet was vigorously triturated with diethyl ether (35 mL) and centrifuged with decantation of supernatant. The product was gradually placed under vacuum to avoid bumping then placed under full vacuum once sufficient solvent had evaporated to give 1.35 g (75%) of the desired compound. 1H NMR (d6-DMSO) 8.16 (t, J =
5.2 Ηζ, ΙΗ), 8.10 (t, J = 5.2 Hz, 1H), 7.82 (t, J = 5.2 Hz, 1H), 7.25 - 7.4 (m, 5H), 5.04 (s, 2H), 3.74 (d, J = 5.6 Hz, 2H), 3.67 (t, J = 6.4 Hz, 4H), 3.25 (q, J = 6.1 Hz, 2H), 2.35 (t, J = 6.8 Hz, 2H), 1.39 (s, 9H). 13C NMR (d6-DMSO) 170.45, 169.61, 169.00, 168.63, 156.49, 136.94, 128.30, 127.76, 127.69, 79.89, 65.51, 43.56, 42.10, 41.90, 34.89, 34.78, 27.70. HRMS (M +Na+) Calc. 473.2012 found 473.1995.
H-Gly-Gly-Gly-P-Ala-OtBu (compound 4)
Figure imgf000058_0001
4
3
To a 250 mL capacity parr shaker flask was added a solution of Z-Gly-Gly-Gly-β- Ala-OtBu (compound 3, 1.3 g, 2.89 mmol) dissolved in 95:5 methanol :deionized water (80 mL) and 10% palladium on carbon (0.12 g) . The flask was shaken under an H2 atmosphere (42 PSI) for 3 h then vacuum filtered through celite filter aid and the filtrate was concentrated by rotary evaporation under vacuum to give 0.88 g (96%) of desired compound 4. 1H NMR
(d6-DMSO) 8.12 (t, J = 1.6Hz 2H), 8.08 (t, J =1.6 Hz, 1H), 3.75 (s,2H), 3.64 (d, J = 5.9 2H), 3.28 (bs, 2H), 3.24 (q, J = 6.0 Hz, 2H), 3.13 (s, 2H), 2.35 (t, J = 6.8 Hz, 2H), 1.39 (s, 9H). 13C NMR (d6-DMSO) 173.38, 170.46, 169.18, 168.70, 79.89, 44.65, 41.95, 34.88, 34.78, 27.71. HRMS (M + H+) Calc. 317.1825, found 317.1801 Mal-Gaba-Gly-Gly-Gly-P-Ala-OtBu (compound 5)
Figure imgf000059_0001
Maleoyl-Gaba-OH [4-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)butanoic acid] (513 mg, 2.8 mmol), H-Gly-Gly-Gly-P-Ala-OtBu [tert-butyl 3-(2-(2-(2- aminoacetamido)acetamido)acetamido)propanoate] (compound 4, 800 mg, 2.8 mmol) and /V- (3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (583 mg, 3.0 mmol) were dissolved in anhydrous dimethyl formamide (12 mL) and stirred for 3 h. The reaction mixture was purified in four equal portions by reverse phase HPLC using a 5.0 cm x 25 cm C18 column. The column was eluted at 100 mL/min with deionized water containing 0.3 % formic acid and 5% acetonitrile for 10 min followed by a 13 min linear gradient from 5% acetonitrile to 33 % acetonitrile. Product fractions (Rt 21 min) were combined and solvent was removed by rotary evaporation under vacuum to give 832 mg (62 %) of compound 5. 1H
NMR (d6-DMSO) 8.10-8.16 (m, 2H), 8.07 (t, J = 4.8 Hz, 1H), 7.0 - 7.15(m, 1H), 3.747 (t, J = 6.0 Hz, 3H), 3.64 (d, J = 5.6 Hz, 2H), 3.41 (t, J = 6.8, 2H), 3.1-3.33 (m, 1H), 3.19-3.26 (m, 2H), 2.348 (t, J = 6.8, 2H), 2.132 (t, J = 1.2 Hz, 2H), 1.67 - 1.76 (m, 2H), 1.39 (s, 9H). 13C NMR (d6-DMSO) 171.80, 170.98, 170.39, 169.48, 168.96, 168.56, 134.37, 79.83, 42.05, 41.83, 37.38, 34.82, 34.71, 32.26, 27.83, 23.95. HRMS (M + Na+) Calc. 504.2070 found 504.2046
Mal-Gaba-Gly-Gly-Gly-P-Ala-OH
Figure imgf000059_0002
Mal-Gaba-Gly-Gly-Gly-P-Ala-OtBu (compound 5, 820 mg, 1.7 mmol) was disolved in 95:5 trifluoroacetic acid: deionized water ( 9.0 mL) and magnetically stirred for 3 h.
Solvent was removed by rotary evaporation under vacuum to give 730 mg (100%) of the desired compound 6. 1H NMR (d6-DMSO) 12.1 (bs, 1H), 8.05-8.20 (m, 3H), 7.82 (t, J = 6.0
Hz, 1H), 7.00 (s, 2H), 3.71 (t, J = 6.0 Hz, 4H), 3.65 (d, J = 6.0 Hz, 2H), 3.41 (t, J = 1.2 Hz, 2H), 3.26 (q, J = 5.6 Hz, 2H), 2.38 (t, J = 7.2 Hz, 2H,), 2.14 (q, J = 8.0 Hz, 2H), 1.67-1.77 (m, 2H). 13C NMR (d6-DMSO) 172.70, 171.83, 171.01, 169.50, 168.99, 168.51, 134.38, 42.07, 41.84, 36.75, 34.70, 33.69, 32.28, 23.97 HRMS (M + Na+) Calc. 448.1444 found 448.1465.
Figure imgf000060_0001
MayNMA-Gly3-mal (compound 7)
To a solution of compound 6 (55mg, 0.130mmol) in DMF (3mL) was added HOBT (34mg, 0.221mmol) and EDC (lOOmg, 0.520mmol). The reaction was stirred until all reagents went into solution. This solution was added to a solution of crude DM' (85mg, 0.130mmol) in ethyl acetate (2.5mL). DM' was synthesized according to methods described in US Patent Nos. 7,301,019 and 7,598,375. The reaction was stirred overnight. The crude reaction mixture was purified by semi-preparative reverse phase HPLC (CI 8, 5-55% ACN over 14 minutes), and rotavaped to give yellow solid, compound 7. Low resolution MS [M+1]+ Calcd. 1057.4; found 1057.3 Low resolution [M-l]" Calcd. 1055.4; 1055.3.
Example 2.
Figure imgf000061_0001
Reaction of commercially available Boc-Gly-Gly-Gly-OH (compound 8) with N- hydroxyxuccinimide and EDC coupling agent affords compound 9. Reaction of compound 9 with l-(2-aminoethyl)-maleimide HCl in the presence of a base such as diisopropyl ethyl amine (DIPEA) followed by Boc deprotection with HCl in methoxymethyl ether gives compound 10. Reaction of compound 10 with glutamic anhydride gives compound 11. Reaction of compound 11 with DM' using EDC as coupling agent will give the desired product compound 12.
Similarly, compounds having different peptide moieties other than Gly-Gly-Gly in compound 12 can be prepared using different Boc protected peptides or Boc protected amino acids. Other cyclic anhydrides or a mono t-Butyl ester protected diacid can be used in place of glutaric anhydride and the t-Bu ester if present can be deprotected with trifluoroacetic acid to give other spacers between the DM' and peptide moieties.
Example 3.
Figure imgf000062_0001
Compounds of the present invention having a maleimide group and a tetrapeptide moiety (e.g., compound 17, wherein R21, R22 and R23 are amino acid side chains or protected amino acid side chains) can be prepared according to the synthetic scheme shown above. The Z (benzyloxy-carbonyl) protected tripeptide (compound 13) can be prepared by standard techniques (see, for example, Lloyd- Williams, P., Albericio, F., and Giralt, E. (1997) Chemical Approaches to the Synthesis of Peptides and Proteins (Boca Raton: CRC Press); and Benoiton, N. L. (2006) Chemistry of Peptide Synthesis, (Boca Raton: Taylor & Francis)). Compound 13 can then react with beta-alanine t-butyl ester to yield compound 14.
Deprotection of the Z group gives compound 15, which can with GMBS followed by t-butyl ester cleavage with trifluoroacetic acid to yield compound 16. The resulting compound 16 can then be coupled to DM' to give the desired maytansinoid peptide maleimide product having the peptide carboxy terminus near the DM' moiety and the peptide N terminus near the maleimide group (compound 17). When the amino acid side chains bear a carboxylic acid, it can be protected, typically as esters such as methyl or ethyl esters. For example, the carboxylic acid can be protected as the 2-(trimethylsilyl)ethanol ester or other photocleavable esters known in the art (Wuts, P. G. M., and Greene, T. W. (2007) Greene's protective groups in organic synthesis, 4th ed. (Hoboken, N.J.: Wiley- Interscience)), which can be removed by Bu4NF or treatment with light of appropriate wavelength. Other photocleavable protecting groups known to one skilled in the art (Greene's protecting groups in organic synthesis fourth ed.) can also be used for protecting the side chain carboxylic acid group. Amino acid side chains bearing a hydroxyl group can be protected, typically as esters such as acetyl or trifluoroacetyl esters. Amino acid side chains bearing an amine group can be protected, typically as a carbamate such as a methyl or ethyl carbamate. Deprotection of the above-mentioned protecting group can be carried out by standard methods known in the art. (Greene)
According to the synthetic methods described above, similar compounds of the present invention having a single amino acid or different peptide moiety can be prepared using t-Bu ester of the amino acid or peptide of interest in place of compound 15.
Example 4.
Figure imgf000063_0001
Compounds of the present invention having a maleimide group and a tripeptide moiety (e.g., compound 23, wherein R2i, R22 and R23 are amino acid side chains or protected amino acid side chains) can be prepared according to the synthetic scheme shown above. The Boc protected peptide (compound 19) can be prepared by standard techniques (see, for example, Lloyd- Williams, P., Albericio, F., and Giralt, E. (1997) Chemical Approaches to the Synthesis of Peptides and Proteins (Boca Raton: CRC Press); and Benoiton, N. L. (2006) Chemistry of Peptide Synthesis, (Boca Raton: Taylor & Francis)). Compound 19 can then be converted to an activated ester (compound 20) by reaction with N-hydroxysuccinimide and EDC. Compound 20 can then react with l-(2-aminoethyl)-maleimide HC1 followed by deprotection with HC1 to yield compound 21. Compound 21 can then react with glutaric anhydride to yield compound 22, which can be coupled to DM' using EDC to form compound 23.
