CA2150785C - Conjugates of methyltrithio antitumor agents and intermediates for their synthesis - Google Patents

Conjugates of methyltrithio antitumor agents and intermediates for their synthesis Download PDF

Info

Publication number
CA2150785C
CA2150785C CA2150785A CA2150785A CA2150785C CA 2150785 C CA2150785 C CA 2150785C CA 2150785 A CA2150785 A CA 2150785A CA 2150785 A CA2150785 A CA 2150785A CA 2150785 C CA2150785 C CA 2150785C
Authority
CA
Canada
Prior art keywords
bond
alkyl
nconhr
ncoor
alk2
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 - Lifetime
Application number
CA2150785A
Other languages
French (fr)
Other versions
CA2150785A1 (en
Inventor
Philip Ross Hamann
Lois Hinman
Irwin Hollander
Ryan Holcomb
William Hallett
Hwei-Ru Tsou
Martin J. Weiss
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.)
Wyeth Holdings LLC
Original Assignee
Wyeth Holdings 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22962074&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2150785(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Wyeth Holdings LLC filed Critical Wyeth Holdings LLC
Publication of CA2150785A1 publication Critical patent/CA2150785A1/en
Application granted granted Critical
Publication of CA2150785C publication Critical patent/CA2150785C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • A61K47/6809Antibiotics, e.g. antitumor antibiotics anthracyclins, adriamycin, doxorubicin or daunomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

This invention describes carrier-drug conjugates prepared from disulfide analogs of the calicheamicin family of potent antitumor antibiotics and their derivatives, as well as similar analogs from related antitumor antibiotics such as the esperamicins. The carrier can be an antibody, growth factor, or steroid which targets an undesired population of cells, such as those of a tumor. Whole protein carriers as well as their antigen-recognizing fragments and their chemically or genetically manipulated counterparts are useful for the targeting portion of the conjugates. This invention includes compounds required for the synthesis of these conjugates, appropriate pharmaceutical compositions of the carrier-drug conjugates, and their method of use.

Description

32, 368 CONJ~ ATES OF MRTHYT~TRTT~IO ANTTTllMOR AGFNTS AND
TNTERMBDIATFS FOR TEIEIR x~ SIS
SUMMA~Y OF T~F. TNV~TION
This invention describes carrier-drug conjugates prepared from disulfide analogs of the calicheamicin family of potent antitumor antibiotics and their derivatives, as well as similar analogs from related antitumor antibiotics such as the esperamicins.
The carrier can be an antibody, growth factor, or steroid which targets an undesired population of cells, such as those of a tumor. Whole protein carriers as well as their antigen-recognizing fragments and their chemically or genetically manipulated counterparts are useful for the targeting portion of the conjugates.
This invention includes compounds required for the synthesis of these conjugates, appropriate pharma--ceutical compositions of the carrier-drug conjugates, and their method of use.
More specifically, one aspect of the invention includes a cytotoxic drug conjugate of the formula:

Z3 [ CO-Alkl -spl -Ar-Sp2 -Alk2 -C ( zl ) =z2 ] m wherein z3 is a protein selected from mono- and polyclonal antibodies, their antigen-recognizing fragments, and their chemically or genetically manipulated counterparts and growth factors and their chemically or genetically manipulated counterparts, wherein a covalent bond to the protein is an amide formed from reaction with ~m~ lysine side ch~; n~, or a steroid, wherein the covalent bond to the steroid is an amide or an ester;
m is an integer from about 0.1 to 15;
Alk1 and Alk2 are independently a bond or branched or unbranched (Cl-C10) alkylene chain;
Spl is a bond, -S-, -0-, -CONH-, -NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar~-Y-(CH2)n-Z wherein X, Y, and Z
are independently a bond, -NR'-, -S-, or -0-, with the proviso that when n = 0, then at least one of Y and Z
must be a bond and Ar~ is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR~, with the proviso that when Alk1 is a bond, Spl is also a bond;
n is an integer from 0 to 5;
R' is a branched or unbranched (C1-C5 ) chain optionally substituted by one or two gr~ups of -OH, (Cl-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, (C1-C3) dialkylamino, or (C1-C3) trialkylammonium-A~ where A-is a pharmaceutically acceptable anion completing a salt;
Sp2 is a bond, -S-, or -0-, with the proviso that when Alk2 is a bond, Sp2 is also a bond;
Zl iS H, (C1-C5) alkyl, or phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as hereinbefore defined;
Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, ha-logen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are - - 21507~5 3 f as hereinbefore defined or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or C~s~

each naphthylidene or phenothiazine optionally substituted with one, two, three, or four groups of tCl-C6) alkyl, (Cl-Cs) alkoxy, (Cl-C4) thioalkoxy~
halogen, nitro, or COOR ', CONHR ', O ( CH2 ) nCOOR ', S(CH2)nCOOR', O(CH2)nCONHR~, or S(CH2)nCONHR~ wherein n and R' are as hereinbefore defined, with the proviso that when Ar is naphthylidene, zl is not hydrogen and with the proviso that when Ar in phenothiazine, spl is a bond only connected to nitrogen;
z2 is Q-Sp-SS-W, wherein W is R~S--~'' HN~

R1 1~ ~SSSSSS or CH3; R2b N~G~ or H;
R~O OCH3 R5 OCH3 C H3~ or H; R4 Is C 1~ or H;

215078~ -;

C H90~C0--R6 w R, is H or J~I~N--H

oD~OCH3 R5 is -CH3, -C2H5, or -CH(CH3)2; X is an iodine or bromine atom; Rs' is a hydrogen or the group RCO, wherein R is hydrogen, branched or unbranched (Cl-Clo) alkyl or (Cl-Clo) alkylene group, a (C6-Cll) aryl group, a (C6-Cll) aryl-alkyl (Cl-Cs) group, or a heteroaryl or heteroaryl-alkyl (Cl-C5) group wherein heteroaryl is defined as 2- or 3-furyl, 2- or 3-thienyl, 2- or 3-(N-methylpyrrolyl), 2-, 3-, or 4-pyridinyl, 2-, 4-, or 5-(N-methylimidazolyl), 2-, 4-, or 5-oxazolyl, 2-, 3-, 5-, or 6-pyrimidinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolyl, or 1-, 3-, 4-, 5-, 6-, - 7-, or 8-isoquinolyl, all aryl and heteroaryl groups optionally substituted by one or more hydrox~, amino, carboxy, halo, nitro, lower (Cl-C3) alkoxy, or lower (Cl-Cs) thioalkoxy groups;
Sp is a straight or branched-chain divalent or trivalent (Cl-Cl8) radical, divalent or trivalent aryl or heteroaryl radical, divalent or trivalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent or trivalent aryl- or heteroaryl-alkyl ~Cl-C18) radical, divalent or trivalent cycloalkyl- or heterocyclo-alkyl-alkyl (Cl-Cl8) radical or divalent or trivalent (C2-Cl8) unsaturated alkyl radical, wherein heteroaryl is furyl, thienyl, N-methylpyrrolyl, pyridinyl, N-methylimidazolyl, oxazolyl, pyrimidinyl, quinolyl, isoquinolyl, N-methylcarbazoyl, aminocoumarinyl, or phenazinyl and wherein if Sp is a trivalent radical, it can be additionally substituted by lower (Cl-C5) dialkylamino, lower (Cl-C5) alkoxy, hydroxy, or lower (Cl-C5) alkylthio groups; and Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NO-and includes the conjugates use as antitumor agents.

A second aspect of this invention involves modified drugs, useful as intermediates for constructing conjugates, of the formula:

Z3 [co-Alkl-spl-Ar-sp2-Alk2-c (zl) =Z2]m wherein zl, z2, Alkl, Spl, Ar, Sp2, and Alk2 are as hereinbefore defined;
z3 is halogen, hydroxy, OM wherein M is a metal completing a salt, -N3, ~ ~ SO,N- ~ ~F, O F F F F
02N~

_ ~ NO2 ,or - ~ NO2 ;

and m is 1.

A third aspect of this invention involves linkers, useful for constructing drug conjugates, of the formula:

Z3[CO-Alkl-Spl-Ar-Sp2-Alk2-C(Zl)=Z2]m wherein Z3 is halogen, hydroxy, OM wherein M i6 a metal completing a salt, -N3, ~1~078~
---ON~ ~ --ON~ ~F, 02N~

--~;}NO2 , or--o~}NO2 Alkl and Alk2 are independently a bond or branched or unbranched (Cl-Clo) alkylene chain;
spl is a bond, -S-, -O-, -CONH-, -NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar'-Y-(CH2) n~Z wherein X, Y, and lS Z are independently a bond, -NR'-, -S-, or -O-, with the proviso that when n = 0, then at least one of Y
and Z must be a bond and Arl is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (Cl-C5) alkyl, (Cl-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR', with the proviso that when Alkl is a bond, spl is a bond;
n is an integer from 0 to 5;
R~ is a branched or unbranched (Cl-Cs ) chain optionally substituted by one or two groups of -OH, (Cl-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, (Cl-C3) dialkylamino, or (Cl-C3) trialkylammonium - A- where A- is a pharmaceutically acceptable anion completing a salt;
Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (Cl-C6) alkyl, (Cl-Cs) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2~nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as hereinbefore defined or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or ~ 5 ~

each naphthylidene or phenothiazine optionally substituted with one, two, three, or four groups of (Cl-C6) alkyl, (Cl-C5) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCR', O(cH2)ncoNHR~ or S(CH2)nCONHR' wherein n and R~ are as hereinbefore defined, with the proviso that when Ar is naphthylidene, zl is not hydrogen and with the proviso that when Ar in phenothiazine, spl is a bond only connected to nitrogen;
lS Sp2 is a bond, -S-, or -O-, with the proviso that when Alk2 is a bond, Sp2 is a bond;
zl is H, (C1-Cs) alkyl, or phenyl optionally substituted with one, two, or three groups of (C1-Cs) alkyl, (C1-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R~ are as hereinbefore defined;
z2 is oxygen; and m is 1, with the proviso that when Ar is unsubstituted 2,6-naphthylene or 1,3- or 1,4-phenylene optionally substituted with one group of (C1-C6) alkyl or (C1-Cs) alkoxy and Alk2 is a bond, then spl is not a bond, -O-, or -NHCO-.
D~.~CRIPTTON OF THF. DR~TNGs ~ig. 1: DNA and amino acid sequences for h-P67.6 light chain.
Fig. 2: DNA and amino acid sequences for h-P67.6 heavy chain.
Fig. 3: Plasmid for h-P67.6 heavy chain expression.
Fig. 4: Plasmid for insertion of h-P67.6 heavy chain.

215078~ 8 Fig. 5: Plasmid for h-P67.6 light chain expression.
Fig. I: The proton magnetic resonance spectrum of 4-formylphensxyacetic acid con~n-~ed with Calirhe~m;cin N-acetyl gamma dimethyl hydrazide.
Fig. II: The proton magnetic rPson~nce spectrum of 4-formylbenzoic acid con~pnced with Caliche~m;cin N-acetyl gamma dimethyl hydrazide.
Fig. III: The proton magnetic r~son~nce spectrum of 4-formyl-3-methoxyrhensxyacetic acid con~Pn~ed with Caliche~m;cin N-acetyl gamma dimethyl hydrazide.
Fig. IV: The proton magnetic resonAnce spectrum of 6-formyl-2-naphthoic acid co~en~ed with Calirhe~m;cin N-acetyl gamma dimethyl hydrazide.
Fig. V. The proton magnetic resonance spectrum of 4-(4-acetylrhPnoxy)butanoic acid co~enced with Calicheamicin N-acetyl gamma dimethyl hydrazide.
Fig. VI: The proton magnetic recon~nce spectrum of 4-(4-acetylphenoxy)butanoic acid co~n~ed with CalichP~m;cin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccinimide ester.
Fig. VII: The proton magnetic resonance spectrum of 4-(4-formylph~noxy)butanoic acid co~en~ed with Cali~he~m;cin N-acetyl gamma dimethyl hydrazide.
` Fig. 1 to Fig. 5 are sometimes referred to as Chart 1 to Chart 5 in the text which follows.

- 21~078~ 9 BAcKGRou~n OF THE INVF.NTTON
Since the discovery of methodology for producing monoclonal antibodies was published in the 1970~s (G. Kohler and C. Milstein, "Nature~ 256, 495 (1975)), numerous attempts have been made to use these proteins to achieve selective targeting of antitumor agents to tumors. (E.g., see T. Ghose and A. H. Blair, UCRC Critical Rev. Drug Carrier Systems~ 3, 263 (1987), G. A. Koppel, ~Bioconjugate Chem.~ 1, 13 (1990), and J.
Upeslacis and L. ~;nm~n, "Ann. Rep. Med. Chem." 23, 151 (1988~.) Although progress continues to be made in this field, most classical antitumor agents produce antibody conjugates which are relatively ineffective for a variety of reasons. Among the reasons for this ineffectiveness is the lack of potency of the chemo-therapeutic agent and its poor utilization due to the lack of efficient release of the drug at its site of action.
The potent family of antibacterial and antitumor agents, known collectively as the cali-cheamicins or the LL-E33288 complex, are described and claimed in U.S. Pat. No. 4,970,198 (1990). The most potent of the agents is designated ~lI, which is herein referred to simply as gamma. The dihydro derivatives of these compounds are described in U.S. Pat. No. 5,037,651 (1991) and the N-acylated derivatives are described in U.S. Pat. No . 5,079,233 (1992). Related compounds which are also useful in this invention include the espera-micins which are described and claimed in U.S. Pat. No.
4,675,187 (1987); 4,539,203; 4,555,162; and European Patent 289,030. All of these compounds contain a methyltrisulfide that can be reacted with a~ Liate thiols to form disulfides, at the same time introducing a functional group such as a hydrazide or similar nucleophile. Examples of this reaction with the .

calicheamicins are given in U.S. Pat. 5,053,394 which also discloses targeted forms of the calicheamicins.
All information in the above-mentioned patent citations is incorporated herein by reference. Two compounds which are useful for the synthesis of conjugates with carrier molecules, as disclosed and claimed in U.S. Pat.
No. 5,053,394, are shown in Table l.
Table l CH3 0 CH3 H2NHN S~ ~0 -o~c~N ~CO

oCH3 o H ,,N
Rs oc~
R5~ = H gamma dimethyl hydrazide R5' = Ac N-acetyl gamma dimethyl hydrazide Included as carrier molecules in U.S. Pat. No.
5,053,394 are steroids, growth factors, antibodies, antibody fragments, and their genetically or enzyma-tically engineered counterparts, hereinafter referred to singularly or as a group as carrier. The essential property of the carrier is its ability to recognize an antigen or receptor associated with an undesired cell line. Examples of carriers are given in U.S. Pat. No.
5,053,394, and such carriers are also appropriate in the present invention. Antibody carriers can be from almost any mammalian species (eg. mouse, human, dog, etc.) and can be produced by various methods (eg. murine antibodies via hybridomas, human antibodies via hybridomas from transgenetic mice, etc).
Specific examples of carriers which are exemplified herein are the antibodies P67.6, A33, CT-M-Ol and the Uanti-Tac~ antibody of Waldman. These antibodies are used here in two forms: a murine form, - 215078~

designated by an ~m~ (e.g., m-P67.6), and a genetically engineered, humanized form, designated by an ~h~ (e.g., h-P67.6) whenever appropriate. The basic technology for humanization is disclosed by Winter in US Patent S,225,539 (1993) and by Adair in WO 91/09967 (1991).
m-P67.6 is disclosed in I.D. Bernstein et al., "J. Clin.
Invest." 79, 1153 (1987) and recognizes the CD33 antigen which is prevalent on certain human myeloid tumors, especially acute non-lymphocytic leukemia (ANLL).
Chart 1 and Chart 2 show the DNA coding and predicted amino acid sequences of the variable regions of one particular h-P67.6 that is particularly suitable for use in the present invention. The framework for this antibody is the EU framework for human IgG4 shown in Gottlieb et al., ~Biochemistry: 9, 3115 and 3161 (1970). The antibody was prepared using the general strategy described in WO 91/09967. With reference to the charts, the overlapping oligonucleotides that were synthesized (Oligo Ll through L8) are shown with double underlines, and the PCR assembly procedure (cf. WO
92/01059) was applied to these sequences. The CDR
regions of the protein are designated with single underlines and other amino acids that were taken from the murine sequences are shown with a double underline.
The restriction sites that were used to splice the sequences into plasmids are indicated at the beg;nn;ng and end of the sequences. The variable portion of the heavy chain was cloned into plasmid pMRR14 (WO
93/06231) to give the plasmid designated pAL63 (Chart 3) and the variable portion of the light chain was cloned into plasmid pMRR15 (Chart 4) to give pAL60 (Chart 5).
Plasmids pMRR14 and pMRR15 contained the constant regions of the heavy and light ch~;nc, respect-ively, and therefore pAL63 and pAL60 contained complete sequences for the P67.6 heavy and light ch~;n~. The plasmids were -- 21S07~ 12 cotransfected into CHO-L761 cells to generate a h-P67.6 producing line from which the h-P67.6 was purified by standard methods. The resultant h-P67.6 bound to HL60 cells in competition with murine antibody with about a 50% loss in immunoaffinity. This binding was inhibited by pre-incubation with soluble CD33 antigen.
The antibody m-CT-M-01 is disclosed in E.P. 86 401482.4/0208615 and recognizes the polyepithelial mucin (PEM) antigen present on many human solid tumors, particularly breast, lung, and ovarian. The hllm~n;zed version of this antibody, h-CT-M-01, is described in WO
93/06231 (1993). The antibody m-A33 is disclosed in US
patent application serial numbers 327,765; 673,153; and lS 671,132 and is a murine antibody which recognizes a glycoprotein antigen present on colon cancer cells. The humanized version of this antibody, h-A33, is disclosed in UK Patent Application 9,315,249.4. Anti-Tac is disclosed in T. A. Waldman et al., ~J. Immunol.~ 126, 1393 (1981) and is a murine antibody reactive with the IL-2 receptor that is found on activated and functionally mature T cells, including abnormally activated leukemia cells.
The two basic types of conjugates disclosed in U.S. Pat. No. 5,053,394 are those which are attached to lysine residues of the antibody and those which are attached to the oxidized carbohydrate residues using the method taught in U.S. Pat. No. 4,671,958. Lysine attachment as it is disclosed in U.S. Pat No. 5,053,394 produces conjugates which are stable to hydrolysis under normal physiological conditions. The carbohydrate-based conjugates, which involve the formation of a hydrazone from a hydrazide or similar derivative, are hydrolyti-cally unstable under certain conditions, and tXat is in many cases an advantage. Some instability is often needed to allow release of the drug once the conjugate has been internalized into the target cell, but a _ - 21~07~5 13 `~

certain degree of stability is important to prevent premature release of the drug from the antibody.
However, these carbohydrate-based conjugates suffer from various drawbacks. First, it is necessary to use periodate to generate aldehydes from the carbohydrate residues of the antibody. Antibodies contain cysteines, cystines, methionines, tryptophans, or tyrosines residues which are necessary for proper functioning of the antibody. However, these same amino acids can be sensitive to periodate oxidation, and if such oxidation takes place to an amino acid which either is part of the antigen binding site of the antibody or a structurally important region near the antigen binding site, its immunoaffinity can.be significantly ~;m;n;shed. A
second drawback of using the carbohydrates for conjugation is the variability of the hydrazones and related structures that are generated from the naturally-occurring sugars and the hydrazide derivative.
Not only are the hydrazones subject to different rates of hydrolysis due to differences in their local structure, but other structures, such as hydrated species, piperadines, etc. can also be generated. Any one conjugate may contain structures that are either too stable or to labile for optimum activity.
Limited examples of how to combine some of the properties of the carbohydrate-based conjugates and the lysine-based conjugates have appeared using other less potent classes of anticancer agents. Cullinan in U.S.
Pat. No. 5,006,652 and 5,094,849 teaches that certain bifunctional compounds cont~;n;ng both carboxylic acid and aldehyde or keto functionality can be used as spacers between the lysines of antibodies and hydrazide derivatives of the Vinca alkaloids, while Johnson in U.S. Pat. No. 5,028,697 and 5,144,012 teaches similar art for methotrexate analogs. Sinam et al. also disclose similar constructs in WO Pat. No. 90/03401. In none of these cases is it demonstrated that this method is useful for preparing conjugates of the methyltri-sulfide antitumor antibiotics, especially the cali-che~m;cins or esperamicins. The cited patents do not demonstrate that these constructs made with either the Vinca alkaloids, the methotrexate analogs, or other agents are superior in their biological profile to conjugates made using lysine-based or carbohydrate-based conjugates.
The present invention describes a series of conjugates prepared from the potent methyltrisulfide antitumor antibiotics made with an improved linker system that gives conjugates which in many cases are vastly superior biologically to conjugates of the same drugs made by other methods.
DT~'.TATT.T~.T) DT;'.C:C~TPTION OF TI~. T~ NTToN
The conjugates of this invention use linkers that can be added to a derivative of a drug, parti-cularly hydrazides and related nucleophiles, prepared from the methyltrisulfide containing antitumor anti-biotics. The linkers require a carbonyl group on one end for formation of a Schiff's base, particularly a hydrazone, and a carboxylic acid on the other end. The carboxylic acid can be activated and subsequently reacted with the lysines of an antibody or other targeting protein or with an amine, alcohol, or other appropriate nucleophile on other targeting agents which have been chosen for their ability to target undesired cell populations. These constructs, which for anti-bodies contain elements of both the lysine-based conjugates and the carbohydrate-based conjugates, not only overcome the disadvantages of previously disclosed constructs, but have the additional advantage that they can be fine-tuned by varying the structure of the linker to ~design inn the optimum amount of hydrolytic stability/instability. This can result in maximum toxicity to the target cells with m;n;~-l toxicity to the non-target cells. The optimum hydrazone stability/instability is not necessarily the same for each drug and targeting agent combination.
The method of constructing the conjugates described in this patent produces conjugates of the methyltrisulfide antitumor antibiotics which are unexpectedly stable relative to the carbohydrate based 10 - conjugates without loss of activity. In some cases, the conjugates are 100 times more potent than the corres-ponding conjugates made by the carbohydrate-based method and, in addition, show reduced cytotoxicity against non-target cell lines. ThiS results in conjugates with up to 10,000-fold selectivity between target and non-target cell lines.
The linkers required for the construction of these conjugates can be represented by the following formula:

Z3[CO-Alkl-Spl-Ar-Sp2-Alk2-C(Zl)=Z2]m Alkl and Alk2 are independently a bond or branched or unbranched ~Cl-C10) alkylene chain. spl is a bond, -S-, -O-, -CONH-,-NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar'-Y-(CH2) n~Z wherein n is an integer from 0 to 5, X, Y, and Z are independently a bond, -NR'-, -S-, or -O-, and Ar' is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (Cl-C5) alkyl, (Cl-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n is as hereinbefore defined, with the proviso that when Alkl is a bond, spl is also a bond. R' is a branched ~r 3S unbranched (Cl-C5 ) chain optionally substituted by one or two groups of -OH, (Cl-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, (Cl-C3) dialkylamino, or (Cl-C3) -- 21507~5 16 -trialkylammonium - A- where A- is a pharmaceutically acceptable anion completing a salt. Sp2 is a bond, -S-, or -0-, with the proviso that when Alk2 is a bond, Sp2 is also a bond. Z3 is a hydroxyl group, and m is 1.
The groups Alkl, Spl, Alk2 and Sp2 in combination, as well as the group Ar discussed below, allow for spacing of the carbonyl group from the carboxylic acid. Furthermore, Alkl and spl can influence the reactivity of the carboxyl group both during and after it has been activated. When Alk2 and Sp2 together are a bond, the spl group also influences the reactivity of the carbonyl group on the other end of the linker and the stability of the product formed from reactions at that carbonyl. The group R' can be used to influence the solubility and other physiochemical properties of these compounds. A preferred embodiment for Alkl is (C2-Cs) alkyl, and for spl is an oxygen atom.
A preferred embodiment for the groups Alk2 and Sp2 - 20 together is a bond.
With reference to the structure shown above, the group z2 is an oxygen atom. The group zl is H, (Cl-C5) alkyl, or phenyl optionally substituted with one, two, or three groups of (Cl-C5) alkyl, (Cl-C4) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, COOR ', CONHR ', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R ' are as hereinbefore defined. The group zl has a pronounced effect on the reactivity of the carbonyl group and on the stability of the products formed from reactions at the carbonyl.
When zl is aryl and the product is, for example, a hydrazone, the hydrazone is relatively stable; when zl is hydrogen, then an intermediate level of stability is obtained, and when zl is (Cl-C6) alkyl, relatively less stable hydrazones are formed. As stated earlier, stability is important to prevent premature release of the drug from the antibody, but some instability is ~1~0785 17 needed to allow release of the drug once the conjugate has been internalized into target cells. A preferred embodiment for the zl group is (Cl to C3).
The group Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (Cl-C6) alkyl, (Cl-C5) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCR~, O(CH2)nCONHRI~ or S(CH2)nCONHR' wherein n and R' are as hereinbefore defined or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or ~5~
I , -wherein spl a bond only connected to the nitrogen of the phenothiazine, each optionally substituted with one, two, three, or four groups of (Cl-C6) alkyl, (Cl-C5) alkoxy, (Cl-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as hereinbefore defined;
The choice of Ar has a significant influence on the stability of the products derived from the carbonyl when Alk2 and Sp2 are together a bond. Both the relative position of spl and Sp2 as well as the presence of additional substituents on Ar can be used to fine-tune the hydrolytic behavior of the product formed from the carbonyl. A preferred embodiment for Ar is 1,2-, 1,3-, or 1,4-phenylene, or 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene.
The structures of specific examples of linkers which are useful in the present invention are as follows:

215078~ 18 OHC~ OHC~ OHC~

~o ~ CH3OJ~O 1 OH
OHC~C~, OHC~O~C~,OHC~O~c~OH

C 1~ HOH~ `OH

OHC~3~ CH3J~

OHC~ , OHC~3 ~OH

O O

CH3 ~ HOOC ~ ~ CH3 HOOC - O HO C~O

CHa~H3 ~1 HOOC - O HOOC - O

lS ~ CHO

HOOC~-~-~^~O ~ OCH3 HOOC - O

HOOC ~ O~"~_,COOH

19 ao COOH HOOC

HOOC~O~CH:i C.~--H~COOH

~ ~! 4 Cl COOMe OHC~O~'COOH ~O--COOH

~COOH
NO2 CH3~ ~q~
O~----COOH H~OMe ~ 28 o rE~r ~0~
CH3~0----COOH

F ~_CH3 3 0 CH3~ COOH ~O~ COOH

2 1 5 0 7 ~ S 2 1 CH3~XN~3 O~{~ COOH

NMe2 ~NMe3~

CH3>{~0--COOH ~O--COO-CH3 ~ 0 ~ NMe Only a few of the more simple of these linkers are commercially available, i.e., linkers 1, ~, 3, 19, ~3, 24, and 33. Linker ~Q is listed by the Chemical Abstract Services with registry number 5084-45-7. Many linkers which contain aryl ethers as a part of their structure, such as 1, 8, lQ, 1~ , 16, 1~, ~Q, 21, , 30, and ~1, can be made by alkylating a phenolic ketone with an electrophile, such as ethyl 4-bromo-butyrate, using an appropriate base, such as potassium carbonate, in an appropriate solvent, such as N,N-dimethyl formamide, and then converting the ester into the required carboxylic acid by hydrolysis with, for example, sodium hydroxide or potassium carbonate in agueous methanol. This strategy can also be used with linkers such as 5, 6, 9, 11, 1~, or ~7, where the - 215078~ 22 carbonyl is carried through the initial steps of the preparation in a masked form, such as an olefin or an alcohol. The carbonyl can then be generated later, as described in the examples, by oxidation with ozone or pyridinium chlorochromate, resp. This procedure is especially valuable when a more reactive carbonyl is present in the final linker.
When necessary, the required carboxylic acid can be introduced in a masked form as in the preparation of linker 26. In this case the phenol is alkylated with 5-bromo-1-pentene and the acid is liberated from the olefin by reaction with ozone followed by pyridinium chlorochromate oxidation. Linkers such as 22 or 32 can be made by alkylating an appropriate secondary amine (a piperazine or phenothiazine derivative, resp.) with an appropriate electrophile and then exposing the required carboxylic acid in a later step, similar to the previously mentioned strategies. Linker 1~ was made by reduction of the corresponding cinnamate with hydrogen.
Although this reaction gave a relatively impure product, the crude mixture was useful for conversion to the required hydrazone because none of the by-products contained aldehyde groups. Structures with more elaborate substituents, such as linkers 33, 34, 35, or 36, can be made from simpler structures by, for example, reacting an ester with an appropriate nucleophile or by quaternizing an amine with an electrophile, such as methyl iodide.
The linkers defined above can be used to form conjugates as follows:

-- 2 1 5 0 7 8 5 23 ?

