CA2627046C - Methods and compounds for preparing cc-1065 analogs - Google Patents

Methods and compounds for preparing cc-1065 analogs Download PDF

Info

Publication number
CA2627046C
CA2627046C CA2627046A CA2627046A CA2627046C CA 2627046 C CA2627046 C CA 2627046C CA 2627046 A CA2627046 A CA 2627046A CA 2627046 A CA2627046 A CA 2627046A CA 2627046 C CA2627046 C CA 2627046C
Authority
CA
Canada
Prior art keywords
compound
substituted
unsubstituted
alkyl
oxycarbonyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2627046A
Other languages
French (fr)
Other versions
CA2627046A1 (en
Inventor
Sanjeev Gangwar
Qian Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ER Squibb and Sons LLC
Original Assignee
ER Squibb and Sons LLC
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 ER Squibb and Sons LLC filed Critical ER Squibb and Sons LLC
Publication of CA2627046A1 publication Critical patent/CA2627046A1/en
Application granted granted Critical
Publication of CA2627046C publication Critical patent/CA2627046C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/58[b]- or [c]-condensed
    • C07D209/60Naphtho [b] pyrroles; Hydrogenated naphtho [b] pyrroles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

Disclosed is a method of making a CBI CC-1065 analog from a compound of formula (I) (see formula I) wherein R1 and R2 are each independently H, alkyl, -C(O)OR', -C(O)NR'R", or a protecting group, wherein R' and R" are independently selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, and unsubstituted heterocycloalkyl; R6 is H, substituted or unsubstituted lower alkyl, cyano, or alkoxy; and X is halogen.

Description

BACKGROUND
CC-1065 is known to be a potent cytotoxin. CC-1065 was first isolated from Streptomyces zelensis in 1981 by the Upjohn Company (Hanka et al., J.
Antibiot. 31: 1211 (1978); Martin et al., J. Antibiot, 33: 902 (1980); Martin et al., J.
Antibiot. 34: 1119 (1981)) and was found to have potent antitumor and antimicrobial activity both in vitro and in experimental animals (Li et al., Cancer Res. 42:999 (1982)). CC-1065 binds to double-stranded B-DNA within the minor groove (Swenson et aL, Cancer Res. 42: 2821 (1982)) with the sequence preference of 5'-d(A/GNTTA)-3' and 5'-d(AAAAA)-3' and allcylates the N3 position of the 3'-adenine by its CPI left-hand unit present in the molecule (Hurley et al., Science 226: 843 (1984)). Despite its potent and broad antitumor activity, CC-1065 cannot be used in humans because it causes delayed death in experimental animals.
Many analogs and derivatives of CC-1065 are known in the art. The research into the structure, synthesis and properties of many of the compounds has been reviewed. See, for example, Boger et al., Angew. Chem. Int. Ed. Engl. 35: 1438 (1996); and Boger et aL, Chem. Rev. 97: 787 (1997).
A group at Kyowa Hakko Kogya Co., Ltd. has prepared a number of CC-1065 derivatives. See, for example, U.S. Pat. No. 5,101, 038; 5,641,780; 5,187,186;
5,070,092;
5,703,080; 5,070,092; 5,641,780; 5,101,038; and 5,084,468; and published PCT
application, WO 96/10405 and published European application 0 537 575 Al.
The Upjohn Company (Pharmacia Upjohn) has also been active in preparing derivatives of CC-1065. See, for example, U.S. Patent No. 5,739,350;
4,978,757, 5,332, 837 and 4,912,227.
BRIEF SUMMARY
One embodiment is a method of making a compound (1) or a salt thereof X
/

SO

(I) where R1 and R2 are each independently H, alkyl, ¨C(0)OR', ¨C(0)NR'R", or a protecting group, wherein R' and R" are independently selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, and unsubstituted heterocycloalkyl; R6 is H, substituted or unsubstituted lower alkyl, cyano, or alkoxy; and X is halogen, preferably Cl or Br. In this method as broadly disclosed, protecting groups R1' and R2' are added to a compound (II) R6 leo NH2 (I1) to form a compound (III) R6 NR1.F1 OR2.
(III)
2 where R3 is H or alkyl, and RI, R2' and R6 are as defined above. A five membered ring is generated comprising the amine nitrogen compound (III). Also, in the invention as claimed, the protecting groups RI and R2' are however different protecting groups.
Preferably again, RI and R1 are the same and R2' and R2 are the same.
In practice, generating the five membered ring may comprise iodination of a carbon adjacent the amine substituent of compound (III) followed by alkylation using 1,3-dihalopropene.
Another embodiment of the invention relates to a method of making a compound (V) or a salt thereof, X
/
R6 N ¨ R1.
*0 R2' (V) wherein Rt and R2' are different protecting groups, R6 is H, a fully saturated straight or branched C1-C6 homoalkyl, cyano, or a fully saturated straight or branched Ci_Cs homoalkoxy; and X is Cl or Br, the method comprising:
(i) adding protecting groups RI and R2' to a compound (II), 2a (II) wherein R3 is a fully saturated straight or branched homoalkyl and R6 is as defined above, to form a compound (IV) R6 NR1.1-1 lee OR2.
(IV) wherein R1', R2' and R6 are as defined above;
(ii) iodinating the compound of formula (IV) obtained from step (i) with N-iodosuccinimide, (iii) alkylating the product obtained from the iodinating step (ii) with 1,3-dibromopropene or 1,3-dichloropropene, and (iv) performing a ring closure on the product obtained from the alkylating step (iii) with tributyltin hydride in the presence of 2,2'-azobisisobutyronitrile to generate a five membered ring comprising the amine nitrogen of compound (V).
Another embodiment of the invention relates to the method defined hereinabove, wherein R3 is methyl.
Another embodiment of the invention relates to the method defined hereinabove, wherein protecting groups RI and R2' are selected from: BOG; FMOC; 2-trimethylsilylethoxycarbonyl; allyloxycarbonyl; 4-methyl-1-piperazinecarbonyl;
1-methyl-1-(4-biphenypethoxycarbonyl; diphenyloxycarbonyl; benzyl; t-butyl;
tetrahydropyran, trimethylsilyl, t-butyldimethylsilyl; triisopropylsilyl; t-butyldiphenylsilyl;
2,2,2-trichloroethyl oxycarbonyl; diisopropylmethyl oxycarbonyl; vinyl oxycarbonyl; methoxy benzyl 2b oxycarbonyl; nitrobenzyl oxycarbonyl; cyclohexyl oxycarbonyl; cyclopentyl oxycarbonyl;
benzyloxycarbonyl; formyl; acetyl; trihaloacetyl; benzoyl; nitrophenylacetyl;

nitrobenzenesulfonyl; phthalimido; and dithiasuccinoyl.
Another embodiment of the invention relates to the method defined hereinabove, wherein RI is tert-butyloxycarbonyl.
Another embodiment of the invention relates to the method defined hereiabove, wherein R2' is -CH2Ph.
Another embodiment is a method of making a CBI CC-1065 analog, or a pharmaceutically acceptable salt thereof, having the following formula:
2c R70 Mir R4' Z Fe.

where X is halo;
X1 and Z are each independently selected from 0, S and NR8, where R8 is a member selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, and acyl;
R4, R4', R5 and R5' are members independently selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, unsubstituted heterocycloalkyl, halogen, NO2, NR9R1 , NC(0)R9, OC(0)NR9R1 , OC(0)0R9, C(0)R9, SR9, OR9, CR9=NR10, and 0(CH2)NRI1Rii', where R9 and R1 are independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycloalkyl, and substituted or unsubstituted peptidyl or where R9 and R1 together with the nitrogen atom to which they are attached are optionally joined to form a substituted or unsubstituted heterocycloalkyl ring system having from 4 to 6 members, optionally containing two or more heteroatorns, and R11 and R11' are each independently H or lower alkyl;
R6 is H, substituted or unsubstituted lower alkyl, cyano, or alkoxy; and R7 is a member selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted heteroalkyl, unsubstituted heteroalkyl, diphosphates, triphosphates, acyl, C(0)R12R13, C(0)0R12, C(0)NR12R13, P(0)(0R12)2, C(0)CHR12R13, SR12 and SiR12R13R14, where R12, R13, and R14 are members independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl and substituted or unsubstituted aryl, or where R12 and R13 together with the nitrogen or carbon atom to which they are attached are optionally joined to form a substituted or unsubstituted
3 heterocycloalkyl ring system having from 4 to 6 members, optionally containing two or more heteroatoms.
The method includes adding protecting groups R1' and R2' to a compound (II) Ra 00 NH2 (11) to form a compound (III) R6 NR1'11 OR2' (III) where R3 is H or alkyl. A five membered ring is generated comprising the amine nitrogen of compound (III). A binding unit is added to compound (III), the binding unit comprising R4' Ail" R4 1--61õ1 XI
R5' Yet another embodiment is a compound of formula (I), or a pharmaceutically acceptable salt thereof:
Br /

(1) where RI and R2 are each independently H, alkyl, -C(0)OR', -C(0)NR'R", or a protecting group, where R' and R" are independently selected from the group consisting of H,
4 substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, and unsubstituted heterocycloalkyl.
Preferably, adding the binding unit comprises deprotecting the amine substituent of compound (I) by removing the protecting groups R1', and reacting a compound comprising the binding unit with the deprotected amine.
Preferably also, adding the binding unit further comprises adding the binding unit to the amine substituent.
BRIEF DESCRIPTION OF THE DRAWINGS
Non-limiting and non-exhaustive embodiments of the present invention are described with reference to the following drawings. In the drawings, like reference numerals refer to like parts throughout the various figures unless otherwise specified.
For a better understanding of the present invention, reference will be made to the following Detailed Description, which is to be read in association with the accompanying drawings, wherein:
Fig. 1 is a synthetic scheme for one embodiment of a method of forming a CBI CC-1065 analog;
FIG. 2 is a synthetic scheme for another embodiment of a method of forming a CBI CC-1065 analog; and FIG. 3 is a synthetic scheme for a third embodiment of a method of forming a CBI C-1065 analog.
DETAILED DESCRIPTION
As used herein, "Boc" refers to t-butyloxycarbonyl.

"CPI" refers to cyclopropapyrroloindole.
"CBI" refers to cyclopropabenzindole.
"Cbz" is carbobenzoxy.
"DCM," refers to dichloromethane.
"DMF" is N,N-dimethylformamide.
"FMOC" refers to 9-fluorenylmethyloxycarbonyl.
"TEA" refers to triethylamine.
"THF" refers to tetrahydrofunan.
"EDC" refers to 1-(3-dimethylaminopropyI)-3-ethylcarbodiimide hydrochloride.
Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one ordinary skill in the art to 5a which this invention belongs. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry and nucleic acid chemistry and hybridization described below are those well known and commonly employed in the art. The techniques and procedures are generally performed according to conventional methods in the art and various general references. The nomenclature used herein and the laboratory procedures in analytical chemistry, and organic synthetic described below are those well known and commonly employed in the art.
Standard techniques, or modifications thereof, are used for chemical syntheses and chemical analyses.
The term "therapeutic agent" is intended to mean a compound that, when present in a therapeutically effective amount, produces a desired therapeutic effect on a mammal. For treating carcinomas, it is desirable that the therapeutic agent also be capable of entering the target cell.
The term "cytotoxin" is intended to mean a therapeutic agent having the desired effect of being cytotoxic to cancer cells. Cytotoxic means that the agent arrests the growth of, or kills, the cells.
The terms "prodrug" and "drug conjugate" are used herein interchangeably. Both refer to a compound that is relatively innocuous to cells while still in the conjugated form but which is selectively degraded to a pharmacologically active form by conditions, e.g., enzymes, located within or in the proximity of target cells.
The symbol '1=11-rx, , whether utilized as a bond or displayed perpendicular to a bond indicates the point at which the displayed moiety is attached to the remainder of the molecule, solid support, substituent, etc.
The term "alkyl," by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated (i.e. Ci-Co means one to ten carbons). Examples of saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. An unsaturated alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers. The term "alkyl," unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below, such as "heteroalkyl." Alkyl groups, which are limited to hydrocarbon groups are termed "homoalkyl".
The term "alkylene" by itself or as part of another substituent means a divalent radical derived from an alkane, as exemplified, but not limited, by ¨CH2CH2CH2CH2-, and further includes those groups described below as "heteroalkylene." Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. A "lower alkyl" or "lower alkylene" is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
The term "heteroalkyl," by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of a number of carbon atoms and at least one heteroatom selected from the group consisting of 0, N, Si and S, and wherein the nitrogen, carbon and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) 0, N, S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule. Examples include, but are not limited to, -C1-CH3, -CH2-CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, -CH2-S-CH2-CH3, -CH2-CH2,-S(0)-CH3, -CH2-CH2-S(0)2-CH3, -CH=CH-0-CH3, -Si(CH3)3, -CH2-CH=N-OCH3, and ¨
CH=CH-N(CH3)-CH3. Up to two heteroatoms may be consecutive, such as, for example, -CH2-NH-OCH3 and ¨CH2-0-Si(CH3)3. Similarly, the term "heteroalkylene" by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH2-CH2-S-C}{2-CH2- and ¨CH2-S-CH2-CH2-NH-CH2-=
For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). The terms "heteroalkyl" and "heteroalkylene" encompass poly(ethylene glycol) and its derivatives (see, for example, Shearwater Polymers Catalog, 2001). Still further, for alkylene and heteroalkylene substituents, no orientation of the substituent is implied by the direction in which the formula of the substituent is written. For example, the formula ¨
C(0)21V- represents both ¨C(0)2R'- and ¨R'C(0)2-.