When the amino acid side chains bear a carboxylic acid, it can be protected, typically as esters such as methyl or ethyl esters. For example, the carboxylic acid can be protected as the 2-(trimethylsilyl)ethanol ester or other photocleavable esters known in the art (Wuts, P. G. M., and Greene, T. W. (2007) Greene's protective groups in organic synthesis, 4th ed. (Hoboken, N.J.: Wiley- Interscience)), which can be removed by Bu4NF or treatment with light of appropriate wavelength. Other photocleavable protecting groups known to one skilled in the art (Greene's protecting groups in organic synthesis fourth ed.) can also be used for protecting the side chain carboxylic acid group. Amino acid side chains bearing a hydroxyl group can be protected, typically as esters such as acetyl or trifluoroacetyl esters. Amino acid side chains bearing an amine group can be protected, typically as a carbamate such as a methyl or ethyl carbamate. Deprotection of the above-mentioned protecting group can be carried out by standard methods known in the art. (Greene)
According to the synthetic methods described above, similar compounds of the present invention having a single amino acid or different peptide moiety can be prepared using Boc protected amino acid or peptide of interest in place of compound 19. The glutaric anhydride can also be replaced by a different cyclic anhydride. Alternatively, compound 22
(or similar compounds having a single amino acid or different peptide moiety) can be prepared by coupling compound 21 (or similar compounds having a single amino acid or different peptide moiety) with a mono t-butyl ester protected diacid with EDC followed by t- butyl ester deprotection with trifluoro acetic acid. Example 5.
Figure imgf000065_0001
Boc protected Ala-Ala-Ala-OH (compound 25) can be activated by reaction with N- hydroxysuccinimde and EDC coupling agent followed by reaction with hydrazine. The resulting hydrazide (compound 26) can be FMoc protected by reaction with FMoc-Cl and the Boc protecting group can be removed by reaction with dilute HC1. The resulting compound 27 can react with l,4-Dioxane-2,6-dione in the presence of DIPEA to give compound 28. DM' can be coupled to compound 28 using EDC and the FMoc protecting group can be removed by reaction with morpholine to give desired compound 29.
The scheme can be generalized by replacing Boc-Ala-Ala-Ala-OH (compound 25) with a Boc protected amino acid or peptide and the glutaric anhydride can also be replaced by a different cyclic anhydride or with a mono protected diacid to give other spacers between the DM' and peptide moieties. Example 6
Figure imgf000066_0001
Figure imgf000066_0002
Figure imgf000066_0003
Figure imgf000066_0004
Boc protected H-Gly-Gly-Gly-OH (compound 8) can be protected by reaction with a 9-Fluorenemethanol DIC coupling agent and dimethylaminopyridine (DMAP). The resulting compound 30 can be Boc deprotected with trifluoroacetic acid to give compound 31, which can the react with l,4-Dioxane-2,6-dione followed by protection reaction with
trimethylsilylethanol in the presence of DIC and DMAP to yield compound 32. Compound 32 can then be treated with morpholine to deprotect the 9-Fluorenemethane ester to give compound 33. DM' can then be coupled to compound 33 with EDC coupling agent and HOBT to afford compound 34. The trimethyl silylethane ester of compound 34 can be removed by treatment with tetrabutyl ammonium fluoride, followed by reaction with N- hydroxy succinimide to activate the carboxylic acid group and then with hydrazine to give desired compound 35.
The method can be generalized by replacing Boc-Gly-Gly-Gly-OH with a Boc- protected amino acid or peptide of choice. The l,4-Dioxane-2,6-dione can also be replaced by a different cyclic anhydride or with a mono trimethylsilylethane protected diacid to give other compounds of the present invention having other spacers between the DM' and the peptide moieties.
Example 7.
Figure imgf000067_0001
Boc protected Ala-Ala-Ala-OH (compound 25) can be activated by reaction with N- hydroxysuccinimide (NHS) and EDC, followed by reaction with 1,2-diaminoethane to give compound 36. Compound 36 can be treated with FMoc-aminoxyacetic acid in the presence EDC coupling agent followed by Boc deprotection with dilute HCI. The resulting compound 37 can react with l,4-Dioxane-2,6-dione to give compound 38.
Coupling of DM' with compound 38 in the presence of EDC followed by hydroxyl amine deprotection with morpholine will give the final product, compound 39. A similar method can be used with a Boc protected amino acid or a Boc protected peptide to generate compounds of the present invention with different peptide side chains. The l,4-Dioxane-2,6-dione can also be replaced by a different cyclic anhydride or with a mono protected diacid to give other spacers between the DM' and peptide moieties.
Example 8.
A. Conjugation of cell-binding agent to compounds of formula (III) in which JCB is a maleimide group
Figure imgf000068_0001
A cell-binding agent bearing one or more thiols will be reacted with a maytansinoid bearing a maleimide moiety. Low molecular weight impurities can be removed from the conjugate by, for example, diafiltration, tangential flow filtration, size exclusion
chromatography or by other chromatographic techniques known to one skilled in the art. If any thiols remain un-modified they can be left unreacted or they can optionally be allowed to form internal disulfide bonds with other free thiols in the cell-binding agent or they can optionally be capped by a thiol-reactive quenching agent, such as N-ethyl maleimide or 2- bromo acetamide. B. Conjugation of cell binding agent to compounds of formula (III) in which J, hydrazide group
Figure imgf000069_0001
A cell binding agent bearing one or more aldehydes or ketones will be reacted with a maytansinoid bearing a hydrazide moiety. Any unreacted aldehydes or ketones can be left unreacted or can optionally be reacted with a separate reagent such as the hydrazide of acetic acid. Low molecular weight impurities can optionally be removed from the conjugate by, for example, diafiltration, tangential flow filtration, size exclusion chromatography or by other chromatographic techniques known to one skilled in the art. The conjugate can optionally be reduced with a reducing agent such as but not limited to sodium cyanoborohydride or sodium triacetoxy borohydride.
C. Conjugation of cell binding agent to compounds of formula III in which JCB is a an alkoxy amine group
Figure imgf000070_0001
A cell binding agent bearing one or more aldehydes or ketones will be reacted with a maytansinoid bearing an alkoxyamine moiety. Any unreacted aldehydes or ketones can be left unreacted or can optionally be reacted with a separate reagent such as the hydrazide of acetic acid. Low molecular weight impurities can optionally be removed from the conjugate by, for example, diafiltration, tangential flow filtration, size exclusion chromatography or by other chromatographic techniques known to one skilled in the art. The conjugate can optionally be reduced with a reducing agent such as but not limited to sodium
cyanoborohydride or sodium triacetoxy borohydride.
Example 9.
Figure imgf000071_0001
Synthesis of compound 36b
Figure imgf000071_0002
Boc-Gly3-OH (25b, 6 g, 20.7 mmol) was dissolved in DMF (40 mL) to which was added N-hydroxysuccinimide (2.4 g, 20.7 mmol) and EDC (4 g, 20.7 mmol) in a flask containing a magnetic stir bar. The reaction was magnetically stirred for 1 hour then slowly poured into a magnteically stirred solution of ethylene diamine (8 g, 133 mmol) in DMF (20 mL) with vigorous magnetic stirring at room temperature. Diethyl ether (200 mL) was added to precipitate a solid. The flask was then vortexed and placed in a sonication bath for 15 min. The material was vacuum filtered then dried under vacuum at room temperature then dissolved in DMF (20 mL) and purified in three runs using approximatly equal injection volumes on a 50 mm x 220 mm load and lock CI 8 column with 220 nm detection eluting at 100 mL/min with deionized water containing 0.2% formic acid and an acetonitrile gradient of 5 % acetonitrile for the first 5 min then a linear gradient of 5 % - 95% acetonitrile from 5 to 32 min. Fractions containing pure desired product were combined, frozen and lyophilized to give 4.5 g (65% yield) of desired product 36b.
Synthesis of compound 36c
Figure imgf000072_0001