HOCO-Alkl-Spl-Ar-Sp2-Alk2-C(Zl)=O (Structure A) Q-Sp-SS-W

Z3-CO-Alkl-Spl-Ar-Sp2-Alk2-C(Zl)=Z2 (Str. B, Z3 = OH
and Str.C, Z3 = e.g. OSu) Carrier Z3[co-Alkl-spl-Ar-sp2-Alk2-c(zl)=z2]m (Structure D, Z3 = Carrier) Scheme With reference to Scheme 1 above, the linker of structure A, wherein zl, Alkl, Spl, Ar, Sp2, and Alk2 are as hereinbefore defined, is condensed with a compound of structure W-SS-Sp-Q, which itself is derived from a methyltrithio antitumor antibiotic, and wherein W is R1S~-- HN~

C
H3 o R~ Is X~5sss~ or CH3; R21s N~ or H;
R40 OCH3 Rs OCH3 215078~ 24 C~ orH; R~l- ~OH ~rH;

C H30~C O--R6 or R, is H or C H30~N~ H
oD~oCH3 Rs is -CH3, -C2Hs, or -CH(CH3)2; X is an iodine or bromine atom; Rs' is a hydrogen or the group RC0, wherein R is hydrogen, branched or unbranched (Cl-Clo) alkyl or (C1-C1o) alkylene group, a (C6-Cl1) aryl group, a (C6-C11) aryl-alkyl (C1-Cs) group, or a heteroaryl or heteroaryl-alkyl (C1-Cs) group wherein heteroaryl is defined as 2- or 3-furyl, 2- or 3-thienyl, 2- or 3-(N-methylpyrrolyl), 2-, 3-, or 4-pyridinyl, 2-, 4-, or 5-(N-methylimidazolyl), 2-, 4-, or S-oxazolyl, 2-, 3-, S-, or 6-pyrimidinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolyl, or 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolyl, all aryl and heteroaryl optionally substituted by one or more hydroxy, amino, carboxy, halo, nitro, lower (Cl-C3) alkoxy, or lower (C1-Cs) thioalkoxy groups; Sp is a straight or branched-chain divalent or trivalent 0 (C1-C18) radical, divalent or trivalent aryl or heteroaryl radical, divalent or trivalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent or trivalent aryl- or heteroaryl-alkyl (C1-C18) radical, divalent or trivalent cycloalkyl- or heterocycl-oalkyl-alkyl (C1-Cl8) radical or divalent or trivalent (C2-C18) unsaturated alkyl radical, wherein heteroaryl is furyl, thienyl, N-methylpyrrolyl, pyridinyl, N-methylimi-dazolyl, oxazolyl, pyrimidinyl, quinolyl, isoquinolyl, N-methylcarbazoyl, aminocoumarinyl, or phenazinyl and wherein if Sp is a trivalent radical, it can be additionally substituted by lower (Cl-C5) dialkylamino, lower (Cl-C5) alkoxy, hydroxy, or lower (Cl-C5) alkylthio groups; and Q is H2NHNCO-, H2NHNCS-, H2NHNCONH-, H2NHNCSNH-, or H2NO-, to produce a compound of structure B, wherein zl, Alkl, Spl, Ar, Sp2, and Alk2 are as hereinbefore defined, z2 is Q-Sp-SS-W, wherein Sp and W
are as herein above defined, Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NO-, and Z3 is -OH.
The condensation can be run in most compatible organic solvents, but is particularly efficient in alcoholic solvents such as methanol or ethanol. This condensation reaction is acid catalyzed. The carboxylic acid in the linkers themselves is sufficient in many cases to catalyze this reaction, but adding a compatible acid catalyst, such as about 5% acetic acid, helps improve the rate of reaction in many cases. The temperature of this reaction can be from about ambient temperature to the reflux temperature of the solvent.
The products are isolated in pure form by removing the volatile solvents and purifying the mixture by chromatography on a suitable medium such as BioSil A, a modified silica gel available from Bio-Rad. It should be understood that the products of structure s, as well as the products from the further transformation of these compounds, exist as easily-interconverted syn and anti isomers at the locus defined as Q, and that these products can exist in different hydrated forms, dep~n~i ng on the exact conditions of solvent and the pH
at which these compounds are examined. Such differing physical forms are also included within the scope of this patent.
The carboxylic acid of structure B (Z3 = -OH) is next converted to an activated ester in preparation for ~ 2 1 5 0 7 8 5 26 conjugation of these intermediates with carrier molecules. Such transformations convert Z3 (structure B) to halogen, -N3, -ON ~ ~ -ON ~ ~F, O F F F F

O N2 , or o~N2 For example, reaction of the carboxyl form of structure B (Z3 = -OH) with a coupling agent, such as 1,3-dicyclohexylcarbodiimide or 1-(3--dimethylamino~ro~l)-3-ethylcarbodiimide hydrochloride, and N-hydroxysuccinimide or other comparable carboxyl-activating group in an inert solvent, such as N,N-dimethylformamide, tetrahydrofuran, or acetonitrile, leads to the formation of an activated ester, such as the N-hydroxysuccinimide ester described herein. These active esters can be isolated in pure form by removal of the volatile solvents and chromatography on an appropriate medium, such as BioSil A. Alternately, the coupling reaction can be quenched with a polymeric carboxylic acid, filtered, and stripped of organic solvents, and the crude product can be used in the following step without further purification. This is especially useful if the active ester is difficult to handle, such as when ,~ ,SO3Na z3 = O~J
O . .

- 215078~ 27 The final step in the construction of the conjugates of this patent involves the reaction of an activated ester (structure C) with a targeting molecule, as shown in Scheme 1. This produces a compound of structure D, wherein zl, z2, Alk1, Spl, Ar, Sp2, and Alk2 are as hereinbefore defined, m is 0.1 to 15, and Z3 is a protein such as a growth factor or a mono- or polyclonal antibody, their antigen-recognizing fragments, or their chemically or genetically manipulated counterparts or a steroid, wherein a covalent bond to a protein is an amide formed from reaction with Um~ lysine side ch~;n~
and the covalent bond to a steroid is an amide or an ester.
This conjugation reaction can be carried out in various ap~o~iate buffers, such as borate, phosphate, or HEPES at slightly basic pH (pH -7.4 to 8.5). The final construct can then be purified by appropriate methods, such as gel-exclusion chromato-graphy, to remove unattached drug and aggregates to yield ~ono~eric conjugates. This sequence of steps constitutes Method A as described in greater detail in the Examples section of this patent.
Alternative methods for constructing the conjugates of Scheme 1 are also contemplated as shown in Scheme 2.

Z3-CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=O (Str. A, Z3 = OH
Str. E , Z3 = e.g. OSu) Carrier Z3-CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=O (Str. F, Z3 = Carrier) Q-Sp-SS-W

Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m (Structure D) Scheme 2 For example, the linker (structure A as defined above) can be converted to an active ester and reacted with the targeting molecule prior to the reaction with the drug. Such manipulations convert structure A into structure E, wherein zl, Alk1, Spl, Ar, Sp2, and Alk2 are as hereinbefore defined, z2 is an oxygen atom, and Z3 is halogen, -N3, ZS ~ ~ SO,N~ ~ ~F, O F F F F

O ~ NO2 ,or O ~ NO2 ;

The activated ester is then reacted with the carrier to produce structure F, wherein zl, Alk1, Spl, Ar, sp2, and Alk2 are as hereinbefore defined, z2 is an ~ 29 oxygen atom, m is about 1 to about 20, and Z3 is a protein selected from mono- and polyclonal antibodies, their antigen-recognizing fragments, and their chemically or genetically manipulated counterparts and growth factors and their chemically or genetically manipulated counterparts, wherein a covalent bond to the protein is an amide formed from reaction with ~m" lysine side ch~;n~, or a steroid, wherein the covalent bond to the steroid is an amide or an ester;.
Once the targeting molecule has been modified with the linker, it can be reacted with a compound of structure Q-Sp-SS-W, which itself is derived from a methyltrithio antitumor antibiotic, and wherein w and Sp are as hereinbefore defined, and Q is H2NHNCO-, H2NHNCS-, H2NHNCONH-, H2NHNCSNH-, or H2NO- to produce a compound of Structure D ( vida supra) .
This sequence of steps in Scheme 2 constitutes Method B in the Examples section of this patent.
Similar antibody-carbonyl constructs are covered in U.S.
Pat. No. 5,144,012 mentioned above. Most of the linkers exemplified herein are new and offer the advantage that a much broader range of structural types and hence a broader range of stabilities is demonstrated. As a specific example, the acetophenone linkers, which are new to this patent, produced conjugates with better hydrolytic release properties of drug and which are more potent when used with the examples of the antibodies shown here. Specifically, the two conjugates prepared from h-P67.6 using 4-formylbenzenepropanoic acid or 4-acetylbenzenebutanoic acid condensed with calicheamicin N-acetyl gamma hydrazide (the two conjugates only differ by having zl = -H and zl = -CH3, respectively, in structure 3 of Figure 1) gave in vitro ICso's of 1.0 and 0.012 ng/mL, and specificity indices of 950 and 26,000, resp. Although the acetophenone based linkers are seen to be superior in this case, it is not necessarily easy - 21507~

to predict which linker will be superior for any given targeting agent-drug construct.

BTnT~oGTc~T CH~ACTFRT7~TTON
Assessment of the biological properties of the conjugates included measuring their ability to recognize the antigen on target cell lines, relative to the unmodified antibody, and determining their selectivity and cytotoxic potentials, using the following methods:
TM~NOAFFT~TTY A~SAYS
Relative immunoaffinities of conjugates are determined in a competitive binding assay in which varying concentrations of test conjugate are allowed to compete with a fixed amount of the same antibody labeled with l25I-Bolton Hunter reagent for binding to a fixed number of cells. For m- or h-P67.6, HEL 92.1.7 human erythroleukemia cells [ATCC (American Type Culture Collection) TIB 180] are used at a concentration of 107 cells/mL; for CT-M-01, cell line A2780DDP (E.M. Newman, et al., ~Biochem. Pharmacol." 37, 443 (1988)) is used;
and for m- or h-A33, cell line COLO 205 (ATCC CCL 222) is used. The concentration of test conjugate required to obtain 5096 inhibition of b;n~;ng of the labeled antibody to target cells is compared with the concentration of a reference preparation of native antibody required for 50% inhibition.
Samples for assay are adjusted to ~300 ~g protein/mL in medium and six serial four-fold dilutions of each are prepared in medium (RPMI-1640 cont~;n;ng 5%
heat-inactivated fetal calf serum), for a total of seven concentrations of each sample. The reference antibody is diluted in the same way. An aliquot of 0.05 mL of each dilution is transferred to a 12 X 75 mm plastic tube, and 0.05 mL of labeled reference antibody at 4 llg/mL is added. The tubes are mixed and chilled at 4C.
Then 0.1 mL of chilled cell suspension is added to each tube. All tubes are mixed again, and incubated for l hr at 4C
Controls to determine maximal binding and non-specific binding are included in each assay. Maximal binding is determined by mixing 0.05 mL of medium, 0.05 mL of l25I-antibody, and O.l mL of cells; non-specific binding is determined by mixing 0.05 mL of 500 ~g/mL of native antibody, 0.05 mL of iodinated antibody, and O.l mL of cells.
At the end of the incubation, cells are washed twice, by centrifugation and resuspension, with 3 mL of cold PBS each time. The cells are resuspended in 0.5 mL
of PBS, transferred to clean tubes, and radioactivity is determined in a gamma-counter.
The percent inhibition of binding is calcu-lated by the following equation:

~I = {[(cp~ -~hinding ~ CPmnon-specific) ~
(Cpmsample - cpmnon-specific)] +
(CPm~l~ax binding ~ CPmnon-specific) }X 100 The percent inhibition values are plotted against sample concentrations, and from the resulting curves the sample concentration that gives 50~ inhibition of binding (IC50) is interpolated. The relative immunoaffinity of each tested conjugate is then determined as follows:

Relative Immunoaffinity =
ICso(reference) + ICso(sample) IN VITRO CYTOTOXICITY ASSAY
Cytotoxic activities are determined in an in vitro pulse assay in which varying concentrations of test conjugate are incubated with antigen-positive and antigen-negative cells for l hr, then cultured for three days. Viability is assessed by [3H]thymidine incorpor-ation during the final 24 hr of culture. As a measure of potency, the concentration of test conjugate required to inhibit [3H]thymidine incorporation by 50% (ICso) is determined from the titration curve. The specificity is determined by comparing ICso values on antigen-positive and antigen-negative cells for P67.6, A33, and m-CT-M-01 or by use of a conjugate of the same drug with the non-targeting antibody P67.6 for h-CT-M-01 conjugates or MOPC-21 for anti-Tac conjugates. MOPC-21 (F. Melchers, "Biochem. J u 119, 765 (1970)) is an antibody which does not recognize any normally occurring, physiologically pertinent antigen.
For P67.6, antigen-positive H~-60 human promyelocytic leukemia cells (ATCC CCL 240) and antigen-negative Raji human Burkitt lymphoma cells (ATCC CCL 86) are used; for A33, antigen-positive COLO 205 cells and antigen-negative Raji cells are used; and for h-cT-M
ZR-75-1 cells (ATCC CRL1500) are used. For m-CT-M-01 antigen-positive A2780DDP cells and antigen-negative Raji cells are used, and for h-CT-M-01, ZR-75-1 cells (ATCC CRL1500) are used. Cells are collected by centrifugation, counted, and resuspended in fresh medium (RPMI-1640 + 5% heat-inactivated fetal calf serum +
antibiotics) at a cell concentration of ~106/mL.
Samples for assay are readjusted to -1 ~g/mL
of drug equivalents in medium and five serial ten-fold dilutions of each are prepared in medium, for a total of six concentrations of each sample. In addition, a medium control is included with each sample set, as well as calicheamicin N-acetyl gamma as a drug control. An aliquot of 0.1 mL of cell suspension is added to 17 X
100 mm plastic tubes containing 0.1 mL of sample; a separate series of tubes is prepared for each cell line.
The tubes are loosely capped and incubated for 1 hr at 37C in a humidified atmosphere of 5% CO2 in air. At the end of the incubation, cells are washed twice by centrifugation and resuspended with 8 mL of medium each time. Cell pellets are resuspended in 1 mL of medium and plated in triplicate in 96-well microtiter plates at 0.2 mL/well. The plates are incubated for 2 days at 37C as above. Then 0.1 mL of medium is removed from each well and replaced with 0.1 mL of fresh medium containing 0.1 ~Ci of [3H]thymidine. Plates are returned to the incubator for one more day. Plates are frozen and thawed, and cells are harvested on glass fiber filter mats. The amount of [3H]thymidine incorporated is determined by liquid scintillation counting.
The measured cpm of the triplicate cultures of each sample dilution are averaged and the percent inhibition of [3H]thymidine incorporation is calculated by the following equation, where the values for no inhibition and maximal inhibition come from the medium controls, and the highest concentration of calicheamicin N-acetyl gamma, respectively:

%I = {[(CPmno inhibition ~ CPmmax inhibition) ~
(Cpmsample ~ Cpmmax inhibition)]
(CPmno inhibition ~ Cp~ Y inhibition)} X 100 The percent inhibition values are plotted against sample concentrations, and from the resulting curves the sample concentration that gives 50% inhi-bition of [3H]thymidine incorporation (ICso) is inter-polated. For P67.6, A33, and m-CT-M-01 conjugates, the specificity of a particular conjugate for antigen-positive cells is calculated by taking the ratio of the IC50 against non-target cells to the ICso against target - cells. The same ratio is calculated for the f~ee drug.
Then, to correct for inherent differences in the sensitivities of the two cell lines to the drug, the 21~0785 34 Specificity Index for each sample is calculated as follows:

Specificity Index =
[IC50 (sample on antigen neg) + IC50 (sample on antigen pos)]:
[IC50 (drug on antigen neg) ~ IC50 (drug on antigen pos)]

For conjugates of Anti-Tac or h-CT-M-01, the Specificity Index is calculated as the ratio of ICs0's for the non-targeting conjugate and the targeting conjugate as follows:

Specificity Index =
IC50 (non-targeting conjugate) + IC50 ttargeting conjugate) T~ VIVO ANTTTUMOR A~SAY
Human tumors (either ~107-108 cells or 5 to 8-2 mm3 fragments of solid tumors) are implanted subcu-taneously into athymic mice (nude mice) and test samples are inoculated intraperitoneally (ip) at several dose levels on a q 4 day x 3 schedule, starting 2-3 days after tumor implantation with 5 mice per test group and 10 in the saline control group. Tumor mass is estimated by measuring the tumor length and width once weekly up to 42 days post tumor implantation with a Fowler ultra CAL II electronic caliper and using the formula: mg tumor = {Length(mm) x Width(mm)}/2. Tumor growth inhibition is calculated as the ratio of the mean tumor mass of treated ~n; m~l S compared with untreated controls and is expressed as ~ T/CU. (0% T/C implies no detectable tumor. All control ~nim~l S routinely develop easily measurable tumor.) ~X VIVO lNHIsITToN OF COTONY FO~M~TTON
For P67.6 conjugates, human leukemic bo~e marrow cells which are CD-33 positive are plated in the presence of 2 ng/mL drug equivalents. The number of colonies which form are counted and reported as the ~ 35 215078~

percent versus a control which consists of a h-CT-M-Ol conjugate which does not recognize the CD-33 antigen.
All the data reported were generated with bone marrow from one patient whose leukemic cells had good antigen expression and good response to this general type of treatment.
For anti-Tac, peripheral blood from CML patients was tested. Progenitor cells for cells of the various hematopoietic lineages can be detected by culturing bone marrow cells and blood cells in a semisolid matrix such as methylcellulose and observing the formation of colonies cont~;n;ng mature differentiated cells. There are progenitor cells that proliferate to form colonies of granulocytes or macrophages, or both, called colony-forming units for granulocytes-macrophages (CFU-GM).
Some CFU-GM form colonies within seven days (D7 CFU-GM);
some require fourteen days for colony formation (D14 CFU-GM) [N. Jacobsen, et al., uslooda 52: 221, (1978), and Ferrero D et al.HProc. Natl. Acad. Sci. USA~ 80:
4114, (1983)]. Inhibition of the growth of D14 CFU-GM on blood cells treated with anti-Tac was compared to those treated with non-targeting MOPC 21 conjugates. The number of D14 CFU-GM colonies are plotted against sample concentrations, and from the resulting curves the sample concentration that gives 50% inhibition of D14 CFU-GM
colony growth is interpolated. Specificity was measured by the ratio of the IC50 of the non-targeting conjugate versus the ICso Of the targeting conjugate.
Normal blood does not produce CFU-GM colonies and normal bone marrow D14 CFU-GM colonies are not inhibited by anti-Tac conjugates.
The invention is further described with the following non-limiting preparations and examples.
(Preparations describe the syntheses of compounds useful in this invention but for which-there is known prior - 215078~ 36 art. Examples describe the syntheses of compounds which are useful and new to this invention.) SYNT~ TS OF ~'l'K~C"l'U~F..~ A (Sch~m~ 1 and Scheme 2) F.XP.MPT.F. 1, COMPOUND 5 4-(2-Oxoethoxv~henz~ne~ro~anoic acid 4-Hydroxybenzenepropanoic acid (500 mg, 3.01 mmol) is allowed to react with 910 mg (7.52 mmol) of allyl bromide by the same procedure described in Example 2 to give 610 mg (82~) of 2-propenyl-4-(2-propenyloxy)-benzenepropanoic ester as a colorless oil. The product is utilized in the next reaction without further purifi-cation. The lH NMR (300 MHz, CDC13) iS consistent with the desired product; IR (neat) 3450, 1740, 1650, 1610, 1510 cm-l; MS (CI low res) m/e 247 (M+H), 229, 215, 187, 175; Analysis calc'd for Cl5Hl8O3: ~, 73-15; H~ 7-37;
found: C, 73.09; H, 6.74.
2-Propenyl-4-(2-propenyloxy)benzenepropanoic ester (271 mg, 1.1 mmol) is treated with 0.14 mL (1.38 mmol) of 10 M sodium hydroxide solution according to the same procedure described for Example 2 to give 196 mg (86%) of 4-(2-propenyloxy)benzenepropanoic acid as a white powder. The product is utilized in the next reaction without further purification: m.p. 88-89; the lH NMR (300 MHz, CDC13) iS consistent with the desired product; IR (KBr) 3200, 1720, 1700, 1610 cm-l; MS (CI
low res) m/e 207 (M+H), 189, 175, 147; Analysis calc'd for C12H1403: C, 69.89; H, 6.84; found: C, 69.87; H, 6.68.
4-(2-Propenyloxy)benzenepropanoic acid (120 mg, 0.58 mmol) is treated with ozone by the procedure described in Example 2 to give 100 mg (82%) of 4-(2-oxoethoxy)benzenepropanoic acid as a white powder: m.p.
95-100; the lH NMR (300 MHZ, CDC13) is consistent with the desired product; IR (KBr) 3400, 1740, 1720, 1610 cm-l; MS (CI low res) m/e 207, 191, 179, 165, 149.

- 21~078S 37 F.~MPT .F. 2, cnMPou~n 6 3-(2-Oxoethoxv)h~n~oic ~cid A mixture of 1.0 g (7.24 mmol) of 3-hydroxy-benzoic acid, 3.0 g (25.3 mmol) of allyl bromide, and 5 g (36.2 mmol) of potassium carbonate in 4 mL of N,N-dimethylformamide is stirred at room temperature for 12 hr. The mixture is diluted with 20 mL of ether and washed five times with 20 mL of water. The organic layer is then washed successively with 20 mL of saturated sodium bicarbonate solution and 20 mL of saturated sodium chloride solution. The organic layer is separated and dried over magnesium sulfate. The mixture is filtered and the organic solution is concentrated in vacuo to give 1.4 g (88~) of 3-(2-propenyloxy)benzoic acid, 2-propenyl ester as a clear colorless oil. The product is utilized in the next reaction without further purification. The lH NMR (300 MHz, CDC13 ) is consistent with the desired product; IR
(KBr) 1720, 1650, 1600 cm-l; MS (CI low res) m/e 219 (M+H), 203, 175, 161; Analysis calc~d for Cl3Hl4O3: C, 71.54; H, 6.47; found: C, 70.31; H, 5.97.
A solution of 917 mg (4.2 mmol) of 3-(2-propenyloxy)benzoic acid, 2-propenyl ester in 9 mL of methanol/water (3:2) at room temperature is treated with 0.53 mL (5.25 mmol) of 10 M sodium hydroxide solution.
The solution is allowed to stir for one hr then acidified with 5 mL of 10% sodium bisulfate solution and extracted with 25 mL of ethyl acetate. The organic layer is washed with saturated sodium chloride solution, dried over magnesium sulfate, and concentrated in vacuo to give 732 mg (97%) of 3-(2-propenyloxy)benzoic acid as a white powder. The product is utilized in the next reaction without further purification; m.p.-78-79;
the lH NMR (300 MHz, CDC13) iS consistent with the desired product; IR (KBr) 3000, 1690, 1620, 1590 cm-l;
MS (CI low res) m/e 179 (M+H), 161, 135; Analysis calc'd _ 38 for CloHl0O3: C, 67.41; H, 5.66; found: C, 67.37; H, 5.59.
A solution of 300 mg (1.68 mmol) of 3-(2-propenyloxy)benzoic acid in 5 mL of methylene chloride is cooled to -78 C. Ozone is introduced by bubbling the gas into the solution through a glass tube until a blue color persists. The solution is then purged with a stream of argon and 1 mL of methyl sulfide is added.
The solution is diluted with 20 mL of ether and washed with water. The organic layer is separated and allowed to stand over magnesium sulfate then concentrated in vacuo to give 283 mg (93%) of 3-(2-oxoethoxy)benzoic acid as a colorless oil. The product is utilized in the next reaction withqut further purification: m.p. 120-130; the lH NMR (300 MHz, CDC13) is consistent with the desired product; IR (KBr) 3400, 3000, 1680, 1590 cm-l;
MS (CI low res) m/e 181 (M+H), 163, 139, 119.
PR~P~ATTON 3. CO~POUND 7 4-(4-Acetvlph~n~yy)hut~noic ac;~ -A solution of 0.90 g (6.61 mmol) of 4'-hydroxyacetophenone, and 1.93 g (9.92 mmol) of ethyl 4-bromobutyrate in 1.80 mL of N,N-dimethylformamide is stirred for 48 hr, under dry conditions with 2.74 g (19.8 mmol) of potassium carbonate and 0.110 g (0.66 mmol) of potassium iodide. The reaction mixture is then evaporated under vacuum, and the residue partitioned between ether and water. The organic phase is separated, washed thrice with water, dried with magnesium sulfate, filtered, and evaporated under vacuum to give a brown solid. This is recrystallized from a warm ether-hexane mixture. The beige crystals are air dried, leaving 0.84 g (51%) of 4-(4-acetylphenoxy)-butanoic acid, ethyl ester: m.p. 59-61 C; IR ~(KBr) 1740, 1670 cm-l; lH-NMR (CDC13) is consistent with the desired product; MS (FAs) m/e 251.1285 ~ = -0.2 mm~ (M+

21~07~5 39 + H). Anal. calc~d. for Cl4Hl8O4: C, 67.18; H, 7.25; o, 25.57. Found: C, 67.16; H, 7.16; O, 25.68.
A sample of 0.25 g (1.00 mmol) of 4-(4-acetylphenoxy)butanoic acid, ethyl ester (example 1) is dissolved in 15 mL of methanol/water (3:2), with stirring. Then, 0.21 g (1.50 mmol) of potassium carbonate is added and the reaction is stirred for 18 hr under an argon atmosphere. Next, the reaction mixture is evaporated under vacuum and the residue dissolved in 20 mL of a 0.1 N solution of sodium hydroxide. This basic solution is washed with ether, the aqueous phase acidified by addition of sodium bisulfate, and the resulting mixture extracted with ethyl acetate. This solution is then dried with magnesium sulfate, filtered and evaporated, leaving an off-white solid. This is crystallized from ethyl acetate with the addition of an equal volume of ether. This provides 0.18 g (80%) of 4-(4-acetylphenoxy)butanoic acid as light beige crystals:
m.p. 148-50 C; IR (KBr) 1730, 1650 cm-l; lH-NMR (CDC13) is consistent with the desired product; MS (FAB) m/e 223.0974 ~ = -0.4 mm~ (M' + H). Anal. calc'd. for Cl2Hl4O4: C, 64.85; H, 6.35; O, 28.80. Found:- C, 64.61; H, 6.36; O, 29.03.
P~PA~ATTON 4 c~MPou~n 8 4-(3-Formyl ph~n~y) hut~nn;c acid 3-Hydroxybenzaldehyde (900 mg, 7.37 mmol) is treated with 2.16 g (11.05 mmol) of ethyl 4-bromo-butyrate, 3.06 g (22.11 mmol) of potassium carbonate, and a catalytic amount (110 mg 0.74 mmol) of sodium iodide under the same conditions as in Preparation 3 to give a yellow oil. Purification by flash chromatography using hexane/ethyl acetate (10:1) gives 1.61 g of 4-(3-formylphenoxy)butanoic acid, ethyl ester as a~light yellow oil. The lH NMR (300 MHz, CDC13) is consistent with the desired product; IR (neat) 1730, 1700, 1600, 1580 cm-l; MS (CI low res) m/e 237 (M+H), 191, 115.