The term "lower" in combination with the terms "alkyl" or "heteroallcyl"
refers to a moiety having from 1 to 6 carbon atoms.
The terms "alkoxy," "allcylamino," "alkylsulfonyl," and "alkylthio" (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, an SO2 group or a sulfur atom, respectively. The term "arylsulfonyl" refers to an aryl group attached to the remainder of the molecule via an SO2 group, and the term "sulfhydryl" refers to an SH
group.
In general, an "acyl" substituent is also selected from the group set forth above. As used herein, the term "acyl" substituent refers to groups attached to, and fulfilling the valence of a carbonyl carbon that is either directly or indirectly attached to the polycyclic nucleus of the compounds of the present invention.
The terms "cycloalkyl" and "heterocycloalkyl", by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of substituted or unsubstituted "alkyl" and substituted or unsubstituted "heteroalkyl", respectively.
Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include, but are not limited to, 1 ¨
(1,2,5,6-tetrahydropyridy1), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1 ¨piperazinyl, 2-piperazinyl, and the like. The heteroatoms and carbon atoms of the cyclic structures are optionally oxidized.
The terms "halo" or "halogen," by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom_ Additionally, terms such as "haloalkyl," are meant to include monohaloalkyl and polyhaloalkyl. For example, the term "halo(Ci-C4)alkyl" is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
The term "aryl" means, unless otherwise stated, a substituted or unsubstituted polyunsaturated, aromatic, hydrocarbon substituent which can be a single ring or multiple rings (preferably from 1 to 3 rings) which are fused together or linked covalently. The term "heteroaryl" refers to aryl groups (or rings) that contain from one to four Iheteroatoms selected from N, 0, and S, wherein the nitrogen, carbon and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized. A heteroaryl group can be attached to the remainder of the molecule through a heteroatom. Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-fury!, 3-fury!, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl, and 6-quinolyl. Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below. "Aryl" and "heteroaryl" also encompass ring systems in which one or more non-aromatic ring systems are fused, or otherwise bound, to an aryl or heteroaryl system.
For brevity, the term "aryl" when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
Thus, the term "arylalkyl" is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like).
Each of the above terms (e.g., "alkyl," "heteroalkyl," "aryl" and "heteroaryl") include both substituted and unsubstituted forms of the indicated radical.
Preferred substituents for each type of radical are provided below.
Substituents for the alkyl, and heteroalkyl radicals (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) are generally referred to as "alkyl substituents" and "heteroalkyl substituents," respectively, and they can be one or more of a variety of groups selected from, but not limited to: -OR', =0, =NR', =N-OR', -NR'R'', -SR', -halogen, -SiR'R"R", -0C(0)R', -C(0)R', -CO2R', -CONR'R", -0C(0)NR'R", -NR"C(0)R', -NR'-C(0)NR"R", -NR"C(0)2R', -NR-C(NR'R"R'")=NR'", -NR-C(NR'R")=NR'", -S(0)R', -S(0)2R', -S(0)2NR'R", -NRSO2R', -CN and -NO2 in a number ranging from zero to (2m'+1), where m' is the total number of carbon atoms in such radical. R', R", R" and R" each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, e.g., aryl substituted with 1-3 halogens, substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylallcyl groups. When a compound of the invention includes more than one R
group, for example, each of the R groups is independently selected as are each R', R", R"
and R'"' groups when more than one of these groups is present. When R' and R"
are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a
5-, 6-, or 7-membered ring. For example, -NR'R" is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term "alkyl" is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., -CF3 and ¨CH2CF3) and acyl (e.g., -C(0)CI-13, -C(0)CF 3, -C(0)CH2OCH3, and the like).
Similar to the substituents described for the alkyl radical, the aryl substituents and heteroaryl substituents are generally referred to as "aryl substituents" and "heteroaryl substituents," respectively and are varied and selected from, for example:
halogen, -OR', =0, =NR', =N-OR', -NR'R", -SR', -halogen, -SiR'R"R", -0C(0)R', -C(0)R', -CO2R', -CONR'R", -0C(0)NR'R", -NR"C(0)R', -NR'-C(0)NR"R", -NR"C(0)2R', -NR-C(NR'R")=NR'", -5(0)R', -S(0)2R', -S(0)2NR'R", -NRSO2R', -CN and ¨NO2, -R', -N3, -CH(Ph)2, fluoro(CI-C4)alkoxy, and fluoro(CI-C4)alkyl, in a number ranging from zero to the total number of open valences on the aromatic ring system; and where R', R", R"' and R"" are preferably independently selected from hydrogen, (C1-C8)alkyl and heteroalkyl, unsubstituted aryl and heteroaryl, (unsubstituted aryI)-(C1-C4)alkyl, and (unsubstituted aryl)oxy-(Ci-C4)alkyl. When a compound of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R', R", R" and R" groups when more than one of these groups is present.
Two of the aryl substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula ¨T-C(0)-(CRR')q-U-, wherein T
and U are independently ¨NR-, -0-, -CRR'- or a single bond, and q is an integer of from 0 to 3. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula ¨A-(CH2),-B-, wherein A and B are independently ¨CRR'-, -0-, -NR-, -S-, -S(0)-, -S(0)2-, -S(0)2NR'- or a single bond, and r is an integer of from 1 to 4. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -(CRR')s-X-(CR"R'")d-, where s and d are independently integers of from 0 to 3, and X is -0-, -NR'-, -S-, -S(0)-, -S(0)2-, or -S(0)2NR'-. The substituents R, R', R"
and R" are preferably independently selected from hydrogen or substituted or unsubstituted (C1-C6) alkyl.
As used herein, the term "diphosphate" includes but is not limited to an ester of phosphoric acid containing two phosphate groups. The term "triphosphate"
includes but is not limited to an ester of phosphoric acid containing three phosphate groups.
For example, particular drugs having a diphosphate or a triphosphate include:
CO2Me R1204=0 N
0,e IP
Di2,1 R11 \i Diphosphate CO2Me 9 oRi2 _ -F,)-oRi2 ,0 Triphosphate As used herein, the term "heteroatom" includes oxygen (0), nitrogen (N), sulfur (S) and silicon (Si).
The symbol "R" is a general abbreviation that represents a substituent group that is selected from substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted heterocyclyl groups.
Regarding the term "protecting group," those of skill in the art will understand how to protect a particular functional group from interfering with a chosen set of reaction conditions. For examples of useful protecting groups, See Greene et al., PROTECTIVE
GROUPS IN ORGANIC SYNTHESIS, John Wiley & Sons, New York, 1991. Examples of suitable protecting groups include, but are not limited to, BOC, FMOC, 2-trimethylsilylethoxycarbonyl, allyloxycarbonyl, 4-methyl-I -piperazinecarbonyl, 1-methyl-1-(4-biphenylypethoxycarbonyl, diphenyloxycarbonyl, benzyl, t-butyl, tetrahydropyran, trimethylsilyl, t-butyldimethylsilyl, triisopropylsilyl, t-butyldiphenylsilyl, 2,2,2-trichloroethyl oxycarbonyl, diisopropylmethyl oxycarbonyl, vinyl oxycarbonyl, methoxy benzyl oxycarbonyl, nitrobenyzl oxycarbonyl, cyclohexyl oxycarbonyl, cyclopentyl oxycarbonyl, benzyloxycarbonyl, formyl, acetyl, trihaloacetyl, benzoyl, nitrophenylacetyl, 2-nitrobenzensulfonyl, phthalimido, and dithiasuccinoyl.
The term "pharmaceutically acceptable carrier", as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent. Pharmaceutically acceptable carriers include pharmaceutically acceptable salts, where the term "pharmaceutically acceptable salts"
includes salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present invention contain relatively basic fiinctionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
The neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
In addition to salt forms, the present invention provides compounds, which are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention. Additionally, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms.
In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, diastereomers, geometric isomers and individual isomers are encompassed within the scope of the present invention.
The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (1251) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. These terms also encompass the term "antibody."
The term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, y-carboxyglutamate, and 0-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an ii carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R
group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
One amino acid that may be used in particular is citrulline, which is a precursor to arginine and is involved in the formation of urea in the liver. 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. The term "unnatural amino acid" is intended to represent the "D" stereochemical form of the twenty naturally occurring amino acids described above. It is further understood that the term unnatural amino acid includes homologues of the natural amino acids, and synthetically modified forms of the natural amino acids. The synthetically modified forms include, but are not limited to, amino acids having alkylene 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.
When attached to a linker or conjugate of the invention, the amino acid is in the form of an "amino acid side chain", where the carboxylic acid group of the amino acid has been replaced with a keto (C(0)) group. Thus, for example, an alanine side chain is -C(0)-CH(NH2)-CH3, and so forth.
Amino acids and peptides may be protected by blocking groups. A blocking group is an atom or a chemical moiety that protects the N-terminus of an amino acid or a peptide from undesired reactions and can be used during the synthesis of a drug-cleavable substrate conjugate. It should remain attached 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, hydrogen, D-amino acid, and carbobenzoxy (Cbz) chloride.
The term "antibody" as referred to herein includes whole antibodies and any antigen binding fragment (i.e., "antigen-binding portion") or single chains thereof. An "antibody" refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, Cm, Cr-2 and CH33 and may be of the mu, delta, gamma, alpha or epsilon isotype. Each light chain is comprised of a light chain variable region (VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL, which may be of the kappa or lambda isotype. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL
is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
The terms "antibody fragment" or "antigen-binding portion" of an antibody (or simply "antibody portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
Examples of binding fragments encompassed within the term "antibody fragment"
or "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and Cm domains; (ii) a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region;
(iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and Vu, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science N2:423-426;
and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
The terms "monoclonal antibody" as used herein refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
"Solid support," as used herein refers to a material that is substantially insoluble in a selected solvent system, or which can be readily separated (e.g., by precipitation) from a selected solvent system in which it is soluble. Solid supports useful in practicing the present invention can include groups that are activated or capable of activation to allow selected species to be bound to the solid support. A solid support can also be a substrate, for example, a chip, wafer or well, onto which an individual, or more than one compound, of the invention is bound.
The compounds of the invention are prepared as a single isomer (e.g., enantiorner, cis-trans, positional, diastereomer) or as a mixture of isomers. In a preferred embodiment, the compounds are prepared as substantially a single isomer. Methods of preparing substantially isomerically pure compounds are known in the art. For example, enantiomerically enriched mixtures and pure enantiomeric compounds can be prepared by using synthetic intermediates that are enantiomerically pure in combination with reactions that either leave the stereochemistry at a chiral center unchanged or result in its complete inversion. Alternatively, the final product or intermediates along the synthetic route can be resolved into a single stereoisomer. Techniques for inverting or leaving unchanged a particular stereocenter, and those for resolving mixtures of stereoisomers are well known in the art and it is well within the ability of one of skill in the art to choose and appropriate method for a particular situation. See, generally, Furniss et al.
(eds.),VoGEL's ENCYCLOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5TH ED., Longman Scientific and Technical Ltd., Essex, 1991, pp. 809-816; and Heller, Acc. Chem. Res. 23: 128 (1990).
Cytotoxic analogs of CC-1065 can be formed using a cyclopropabenzindole (CBI) moiety as an alkylating unit instead of the cyclopropapyrolloindole (CPI) moiety of CC-1065. As one example, CC-1065 CBI analogs include, but are not limited to compounds having the formula (or a pharmaceutically acceptable salt thereof):