Fmoc aminoxyacetic acid (187 mg, 0.60 mmol) was dissolved in anhydrous DMF (1 mL) to which was added TBTU (243 mg, 0.76 mmol) and DIPEA (95 μί, 0.54 mmol) and magnetically stirred for 3 min. A solution of Boc-Gly3NH-(CH2)2-NH2 (200 mg, 0.54 mmol) in anhydrous DMF (1 mL) was then added with magnetic stirring. After 1 h the reaction was directly purified on a 27 mm x 220 mm load and lock CI 8 column 40 mL/min with 254 nm detection eluting with deionized water containing 0.2% formic acid and an acetonitrile gradient of 10% acetonitrile for the first 5 min then a linear gradient of l0% - 95%
acetonitrile from 5 to 32 min. Fractions containing pure desired product were combined, frozen and lyophilized to give 60 mg ( 17 % yield) of desired product 36c. 1H NMR (400 MHz, DMSO- 6) δ 1.37 (s, 9H), 3.15 (dt, J = 10.0, 5.6 Hz, 4H), 3.58 (d, J = 6.0 Hz, 2H), 3.67 (d, J = 5.8 Hz, 2H), 3.74 (d, J = 5.6 Hz, 2H), 4.16 (s, 2H), 4.26 (t, J = 6.7 Hz, 1H), 4.43 (d, J = 6.7 Hz, 2H), 7.01 (t, J = 6.0 Hz, 1H), 7.33 (td, J = 7.4, 1.2 Hz, 2H), 7.42 (t, J = 7.4 Hz, 2H), 7.68 (d, J = 7.4 Hz, 2H), 7.85 (d, J = 6.0 Hz, 1H), 7.89 (d, J = 7.5 Hz, 2H), 7.97 (d, J = 5.7 Hz, 1H), 8.05 (t, J = 5.6 Hz, 1H), 8.10 (t, J = 6.0 Hz, 1H). HRMS calcd. 649.2592, found 649.2588.
Synthesis of compound 37b
Figure imgf000072_0002
A 25 mL round bottom flask was charged with Boc-Gly3-NH(CH2)2NHCOCH2-0- NH-FMoc (76 mg, 0.122 mmol) and dissolved in a solution of 95/5 trifluoroacetic acid in water (10 mL). The reaction was magnetically stirred at room temperature for 1 hour. The reaction volume was reduced in vacuo and the crude residue was redissolved in a 1: 1 mixture of methylene chloride and toluene (10 mL) then concentrated in vacuo. The coevaporation was repeated three times and the resulting product (37b) was used without further purification. MS (m + H+) found: 527.4, calcd. 527.2; (m-1).
Sythesis of compound 38b
Figure imgf000073_0001
A 10 mL round bottom flask was charged with the t-butyl deprotected H-Gly3- NH(CH2)2NHCOCH2ONH-Fmoc (64 mg, 0.122 mmol) and DMF (3 mL). The solution was stirred as diglycolic anhydride (15.52 mg, 0.134 mmol) and N,N-diisopropylethylamine (0.042 mL, 0.243 mmol) were sequentially added. The flask was equipped with a septum and magnetically stirred at room temperature for 1 hour. The desired product was isolated by semi-preparative CI 8 HPLC eluting with deionized water containing 0.1% formic acid and a linear gradient of acetonitrile 5 - 95% over 25 min. Product containing fractions were combined and concentrated in vacuo to give 40 mg (0.062 mmol, 51.2% yield) of the desired product (38b) as a white residue. MS (m + H+) found: 643.4, calcd. 643.2; (m-1) found: 641.2, calculated: 641.2.
Figure imgf000073_0002
A 10 mL round bottom flask was charged with a solution of May-NMA (5.06 mg, 7.78 μιηοΐ) in ethyl acetate (1.500 mL). N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (EDC, 3.73 mg, 0.019 mmol) and N,N-diisopropylethylamine (3.39 μΐ, 0.019 mmol) were then added followed by a solution of HOCOCH2OCH2CO- Gly3NH(CH2)2NHCOCH2ONH-FMoc (10 mg, 0.016 mmol) in N,N-dimethylformamide (1.5 mL). The reaction was stirred for 1 hour at room temperature. The reaction volume was reduced by half in vacuo and the product was isolated by semi-preparative C18 HPLC eluting with deionized water containing 0.1% formic acid and a linear gradient of acetonitrile 5 - 95% over 25 min. Product containing fractions were combined, frozen and lyophilized to give desired product (38c) 1.4 mg (13 % yield) as a white solid. MS (m + Na+) found:
1296.7, calcd: 1296.5.
Synthesis of compound 39b
Figure imgf000074_0001
May-NMA-COCH2OCH2CO-Gly3NH(CH2)2NHCH2ONH-Fmoc (1.4 mg, 1.098 μιηοΐ) was dissolved in N,N-dimethylformamide (1.5 mL) and transferred to a 3 mL glass vial equipped with a stir bar. The solution was stirred as 20% v/v morpholine (300 μί, 3.44 mmol) was added. The reaction proceeded with stirring at room temperature until determined complete. The crude reaction mixture was purified by semi-preparative C18 HPLC over 3 injections. Product containing fractions were combined, transferred to a scintillation vial, frozen in a bath of dry ice and acetone and lyophilized to give 1.1 mg ( 95% yield) of desired product (39b). MS (m + Na+) found: 1074.7, calculated: 1074.4; high-resolution MS (m+H)+ found: 1052.4331, calculated: 1052.4338; (m + C1-) found: 1086.5, calculated: 1086.4.
Figure imgf000075_0001
Figure imgf000075_0002
H-Gly3-beta-Ala-0-tBu (150 mg, 0.47 mmol), 5-azidopentanoic acid (88 mg, 0.62 mmol), HOBT (145 mg, 0.95 mmol) and EDC (270 mg, 1.4 mmol) were weighed into a flask containing a magnetic stir bar and dissolved in anhydrous DMF (2 mL). The reaction was magnetically stirred for 2 hours then purified on the 27 mm x 220 mm load and lock C18 column 40 mL/min with 220 nmdetection eluting with deionized water containing 0.2% formic acid using 5% acetonitrile for the first 5 min then a linear ramp of 5% to 95% acetonitrile from 5 min to 32 min. Detection 220 nm and Mass spec. Fractions containing pure desired product were combined, frozen and lyophilized to give 70 mg (33% yield) of desired product. 1H NMR (400 MHz, DMSO- 6) δ 1.40 (s, 9H), 1.49 - 1.59 (m, 5H), 2.17 (t, / = 6.8 Hz, 3H), 2.35 (t, / = 7.1 Hz, 4H), 3.24 (q, / = 6.8 Hz, 4H), 3.64(d, / = 5.8 Hz, 2H), 3.69 - 3.75 (m, 4H). HRMS calcd. 464.2228, found 464.2227.
Figure imgf000075_0003
A 10 mL round bottom flask was charged with N3(CH2)4CO-Gly3NH(CH2)2COO-t-Bu (50 mg, 0.113 mmol) and a solution of 95/5 trifluoroacetic acid in water (5 mL) was magnetically stirred for 1 hour at room temperature then concentrated in vacuo. The crude residue was redissolved in a 1 :1 solution of methylene chloride and toluene (10 mL) then concentrated in vacuo. The coevaporation was repeated three times and the resulting product was used without further purification.
A 10 mL round bottom flask was charged with May-NMA (9.62 mg, 0.015 mmol) in ethyl acetate and a stir bar. The solution was stirred as JV-(3-Dimethylaminopropyl)-JV- ethylcarbodiimide hydrochloride (EDC, 7.09 mg, 0.037 mmol) and JV,JV-diisopropylethylamine (4.78 mg, 0.037 mmol) were then added. A solution of HOOC(CH2)2-NH-Gly3-(CH2)4-N3 (11.4 mg, 0.030 mmol) in , -dimethylformamide (1.5 mL) was then added and the reaction was magnetically stirred at room temperature for 1 hour. The reaction volume was reduced by half in vacuo and the remaining crude JV,JV-dimethylformamide mixture was purified by semi-preparative CI 8 HPLC eluting with deionized water containing 0.1 % formic acid using a gradient of acetonitrile from 5% - 95% over 30 min. Fractions containing desired product were combined, frozen and lyophilized to give 7.6 mg (51% yield) of desired product as a white solid. MS (m + Na+) found: 1039.7, calcd: 1039.4; (m + C1-) found: 1052.5, calculated: 1051.4; high-resolution MS (m+H)+ found: 1017.4415; calcd: 1017.4443..
Example 11. Synthesis of aminoxy-Gl rMayNMA
Figure imgf000076_0001
Figure imgf000077_0001
Fmoc-Gly3-MayNMA
A 100 mL flask was charged with a solution of MayNMA (200mg, 0.308mmol) in ethyl acetate (19 mL). The reaction flask was concentrated in vacuo to remove EtOAC. The material was re-dissolved in DMSO (10 mL), treated with FMoc-GlyrOH (165 mg, 0.400mmol), HOBT (12.25mg, 0.080mmol), EDC (77mg, 0.400mmol) and DIPEA (53.ΊμΙ,, 0.308mmol). The reaction was allowed to proceed at room temperature under argon. After 1 hour the reaction was purified using a Combiflash Rf 200i using C18 high performance gold 30g column, at 35 mL/min. Eluting with deionized water containing 0.1% formic acid and an acetonitrile gradient of 5 - 95% over 20 min. Fractiosn containing desired product were combined, frozen and lyophilized to give desired product 296 mg (93% yield). MS (M)+ found 1043.4, calcd.: 1043.4
Figure imgf000077_0002
H-Gly3-MayNMA
FMoc-Gly3-MayNMA (37.7mg, 0.036mmol) was dissolved in 20% morpholine in DMSO (5 mL) and magnetically stirred for 1 hour. The reaction was purified using a Combiflash Rf 200i using CI 8 high performance gold 30 g column at 35 mL/min. Eluting with deionized water containing 0.1% formic acid with an acetonitrile gradient 2-95% over 25 min. Fractions containing desired product were combined, frozen and lyophilized to desired product 20 mg ( 75 % yield). LRMS (M)+ found 821.40, calculated: 821.34
Figure imgf000078_0001
Fmoc-Aminoxy-Gly3-MayNMA