A solution of 385 mg (1.63 mmol) of 4-(3-formylphenoxy)butanoic acid, ethyl ester and 850 mg (6.15 mmol) of potassium carbonate is stirred in 6 mL of methanol/water (3:2) at room temperature for 8 hr. The solution is then concentrated in vacuo. The residue is dissolved in 10 mL of 0.1N sodium hydroxide solution and washed with 20 mL of ether. The aqueous layer is separated and acidified with sodium bisulfate and extracted with ethyl acetate. The organic layer is washed with saturated sodium chloride solution, then dried over magnesium sulfate. The mixture is then filtered and concentrated in vacuo to give 315 mg of 4-(3-formylphenoxy)butanoic acid as a white solid. The product is utilized in the next reaction without further purification. m.p. 62-63; the lH NMR (300 MHz, CDCl3) is consistent with the desired produ-ct; IR (KBr) 3400, 3000, 1700, 1690, 1590 cm-l; MS (CI low res) m/e 209 (M+H), 191, 123.
PREP~ATTON 5, COMPoU~n g 4-(4-Formvl~h~nnxv)hut~nn;c ac;~
4-Hydroxybenzyl alcohol (1 g, 8.06 mmol) is treated with 1.73 g (8.86 mmol) of ethyl 4-bromo-butyrate, 3.34 g (24.2 mmol) of potassium carbonate and a catalytic amount (120 mg 0.81 mmol) of sodium iodide under the same conditions as described in Preparation 3 to give 1.73 g of 4-[4-(hydroxymethyl)phenoxy]butanoic acid, ethyl ester as a light brown oil (90%). The product is utilized in the next reaction without further purification. The lH NMR (300 MHz, CDCl3) is consistent with the desired product; IR (neat) 3400, 1730, 1610, 1580, 1510 cm-l; MS (CI) m/e 238, 221, 115.
A mixture of 230 mg (0.97 mmol) of 4-[4-(hydroxymethyl)phenoxy]butanoic acid, ethyl est-er, 624.2 mg (2.9 mmol) of pyridinium chlorochromate, and a catalytic amount of 4 A molecular sieve is stirred in 2 mL of methylene chloride at room temperature for 3 hr.

215078~

The mixture is diluted with 20 mL of ether, filtered and concentrated in vacuo to give 175 mg (76%) of a light yellow oil. The oil ~150 mg, 0.63 mmol) is dissolved in 2.3 mL of methanol/water (3:2) and treated with 307 mg (2.22 mmol) of potassium carbonate according to the procedure described for Example 4 to give 100 mg (75%) of 4-(4-formylph~noxy)butanoic acid as a white powder.
The product is used without further purification. The lH NMR (300 MHz, CDC13) iS consistent with the desired product; IR (KBr) 3000, 1740, 1660 cm-l; MS (CI (low res)) m/e 209 (M+H), 191, 123.

PREPARATTON 6, COMPOUND 1O
4-(4-Acetvl-2-methvl~henoxY)hllt~n~;c ac;d Utilizing the procedure of Preparation 3, 2.00 g (13.32 mmol) of 4-hydroxy-3-methylacetophenone is alkylated with ethyl 4-bromobutyrate. This produces 3.45 g (98%) of 4-(4-acetyl-2-methylphenoxy)butanoic acid, ethyl ester as a golden oil, after drying at 75 C, under vacuum: IR (neat) 1740, 1675 cm-l; 1~ KR
(CDC13 ) iS consistent with the desired product; MS (FAB) m/e 265 (M+ + H). Anal. calc'd. for C1sH20o4: C, 68.16;
H, 7.63; O, 24.21. Found: C, 67.92; H, 7.44; O, 24.64.
Following the method of Preparation 3, 2.50 g (9.46 mmol) of 4-(4-acetyl-2-methylphenoxy)butanoic acid, ethyl ester is saponified to give the desired compound as a solid. It is recrystallized from ethyl acetate/ether leaving 1.32 g (59%) of 4-(4-acetyl-2-methylphenoxy)butanoic acid as white crystals: m.p.
114-16 C; IR (KBr) 1730, 1650 cm-l; 1H-NMR (CDC13) is consistent with the desired product; MS (FAB) m/e 237 (M+ + H). Anal. calc'd. for C13H16O4: C, 66.08; H, 6.83; O, 27.09. Found: C, 65.88; H, 6.92; 0,--27.20.

_ 42 21~078S

P~FPA~ATTON 7, cnMPouND 11 4-(4-Formyl-2-methnyyph~n~y)hllt~no;c ~c;d 4-Hydroxy-3-methoxybenzyl alcohol (1 g, 6.49 mmol) is treated with 1.73 g (7.13 mmol) of ethyl 4-bromobutyrate, 2.69 g (19.46 mmol) of potassium carbonate and a catalytic amount (97.22 mg 0.65 mmol) of sodium iodide as described in Preparation 3 to give 821 mg of 4-[4-(hydroxymethyl)-2-methoxyphenoxy]butanoic acid, ethyl ester as a light brown oil (47%). The lH
NMR (300 MHz, CDC13) is consistent with the desired product; IR (neat) 3500, 1730, 1620, 1600 cm-l; MS (CI
(low res)) m/e 269 (M+H), 251, 223, 195.
4-[4-(Hydroxymethyl)-2-methoxy~henoxy]butanoic acid, ethyl ester (431 mg, 1.61 mmol) is treated with 1.0 g (4.8 mmol) of pyridinium chlorochromate by the procedure described in Example 5 to give 280 mg (65%) of 4-(4-formyl-2-methoxyphenoxy)butanoic, ethyl ester as a colorless oil. The lH NMR (300 MHz, CDC13) is consistent with the desired product; IR (neat) 1730, 1690, 1600, 1580 cm-l.
4-(4-Formyl-2-methoxyphenoxy)butanoic acid, ethyl ester (240 mg, 0.90 mmol) is dissolved in 3 mL of methanol/water (3:2) and treated with 435 mg (3.15 mmol) of potassium carbonate according to the procedure described for Example 4 to give 125 mg (58%) of 4-(4-formyl-2-methoxyphenoxy)butanoic acid as a white powder:
m.p. 143-148; the lH NMR (300 MHz, CDC13) is consistent with the desired product; IR (KBr) 3575, 3500, 1720, 1700, 1680, 1600, 1585 cm-l; MS (CI (low res)) m/e 239 (M+H), 221, 207, 153.
p~FPA~ATTON 8 C~MpouND 1~
4-Formylhenzene~ro~no;c ac;d A mixture of 253 mg (1.44 mmoi) of 4-formylc;nn~m;c acid and 32.61 mg of platinum oxide in 10 mL of methanol is stirred overnight at room temperature _ 43 under an atmosphere of hydrogen supplied by a balloon.
The mixture is filtered through celite and concentrated in vacuo. The residue is dissolved in 0.lN sodium hydroxide solution and washed with ether. The aqueous layer is then acidified and the product is extracted with ethyl acetate. The organic layer is washed with saturated sodium chloride solution and dried over magnesium sulfate. The solvent is removed in vacuo to afford an inseparable mixture of 4-formylphenylpropanoic acid and other reduction products. The mixture is utilized in the next reaction without characterization or further purification.

P~PA~ATTON 9 COMPOUND 13 4-(2 3-D;methoxv-5-formvl~henoxv)hutano;c ~c;d Employing the method of Preparation 3, 3.30 g (18.41 mmol) of 3,4-dimethoxy-5-hydroxybenzaldehyde is alkylated with ethyl 4-bromobutyrate. 4-(2,3-Dimethoxy-5-formylphenoxy)butanoic acid, ethyl ester is obtained as a yellow-orange oil after drying under high vacuum at 60 C (5.45 g, 100%): IR (neat) 1735, 1690 cm-l; lH-NMR
(CDCl3) is consistent with the desired product; MS (FAB) m/e 297 (M+ + H). Anal. calc~d. for Cl5H20o6: C, 60.80;
H, 6.80; O, 32.40. Found: C, 60.51; H, 6.86; O, 32.63.
Following the procedure of Preparation 3, a sample of 4.70 g tl5.86 mmol) of 4-(2,3-dimethoxy-5-formylphenoxy)butanoic acid, ethyl ester is saponified giving the desired compound as a cream colored solid.
This is recrystallized from ethyl acetate/ether, leaving 3.65 g (86%) of 4-(2,3-dimethoxy-5-formylphenoxy)butanoic acid as off-white crystals: m.p.
90-92 C; IR (KBr) 1710, 1690 cm-l; lH-NMR (CDC13) is consistent with the desired product; MS (FAB) m/e 269 (M+ + H). Anal. calc'd. for Cl3Hl6O6: C, 58.20; H, 6.01; O, 35.79. Found: C, 58.10; H, 6.09; O, 35.81.

_ 44 21~078~

PRF.PARATTON 10, c~MpouNn 14 4-(4-Acetyl-2.6-~;methoxyghenoxy)but~noic ~c;d Utilizing the procedure of Preparation 3, 2.61 g (13.32 mmol) of 4-acetyl-2,6-dimethoxyphenol is treated with ethyl 4-bromobutyrate. This gives the desired product after drying at -70 C, under high vacuum, as a brown oil. This is chromatographed on a column of silica gel, and eluted with a 1:1 mixture of ether/hexane leaving 0.40 g (10%) of 4-(4-acetyl-2,6-dimethoxyphenoxy)butanoic acid, ethyl ester as a colorless oil: IR (neat) 1735, 1675 cm-1; lH-NMR
(CDC13) is consistent with the desired product; MS (FAB) m~e 311.1489 a = +o . 6 mm~ (M+ + H). Anal. calc~d. for C16H22O6: C, 61.92; H, 7.14; O, 30.94. Found: C, 61.48; H, 7.04; O, 31.48.
Following the method of Preparation 3, 0.179 g (O.577 mmol) of 4-(4-acetyl-2,6-dimethoxyphenoxy)-butanoic acid, ethyl ester is treated with potassium carbonate, producing an off-white solid. Recrystalli-zation from ethyl acetate/hexane gives 4-(4-acetyl-2,6-dimethoxyphenoxy)butanoic acid as white crystals (0.14 g, 88%): m.p. 122-24 C; IR (KBr) 1735, 1660 cm-1; 1H-NMR (CDCl3) is consistent with the desired product; MS
(FAB) m/e 283 (M+ + H). Anal. calc'd. for C14H1gO6: C, 59.57; H, 6.43; O, 34.01. Found: C, 59.34; H, 6.40; O, 34.26.
PRFP~ATTON 11, coMpou~n 15 4-(4-Acetvl-2-meth~xvph~nnxv)hut~no;c ac;d Employing the procedure of Preparation 3, 2.21 g (13.32 mmol) of 4-hydroxy-3-methoxyacetophenone is alkylated, producing a solid. This is recrystallized as in Preparation 3, leaving 3.23 g (86%) of 4-(4-acetyl-2-methoxyphenoxy)butanoic acid, ethyl ester as white crystals: m.p. 53-55 C; IR (KBr) 1745, 1675 cm-1; 1H-NMR (CDCl3) is consistent with the desired product; MS
(FAs) m/e 281 (M+ + H). Anal. calc~d. for C1sH20os: C, _ 45 64.27; H, 7.19; O, 28.54. Found: C, 64.26; H, 7.05; O, 28.69.
Following the method of Preparation 3, 2.74 g (9.78 mmol) of 4-(4-acetyl-2-methoxyphenoxy)butanoic acid, ethyl ester is saponified. This produces 4-(4-acetyl-2-methoxyphenoxy)butanoic acid as off-white crystals after recrystallization from ethyl acetate (1.61 g, 87%): m.p. 161-63 C; IR (KBr) 1720, 1670 cm-l; lH-NMR (CDC13) is consistent with the desired product; MS (FAB) m/e 253 (M+ + H). Anal. calc'd. for Cl3Hl6Os: C, 61.90; H, 6.39; O, 31.71. Found: C, 61.75; H, 6.37; O, 31.88.

p~FPA~ATToN 12 C~MPOUND 16 4- r 4-(3-Oxobutvl)~henoxvlbutano;c ac;d 4-Hydroxybenzylacetone (2 g, 12.18 mmol) is treated with 2.61 g (13.4 mmol) of ethyl 4-bromo-butyrate, 5.05 g (36.5 mmol) of potassium carbonate and a catalytic amount (182 mg 1.22 mmol) of sodium iodide in 2 mL N,N-dimethylformamide as described in Prepara-tion 3 to give 2.73 g of 4-[4-(3-oxobutyl)phenoxy]-butanoic, ethyl ester as a light brown oil (80~): m.p.
32-34; the lH NMR (300 MHz, CDCl3) is consistent with the desired product; IR (neat) 1730, 1720, 1610, 1580, 1510 cm-l; MS (CI (low res)) m/e 279 (M+H), 233, 221;
Analysis calc~d for Cl6H22O4: C, 69.04; H, 7.97; found:
C, 68.33; H, 7.68.
4-[4-(3-Oxobutyl)phenoxy]butanoic acid, ethyl ester (716 mg, 2.57 mmol) is dissolved in 5 mL of methanol/water (3:2) and treated with 1.24 g (9.0 mmol) of potassium carbonate according to the procedure described for Example 4 to give 385 mg (60%) of 4-[4-(3-oxobutyl)phenoxy]butanoic acid as a white powder: m.p.
97-99; the lH NMR (300 MHz, CDCl3) is consistent with the desired product; IR (neat) 1730, 1700, 1620, 1520 _ 46 cm-l; Analysis calc~d for Cl4HlgO4: C, 67.18; H, 7.25;
found: C, 66.55; H, 7.09.
~Z~MPT F 1~ . COMPOuNn 1 7 4-(2-Acetyl-5-metho~ypheno~y)hllt~noic ac;d Following the procedure of Preparation 3, 2.21 g (13.32 mmol) of 2-hydroxy-4-methoxyacetophenone is alkylated and worked up as before to leave 3.40 g (91%) of 4-(2-acetyl-5-methoxyphenoxy)butanoic acid, ethyl ester as a yellow oil: IR (neat) 1740, 1665 cm-l; lH-NMR (CDC13) iS consistent with the desired product; MS
(FAB) m/e 281 (M+ + H). Anal. calc'd. for ClsH20os: C, 64.27; H, 7.19; O, 28.54. Found: C, 64.06; H, 7.24; O, 28.70.
Utilizing the method of Preparation 3, 2.50 g (8.92 mmol) of 4-(2-acetyl-5-methoxyphenoxy)butanoic acid, ethyl ester is treated with potassium carbonate, producing a white solid. This is recrystallized from ethyl acetate/ether leaving 1.61 g (71%) of 4-(2-acetyi-5-methoxyphenoxy)butanoic acid as colorless crystals:
m.p. 127-29 C; IR (KBr) 1720, 1655 cm-l; lH-NM~ (CDC13) is consistent with the desired product; MS (FAB) m/e 253 (M+ + H). Anal. calc'd. for Cl3Hl6Os: C, 61.90; H, 6.39; O, 31.71. Found: C, 61.82; H, 6.37; O, 31.81.
p~?FP.~l~ATTON 14, COMPOU~) 18 4-~4-(3-Oxo~o~yl)Dhenoxvlhllt~n~;c ~c;~
Following the procedure of Preparation 3, 2.80 g (18.41 mmol) of 3-(4-hydroxyphenyl-1-propanol) is alkylated with ethyl 4-bromobutyrate. The product is dried at 70 C, under high vacuum, leaving 4.70 g (96%) of 4-[4-(3-hydroxypropyl)phenoxy]butanoic acid, ethyl ester as a colorless oil: IR (neat) 3400 (br.), 1735 cm-l; lH-NMR (CDC13 ) is consistent with the desired product; MS (CI) m/e 267 (M+ + H). Anal. calc'a. for ClsH22O4: C, 67.65; H, 8.33; O, 24.03. Found: C, 67.40; H, 8.20; O, 24.38.

_ 47 215078~

After the method of Preparation 3, 4.13 g (15.51 mmol) of 4-[4-(3-hydroxypropyl)phenoxy]butanoic acid, ethyl ester is saponified with potassium carbonate to produce a solid. This is recrystallized from an ethyl acetate-hexane mixture giving 2.45 (66~) of 4-[4-(3-hydroxypropyl)phenoxy]butanoic acid as white crystals: m.p. 92-94 C; IR (KBr) 3420 (br.), 1710 cm-l; lH-NMR (CDC13 ) is consistent with the desired product; MS (CI) m/e 239 (M+ + H). Anal. calc~d. for Cl3HlgO4: C, 65.53; H, 7.61; O, 26.86. Found: C, 65.75; H, 7.87; O, 26.38.
A 1.19 g (5.00 mmol) sample of 4-[4-(3-hydroxypropyl)phenoxy]butanoic acid is dissolved with stirring in 250 mL of methylene dichloride. Next, 3.77 g (17.49 mmol) of pyridinium chlorochromate is added, the mixture is stirred for 4 hr, and then filtered through a celite pad. The reaction mixture is then diluted with an equal volume of ether, precipitating out salts. This mixture is then filtered through a silica gel pad, and the filtrate evaporated, giving a brown solid. The solid is recrystallized from an ether-hexane mixture producing 0.21 (18%) of 4-[4-(3-oxopropyl)phenoxy]butanoic acid as off-white crystals:
m.p. 100-03 C; IR (KBr) 1715 cm-l; lH-NMR (CDCl3) is consistent with the desired product; MS (CI) m/e 237 (M+
+ H). Anal. calc~d. for C13H16O4: C, 66.09; H, 6.83; O, 27.09. Found: C, 65.91; H, 6.72; O, 27.35.
F.~AMPT.F. 15, cnMpouND 20 4- r ( 7-Acetvl-l-na~hth~lenvl)oxvlhllt~n~;c ac;d A 3.42 g (18.37 mmol) sample-of 1-hydroxy-2-acetonapthone is alkylated as in Preparation 3. The crude product is dried under high vacuum at 60 C to give 5.21 g (94%) of 4-[(2-acetyl-1-naphthalenyl)-oxy]butanoic acid, ethyl ester as a golden liquid: IR
(neat) 1730, 1665 cm-l; lHNMR (CDC13) is consistent with the desired product; MS (FAB) m/e 301(M+ + H). Anal.

_ 48 215078~

calc'd. for Cl8H20o4: C, 71.98; H, 6.71; O, 21.31.
Found: C, 72.11; H, 6.58; O, 21.31.
Utilizing the method of Preparation 3, 2.84 g (9.46 mmol) of 4-[(2-acetyl-1-naphthalenyl)oxy]butanoic acid, ethyl ester is saponified. The crude product is recrystallized from ethyl acetate/ether to give 1.15 g (45~) of 4-[t2-acetyl-1-naphthalenyl)oxy]butanoic acid as golden crystals: m.p. 104-06 C; IR (KBr) 1720, 1640 cm-l; lHMMR (CDC13) iS consistent with the desired product; MS (FAB) m/e 273 lM+ + H). Anal. calc'd. for C16H16O4: C, 70.58; H, 5.92; O, 23.50. Found: C, 70.40; H, 5.89; O, 23.71.

PR~P~ATTON 16, CoMPou~n ~1 4-r4-(4-Fluoroh~n~ovl)phenoxylhut~no;c ac;d Following the method of Preparation 3, 3.98 g (18.41 mmol) of 4-fluoro-4~-hydroxybenzophenone is alkylated with ethyl 4-bromobutyrate. The crude yellow solid product is recrystallized from ether providing 2.97 g (49%) of 4-[4-(4-fluorobenzoyl)phenoxy]butanoic acid, ethyl ester as white crystals: m.p. 57-59 C; IR
(KBr) 1735, 1645 cm-l; lHNMR (CDC13) is consistent with the desired product; MS (FAB) m/e 311 (M+ + H). Anal.
calc'd. for ClgHlgO4F: C, 69.08; H, 5.80; F, 5.75; O, 19.37. Found: C, 69.09; H, 5.62; F, 5.95; O, 19.34.
Utilizing the procedure of Preparation 3, 0.48 g (1.45 mmol) of 4-[4-fluorobenzoyl)phenoxy]butanoic acid, ethyl ester is saponified. The crude white solid product is recrystallized from an ether-hexane mixture leaving 0.16 g (36~) of 4-[4-(4-fluorobenzoyl)phenoxy]-butanoic acid as white crystals: m.p. 109-111 C; IR
(KBr) 1735, 1700 1640 cm-l; lHNMR (CDCl3) is consistent with the desired product; MS (FAs) m/e 303 (M+ `+ H).
Anal. calc'd. for Cl7HlsO4F: C, 67.54; H, 5.00; F, 6.28 O, 21.18. Found: C, 67.28; H, 4.89; F, 6.41; O, 21.42 _ 49 21507~5 F.XAMPr.F. 17. COMPouND ~-4-(4-Acetvl~henyl)-1-D;~eraz;nev~ler;c ac;d 4'-Piperazinoacetophenone (102 mg) is dissolved in 1 mL of N,N-dimethylformamide. After addition of methyl 5-bromovalerate (0.077 mL) and potassium carbonate (69 mg), the mixture is stirred at room temperature for 65 hr. TLC (10% MeOH/CH2Cl2) should show a single product spot without residual starting material. The reaction solution is evaporated under vacuum. The residue is taken up in methylene chloride, washed twice with water and dried over sodium sulfate. Evaporation of the solvent yields 137 mg of 4-(4-acetylphenyl)-1-piperazinevaleric acid, methyl ester as yellow crystals whose 1H-NMR (CDCl3) spectrum is consistent with the assigned structure.
4-(4-Acetylphenyl)-1-piperazinevaleric acid, methyl ester (15.3 mg) is suspended in 0.1 mL of potassium hydroxide solution (33.2 mg/mL). After heating at 100 C for 150 min, the starting material is completely dissolved and absent by TLC (10%
MeOH/CH2Cl2). After acidifying the reaction solution to pH 4 by adding 0.2 N HCl, the aqueous solution is extracted with methylene chloride. After evaporation of the organic layer to dryness, the residue is dissolved in methylene chloride and filtered. Evaporation of the organic layer gives 7 mg of 4-(4-acetylphenyl)-1-piperazinevaleric acid as a white solid. 1H-NMR (CDC13) spectrum is consistant with the assigned structure. MS
3 (FAB) m/e 305 (M+ + H), 327 (M+ + Na), 348 (M+ + 2Na-H).
PRFP~ATTON 18, COMPOU~ 25 4-(~-Chloro-4-formvl~henoxv)hllt~n~;c ~c;d Following the procedure of Preparation 3, 2.88 g (18.41 mmol) of 3-chloro-4-hydroxybenzaldehyde is alkylated as before. This produces 4.65 g (93%) of 4-(2-chloro-4-formylphenoxy)butanoic acid, ethyl ester as an orange oil: IR (neat) 1730, 1685 cm-l; lHNMR (CDC13) _ - 2150785 50 is consistent with the desired product; MS (CI) mJe 271 (M+ + H). Anal. calc~d. for Cl3Hl5O4Cl: C, 57.68; H, 5.58; Cl, 13.10; O, 23.64. Found: C, 58.05; H, 5.37;
Cl, 12.43; O, 24.15.
After the method of Preparation 3, 3.52 g (13.00 mmol) of 4(2-chloro-4-formYlphenoxy)butanoic acid, ethyl ester is saponified to give a white solid.
This is recrystallized from ethyl acetate resulting in 1.78 g (56%) of 4-(2-chloro-4-formylphenoxy)butanoic acid as white crystals: m.p. 128-31 C; IR (KBr) 1730, 1650 cm-l; lHNMR (CDC13 ) is consistent with the desired product; MS (CI) m/e 243 (M+ + H). Anal. calc'd. for CllHllO4Cl: C, 54.45; H, 4.57; Cl, 14.61; O, 26.37.
Found: C, 54.61; H, 4.70; Cl, 14.25; O, 26.42.
FXAMPT.F. 1 9, COMPOUND ~6 5-Acetvl-2-(3-c~rboxY~ro~oxY)henzoic ~ci~ methvl ester Under dry condition, 3.58 g (18.41 mmol) of 5-acetylsalicylic acid, methyl ester is dissolved in 25`mL
of dry N,N-dimethylformamide. To this solution is added 3.07 g (20.58 ~mol) of 5-bromo-1-pentene, 6.83 (20.58 mmol) of potassium carbonate, and 0.246 g (1.65 mmol) of potassium iodide, and the reaction mixture is stirred for 24 hr at ambient temperature. Another portion of 5-bromopentene is added to the reaction, followed bY one-half portions of the other two reagents above, and stirring is continued for 72 hr. The mixture is then evaporated under high vacuum-at 70 C. The residue is partitioned between ether/water and the organic phase is separated, dried with magnesium sulfate, filtered, and evaporated under vacuum to leave 4.60 g (95%) of 5-acetyl-2-(4-pentenyloxy)benzoic acid, methyl ester as a yellow liquid: IR (neat) 1735, 1710, 1680 cm-l; lHNMR
(CDCl3) is consistent with the desired product; MS (FAB) m/e 263 (M+ + H) . Anal. calc'd. for ClsHlgO4: C, 68.69;
H, 6.92; O, 24.40. Found: C, 68.60; H, 6.92; O, 24.46.