Ix R70 VIV R4' N Rd C
X1; ________ Z 01 R5, where X is halo. Preferably, X is Cl or Br and, more preferably, X is Br.
X1 and Z are each independently selected from 0, S and NR8 where R8 is a member selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, and acyl.
R4, R4', R5 and R5' are members independently selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, unsubstituted heterocycloalkyl, halogen, NO2, NR9RI , NC(0)R9, OC(0)NR9R1 , OC(0)0R9, C(0)R9, SR9, OR9, CR9=NR10, and 0(CH2),INR11R11', where R9 and RI are independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycloalkyl, and substituted or unsubstituted peptidyl, or where R9 and R1 together with the nitrogen atom to which they are attached are optionally joined to form a substituted or unsubstituted heterocycloalkyl ring system having from 4 to 6 members, optionally containing two or more heteroatoms, and R" and R11' are each independently H or lower alkyl.
R6 is H, substituted or unsubstituted lower alkyl, cyano, or alkoxy.
Preferably R6 is methyl, cyano or H. More preferably, R6 is H.

R7 is a member selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted heteroalkyl, unsubstituted heteroalkyl, diphosphates, triphosphates, acyl, C(0)R12R13, C(0)0R12, C(0)NR12.'sK13, P(0)(0R1)2, C(0)CHR12R13, SR12 and SiR12R13R14, in which R12, R13, and R14 are members independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl and substituted or unsubstituted aryl, or where R12 and R13 together with the nitrogen or carbon atom to which they are attached are optionally joined to form a substituted or unsubstituted heterocycloalkyl ring system having from 4 to 6 members, optionally containing two or more heteroatoms.
Examples of CBI CC-1065 analogs are described in co-owned U.S. Patent Nos. 7,129,261; 6,989,452; 7,087,600; 7,517,903 and 7,691,962. These references also describe examples of synthesis and uses for these compounds. These compounds can be used as therapeutic agents (e.g., drugs) and as prodrugs. In at least some embodiments, the CBI CC-1065 analogs can be conjugated to targeting agents, such as an antibody, receptor, peptide, lectin, saccharide, nucleic acid or a combination thereof, for use in pharmaceutical compositions that selectively deliver the cytotoxic CBI CC-1065 analogs to desired target cells, such as carcinoma cells.
Representative examples of precancerous conditions that may be targeted by these compounds, include, but are not limited to: metaplasia, hyperplasia, dysplasia, colorectal polyps, actinic ketatosis, actinic cheilitis, human papillomaviruses, leukoplakia, lychen planus and Bowen's disease.
Representative examples of cancers or tumors that may be targeted by these compounds include, but are not limited to: lung cancer, colon cancer, prostate cancer, lymphoma, melanoma, breast cancer, ovarian cancer, testicular cancer, CNS
cancer, renal cancer, kidney cancer, pancreatic cancer, stomach cancer, oral cancer, nasal cancer, cervical cancer and leukemias. It will be readily apparent to the ordinarily skilled artisan that the particular targeting agent can be chosen such that it targets the drug to the tumor tissue to be treated with the drug (i.e., a targeting agent specific for a tumor-specific antigen is chosen). Examples of such targeting agents are well known in the art, non-limiting examples of which include anti-Her2 for treatment of breast cancer, anti-CD20 for 18a treatment of lymphoma, anti-PSMA for treatment of prostate cancer and anti-CD30 for treatment of lymphomas, including non-Hodgkin's lymphoma.
These compounds provide a method of killing a cell. The method includes administering to the cell an amount of a compound of the invention sufficient to kill said cell. In an exemplary embodiment, the compound is administered to a subject bearing the cell. In a further exemplary embodiment, the administration serves to retard or stop the growth of a tumor that includes the cell (e.g., the cell can be a tumor cell).
For the administration to retard the growth, the rate of growth of the cell should be at least 10%
less than the rate of growth before administration. Preferably, the rate of growth will be retarded at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or completely stopped.
Pharmaceutical compositions include compositions wherein the active ingredient is contained in a therapeutically effective amount, i.e., in an amount effective to achieve its intended purpose. The actual amount effective for a particular application will depend, inter alia, on the condition being treated. Determination of an effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure herein.
For any compound described herein, the therapeutically effective amount can be initially determined from cell culture assays. Target plasma concentrations will be those concentrations of active compound(s) that are capable of inhibition cell growth or division.
In preferred embodiments, the cellular activity is at least 25% inhibited.
Target plasma concentrations of active compound(s) that are capable of inducing at least about 50%, 75%, or even 90% or higher inhibition of cellular activity are presently preferred.
The percentage of inhibition of cellular activity in the patient can be monitored to assess the appropriateness of the plasma drug concentration achieved, and the dosage can be adjusted upwards or downwards to achieve the desired percentage of inhibition.
As is well known in the art, therapeutically effective amounts for use in humans can also be determined from animal models. For example, a dose for humans can be formulated to achieve a circulating concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring cellular inhibition and adjusting the dosage upwards or downwards, as described above.
A therapeutically effective dose can also be determined from human data for compounds which are known to exhibit similar pharmacological activities. The applied dose can be adjusted based on the relative bioavailability and potency of the administered compound as compared with the known compound.
Adjusting the dose to achieve maximal efficacy in humans based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.
In the case of local administration, the systemic circulating concentration of administered compound will not be of particular importance. In such instances, the compound is administered so as to achieve a concentration at the local area effective to achieve the intended result.
For use in the prophylaxis and/or treatment of diseases related to abnormal cellular proliferation, a circulating concentration of administered compound of about 0.001 1.1ivI to 1AM is preferred, with about 0.01111VI to 5 I.LM being preferred.
Patient doses for oral administration of the compounds described herein, typically range from about 1 mg/day to about 10,000 mg/day, more typically from about 10 mg/day 15 to about 1,000 mg/day, and most typically from about 50 mg/day to about 500 mg/day.
Stated in terms of patient body weight, typical dosages range from about 0.01 to about 150 mg/kg/day, more typically from about 0.1 to about 15 mg/kg/day, and most typically from about 1 to about 10 mg/kg/day, for example 5 mg/kg/day or 3 mg/kg/day.
In at least some embodiments, patient doses that retard or inhibit tumor growth can 20 be 1 nmol/kg/day or less. For example, the patient doses can be 0.9, 0.6, 0.5, 0.45, 0.3, 0.2, 0.15, or 0.1 Umol/kg/day or less (referring to moles of the drug) of the drug or a drug conjugate, such as an antibody-drug conjugate. Preferably, the drug or drug conjugate growth of the tumor when administered in the daily dosage amount over a period of at least five days. In at least some embodiments, the tumor is a human-type tumor in a SCID
mouse. As an example, the SCID mouse can be a CB17.SCID mouse (available from Taconic, Germantown, NY).
For other modes of administration, dosage amount and interval can be adjusted individually to provide plasma levels of the administered compound effective for the particular clinical indication being treated. For example, in one embodiment, a compound according to the invention can be administered in relatively high concentrations multiple times per day. Alternatively, it may be more desirable to administer a compound of the invention at minimal effective concentrations and to use a less frequent administration regimen. This will provide a therapeutic regimen that is commensurate with the severity of the individual's disease.
Utilizing the teachings provided herein, an effective therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the clinical symptoms demonstrated by the particular patient. This planning should involve the careful choice of active compound by considering factors such as compound potency, relative bioavailability, patient body weight, presence and severity of adverse side effects, preferred mode of administration and the toxicity profile of the selected agent.
Generally, the CBI moiety has the formula:
X

etql =
-where substituents can be attached to the oxygen and nitrogen atoms, X is halo, and R6 is H, substituted or unsubstituted lower alkyl, cyano, or alkoxy. Preferably, R6 is H, methyl, or cyano. More preferably, R.6 is H. In addition, X is preferably Cl or Br and, more preferably, X is Br. Generally, a binding unit can be attached to the amine substituent of the CBI moiety. Examples of suitable binding units include, but are not limited to, R4`
ilso R4 Xi F25' where X, Z, R4, R4', R5, and R5' are as defined above.
Examples of suitable binding units are illustrated and described in U.S.
Patent , .
Nos. 7,129,261; 6,989,452; 7,087,600; 7,517,903 and 7,691,962 as well as in U.S.
Patent No. 6,534,660. Suitable binding units within this formula also include binding units with multiple fused rings such as:
21a R4"
R4' R15 40 Ram N
Z' "

R5,"

where Z' is independently selected from 0, S and NR8 where R8 is a member selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, and acyl.
R4", R4", R5", and R5" are members independently selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, unsubstituted heterocycloalkyl, halogen, NO2, NR9R10, NC(0)R9, OC(0)NR9R1 , OC(0)0R9, C(0)R9, SR9, OR9, CR9=NRI , and 0(CH2)NRI IR11' where R9 and R1 are independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycloalkyl, and substituted or unsubstituted peptidyl, or where R9 and R1 together with the nitrogen atom to which they are attached are optionally joined to form a substituted or unsubstituted heterocycloalkyl ring system having from 4 to 6 members, optionally containing two or more heteroatoms, and R11 and RI1' are each independently H or lower alkyl.
R15 can be H, substituted or unsubstituted alkyl, or R15 and R4' or R5' can be combined to form a ring (e.g., a five- or six-membered ring.) One intermediate compound useful in the formation of CC-1065 CBI analogs has the formula (I):
X

(I) where RI and R2 are each independently H, alkyl, -C(0)OR', -C(0)NR'R", or a protecting group, where R' and R" are independently selected from the group consisting of H, substituted alkyl, unsubstituted alkyl, substituted aryl, unsubstituted aryl, substituted heteroaryl, unsubstituted heteroaryl, substituted heterocycloalkyl, and unsubstituted heterocycloalkyl, and X is halogen.
In one conventional synthetic method for compound (I), the starting material is 1,3-dihydroxynaphthalene which is then reacted with ammonia in a pressurized chamber (e.g., a bomb) to replace the hydroxy group at the 3-position of the naphthalene with an amine.
Examples of this synthetic method can be found in U.S. Patent No. 6,534,660 and D.L.
Boger et al., J. Org. Chem. 57, 2873-2876 (1992). The amination reaction is followed by the addition of protecting groups to both the hydroxy and amine moieties.
Although the amination reaction may have acceptable yield on a small scale, it can be difficult to scale-up the reaction because of the use of a bomb to contain this pressurized reaction which typically occurs at a pressure substantially greater than 1 atmosphere (about 1.01 x 105 Pa) and generally at a pressure of at least 1.5 atmospheres (1.52 x 105 Pa). This synthetic method has been found to result in a substantially lower yield when scaled-up.
In contrast to the conventional methods, the starting material can be compound (II), as illustrated in the synthetic schemes of Figures 1 and 2:
Re 00401 NH2 oR3 (n) where R3 is H or alkyl. Preferably, R3 is C1-5 alkyl and, more preferably, methyl. For example, 4-methoxy-2-naphthylamine is available commercially from Aldrich Chemical Company, Inc., Milwaukee, WI. It is relatively easy to hydrolyze the compound, if R3 is alkyl, and add protecting groups, Ry, to both the hydroxy and amine moieties to form compound (III) R6 NeH
=R1' (III') In some embodiments, as illustrated in Figures I and 2, it is desirable to provide different protecting groups on the amine and hydroxy functionalities.
Accordingly, the initial protecting group, can be removed from one of these moieties (e.g., from the hydroxy moiety) and replaced with a second, different protecting group, R2', to provide compound (III) Nal'H

One specific example of compound (IV) has the formula:
ipso NH y0 Having different protecting groups on the hydroxyl and amine substituents can facilitate later reaction steps where one of the protecting groups can be selectively removed while leaving the other protecting group. Alternatively, different protecting groups can be initially added to the hydroxyl and amine substituents.
Schemes 1 and 2 (Figs. I and 2) illustrate one embodiment of the remaining steps in forming compound (I) from compound (IV). These steps can include, for example, forming a ring using the nitrogen of the amine group. This can be accomplished, for example, by allcylation of the aryl ring adjacent the nitrogen followed by a ring closure step. In one embodiment, iodination of compound (TV) by N-iodosuccinimide produces a compound which can then be alkylated using 1,3-dibromopropene or 1,3-dichloropropene.