H-Gly3-MayNMA (15 mg, 0.018mmol) was dissolved in DMSO (2 mL), to which FMoc- aminoxyacetic acid (11.44mg, 0.037mmol), DIPEA (3.19μΕ, 0.018mmol) and EDC (7.0mg, 0.037mmol) were added. After 1 hour the crude material was purified by semi-preparative CI 8 HPLC using a XB-C18 21.2x150mm, 5μιη column with a flow rate of 21.2mL/min. Eluting with deionized water containing 0.1% formic acid and 5% acetonitrile for 3 min then a linear gradient of 5% - 95% acetonitrile from 5 - 25 min. Fractions containing desired product were combined, frozen and lyophilized to give 2 mg (8% yield) of desired product. MS (M+Na)+ found 1138.6, calculated: 1138.4
Figure imgf000078_0002
Aminoxy-Gly3-MayNMA
FMoc-aminoxy-Gly3-MayNMA (2 mg, 18 μιηοΐ) was treated with a solution of 20% morpholine in DMSO (1 mL) with magnetic stirring at room temperature for 2 hours. The reaction was purified by semi-preparative CI 8 HPLC using a XB-C18 21.2x150mm, 5μιη column with a flow rate of 21.2 mL/min. Eluting with deionized water containing 0.1% formic acid and 5% acetonitrile for 5 min then a linear gradient of acetonitrile 5% - 95% from 5 min - 25 min. Fractions containing desired product was immediately collected, frozen and lyophilized to yield aminoxy-Gly3-MayNMA (0.2mg, 0.224μηιο1). LRMS (M+Na)+ found 916.60, calcd: 916.36; HRMS (M+Na)+ found: 916.3466; calcd: 916.3466. -amido-norbornene
Figure imgf000079_0001
To a 0.088M solution of MayNMA (28.8mg, 0.044 mmol) in DMF was added a solution of (lR,4R)-bicyclo[2.2.1]hept-5-ene-2-carboxylic acid (0.014 g, 0.102 mmol) and EDC (0.021 g, 0.111 mmol) in an equal volume of DMF. The reaction mixture was magnetically stirred under argon at room temperature for 3h. The crude reaction mixture was purified by reverse-phase HPLC (C18, 21.2 x 250mm) eluting with deionized water containing 0.1% formic acid using an acetonitrile gradient 5-95% over 30 min. Desired product was collected, frozen then lyophilized to give desired product 9 mg (26% yield) as a white solid. MS (M+l)+ found: 770.30, calculated: 770.3414. 1H NMR (400 MHz,
Chloroform-d) δ 0.79 (d, 6H), 1.67 - 1.74 (m, 1H), 0.99 - 1.09 (m, 1H), 1.13 - 1.20 (m, 2H), 1.20 - 1.26 (m, 5H), 1.29 (d, J = 6.3 Hz, 9H), 1.32 - 1.40 (m, 1H), 1.38 - 1.51 (m, 4H), 1.50
- 1.57 (m, 3H), 1.57 - 1.63 (m, 4H), 1.65 (s, 3H), 1.67 - 1.73 (m, 1H), 2.12 - 2.23 (m, 3H), 2.54 - 2.68 (m, 2H), 2.78 - 2.83 (m, 1H), 2.86 (s, 5H), 2.98 (s, 3H), 3.02 (dd, J = 9.5, 5.5 Hz, 4H), 3.06 - 3.19 (m, 3H), 3.21 (s, 6H), 3.25 - 3.29 (m, 1H), 3.33 (s, 6H), 3.48 (t, J = 8.6 Hz, 3H), 3.58 (d, J = 12.7 Hz, 1H), 3.77 (d, J = 12.6 Hz, 1H), 3.98 (d, J = 5.3 Hz, 6H), 4.23 - 4.34 (m, 2H), 4.72 - 4.85 (m, 2H), 5.21 (s, 1H), 5.35 (q, J = 6.8 Hz, 1H), 5.59 - 5.71 (m, 2H), 5.88
- 5.95 (m, 1H), 6.09 - 6.15 (m, 1H), 6.15 - 6.20 (m, 1H), 6.23 (d, J = 4.9 Hz, 2H), 6.31 - 6.48 (m, 3H), 6.57 (s, 1H), 6.64 (d, J = 11.1 Hz, 1H), 6.74 - 6.88 (m, 4H). Example 13. MayNMA-amidopent;
Figure imgf000080_0001
To a 0.088M solution of MayNMA (28.8mg, 0.044 mmol) in DMF was added a solution of 5-azidopentanoic acid (15.0 mg, 0.105 mmol) and EDC (21.2 mg, 0.111 mmol) in an equal volume of DMF. The reaction mixture was magnetically stirred under argon at room temperature for 3h. The crude reaction mixture was purified by reverse-phase HPLC (CI 8, 21.2 x 250mm) eluting with deionized water containing 0.1% formic acid using an acetonitrile gradient 5-95% over 30 min to give 11.6 mg (34% yield) of desired product as a white solid. MS (M+l)+ found: 775.35, calculated: 775.3428. 1H NMR (400 MHz,
Chloroform-d) δ 0.80 (s, 3H), 1.18 - 1.27 (m, 2H), 1.27 - 1.33 (m, 6H), 1.38 - 1.54 (m, 2H),
I.54 - 1.67 (m, 7H), 1.66 - 1.84 (m, 3H), 2.00 (s, OH), 2.18 (dd, J = 14.4, 3.2 Hz, 1H), 2.21 - 2.33 (m, 1H), 2.35 - 2.49 (m, 1H), 2.61 (dd, J = 14.4, 12.1 Hz, 1H), 2.84 (s, 3H), 2.98 - 3.07 (m, 1H), 3.12 (d, J = 12.7 Hz, 1H), 3.19 (d, J = 2.9 Hz, 3H), 3.21 - 3.29 (m, 2H), 3.35 (d, J = 3.4 Hz, 4H), 3.50 (d, J = 9.1 Hz, 1H), 3.65 (d, J = 12.7 Hz, 1H), 3.98 (s, 3H), 4.28 (td, J =
I I.3, 10.3, 2.1 Hz, 1H), 4.78 (dd, J = 12.0, 3.1 Hz, 1H), 5.38 (q, J = 6.8 Hz, 1H), 5.66 (dd, J = 15.3, 9.1 Hz, 1H), 6.23 (s, 1H), 6.43 (dd, J = 15.4, 11.1 Hz, 1H), 6.66 (d, J = 1.8 Hz, 1H), 6.72 (d, J = 11.1 Hz, 1H), 6.83 (d, J = 1.9 Hz, 1H).
Example 14. May-NMA-amidopent-4-yne
Figure imgf000080_0002
To a 0.066M solution of MayNMA (21.6mg, 0.033 mmol) in DMF was added a solution of pent-4-ynoic acid (4.9 mg, 0.050 mmol) and EDC (12.7 mg, 0.066 mmol) in an equal volume of DMF. The reaction mixture was magnetically stirred under argon at room temperature for lh. The crude reaction mixture was purified by reverse-phase HPLC (CI 8, 21.2 x 250mm) eluting with deionized water containing 0.1% formic acid using an acetonitrile gradient 5-80% over 30 min. Fractions containing desired product were combined, frozen and lyophilized to give 5 mg (21% yield) of desired product as a white solid. MS (M+l)+ found: 730.50, calculated: 730.3101. 1H NMR (400 MHz, Chloroform-d) δ 0.79 (s, 3H), 1.17 - 1.27 (m, 2H), 1.30 (dd, J = 6.6, 4.7 Hz, 6H), 1.40 - 1.53 (m, 2H), 1.56 (dt, J = 13.8, 1.8 Hz, 1H), 1.64 (s, 3H), 1.83 (t, J = 2.5 Hz, 1H), 2.18 (dd, J = 14.3, 3.1 Hz, 1H), 2.38 - 2.52 (m, 3H), 2.52 - 2.72 (m, 4H), 2.85 (s, 3H), 3.00 - 3.06 (m, 1H), 3.10 (d, J = 12.7 Hz, 1H), 3.21 (s, 3H), 3.35 (d, J = 3.0 Hz, 3H), 3.50 (d, J = 9.0 Hz, 1H), 3.66 (d, J = 12.7 Hz, 1H), 3.98 (s, 3H), 4.22 - 4.35 (m, 2H), 4.76 (dd, J = 12.1, 3.1 Hz, 1H), 5.42 (q, J = 6.8 Hz, 1H), 5.63 (dd, J = 15.4, 9.1 Hz, 1H), 6.37 (s, 1H), 6.43 (dd, J = 15.4, 11.2 Hz, 2H), 6.64 (d, J = 1.9 Hz, 1H), 6.73 (d, J = 11.1 Hz, 1H), 6.82 (d, J = 1.9 Hz, 1H), 8.01 (s, 1H).
Figure imgf000081_0001
Example 15. Synthesis of Aminoxy-MayNMA
Figure imgf000081_0002
FMoc protected aminoxy MayNMA
A lOOmL flask was charged with MayNMA (150mg, 0.231mmol) in ethyl acetate (35 mL). The reaction flask was concentrated in vacuo to remove EtOAC. The material was re- dissolved in DMF (5 mL), treated with FMoc-aminoxyacetic acid (72.3 mg, 0.231 mmol) followed by EDC (44.2 mg, 0.231 mmol) with magnetic stirring. The reaction was allowed to proceed under argon at room temperature for 4 hours then purified by semi-preparative CI 8 HPLC using a XB-C18 21.2x150mm, 5μιη column with a flow rate of 21.2mL/min. Eluting with deionized water containing 0.1% formic acid and an acetonitrile gradient of 5% for the first 5 min then 5% - 95% from 5 min to 25. Fractions containing desired product were combined frozen and lyophilized to give 24.2mg (11 % yield) of desired product. LRMS (M)+ found 945.35, calculated: 945.36
Figure imgf000082_0001
Aminoxy-MayNMA
FMoc protected aminoxy MayNMA (24.2mg, 0.026mmol) was treated 20% morpholine in DMF (2 mL). After 1 hour with magnetic stirring the reaction was purified by semi-preparative C18 HPLC using a XB-C18 21.2x150mm, 5μιη column with a flow rate of 21.2mL/min. Eluting with deionized water containing 0.1% formic acid and an acetonitrile gradient of 5% for 5 min then a linear gtradient of 5% - 95% over 25 min. Fractions containing desired product were combined, frozen and lyophilized to give lOmg (54.0% yield) of desired product. MS (M)+ found: 723.3, calculated: 723.3; high-resolution MS found: 723.2995; calculated: 723.3003.
Example 16. Preparation of chB38.1-SFB-MayNMA
40 mM stock solution of SFB (Succinimidyl 4-formylbenzoate) (Pierce) in DMA
(dimethylacetamide) and 20 mM stock of aminooxy-acetyl-MayNMA in DMA were prepared. 10 molar equivalents of SFB linker was added to a buffered solution of chB38.1 antibody at 4 mg/ml in 50 mM EPPS (4-(2-hydroxyethyl)piperazine-l-propanesulfonic acid), pH 8.5. Reaction proceeded for 90 min room temperature followed by desalting in NAP columns (Illustra Sephadex G-25 DNA Grade, GE Healthcare) equilibrated in PBS pH 7.4 to remove unconjugated linker.
Then 2 molar equivalents of aminooxy-acetyl-MayNMA were added per SFB linker and the oxamination reaction was allowed to proceed at 37 °C for 5 hr. Again, the resulting conjugate was desalted in NAP columns (Illustra Sephadex G-25 DNA Grade, GE
Healthcare) equilibrated in phosphate buffereed saline pH 7.4 (PBS) to remove unconjugated maytansinoid. Dialysis of conjugate into 10 mM histidine, 250 mM glycine, 1% sucrose pH 5.5 was then carried out at 4 °C using Slide-a-Lyzer dialysis cassettes (ThermoScientific, 20,000 MWCO).
The purified conjugate was found to have an average of 4.9 maytansinoid molecules linked per antibody (by UV-VIS using molar extinction coefficients £252 nm= 33,874 cm^M"1 and e28o nm= 9230 cm^M"1 for SFB-MayNMA, and e28o nm= 213,445 cm^M"1 and e252 nm= 72,000 cm" M_1 for chB38.1 antibody), 97% monomer (by size exclusion chromatography), 2% unconjugated maytansinoid (by HISEP mixed mode chromatography), a final protein concentration of 2.3 mg/ml, and an overall 78% protein yield.
Figure imgf000083_0001