-- 215078~ 51 A sample of 0.203 g (0.775 mmol) of 5-acetyl-2-(4-pentenyloxy)benzoic acid, methyl ester is dissolved in 5 mL of methylene dichloride, under an argon atmos-phere, and cooled to -78 C in a dry ice acetone bath, with stirring. Next, ozone gas is passed through this solution for 10 min, until it turns a light bluish color. Then 0.5 mL of dimethyl sulfide is added to quench the reaction and it is allowed to warm to room temperature for 2 hr. The mixture is then evaporated under high vacuum, leaving the crude aldehyde product as an oil which is used "as is~ for the second step. It is dissolved in 5 mL of N,N-dimethylformamide, and 1.02 g (2.71 mmol) of pyridinium dichromate is added. This reaction mixture is sealed and allowed to stand for 20 hr. It is next poured into 50 m~ of water, extracted with ether, and the organic phase is washed with water again, dried with magnesium sulfate, filtered, and evaporated, which gives oily crystals. These are recrystallized from a mixture of ethyl acetate and hexane, producing 0.109 g (50%) of 5-acetyl-2-(3-carboxypropoxy)benzoic acid, methyl ester as white crystals: m.p. 111-113 C; IR (KBr) 1725, 1645 cm-l;
lHNMR (CDCl3) is consistent with the desired product; MS
(FAB) m/e 281 (M+ + H). Anal. calc~d. for Cl~H1606: C, 60.00; H, 5.75; O, 34.25. Found: C, 59.96; H, 5.75; O, 34.27.
PR~PA~ATION 20 COMPOUNn 27 4-(4-Formvl-2-n;tro~henoxv)but~n~;c ac;d 4-Hydroxy-3-nitrobenzyl alcohol (1 g, 5.91 mmol) is treated with 1.44 g (7.39 mmol) of ethyl 4-bromobutyrate, 2.86 g (20.69 mmol) of potassium carbonate and a catalytic amount (88 mg .59 mmol) of sodium iodide as described in Preparation 3 to-give 1.45 g of 4-[4-(hydroxymethyl)2-nitroph~noxy]butanoic acid, ethyl ester as a light yellow oil (86%). The lH NMR
(300 MHz, CDCl3) is consistent with the desired product;

- ~ 21S078~ 52 IR (neat) 3400, 1730, 1710, 1630, 1580 cm-l; MS (CI) m/e 284 (M+H), 238; Analysis calc~d for C13H1706N: C, 55.12;
H, 6.05; found: C, 55.36; H, 6.03.
4-[4-(Hydroxymethyl)2-nitrophPnoxy]butanoic acid, ethyl ester (300 mg, 1.06 mmol) is treated with 799 mg (3.71 mmol) of pyridinium chlorochromate by the procedure described in Example 5 to give 188 mg (63%) of 4-(4-formyl-2-nitrophenoxy)butanoic acid, ethyl ester as a colorless oil. The lH NMR (300 MHz, CDC13) is consistent with the desired product; IR (neat) 1730, liOO, 1610, 1570 cm-l; MS (CI) m/e 282 (M+H).
4-(4-Formyl-2-nitrophenoxy)butanoic acid, ethyl ester (135 mg, 0.48 mmol) is dissolved in ~ mL of methanol/water (3:2) and treated with 232 mg (1.68 mmol) of potassium carbonate according to the procedure described for Example 4 to give 84 mg (69%) of 4-(4-formyl-2-nitrophenoxy)butanoic acid as a yellow powder:
m.p. 136-139; the lH NMR (300 MHz, CDCl3) is consistent with the desired product; IR (KBr) 3400, 1730, 1700, 1650, 1600, 1570 cm-l; MS (CI) m/e 254, 236, 224, 208, 196, 168.
~MPT.F. 21 CO~POUND ~8 4- r~- r r (4-Acetyl~he~yl)~m'nolm~th,yll-6-methn~y-phenoxvlhlltano;c ~c;d 4'-(2-Hydroxy-3-methoxybenzyl~;no)aceto-phenone (500 mg, 1.84 mmol) is treated with 629 mg (3.22 mmol) of ethyl 4-bromobutyrate, 764 mg (5.53 mmol) of potassium carbonate and a catalytic amount (182 mg, 1.22 mmol) of sodium iodide in 2 mL N,N-dimethylformamide as described in Preparation 3 to give 680 mg of 4-[2-[[(4-acetylphenyl)amino]methyl]-6-methoxyphenoxy]butanoic acid, ethyl ester as a light brown oil (95%). The lH
NMR (300 MHz, CDCl3) is consistent with the desired product; IR (neat) 3400, 1730, 1660, 1600 cm-l; MS (CI) m/e 386 (M+H), 115; Analysis calc'd for C22H270sN: C, ~ J 2150785 53 _ 68.55; H, 7.06; N, 3.63; found: C, 68.27; H, 6.81; N, 3.54.
4-[2-[[(4-Acetylphenyl)amino]methyl]-6-methoxy-phenoxy]butanoic acid, ethyl ester (250 mg, 0.65 mmol) is dissolved in 5 mL of methanol/water (3:2) and treated with 313 mg (2.27 mmol) of potassium carbonate according to the procedure described for Example 4 to give 166 mg (71%) of 4-[2-[[(4-acetylphenyl)amino]-methyl]-6-methoxy-phenoxy]butanoic acid as a red colored solid: m.p. 85-95 C; The lH NMR (300 MHz, CDCl3) is consistent with the desired product; IR (KBr) 3400, 1720, 1630, 1580 cm-1; MS (CI) m/e calc~d for C20H23OsNNa: 380.1473, found 380.1482; 358 (M+H), 233, 223, 221, 136.

~X~MpT.F. 22. COMPOUND 29 5-Acetvl-2-(3-carhoxvDro~oxY)-benzoic acid, 1-(3-~ u~ouvl) ester To a solution of 0.744 g (3.00 mmol) of 5-acetyl-2-(4-pentenyloxy)-benzoic acid (Example 19), under an argon atmosphere, with stirring, in 36 mL of methylene dichloride, is added 1.67 g (12.0 mmol) of 3-bromopropanol. This is followed by 0.912 g (9.0 mmol) of triethyl amine and by 1. 66 g (3.-75 mmol) of benzotriazol-l-yloxytris(dimethylamino) phosphonium hexafluorophosphate and the reaction is stirred for 20 hr. The mixture is then evaporated, under high vacuum at 65 C. The residue is partitioned between ether and water, the ether phase is washed twice more with water, dried with magnesium sulfate, filtered, and evaporated leaving a gum. This is chromatographed on a column of silica gel, and eluted with ethyl acetate/hexane (1:2) to give 0.80 g (72%) of 5-acetyl-2-(4-pentenyloxy)-benzoic acid, 1-(3-bromopropyl) ester as a colorless oil: IR (neat) 1730, 1700, 168Q cm-l; lHNMR (CDC13) is consistent with the desired product; MS (FAB) m/e 369 (M+ + H). Anal. calc~d. for C17H21O4Br: C, 55.30; H, 5.73; O, 17.33; Br 21.64. Found: C, 55.34; H, 5.44; o, 17.34; sr 21.88.
Following the procedure of Example 19, 0.377 g (1.02 mmol) of 5-acetyl-2-(4-pentenyloxy)-benzoic acid, 1-(3-bromo~Lo~l) ester is ozonized, and then further oxidized with pyridinium dichromate producing a colorless gum which partially crystallizes. This is recrystallized from a mixture of equal parts of ethyl acetate and hexane leaving 0.277 g (70%) of 5-acetyl-2-(3-carboxypropoxy)-benzoic acid, 1-(3-bromopropyl) ester as white crystals: m.p. 103-05 C; IR (KBr) 1730, 1645 cm-1; lHNMR (CDC13 ) is consistent with the title product; MS (CI) m/e 389 (M~ + H). Anal. calc~d. for C16H1gO6Br: C, 49.63; H, 4.95; O, 24.79; Br, 20.63.
Found: C, 49.90; H, 4.75; O, 24.94, Br, 20.39.

p~FP~ATTON 23. coMpou~n 30 4-(4-Acetyl-3-fllloro~h~noxv)hut~no;c ~c;d A solution of 2-fluoro-4-methoxyacetophenone in 5 mL of DMSO is stirred at 100 C in the presence of 730 mg (15 mmol) of sodium cyanide to give a dark viscous sludge. The mixture is allowed to cool, then poured into 50 mL of ice water and acidified with 6N
aqueous HCl. The acidic solution is extracted with ethyl acetate (50 mL x 2) and the organic layers are combined and washed with water. The organic layer is then extracted twice with 1.0 N aqueous sodium hydroxide solution. The basic layer is washed once with ether, then acidified with solid sodium bisulfate and extracted with ethyl acetate twice. The ethyl acetate layers are combined, then washed with 10% sodium bisulfate solution and saturated sodium chloride solution. The organic phase is dried over magnesium sulfate and concentrated in vacuo at ambient temperature to give 143 mg (31%) of - an oil.

- 215078~ 55 The oil isolated above is dissolved in 1 mL of - N,N-dimethylformamide and treated with 205 mg (1.05 mmol) of ethyl 4-bromobutyrate, 4.07 g (2.95 mmol) of potassium carbonate and a catalytic amount (1.26 mg, 0.008 mmol) of sodium iodide according to the procedure described in Preparation 3 to give 39 g of 4-(4-acetyl-3-fluorophenoxy)butanoic acid, ethyl ester as a light brown oil (17%). The lH NMR (300 MHz, CDCl3) iS
consistent with the desired product; MS (CI (low res)) m/e calc~d for Cl4Hl8O4F: 269.1189, found 269.1191.
4-(4-Acetyl-3-fluorophenoxy)butanoic acid, ethyl ester (20 mg, 0.0745 mmol) is dissolved in 1 mL of methanol/water (3:2) and treated with 30.91 mg (0.22 mmol) of potassium.carbonate according to the procedure described for Example 4 to give 14 mg (82%) of 4-(4-acetyl-3-fluorophenoxy)butanoic acid as a white powder:
- m.p. 110-111 C; The lH NMR (300 MHZ, CDC13) iS
consistent with the desired product; IR (KBr) 1710, 1670, 1610 cm-l; MS m/e calc~d for Cl2Hl3O4FNa:
263.0695, found 263.0699.
EX~MPT.F. 24 COMPOUND 31 (2-Acetvl~henoxv)hl~t~noic ac;d 2-Acetylphenol (1 g, 7.34 mmol) is treated with 1.79 g (9.18 mmol) of ethyl 4-bromobutyrate, 3.55 g (25.71 mmol) of potassium carbonate and a catalytic amount (11 mg, 0.07 mmol) of sodium iodide as described in Preparation 3 to give 1.84 g of (2-acetylphenoxy)-butyric acid, ethyl ester as a light yellow oil which solidified upon standing: m.p. 43-45 C; The lH NMR
(300 MHz, CDCl3) iS consistent with the desired product;
IR (neat) 1730, 1660, 1600 cm-l; MS (CI) m/e 251 (M+H), 232, 205.
(2-Acetylphenoxy)butanoic acid, ethyl ester (500 mg, 2.00 mmol) is dissolved in 3 mL of methanol/water (3:2) and-treated with 828 mg (5.99 mmol?
of potassium carbonate according to the procedure -described for Example 4 to give 412 mg (93%) of (2-acetylphenoxy)butanoic acid as a white powder. The 1H
NMR (300 MHz, CDC13) is consistent with the desired product; IR (KBr) 1710, 1670, 1590 cm-1; MS m/e calc~d for C12H1sO4: 223.0970, found 223.0971.
EX~MPLE 25, COMPOUND 32 2-Acetvl-lOH-~henothiazine-10-hexanoic acid A solution of 500 mg (2.07 mmol) of 2-acetylphenothiazine in 8 mL of tetrahydrofuran is cooled to -78 C and 4.14 mL (2.07 mmol) of a 0.5 M solution of potassium bis(trimethylsilyl)amide in toluene is added.
After five minutes, a solution of [(6-bromohexyl)oxy]-(1,1-dimethylethyl)dimethyl silane in 2 mL of tetra-lS hydrofuran is added and the reaction is allowed to warm to room temperature. The mixture is diluted with 25 mL
of ethyl acetate and washed with 10% sodium bisulfate solution and saturated sodium chloride solution, then dried over magnesium sulfate and concentrated in vacuo to give a dark colored residue. Flash chromatography (3:1 hexane/ethyl acetate) provides 318 mg (33%) of 1-[10-[6-[[(1,1-dimethylethyl)dimethylsilyl]oxy]hexyl]-lOH-phenothiazin-2-yl]ethanone as a dark colored oil.
The 1H NMR (300 MHz, CDC13) is consistent with the desired product; IR (neat) 1680, 1600, 1560 cm-1; MS m/e calc~d for C26H37NO2SSi: 455.2314, found 455.2312.
A solution of 150 mg (0.33 mmol) of 1-[10-[6-[[(1,1-dimethylethyl)dimethylsilyl]oxy]hexyl]-lOH-phenothiazin-2-yl]ethanone in 0.6 mL of tetrahydrofuran is treated with 0.41 mL (0.41 mmol) of 1 M tetra-butylammonium fluoride in tetrahydrofuran. The reaction is stirred for 3 hr at room temperature, then diluted with 20 mL of ethyl acetate. The organic layer is washed successively with 10% sodium bisulfate solution and saturated sodium chloride solution, then dried over magnesium sulfate and concentrated in v~cuo to give 114 mg of 1-[10-(6-hydroxyhexyl)-lOH-phenothiazin-2--yl]ethanone as a dark oil. The 1H NMR (300 MHz, CDCl3) is consistent with the desired product; IR (neat) 3400, 1680, 1590, 1560 cm-1; MS m/e calc~d for C2oH23No2s:
341.1449, found 341.1456.
A solution of 41 mg tO.12 mmol) of 1-[10-(6-hydroxyhexyl)-lOH-phenothiazin-2-yl]ethanone in 0.16 mL
of N,N-dimethylformamide is treated with 158 mg (0.42 mmol) of pyridinium dichromate and stirred at room temperature for 12 hr. The mixture is diluted with ether and filtered through a pad of celite with the aid of 100 mL of ether. The filtrate is washed successively with 10% sodium bisulfate solution and saturated sodium chloride solution, then dried over magnesium sulfate and concentrated in vacuo to give 10 mg (23%) of 2-acetyl-lOH-phenothiazine-10-hexanoic acid as a dark residue.
The 1H NMR (300 MHz, CDC13) is consistent with the desired product. MS (CI) m/e 323 (M+ + H).

EXAMPLE 26, COMPOUND 34 5-Acetyl-2-(3-carboxyDro~oxv)-N-(2-~;methvlaminoethyl)-benzamide A sample of 0.140 g (0.50 mmol) of 5-acetyl-2-(3-carboxypropoxy)-benzoic acid, methyl ester (Example 19) is heated on a steam bath, under dry conditions with 5.49 mL (50.0 mmol) of N,N-dimethylethylenediamine for 5 hr. The mixture is allowed to cool for 20 hr to ambient temperature and evaporated under vacuum at 55 C. The brown gum produced is triturated with ether, and the r~m~ining residue taken up in water and acidified with hydrochloric acid. This is then extracted with ethyl acetate, and the aqueous solution is evaporated under vacuum, leaving a gum. It is next triturated with hot chloroform and this solution is evaporated to give a brown glass. This is chromatographed on a preparatory silica gel plate which is eluted with a 9/1 mixture of chloroform to methanol. The product band is cut from -the plate, triturated with the above solvent mixture, filtered, and evaporated leaving 0.025 g (15%) of 5-acetyl-2-(3-carboxypropoxy)-N-(2-dimethylaminoethyl)-benzamide as a light brown gum: MS (FAB) m/e 337.1753 +0.9 mmll (M+ + H), 359 (M+ + Na). lHNMR (CDC13) is consistent with the desired product.
EXAMPLE 27, COMPOUND 35 5-Acetvl-2-(3-carboxv~ro~oxv)-N-(2-trimethvlaminoethvl~-benzamide, internal salt To 100 mg of 5-acetyl-2-(3-carboxypropoxy)-N-(2-dimethylaminoethyl)-benzamide in 2 mL of methanol and 8 mL of pH 8.6 phosphate buffer is added 0.5 mL of dimethyl sulfate. The reaction pH is monitored about every 30 min and 0.1 N sodium hydroxide is added as needed to return the pH to -8.5. After 4 hr the solvents are removed under vacuum and the product ls purified on BioSil A with a methanol-in-chloroform gradient to give 5-acetyl-2-(3-carbomethoxypropoxy)-N-(2-trimethylaminoethyl)-benzamide, chloride which is taken on to the next step. lH-NMR (CDC13): 8.6 ppm (lH, d), 8.1 ppm (lH, dd), 7.1 ppm (lH, d), 4.3 ppm (2H, t), 4.0 ppm (2H, br t), 3.9 ppm (2H, br s), 3.7 ppm (3H, s), 3.7 ppm (lH, t), 3.3 ppm (9H, s), 2.1 ppm (3H, s), 2.1 ppm (2H, t), 2.3 ppm (2H, m).
The above product is dissolved in 2 mL of tetrahydrofuran and treated with an excess of 1 N sodium hydroxide for 16 hr at ambient temperature. The organic cosolvent is removed under vacuum and the aqueous solution which remains is acidified with 1 N HCl to a pH
of about 5. The solution is then evaporated under vacuum to give a glass which crystallizes on standing.
The resultant 5-acetyl-2-(3-carboxypropoxy)-N-(2-trimethylaminoethyl)benzamide, internal salt can be used without further purification. MS (FAB) m/e 351 (M+ +
H).

_ 59 5-Acetyl-2-~N-(2-dimethylaminoethvl)-3-c~rbox~midoproDoxylbenzoic acid internal salt To 1.16 g of 5-acetylsalicylic acid, methyl ester in 10 mL of N,N-dimethylformamide is added 1 g of chloroacetic acid, methyl ester and 1.2 g of potassium carbonate. After stirring this mixture at ambient temperature for 16 hr the reaction is filtered, diluted with ethyl acetate, and washed once with water and twice with brine. The ethyl acetate is dried with magnesium sulfate, filtered, and evaporated to give 5-acetyl-2-(carboxymethoxy)benzoic acid as a crude product.
Crystallization from methanol at -15 C gives 0.6 g of white crystals. lH-NMR (CDCl3): 8.5 ppm (lH, d), 8.1 ppm (lH, dd), 6.9 ppm (lH, d), 4.8 ppm (2H, s), 4.0 ppm (3H, s), 3.8 ppm (3H, s), 2.6 ppm (3H, s).
450 mg of the above product is stirred in 1 mL
of N,N-dimethylethylenediamine at ambient temperature for 16 hr. The reaction is then diluted with ethyl acetate and water. The water layer is extracted five times with ethyl acetate and the ethyl acetate from the various extractions is pooled, dried with magnesium sulfate, filtered, and evaporated to give 380 mg of 5-acetyl-2-[N-(2-dimethylaminoethyl)-3-carboxamido-propoxy]benzoic acid , methyl ester as a yellowish oil which is pure enough for further use. lH-NMR (CDCl3):
8.6 ppm (lH, d), 8.2 ppm (lH, dd), 8.1 ppm (lH, br t), 7.0 ppm (lH, d), 4.7 ppm (2H, s), 4.0 ppm (3H, s), 3.5 ppm (2H, q), 2.7 ppm (3H, s), 2.6 ppm (2H, t), 2.3 ppm (6H, s).
To 280 mg of the above compound in 15 mL of methanol and 5 mL of chloroform is added 1 mL of methyl iodide. After 3 hr at ambient temperature the~volatile components are removed. lH-NMR indicates the presence of the desired 5-acetyl-2-[N-(2-trimethylaminoethyl)-3-carboxamidopropoxy]benzoic acld, methyl ester, iodide.

_ 60 lH-NMR (CDC13+CD30D): 8.8 ppm (lH, br t), 8.6 ppm (lH, d), 8.2 ppm (lH, dd), 7.1 ppm (lH, d), 4.7 ppm (2H, s), 4.0 ppm (3H, s), 3.9 ppm (2H, q), 3.8 ppm (2H, t), 3.4 5(9H, s), 2.6 ppm (3H, s).
The above compound is dissolved in - 5 mL of methanol. Five equivalents of sodium hydroxide is added as a 5N solution in water. After 5 hr at ambient temperature the pH is adjusted to -7.5 with dilute HCl and the volatile components are removed under vacuum to give a crude product containing 5-acetyl-2-[N-(2-trimethylaminoethyl)-3-carboxamidopropoxy]benzoic acid, internal salt. MS (CI) m/e 323 (M+ + H).
SYNTHESIS OF STRUCTURES B (Scheme 1) General Procedure The drug-hydrazide derivative (Q-Sp-SS-W
wherein Q = H2NHN-) is dissolved in alcohol or other compatible organic solvent containing ~3 to 10 equivalents of the carbonyl linker and ~1-10~ acetic acid or other appropriate acid catalyst. A m;n;m~l amount of solvent gives a faster reaction. Anhydrous conditions give the best results as the condensation is an equilibrium reaction. The reaction is allowed to proceed at a temperature of ~20-60 C until complete by HPLC or alternately by TLC. This requires from a few hours to a day or more depending on the linker and the specific reaction conditions. The solvents are removed in vacuo and the crude product is purified on an appropriate silica gel, such as Biosil-A, using an appropriate solvent system, such as a gradient of 0 to 20% methanol in either chloroform or ethyl acetate. The products are sufficiently pure for subsequent steps.
E~AMPT . E 2 9 4-Formylphenoxyacetic acid (1) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 4.4 min, 215078~

FAB MS: 1641 (M+H), W max at 291 and 305 nm (acetate), H-NMR: See Fig. I.
P~FPARATION 30 4-Formylbenzoic acid (2) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.2 min, FAB MS: 1611 (M+H), W max at 292 and 302 nm (ethanol), H-NMR: See Fig. II.
P~FPARATION 31 4-Formyl-3-methoxyphenoxyacetic acid -(3) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 4.7 min, FAB MS: 1671 (M+H), W max at 282, 291, and 325 nm (ethanol), H-NMR: See Fig. III.

6-Formyl-2-naphthoic acid (4) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.1 min, FAB MS: 1661 (M+H), W max at 257, 267, 277, 313, and 321 nm (ethanol), lH-NMR: See Fig. IV.

4-(2-Oxoethoxy)benzenepropanoic acid (5) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.0 min, FAB MS: 1669 (M+H), W - no maxima.

3-(2-Oxoethoxy)benzoic acid (6) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.

HPLC retention time: 5.5 min, FAB MS: 1641 (M+H), W - no maxima.

-PRFP~RATION 35 4-(4-Acetylphenoxy)butanoic acid (7) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
s HPLC retention time: 6. 4 min, FAB MS: 1683 (M+H), W max at 285 nm (ethanol), H-NMR: See Fig. V.

4-(4-Acetylphenoxy)butanoic acid (7) condensed with calicheamicin gamma dimethyl hydrazide.
HPLC retention time: 6.2 min, W max at 285 nm (ethanol).
PRF.PARATION 37 4-(3-Formylphenoxy)butanoic acid (8) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.3 min, FAB MS: 1669 (M+H).
PRF.PARATION 3 8 4-(4-Formylphenoxy)butanoic acid (~) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.1 min, FAB MS: 1669 (M+H), W max at 291 nm (ethanol), 1H-NMR: See Fig. VII.
PREP~ATION 39 4-(4-Acetyl-2-methylphenoxy)butanoic acid (10) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6. 8 min, FAB MS: 1697 (M+H).
FX,~MPT.F. 40 4-(4-Formyl-2-methoxyphenoxy)butanoic acid (11) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.5 min, FAB MS: 1699 (M+H), W max at 284, 300, and 316 nm (acetonitrile).

4-Formylbenzenepropanoic acid (12) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.6 min, EAB MS: 1639 (M+H).
FX~MPLE 42 4-(2,3-Dimethoxy-5-formylphenoxy)butanoic acid (13) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.8 min, FAs MS: 1729 (M+H), W max at 302 nm (ethanol).
~X~MPLE 43 4-(4-Acetyl-2,6-dimethoxyphenoxy)butanoic acid (14) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.0 min, FAs MS: 1743 (M+H), W max at 287 nm (ethanol),.
F.XAMPT.~ 44 4-(4-Acetyl-2-methoxyphenoxy)butanoic acid (1~) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.1 min, FAs MS: 1713 (M+H), W max at 284 nm (ethanol).
~XAMPT.E 45 4-[4-(3-Oxobutyl)phenoxy]butanoic acetic acid (16) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.6 min, FAB MS: 1611 (M+H), W - no maxima.

4-(2-Acetyl-5-methoxyphenoxy]butanoic acid (17) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.5 min, FAB MS: 1713 (M+H), W - no maxima.
EXAMpTF 47 4-[4-(3-Oxopropyl)phenoxy]butanoic acid (18) condensed wlth Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 9.8 min, FAB MS: 1697 (M+H), W - no maxima.

4-Acetylbenzenebutanoic acid (19) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.4 min, FAB MS: 1667 (M+H), W max at 281 nm (ethanol).
EXAMPT.F. 49 4-[(2-Acetyl-1-naphthalenyl)oxy] butanoic acid (20) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 7.8 min, FAB MS: 1733 (M+H), W - no maxima.
EXAMpLF. 50 4-[4-(4-Fluorobenzoyl)phenoxy]butanoic acid (21) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 8.4 min, FAB MS: 1763 (M+H), W max at 284 nm (ethanol).

_ 65 ~X~MPLE 51 4-(4-Acetylphenyl)-l-piperazinepentanoic acid (22) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.0 min, FAB MS: 1641 (M+H), W max at 322 nm (ethanol).

11-(4-Acetylphenoxy)undecanoic acid (23) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 4.8 min (65% acetonitrile-isocratic), FAB MS: 1781 (M+H), W max at 286 nm (ethanol).
EXAMPT.F. 53 5-[(4-Acetylphenyl)amino]-5-oxopentanoic acid (24) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.2 min, FAB MS: 1710 (M+H), W max at 295 nm (ethanol).
F~x~MpT~E 54 4-(2-Chloro-4-formylphenoxy)butanoic acid (25) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.5 min, FAB MS: 1704 (M+H), W max at 292 nm (ethanol).

5-Acetyl-2-(3-carboxypropoxy)benzoic acid (26), methyl ester condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.1 min, FAB MS: 1741 (M+H), W max at 285 nm (ethanol).

4-(4-Formyl-2-nitrophenoxy)butanoic acid (27) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.2 min, FAB MS: 1741 (M+H), W max at 294 nm (ethanol).

4-[2-[[(4-Acetylphenyl)amino]methyl]-6-methoxyphenoxy]butanoic acid (28) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 7.7 min, FAB MS: 1818 (M+H), W max at 323 nm (ethanol).
FX~MPLE 58 4-(4-Acetyl-3-fluorophenoxy)butanoic acid (30) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.1 min, FAB MS: 1701 (M+H), W max at 273 nm (ethanol).
~X~MPTE 59 4-(2-Acetylphenoxy)butanoic acid (31) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.1 min, FAB MS: 1683 (M+H), W - no maxima.
EX~MPTE 60 2-Acetyl-lOH-phenothiazine-10-hexanoic acid (32) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 6.2 min, FAB MS: 1833 (M+NH4), W max at 281, strong shoulder at 356 nm (CH3CN).
FX~MPT~F~ 61 4-Acetylphenylacetic acid (33) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide.
HPLC retention time: 5.0 min, _ 67 -FAB MS: 1639 ~M+HJ, W max at 281 nm tacetonitrile).

SYNTHESIS OF STRUCTURES C (Scheme 1) General Procedure The carboxylic acid-hydrazones as obtained above are converted to the OSu esters (Z3=N-succinimidyloxy) by dissolving them in an appropriate solvent such as acetonitrile or acetonitrile containing 10-20% N,N-dimethylforamide or tetrahydrofuran for better solubilization and adding ~2-5 equivalents of N-hydroxysuccinimide and ~2-10 equivalents of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide (EDCI) as the hydrochloride salt. The reaction is allowed to proceed at ambient temperature until complete as measured by HPLC or alternately by TLC, which is usually 1 to 8 hours. The solvents are then removed and the crude product is purified on an appropriate silica gel, such as Biosil-A, using an appropriate solvent system, such as a gradient of 0 to 20~ methanol in either chloroform or ethyl acetate. The products are then sufficiently pure for the conjugation step.

4-Formylphenoxyacetic acid (1) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 6.5 min.
EXAMPT.E 63 4-Formylbenzoic acid (2) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 6.6 min, FAB MS: 1708 (M+H), W max at 310 nm (acetonitrile).

4-Formyl-3-methoxyphenoxyacetic acid (3) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.0 min.
FAB MS: 1768 (M+H), W max at 279, 288, and 320 nm (acetonitrile).
F.X~MPT .F~ 65 6-Formyl-2-naphthoic acid (4) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.4 min, FAB MS: 1758 (M+H), W max at 272 and 323 nm (ethanol).
FXi~MPrE 66 4-(2-Oxoethoxy)benzenepropanoic acid (5) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.1 min.
FAB MS: 1766 (M+H), W - no maxima.
~X~MPLE 67 3-(2-Oxoethoxy)benzoic acid (6) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.7 min, W - no maxima.
F.XAMPT.F. 68A
4-(4-Acetylphenoxy)butanoic acid (7) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.5 min, FAB MS: 1780 (M+H), W max at 283 nm (acetonitrile), lH-NMR: See Fig. VI.

-4-(4-Acetylphenoxy)butanoic acid (7) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, (1-hydroxy-2,5-dioxo-3-pyrrolidinesulfonic acid, monosodium salt) ester (i.e. ester with ~sulfonato-N-hydroxysuccimide~
HPLC retention time: 5.2 min, W max at 278 nm (ethanol).
F.X~MPLE 69 4-(4-Acetylphenoxy)butanoic acid (7) condensed with calicheamicin gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.6 min, w max at 283 nm (acetonitrile).
FxAMPLE 70 4-(3-Formylphenoxy)butanoic acid (8) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.4 min, EAB MS: 1766 (M+H), W max at 283 nm (acetonitrile).
F.XAMpLE 71 4-(4-Formylphenoxy)butanoic acid (9) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.0 min, EAB MS: 1766 (M+H) t W max at 289 nm (acetonitrile).
o F.XAMPT.F 72 4-(4-Acetyl-2-methylphenoxy)butanoic acid (10) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 8.2 min, FAB MS: 1794 (M+H).