Ring closure can then be performed using tributyltin hydride in the presence of 2,2'-azobisisobutyronitrile (AIBN) to give the mcemic CBI-derivative, compound (V):
X
Re NRI' (V) If desired, the protecting groups can he removed to form CBI. It will be understood that different reactants and catalysts can be used in these reaction steps.
Examples can be found in Boger, Chemical Reviews, 97, 787-828 (1997).
The rac,emic CBI-derivative can be separated using known techniques of the separation of enantiomers including the use of chromatographic methods. One particularly useful technique is high pressure liquid chromatography (HPLC) using a chiral column.
For example, separation of such enantiomers has been performed using a HPLC
Chiralcel column and hexane/isopropanol (99:1) eluent to give compound (I).
Specific examples of Compound (I) include, but are not limited to, Br Br =
= H

Cl Br 4 =
00 NBoc H 4110 OBoc = H

400 Boc = Boc jCI /Br /0õ
NBoc NBoc ..
Compound (I) can be used to form CBI CC-1065 analogs as described, for example, in co-owned U.S. Patent Nos. 7,129,261; 6,989,452; 7,087,600; 7,517,903 and 7,691,962. For example, a binding unit can be added to Compound (I) by deprotecting the amine substituent and reacting a compound comprising the binding unit with the deprotected amine. Additional substituents can be added to the oxygen atom of the CBI
compound by deprotecting the oxygen and reacting it with appropriate reactant(s).

EXAMPLES
Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent.
The following preferred specific embodiments are, therefore, to be construed as merely illustrative, and not !imitative of the remainder of the disclosure in any way whatsoever.
26a In the foregoing and in the following examples, all temperatures are set forth uncorrected in degrees Celsius and, all parts and percentages are by weight, unless otherwise indicated.
EXAMPLE 1 - Scheme 1 (Fig. 1) Synthesis of N-(tert-butyloxycarbony1)-4-0-(tert-butyloxycarbony1)-2-naphthylamine (2) A solution of 4-methoxy-2-naphthylamine (230 mg, 1.33 mmol) in glacial acetic acid (9.6 mL) and hydrobromic acid in water (16 mL, 48%) was refluxed under N2 for 4 h.
A small amount of sample (0.1 mL) was diluted with ethyl acetate (0.5 mL), and then water (0.5 mL) and TEA (0.1 mL) were added. TLC (20:1 DCM/methanol) of the organic layer showed no starting material and a new much lower spot (Rf =0.1). The solvent was removed under reduced pressure and the product was dried under vacuum to yield the intermediate 4-hydroxy-2-naphthylamine which was used for the next step without any purification. To a solution of 4-hydroxy-2-naphthylamine in dioxane (10 mL) was added TEA (1 mL) and di-tert-butyl dicarbonate (1.149 g, 5.27 mmol). The reaction mixture was refluxed under N2 for 4 h. TLC (4:1 hexane/ethyl acetate) showed no starting material and a new higher spot (Rf=0.55). The reaction mixture was diluted with ethyl acetate (50 mL) and washed with water. The aqueous layer was extracted with ethyl acetate (2 x 50 mL) and the organics were combined and washed with brine. The organics were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to yield N-(tert-butyloxycarbony1)-4-0-(tert-butyloxycarbony1)-2-naphthylamine (2, 80% yield) as oil.
Synthesis of compound 3 To a solution of compound 2 in acetone (10 mL) was added NaOH solution in water (10 mL, 1 M). The reaction mixture was stirred at room temperature for overnight.
TLC (4:1 hexane/ethyl acetate) showed no starting material and a new lower spot. The reaction mixture was extracted with ethyl acetate (50 mL) and washed with water. The aqueous layer was extracted with ethyl acetate (2 x 50 mL) and the organics were washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified on 10 g silica gel column with 10-20% ethyl acetate in hexane to yield compound 3 (181 mg, 53%) as an oil.

Synthesis of compound 4:
A solution of compound 3 (5g, 19.3 mrnol) in anhydrous DMF (50 ml) under nitrogen atmosphere was treated with benzyl bromide (4 g, 23.1 moles), potassium carbonate (3.7 g, 27 moles) and tetrabutylammonium iodide (70 mg, 0.01 mmoles). The reaction mixture was stirred at room temperature for 8 h. The reaction mixture was concentrated under reduced pressure. Chromatographic (4x 10 cm Si02, 10-20%
Et0Ac-hexane gradient elution) separation provided pure compound 4 (5.48 g, 83%) as a cream powder. 111 NMR (CDC13, 400 MHz, ppm) 8.22 (d, 1H J=8.1 Hz, C5-H), 7.68 (d, 1H, J=8.2 Hz, C8-H), 7.3-7.5 (m, 8H, Cl-H, C6-H, C7-H, CH2C6H5), 7.06 (d, IH, J=1.1 Hz, C3-H), 6.62 (br s, 1H, NH), 5.23 (S, 2H, OCH2(C6H5), 1.55 (s, 9H, OC(CH3)3).
Synthesis of compound 5:
To a 1000 ml round bottom flask equipped with a stir bar and a rubber septum was combined compound 4 (13 g, 0.0372 moles) and THF (300 m1).The clear yellow solution was cooled to -20 C with a dry ice bath under a nitrogen atmosphere. p-Toluenesulfonic acid (0.10 g, 0.0005 moles) was added to the reaction and the solution was stirred for 10 minutes. N-Iodosuccinimide (10 g, 0.0446 moles) was dissolved in THF (50 ml) and added to the reaction by cannula (approximately 1 hr). The solution was stirred in the ice bath for 2 hr and turned brownish. The solution was then removed from the ice bath and let warm to room temperature under nitrogen for 1.5 hr. TLC (2:1 hexane(DCM) showed no starting material and a new higher spot. The reaction was quenched with saturated NaHCO3 (200 ml) and a white solid formed. After stirring the solution for 10 minutes, added Et0Ac (200 ml) and water (100 ml) to the reaction. The aqueous layer was extracted with Et0Ac (2 x 100 ml) and the organics were combined and extracted with brine (100 m1). The organics were dried over MgSO4, filtered, and concentrated under vacuum to a dark red-brown solid. The solid was purified by column chromatography using 2:1 hexane/DCM as eluant to yield compound 5 (14 g, 79%) as a brown solid.
Synthesis of compound 6:
To a 500 ml round bottom flask equipped with stir bar and nitrogen inlet was combined compound 5 (22.5 g, 0.0473 moles) and anhydrous DMF (250 m1). The yellow-orange solution was cooled to 0 C with art ice/salt bath under a nitrogen atmosphere.
NaH (60%, 5.6 g, 0.146 moles) was added to the reaction in one portion. The solution turned cloudy and a gas was formed. The reaction was stirred in the ice bath for 15 minutes and then the ice bath was removed and the solution was stirred for another 15 minutes.
cis/trans-1,3-Dibromopropene (14 ml, 0.14 moles) was added to the reaction in portions by syringe.
The reaction was stirred under nitrogen at room temperature for 1 hr and turned a cloudy brown. The temperature rose to 40 C. The reactions was allowed to cool to room temperature. TLC (4:1 hexane/Et0Ac) showed no starting material and a new lower spot.
The reaction was quenched with water (500 ml). The aqueous layer was extracted with Et0Ac (4 x 100 ml) and the organics were washed with brine (2 x 75 ml). The organics were dried over MgSO4, filtered and concentrated under vacuum to a brown oil.
The product was purified by column chromatography using 1:1 DCM/hexane as eluant to yield compound 6 (25 g, 89%) as a brown oil.
Synthesis of compound 7:
To a 1000 ml three necked round bottom flask equipped with stir bar, temperature probe, reflux condenser and nitrogen inlet was combined compound 6 (25 g, 0.0421 moles), toluene (500 ml), 2,2'-azobis(2-methylpropionitrile) (0.15 g, 0.0009 moles) and tributyltin hydride (3.4 ml, 0.0126 moles) [by syringe]. Nitrogen was bubbled through the solution for 15 minutes and then the reaction was heated to 80 C under nitrogen. After heating at 80 C for 15 minutes, tributyltin hydride (3.4 ml, 0.0126 moles) was added to the reaction by syringe. After heating for another 15 minute tributyltin hydride (3.4 ml, 0.0126 moles) was added to the reaction by syringe. After a further 15 minutes, tributyltin hydride (3.4 ml, 0.0126 moles) was added to the reaction by syringe. The total amount of tributyltin hydride added was 13.6 ml, 0.0505 moles. The reaction was heated at 80 C for 30 minutes and then allowed to cool to room temperature. TLC (10%
Et0Ae/hexane) showed starting material and a new higher spot. The solution was concentrated under vacuum to a yellow solid. The solid was purified by column chromatography using as eluant 1:1 diclorornethane/hexane to give a yellow solid. The solid was recrystallized from hexane (200 ml, 45 C for 30 min, cooled in fridge for 2 hr, collected by filtration, dried under vacuum) to yield compound 7 (11.70 g, 59% yield) as a pale yellow solid.
NIVIR(1H, CDCI3, 400 MHz): El 1.61 (9H, s, C-(CH3)3); 3.30 (1H, t, J = 26 Hz, N); 3.82 (1H, d, J -= 26 Hz, Br-CH2-CH); 4.04 (1H, d, J = 19 Hz, Br-CH2-CH) 4.14 (1H, t, J = 26 Hz, CH-CH2-N); 4.21 (111, m, CH2-CH-CH2); 5.26 (2H, s, 0-CH2-C6H5); 7.3-7.55 (8H, m, 0-CH2-C6H5, C10H5); 7.63 (1H, d, J= 21 Hz, C10H5); 8.3 (1H, d, J = 21 Hz, C10H5) Resolution of compound 7:
The racemic compound 7 was dissolved in DCM (50mg, 1 ml). The solution was then diluted with hexane (9 ml). The solution was then loaded onto a Chiralcel OD prep column (10 micron, 20 x 250 mm) and separated using hexane/isopropanol (99:1, ml/min). The first enantiomer ('7a) elutes from 10 to 15 mm and the second enantiomers (7b) elutes from 17.5 to 25 min. The analytical column (Chiralcel OD, 0.46 x 25 cm, 20 micrometers) gives a retention tirne of 7.71 min for 7a and 12.9 mm for 7b (99:1 hexane/IPA, 1 ml/min, 15 minute run). NMR(1H, CDC13, 400 MHz): 0 1.61 (9H, s, C-(CH3)3); 3.30 (1H, t, J = 26 Hz, CH-CH2-N); 3.82 (1H, d, .1= 26 Hz, Br-CH2-CH); 4.04 (1H, d, J = 19 Hz, Br-CH2-CH) 4.14 (1H, t, J = 26 Hz, CH-C112-N); 4.21 (1H, m, CH2); 5.26 (2H, s, 0-CH2-C61-15); 7.3-7.55 (8H, m, 0-CH2-C6H5, C1055); 7.63 (1H, d, J =
21 Hz, C10H5); 8.3 (1H, d, J = 21 Hz, C101/5).
EXAMPLE 2 - Scheme 2 (Fig. 2) The synthetic method procedure is as described above in Example 1 through the synthesis of compound 5.
Synthesis of compound 8:
To a 250 ml round bottom flask equipped with stir bar and nitrogen inlet was combined compound 5 (8.4 g, 0.0177 moles) and anhydrous DMF (125 ml). The yellow-orange solution was cooled to 0 C with an ice/salt bath under a nitrogen atmosphere.
NaH (60%, 2.22 g, 0.0554 moles) was added to the reaction in one portion. The solution turned cloudy and a gas was formed. The reaction was stirred in the ice bath for 15 minutes and then the ice bath was removed and the solution was stirred for another 15 minutes.
cis/trans-1,3-Dichloropropene (5.3 ml, 0.0571 moles) was added to the reaction in portions by syringe.
The reaction was stirred under nitrogen at room temperature for 3 hr and turned a cloudy brown. TLC (4:1 hexane/Et0Ac) showed no starting material and a new lower spot. The reaction was quenched with water (250 m1). The aqueous layer was extracted with Et0Ac (3 x 100 ml) and the organics were washed with brine (2 x 50 ml). The organics were dried over MgSO4, filtered and concentrated under vacuum to a brown oil. The product was purified by column chromatography using 1:1 DCM/hexane as eluant to yield (E/Z)-tert-butyl 4-(benzyloxy)-1-iodonaphthalene-2-y1(3-chloroallypcarbamate (8) (9 g, 93%) as a yellow oil.