Claims

We claim:
1. A conjugate represented by the following formula, or a pharmaceutically acceptable salt thereof:
Figure imgf000084_0001
wherein:
CBA is a cell binding agent;
DM is a drug moiet represented by the following formula:
Figure imgf000084_0002
R, R' and R", for each occurrence, are independently H or an optionally substituted alkyl;
one of Ei and E2 is -C(=0)-, and the other is -NR9-; or one of Et and E2 is - C(=0)- or -NR9-, and the other is absent;
P is [XX] 1-10, in which each XX is a residue of an independently selected amino acid, or P is -(NRm-CH2CH2)s-;
s is an integer between 1 and 5; Rmis H, or alkyl optionally substituted with a charged substituent or an ionizable group;
Zi and Z2 are each independently absent, -S02NR9-, -NR9S02-, -C(=0)-NR9-, -NR9-C(=0)-, -C(=0)-0-, -0-C(=0)-, -CH2-0-, -0-CH2-, -(CH2CH20)p- or - (OCH2CH2)p-, -NR9-C(=0)-CH2-, or -CH2-C(=0)-NR9- wherein p and p' are independently an integer from 1 to 1000;
Figure imgf000085_0001
Figure imgf000086_0001
which si is the site covalently linked to the CBA, s2 is the site covalently linked to the group X\ in formula (Γ) or (CR3R4)nin formula (I),
JCBI' for Formula (V) is
Figure imgf000086_0002
N-NRe— ^ s2 s1 =N-NRe-C(=0)— s2 s1 -N"NRe-C(=0)— s2
N-NRe— Ar J s2 s1 — N_NRe— Ar s2 s1 ^=N-0— j s2
Figure imgf000086_0003
Figure imgf000087_0001
which si is the site covalently linked to the CBA, s2 is the site covalently linked to the group Z1;
Ar is an optionally substituted arylene or an optionally substituted
heteroarylene, Cy is the non-alkyne residue of an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne;
R201, R202 and R203 each are independently H or an optionally substituted alkyl;
Z3 is pyridyl or
Figure imgf000087_0002
, wherein R2o4 and R205 are each
independently optionally substituted alkyl;
Cy' is the non-alkene residue of an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene;
R301 is H or optionally substituted alkyl; Ra, Rb, Rc, Re, Ri to R6, and R9, for each occurrence, are independently H or an optionally substituted alkyl;
m and n, for each occurrence, are independently an integer between 0 and 10; and
w is an integer between 1 and 20.
2. The conjugate of claim 1 , wherein the conjugate represented by the following
formula, or a pharmaceutically acceptable salt thereof:
Figure imgf000088_0001
wherein:
CBA is a cell binding agent;
DM is a drug moiety represented by the following formula:
Figure imgf000088_0002
R, R' and R", for each occurrence, are independently H or an optionally substituted alkyl;
one of Ei and E2 is -C(=0)-, and the other is -NRg-; P is [XX]1-10, in which each XX is a residue of an independently selected amino acid, or P is -(NRm-CH2CH2)s-;
s is an integer between 1 and 5;
Rmis H, or alkyl optionally substituted with a charged substituent or an ionizable group;
Z\ and Z2 are each independently absent, -S02NR9-, -NR9S02-, C(=0)-NR9-, -NR9-C(=0)-, -C(=0)-0-, -0-C(=0)-, -CH2-0-, -0-CH2-, -(CH2CH20)P- or - (OCH2CH2)p -, wherein p and p' are independently an integer from 1 to 1000;
JCB' for Formula (Γ) or (I) is
Figure imgf000089_0001
s1
Figure imgf000089_0002
H
s1 §=N-NRe~Ar— s2 s1 -N-NRe— Ar < s2 s1 s=N-0— $ s2
Figure imgf000089_0003
Figure imgf000090_0001
which si is the site covalently linked to the CBA, s2 is the site covalently linked to the group X\ in formula (Γ) or (CR3R4)nin formula (I),
JCBI' for Formula (V) is
Figure imgf000090_0002
N-NRe— ^ s2 s1 =N-NRe-C(=0)— s2 s1 -N"NRe-C(=0)— s2
N-NRe— Ar J s2 s1 — N_NRe— Ar s2 s1 ^=N-0— j s2
Figure imgf000090_0003
Figure imgf000091_0001
which si is the site covalently linked to the CBA, s2 is the site covalently linked to the group Z1;
Ar is an optionally substituted arylene or an optionally substituted
heteroarylene, Cy is the non-alkyne residue of an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne;
R201, R202 and R203 each are independently H or an optionally substituted alkyl;
Z3 is pyridyl or
Figure imgf000091_0002
, wherein R2o4 and R205 are each
independently optionally substituted alkyl;
Cy' is the non-alkene residue of an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene;
R301 is H or optionally substituted alkyl; Ra, Rb, Rc, Re, Ri to R6, and R9, for each occurrence, are independently H or an optionally substituted alkyl;
m and n, for each occurrence, are independently an integer between 0 and 10; and
w is an integer between 1 and 20.
3. The conjugate of claim 1 or 2, wherein w is an integer between 1 and 4.
4. The conjugate of any one of claims 1 to 3, wherein the ionizable group is -SO3H, -Z'- SO3H, -OP03H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, -C02H, -Z'C02H, -NRnRi2, or - Z'-NRuRi2, and the charged group is -N+R13R14R15X~, or -Z'-N+R13R14R15X~; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R12, for each occurrence, are independently H or an optionally substituted alkyl; R1 to R15 are each independently an optionally substituted alkyl; and X" is a pharmaceutically acceptable anion.
5. The conjugate of any one of claims 1 to 4, wherein Rmis H or -CH2CH2S03H or a pharmaceutically acceptable salt thereof.
6. The conjugate of any one of claims 1 to 5, wherein P is [XX] 2-4.
7. The conjugate of claim 6, wherein P is [XX] 3.
8. The conjugate of any one of claims 1 to 7, wherein each XX is the residue of an
independently selected amino acid selected from: a naturally occurring amino acid, an unnatural amino acid, an amino acid analog, or an amino acid mimetic that functions in a manner similar to the naturally occurring amino acids.
9. The conjugate of claim 8, wherein each XX is the residue of an independently
selected amino acid selected from the group consisting of: Histidine, Alanine,
Isoleucine, Arginine, Leucine, Asparagine, Lysine, Aspartic acid, Methionine,
Cysteine, Phenylalanine, Glutamic acid, Threonine, Glutamine, Tryptophan, Glycine,
Valine, Proline, Serine, Tyrosine, N-methyl-Histidine, N-methyl-Alanine, N-methyl-
Isoleucine, N-methyl-Arginine, N-methyl-Leucine, N-methyl-Asparagine, N-methyl-
Lysine, N-methyl-Aspartic acid, N-methyl-Methionine, N-methyl-Cysteine, N- methyl-Phenylalanine, N-methyl-Glutamic acid, N-methyl-Threonine, N-methyl-
Glutamine, N-methyl-Tryptophan, N-methyl-Glycine, N-methyl- Valine, N-methyl-
Proline, N-methyl-Serine, N-methyl-Tyrosine, hydroxyproline, γ-carboxyglutamate, selinocysteine, 0-phosphoserine, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, citrulline, Ornithine, cysteine sulfonic acid, cysteine sulfinic acid, 3-aminoalanine, 3-dimethylaminoalanine, 2-amino-4- (dimethylamino)butanoic acid, 2,4-diaminobutanoic acid, 2-amino-6- (dimethylamino)hexanoic acid, 2-amino-5-(dimethylamino)pentanoic acid, and β- alanine, each independently as an L or D isomer.
10. The conjugate of claim 9, wherein each XX is the residue of an independently
selected glycine or alanine.
11. The conjugate of any one of claims 1 to 10, wherein P is a peptide cleavable by a protease.
12. The conjugate of claim 11, wherein P is a peptide cleavable by a protease expressed in tumor tissue.
13. The conjugate of claim 11, wherein P is a peptide cleavable by a lysosomal protease.
14. The conjugate of any one of claims 1 to 6 and 8 to 13, wherein P is selected from the group consisting of: Val-Cit, Val-Lys, Phe-Lys, Lys-Lys, Ala-Lys, Phe-Cit, Leu-Cit, Lle-Cit, Trp, Cit, Phe-Ala, Phe-N9-tosyl-Arg, Phe-N9-nitro-Arg, Phe-Phe-Lys, D-Phe- Phe-Lys, Gly-Phe-Lys, Leu- Ala- Leu, He- Ala- Leu, Val-Ala-Val, Ala-Leu- Ala- Leu, β- Ala-Leu- Ala-Leu, Gly-Phe-Leu-Gly, Val-Arg, Arg-Val, Arg-Arg, Val-D-Cit, Val-D- Lys, Val-D-Arg, D- Val-Cit, D- Val-Lys, D- Val-Arg, D-Val-D-Cit, D-Val-D-Lys, D- Val-D-Arg, D-Arg-D-Arg, Ala-Ala, Ala-D-Ala, D-Ala-Ala, and D-Ala-D-Ala, Gly- Gly-Gly, Ala- Ala- Ala, D-Ala-Ala-Ala, Ala-D-Ala-Ala, Ala-Ala-D-Ala, Ala-Val-Cit, and Ala-Val-Ala.
15. The conjugate of any one of claims 1 to 14, wherein P is Gly-Gly-Gly, Ala-Ala-Ala, D-Ala- Ala-Ala, Ala-D-Ala-Ala, or Ala-Val-Ala. The conjugate of any one of claims 1 to 15, wherein
Figure imgf000094_0001
N-NRe--j s2 s1 j-N-NRe— ^ s2 s1 ^=N-NRe-phenylene— J s2 N-NRe-phenylene— 1 s2 s1 =N-0— I s2 s1 -N-0— 1 s2
Figure imgf000094_0002
Figure imgf000095_0001
94
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000097_0002
Figure imgf000097_0003
s1 i=N-NRe— I s2
18. The conjugate of claim 16, wherein JCB' and JCBI' is * * , s1 |-N-NRe— s2 s1 =N-0—
* ¾ , ¾ \ S2 s 1
? , or f ?— N~ O— I s2
? .
19. The conjugate of any one of claims 1 to 16, wherein Ra, Rb, Rc, and Re are each H.
20. The conjugate of any one of claims 1 to 19, wherein one of X\ and Z2 is absent and the other is -CH2-0- or -0-CH2-.
21. The conjugate of any one of claims 1 to 19, wherein one of X\ and Z2 is absent and the other is -(CH2CH20)p- or -(OCH2CH2)p-.
22. The conjugate of claim 21, wherein p or ' is an integer from 2 to 20.
23. The conjugate of claim 22, wherein p or p' is an integer from 2 to 8.
24. The conjugate of any one of claims 1 to 19, wherein Z2 is absent, and X\
is -S02NR9-, -NR9S02-, C(=0)-NR9- or -NR9-C(=0)-.
25. The conjugate of any one of claims 1 to 19, wherein Z2 is -0-CH2- and Z\ is -CH2- C(=0)-NR9-.
26. The conjugate of claim 25, wherein R9 is H.
27. The conjate of any one of claims 1 to 26, wherein R3, R4, R5 an R6 are all H; m and n are each independently an integer from 1 to 5.
28. The conjugate of claim 1, wherein the conjugate is represented by any one of the following formulae, or a pharmaceutically acceptable salt thereof:
Figure imgf000098_0001
wherein DM is a drug moiety represented by the following formula:
Figure imgf000098_0002
The conjugate of any one of claims 1 to 28, wherein the cell-binding agent is an antibody, a single chain antibody, an antibody fragment that specifically binds to the target cell, a monoclonal antibody, a single chain monoclonal antibody, a monoclonal antibody fragment that specifically binds to a target cell, a chimeric antibody, a chimeric antibody fragment that specifically binds to the target cell, a domain antibody, a domain antibody fragment that specifically binds to the target cell, a lymphokine, a hormone, a vitamin, a growth factor, a colony stimulating factor, or a nutrient-transport molecule.