_- 70 F.XAMpT.F. 73 4-(4-Formyl-2-methoxyphenoxy)butanoic acid (11) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 6.6 min, FAB MS: 17 96 ( M+H).
EX~MPLE 7 4 4-Formylbenzenepropanoic acid (12) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC ~etention time: 6. 7 min, FAB MS: 173 6 (M+H).

lS 4-(2, 3 -Dimethoxy-5-formylphenoxy)butanoic acid (13) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 6. 7 min, FAB MS: 1826 (M+H), W max at 298 nm (ethanol).
F.XAMPT.F 76 4-(4-Acetyl-2,6-dimethoxyphenoxy)butanoic acid (14) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.7 min, -FAB MS: 1840 (M+H), W max at 286 nm (acetonitrile).
EX~MPLE 77 4-(4-Acetyl-2-methoxyphenoxy)butanoic acid (15) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7. 2 min, FAB MS: 1810 (M+H), W max at 284 nm (acetonitrile).

_ 71 ` 21~0785 FX~MPTF 78 4-[4-(3-Oxobutyl)phenoxy]butanoic acid (1) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.9 min, FAs MS: 1808 (M+H), W - no maxima.
F.XZ~MPT.F. 79 4-~2-Acetyl-5-methoxyphenoxy]butanoic acid (17) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.4 min, FAs MS: 1810 (M+H), W - no maxima.
F.X ;~MPT .F. 80 4-[4-(3-Oxopropyl)phenoxy]butanoic acid (18) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 13.1 min, FAs MS: 1794 (M+H), W - no maxima.
F.XAMPT.F. 81 4-Acetylbenzenebutanoic acid (19) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.7 min.
F.XAMPT .F. 82 4-[(2-Acetyl-1-naphthalenyl)oxy] butanoic acid (20) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 9.4 min, FAs MS: 1830 (M+H), W - no maxima.

215078~

4-[4-(4-Fluorobenzoyl)phenoxy]butanoic acid (21) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 9.3 min, FAB MS: 1860 (M+H), W max at 284 nm (ethanol).

4-(4-Acetylphenyl)-1-piperazinepentanoic acid (22) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 6.3 min, FAB MS: 1863 (M+.H), W max at 306 nm (1:1 acetonitrile/chloroform).
~X~MPLE 85 11-(4-Acetylphenoxy)undecanoic acid (23) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 15.5 min, FAB MS: 1878 (M+H), W max at 284 nm (ethanol).
FXi~PT.F. 86 5-[(4-Acetylphenyl)amino]-5-oxopentanoic acid (24) 2S condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 6.2 min, FAB MS: 1807 (M+H), UV max at 292 nm (acetonitrile~.
~X~MPLF. 87 4-(2-Chloro-4-formylphenoxy)butanoic acid (25) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.5 min, FAB MS: 1800 (M+H), W max at 290 nm (acetonitrile).

21507~5 F.X~MPT.F. 88 5-Acetyl-2-(3-carboxypropoxy)benzoic acid (26), methyl ester condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.2 min, FAB MS: 1838 (M+H), W max at 284 nm (acetonitrile).

4-(4-Formyl-2-nitrophenoxy)butanoic acid (27) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7.1 min, FAs MS: 1811 (M+H), W max at 293 nm (ethanol).

4-[2-[[(4-Acetylphenyl)amino]methyl]-6-methoxyphenoxy]butanoic acid (28) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 9.2 min, FAB MS: 1916 (M+H), W max at 309 nm (acetonitrile).
F.X~MPT.E 9 1 4-(4-Acetyl-3-fluorophenoxy)butanoic acid (30) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N- -hydroxysuccimide ester.
HPLC retention time: 8.2 min, FAB MS: 1798 (M+H), W max at 270 nm (ethanol).

4-(2-Acetylphenoxy)butanoic acid (31) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 8.1 min, EAB MS: 1780 (M+H), w - no maxima.

2-Acetyl-lOH-phenothiazine-10-hexanoic acid (32) condensed with Calicheamicin N-acetyl gamma dimethyl s hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 8. 3 min, FAB MS : 193 0 (M+NH4), W max at 281 nm (acetonitrile).
E~AMPT.~ 94 4-Acetylphenylacetic acid (33) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide, N-hydroxysuccimide ester.
HPLC retention time: 7 . 2 min, FAB MS : 1736 (M+H), W max at 280 nm (acetonitrile).

SYNTHESIS OF STRUCTURES D (Method A-Scheme 1) General Procedure The activated ester from above is dissolved in an appropriate organic solvent, such as dimethylformamide, and added to a solution of antibody at ~1-15 mg/mL in an appropriate buffer, such as pH 7.4 phosphate (50 mM, 100 mM salt) such that the concentration of organic co-solvent is ~10-30% and ~2-10 equivalents of active ester are used per mole of antibody. The conjugation reaction is allowed to proceed at ambient temperature for ~4-24 hours. The solution is concentrated by use of a semipermeable membrane, if necessary, and purified by standard size-exclusion chromatography, such as with Sephacryl S-200 gel. The monomer fractions are pooled and the loading of drug on the antibody is estimated by W -VIS absorbence at 280 nm for antibody and 333 nm or other appropriate wavelength for the calicheamicin hydrazones.

SYNT~F.~IS OF STRUCTU~ E (Scheme 2) General Procedure The carboxylic acids of the spacers are activated as the OSu esters (Z3=N-succinimidyloxy) by dissolving them in an appropriate solvent such as tetrahydrofuran containing 10-20% dimethylformamide and adding ~2-3 equivalents of N-hydroxysuccinimide and ~2-5 equivalents of l-t3-dimethylaminopropyl)-3-ethylcarbodiimide (EDCI) as the hydrochloride salt. The reaction is allowed to proceed at ambient temperature until complete as assessed by TLC, which is usually 1 to 8 hours. The solvents are then removed and the crude product is purified on an appropriate silica gel, such as Biosil-A, using an appropriate solvent system, such as a gradient of 0 to 5% methanol in chloroform. The products are generally purified further by recrystallization from a mixture of ethyl acetate-hexanes or other appropriate solvents.
The following preparations were made by the above procedure:
(SuOH = N-hydroxysuccinimide) 4-Formylphenoxy acetic acid (1), N-hydroxysuccinimide ester.
CI MS: 278 (MH+), NMR (CDCl3+D6-DMSO): 9.9 ppm (lH, s, CH=O), 7.9 and 7.1 (2H eachi d, ArH), 5.2 (2H, s, OCH2), 2.9 (4H, s, CH2cH2).

4-Formyl-3-methoxyphenoxy acetic acid (3), N-hydroxysuccinimide ester.
CI MS: 308 (MH+), NMR (CDC13): 10.3 ppm (lH, s, CH=O), 7.8 (lH, d, ArH), 6.6 (lH, dt, ArH), 6.55 (lH,-d, ArH), 5.1 (2H, s, OCH2), 3.95, (3H, s, OCH3), 2.9 (4H, s, CH2CH2 ) --~ - 2 1~ 0 7 8 S 76 4-(4-Acetylphenoxy)butanoic acid (7), N-hydroxysuccinimide ester.
CI MS: 320 (MH+), NMR (CDCl3): 7.9 and 7.0 (2H each, d, ArH), 4.2 (2H, s, OCH2), 2.9 (6H, m, CH2CH2 + O=CCH2), 2.6 (3H, s, O=CCH3), 2.3 (2H, m, CH2).

SYNTH~sIs OF STRUCTURES F (Scheme 2) General Procedure The activated ester from above is dissolved in an appropriate organic solvent, such as N,N-dimethylformamide, and added to a solution of antibody at ~1-15 mg/mL in an appropriate buffer, such as pH 7.4 phosphate (50 mM, 100 mM salt) such that the concentration of organic co-solvent is ~10-25% and ~2-20 equivalents of active ester are used per mole of antibody. The conjugation reaction is allowed to proceed at ambient temperature for ~4-24 hours. The buffer is exchanged and the organic co-solvents and by-products are removed by use of a desalting column such as a PD-10 using pH 5.5 acetate buffer (25 mM acetate, 100 mM NaCl). The solution is concentrated by use of a semipermeable membrane, if necessary, and the product is used without further purification for the following step. The number of carbonyl groups incorporated per antibody is usually about half the number of equivalents of OSu ester used and can be further quantified by use of p-nitrophenyl hydrazine or other comparable method, if desired.

SYNTHESIS OF STRUCTURES D (Method B-Scheme 2) General Procedure The drug hydrazide derlvative is dissolved in an appropriate organic solvent, such as N,N-_ 77 dimethylformamide, and added to a solution of antibody-linker conjugate (structure F) from the previous step at -1-15 mg/mL in an appropriate buffer, such as pH acetate (25 mM, 100 mM salt) such that the concentration of organic co-solvent is ~10-15% and ~2-15 equivalents of hydrazide are used per mole of antibody. The conjugation reaction is allowed to proceed at ambient temperature for ~4-24 hours. The buffer is exchanged and the organic co-solvents and by-products are removed by use of a desalting column such as a PD-10 using pH
7.4 buffer (50 mM phosphate, 100 mM NaCl). The solution is concentrated by use of a semipermeable membrane, if necessary, and purified by standard size-exclusion chromatography, such as with Sephacryl S-200 gel. The monomer fractions are pooled and the loading of drug on the antibody is estimated by W -VIS absorbence at 280 nm for antibody and 333 nm or other appropriate wavelength for the calicheamicin hydrazones.
EXAMPTF. 98 (~THOD A AND B) Conjugate of 4-formylphenoxyacetic acid (1) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.0 M/M, Rel. Affinity: 0.65, In vitro ICso: 0.23 ng/mL, Spec. Index: 1,600, In vivo: 29% T/C (2 ~g x 3 doses -- 5/5 alive-28d), Ex vi vo: 9 5% inhibition.
EXAMPT.F 99 (MFTHOD A AND B) Conjugate of 4-formylphenoxyacetic acid (1) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.5 M/M, Rel. Affinity: 0.77, In vitro ICso: 0.068 ng/mL, Spec. Index: 3,600, Ex vivo: 90% inhibition.

_ 78 21S0~8~

FXAMPLE 100 (METHOD A) Conjugate of 4-formylphenoxyacetic acid (1) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-CT-M-01.
Loading: 2.0 M/M, Rel. Affinity: 0.84, In vitro ICso: 1.5 ng/mL, Spec. Index: 59.
FX~MPLE 101 (MFTHOD A) Conjugate of 4-formylbenzoic acid (2) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 4.8 M/M, Rel. Affinity: 0.99, In vitro ICso: 4.8 ng/mL, Spec. Index: >125, Ex vivo: 63% inhibition.
EXAMPLE 102 (METHOD A) Conjugate of 4-formylbenzoic acid (2) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 4.0 M/M, Rel. Affinity: 1.05, In vitro ICso: 4.0 ng/mL, Spec. Index: >125.
EXAMPLE 103 (METHOD A) Conjugate of 4-formylbenzoic acid (2) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-CT-M-01.
Loading: 2.3 M/M, Rel. Affinity: 0.90, In vitro ICso: 5.6 ng/mL, Spec. Index: 32.
FXAMPLE 104 (MFTHOD A ~D B) Conjugate of 4-formyl-3-methoxyphenoxyacetic acid (3) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 0.8 M/M, Rel. Affinity: 0.81, In vitro ICso: 0.30 ng/mL, Spec. Index: 375.
EXAMPTF 105 (METHOD A AND B) Conjugate of 4-formyl-3-methoxyphenoxyacetic acid (3) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 0.9 M/M, - Rel. Affinity: 0.76, In vitro ICso: 0.12 ng/mL, Spec. Index: 1,200, Ex vi vo: 90% inhibition.
EXAMPLE 106 (MF.THOD A) Conjugate of 4-formyl-3-methoxyphenoxyacetic acid tl) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-CT-M-01.
Loading: 2.1 M/M, Rel. Affinity: 0.88, In vitro ICso: 5.6 ng/mL, Spec. Index: 12.
FX~MpTF 107 (MFTHOD A) Conjugate of 6-formyl-2-naphthoic acid (4) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 2.0 M/M, Rel. Affinity: 0.73, In vitro ICso: 0.047 ng/mL, Spec. Index: 675.
EXAMPLE 10~ ~METHOD A) Conjugate of 4-(2-oxoethoxy)benzenepropanoic acid (5) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.1 M/M, Rel. Affinity: 1.09, In vitro IC~o: 2.22 ng/mL, Spec. Index: 125.
EXAMPLE 109 (METHOD A) Conjugate of 4-(2-oxoethoxy)benzenepropanoic acid (5) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 0.6 M/M, Rel. Affinity: 1.11, In vitro ICso: 0.45 ng/mL, Spec. Index: 200, Ex vivo: 71% inhibition.
FXAMPLE 110 (MFTHOD A) Conjugate of 3-(2-oxoethoxy)benzoic acid (6) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.3 M/M, Rel. Affinity: 1.19, In vitro ICso: 0.69 ng/mL, Spec. Index: 100.

-_ 80 ` 215078~

FX~MpT.F. 111 (MFTHOD A) Conjugate of 3-(2-oxoethoxy)benzoic acid (6) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 0.8 M/M, Rel. Affinity: 1.91, In vitro ICso: 0.32 ng/mL, Spec. Index: 175.
EX~MPLE 112 (MFTHOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.7 M/M, Rel. Affinity: 0.75, In vitro ICso: 0.098 ng/mL, Spec. Index: 6,400, In vivo: 0% T/C (1 ~g x 3 doses, 5/5 alive-28d), 0% T/C (3 ~g x 3 doses, 5/5 alive-28d), 0% T/C (6 ~g x 3 doses, 5/5 alive-28d), Ex vivo: 96% inhibition.
FX~Mp~E 113 (MFTHOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with calicheamicin gamma dimethyl hydrazide and h-P67.6.
Loading: 3.2 M/M, Rel. Affinity: 0.78, In vitro ICso: 0.001 ng/mL, Spec. Index: 10,000 F.XAMPT.~ 114 (MFTHOD A OR B) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.7 M/M, Rel. Affinity: 0.96, In vitro ICso: 0.017 ng/mL, Spec. Index: 29,500, In vivo: 22% T/C (0.5 ~g x 3 doses, 5/5 alive-28d), 0% T/C ~1 ~g x 3 doses, 5/5 alive-28d), 1% T/C (3 ~g x 3 doses, 5/5 alive-28d), 0% T/C t6 ~g x 3 doses, 2/5 alive-28d), Ex vivo: 98% inhibition.

F.XAMPT .~ 115 (M~THOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with Calicheamicin N-acetyl gamma dimethyl s hydrazide and h-CT-M-01.
Loading: 3.4 M/M, Rel. Affinity: not determined, In vitro ICso: 0.048 ng/mL, Spec. Index: 6,200.
FXAMpTF 116 (METHOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with calicheamicin N-acetyl gamma dimethyl hydrazide and m-A33.
Loading: 1.1 M/M, Rel. Affinity: 0.68, In vitro ICso: 3.32 ng/mL, Spec. Index: 0.72.
In vivo: 4% T/C (3 ~g x 3 doses, 5/5 alive-28d), 5% T/C (6 ~g x 3 doses, 5/5 alive-28d).
EXAMPTF 117 (MFTHOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with calicheamicin N-acetyl gamma dimethyl hydrazide and h-A33.
Loading: 1.8 M/M, Rel. Affinity: 1.13, In vitro ICso: 4.03 ng~mL, Spec. Index: 0.96.
EX~MPT .F. 118 (METHOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with calicheamicin gamma dimethyl hydrazide and h-A33.
Loading: 2.8 M/M, Rel. Affinity: 0.91, In vitro ICso: 3.55 ng/mL, Spec. Index: 2.6.
EXAMPTF 119 (METHOD A) Conjugate of 4-(4-acetylphenoxy)butanoic acid (7) condensed with calicheamicin N-acetyl gamma dimethyl hydrazide and anti-Tac.
Loading: 2.1 M/M, Rel. Affinity: not determined, In vitro ICso: 0.004 ng/mL, Spec. Index: 250, Ex vivo: ICso:1.0 ng/mL, Spec. Index: 100.

-EXAMPTF 120 (MFTHOD A) Conjugate of 4-(3-formylphenoxy)butanoic acid (8) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.7 M/M, Rel. Affinity: 1.00, In vitro ICso: 0.38 ng/mL, Spec. Index: 1,700.
EXAMPLE 121 (MFTHOD A) Conjugate of 4-(4-formylphenoxy)butanoic acid (9) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.8 M/M, Rel. Affinity: 0.56, In vitro ICso: 0.52 ng/mL, Spec. Index: 2,900, In vivo: 12% T/C (1 ~g x 3 doses, 5/5 alive-28d), 9% T/C (3 ~g x 3 doses, 5/5 alive-28d), 3% T/C (6 ~g x 3 doses, 4/5 alive-28d), Ex vi vo: 9 8% inhibition.
EXAMPLE 122 (MFTHOD A) Conjugate of 4-(4-formylphenoxy)butanoic acid (9) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.8 M/M, Rel. Affinity: 0.70, In vitro ICso: 0.087 ng/mL, Spec. Index: 11,000, In vivo: 17% T/C (0.5 ~g x 3 doses, 5/5 alive-28d), 23% T/C (1 ~g x 3 doses, 5/5 alive-28d), 9% T/C (3 ~g x 3 doses, 5/5 alive-28d), 0% T/C (6 ~g x 3 doses, 5/5 alive-28d).
EXAMPLE 123 (~FTHOD A) Conjugate of 4-(4-acetyl-2-methylphenoxy)butanoic acid (10) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 3.5 M/M, Rel. Affinity: 1.16, In vitro ICso: 0.45 ng/mL, Spec. Index: 2,900.
EXAMPLE 124 (METHOD A) Conjugate of 4-(4-acetyl-2-methylphenoxy)butanoic acid (10) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.

- 21aO78S

Loading: 1.6 M/M, Rel. Affinity: 1.07, In vitro ICso: 0.041 ng/mL, Spec. Index: 5,100.
FX~MpLE 125 (MFTHOD A) Conjugate of 4-(4-formyl-2-methoxyphenoxy)butanoic acid (11) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.6 M/M, Rel. Affinity: 0.73, In vitro ICso: 3.8 ng/mL, Spec. Index: 575.
EXAMPLE 126 (METHOD A) Conjugate of 4-(4-formyl-2-methoxyphenoxy)butanoic acid (11) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.9 M/M, Rel. Affinity: 0.22, In vitro ICso: 0.13 ng/mL, Spec. Index: 1,800.
EXAMPLE 127 (METHOD A) Conjugate of 4-formylbenzenepropanoic acid (12) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.5 M/M, Rel. Affinity: 0.73, In vi~ro ICso: 1.0 ng/mL, Spec. Index: 950.
EXAMPLE 128 (METHOD A) Conjugate of 4-formylbenzenepropanoic acid (12) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.6 M/M, Rel. Affinity: 0.73, In vitro ICso: 0.12 ng/mL, Spec. Index: 2,000.
EXAMPLE 129 (METHOD A) Conjugate of 4-(2,3-dimethoxy-5-formylphenoxy)butanoic acid (13) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.0 M/M, Rel. Affinity: 1.16, In vitro ICso: 1.1 ng/mL, Spec. Index: >375.
EXAMPLE 130 (M~THOD A) Conjugate of 4-(2,3-Dimethoxy-5-formylphenoxy)butanoic acid (13) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.

-_ 84 Loading: 1.8 M/M, Rel. Affinity: 1.08, In vitro ICso: 0.062 ng/mL, Spec. Index: >9,800.
EXAMPTF 131 (METHOD A) Conjugate of 4-(4-acetyl-2,6-dimethoxyphenoxy)butanoic acid (14) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.6 M/M, Rel. Affinity: 1.07, In vitro ICso: 0.24 ng/mL, Spec. Index: >1,700.
EXAMPLE 132 (METHOD A) Conjugate of 4-(4-acetyl-2,6-dimethoxyphenoxy)butanoic acid (14) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.7 M/M, Rel. Affinity: 1.18, In vitro ICso: 0.015 ng/mL, Spec. Index: >40,500.
FXAMPLE 133 (METHOD A) Conjugate of 4-(4-acetyl-2-methoxyphenoxy)butanoic acid (15) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6-.
Loading: 2.3 M/M, Rel. Affinity: 0.78, In vitro ICso: 0.23 ng/mL, Spec. Index: 875.
EXAMPLE 134 (METHOD A) Conjugate of 4-(4-acetyl-2-methoxyphenoxy)butanoic acid (15) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.7 M/M, Rel. Affinity: 0.80, In vitro ICso: 0.029 ng/mL, Spec. Index: 13,500.
EX~MPT .F. 135 (METHOD A) Conjugate of 4-[4-(3-oxobutyl)phenoxy]butanoic acid tl6) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 0.5 M/M, Rel. Affinity: not determined, In vitro ICso: 9 ng/mL, Spec. Index: 2.
EXAMPLE 136 (METHOD A) Conjugate of 4-(2-acetyl-5-methoxyphenoxy]butanoic acid (17) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.

-_ 85 Loading: 2.3 M/M, Rel. Affinity: 0.98, In vitro ICso: 0.088 ng/mL, Spec. Index: 1,100.
EXAMPLE 137 (METHOD A) Conjugate of 4-(2-acetyl-5-methoxyphenoxy]butanoic acid (17) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.6 M/M, Rel. Affinity: 1.20, In vitro ICso: 0.0098 ng/mL, Spec. Index: 21,500.
EXAMPLE 138 (METHOD A) Conjugate of 4-[4-(3-oxopropyl)phenoxy]butanoic acid (18) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.0 M/M, Rel. Affinity: 0.80, In vitro ICso: 1.1 ng/mL, Spec. Index: 80.
EXAMPLE 139 (METHOD A) Conjugate of 4-[4-(3-oxopropyl)phenoxy]butanoic acid (18) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 0.6 M/M, Rel. Affinity: 1.21, In vitro ICso: Q.62 ng/mL, Spec. Index: 90.
EXAMPLE 140 (MFTHOD A) Conjugate of 4-acetylbenzenebutanoic acid (19) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.6 M/M, Rel. Affinity: 0.50, In vitro ICso: 0.012 ng/mL, Spec. Index: 2,600, In vivo: 23% T/C (0.5 ~g x 3 doses, 5/5 alive-28d), 10% T/C (1 ~g x 3 doses, 5/5 alive-28d), 4~ T/C (3 ~g x 3 doses, 4/5 alive-28d), 0~ T/C (6 ~g x 3 doses, 2/5 alive-28d), Ex vivo: 99% inhibition.
~xAMpr .F 141 (MFTHOD A) Conjugate of 4-acetylbenzenebutanoic acid (19)- condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 2.2 M/M, Rel. Affinity: 0.42, In vitro ICso: 0.0082 ng/mL, Spec. Index: 31,500, In vivo: 21% T/C (0.5 ~g-x 3 doses, 5/5 alive-28d), 25% T/C (1 ~g x 3 doses, 5/5 alive-28d), 0% T/C (3 ~g x 3 doses, 4/5 alive-28d), 0% T/C (6 ~g x 3 doses, 1/5 alive-28d), Ex vivo: 9 9 % inhibition.
EXAMPTF 142 (~ETHQD A) Conjugate of 4-[(2-acetyl-1-naphthalenyl)oxy] butanoic acid (20) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.5 M/M, Rel. Affinity: 0.50, In vitro ICso: 0.061 ng/mL, Spec. Index: 5,000, In vivo: 36% T/C (1 ~g x 3 doses, 5/5 alive-28d), 15- - 22% T/C (3 ~g x 3 doses, 5/5 alive-28d), 11% T/C (6 ~g x 3 doses, 4/5 alive-28d), Ex vivo: 76% inhibition.
EXAMPT F 143 (METHOD A) Conjugate of 4-[(2-acetyl-1-naphthalenyl)oxy] butanoic acid (20) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 1.8 M/M, Rel. Affinity: 0.66, In vitro ICso: 0.0067 ng/mL, Spec. Index: 105,000.
EXAMPLE 144 (METHOD A) Conjugate of 4-[4-(4-fluorobenzoyl)phenoxy]butanoic acid (21) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.5 M/M, Rel. Affinity: 0.67, In vitro ICso: 99 ng/mL, Spec. Index: 3.
EXAMPLE 145 (METHOD A) Conjugate of 4-[4-(4-fluorobenzoyl)phenoxy]butanoic acid (21) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.8 M/M, Rel. Affinity:` 0.76, In vitro ICso: 63 ng/mL, Spec. Index: 9.

~ 87 a 1 5~85 EXAMPLE 146 (METHOD A) Conjugate of 4-(4-acetylphenyl)-1-piperazinepentanoic acid (22) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 0.1 M/M, Rel. Affinity: 0.98, In vitro ICso: 12 ng/mL, Spec. Index: 2.
~X~MPT .~ 147 (~T~OD A) Conjugate of 11-(4-acetylphenoxy)undecanoic acid (23) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 0.5 M/M, Rel. Affinity: 0.80, In vitro ICso: 0.43 ng/mL, Spec. Index. 175.
FX~MPLE 148 ~MFT~OD A) Conjugate of 11-(4-acetylphenoxy)undecanoic acid (23) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 0.4 M/M, Rel. Affinity: 1.16, In vitro ICso: 0.47 ng/mL, - Spec. Index: 125.
EXAMPLE 149 (M~THOD A) Conjugate of 5-[(4-acetylphenyl)amino]-5-oxopentanoic acid (24) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and m-P67.6.
Loading: 2.0 M/M, Rel. Affinity: 0.73, In vitro ICso: c0.005 ng/mL, Spec. Index: >1,200.
EXAMPLE 150 (MFTHOD A) Conjugate of 4-(2-chloro-4-formylphenoxy)butanoic acid (25) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.0 M/M, Rel. Affinity: 0.31, In vitro ICso: 0.0071 ng/mL, Spec. Index: 1,500.
EXAMPLE 151 (M~THOD A) Conjugate of 5-acetyl-2-(3-carboxypropoxy)benzoic acid (26), methyl ester condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.0 M/M, Rel. Affinity: 0.79, In vitro ICso: <0.005 ng/mL, Spec. Index: >9,600.

- 215078~

EXAMPLE 152 (METHOD A) Conjugate of 4-(4-formyl-2-nitrophenoxy)butanoic acid (27) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.5 M/M, Rel. Affinity: 1.3, In vitro ICso: 0.023 ng/mL, Spec. Index: >4,500.
FXAMPLE 153 (METHOD A) Conjugate of 4-[2-[[(4-acetylphenyl)amino]methyl]-6-methoxyphenoxy]butanoic acid (28) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 2.0 M/M, Rel. Affinity: 0.85, In vitro ICso: <0.005 ng/mL, Spec. Index: >5,000.
FX~MPTF 154 (M~THnD A) Conjugate of 4-(4-acetyl-3-fluorophenoxy)butanoic acid (30) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.5 M/M, Rel. Affinity: 1.01, In vitro ICso: 0.005 ng/mL, Spec. Index: 4,800.
EXAMPLE 155 (METHOD A) Conjugate of 4-(2-Acetylphenoxy)butanoic acid (31) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.5 M/M, Rel. Affinity: 0.95, In vitro ICso: <0.005 ng/mL, Spec. Index: >7,000.
EXAMPTF 156 (MFTHOD A) Conjugate of 2-acetyl-lOH-phenothiazine-10-hexanoic acid (32) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.5 M/M, Rel. Affinity: 1.25, In vitro ICso: 0.021 ng/mL, Spec. Index: 2,300.
EXAMPLE 157 (METHOD A) Conjugate of 4-acetylphenylacetic acid (33) condensed with Calicheamicin N-acetyl gamma dimethyl hydrazide and h-P67.6.
Loading: 1.4 M/M, Rel. Affinity: 0.91, In vitro ICso: <0.005 ng/mL, Spec. Index: 4,700.