Synthesis of compound 9:
To a 500 ml three necked round bottom flask equipped with stir bar, temperature probe, reflux condenser and nitrogen inlet was combined compound 8 (9 g, 0.0164 moles),toluene (200 ml), 2,2'-azobis(2-methylpropionitrile) (0.15 g, 0.0009 moles) and tributyltin hydride (1.5 ml, 0.0056 moles) [by syringe]. Nitrogen was bubbled through the solution for 15 minutes and then the reaction was heated to 80 C under nitrogen. After heating at 80 C for 15 minutes, tributyltin hydride (1.5 ml, 0.0056 moles) was added to the reaction by syringe. After heating for another 15 minute tributyltin hydride (1.5 ml, 0.0056 moles) was added to the reaction by syringe. After a further 15 minutes, tributyltin hydride (1.0 ml, 0.0037 moles) was added to the reaction by syringe. The total amount of tributyltin hydride added was 5.5 ml, 0.0204 moles. The reaction was heated at 80 C for 30 minutes and then allowed to cool to room temperature. TLC (10%
Et0Ac/hexane) showed no starting material and a new higher spot. The solution was concentrated under vacuum to a yellow oil. The oil was purified by column chromatography using as eluant 100% Hexane to 5% Et0Ac/Hexane to 10% Et0Ac/Hexane to give a pale yellow solid.
The solid was recrystallized from hexane (100 ml, 45 C for 30 min, cooled in fridge for 2 hr, collected by filtration, dried under vacuum) to yield compound 9 (4.16 g, 60% yield) as a white solid.
Resolution of compound 9:
The racemic compound 9 was dissolved in DCM (50mg, 1 m1). The solution was then diluted with hexane (9 ml). The solution was then loaded onto a Chiralcel OD prep column (10 micron, 20 x 250 mm) and separated using hexane/isopropanol (99:1, ml/min). The first enantiomer (9a) elutes from 11.5 to 15 min and the second enantiomers (9b) elutes from 17.5 to 25 min. The analytical column (Chiralcel OD, 0.46 x 25 cm, 20 microns) gives a retention time of 6.5 min for 9a and 10.6 min for 9b (99:1 hexane/IPA, 1 ml/min, 15 minute run). NMR(1H, CDC13, 400 MHz): d 1.61 (91-1, s, C-(CH3)3);
3.44 (111, t, J = 25 Hz, CH-CH2-N); 3.9-4.0 (2H, m, Cl-CH2-CH); 4.12 (1H, t, J = 26 Hz, N); 4.25 (1H, m, C1-12-CFI-CH2); 5.26 (2H, s, 0-CH2-C6115); 7.2-7.5 (811, m, 0-CH2-C6H5, C10H5); 7.63 (1H, d, J = 21 Hz, C10H5); 8.3(114, d, J = 21 Hz, C10H5).
EXAMPLE 3- Scheme 3 (Fig. 3) The synthetic method procedure is as described above in Example 1 through the synthesis of compound 3.

Synthesis of compound 10:
A solution of tert-butyl-4-hydroxynaphthalen-2-ylcarbamate (3) (500 mg, 2.89 mmol), 4-methyl-l-piperazinecarbonyl chloride hydrochloride (858 mg, 4.34 mmol), anhydrous pyridine (4.98 ml, 57.8 mmol), and ally! alcohol (4.98 ml, 73.2 mmol) in anhydrous DCM (20 ml) was stirred at room temperature for overnight. TLC (9:1 DCM/Me0H) showed no starting material and a much lower spot. The reaction mixture was quenched with water. The aqueous layer was extracted with Et0Ac (3 x 50 ml) and the organics were washed with brine (2x 50 ml). The organics were dried over Na2SO4, filtered and concentrated under vacuum to brown oil. The crude product was purified by column chromatography with 1-5% methanol in DCM to yield 10 (602 mg, 82%) as yellow solid.
11-1NMR. (DMSO-d6) 5 9.68 (s, 111), 7.91 (s, 1H), 7.80 (d, 11-1), 7.69 (s, 1H), 7.47 (t, 1 H), 7.42 (s, 1H), 7.39 (t, 1H), 3.78 (s, 211), 3.42 (s, 2H), 2.44 (s, 2H), 2.39 (s, 211), 2.21 (s, 3H), 1.50 (s, 911).
Synthesis of compound 11:
Compound 10 (82 mg, 0.21 mmol),p-Toluenesulfonic acid (10 mg, 0.05 mmol), and N-Iodosuccinimide (96 mg, 0.43 mmol) in anhydrous THF (5 ml) was stirred at room temperature overnight. TLC (9:1 DCM/Me0H) showed a small amount of starting material and a higher spot. The reaction mixture was quenched with saturated NaHCO3 (10 ml). After stirring at room temperature for 10 min, the reaction mixture was extracted with Et0Ac (3 x 20 ml) and the organics were washed with brine (2x 20 m1). The organics were dried over Na2SO4, filtered and concentrated under vacuum to brown oil.
The crude product was purified by column chromatography with 1-5% methanol in DCM to yield 11 (52 mg, 48%) as yellow oil.
11-INMR (DMSO-d6) 5 8.81 (s, 111), 8.14 (d, 111), 7.79 (d, 1H), 7.65 (t, 1 H), 7.58 (t, 1H), 7.45 (t, 1H), 3.78 (s, 211), 3.42 (s, 211), 2.44 (s, 211), 2.39 (s, 2H), 2.21 (s, 31-1), 1.50 (s, 9H).
Synthesis of compound 12:
A solution of compound 11 (102 mg, 0.2 mmol) in anhydrous DMF (5 ml) was cooled in an ice bath. Sodium hydride (60% in mineral oil, 32 mg, 0.8 mmol) was added to the reaction. The reaction mixture was stirred at 0 C for 15 min and at room temperature for 15 min. cis/trans-1,3-Dichloroprepene (83.36 pl, 0.9 mmol) was added to the reaction. The reaction mixture was stirred at room temperature for 1 hr.
TLC (9:1 DCM/Me0H) showed no starting material. The reaction mixture was quenched with water. The aqueous layer was extracted with Et0Ac (3 x 10 ml) and the organics were washed with brine (2 x 10 ml). The organics were dried over Na2SO4, filtered and concentrated under vacuum to brown oil. The crude product was purified by column chromatography with 1-5% methanol in DCM to yield 12 (82 mg, 70%) as yellow solid.
11-INMR (DMSO-d6) 6 8.20 (d, 1H), 7.82 (d, 1H), 7.62 (m, 2H), 7.38 (d, 1H),
6.38 (m, 1H), 6.18 (m, 1H), 3.98-4.46 (dd, 2H), 3.78 (s, 2H), 3.42 (s, 2H), 2.44 (s, 2H), 2.39 (s, 2H), 2.21 (s, 3H), 1.50 (s, 9H).
Synthesis of compound 13:
To a solution of 12 (82mg, 0.14 mmol) in anhydrous toluene (3 ml), dry nitrogen was bubbled for 15 min. Tributyltin hydride (47.1 pl, 0.18 mmol) and 2,2'-azobisisobutyronitrile (10 mg, 0.06 mmol) were added to the reaction. The reaction mixture was heated at 80 C for 15 min under nitrogen. TLC (9:1 DCM/Me0H) showed new blue spot and no starting material. The reaction mixture was concentrated under vacuum to yellow oil. The crude product was purified by column chromatography with 1-5% methanol in DCM to yield 13 (52 mg, 82%) as white solid.
11-1NMR (DMSO-d6) 6 7.92 (d, 1H), 7.83 (m, 1H), 7.78 (d, 1H), 7.58 (t, 1H),
7.42 (t, 1H), 4.20 (m, 2H), 4.04 (m, 2H), 3.92 (m, 1H), 3.78 (s, 2H), 3.42 (s, 2H), 2.44 (s, 2H), 2.39 (s, 2H), 2.21 (s, 3H), 1.50 (s, 9H).

Resolution of compound 13:
The racemic compound 13 is dissolved in methanol. The solution is then loaded onto a CHIRALPAK* AD prep column (20 micron, 20 x 250 mm) and separated using methanol (15 ml/min). The first enantiomer (13a) elutes from 5.1 min and the second enantiomers (13b) elutes from 7.1 min. 11-INMR (DMSO-d6) 6 7.92 (d, 1H), 7.83 (m, 1H), 7.78 (d, 1H), 7.58 (t, 1H), 7.42 (t, 1H), 4.20 (m, 2H), 4.04 (m, 2H), 3.92 (m, 1H), 3.78 (s, 2H), 3.42 (s, 2H), 2.44 (s, 2H), 2.39 (s, 2H), 2.21 (s, 3H), 1.50 (s, 9H).
Example 4 - Synthesis of CBI CC-1065 Analog o rci 401 r a 40 ,C1 HO2C /N
Bn0 40 lel H2, Pd-C, 0 4N HCI in Et0Ac H
= =
EDC
N HO N HO el N
µ
Boc Boc H
9b 14 15 401 ,CI 0 401 HO lei r C I
J-L
(N Cl/y I 00 0 rsL) 0 N ir i-N 0 N
b / 00 DCM __ N
/ N e N
H H

0.0õ.;;-...N..1c.õ---..N ...1( TFA 0 H \ 401 ,CI
H2N N)c.c.-- H
so.......) 5% AcOH in DCM el I
'N N )-LO \
e N
H