30. The conjugate of claim 29, wherein the cell-binding agent is a monoclonal antibody, a single chain monoclonal antibody, or a monoclonal antibody fragment that specifically binds to a target cell.
31. The conjugate of claim 29 or 30, wherein the antibody is a resurfaced antibody, a resurfaced single chain antibody, or a resurfaced antibody fragment. 32. The conjugate of any one of claims 29 to 31, wherein the antibody is a humanized antibody, a humanized single chain antibody, or a humanized antibody fragment. 33. The conjugate of any one of claims 1 to 29, wherein the cell-binding agent is a
minibody, a diabody, a tribody, a tetrabody, a nanobody, a probody, a domain body, a unibody, a bispecific antibody, an ankyrin repeat protein or a DARPin, a Centyrin, or an Avibody.
34. A cytotoxic com ound represented by the following formula, or a salt thereof:
Figure imgf000099_0001
Figure imgf000099_0002
wherein:
DM is a drug moiety represented by the following formula:
Figure imgf000100_0001
R, R' and R", for each occurrence, are independently H or an optionally substituted alkyl;
one of Ei and E2 is -C(=0)-, and the other is -NR9-; or one of Έ\ and E2 is - C(=0)- or -NR9-, and the other is absent;
P is [XX] 1-10, in which each XX is a residue of an independently selected amino acid, or P is -(NRm-CH2CH2)s-;
s is an integer between 1 and 5;
Rm is H, or alkyl optionally substituted with a charged substituent or an ionizable group;
Z\ and Z2 are each independently absent, -S02NR9-, -NR9S02-, -C(=0)-NR9-, -NR9-C(=0)-, -C(=0)-0-, -0-C(=0)-, -CH2-0-, -0-CH2-, -(CH2CH20)P- or - (OCH2CH2)p-, -NR9-C(=0)-CH2-, or -CH2-C(=0)-NR9- wherein p and p' are independently an integer from 1 to 1000;
' -CRBRC-C(=0)-, X' -CRBRC-C(=0)-NRE- A-C(=0)-, RA-
Figure imgf000100_0002
JCBI is Ra-C(=0)-, Ra-C(=0)-Ar-, NH2-NRe-, NH2-NRe-Ar-, NH2-NRe-C(=0)-
Figure imgf000101_0001
Ar is an optionally substituted arylene or an optionally substituted heteroarylene;
' is a halogen;
Figure imgf000101_0002
an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne,
Figure imgf000101_0003
is an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene;
R2oi, R2o2 and R203 each are independently H or an optionally substituted alkyl;
Z3 is pyridyl or
Figure imgf000101_0004
? wherein R2o4 and R2os are each independently optionally substituted alkyl;
R3oi is H or optionally substituted alkyl;
Ra, Rb, Rc, Re, Ri to R6, and R9, for each occurrence, are independently H or an optionally substituted alkyl; and
m and n, for each occurrence, are independently an integer between 0 and 10.
35. The cytotoxic compound of claim 34, wherein the cytotoxic compound is represented by the following formula, or a salt thereof: ^4 R5 R6 o (ΠΓ ),
Figure imgf000102_0001
wherein:
DM is a drug moiet represented by the following formula:
Figure imgf000102_0002
R, R' and R", for each occurrence, are independently H or an optionally substituted alkyl;
one of Ei and E2 is -C(=0)-, and the other is -NR9-;
P is [XX] 1-10, in which each XX is a residue of an independently selected amino acid, or P is -(NRm-CH2CH2)s-;
s is an integer between 1 and 5;
Rm is H, or alkyl optionally substituted with a charged substituent or an ionizable group;
JCB is maleimide, X'-CRbRc-C(= -, X'-CRbRc-C(=0)-NRe- a-C(=0)-, Ra-
C(=0)-Ar-, NH2-NR6-, NH2-NRe-Ar-,
Figure imgf000102_0003
Figure imgf000103_0001
JCBI is Ra-C(=0)-, Ra-C(=0)-Ar-, NH2-NR6-, NH2-NRe-Ar-, NH2-NRe-C(=0)-
Figure imgf000103_0002
Ar is an optionally substituted arylene or an optionally substituted
heteroarylene;
' is a halogen;
Figure imgf000103_0003
an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne,
Figure imgf000103_0004
is an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene;
R2oi, R202 and R2Q3 each are independently H or an optionally substituted alkyl; R2o4o^ ll
Z3 is pyridyl or R205O o ? wherein R2o4 and R205 are each independently optionally substituted alkyl;
R3oi is H or optionally substituted alkyl;
Ra, Rb, Rc, Re, Ri to R6, and R9, for each occurrence, are independently H or an optionally substituted alkyl; and
m and n, for each occurrence, are independently an integer between 0 and 10.
36. The cytotoxic compound of claim 34 or 35, wherein the ionizable group is -S03H, - Z'-S03H, -OP03H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, -C02H, -Z'C02H, -NRnRi2, or -Z'-NRuRi2, and the charged group is -N+R13R14R15X", or -Z'-N+R13R14R15X~; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R12, for each occurrence, are independently H or an optionally substituted alkyl; R1 to R15 are each independently an optionally substituted alkyl; and X" is an anion.
37. The cytotoxic compound of claim 34 or 36, wherein Rm is H, or the charged
substituent is -CH2CH2S03H or a pharmaceutically acceptable salt thereof.
38. The cytotoxic compound of any one of claims 34 to 36, wherein P is [XX]2_4.
39. The cytotoxic compound of claim 38, wherein P is [XX]3.
40. The cytotoxic compound of any one of claims 34 to 39, wherein each XX is the
residue of an independently selected amino acid selected from: a naturally occurring amino acid, a synthetic amino acid, an amino acid analog, or an amino acid mimetic that functions in a manner similar to the naturally occurring amino acids.
41. The cytotoxic compound of claim 40, wherein each XX is the residue of an
independently selected amino acid selected from the group consisting of: Histidine, Alanine, Isoleucine, Arginine, Leucine, Asparagine, Lysine, Aspartic acid,
Methionine, Cysteine, Phenylalanine, Glutamic acid, Threonine, Glutamine,
Tryptophan, Glycine, Valine, Proline, Serine, Tyrosine, N-methyl-Histidine, N- methyl- Alanine, N-methyl-Isoleucine, N-methyl-Arginine, N-methyl-Leucine, N- methyl-Asparagine, N-methyl-Lysine, N-methyl-Aspartic acid, N-methyl-Methionine, N-methyl-Cysteine, N-methyl-Phenylalanine, N-methyl-Glutamic acid, N-methyl- Threonine, N-methyl-Glutamine, N-methyl-Tryptophan, N-methyl-Glycine, N- methyl- Valine, N-methyl-Proline, N-methyl-Serine, N-methyl-Tyrosine,
hydroxyproline, γ-carboxyglutamate, selinocysteine, O-phosphoserine, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, citrulline, Ornithine, cysteine sulfonic acid, cysteine sulfinic acid, 3-aminoalanine, 3- dimethylaminoalanine, 2-amino-4-(dimethylamino)butanoic acid, 2,4- diaminobutanoic acid, 2-amino-6-(dimethylamino)hexanoic acid, 2-amino-5- (dimethylamino)pentanoic acid, and β-alanine, each independently as an L or D isomer.
42. The cytotoxic compound of claim 41, wherein each XX is the residue of an
independently selected glycine or alanine.
43. The cytotoxic compound of any one of claims 34 to 42, wherein P is a peptide
cleavable by a protease.
44. The cytotoxic compound of claim 43, wherein P is a peptide cleavable by a protease expressed in tumor tissue.
45. The cytotoxic compound of claim 43, wherein P is a peptide cleavable by a lysosomal protease.
46. The cytotoxic compound of any one of claims 34 to 38 and 40 to 45, wherein P is selected from the group consisting of: Val-Cit, Val-Lys, Phe-Lys, Lys-Lys, Ala-Lys, Phe-Cit, Leu-Cit, Lle-Cit, Trp, Cit, Phe-Ala, Phe-N9-tosyl-Arg, Phe-N9-nitro-Arg, Phe-Phe-Lys, D-Phe-Phe-Lys, Gly-Phe-Lys, Leu-Ala-Leu, Ile-Ala-Leu, Val-Ala-Val, Ala-Leu- Ala-Leu, β-Ala-Leu- Ala-Leu, Gly-Phe-Leu-Gly, Val-Arg, Arg-Val, Arg- Arg, Val-D-Cit, Val-D-Lys, Val-D-Arg, D-Val-Cit, D-Val-Lys, D- Val-Arg, D-Val-D- Cit, D- Val-D-Lys, D- Val-D-Arg, D-Arg-D-Arg, Ala-Ala, Ala-D-Ala, D-Ala-Ala, and D-Ala-D-Ala, Gly-Gly-Gly, Ala- Ala- Ala, D-Ala-Ala-Ala, Ala-D-Ala- Ala, Ala- Ala- D-Ala, Ala- Val-Cit, and Ala-Val-Ala.
47. The cytotoxic compound of any one of claims 34 to 46, wherein P is Gly-Gly-Gly, Ala- Ala- Ala, D-Ala-Ala-Ala, Ala-D-Ala- Ala, or Ala-Val-Ala. The cytotoxic compound of any one of claims 34 to 47, wherein
JCB is maleimide, X'-CRbRc-C(=0)-, X'-CRbRc-C(=0)-NRe-, Ra-C(=0)-, R: =0)-phenylene-, NH2-NR6-, NH2-NRe-phenylene-, ΝΗ2-0-, -N3, -C≡CH,
Figure imgf000106_0001
49. The cytotoxic compound of claim 48, wherein JCB is maleimide.
50. The cytotoxic compound of claim 48, wherein JCB and JCB I is NH2-NRe- or ΝΗ2-0-,
51. The cytotoxic compound of any one of claims 34 to 48, wherein Ra, Rb, Rc, and Re are each H.
52. The cytotoxic compound of any one of claims 34 to 51, wherein one of X\ and Z2 is absent and the other is -CH2-0- or -0-CH2-.
53. The cytotoxic compound of any one of claims 34 to 51, wherein one of X\ and Z2 is absent and the other is -(CH2CH20)p- or -(OCH2CH2)p<-.
54. The cytotoxic compound of claim 53, wherein p or p' is an integer from 2 to 20.
55. The cytotoxic compound of claim 54, wherein p or p' is an integer from 2 to 8.
56. The cytotoxic compound of any one of claims 34 to 51, wherein Z2 is absent, and X\ is -S02NR9-, -NR9S02-, -C(=0)-NR9- or -NR9-C(=0)-.
57. The cytotoxic compound of any one of claims 34 to 51, wherein Z2 is -0-CH2- and X\ is -CH2-C(=0)-NR9-.
58. The cytotoxic compound of claim 57, wherein R9 is H.
59. The cytotoxic compound of any one of claims 34 to 58, wherein R3, R4, R5 an R6 are all H; m and n are each independently an integer from 1 to 5.
60. The cytotoxic compound of claim 34, wherein the cytotoxic compound is represented by any one of the following formulae, or a salt thereof:
Figure imgf000107_0001
wherein DM is a drug moiety represented by the following formula:
Figure imgf000108_0001
A linker compound represented by the following formula, or a salt thereof:
R3 R4 R5 Re O (IV),
Figure imgf000108_0002
one of Ei and E2 is -C(=0)-, and the other is -NR9-;or one of Ei and E2 is - C(=0)- or -NR9-, and the other is absent;
P is [ΧΧ] 1-10, in which each XX is a residue of an independently selected amino acid, or P is -(NRm-CH2CH2)s-;
s is an integer between 1 and 5;