The described conjugates are useful for inhibiting the growth of unwanted cells which is an important part of the invention. Accordingly, the invention also includes pharmaceutical compositions, most preferably a parenteral composition suitable for injection into the body of a warm-blooded m~mm~l. Such compositions are formulated by methods which are commonly used in pharmaceutical chemistry. The conjugates are acceptably soluble in physiologically-acceptable fluids, such as physiological saline solutions and other aqueous solutions which can safely be administered parenterally.
Products for parenteral administration are often formulated and distributed in solid, preferably freeze-dried form, for reconstitution immediately before use.
Such formulations are useful compositions of the present invention. Their preparation is well understood by pharmaceutical chemists; in general, they comprise mixtures of inorganic salts, to confer isotonicity, and dispersing agents, such as sucrose, to allow the dried preparation to dissolve quickly upon reconstitution.
Such formulations are reconstituted with highly purified water or physiologically acceptable buffers to a known concentration, based on the drug.
The optimum dosage and administration schedule of conjugates of the invention must be determined by the treating physician, in light of the patient's condition.
It is customary, of course, to administer cytotoxic drugs in the form of divided doses, with intervals of days or weeks between each series of doses.
~ The conjugates are effective over a wide dosage range, and dosages per week will usually fall within the range from about 1 to about 10,000 ~g/m2 of drug, more - ~150785 90 -preferably in the range from about 10 to about 200 ~g/m2 .

Claims (44)

1. A cytotoxic drug conjugate of formula:
Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m wherein Z3 is a protein selected from mono- and polyclonal antibodies, their antigen-recognizing fragments, and their chemically or genetically manipulated counterparts and/growth factors and their chemically or genetically manipulated counterparts, wherein a covalent bond to the protein is an amide formed from reaction with "m" lysine side chains, or a steroid, wherein the covalent bond to the steroid is an amide or an ester;
Alk1 and Alk2 are independently a bond or branched or unbranched (C1-C10) alkylene chain;
Sp1 is a bond, -S-, -O-, -CONH-, -NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar'-Y-(CH2)n-Z wherein X, Y, and Z are independently a bond, -NR'-, -S-, or -O-, with the proviso that when n = 0, then at least one of Y
and Z must be a bond and Ar' is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR', with the proviso that when Alk1 is a bond, SP1 is a bond;
n is an integer from 0 to 5;
R' is a branched or unbranched (C1-C5 ) chain optionally substituted by one or two groups of -OH, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, (C1-C3) dialkylamino, or (C1-C3) trialkylammonium -A- where A- is a pharmaceutically acceptable anion completing a salt;
Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1 or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or , each naphthylidene or phenothiazine optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1, with the proviso that when Ar is naphthylidene, Z1 is not hydrogen and with the proviso that when Ar in phenothiazine, Sp1 is a bond only connected to nitrogen;
Sp2 is a bond, -S-, or -O-, with the proviso that when Alk2 is a bond, Sp2 is a bond;
Z1 is H, (C1-C5) alkyl, or phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1;
Z2 is Q-Sp-SS-W, wherein W is or CH3; R2 is or H;

or H; or H;
R6 or R7 is H or ;

R5 is -CH3, -C2H5, or -CH (CH3)2; X is an iodine or bromine atom; R5' is a hydrogen or the group RCO, wherein R is hydrogen, branched or unbranched (C1 -C10) alkyl or (C1 - C10) alkylene group, a (C6-C11) aryl group, a (C6-C11) aryl-alkyl (C1 - C5) group, or a heteroaryl or heteroaryl-alkyl (C1 - C5) group wherein heteroaryl is 2- or 3-furyl, 2- or 3-thienyl,
2- or 3-(N-methylpyrrolyl), 2-, 3-, or 4-pyridyl, 2-, 4-, or 5-(N-methylimidizolyl), 2-, 4-, or 5-oxazolyl, 2-, 3-, 5-, or 6-pyrimidinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolyl, or 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolyl, all aryl and heteroaryl optionally substituted by one or more hydroxy, amino, carboxy, halo, nitro, lower (C1 - C3) alkoxy, or lower (C1 -C5) thioalkoxy groups;
Sp is a straight or branched-chain divalent or trivalent (C1 - C18) radical, divalent or trivalent aryl or heteroaryl radical, divalent or trivalent (C3 - C18) cycloalkyl or heterocycloalkyl radical, divalent or trivalent aryl- or heteroaryl-alkyl (C1 - C18) radical, divalent or trivalent cycloalkyl- or heterocycloalkyl-alkyl (C1 - C18) radical or divalent or trivalent (C2 - C18) unsaturated alkyl radical, wherein heteroaryl is furyl, thienyl, N-methylpyrrolyl, pyridinyl, N-methylimidizolyl, oxazolyl, pyrimidinyl, quinolyl, isoquinolyl, N-methylcarbazoyl, aminocoumarinyl, or phenazinyl and wherein if Sp is a trivalent radical, Sp can be additionally substituted by lower (C1 - C5) dialkylamino, lower (C1 - C5) alkoxy, hydroxy, or lower (C1 - C5) alkylthio groups; and Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NHO-m is an integer from about 0.1 to 15.
2. A cytotoxic drug conjugate according to claim 1 wherein Alk2 is a branched or unbranched (C1-C10) alkylene chain and Z1 is phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1.
3. A cytotoxic drug conjugate according to claim 1 wherein Alk2 is a branched or unbranched (C1-C10) alkylene chain and Z1 is H or (C1-C5) alkyl.
4. A cytotoxic drug conjugate according to claim 1 wherein Alk2 and Sp2 are together a bond and Z1 is H or (C1-C5) alkyl.
5. A cytotoxic drug conjugate according to claim 1 wherein Alk2 and Sp2 are together a bond and Z1 is phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1.
6. A cytotoxic drug conjugate according to claim 2, claim 3, claim 4, or claim 5 wherein Sp1 is a bond, -S-, -O-, -CONH-, - NHCO-, or -NR' wherein n and R' are as defined in claim 1-, with the proviso that when Alk1 is a bond, Sp1 is a bond.
7. A cytotoxic drug conjugate according to claim 6 wherein Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1 or Ar is a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene each optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1;
8. A cytotoxic drug conjugate according to claim 7 wherein Z3 is a protein selected from mono- and polyclonal antibodies, their antigen-recognizing fragments, and their chemically or genetically manipulated counterparts and growth factors and their chemically or genetically manipulated counterparts, wherein a covalent bond to the protein is an amide formed from reaction with "m" lysine side chains.
9. A cytotoxic drug conjugate according to claim 8 wherein Z2 is Q-Sp-SS-W, wherein W is R1 is ; R2 is or H;

R4 is or H;

R5, X, R5', R, and Sp are as defigned in claim 1; and Q is =NHNCO-.
10. A cytotoxic drug conjugate according to claim 9 wherein Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1, Alk2 and Sp2 are together a bond, and Z1 is H or (C1-C5) alkyl.
11. A cytotoxic drug conjugate according to claim 10 wherein Sp1 is -O-, Alk1 is C4 alkyl, Ar is 1,4-phenylene, and Z1 is C1 alkyl.
12. A cytotoxic drug conjugate according to claim 11 wherein Z3 is antibody h- or m-P67.6, h- or m-CT-M-01, h- or m-A33, or anti-Tac and Z2 is calicheamicin gamma dimethyl hydrazide or calicheamicin N-acetyl gamma dimethyl hydrazide.
13. A cytotoxic drug conjugate according to claim 12 wherein Z3 is antibody h-CT-M-01 and Z2 is Calicheamicin N-acetyl gamma dimethyl hydrazide.
14. A cytotoxic drug conjugate according to claim 12 wherein Z3 is antibody h-P67.6 and Z2 is Calicheamicin N-acetyl gamma dimethyl hydrazide.
15. A compound of the formula:

Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m Z3 is halogen, hydroxy, OM wherein M is a metal completing a salt, -N3, , , , , ;
,or Alk1 and Alk2 are independently a bond or branched or unbranched (C1-C10) alkylene chain;
Sp1 is a bond, -S-, -O-, -CONH-, -NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar'-Y-(CH2)n-Z wherein X, Y, and Z are independently a bond, -NR'-, -S-, or -O-, with the proviso that when n = 0, then at least one of Y
and Z must be a bond and Ar' is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR', with the proviso that when Alk1 is a bond, Sp1 is a bond;
n is an integer from 0 to 5;
R' is a branched or unbranched (C1-C5) chain optionally substituted by one or two groups of -OH, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, (C1-C3) dialkylamino, or (C1-C3) trialkylammonium - A- where A- is a pharmaceutically acceptable anion completing a salt;
Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1 or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or each naphthylidene or phenothiazine optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR' or S(CH2)nCONHR' wherein n and R' are as defined in claim 1, with the proviso that when Ar is naphthylidene, Z1 is not hydrogen and with the proviso that when Ar in phenothiazine, Sp1 is a bond only connected to nitrogen;

Sp2 is a bond, -S-, or -O-, with the proviso that when Alk2 is a bond, Sp2 is a bond;
Z1 is H, (C1-C5) alkyl, or phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1;
Z2 is Q-Sp-SS-W, wherein W is R1 is or CH3; R2 is or H;

R3 is or H; R4 is or H;

R6 or R7 is H or ;

R5 is -CH3, -C2H5, or -CH(CH3)2; X is an iodine or bromine atom; R5' is a hydrogen or the group RCO, wherein R is hydrogen, branched or unbranched (C1 -C10) alkyl or (C1 - C10) alkylene group, a (C6-C11) aryl group, a (C6-C11) aryl-alkyl (C1 - C5) group, or a heteroaryl or heteroaryl-alkyl (C1 - C5) group wherein heteroaryl is 2- or 3-furyl, 2- or 3-thienyl, 2- or 3-(N-methylpyrrolyl), 2-, 3-, or 4-pyridyl, 2-, 4-, or 5-(N-methylimidizolyl), 2-, 4-, or 5-oxazolyl, 2-, 3-, 5-, or 6-pyrimidinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolyl, or 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolyl, all aryl and heteroaryl optionally substituted by one or more hydroxy, amino, carboxy, halo, nitro, lower (C1 - C3) alkoxy, or lower (C1 -C5) thioalkoxy groups;
Sp is a straight or branched-chain divalent or trivalent (C1 - C18) radical, divalent or trivalent aryl or heteroaryl radical, divalent or trivalent (C3 - C18) cycloalkyl or heterocycloalkyl radical, divalent or trivalent aryl- or heteroaryl-alkyl (C1 - C18) radical, divalent or trivalent cycloalkyl- or heterocycloalkyl-alkyl (C1 - C18) radical or divalent or trivalent (C2 - C18) unsaturated alkyl radical, wherein heteroaryl is furyl, thienyl, N-methylpyrrolyl, pyridinyl, N-methylimidizolyl, oxazolyl, pyrimidinyl, quinolyl, isoquinolyl, N-methylcarbazoyl, aminocoumarinyl, or phenazinyl and wherein if Sp is a trivalent radical, Sp can be additionally substituted by lower (C1 - C5) dialkylamino, lower (C1 - C5) alkoxy, hydroxy, or lower (C1 - C5) alkylthio groups;
Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NHO-;
and m is 1.
16. A compound according to claim 15 wherein Z3 is hydroxy, , or
17. A compound according to claim 16 wherein Alk2 is a branched or unbranched (C1-C10) alkylene chain and Z1 is phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1.
18. A compound according to claim 17 wherein Alk2 is a branched or unbranched (C1-C10) alkylene chain and Z1 is H or (C1-C5) alkyl.
19. A compound according to claim 18 wherein Alk2 and Sp2 are together a bond and Z1 is H or (C1-C5) alkyl.
20. A compound according to claim 19 wherein Alk2 and Sp2 are together a bond and Z1 is phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1.
21. A compound according to claim 17, claim 18, claim 19, or claim 20 wherein Sp1 is a bond, -S-, -O-, -CONH-, - NHCO-, or -NR' wherein R' is as hereinbefore defined-, with the proviso that when Alk1 is a bond, Sp1 is a bond.
22. A compound according to claim 21 wherein Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1 or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene each optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1;
23. A compound according to claim 22 wherein Z2 is Q-Sp-SS-W, wherein W is ; R2 is or H;
R1 is R4 is or H;

R5, X, R5', R, and Sp are as defined in claim 1; and Q is =NHNCO-.
24. A compound according to claim 23 wherein Alk2 and Sp2 are together a bond, Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1, and Z1 is H or (C1-C5) alkyl.
25. A compound according to claim 24 wherein Sp1 is -O-, Alk1 is C4 alkyl.
Ar is 1,4-phenylene, and Z1 is C1 alkyl.
26. A compound according to claim 25 wherein Z2 is calicheamicin gamma dimethyl hydrazide or calicheamicin N-acetyl gamma dimethyl hydrazide.
27. A compound of the formula:

Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m Z3 is halogen, hydroxy, OM wherein M is a metal completing a salt, -N3, , , , ,or ;

Alk1 and Alk2 are independently a bond or branched or unbranched (C1-C10) alkylene chain;
Sp1 is a bond, -S-, -O-, -CONH-, -NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar'-Y-(CH2)n-Z wherein X, Y, and Z are independently a bond, -NR'-, -S-, or -O-, with the proviso that when n = 0, then at least one of y and z must be a bond and Ar' is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR', with the proviso that when Alk1 is a bond, Sp1 is a bond;
n is an integer from 0 to 5;
R' is a branched or unbranched (C1-C5) chain optionally substituted by one or two groups of -OH, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, (C1-C3) dialkylamino, or (C1-C3) trialkylammonium - A- where A- is a pharmaceutically acceptable anion completing a salt;
Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as hereinbefore defined or Ar is a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or each naphthylidene or phenothiazine optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1, with the proviso that when Ar is naphthylidene, Z1 is not hydrogen and with the proviso that when Ar in phenothiazine, Sp1 is a bond only connected to nitrogen;

Sp2 is a bond, -S-, or -O-, with the proviso that when Alk2 is a bond, Sp2 is a bond;
Z1 is H, (C1-C5) alkyl, or phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 1;
Z2 is oxygen; and m is 1, with the proviso that when Ar is unsubstituted 2,6-naphthylene or 1,3- or 1,4-phenylene optionally substituted with one group of (C1-C6) alkyl or (C1-C5) alkoxy and Alk2 is a bond, then Sp1 is not a bond, -O-, or -NHCO-.
28. A compound according to claim 27 wherein Z3 is hydroxy, , or
29. A compound according to claim 28 wherein Alk2 is a branched or unbranched (C1-C10) alkylene chain and Z1 is phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 27.
30. A compound according to claim 28 wherein Alk2 is a branched or unbranched (C1-C10) alkylene chain and Z1 is H or (C1-C5) alkyl.
31. A compound according to claim 28 wherein Alk2 and Sp2 are together a bond and Z1 is H or (C1-C5) alkyl.
32. A compound according to claim 28 wherein Alk2 and Sp2 are together a bond and Z1 is phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 27.
33. A compound according to claim 29, claim 30, claim 31, or claim 32 wherein Sp1 is a bond, -S-, -O-, -CONH-, - NHCO-, or -NR' wherein R' is as hereinbefore defined-, with the proviso that when Alk1 is a bond, Sp1 is a bond.
34. A compound according to claim 33 wherein Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 27 or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene each optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 27;
35. A compound according to claim 34 wherein Alk2 and Sp2 are together a bond, Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or COOR', CONHR', O(CH2)nCOOR', S(CH2)nCOOR', O(CH2)nCONHR', or S(CH2)nCONHR' wherein n and R' are as defined in claim 27, and Z1 is H or (C1-C5) alkyl.
36. A pharmaceutical composition comprising a conjugate of claim 1 and a parentally-administrable medium.
37. A pharmaceutical composition comprising a conjugate of claim 13 or 14 and a parentally-administrable medium.
38. A method of controlling the growth of an undesirable cell comprising administering a conjugate of claim 1 to a patient.
39. A method of controlling the growth of an undesirable cell comprising administering a conjugate of claim 13 or 14 to a patient.
40. A method of claim 45 or 46 wherein the undesired cells are cancer cells in a mammalian species.
41. A freeze-dried pharmaceutical composition comprising a conjugate of claim 13 or 14 which is obtained by freeze-drying an approximately 1 mg/mL
solution of the conjugate dissolved in about 5 mM
sodium phosphate buffer at a pH of about 7.4 containing about 100 mM sodium chloride and about 100 mM sucrose.
42. A process for preparing the targeted derivatives Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m of compounds of formula Z2, wherein Z2 is Q-Sp-SS-W and W is R1 is or CH3; R2 is or H;

R3 is or H; R4 is or H;

R6 or R7 is H or ;

R5 is -CH3, -C2H5, or -CH (CH3)2; X is an iodine or bromine atom; R5' is a hydrogen or the group RCO, wherein R is hydrogen, branched or unbranched (C1-C10) alkyl or (C1-C10) alkylene group, a (C6-C11) aryl group, a (C6-C11) aryl-alkyl (C1-C5) group, or a heteroaryl or heteroaryl-alkyl (C1-C5) group wherein heteroaryl is 2- or 3-furyl, 2- or 3-thienyl, 2- or 3-(N-methylpyrrolyl), 2-, 3-, or 4-pyridyl, 2-, 4-, or 5-(N-methylimidazolyl), 2-, 4-, or 5-oxazolyl, 2-, 3-, 5-, or 6-pyrimidinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolyl, or 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolyl, all aryl and heteroaryl optionally substituted by one or more hydroxy, amino, carboxy, halo, nitro, lower (C1-C3) alkoxy, or lower (C1-C5) thioalkoxy groups;
Sp is a straight or branched-chain divalent or trivalent (C1-C18) radical, divalent or trivalent aryl or heteroaryl radical, divalent or trivalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent or trivalent aryl- or heteroaryl-alkyl (C1-C18) radical, divalent or trivalent cycloalkyl- or heterocycloalkyl-alkyl (C1-C18) radical or divalent or trivalent (C2-C18) unsaturated alkyl radical, wherein heteroaryl is furyl, thienyl, N-methylpyrrolyl, pyridinyl, N-methylimidazolyl, oxazolyl, pyrimidinyl, quinolyl, isoquinolyl, N-methylcarbazoyl, aminocoumarinyl, or phenazinyl and wherein if Sp is a trivalent radical, Sp may be additionally substituted by lower (C1-C5) dialkylamino, lower (C1-C5) alkoxy, hydroxy, or lower (C1-C5) alkylthio groups; and Q is H2NHNCO-, H2NHNCS-, H2NHNCONH-, H2NHNCSNH-, or H2NO-,comprising reacting Z2 with a compound of formula HOCO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=O, wherein Alk1 and Alk2 are independently a bond or branched or unbranched (C1-C10) alkylene chain;
Sp1 is a bond, -S-, -O-, -CONH-, -NHCO-, -NR'-, -N(CH2CH2)2N-, or -X-Ar'-Y-(CH2)n-Z wherein X, Y, and Z are independently a bond, -NR'-, -S-, or -O-, with the proviso that when n = 0, then at least one of Y
and z must be a bond and Ar' is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, -COOR', -CONHR', -O(CH2)nCOOR', -S(CH2)nCOOR', -O(CH2)nCONHR', or -S(CH2)nCONHR', with the proviso that when Alk1 is a bond, Sp1 is a bond;
n is an integer from 0 to 5;
R' is a branched or unbranched (C1-C5) chain optionally substituted by one or two groups of -OH, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, (C1-C3) dialkylamino, or (C1-C3) trialkylammonium - A- where A- is a pharmaceutically acceptable anion completing a salt;
Sp2 is a bond, -S-, or -O-, with the proviso that when Alk2 is a bond, Sp2 is a bond;
Z1 is H, (C1-C5) alkyl, or phenyl optionally substituted with one, two, or three groups of (C1-C5) alkyl, (C1-C4) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, -COOR', -CONHR', -O(CH2)nCOOR', -S(CH2)nCOOR', -O(CH2)nCONHR', or -S(CH2)nCONHR' wherein n and R' are as hereinbefore defined;

Ar is 1,2-, 1,3-, or 1,4-phenylene optionally substituted with one, two, or three groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, or -COOR', -CONHR', -O(CH2)nCOOR', -S(CH2)nCOOR', -O(CH2)nCONHR', or -S(CH2)nCONHR' wherein n and R' are as hereinbefore defined or a 1,2-, 1,3-, 1,4-, 1,5-, 1,6-, 1,7-, 1,8-, 2,3-, 2,6-, or 2,7-naphthylidene or each naphthylidene or phenothiazine optionally substituted with one, two, three, or four groups of (C1-C6) alkyl, (C1-C5) alkoxy, (C1-C4) thioalkoxy, halogen, nitro, -COOR', -CONHR', -O(CH2)nCOOR', -StCH2)nCOOR', -O(CH2)nCONHR', or -S(CH2)nCONHR' wherein n and R' are as hereinbefore defined, with the proviso that when Ar is naphthylidene, Z1 is not hydrogen and with the proviso that when Ar in phenothiazine, Sp1 is a bond only connected to nitrogen, in an alcoholic solvent with a boiling point of less than about 100°C in the presence of about 5% acetic acid or other similar acid catalyst at about 20° to 70°C for about 1-24 hours and isolating the intermediate of formula HOCO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2, wherein Alk1, Sp1, Ar, Sp2, Alk2, and Z1 are as hereinbefore defined;
Z2 is Q-Sp-SS-W wherein Sp and W are as hereinbefore defined; and Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NO-, then reacting the compound of formula HOCO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2 with N-hydroxysuccinimide, 2, 3, 5, 6-tetrafluorophenol, pentafluorophenol, 4-nitrophenol, 2,4-dinitrophenol, or other suitable activating group in the presence of DCC, EDCI, or other coupling agent in an inert organic solvent such as acetonitrile or acetonitrile containing 5-50% DMF to generate the compound Z3'CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2, wherein Alk1, Sp1, Ar, Sp2, Alk2, and Z1, are as hereinbefore defined;
Z2 is Q-Sp-SS-W wherein Sp and W are as hereinbefore defined;
Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NO-;
and , , , , , , or other comparable acid activating group, and finally reacting the compound Z3'CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2 with a carrier Z3, wherein Z3 is a protein selected from mono- and polyclonal antibodies, their antigen-recognizing fragments, and their chemically or genetically manipulated counterparts and growth factors and their chemically or genetically manipulated counterparts in an aqueous, buffered solution at a pH of between 6.5 and 9.0 and a temperature of 4° to 40°C for 1-48 hours to generate a compound of formula Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m, wherein Alk1, Sp1, Ar, Sp2, Alk2, Z1 and Z3, are as hereinbefore defined;
Z2 is Q-Sp-SS-W wherein Sp and W are as hereinbefore defined;
Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NO-;
and m is an integer from about 0.1 to 15 or by reacting a compound of formula HOCO-Alk1-Sp1-Ar-Sp2-Alk2-C(H)=O, wherein Alk1, Sp1, Ar, Sp2, and Alk2 are as hereinbefore defined, with N-hydroxysuccinimide, 2, 3, 5, 6-tetrafluorophenol, pentafluorophenol, 4-nitrophenol, 2,4-dinitrophenol, of other suitable activating group in the presence of DCC, EDCI, or other coupling agent in an inert organic solvent such as acetonitrile or acetonitrile containing 5-50% DMF to generate the compound Z3'CO-Alk1-Sp1-Ar-Sp2-Alk2-C(H)=O

wherein Alk1, Sp1, Ar, Sp2, Alk2, and Z3' are as hereinbefore defined, and then reacting the compound Z3'CO-Alk1-Sp1-Ar-Sp2-Alk2-C(H)=O

with a carrier Z3, wherein Z3 is as hereinbefore defined, in an aqueous, buffered solution at a pH of between 6.5 and 9.0 and a temperature of 4° to 40°C
for 1-48 hours to generate a compound of formula Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(H)=O]m wherein Alk1, Sp1, Ar, Sp2, Alk2, Z3, and m are as hereinbefore defined, and finally reacting the compound Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(H)=O]m with a compound of formula Z2 wherein Z2 is Q-Sp-SS-W wherein Sp and W are as hereinbefore defined and Q is H2NHNCO-, H2NHNCS-, H2NHNCONH-, H2NHNCSNH-, or H2NO-,in an aqueous, buffered solution at a pH of between 4.0 and 6.0 and a temperature of 4° to 40°C for 1-48 hours to generate a compound of formula Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(H)=Z2]m wherein Alk1, Sp1, Ar, Sp2, and Alk2, Z3, and m are as hereinbefore defined, Z2 is Q-Sp-SS-W wherein Sp and W are as hereinbefore defined, and Q is =NHNCO-, =NHNCS-, =NHNCONH-, =NHNCSNH-, or =NO-.
43. A cytotoxic drug conjugate according to claim 10 wherein Sp1 is -O- or a bond, Alk1 is a bond or branched or unbranched (C1-C10) alkylene chain, with the proviso that when Alk1 is a bond, Sp1 is a bond, Z1 is (C1-C5) alkyl, and Z2 is Calicheamicin N-acetyl gamma dimethyl hydrazide.
44. A protein carrier of formula:
Z3[CO-Alk1-Sp1-Ar-Sp2-Alk2-C(Z1)=Z2]m wherein Z3 is humanized P67.6 (h-P67.6) and m is zero.
CA2150785A 1994-06-03 1995-06-01 Conjugates of methyltrithio antitumor agents and intermediates for their synthesis Expired - Lifetime CA2150785C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/253,877 US5773001A (en) 1994-06-03 1994-06-03 Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US08/253,877 1994-06-03

Publications (2)

Publication Number Publication Date
CA2150785A1 CA2150785A1 (en) 1995-12-04
CA2150785C true CA2150785C (en) 2010-07-06

Family

ID=22962074

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2150785A Expired - Lifetime CA2150785C (en) 1994-06-03 1995-06-01 Conjugates of methyltrithio antitumor agents and intermediates for their synthesis

Country Status (20)

Country Link
US (4) US5773001A (en)
EP (1) EP0689845B1 (en)
JP (2) JP3650165B2 (en)
KR (1) KR100408376B1 (en)
AT (1) ATE223234T1 (en)
AU (1) AU697280B2 (en)
BR (2) BR1101078A (en)
CA (1) CA2150785C (en)
CY (1) CY2334B1 (en)
DE (1) DE69528016T2 (en)
DK (1) DK0689845T3 (en)
ES (1) ES2181752T3 (en)
FI (1) FI117690B (en)
IL (1) IL113984A (en)
NO (1) NO313617B1 (en)
NZ (3) NZ536440A (en)
PT (1) PT689845E (en)
SI (1) SI0689845T1 (en)
TW (1) TW394778B (en)
ZA (1) ZA954570B (en)