*trademark Synthesis of Compound (14). A solution of 9b (100 mg, 0.24 mmol) and 10%
Pd¨C (35 mg) in Me0H/CH2C12 (1/2, 10 ml) was degassed in vacuo for 40 s. The resulting mixture was placed under an atmosphere of hydrogen and stirred at 25 C
for 7 h. The reaction mixture was filtered through Celite* (CH2Cl2 wash). The solvent was removed in vacuo. Chromatography on silica gel eluted with Et0Ac/Hex (2/8) afforded 14 (77 mg, 98%). 1NMR (DMSO-d6) 6 10.36 (s, 1H), 8.04 (d, 1H, J=8.2 Hz), 7.72 (d, 1H, J=8.2 Hz), 7.61 (br s, 1H), 7.45 (t, 1H, J=8.4 Hz), 7.261 (t, 1H, J=8.4 Hz), 4.06 (m, 4H), 3.73 (m, 1H), 1.52 (s, 9H).
Synthesis of Compound (16). A solution of 14 (35 mg, 0.1 mmol) in 4 M HCI-Et0Ac (5 ml) was stirred at 25 C under Ar for 30 min. The solvent was removed in vacuo. To the residue was added 5-acetylindone-2-carboxylic acid (24.4 mg, 0.12 mmol). A solution of EDC (22.9 mg, 0.12 mmol) in DMF (3 ml) was added and the reaction mixture was stirred * trademark 34a at 25 C for 5 h. The solvent was removed. The crude product was chromatographed on silica gel eluted with 10% Me0H in CH2C12to give 16 (40.7mg, 93%). IHNMR DMSO-d6) 812.13 (s, 1H), 10.47 (s, 1H), 8.45 (s, 11-1), 8.10 (d, 1H, J=8.4 Hz), 7.96 (br s, 1H), 7.85 (d, 2H, J=8.4 Hz), 7.54 (d, 1H, J=8.4 Hz), 7.51 (t, 1H, J=8.2 Hz), 7.36 (t, 1H, J=7.6), 7.35 (s, 111), 4.81 (t, 1H, 11.2 Hz), 4.54 (dd, 1H, 8.8 Hz), 4.23 (m, 1H), 4.01 (dd, 1H, J=10.2 Hz), 3.86 (dd, 1H, J=10.7 Hz), 2.61 (s, 31-1).
Synthesis of Compound (17). 4-Methyl-l-piperazinecarbonyl chloride hydrochloride (19.9 mg, 0.1 mmol) was added to a solution of 16 (20 mg, 0.05 mmol) and anhydrous pyridine (25 uml, 0.3 mrnol) in 3% allyl alcohol in dry methylene chloride (4 ml) and the mixture was stirred for 16 h. Purification of the crude product on silica gel yielded 17 (23.6 mg, 91%). INMR DMSO-d6) 8 12.03 (s, 1H), 8.41 (s, 111), 8.21 (s, 11-1), 8.01 (d, 1H, J=8.4 Hz), 7.88 (d, 1H, J=8.4 Hz), 7.82 (dd, 1H, J=8.4 Hz), 7.58 (t, 1H, J=8.1 Hz), 7.51 (d, 1H, J=8.4 Hz), 7.46 (t, 1H, J=7.6 Hz), 7.37 (s, 1H), 4.86 (t, 1H, J=10.8 Hz), 4.57 (dd, 1H, .1=10.8 Hz), 4.38 (in, 1H), 4.06 (dd, 111, J=10.8 Hz), 3.86 (dd, 1H, J=11 Hz), 3.41 (br, 4H), 3.29 (br, 4H), 2.82 (s, 3H), 2.57 (s, 3H).
Synthesis of Compound (19). A solution of 17 (13 mg, 24 umol) and linker 18 (16.9 mg, 31 umol) in 5% acetic acid in dry methylene chloride (1 ml) was stirred for 30 min at 25 C. The solvent was completely removed in vacuo and purified by HPLC
(SymmetryPrep C18,7ttm, 19 x 150 mm column) to give 19 (18.5 mg, 81%). MS: calcd for C48H57C1.1\18011 (M+H) m/z 958.38, found 958.10.
EXAMPLE 5: Proliferation Assays The biological activity of the cytotoxic compounds of the invention can be assayed using the well established 3H-thymidine proliferation assay. This is a convenient method for quantitating cellular proliferation, as it evaluates DNA synthesis by measuring the incorporation of exogenous radiolabeled 3H-thymidine. This assay is highly reproducible and can accommodate large numbers of compounds.
To carry out the assay, promyelocytic leukemia cells, HL-60, are cultured in RPMI
media containing 10% heat inactivated fetal calf serum (FCS). On the day of the study, the cells are collected, washed and resuspended at a concentration of 0.5 x 106cells/m1 in RPMI containing 10% FCS. 100 I of cell suspension is added to 96 well plates.
Serial dilutions (3-fold increments) of doxorubicin (as a positive control) or test compounds are made and 100 ill of compounds are added per well. Finally 10 JIl of a 100 Ci/m131-1-thymidine is added per well and the plates are incubated for 24 hours. The plates are harvested using a 96 well Harvestd(Packard Instruments) and counted on a Packard Top Count counter. Four parameter logistic curves are fitted to the 3H-thymidine incorporation as a function of drug molarity using Prism software to determine ICso values.
The CBI CC-1065 analogs (e.g., compound 19 of Example 4) generally have an ICso value in the above assay of from about 1 pM to about 100 n.M, preferably from about pM to about 10 nM.
EXAMPLE 6: Conjugation of Drug Molecules to Antibodies This example describes reaction conditions and methodologies for conjugating a drug molecule of the invention (optionally including other groups, such as spacers, reactive functional groups and the like) to an antibody as a targeting agent. The conditions and methodologies are intended to be exemplary only and non-limiting. Other approaches for conjugating drug molecules to antibodies are known in the art.
The conjugation method described herein is based on introduction of free thiol groups to the antibody through reaction of lysines of the antibody with 2-iminothiolane, followed by reaction of the drug-linker molecule with an active maleimide group. Initially the antibody to be conjugated is buffer exchanged into 0.1M phosphate buffer pH 8.0 containing 50mM NaCI, 2mM DTPA, pH 8.0 and concentrated to 5-10 mg/ml.
Thiolation is achieved through addition of 2-iminothiolane to the antibody. The amount of iminothiolane to be added is determined in preliminary experiments and varies from antibody to antibody. In the preliminary experiments, a titration of increasing amounts of 2-iminothiolane is added to the antibody, and following incubation with the antibody for one hour at room temperature, the antibody is desalted into 50mM HEPES buffer pH 6.0 *trademark using a Sephadex G-25*column and the number of thiol groups introduced determined rapidly by reaction with dithiodipyridine (DTDP). Reaction of thiol groups with DTDP
results in liberation of thiopyridine which is monitored at 324nm. Samples at a protein concentration of 0.5-1.0 mg/ml can be used. The absorbance at 280nm is used to accurately determine the concentration of protein in the samples, and then an aliquot of each sample (0.9m1) is incubated with 0.1 ml DTDP (5mM stock solution in ethanol) for 10 minutes at room temperature. Blank samples of buffer alone plus DTDP are also incubated alongside.
After 10 minutes, absorbance at 324nm is measured and the number of thiols present quantitated using an extinction coefficient for thiopyridine of 19800M-I.
Typically a thiolation level of three thiol groups per antibody is desired.
For example, with one particular antibody this can be achieved through adding a 15 fold molar excess of 2-iminothiolane followed by incubation at room temperature for 1 hour.
Antibody to be conjugated is therefore incubated with 2-iminothiolane at the desired molar ratio and then desalted into conjugation buffer (50mM HEPES buffer pH 6.0 containing 5mM glycine, 3% Glycerol and 2mM DTPA). The thiolated material is maintained on ice whilst the number of thiols introduced is quantitated as described above.
After verification of the number of thiols introduced, the drug molecule containing an active maleimide group (e.g., compound 15 of Example 3) is added at a 3-fold molar excess per thiol. The conjugation reaction is carried out in conjugation buffer also containing a final concentration of 5% ethylene glycol dimethyl ether (or a suitable alternative solvent). Commonly, the drug stock solution is dissolved in 90%
ethylene glycol dimethyl ether, 10% dimethyl sulfoxide. For addition to antibody, the stock solution is added directly to the thiolated antibody, which has enough ethylene glycol dimethyl ether added to bring the final concentration to 5%, or pre-diluted in conjugation buffer containing a final concentration of 10% ethylene glycol dimethyl ether, followed by addition to an equal volume of thiolated antibody.
*trademark The conjugation reaction is incubated at room temperature for 2 hours with mixing.
Following incubation the reaction mix is centrifuged at 14000 RPM for 15 minutes and the can be adjusted to 7.2 if purification was not immediate. Purification of conjugate can be achieved through chromatography using a number of methods. Conjugate can be purified using size-exclusion chromatography on a Sephacryl S200*column pre-equilibrated with 50mM HEPES buffer pH 7.2 containing 5mM glycine, 50mM NaC1 and 3% glycerol. Chromatography can be carried out at a linear flow rate of 28 cm/h.
Fractions containing conjugate can be collected, pooled and concentrated.
Alternatively purification can be achieved through ion-exchange chromatography. Conditions vary from antibody to antibody and need to be optimized in each case. For example, antibody-drug conjugate reaction mix can be applied to an SP-Sepharose column pre-equilibrated in 50mM HEMS, 5mM glycine, 3% glycerol, pH 6Ø The antibody conjugate can be eluted using a gradient of 0-1M NaC1 in equilibration buffer. Fractions containing the conjugate can be pooled, the pH can be adjusted to 7.2 and the sample concentrated as required.
The preceding examples can be repeated with similar success by substituting the generically or specifically described reactants and/or operating conditions of this invention for those used in the preceding examples.
Of course, the scope of the claims should not be limited by the preferred embodiments set forth in the examples, but should be given the broadest interpretation consistent with the description as a whole.
* trademarks

Claims (5)

1. A method of making a compound (V) or a salt thereof, wherein R1' and R2' are different protecting groups; R6 is H, a fully saturated straight or branched C1-C6 homoalkyl, cyano, or a fully saturated straight or branched C1-C6 homoalkoxy; and X is CI or Br, the method comprising:
(i) adding protecting groups R1' and R2' to a compound (II), wherein R3 is a fully saturated straight or branched homoalkyl and R6 is as defined above, to form a compound (IV) wherein R1', R2' and R6 are as defined above;
(ii) iodinating the compound of formula (IV) obtained from step (i) with N-iodosuccinimide, (iii) alkylating the product obtained from the iodinating step (ii) with 1,3-dibromopropene or 1,3-dichloropropene, and (iv) performing a ring closure on the product obtained from the alkylating step (iii) with tributyltin hydride in the presence of 2,2'-azobisisobutyronitrile to generate a five membered ring comprising the amine nitrogen of compound (V).
2. The method of claim 1, wherein R3 is methyl.
3. The method of claim 1, wherein protecting groups R1' and R2' are selected from: BOC; FMOC; 2-trimethylsilylethoxycarbonyl; allyloxycarbonyl; 4-methyl-1-piperazinecarbonyl; 1-methyl-1-(4-biphenyl)ethoxycarbonyl;
diphenyloxycarbonyI;
benzyl; t-butyl; tetrahydropyran, trimethylsilyl, t-butyldimethylsilyl;
triisopropylsilyl; t-butyldiphenylsilyl; 2,2,2-trichloroethyl oxycarbonyl; diisopropylmethyl oxycarbonyl;
vinyl oxycarbonyl; methoxy benzyl oxycarbonyl; nitrobenzyl oxycarbonyl;
cyclohexyl oxycarbonyl; cyclopentyl oxycarbonyl; benzyloxycarbonyl; formyl; acetyl;
trihaloacetyl; benzoyl; nitrophenylacetyl; 2-nitrobenzenesulfonyl;
phthalimido; and dithiasuccinoyl.
4. The method of claim 1, wherein R1' is tert-butyloxycarbonyl.
5. The method of claim 4, wherein R2' is -CH2Ph.
CA2627046A 2005-10-26 2006-10-18 Methods and compounds for preparing cc-1065 analogs Expired - Fee Related CA2627046C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US73080405P 2005-10-26 2005-10-26
US60/730,804 2005-10-26
PCT/US2006/060050 WO2007051081A1 (en) 2005-10-26 2006-10-18 Methods and compounds for preparing cc-1065 analogs

Publications (2)

Publication Number Publication Date
CA2627046A1 CA2627046A1 (en) 2007-05-03
CA2627046C true CA2627046C (en) 2015-09-15

Family

ID=37766648

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2627046A Expired - Fee Related CA2627046C (en) 2005-10-26 2006-10-18 Methods and compounds for preparing cc-1065 analogs

Country Status (23)