Rm is H, or alkyl optionally substituted with a charged substituent or an ionizable group;
Zi and Z2 are each independently absent, -S02NR9-, -NR9S02-, -C(=0)-NR9 -NR9-C(=0)-, -C(=0)-0-, -0-C(=0)-, -CH2-0-, -0-CH2-, -(CH2CH20)p- or
(OCH2CH2)p-, -NR9-C(=0)-CH2-, or -CH2-C(=0)-NR9- wherein p and p' are independently an integer from 1 to 1000;
JCB is maleimide, X'-CRbRc-C(= -, X'-CRbRc-C(=0)-NRe- a-C(=0)-, Ra
C(=0)-Ar-, NH2-NR6-, NH2-NRe-Ar-,
Figure imgf000108_0003
Figure imgf000109_0001
JCBI is Ra-C(=0)-, Ra-C(=0)-Ar-, NH2-NR6-, NH2-NRe-Ar-, NH2-NRe-C(=0)-
Figure imgf000109_0002
Ar is an optionally substituted arylene or an optionally substituted
heteroarylene;
' is a halogen;
Figure imgf000109_0003
is an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne,
Figure imgf000109_0004
is an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene;
R2oi, R2o2 and R2o3 each are independently H or an optionally substituted alkyl; Z3 is pyridyl or
Figure imgf000110_0001
, wherein R2o4 and R 2o5 are each
independently optionally substituted alkyl;
R301 is H or optionally substituted alkyl;
Ra, Rb, Rc, Re, Ri to R6, and R9, are each independently H or an optionally substituted alkyl;
m and n, for each occurrence, are independently an integer between 0 and 10; JD is -OH or halogen; or -C(=0)-JD is a reactive ester.
The linker compound of claim 61, wherein the linker compound is represented by the following formula, or a salt thereof:
Figure imgf000110_0002
one of Ei and E2 is -C(=0)-, and the other is -NR9-;
P is [XXJ i-io, in which each XX is a residue of an independently selected amino acid, or P is -(NRm-CH2CH2)s-;
s is an integer between 1 and 5;
Rm is H, or alkyl optionally substituted with a charged substituent or an ionizable group;
JCB is maleimide, X'-CRbRc-C(= -, X'-CRbRc-C(=0)-NRe- a-C(=0)-, Ra
Figure imgf000110_0003
Figure imgf000111_0001
JCBI is Ra-C(=0)-, Ra-C(=0)-Ar-, NH2-NR6-, NH2-NRe-Ar-, NH2-NRe-C(=0)-
Figure imgf000111_0002
Ar is an optionally substituted arylene or an optionally substituted
heteroarylene;
' is a halogen;
Figure imgf000111_0003
an optionally substituted cycloalkyne or an optionally substituted heterocycloalkyne,
Figure imgf000111_0004
is an optionally substituted strained cycloalkene or an optionally substituted strained heterocycloalkene;
R2oi, R2o2 and R2o3 each are independently H or an optionally substituted alkyl;
0
R2o4o^ ll
Z is pyridyl or 205O o ? wherein R204 and R205 are each
independently optionally substituted alkyl;
R3oi is H or optionally substituted alkyl; Ra, Rb, Rc, Re, Ri to R6, and R9, are each independently H or an optionally substituted alkyl;
m and n, for each occurrence, are independently an integer between 0 and 10; JD is -OH or halogen; or -C(=0)-JD is a reactive ester.
63. The linker compound of claim 61 or 62, wherein the ionizable group is -SO3H, -Z'- SO3H, -OPO3H2, -Z'-OP03H2, -P03H2, -Z'-P03H2, -C02H, -Z'C02H, -NRnRi2, or - Z'-NRuRi2, and the charged group is -N+R13R14R15X~, or -Z'-N+R13R14R15X~; Z' is an optionally substituted alkylene, an optionally substituted cycloalkylene or an optionally substituted phenylene; Rn and R12, for each occurrence, are independently H or an optionally substituted alkyl; R13 to R15 are each independently an optionally substituted alkyl; and X" is an anion.
64. The linker compound of any one of claims 61 to 63, wherein Rm is H, or the charged substituent is -CH2CH2S03H or a pharmaceutically acceptable salt thereof.
65. The linker compound of any one of claims 61 to 64, wherein P is [XX]2^.
66. The linker compound of claim 65, wherein P is [XX] 3.
67. The linker compound of any one of claims 61 to 66, wherein each XX is the residue of an independently selected amino acid selected from: a naturally occurring amino acid, a synthetic amino acid, an amino acid analog, or an amino acid mimetic that functions in a manner similar to the naturally occurring amino acids.
68. The linker compound of claim 67, wherein each XX is the residue of an
independently selected amino acid selected from the group consisting of: Histidine, Alanine, Isoleucine, Arginine, Leucine, Asparagine, Lysine, Aspartic acid,
Methionine, Cysteine, Phenylalanine, Glutamic acid, Threonine, Glutamine,
Tryptophan, Glycine, Valine, Proline, Serine, Tyrosine, N-methyl-Histidine, N- methyl- Alanine, N-methyl-Isoleucine, N-methyl-Arginine, N-methyl-Leucine, N- methyl-Asparagine, N-methyl-Lysine, N-methyl-Aspartic acid, N-methyl-Methionine, N-methyl-Cysteine, N-methyl-Phenylalanine, N-methyl-Glutamic acid, N-methyl- Threonine, N-methyl-Glutamine, N-methyl-Tryptophan, N-methyl-Glycine, N- methyl- Valine, N-methyl-Proline, N-methyl-Serine, N-methyl-Tyrosine,
hydroxyproline, γ-carboxyglutamate, selinocysteine, O-phosphoserine, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, citrulline, Ornithine, cysteine sulfonic acid, cysteine sulfinic acid, 3-aminoalanine, 3- dimethylaminoalanine, 2-amino-4-(dimethylamino)butanoic acid, 2,4- diaminobutanoic acid, 2-amino-6-(dimethylamino)hexanoic acid, 2-amino-5- (dimethylamino)pentanoic acid, and β-alanine, each independently as an L or D isomer.
69. The linker compound of claim 68, wherein each XX is the residue of an
independently selected glycine or alanine.
70. The linker compound of any one of claims 61 to 69, wherein P is a peptide cleavable by a protease.
71. The linker compound of claim 70, wherein P is a peptide cleavable by a protease expressed in tumor tissue.
72. The linker compound of claim 70, wherein P is a peptide cleavable by a lysosomal protease.
73. The linker compound of any one of claims 71 to 65 and 67 to 72, wherein P is
selected from the group consisting of: Val-Cit, Val-Lys, Phe-Lys, Lys-Lys, Ala-Lys, Phe-Cit, Leu-Cit, Lle-Cit, Trp, Cit, Phe-Ala, Phe-N9-tosyl-Arg, Phe-N9-nitro-Arg, Phe-Phe-Lys, D-Phe-Phe-Lys, Gly-Phe-Lys, Leu-Ala-Leu, Ile-Ala-Leu, Val-Ala-Val, Ala-Leu- Ala-Leu, β-Ala-Leu- Ala-Leu, Gly-Phe-Leu-Gly, Val-Arg, Arg-Val, Arg- Arg, Val-D-Cit, Val-D-Lys, Val-D-Arg, D-Val-Cit, D-Val-Lys, D- Val-Arg, D-Val-D- Cit, D- Val-D-Lys, D- Val-D-Arg, D-Arg-D-Arg, Ala-Ala, Ala-D-Ala, D-Ala-Ala, and D-Ala-D-Ala., Gly-Gly-Gly, Ala-Ala-Ala, d-Ala-Ala-Ala, Ala-d-Ala-Ala, Ala-Ala-d- Ala, Ala-Val-Cit, and Ala-Val-Ala.
74. The linker compound of any one of claims 61 to 73, wherein P is Gly-Gly-Gly, Ala- Ala- Ala, d- Ala- Ala- Ala, Ala-d-Ala-Ala, or Ala-Val-Ala.
75. The linker compound of claims 61 to 74, wherein JCB is maleimide, X'-CRbRc- C(=0)-, X'-CRbRc-C(=0)-NRe-, Ra-C(=0)-, a-C =0)-phenylene-, NH2-NRe-,
NH2-NRe-phenylene-, ΝΗ2-0-, -N3, -C≡CH,
Figure imgf000113_0001
Figure imgf000114_0001
a-C(=0)-, Ra-C(=0)-phenylene-, NH2-NRE-, NH2-NRe-phenylene-,
Figure imgf000114_0002
76. The linker compound of claim 75, wherein JCB is maleimide.
77. The linker compound of claim 75, wherein JCB and JCBI is NH2-NRE- or
NH2-0-.
78. The linker compound of any one of claims 61 to 75, wherein RA, RB, RC, and RE are each H.
79. The linker compound of any one of claims 61 to 78, wherein one of X\ and Z2 is absent and the other is -CH2-0- or -0-CH2-.
80. The linker compound of any one of claims 61 to 78, wherein one of X\ and Z2 is absent and the other is -(CH2CH20)p- or -(OCH2CH2)p<-.
81. The linker compound of claim 80, wherein p or p' is an integer from 2 to 20.
82. The linker compound of claim 81, wherein p or p' is an integer from 2 to 8.
83. The linker compound of any one of claims 61 to 78, wherein Z2 is absent, and Z\ is -S02NR9-, -NR9S02-, C(=0)-NR9- or -NR9-C(=0)-.
84. The linker compound of any one of claims 61 to 78, wherein Z2 is -0-CH2- and Z\ is -CH2-C(=0)-NR9-.
85. The linker compound of claim 84, wherein R9 is H.
86. The linker compound of any one of claims 61 to 85, wherein wherein R3, R4, R5 an R6 are all H; m and n are each independently an integer from 1 to 5.
87. The linker compound of claim 61, represented by any one of the following formulae, or a salt thereof:
Figure imgf000115_0001
88. The linker compound of any one of claims 61 to 87, wherein -C(=0)-JD is N- hydroxysuccinimide ester.
89. A pharmaceutical composition comprising the conjugate of any one of claims 1 to 33 or the compound of any one of claims 34 to 59; and a pharmaceutically acceptable carrier.
90. A method of inhibiting abnormal cell growth or treating a proliferative disorder, an autoimmune disorder, a destructive bone disorder, a graft versus host disease, a transplant rejection, an immune deficiency, an inflammatory disease, an infectious disease, a viral disease, a fibrotic disease, a neurodegenerative disorder, pancreatitis, or a kidney disease in a mammal comprising administering to said mammal a therapeutically effective amount of the conjugate of any one of claims 1 to 33, the compound of any one of claims 34 to 59, or the pharmaceutical composition of claim 89; and, optionally, a second therapeutic agent.
91. The method of claim 90, wherein said second therapeutic agent is administered to said mammal sequentially or consecutively.
92. The method of claim 90 or 91, wherein the method is for treating a condition selected from the group consisting of cancer, rheumatoid arthritis, multiple sclerosis, a graft versus host disease, a transplant rejection, lupus, myositis, an infectious disease, and an immune deficiency.
93. The method of claim 92, wherein the condition is cancer.
94. The method of claim 93, wherein the cancer is selected from the group consisting of breast cancer, colon cancer, brain cancer, prostate cancer, kidney cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, colorectal cancer, gastric cancer, squamous cancer, small-cell lung cancer, nonsmall-cell lung cancer, testicular cancer, Merkel cell carcinoma, glioblastoma, neuroblastoma, a cancer of a lymphatic organ, and a hematological malignancy.
PCT/US2014/019434 2013-02-28 2014-02-28 Conjugates comprising cell-binding agents and cytotoxic agents WO2014134457A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361770796P 2013-02-28 2013-02-28
US61/770,796 2013-02-28