Families Citing this family (699)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5773001A (en) * 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
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
AU2335899A (en) * 1998-01-29 1999-08-16 Pharmacopeia, Inc. Acid cleavable phenoxyalkyl linker for combinatorial synthesis
US6008321A (en) * 1998-03-16 1999-12-28 Pharmacopeia, Inc. Universal linker for combinatorial synthesis
US6914130B2 (en) * 1998-06-17 2005-07-05 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US6733998B1 (en) 1998-12-07 2004-05-11 Sloan-Kettering Institute For Cancer Research Micromonospora echinospora genes coding for biosynthesis of calicheamicin and self-resistance thereto
US20050159588A1 (en) * 1999-06-02 2005-07-21 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1189641B1 (en) 1999-06-25 2009-07-29 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
US20060228364A1 (en) * 1999-12-24 2006-10-12 Genentech, Inc. Serum albumin binding peptides for tumor targeting
US20040001827A1 (en) * 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
ATE337403T1 (en) * 1999-12-24 2006-09-15 Genentech Inc METHOD AND COMPOUNDS FOR EXTENSING THE HALF-LIFE TIMES IN THE EXCRETION OF BIOACTIVE COMPOUNDS
US20050287153A1 (en) * 2002-06-28 2005-12-29 Genentech, Inc. Serum albumin binding peptides for tumor targeting
AU2001233027A1 (en) * 2000-01-27 2001-08-07 Genetics Institute, Llc Antibodies against ctla4 (cd152), conjugates comprising same, and uses thereof
ES2528794T3 (en) 2000-04-11 2015-02-12 Genentech, Inc. Multivalent antibodies and uses thereof
DE60144198D1 (en) 2000-12-28 2011-04-21 Wyeth Llc RECOMBINANT PROTECTION PROTEIN FROM STREPTOCOCCUS PNEUMONIAE
US6652853B2 (en) * 2001-03-08 2003-11-25 Ludwig Institute For Cancer Research Method for treating cancer using A33 specific antibodies and chemotherapeutic agents
NZ511705A (en) * 2001-05-14 2004-03-26 Horticulture & Food Res Inst Methods and rapid immunoassay device for detecting progesterone and other steroids
US7129261B2 (en) 2001-05-31 2006-10-31 Medarex, Inc. Cytotoxic agents
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
EP1992643A3 (en) 2001-06-20 2008-12-10 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
US20050107595A1 (en) * 2001-06-20 2005-05-19 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20040086504A1 (en) * 2001-06-21 2004-05-06 Deepak Sampath Cyr61 as a target for treatment and diagnosis of breast cancer
US20040235068A1 (en) * 2001-09-05 2004-11-25 Levinson Arthur D. Methods for the identification of polypeptide antigens associated with disorders involving aberrant cell proliferation and compositions useful for the treatment of such disorders
WO2003024392A2 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20040023910A1 (en) * 2001-09-28 2004-02-05 Zhiming Zhang Use of cyr61 in the treatment and diagnosis of human uterine leiomyomas
US20110045005A1 (en) 2001-10-19 2011-02-24 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic origin
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1575571A4 (en) 2002-01-02 2008-06-25 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
US20090042291A1 (en) * 2002-03-01 2009-02-12 Xencor, Inc. Optimized Fc variants
US7662925B2 (en) * 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US20100311954A1 (en) * 2002-03-01 2010-12-09 Xencor, Inc. Optimized Proteins that Target Ep-CAM
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
HUE030806T2 (en) 2002-05-02 2017-05-29 Wyeth Holdings Llc Calicheamicin derivative-carrier conjugates
PT1585966E (en) 2002-07-15 2012-02-20 Hoffmann La Roche Treatment of cancer with the anti-erbb2 antibody rhumab 2c4
BRPI0314814C1 (en) 2002-09-27 2021-07-27 Xencor Inc antibody comprising an fc variant
EP1551876B1 (en) 2002-10-16 2011-03-16 Purdue Pharma L.P. Antibodies that bind cell-associated ca 125/0722p and methods of use thereof
US20040152632A1 (en) * 2002-11-06 2004-08-05 Wyeth Combination therapy for the treatment of acute leukemia and myelodysplastic syndrome
US20090010920A1 (en) * 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
SI1641822T1 (en) 2003-07-08 2013-08-30 Genentech, Inc. Il-17 a/f heterologous polypeptides and therapeutic uses thereof
PL1651612T3 (en) 2003-07-22 2012-09-28 Astex Therapeutics Ltd 3,4-disubstituted 1h-pyrazole compounds and their use as cyclin dependent kinases (cdk) and glycogen synthase kinase-3 (gsk-3) modulators
JP2007524649A (en) * 2003-07-29 2007-08-30 イミューノメディクス、インコーポレイテッド Fluorinated carbohydrate complex
KR20060069825A (en) * 2003-08-01 2006-06-22 제넨테크, 인크. Antibody cdr polypeptide sequences with restricted diversity
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US8101720B2 (en) * 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US20050282168A1 (en) * 2003-09-29 2005-12-22 Wyeth Cell surface molecules as markers and therapeutic agents against kidney cancers
KR101520209B1 (en) 2003-11-06 2015-05-13 시애틀 지네틱스, 인크. Monomethylvaline compounds capable of conjugation to ligands
EP1689432B1 (en) 2003-11-17 2009-12-30 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
PT1718677E (en) 2003-12-19 2012-07-18 Genentech Inc Monovalent antibody fragments useful as therapeutics
TW200539855A (en) * 2004-03-15 2005-12-16 Wyeth Corp Calicheamicin conjugates
MXPA06011199A (en) * 2004-03-31 2007-04-16 Genentech Inc Humanized anti-tgf-beta antibodies.
CA2559870A1 (en) * 2004-04-07 2005-10-27 Genetech,Inc. Mass spectrometry of antibody conjugates
US7691962B2 (en) * 2004-05-19 2010-04-06 Medarex, Inc. Chemical linkers and conjugates thereof
EP1747021B1 (en) * 2004-05-19 2015-09-09 E. R. Squibb & Sons, L.L.C. Self-immolative linkers and drug conjugates
NZ551180A (en) 2004-06-01 2009-10-30 Genentech Inc Antibody drug conjugates and methods
EP1919950A1 (en) 2004-07-15 2008-05-14 Xencor, Inc. Optimized fc variants
ZA200701183B (en) 2004-07-20 2008-05-28 Genentech Inc Inhibitors of angiopoietin-like 4 protein, combinations, an their use
NZ551908A (en) 2004-07-26 2009-10-30 Genentech Inc Methods and compositions for modulating hepatocyte growth factor HGF activation
CA2486285C (en) * 2004-08-30 2017-03-07 Viktor S. Goldmakher Immunoconjugates targeting syndecan-1 expressing cells and use thereof
RS57636B1 (en) * 2004-09-03 2018-11-30 Genentech Inc Humanized anti-beta7 antagonists and uses therefor
PA8645301A1 (en) * 2004-09-10 2006-07-03 Wyeth Corp ANTIBODY ANTI-5T4 HUMANIZED AND CONJUGATED ANTIBODY ANTI-5T4 / CALICHEAMICINA
EP1791565B1 (en) 2004-09-23 2016-04-20 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2006041641A2 (en) * 2004-10-05 2006-04-20 Genentech, Inc. Therapeutic agents with decreased toxicity
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US8802820B2 (en) * 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US20070135620A1 (en) * 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
CN102746404B (en) 2004-11-12 2016-01-20 赞科股份有限公司 To FcRn in conjunction with reformed Fc variant
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
JP4993645B2 (en) 2004-12-01 2012-08-08 ジェネンテック, インコーポレイテッド Antibody drug conjugates and methods
US7964195B2 (en) 2005-01-07 2011-06-21 Diadexus, Inc. Ovr110 antibody compositions and methods of use
US8404718B2 (en) 2005-01-21 2013-03-26 Astex Therapeutics Limited Combinations of pyrazole kinase inhibitors
AR054425A1 (en) 2005-01-21 2007-06-27 Astex Therapeutics Ltd PIPERIDIN ADDITION SALTS 4-IL-ACID AMID 4- (2,6-DICLORO-BENZOILAMINO) 1H-PIRAZOL-3-CARBOXILICO.
EP3698807A1 (en) 2005-01-21 2020-08-26 Genentech, Inc. Fixed dosing of her antibodies
ES2552338T3 (en) 2005-01-21 2015-11-27 Astex Therapeutics Limited Pharmaceutical compounds
AU2006214031A1 (en) * 2005-02-18 2006-08-24 Medarex, Inc. Human monoclonal antibodies to prostate specific membrane antigen (PSMA)
AU2006216732C1 (en) 2005-02-23 2017-07-20 Genentech, Inc. Extending time to disease progression or survival in cancer patients using a HER dimerization inhibitor
WO2006096861A2 (en) * 2005-03-08 2006-09-14 Genentech, Inc. METHODS FOR IDENTIFYING TUMORS RESPONSIVE TO TREATMENT WITH HER DIMERIZATION INHIBITORS (HDIs)
EP1865981A2 (en) 2005-04-07 2007-12-19 Chiron Corporation Cacna1e in cancer diagnosis, detection and treatment
EP1871911A2 (en) 2005-04-07 2008-01-02 Chiron Corporation Cancer-related genes (prlr)
US7714016B2 (en) * 2005-04-08 2010-05-11 Medarex, Inc. Cytotoxic compounds and conjugates with cleavable substrates
US7858843B2 (en) 2005-06-06 2010-12-28 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
WO2007094842A2 (en) * 2005-12-02 2007-08-23 Genentech, Inc. Binding polypeptides and uses thereof
US20070003559A1 (en) * 2005-07-01 2007-01-04 Wyeth Methods of determining pharmacokinetics of targeted therapies
WO2007008943A2 (en) 2005-07-08 2007-01-18 Xencor, Inc. Optimized anti-ep-cam antibodies
AU2006280321A1 (en) 2005-08-15 2007-02-22 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
US8795674B2 (en) 2005-09-30 2014-08-05 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Methods and compositions for modulating immune tolerance
EP1931709B1 (en) 2005-10-03 2016-12-07 Xencor, Inc. Fc variants with optimized fc receptor binding properties
ES2375843T3 (en) 2005-10-26 2012-03-06 Medarex, Inc. PROCEDURES AND COMPOUNDS FOR THE PREPARATION OF ANC? LOGOS OF CC-1065.
ES2577292T3 (en) 2005-11-07 2016-07-14 Genentech, Inc. Binding polypeptides with diversified VH / VL hypervariable sequences and consensus
WO2007059404A2 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
AU2006335053A1 (en) 2005-11-21 2007-07-19 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
CN105859886A (en) 2005-12-02 2016-08-17 健泰科生物技术公司 Compositions and methods associated with antibodies that bind to IL-22 and IL-22R
US20070237764A1 (en) * 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
BRPI0619476A2 (en) * 2005-12-05 2011-10-04 Wyeth Corp Interleukin-11 compositions and methods of use
US7763245B2 (en) 2005-12-15 2010-07-27 Genentech, Inc. Methods and compositions for targeting polyubiquitin
RU2450020C2 (en) 2006-01-05 2012-05-10 Дженентек, Инк. ANTI-EphB4 ANTIBODIES AND METHODS OF USING SAID ANTIBODIES
CA2638821A1 (en) 2006-02-17 2007-10-11 Genentech, Inc. Gene disruptons, compositions and methods relating thereto
TW200806685A (en) * 2006-02-21 2008-02-01 Wyeth Corp Processes for the convergent synthesis of calicheamicin derivatives
AU2014202414B2 (en) * 2006-02-21 2016-06-02 Wyeth Llc Processes for the convergent synthesis of calicheamicin derivatives
AU2012261705B2 (en) * 2006-02-21 2014-02-06 Wyeth Llc Processes for the convergent synthesis of calicheamicin derivatives
EP1999269B1 (en) 2006-03-08 2014-12-10 Wake Forest University Health Sciences Soluble monomeric ephrin a1
DK1994055T3 (en) 2006-03-10 2014-08-25 Wyeth Llc ANTI-5T4 ANTIBODIES AND APPLICATIONS THEREOF
AR059851A1 (en) 2006-03-16 2008-04-30 Genentech Inc ANTIBODIES OF EGFL7 AND METHODS OF USE
EP2389948A1 (en) 2006-03-23 2011-11-30 Novartis AG Anti-tumor cell antigen antibody therapeutics
EP2614839A3 (en) 2006-04-05 2015-01-28 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
JP2009541208A (en) 2006-04-13 2009-11-26 ノバルティス・バクシーンズ・アンド・ダイアグノスティクス・インコーポレイテッド How to treat, diagnose, or detect cancer
EP2082645A1 (en) 2006-04-19 2009-07-29 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
WO2007134132A2 (en) * 2006-05-12 2007-11-22 Genentech, Inc. Compositions and methods for the diagnosis and treatment of bladder and urinary tract tumors
AR060978A1 (en) * 2006-05-30 2008-07-23 Genentech Inc ANTIBODIES AND IMMUNOCATE PLAYERS AND THEIR USES
DK2383297T5 (en) 2006-08-14 2022-07-04 Xencor Inc Optimized antibodies directed against CD19
CA2662236A1 (en) 2006-09-12 2008-03-20 Genentech, Inc. Methods and compositions for the diagnosis and treatment of cancer
US8916552B2 (en) 2006-10-12 2014-12-23 Astex Therapeutics Limited Pharmaceutical combinations
EP2073807A1 (en) 2006-10-12 2009-07-01 Astex Therapeutics Limited Pharmaceutical combinations
SI2502938T1 (en) 2006-10-27 2015-05-29 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
JP5391073B2 (en) 2006-11-27 2014-01-15 ディアデクサス インコーポレーテッド Ovr110 antibody compositions and methods of use
TWI412367B (en) 2006-12-28 2013-10-21 Medarex Llc Chemical linkers and cleavable substrates and conjugates thereof
CA2676766A1 (en) 2007-02-09 2008-08-21 Genentech, Inc. Anti-robo4 antibodies and uses therefor
WO2008103693A2 (en) 2007-02-21 2008-08-28 Medarex, Inc. Chemical linkers with single amino acids and conjugates thereof
WO2008109440A2 (en) 2007-03-02 2008-09-12 Genentech, Inc. Predicting response to a her dimerisation inhibitor based on low her3 expression
JP2010521180A (en) 2007-03-14 2010-06-24 ノバルティス アーゲー APCDD1 inhibitor for treating, diagnosing or detecting cancer
US7960139B2 (en) 2007-03-23 2011-06-14 Academia Sinica Alkynyl sugar analogs for the labeling and visualization of glycoconjugates in cells
WO2008127656A1 (en) 2007-04-12 2008-10-23 The Brigham And Women's Hospital, Inc. Targeting abcb5 for cancer therapy
LT2176298T (en) 2007-05-30 2018-04-10 Xencor, Inc. Methods and compositions for inhibiting cd32b expressing cells
PE20090321A1 (en) 2007-06-04 2009-04-20 Genentech Inc ANTI-NOTCH1 NRR ANTIBODIES, METHOD OF PREPARATION AND PHARMACEUTICAL COMPOSITION
JP5469600B2 (en) 2007-07-16 2014-04-16 ジェネンテック, インコーポレイテッド Anti-CD79b antibody and immunoconjugate and method of use thereof
TW200918089A (en) 2007-07-16 2009-05-01 Genentech Inc Humanized anti-CD79b antibodies and immunoconjugates and methods of use
CL2008002886A1 (en) 2007-09-26 2009-12-04 Chugai Pharmaceutical Co Ltd Constant region of a human antibody; anti-interleukin-6 (yl-6) receptor antibody and pharmaceutical composition comprising it.
US8691222B2 (en) 2007-10-02 2014-04-08 Genentech, Inc. NLRR-1 antagonists and uses thereof
KR20100097691A (en) 2007-11-12 2010-09-03 테라클론 사이언시스, 아이엔씨. Compositions and methods for the therapy and diagnosis of influenza
US20110033476A1 (en) * 2007-11-12 2011-02-10 Theraclone Sciences Inc. Compositions and methods for the therapy and diagnosis of influenza
AR069501A1 (en) 2007-11-30 2010-01-27 Genentech Inc ANTI-VEGF ANTIBODIES (VASCULAR ENDOTELIAL GROWTH FACTOR)
PT2242772E (en) * 2007-12-26 2015-02-09 Biotest Ag Immunoconjugates targeting cd138 and uses thereof
EP2801584B1 (en) 2007-12-26 2019-07-10 Biotest AG Agents targeting CD138 and uses thereof
US9011864B2 (en) * 2007-12-26 2015-04-21 Biotest Ag Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
JP2011508738A (en) * 2007-12-26 2011-03-17 バイオテスト・アクチエンゲゼルシヤフト Method and agent for improving targeting of CD138 expressing tumor cells
SI2808343T1 (en) 2007-12-26 2019-10-30 Xencor Inc Fc variants with altered binding to FcRn
AU2009205995B2 (en) 2008-01-18 2014-04-03 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
SI2657253T1 (en) 2008-01-31 2017-10-30 Genentech, Inc. Anti-CD79b antibodies and immunoconjugates and methods of use
AU2009223688B2 (en) 2008-03-10 2014-12-11 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of cytomegalovirus infections
EP2631302A3 (en) * 2008-03-31 2014-01-08 Genentech, Inc. Compositions and methods for treating and diagnosing asthma
SG10201402815VA (en) 2008-04-09 2014-09-26 Genentech Inc Novel compositions and methods for the treatment of immune related diseases
JP6219556B2 (en) * 2008-05-16 2017-10-25 ジェネンテック, インコーポレイテッド Use of biomarkers for evaluation of treatment of gastrointestinal inflammatory disorders using beta7 integrin antagonists
JP5986745B2 (en) 2008-07-15 2016-09-06 アカデミア シニカAcademia Sinica Glycan arrays on PTFE-like aluminum-coated glass slides and related methods
AU2009274512A1 (en) 2008-07-25 2010-01-28 The Regents Of The University Of Colorado Clip inhibitors and methods of modulating immune function
AR073717A1 (en) 2008-10-01 2010-11-24 Genentech Inc ANTI-NOTCH2 ANTIBODIES OF MURINE AND HUMAN, AND METHODS OF USE
KR20110101212A (en) 2008-12-17 2011-09-15 제넨테크, 인크. Hepatitis c virus combination therapy
JP5936112B2 (en) 2009-02-11 2016-06-15 アルブミディクス アクティーゼルスカブ Albumin variants and complexes
SI3260136T1 (en) 2009-03-17 2021-05-31 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv) -neutralizing antibodies
ES2572728T3 (en) 2009-03-20 2016-06-02 F. Hoffmann-La Roche Ag Bispecific anti-HER antibodies
CA2754163C (en) 2009-03-25 2019-04-09 Genentech, Inc. Anti-fgfr3 antibodies and methods using same
NZ594343A (en) 2009-03-25 2013-10-25 Genentech Inc Novel anti-alpha5beta1 antibodies and uses thereof
AU2010236787A1 (en) 2009-04-01 2011-11-10 Genentech, Inc. Anti-FcRH5 antibodies and immunoconjugates and methods of use
CA2756244A1 (en) 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US8722860B2 (en) 2009-04-16 2014-05-13 Abbvie Biotherapeutics Inc. Anti-TNF-α antibodies and their uses
WO2010121125A1 (en) 2009-04-17 2010-10-21 Wake Forest University Health Sciences Il-13 receptor binding peptides
CA2759506A1 (en) * 2009-04-23 2010-10-28 Theraclone Sciences, Inc. Granulocyte-macrophage colony-stimulating factor (gm-csf) neutralizing antibodies
AU2010249787A1 (en) 2009-05-20 2011-12-22 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
EP3248619A3 (en) 2009-06-04 2018-03-07 Novartis AG Methods for identification of sites for igg conjugation
MX346002B (en) 2009-06-17 2017-03-01 Abbvie Biotherapeutics Inc Anti-vegf antibodies and their uses.
WO2011005481A1 (en) 2009-06-22 2011-01-13 Medimmune, Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
NZ597531A (en) 2009-07-31 2014-05-30 Genentech Inc Inhibition of tumor metastasis using bv8- or g-csf-antagonists
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US9321823B2 (en) 2009-09-02 2016-04-26 Genentech, Inc. Mutant smoothened and methods of using the same
RU2015153109A (en) 2009-09-16 2019-01-15 Дженентек, Инк. SUPERSPIRAL AND / OR BINDING PROTEIN COMPLEXES AND THEIR APPLICATIONS
EP2486023A4 (en) 2009-10-06 2014-05-07 Immunogen Inc Potent conjugates and hydrophilic linkers
PL2488873T3 (en) 2009-10-16 2016-01-29 Novartis Ag Biomarkers of tumor pharmacodynamic response
JP5889794B2 (en) 2009-10-19 2016-03-22 ジェネンテック, インコーポレイテッド Regulation of hepatocyte growth factor activator
NZ599337A (en) 2009-10-22 2013-05-31 Genentech Inc Anti-hepsin antibodies and methods using same
BR112012009409A2 (en) 2009-10-22 2017-02-21 Genentech Inc method of identifying an inhibitory substance, antagonist molecule, isolated nucleic acid, vector, host cell, method of making the molecule, composition, article of manufacture, method of inhibiting a biological activity, method of treating a pathological condition, method for detect msp in a sample and method to detect hepsin in a sample
WO2011056497A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor type iib compositions and methods of use
WO2011056494A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor-like kinase-1 antagonist and vegfr3 antagonist combinations
WO2011056502A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Bone morphogenetic protein receptor type ii compositions and methods of use
TW201122101A (en) 2009-10-28 2011-07-01 Facet Biotech Corp Anti-EGFR antibodies and their uses
CN105567699A (en) 2009-10-30 2016-05-11 诺维信生物制药丹麦公司 Albumin variants
RU2585488C2 (en) 2009-11-05 2016-05-27 Дженентек, Инк. Methods and composition for secretion of heterologous polypeptides
AR079217A1 (en) 2009-11-30 2012-01-04 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSIS AND TUMOR TREATMENT
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
EP2507381A4 (en) 2009-12-04 2016-07-20 Hoffmann La Roche Multispecific antibodies, antibody analogs, compositions, and methods
ES2722300T3 (en) 2009-12-10 2019-08-09 Hoffmann La Roche Antibodies that preferentially bind to extracellular domain 4 of CSF1R and its use
TWI505836B (en) 2009-12-11 2015-11-01 Genentech Inc Anti-vegf-c antibodies and methods using same
EP2513148B1 (en) 2009-12-16 2016-08-31 AbbVie Biotherapeutics Inc. Anti-her2 antibodies and their uses
US9023996B2 (en) 2009-12-23 2015-05-05 Synimmune Gmbh Anti-FLT3 antibodies
SI2516465T1 (en) 2009-12-23 2016-08-31 F. Hoffmann-La Roche Ag Anti-bv8 antibodies and uses thereof
WO2011089211A1 (en) 2010-01-22 2011-07-28 Synimmune Gmbh Anti-cd133 antibodies and methods of using the same
KR101923235B1 (en) 2010-02-04 2018-11-28 에이자이 아이엔씨. Chlorotoxin polypeptides and conjugates and uses thereof
KR101510413B1 (en) 2010-02-08 2015-04-08 어젠시스 인코포레이티드 Antibody drug conjugates (adc) that bind to 161p2f10b proteins
JP5981853B2 (en) 2010-02-18 2016-08-31 ジェネンテック, インコーポレイテッド Neuregulin antagonists and their use in the treatment of cancer
MX2012009215A (en) 2010-02-23 2012-11-23 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
CA2789076C (en) 2010-03-05 2017-11-21 F. Hoffmann-La Roche Ag Antibodies against human colony stimulating factor-1 receptor and uses thereof
BR112012022046A2 (en) 2010-03-05 2017-02-14 F Hoffamann-La Roche Ag "antibody, pharmaceutical composition, nucleic acid, expression vectors, host cell and method for producing a recombinant antibody".
US8642557B2 (en) 2010-03-12 2014-02-04 Abbvie Biotherapeutics Inc. CTLA4 proteins and their uses
NZ602040A (en) 2010-03-24 2014-12-24 Genentech Inc Anti-lrp6 antibodies
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
CN106977608A (en) 2010-04-09 2017-07-25 阿尔布麦狄克斯公司 Albumin derivant and variant
WO2011130332A1 (en) 2010-04-12 2011-10-20 Academia Sinica Glycan arrays for high throughput screening of viruses
ES2617777T5 (en) 2010-04-23 2022-10-13 Hoffmann La Roche Production of heteromultimeric proteins
MA34291B1 (en) 2010-05-03 2013-06-01 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING A TUMOR
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
BR112012029866A2 (en) 2010-06-03 2017-03-07 Genentech Inc method for determining the presence of a steap-1 protein
CA3220104A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
RU2577986C2 (en) 2010-06-18 2016-03-20 Дженентек, Инк. Antibodies against axl and their application
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2011161189A1 (en) 2010-06-24 2011-12-29 F. Hoffmann-La Roche Ag Anti-hepsin antibodies and methods of use
JP5953303B2 (en) 2010-07-29 2016-07-20 ゼンコア インコーポレイテッド Antibodies with modified isoelectric points
CN103153341B (en) 2010-08-03 2015-05-27 霍夫曼-拉罗奇有限公司 Chronic lymphocytic leukemia (Cll) biomarkers
JP2013540701A (en) 2010-08-12 2013-11-07 セラクローン サイエンシーズ, インコーポレイテッド Anti-hemagglutinin antibody composition and method of use thereof
SG187746A1 (en) 2010-08-13 2013-03-28 Roche Glycart Ag Anti-fap antibodies and methods of use
BR112013002444A2 (en) 2010-08-13 2016-05-24 Roche Glycart Ag isolated antibody, polynucleotide and polypeptide, composition, vector, host cell, antibody conjugate, pharmaceutical formulation, use of the antibody, methods of producing an antibody, treating an individual, inducing cell lysis of a tumor cell and diagnosing a disease in an individual
BR112013003279A2 (en) 2010-08-13 2016-06-14 Genentech In "Methods for treating a disease, method for neutralizing or blocking il-1ß and / or il-18 activity, antibody, uses of an antibody and uses of a monoclonal antibody"
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
SG10201408229WA (en) 2010-08-31 2015-02-27 Genentech Inc Biomarkers and methods of treatment
PT3556396T (en) 2010-08-31 2022-07-04 Scripps Research Inst Human immunodeficiency virus (hiv)-neutralizing antibodies
EP2614085B1 (en) 2010-09-10 2016-04-20 Apexigen, Inc. Anti-il-1 beta antibodies and methods of use
PT2621526T (en) 2010-09-29 2018-08-02 Seattle Genetics Inc Antibody drug conjugates (adc) that bind to 191p4d12 proteins
US9228023B2 (en) 2010-10-01 2016-01-05 Oxford Biotherapeutics Ltd. Anti-ROR1 antibodies and methods of use for treatment of cancer
UA112062C2 (en) 2010-10-04 2016-07-25 Бьорінгер Інгельхайм Інтернаціональ Гмбх CD33-Binding Agent
WO2012047968A2 (en) 2010-10-05 2012-04-12 Genentech, Inc. Mutant smoothened and methods of using the same
WO2012064836A1 (en) 2010-11-10 2012-05-18 Genentech, Inc. Methods and compositions for neural disease immunotherapy
US9309322B2 (en) 2010-11-12 2016-04-12 Scott & White Healthcare (Swh) Antibodies to tumor endothelial marker 8
AU2011343570B2 (en) 2010-12-16 2016-11-03 Genentech, Inc. Diagnosis and treatments relating to TH2 inhibition
MX345519B (en) 2010-12-20 2017-02-01 Genentech Inc Anti-mesothelin antibodies and immunoconjugates.
MA34818B1 (en) 2010-12-22 2014-01-02 Genentech Inc ANTI-PCSK9 ANTIBODIES AND METHODS OF USE
SG191153A1 (en) 2010-12-23 2013-07-31 Hoffmann La Roche Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
JOP20210044A1 (en) 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