Country Link
US (1) US7847105B2 (en)
EP (1) EP1940789B1 (en)
JP (1) JP5116686B2 (en)
KR (1) KR20080068084A (en)
CN (1) CN101365679B (en)
AT (1) ATE534629T1 (en)
AU (1) AU2006305842B2 (en)
BR (1) BRPI0619331A2 (en)
CA (1) CA2627046C (en)
CY (1) CY1112533T1 (en)
DK (1) DK1940789T3 (en)
EA (1) EA015324B1 (en)
ES (1) ES2375843T3 (en)
HK (1) HK1118549A1 (en)
HR (1) HRP20120058T1 (en)
IL (1) IL190971A (en)
NO (1) NO20082243L (en)
PL (1) PL1940789T3 (en)
PT (1) PT1940789E (en)
RS (1) RS52100B (en)
SI (1) SI1940789T1 (en)
WO (1) WO2007051081A1 (en)
ZA (1) ZA200804521B (en)

Families Citing this family (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100056762A1 (en) 2001-05-11 2010-03-04 Old Lloyd J Specific binding proteins and uses thereof
DE60234094D1 (en) 2001-05-11 2009-12-03 Ludwig Inst For Cancer Res Ltd SPECIFIC TIE PROTEINS AND ITS USE
AU2006294663B2 (en) 2005-09-26 2012-03-22 Medarex, Inc. Human monoclonal antibodies to CD70
CN104013956B (en) 2007-01-25 2018-12-18 达娜-法勃肿瘤研究所公司 Purposes of the anti-egfr antibodies in the mutant mediated disease for the treatment of EGFR
WO2008115404A1 (en) 2007-03-15 2008-09-25 Ludwing Institute For Cancer Research Treatment method using egfr antibodies and src inhibitors and related formulations
US9901567B2 (en) 2007-08-01 2018-02-27 Syntarga B.V. Substituted CC-1065 analogs and their conjugates
MX2010001757A (en) 2007-08-14 2010-09-14 Ludwig Inst Cancer Res Monoclonal antibody 175 targeting the egf receptor and derivatives and uses thereof.
WO2009026274A1 (en) 2007-08-22 2009-02-26 Medarex, Inc. Site-specific attachment of drugs or other agents to engineered antibodies with c-terminal extensions
CN101951946B (en) 2007-10-01 2014-12-10 百时美施贵宝公司 Human antibodies that bind mesothelin, and uses thereof
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
CN106967062A (en) * 2008-11-03 2017-07-21 辛塔佳股份有限公司 The analogs of novel C C 1065 and its conjugate
WO2010096394A2 (en) 2009-02-17 2010-08-26 Redwood Biosciences, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
BRPI1009232B1 (en) 2009-03-05 2022-05-03 E.R. Squibb & Sons, Llc. Isolated monoclonal antibody or an antigen-binding portion thereof, or an antibody fragment, composition comprising them, nucleic acid molecule, hybridoma and methods for preparing an anti-cadm1 antibody
US8394922B2 (en) 2009-08-03 2013-03-12 Medarex, Inc. Antiproliferative compounds, conjugates thereof, methods therefor, and uses thereof
TR201802539T4 (en) 2010-04-21 2018-03-21 Syntarga Bv Conjugates of CC-1065 analogs and bifunctional binders.
US8956859B1 (en) 2010-08-13 2015-02-17 Aviex Technologies Llc Compositions and methods for determining successful immunization by one or more vaccines
JOP20210044A1 (en) 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
CA2824143C (en) 2011-01-14 2018-12-18 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
AU2013221873B2 (en) 2012-02-13 2016-11-17 Bristol-Myers Squibb Company Enediyne compounds, conjugates thereof, and uses and methods therefor
AR091649A1 (en) 2012-07-02 2015-02-18 Bristol Myers Squibb Co OPTIMIZATION OF ANTIBODIES THAT FIX THE LYMPHOCYTE ACTIVATION GEN 3 (LAG-3) AND ITS USES
EP2956173B1 (en) 2013-02-14 2017-03-29 Bristol-Myers Squibb Company Tubulysin compounds, methods of making and use
CA2935430C (en) 2014-01-10 2018-09-18 Synthon Biopharmaceuticals B.V. Duocarmycin adcs for use in treatment of endometrial cancer
AU2015205509B2 (en) 2014-01-10 2019-08-15 Byondis B.V. Duocarmycin ADCs showing improved in vivo antitumor activity
EP3092010B1 (en) 2014-01-10 2018-07-11 Synthon Biopharmaceuticals B.V. Method for purifying cys-linked antibody-drug conjugates
EP3647322B1 (en) 2014-03-20 2021-10-20 Bristol-Myers Squibb Company Stabilized fibronectin based scaffold molecules
RS59643B1 (en) 2014-06-06 2020-01-31 Bristol Myers Squibb Co Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
US10077287B2 (en) 2014-11-10 2018-09-18 Bristol-Myers Squibb Company Tubulysin analogs and methods of making and use
CN113773388A (en) 2014-11-21 2021-12-10 百时美施贵宝公司 anti-CD 73 antibodies and uses thereof
CN107250157B (en) 2014-11-21 2021-06-29 百时美施贵宝公司 Antibodies comprising modified heavy chain constant regions
JP6622814B2 (en) 2014-11-25 2019-12-18 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Novel PD-L1 binding polypeptides for imaging
BR112017010414A2 (en) 2014-11-25 2018-05-15 Bristol-Myers Squibb Company methods and compositions for 18f radiolabeling of biological substances
CA2971542A1 (en) 2014-12-19 2016-06-23 Regenesance B.V. Antibodies that bind human c6 and uses thereof
MX2017009144A (en) 2015-01-14 2017-11-22 Bristol Myers Squibb Co Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using.
ES2809125T3 (en) 2015-09-23 2021-03-03 Bristol Myers Squibb Co Glypican-3 binding fibronectin-based scaffold molecules
CN108738324B (en) 2015-11-19 2022-06-21 百时美施贵宝公司 Anti-glucocorticoid-induced tumor necrosis factor receptor (GITR) antibodies and uses thereof
AU2017228470A1 (en) 2016-03-04 2018-08-30 Bristol-Myers Squibb Company Combination therapy with anti-CD73 antibodies
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11344639B2 (en) 2016-06-01 2022-05-31 Bristol-Myers Squibb Company PET imaging with PD-L1 binding polypeptides
WO2017210335A1 (en) 2016-06-01 2017-12-07 Bristol-Myers Squibb Company Imaging methods using 18f-radiolabeled biologics
PE20190418A1 (en) 2016-07-14 2019-03-19 Bristol Myers Squibb Co ANTIBODIES AGAINST PROTEIN 3 CONTAINING THE MUCIN AND IMMUNOGLOBULIN T-LYMPHOCYTE DOMAIN (TIM3) AND THEIR USES
ES2902179T3 (en) 2016-08-19 2022-03-25 Bristol Myers Squibb Co Seco-cyclopropapyrroloindole compounds, antibody-drug conjugates thereof, and methods of making and use
US10398783B2 (en) 2016-10-20 2019-09-03 Bristol-Myers Squibb Company Antiproliferative compounds and conjugates made therefrom
SG11201906947SA (en) 2017-02-17 2019-08-27 Bristol Myers Squibb Co Antibodies to alpha-synuclein and uses thereof
US11339218B2 (en) 2017-05-10 2022-05-24 Zhejiang Shimai Pharmaceutical Co., Ltd. Human monoclonal antibodies against LAG3 and uses thereof
CN116333129A (en) 2017-05-25 2023-06-27 百时美施贵宝公司 Antibodies comprising modified heavy chain constant regions
US10472361B2 (en) 2017-08-16 2019-11-12 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a benzotriazole moiety, conjugates thereof, and methods and uses therefor
US10457681B2 (en) 2017-08-16 2019-10-29 Bristol_Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a tricyclic moiety, conjugates thereof, and methods and uses therefor
US10487084B2 (en) 2017-08-16 2019-11-26 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a heterobiaryl moiety, conjugates thereof, and methods and uses therefor
US10494370B2 (en) 2017-08-16 2019-12-03 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a pyridine or pyrazine moiety, conjugates thereof, and methods and uses therefor
US10508115B2 (en) 2017-08-16 2019-12-17 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having heteroatom-linked aromatic moieties, conjugates thereof, and methods and uses therefor
WO2019075090A1 (en) 2017-10-10 2019-04-18 Tilos Therapeutics, Inc. Anti-lap antibodies and uses thereof
CN111886255A (en) 2018-01-12 2020-11-03 百时美施贵宝公司 anti-TIM 3 antibodies and uses thereof
TW202003565A (en) 2018-03-23 2020-01-16 美商必治妥美雅史谷比公司 Antibodies against MICA and/or MICB and uses thereof
JOP20200240A1 (en) 2018-04-02 2020-09-27 Bristol Myers Squibb Co Anti-trem-1 antibodies and uses thereof reference to sequence listing submitted electronically via efs-web
WO2019209811A1 (en) 2018-04-24 2019-10-31 Bristol-Myers Squibb Company Macrocyclic toll-like receptor 7 (tlr7) agonists
US11884729B2 (en) 2018-06-29 2024-01-30 ApitBio, Inc Anti-L1CAM antibodies and uses thereof
US11554120B2 (en) 2018-08-03 2023-01-17 Bristol-Myers Squibb Company 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor
WO2020076969A2 (en) 2018-10-10 2020-04-16 Tilos Therapeutics, Inc. Anti-lap antibody variants and uses thereof
WO2020112781A1 (en) 2018-11-28 2020-06-04 Bristol-Myers Squibb Company Antibodies comprising modified heavy constant regions
US20210403602A1 (en) 2018-12-03 2021-12-30 Bristol-Myers Squibb Company Anti-ido antibody and uses thereof
CN109762067B (en) 2019-01-17 2020-02-28 北京天广实生物技术股份有限公司 Antibodies that bind human Claudin18.2 and uses thereof
EP3914622A1 (en) 2019-01-22 2021-12-01 Bristol-Myers Squibb Company Antibodies against il-7r alpha subunit and uses thereof
CN114144435A (en) 2019-07-15 2022-03-04 百时美施贵宝公司 Antibodies against human TREM-1 and uses thereof
CN114174536A (en) 2019-07-15 2022-03-11 百时美施贵宝公司 anti-TREM-1 antibodies and uses thereof
IL294663A (en) 2020-01-13 2022-09-01 Neoimmunetech Inc Method of treating a tumor with a combination of il-7 protein and a bispecific antibody
AU2021228061A1 (en) 2020-02-27 2022-10-13 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Antibodies binding IL4R and uses thereof
EP4132971A1 (en) 2020-04-09 2023-02-15 Merck Sharp & Dohme LLC Affinity matured anti-lap antibodies and uses thereof
CN114685669A (en) 2020-12-30 2022-07-01 和铂医药(苏州)有限公司 Antibodies that bind TROP2 and uses thereof
EP4314068A1 (en) 2021-04-02 2024-02-07 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses thereof