Publications (2)

Publication Number Publication Date
WO2014134457A2 true WO2014134457A2 (en) 2014-09-04
WO2014134457A3 WO2014134457A3 (en) 2014-12-04

Family

ID=50288321

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/019434 WO2014134457A2 (en) 2013-02-28 2014-02-28 Conjugates comprising cell-binding agents and cytotoxic agents

Country Status (1)

Country Link
WO (1) WO2014134457A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3077402A4 (en) * 2013-12-02 2017-06-28 Hong Kong Baptist University Anticancer maytansinoids with two fused macrocyclic rings
WO2018159582A1 (en) 2017-02-28 2018-09-07 学校法人近畿大学 Method for treating egfr-tki-resistant non-small cell lung cancer by administration of anti-her3 antibody-drug conjugate
US10287256B2 (en) 2016-11-23 2019-05-14 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
US10449258B2 (en) 2015-06-09 2019-10-22 Xdcexplorer (Shanghai) Co., Ltd. Antibody drug conjugate, intermediate, preparation method, pharmaceutical composition and uses thereof
WO2020059772A1 (en) 2018-09-20 2020-03-26 第一三共株式会社 Treatment of her3 mutant cancer by administration of anti-her3 antibody-drug conjugate
US10898579B2 (en) 2015-06-29 2021-01-26 Immunogen, Inc. Conjugates of cysteine engineered antibodies
CN112826940A (en) * 2014-11-11 2021-05-25 杭州多禧生物科技有限公司 Conjugates of cytotoxic molecules and cell-binding receptor molecules
US11202837B2 (en) 2014-09-25 2021-12-21 Antikor Biopharma Limited Biological materials and uses thereof
WO2022014698A1 (en) 2020-07-17 2022-01-20 第一三共株式会社 Method for producing antibody-drug conjugate

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009134977A1 (en) * 2008-04-30 2009-11-05 Immunogen, Inc. Cross-linkers and their uses
WO2012061590A1 (en) * 2010-11-03 2012-05-10 Immunogen, Inc. Cytotoxic agents comprising new ansamitocin derivatives
WO2012135517A2 (en) * 2011-03-29 2012-10-04 Immunogen, Inc. Preparation of maytansinoid antibody conjugates by a one-step process
WO2012138749A1 (en) * 2011-04-04 2012-10-11 Immunogen, Inc. Methods for decreasing ocular toxicity of antibody drug conjugates
WO2012145112A2 (en) * 2011-04-18 2012-10-26 Immunogen, Inc. Novel maytansinoid derivatives with sulfoxide linker

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009134977A1 (en) * 2008-04-30 2009-11-05 Immunogen, Inc. Cross-linkers and their uses
WO2012061590A1 (en) * 2010-11-03 2012-05-10 Immunogen, Inc. Cytotoxic agents comprising new ansamitocin derivatives
WO2012135517A2 (en) * 2011-03-29 2012-10-04 Immunogen, Inc. Preparation of maytansinoid antibody conjugates by a one-step process
WO2012138749A1 (en) * 2011-04-04 2012-10-11 Immunogen, Inc. Methods for decreasing ocular toxicity of antibody drug conjugates
WO2012145112A2 (en) * 2011-04-18 2012-10-26 Immunogen, Inc. Novel maytansinoid derivatives with sulfoxide linker

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LAU A ET AL: "CONJUGATION OF DOXORUBICIN TO MONOCLONAL ANTI-CARCINOEMBRYONIC ANTIGEN ANTIBODY VIA NOVEL THIOL-DIRECTED CROSS-LINKING REAGENTS", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 3, no. 10, 1 January 1995 (1995-01-01), pages 1299-1304, XP001193824, ISSN: 0968-0896, DOI: 10.1016/0968-0896(95)00125-Z *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3077402A4 (en) * 2013-12-02 2017-06-28 Hong Kong Baptist University Anticancer maytansinoids with two fused macrocyclic rings
US11202837B2 (en) 2014-09-25 2021-12-21 Antikor Biopharma Limited Biological materials and uses thereof
CN112826940A (en) * 2014-11-11 2021-05-25 杭州多禧生物科技有限公司 Conjugates of cytotoxic molecules and cell-binding receptor molecules
US10449258B2 (en) 2015-06-09 2019-10-22 Xdcexplorer (Shanghai) Co., Ltd. Antibody drug conjugate, intermediate, preparation method, pharmaceutical composition and uses thereof
US10898579B2 (en) 2015-06-29 2021-01-26 Immunogen, Inc. Conjugates of cysteine engineered antibodies
US10287256B2 (en) 2016-11-23 2019-05-14 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
US10494348B2 (en) 2016-11-23 2019-12-03 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
US10822313B2 (en) 2016-11-23 2020-11-03 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
US11572349B2 (en) 2016-11-23 2023-02-07 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
WO2018159582A1 (en) 2017-02-28 2018-09-07 学校法人近畿大学 Method for treating egfr-tki-resistant non-small cell lung cancer by administration of anti-her3 antibody-drug conjugate
WO2020059772A1 (en) 2018-09-20 2020-03-26 第一三共株式会社 Treatment of her3 mutant cancer by administration of anti-her3 antibody-drug conjugate
WO2022014698A1 (en) 2020-07-17 2022-01-20 第一三共株式会社 Method for producing antibody-drug conjugate

Also Published As

Publication number Publication date
WO2014134457A3 (en) 2014-12-04

Similar Documents

Publication Publication Date Title
USRE49918E1 (en) Cytotoxic benzodiazepine derivatives
US9999680B2 (en) Conjugates comprising cell-binding agents and maytansinoids as cytotoxic agents
US9624240B2 (en) Maytansinoid derivatives with sulfoxide linker
WO2014194030A2 (en) Conjugates comprising cell-binding agents and cytotoxic agents
AU2012272930B2 (en) Novel maytansinoid derivatives with peptide linker and conjugates thereof
WO2014134457A2 (en) Conjugates comprising cell-binding agents and cytotoxic agents
US20150315193A1 (en) Cytotoxic benzodiazepine derivatives
EP2961435B1 (en) Conjugates comprising cell-binding agents and cytotoxic agents

Legal Events

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

Ref document number: 14710741

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 14710741

Country of ref document: EP

Kind code of ref document: A2