Family Cites Families (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4169888A (en) 1977-10-17 1979-10-02 The Upjohn Company Composition of matter and process
US4391904A (en) 1979-12-26 1983-07-05 Syva Company Test strip kits in immunoassays and compositions therein
US4671958A (en) 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
US4978757A (en) 1984-02-21 1990-12-18 The Upjohn Company 1,2,8,8a-tetrahydrocyclopropa (C) pyrrolo [3,2-e)]-indol-4(5H)-ones and related compounds
US4912227A (en) 1984-02-21 1990-03-27 The Upjohn Company 1,2,8,8A-tetrahydrocyclopropa(c)pyrrolo(3,2-e)-indol-4-(5H)-ones and related compounds
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
JPH0684377B1 (en) 1986-04-17 1994-10-26 Kyowa Hakko Kogyo Kk
US5332837A (en) 1986-12-19 1994-07-26 The Upjohn Company CC-1065 analogs
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US5773435A (en) 1987-08-04 1998-06-30 Bristol-Myers Squibb Company Prodrugs for β-lactamase and uses thereof
US4952394A (en) 1987-11-23 1990-08-28 Bristol-Myers Company Drug-monoclonal antibody conjugates
US4994578A (en) 1987-11-27 1991-02-19 Meiji Seika Kaisha, Ltd. Certain anti-tumor duocarmycin antibiotics from streptomyces
US5147786A (en) 1988-04-22 1992-09-15 Monsanto Company Immunoassay for the detection of α-haloacetamides
JP2642165B2 (en) 1988-07-22 1997-08-20 協和醗酵工業株式会社 Novel DC-89 compound and method for producing the same
US5084468A (en) 1988-08-11 1992-01-28 Kyowa Hakko Kogyo Co., Ltd. Dc-88a derivatives
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5176996A (en) 1988-12-20 1993-01-05 Baylor College Of Medicine Method for making synthetic oligonucleotides which bind specifically to target sites on duplex DNA molecules, by forming a colinear triplex, the synthetic oligonucleotides and methods of use
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
JP2598116B2 (en) 1988-12-28 1997-04-09 協和醗酵工業株式会社 New substance DC113
US5187186A (en) 1989-07-03 1993-02-16 Kyowa Hakko Kogyo Co., Ltd. Pyrroloindole derivatives
JP2510335B2 (en) 1989-07-03 1996-06-26 協和醗酵工業株式会社 DC-88A derivative
US5495009A (en) 1989-10-24 1996-02-27 Gilead Sciences, Inc. Oligonucleotide analogs containing thioformacetal linkages
US5334528A (en) 1989-10-30 1994-08-02 The Regents Of The University Of California Monoclonal antibodies to cyclodiene insecticides and method for detecting the same
ATE139258T1 (en) 1990-01-12 1996-06-15 Cell Genesys Inc GENERATION OF XENOGENE ANTIBODIES
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
AU648313B2 (en) 1990-04-25 1994-04-21 Pharmacia & Upjohn Company Novel CC-1065 analogs
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5137877B1 (en) 1990-05-14 1996-01-30 Bristol Myers Squibb Co Bifunctional linking compounds conjugates and methods for their production
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
DE69121334T2 (en) 1990-07-26 1997-03-20 Kyowa Hakko Kogyo Kk DC-89 derivatives as anti-tumor agents
GB9017024D0 (en) 1990-08-03 1990-09-19 Erba Carlo Spa New linker for bioactive agents
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
KR100272077B1 (en) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 Transgenic non-human animals capable of producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
JPH0597853A (en) 1991-10-07 1993-04-20 Kyowa Hakko Kogyo Co Ltd Hydrobromic acid salt of dc-89 derivative
PT1696031E (en) 1991-12-02 2010-06-25 Medical Res Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
ES2149768T3 (en) 1992-03-25 2000-11-16 Immunogen Inc CONJUGATES OF BINDING AGENTS OF CELLS DERIVED FROM CC-1065.
GB9314960D0 (en) 1992-07-23 1993-09-01 Zeneca Ltd Chemical compounds
JP3514490B2 (en) 1992-08-21 2004-03-31 杏林製薬株式会社 Trifluoromethylpyrroloindole carboxylate derivative and method for producing the same
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5324483B1 (en) 1992-10-08 1996-09-24 Warner Lambert Co Apparatus for multiple simultaneous synthesis
DE4314091A1 (en) 1993-04-29 1994-11-03 Boehringer Mannheim Gmbh Immunological detection method for triazines
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5786377A (en) 1993-11-19 1998-07-28 Universidad De Santiago De Compostela Pyrrolo 3,2-E!indol derivatives, process for the preparation thereof and applications
ATE271557T1 (en) 1994-04-22 2004-08-15 Kyowa Hakko Kogyo Kk DC-89 DERIVATIVE
JPH07309761A (en) 1994-05-20 1995-11-28 Kyowa Hakko Kogyo Co Ltd Method for stabilizing duocamycin derivative
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
ATE380559T1 (en) 1994-08-19 2007-12-15 Wallone Region CONJUGATES CONTAINING AN ANTITUMORAL AGENT AND USE THEREOF
EP0786252A4 (en) 1994-09-30 1998-01-07 Kyowa Hakko Kogyo Kk Antitumor agent
AU693283B2 (en) 1994-11-29 1998-06-25 Kyorin Pharmaceutical Co. Ltd. Acrylamide derivatives and process for producing the same
DE69637383T2 (en) 1995-05-10 2008-12-11 Kyowa Hakko Kogyo Co., Ltd. CYTOTOXIN CONJUGATE COMPRISING DIPEPTIDE
US5686237A (en) 1995-06-05 1997-11-11 Al-Bayati; Mohammed A. S. Use of biomarkers in saliva to evaluate the toxicity of agents and the function of tissues in both biomedical and environmental applications
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5646298A (en) * 1995-06-07 1997-07-08 Procoron, Inc. Cyclopropylindole prodrugs
CA2233936A1 (en) * 1995-10-03 1997-04-10 The Scripps Research Institute Cbi analogs of cc-1065 and the duocarmycins
EP0774464B1 (en) 1995-10-17 2004-07-28 Combichem, Inc. A template for solution phase synthesis of combinatorial libraries
DE69626849T2 (en) 1995-12-22 2003-12-24 Bristol Myers Squibb Co BRANCHED HYDRAZONE GROUPS OF COUPLERS
US6143901A (en) 1996-07-31 2000-11-07 Genesoft, Inc. Complex formation between dsDNA and pyrrole imidazole polyamides
JP2000506168A (en) * 1996-03-08 2000-05-23 ザ スクリップス リサーチ インスティテュート MCBI analogs of CC-1065 and duocamycin
WO1997045411A1 (en) * 1996-05-31 1997-12-04 The Scripps Research Institute Analogs of cc-1065 and the duocarmycins
US6130237A (en) 1996-09-12 2000-10-10 Cancer Research Campaign Technology Limited Condensed N-aclyindoles as antitumor agents
JPH1087666A (en) 1996-09-18 1998-04-07 Kyorin Pharmaceut Co Ltd Intermediate for producing duocarmycin sa and its derivative and production thereof
US6759509B1 (en) 1996-11-05 2004-07-06 Bristol-Myers Squibb Company Branched peptide linkers
WO1998043663A1 (en) 1997-03-28 1998-10-08 The Scripps Research Institute Sandramycin analogs
WO1998050432A1 (en) 1997-05-07 1998-11-12 Bristol-Myers Squibb Company Recombinant antibody-enzyme fusion proteins
JP2002503228A (en) 1997-05-22 2002-01-29 ザ スクリップス リサーチ インスティテュート Duocarmycin and analogs of CC-1065
WO1999019298A1 (en) 1997-10-14 1999-04-22 The Scripps Research Institute iso-CBI AND iso-CI ANALOGS OF CC-1065 AND THE DUOCARMYCINS
IL136489A0 (en) 1997-12-08 2001-06-14 Scripps Research Inst Synthesis of cc-1065/duocarmycin analogs
JP3045706B1 (en) 1998-09-14 2000-05-29 科学技術振興事業団 Compound for alkylating a specific base sequence of DNA and method for synthesizing the same
US7425541B2 (en) 1998-12-11 2008-09-16 Medarex, Inc. Enzyme-cleavable prodrug compounds
US6909006B1 (en) * 1999-08-27 2005-06-21 Spirogen Limited Cyclopropylindole derivatives
NZ518764A (en) 1999-12-29 2004-02-27 Immunogen Inc Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
US6559125B1 (en) * 2000-01-28 2003-05-06 California Institute Of Technology Polyamide-alkylator conjugates and related products and method
JP2003529609A (en) 2000-03-16 2003-10-07 ジーンソフト インコーポレイティッド Charged compounds containing nucleic acid binding moieties and uses thereof
JP4061819B2 (en) 2000-05-12 2008-03-19 独立行政法人科学技術振興機構 Method for synthesizing interstrand crosslinker
AU2001266853B2 (en) 2000-06-14 2005-02-17 Medarex, Inc. Prodrug compounds with an oligopeptide having an isoleucine residue
AU7552501A (en) 2000-06-14 2002-01-08 Corixa Corp Tripeptide prodrug compounds
EP1243276A1 (en) 2001-03-23 2002-09-25 Franciscus Marinus Hendrikus De Groot Elongated and multiple spacers containing activatible prodrugs
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US7256257B2 (en) 2001-04-30 2007-08-14 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US20030083263A1 (en) 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
WO2002096910A1 (en) 2001-05-31 2002-12-05 Medarex, Inc. Cytotoxins, prodrugs, linkers and stabilizers useful therefor
MXPA03010961A (en) 2001-05-31 2004-02-27 Vertex Pharma Thiazole compounds useful as inhibitors of protein kinases.
WO2002100353A2 (en) 2001-06-11 2002-12-19 Medarex, Inc. Cd10-activated prodrug compounds
EP1423110A4 (en) 2001-09-07 2005-04-27 Scripps Research Inst Cbi analogues of cc-1065 and the duocarmycins
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
US6756397B2 (en) * 2002-04-05 2004-06-29 Immunogen, Inc. Prodrugs of CC-1065 analogs
US6534660B1 (en) 2002-04-05 2003-03-18 Immunogen, Inc. CC-1065 analog synthesis
US7776814B2 (en) 2002-07-09 2010-08-17 R&D-Biopharmaceuticals Gmbh Tubulysin conjugates
AU2003266233A1 (en) 2002-07-09 2004-01-23 Morphochem Aktiengesellschaft Fur Kombinatorische Chemie Novel tubulysin analogues
US20050026987A1 (en) 2003-05-13 2005-02-03 The Scripps Research Institute CBI analogues of the duocarmycins and CC-1065
US7517903B2 (en) * 2004-05-19 2009-04-14 Medarex, Inc. Cytotoxic compounds and conjugates
US7714016B2 (en) * 2005-04-08 2010-05-11 Medarex, Inc. Cytotoxic compounds and conjugates with cleavable substrates

Also Published As

Publication number Publication date
NO20082243L (en) 2008-07-21
AU2006305842B2 (en) 2011-11-03
DK1940789T3 (en) 2012-03-19
EP1940789B1 (en) 2011-11-23
CA2627046A1 (en) 2007-05-03
PL1940789T3 (en) 2012-04-30
CY1112533T1 (en) 2015-12-09
IL190971A0 (en) 2008-12-29
ATE534629T1 (en) 2011-12-15
JP5116686B2 (en) 2013-01-09
HK1118549A1 (en) 2009-02-13
CN101365679A (en) 2009-02-11
EA015324B1 (en) 2011-06-30
ZA200804521B (en) 2009-04-29
US20080281102A1 (en) 2008-11-13
PT1940789E (en) 2012-02-01
IL190971A (en) 2013-02-28
SI1940789T1 (en) 2012-03-30
KR20080068084A (en) 2008-07-22
BRPI0619331A2 (en) 2011-09-27
JP2009513676A (en) 2009-04-02
EA200801176A1 (en) 2009-02-27
AU2006305842A1 (en) 2007-05-03
ES2375843T3 (en) 2012-03-06
WO2007051081A1 (en) 2007-05-03
RS52100B (en) 2012-06-30
CN101365679B (en) 2012-11-14
WO2007051081A8 (en) 2010-12-16
HRP20120058T1 (en) 2012-02-29
US7847105B2 (en) 2010-12-07
EP1940789A1 (en) 2008-07-09
WO2007051081B1 (en) 2007-07-19

Similar Documents

Publication Publication Date Title
CA2627046C (en) Methods and compounds for preparing cc-1065 analogs
EP1948242B1 (en) Cytotoxic compounds
US7714016B2 (en) Cytotoxic compounds and conjugates with cleavable substrates
CA2623652C (en) Antibody-drug conjugates and methods of use
US7517903B2 (en) Cytotoxic compounds and conjugates
CA2678514A1 (en) Chemical linkers with single amino acids and conjugates thereof
IL261759A (en) Process for the preparation of pegylated drug-linkers and intermediates thereof
WO2021098859A1 (en) Aza seven-membered ring inhibitor, and preparation method therefor and use thereof
KR20230035332A (en) Antifolate Linker-Drug and Antibody-Drug Conjugates
KR20220044753A (en) Small molecule inhibitor of acetyl coenzyme A synthetase short chain 2 (ACSS2)
CN107660208B (en) Bifunctional cytotoxic agents comprising a CTI pharmacophore
EP3917933A1 (en) Duocarmycin analogues
AU2006315252A1 (en) Duocarmycin derivatives as novel cytotoxic compounds and conjugates

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20171018