WO2012092539A2 (en) 2010-12-31 2012-07-05 Takeda Pharmaceutical Company Limited Antibodies to dll4 and uses thereof
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
CA2825064C (en) 2011-02-04 2022-08-30 Genentech, Inc. Fc variants and methods for their production
US20140088019A1 (en) 2011-02-11 2014-03-27 Zyngenia, Inc. Monovalent and Multivalent Multispecific Complexes and Uses Thereof
AU2012217867A1 (en) 2011-02-14 2013-09-05 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
JP6097702B2 (en) 2011-03-03 2017-03-15 アペクシジェン, インコーポレイテッド Anti-IL-6 receptor antibody and method of use thereof
CA2827759C (en) 2011-03-10 2018-10-16 Omeros Corporation Generation of anti-fn14 monoclonal antibodies by ex-vivo accelerated antibody evolution
EP2685968A1 (en) 2011-03-15 2014-01-22 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
CN103561771B (en) 2011-03-17 2019-01-04 伯明翰大学 The immunization therapy redirected
PT2691417T (en) 2011-03-29 2018-10-31 Roche Glycart Ag Antibody fc variants
JP2014516511A (en) 2011-04-07 2014-07-17 ジェネンテック, インコーポレイテッド Anti-FGFR4 antibody and method of use
JP2014514313A (en) 2011-04-20 2014-06-19 ロシュ グリクアート アクチェンゲゼルシャフト Methods and constructs for pH-dependent passage of the blood brain barrier
EP3508500A1 (en) 2011-04-29 2019-07-10 Apexigen, Inc. Anti-cd40 antibodies and methods of use
WO2012158704A1 (en) 2011-05-16 2012-11-22 Genentech, Inc. Fgfr1 agonists and methods of use
EP2714738B1 (en) 2011-05-24 2018-10-10 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
PT3415531T (en) 2011-05-27 2023-09-12 Glaxo Group Ltd Bcma (cd269/tnfrsf17) - binding proteins
ES2622578T3 (en) 2011-06-10 2017-07-06 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
WO2013003507A1 (en) 2011-06-27 2013-01-03 Morphotek, Inc. Multifunctional agents
WO2013003680A1 (en) 2011-06-30 2013-01-03 Genentech, Inc. Anti-c-met antibody formulations
WO2013022855A1 (en) 2011-08-05 2013-02-14 Xencor, Inc. Antibodies with modified isoelectric points and immunofiltering
JP5944994B2 (en) 2011-08-12 2016-07-05 オメロス コーポレーション Anti-FZD10 monoclonal antibodies and methods for their use
MX2014001766A (en) 2011-08-17 2014-05-01 Genentech Inc Neuregulin antibodies and uses thereof.
CN103890008A (en) 2011-08-17 2014-06-25 霍夫曼-拉罗奇有限公司 Inhibition of angiogenesis in refractory tumors
KR101870555B1 (en) 2011-08-23 2018-06-22 로슈 글리카트 아게 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
RU2617970C2 (en) 2011-08-23 2017-04-28 Рош Гликарт Аг ANTIBODIES WITHOUT Fc-FRAGMENT INCLUDING TWO FAB-FRAGMENT AND METHODS OF APPLICATION
US8822651B2 (en) 2011-08-30 2014-09-02 Theraclone Sciences, Inc. Human rhinovirus (HRV) antibodies
KR20140068062A (en) 2011-09-15 2014-06-05 제넨테크, 인크. Methods of promoting differentiation
MX2014002990A (en) 2011-09-19 2014-05-21 Genentech Inc Combination treatments comprising c-met antagonists and b-raf antagonists.
MX2014002996A (en) 2011-09-23 2014-05-28 Roche Glycart Ag Bispecific anti-egfr/anti igf-1r antibodies.
US9663573B2 (en) 2011-10-05 2017-05-30 Genentech, Inc. Methods of treating liver conditions using Notch2 antagonists
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
JP6310394B2 (en) 2011-10-10 2018-04-11 ゼンコア インコーポレイテッド Methods for purifying antibodies
KR102102862B1 (en) 2011-10-14 2020-04-22 제넨테크, 인크. ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
JP6134725B2 (en) 2011-10-14 2017-05-24 ジェネンテック, インコーポレイテッド BACE1 peptide inhibitors
KR20140084164A (en) 2011-10-15 2014-07-04 제넨테크, 인크. Scd1 antagonists for treating cancer
WO2013063312A1 (en) 2011-10-25 2013-05-02 Memorial Sloan-Kettering Cancer Center Free psa antibodies as diagnostics, prognostics and therapeutics for prostate cancer
AU2012328980A1 (en) 2011-10-28 2014-04-24 Genentech, Inc. Therapeutic combinations and methods of treating melanoma
TWI571473B (en) 2011-11-02 2017-02-21 埃派斯進有限公司 Anti-kdr antibodies and methods of use
EP2589609A1 (en) 2011-11-03 2013-05-08 Pierre Fabre Medicament Antigen binding protein and its use as addressing product for the treatment of cancer
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
BR112014012005A2 (en) 2011-11-21 2017-12-19 Genentech Inc compositions, methods, pharmaceutical formulation and article
BR112014013694A2 (en) 2011-12-08 2017-06-13 Biotest Ag method to treat a disease and kit
CN104159921B (en) 2011-12-15 2018-05-04 霍夫曼-拉罗奇有限公司 Antibody for people CSF-1R and application thereof
US20130195851A1 (en) 2011-12-23 2013-08-01 Genentech, Inc. Articles of manufacture and methods for co-administration of antibodies
WO2013106489A1 (en) 2012-01-09 2013-07-18 The Scripps Research Institute Humanized antibodies with ultralong cdr3s
WO2013106485A2 (en) 2012-01-09 2013-07-18 The Scripps Research Institute Ultralong complementarity determining regions and uses thereof
JP6242813B2 (en) 2012-01-18 2017-12-06 ジェネンテック, インコーポレイテッド Anti-LRP5 antibody and method of use
CN104168920A (en) 2012-01-18 2014-11-26 霍夫曼-拉罗奇有限公司 Methods of using FGF19 modulators
WO2013116686A1 (en) 2012-02-02 2013-08-08 Massachusetts Institute Of Technology Methods and products related to targeted cancer therapy
KR20140127854A (en) 2012-02-10 2014-11-04 제넨테크, 인크. Single-chain antibodies and other heteromultimers
BR112014019741A2 (en) 2012-02-11 2020-12-22 Genentech, Inc USES OF AN ANTAGONIST OF THE WNT VIA, USE OF ANTI-CANCER THERAPY, METHOD OF IDENTIFICATION OF AN INDIVIDUAL WITH CANCER, METHODS FOR PREVENTING, METHOD OF INHIBITION OF A CANCER CELL PROLIFERATION, USE OF AN ANGONIST ANTAGONIST TRANSLOCATION OF ISOLATED R-SPONDINA
JP6152120B2 (en) 2012-02-15 2017-06-21 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Affinity chromatography based on Fc receptors
GB201203442D0 (en) 2012-02-28 2012-04-11 Univ Birmingham Immunotherapeutic molecules and uses
BR112014018679A2 (en) 2012-03-16 2017-07-04 Novozymes Biopharma Dk As albumin variants
US10024860B2 (en) 2012-03-28 2018-07-17 Massachusetts Institute Of Technology Cancer-related extracellular matrix signatures and related methods and products
WO2013148259A1 (en) 2012-03-28 2013-10-03 Massachusetts Institute Of Technology Cancer-related extracellular matrix signatures and related methods and products
AR090549A1 (en) 2012-03-30 2014-11-19 Genentech Inc ANTI-LGR5 AND IMMUNOCATE PLAYERS
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
RU2014148162A (en) 2012-05-01 2016-06-20 Дженентек, Инк. ANTI-PMEL17 ANTIBODIES AND THEIR IMMUNO CONJUGATES
WO2013170191A1 (en) 2012-05-11 2013-11-14 Genentech, Inc. Methods of using antagonists of nad biosynthesis from nicotinamide
CN104364266A (en) 2012-06-15 2015-02-18 霍夫曼-拉罗奇有限公司 Anti-PCSK9 antibodies, formulations, dosing, and methods of use
WO2014001324A1 (en) 2012-06-27 2014-01-03 Hoffmann-La Roche Ag Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
BR112014029888A2 (en) 2012-06-27 2020-05-12 Hoffmann La Roche METHODS OF PRODUCTION OF AN ANTIBODY, DETERMINATION OF A COMBINATION OF BINDING SITES AND TREATMENT OF AN INDIVIDUAL WITH CANCER, PHARMACEUTICAL FORMULATION, ANTIBODY AND USE OF AN ANTIBODY
RU2015100656A (en) 2012-06-27 2016-08-20 Ф. Хоффманн-Ля Рош Аг METHOD FOR PRODUCING ANTIBODY FC-FRAGMENT CONNECTING, INCLUDING AT LEAST ONE CONNECTING GROUP, WHICH SPECIALLY RELATED TO THE TARGET, AND THEIR APPLICATION
ES2604012T3 (en) 2012-07-04 2017-03-02 F. Hoffmann-La Roche Ag Covalently bound antigen-antibody conjugates
CA2872192A1 (en) 2012-07-04 2014-01-09 F. Hoffmann-La Roche Ag Anti-biotin antibodies and methods of use
MX353951B (en) 2012-07-04 2018-02-07 Hoffmann La Roche Anti-theophylline antibodies and methods of use.
HUE056217T2 (en) 2012-07-13 2022-02-28 Roche Glycart Ag Bispecific anti-vegf/anti-ang-2 antibodies and their use in the treatment of ocular vascular diseases
AU2013306098A1 (en) 2012-08-18 2015-02-12 Academia Sinica Cell-permeable probes for identification and imaging of sialidases
SG10201701424QA (en) 2012-08-23 2017-04-27 Agensys Inc Antibody drug conjugates (adc) that bind to 158p1d7 proteins
MX2015003541A (en) 2012-09-19 2015-10-26 Abbvie Biotherapeutics Inc Methods for identifying antibodies with reduced immunogenicity.
WO2014056783A1 (en) 2012-10-08 2014-04-17 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
NZ746440A (en) 2012-10-11 2019-11-29 Daiichi Sankyo Co Ltd Glycinamide derivatives and production methods thereof
WO2014061277A1 (en) 2012-10-19 2014-04-24 第一三共株式会社 Antibody-drug conjugate produced by binding through linker having hydrophilic structure
CA2889756C (en) 2012-10-26 2023-03-14 Memorial Sloan-Kettering Cancer Center Thiohydantoin compounds as androgen receptor modulators
CN104918957B (en) 2012-10-30 2018-11-16 埃派斯进有限公司 Anti-CD 40 antibodies and its application method
US10189906B2 (en) 2012-11-01 2019-01-29 Max-Delrück-Centrum Für Molekulare Medizin Antibody that binds CD269 (BCMA) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
JP6445444B2 (en) 2012-11-05 2018-12-26 ピエール、ファーブル、メディカマン Novel antigen binding proteins and their use as addressing products (ADDRESSINGPRODUCT) for the treatment of cancer
CA2884431A1 (en) 2012-11-08 2014-05-15 F. Hoffmann-La Roche Ag Her3 antigen binding proteins binding to the beta-hairpin of her3
MX2015005363A (en) 2012-11-08 2015-11-06 Novozymes Biopharma Dk As Albumin variants.
WO2014078268A2 (en) 2012-11-13 2014-05-22 Genentech, Inc. Anti-hemagglutinin antibodies and methods of use
US10131682B2 (en) 2012-11-24 2018-11-20 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and their uses for conjugation of drugs to a cell binding molecules
US20140154255A1 (en) 2012-11-30 2014-06-05 Abbvie Biotherapeutics Inc. Anti-vegf antibodies and their uses
JP6334553B2 (en) 2012-12-10 2018-05-30 メルサナ セラピューティクス,インコーポレイティド Protein-polymer-drug conjugate
US9872918B2 (en) 2012-12-12 2018-01-23 Mersana Therapeutics, Inc. Hydroxyl-polymer-drug-protein conjugates
WO2014107739A1 (en) 2013-01-07 2014-07-10 Eleven Biotherapeutics, Inc. Antibodies against pcsk9
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
AU2014205086B2 (en) 2013-01-14 2019-04-18 Xencor, Inc. Novel heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
EP2945969A1 (en) 2013-01-15 2015-11-25 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
JP2016509045A (en) 2013-02-22 2016-03-24 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト How to treat cancer and prevent drug resistance
CA2896259A1 (en) 2013-02-26 2014-09-04 Roche Glycart Ag Anti-mcsp antibodies
US9925240B2 (en) 2013-03-06 2018-03-27 Genentech, Inc. Methods of treating and preventing cancer drug resistance
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
CA2905070A1 (en) 2013-03-14 2014-09-25 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
MX2015012872A (en) 2013-03-15 2016-02-03 Ac Immune Sa Anti-tau antibodies and methods of use.
RU2015144033A (en) 2013-03-15 2017-04-26 Эббви Байотекнолоджи Лтд. ANTIBODIES AGAINST CD25 AND THEIR APPLICATION
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
MX2015011899A (en) 2013-03-15 2016-05-05 Genentech Inc Methods of treating cancer and preventing cancer drug resistance.
BR112015021521A2 (en) 2013-03-15 2017-10-10 Genentech Inc anti-crth2 antibodies and methods for their use
JP6527132B2 (en) 2013-03-15 2019-06-05 ジェネンテック, インコーポレイテッド Compositions and methods for diagnosis and treatment of liver cancer
CN111138543A (en) 2013-03-15 2020-05-12 Xencor股份有限公司 Heterodimeric proteins
JP2016515524A (en) 2013-03-15 2016-05-30 アッヴィ バイオテクノロジー リミテッド Anti-CD25 antibodies and their use
EP3421495A3 (en) 2013-03-15 2019-05-15 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions
EP2972373B1 (en) 2013-03-15 2019-10-09 F.Hoffmann-La Roche Ag Biomarkers and methods of treating pd-1 and pd-l1 related conditions
CN105143257B (en) 2013-03-15 2020-10-27 艾伯维生物医疗股份有限公司 FC variants
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
EA201890895A1 (en) 2013-03-15 2019-02-28 Зинджения, Инк. MULTIVALENT AND MONOVALENT MULTIS-SPECIFIC COMPLEXES AND THEIR APPLICATION
AR095882A1 (en) 2013-04-22 2015-11-18 Hoffmann La Roche ANTIBODY COMBINATION THERAPY AGAINST HUMAN CSF-1R WITH A TLR9 AGONIST
RU2687043C2 (en) 2013-04-29 2019-05-06 Ф. Хоффманн-Ля Рош Аг Fc-RECEPTOR BINDING MODIFIED ASYMMETRIC ANTIBODIES AND METHODS OF USE
EP2992329A1 (en) 2013-05-03 2016-03-09 Eleven Biotherapeutics, Inc. Albumin variants binding to fcrn
EP2999716A2 (en) 2013-05-20 2016-03-30 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
US10086054B2 (en) 2013-06-26 2018-10-02 Academia Sinica RM2 antigens and use thereof
EP3013347B1 (en) 2013-06-27 2019-12-11 Academia Sinica Glycan conjugates and use thereof
EP3022224A2 (en) 2013-07-18 2016-05-25 Fabrus, Inc. Antibodies with ultralong complementarity determining regions
CN105814074B (en) 2013-07-18 2020-04-21 图鲁斯生物科学有限责任公司 Humanized antibodies with ultralong complementarity determining regions
JP6510518B2 (en) 2013-08-01 2019-05-08 アジェンシス,インコーポレイテッド Antibody-drug conjugate (ADC) that binds to the CD37 protein
WO2015035044A2 (en) 2013-09-04 2015-03-12 Abbvie Biotherapeutics Inc. Fc VARIANTS WITH IMPROVED ANTIBODY-DEPENDENT CELL-MEDIATED CYTOTOXICITY
CN105682666B (en) 2013-09-06 2021-06-01 中央研究院 Activation of human iNKT cells using glycolipids
AR097584A1 (en) 2013-09-12 2016-03-23 Hoffmann La Roche ANTIBODY COMBINATION THERAPY AGAINST HUMAN CSF-1R AND ANTIBODIES AGAINST HUMAN PD-L1
CN105518027A (en) 2013-09-17 2016-04-20 豪夫迈·罗氏有限公司 Methods of using anti-LGR5 antibodies
MX2016004579A (en) 2013-10-10 2016-12-09 Beth Israel Deaconess Medical Ct Inc Tm4sf1 binding proteins and methods of using same.
PT3055331T (en) 2013-10-11 2021-04-05 Oxford Bio Therapeutics Ltd Conjugated antibodies against ly75 for the treatment of cancer
AU2014331714B2 (en) 2013-10-11 2019-05-02 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
CN105813655B (en) 2013-10-11 2022-03-15 阿萨纳生物科技有限责任公司 Protein-polymer-drug conjugates
MX2016004802A (en) 2013-10-18 2016-07-18 Genentech Inc Anti-rsp02 and/or anti-rsp03 antibodies and their uses.
WO2015063680A1 (en) * 2013-11-04 2015-05-07 Pfizer Inc. Intermediates and methods for synthesizing calicheamicin derivatives
KR20160072268A (en) 2013-11-04 2016-06-22 화이자 인코포레이티드 Anti-efna4 antibody-drug conjugates
MA39095A1 (en) 2013-12-13 2018-08-31 Genentech Inc Anti-cd33 antibodies and immunoconjugates
EP3082875B1 (en) 2013-12-16 2020-11-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015100113A2 (en) 2013-12-23 2015-07-02 Memorial Sloan-Kettering Cancer Center Methods and compositions for treating cancer using peptide nucleic acid-based agents
TWI728373B (en) 2013-12-23 2021-05-21 美商建南德克公司 Antibodies and methods of use
BR112016012666A2 (en) 2014-01-03 2017-09-26 Hoffmann La Roche conjugate, antibodies, pharmaceutical formulation and uses of conjugate
JP6521464B2 (en) 2014-01-03 2019-05-29 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Covalently linked polypeptide toxin-antibody conjugates
EP3089996B1 (en) 2014-01-03 2021-07-28 F. Hoffmann-La Roche AG Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
US9982041B2 (en) 2014-01-16 2018-05-29 Academia Sinica Compositions and methods for treatment and detection of cancers
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
US10562977B2 (en) 2014-01-29 2020-02-18 Shanghai Hengrui Pharmaceutical Co., Ltd. Ligand-cytotoxic drug conjugate, preparation method thereof, and uses thereof
WO2015120075A2 (en) 2014-02-04 2015-08-13 Genentech, Inc. Mutant smoothened and methods of using the same
AU2015217271B2 (en) 2014-02-12 2018-10-25 Genentech, Inc. Anti-Jagged1 antibodies and methods of use
BR112016018980A2 (en) 2014-02-21 2017-10-10 Genentech Inc method of treating a disorder, multispecific antibody, isolated nucleic acid, host cell, methods of producing an antibody, producing an antibody half or multispecific antibody, and producing a multispecific, immunoconjugate antibody and pharmaceutical formulation
US10464955B2 (en) 2014-02-28 2019-11-05 Hangzhou Dac Biotech Co., Ltd. Charged linkers and their uses for conjugation
MA39746A (en) 2014-03-14 2021-04-28 Hoffmann La Roche HETEROLOGICAL POLYPEPTIDE SECRETION COMPOSITIONS AND ASSOCIATED PROCESSES
TW202214691A (en) 2014-03-21 2022-04-16 美商艾伯維有限公司 Anti-egfr antibodies and antibody drug conjugates
BR112016021383A2 (en) 2014-03-24 2017-10-03 Genentech Inc METHOD TO IDENTIFY A PATIENT WITH CANCER WHO IS LIKE OR LESS LIKELY TO RESPOND TO TREATMENT WITH A CMET ANTAGONIST, METHOD TO IDENTIFY A PATIENT WITH PREVIOUSLY TREATED CANCER, METHOD TO DETERMINE THE EXPRESSION OF THE HGF BIOMARKER, ANTI-C-MET ANTAGONIST AND ITS USE, DIAGNOSTIC KIT AND ITS PREPARATION METHOD
TWI687428B (en) 2014-03-27 2020-03-11 中央研究院 Reactive labelling compounds and uses thereof
EP3954713A3 (en) 2014-03-28 2022-03-30 Xencor, Inc. Bispecific antibodies that bind to cd38 and cd3
EP3632934A1 (en) 2014-03-31 2020-04-08 F. Hoffmann-La Roche AG Anti-ox40 antibodies and methods of use
WO2015153514A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
US10160812B2 (en) 2014-04-11 2018-12-25 Medimmune, Llc Bispecific HER2 antibodies
KR102357032B1 (en) 2014-04-25 2022-02-08 피에르 파브르 메디카먼트 IGF-1R antibody and its use as addessing vehicle for the treatment of cancer
LT3137114T (en) 2014-04-30 2021-03-25 Pfizer Inc. Anti-ptk7 antibody-drug conjugates
BR112016024462B1 (en) 2014-05-06 2022-12-27 Genentech, Inc METHODS FOR PREPARING AN ANTIBODY
MX2016015162A (en) 2014-05-22 2017-03-03 Genentech Inc Anti-gpc3 antibodies and immunoconjugates.
CN106661622B (en) 2014-05-23 2020-08-21 豪夫迈·罗氏有限公司 MIT biomarkers and methods of using the same
CN106573971A (en) 2014-05-27 2017-04-19 中央研究院 Anti-CD20 glycoantibodies and uses thereof
AU2015267045B2 (en) 2014-05-27 2021-02-25 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
EP3149161B1 (en) 2014-05-27 2021-07-28 Academia Sinica Fucosidase from bacteroides and methods using the same
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
TWI732738B (en) 2014-05-28 2021-07-11 中央研究院 Anti-tnf-alpha glycoantibodies and uses thereof
JP2017517552A (en) 2014-06-13 2017-06-29 ジェネンテック, インコーポレイテッド Treatment and prevention of anticancer drug resistance
KR20170026362A (en) 2014-06-26 2017-03-08 에프. 호프만-라 로슈 아게 Anti-brdu antibodies and methods of use
EP3166627A1 (en) 2014-07-11 2017-05-17 Genentech, Inc. Notch pathway inhibition
EP3194449A1 (en) 2014-07-24 2017-07-26 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
AU2015308818B2 (en) 2014-08-28 2021-02-25 Bioatla Llc Conditionally active chimeric antigen receptors for modified T-cells
TWI751102B (en) 2014-08-28 2022-01-01 美商奇諾治療有限公司 Antibodies and chimeric antigen receptors specific for cd19
WO2016040369A2 (en) 2014-09-08 2016-03-17 Academia Sinica HUMAN iNKT CELL ACTIVATION USING GLYCOLIPIDS
EP3191518B1 (en) 2014-09-12 2020-01-15 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
EA201790545A1 (en) 2014-09-12 2017-07-31 Дженентек, Инк. ANTIBODIES AND IMMUNOCONJUGATES AGAINST HER2
US9751946B2 (en) 2014-09-12 2017-09-05 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
CA2957354A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
PL3262071T3 (en) 2014-09-23 2020-08-10 F. Hoffmann-La Roche Ag Method of using anti-cd79b immunoconjugates
JP6863888B2 (en) 2014-10-01 2021-04-21 メディミューン,エルエルシー How to conjugate a polypeptide
WO2016061389A2 (en) 2014-10-16 2016-04-21 Genentech, Inc. Anti-alpha-synuclein antibodies and methods of use
JP6723509B2 (en) 2014-10-24 2020-07-15 日油株式会社 Antibody-drug conjugates with cyclic benzylidene acetal linker
CN107148283A (en) 2014-10-31 2017-09-08 豪夫迈·罗氏有限公司 Anti- IL 17A and IL 17F cross reacting antibodies variant, the composition comprising it and its preparation and application
CA2966523A1 (en) 2014-11-03 2016-05-12 Genentech, Inc. Assays for detecting t cell immune subsets and methods of use thereof
CN114381521A (en) 2014-11-03 2022-04-22 豪夫迈·罗氏有限公司 Methods and biomarkers for efficacy prediction and assessment of OX40 agonist treatment
CA2961439A1 (en) 2014-11-05 2016-05-12 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
WO2016073157A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Anti-ang2 antibodies and methods of use thereof
CA2960797A1 (en) 2014-11-06 2016-05-12 F. Hoffmann-La Roche Ag Fc-region variants with modified fcrn-binding and methods of use
CR20170240A (en) 2014-11-10 2018-04-03 Genentech Inc ANTI-INTERLEUCINA-33 ANTIBODIES AND THEIR USES
US10508151B2 (en) 2014-11-19 2019-12-17 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
US11008403B2 (en) 2014-11-19 2021-05-18 Genentech, Inc. Anti-transferrin receptor / anti-BACE1 multispecific antibodies and methods of use
WO2016081639A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Antibodies against bace1 and use thereof for neural disease immunotherapy
BR112017011166A2 (en) 2014-11-26 2018-02-27 Xencor, Inc. heterodimeric antibodies that bind to cd3 and cd38
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
CA2967426A1 (en) 2014-11-26 2016-06-02 Xencor, Inc. Heterodimeric antibodies that bind cd3 and tumor antigens
EP3227332B1 (en) 2014-12-03 2019-11-06 F.Hoffmann-La Roche Ag Multispecific antibodies
US9975949B2 (en) 2014-12-05 2018-05-22 Genentech, Inc. Anti-CD79b antibodies and methods of use
RU2017120039A (en) 2014-12-10 2019-01-10 Дженентек, Инк. ANTIBODIES TO HEMATOENCEPHALIC BARRIER RECEPTORS AND METHODS OF APPLICATION
RU2746356C2 (en) 2014-12-19 2021-04-12 Чугаи Сейяку Кабусики Кайся C5 antibodies and their application methods
JP6862343B2 (en) 2014-12-19 2021-04-21 アブリンクス エン.ヴェー. Cysteine-bonded nanobody dimer
WO2016105450A2 (en) 2014-12-22 2016-06-30 Xencor, Inc. Trispecific antibodies
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
US20160208018A1 (en) 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
GB201501004D0 (en) 2015-01-21 2015-03-04 Cancer Rec Tech Ltd Inhibitors
WO2016117346A1 (en) 2015-01-22 2016-07-28 Chugai Seiyaku Kabushiki Kaisha A combination of two or more anti-c5 antibodies and methods of use
WO2016118961A1 (en) 2015-01-24 2016-07-28 Academia Sinica Cancer markers and methods of use thereof
EP3248005B1 (en) 2015-01-24 2020-12-09 Academia Sinica Novel glycan conjugates and methods of use thereof
AU2016215604B2 (en) 2015-02-02 2020-08-13 The University Of Birmingham Targeting moiety peptide epitope complexes having a plurality of T-cell epitopes
US10330683B2 (en) 2015-02-04 2019-06-25 Genentech, Inc. Mutant smoothened and methods of using the same
KR20170110129A (en) 2015-02-05 2017-10-10 추가이 세이야쿠 가부시키가이샤 Antibodies comprising ionic concentration dependent antigen binding domains, Fc region variants, antibodies that bind to IL-8, and their use
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
JP2018515457A (en) * 2015-04-21 2018-06-14 アッヴィ・ステムセントルクス・エル・エル・シー Calicheamicin constructs and methods of use
JP7044553B2 (en) 2015-04-24 2022-03-30 ジェネンテック, インコーポレイテッド How to identify bacteria containing bound polypeptides
CN107787332B (en) 2015-04-24 2022-09-09 豪夫迈·罗氏有限公司 Multispecific antigen binding proteins
JP2018520642A (en) 2015-05-01 2018-08-02 ジェネンテック, インコーポレイテッド Mask anti-CD3 antibody and method of use thereof
EP4238994A3 (en) 2015-05-11 2024-02-07 F. Hoffmann-La Roche AG Compositions and methods of treating lupus nephritis
ES2835866T3 (en) 2015-05-12 2021-06-23 Hoffmann La Roche Therapeutic and diagnostic procedures for cancer
KR20180012753A (en) 2015-05-29 2018-02-06 제넨테크, 인크. Treatment and Diagnosis Methods for Cancer
EP3302563A1 (en) 2015-05-29 2018-04-11 H. Hoffnabb-La Roche Ag Humanized anti-ebola virus glycoprotein antibodies and methods of use
CA2988420A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists
EP3303399A1 (en) 2015-06-08 2018-04-11 H. Hoffnabb-La Roche Ag Methods of treating cancer using anti-ox40 antibodies
TWI731861B (en) 2015-06-16 2021-07-01 美商建南德克公司 HUMANIZED AND AFFINITY MATURED ANTIBODIES TO FcRH5 AND METHODS OF USE
WO2016204966A1 (en) 2015-06-16 2016-12-22 Genentech, Inc. Anti-cd3 antibodies and methods of use
CN107847568B (en) 2015-06-16 2022-12-20 豪夫迈·罗氏有限公司 anti-CLL-1 antibodies and methods of use
JP2018524312A (en) 2015-06-17 2018-08-30 ジェネンテック, インコーポレイテッド Anti-HER2 antibody and method of use
DK3313879T3 (en) 2015-06-24 2022-03-14 Hoffmann La Roche Anti-transferrin receptor antibodies with adapted affinity
JP2018520153A (en) 2015-06-29 2018-07-26 ジェネンテック, インコーポレイテッド Type II anti-CD20 antibody for use in organ transplantation
CN113350518A (en) 2015-07-12 2021-09-07 杭州多禧生物科技有限公司 Conjugated bridge linkers to cell binding molecules
US9839687B2 (en) 2015-07-15 2017-12-12 Suzhou M-Conj Biotech Co., Ltd. Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
ES2944982T3 (en) 2015-08-05 2023-06-27 Janssen Biotech Inc Anti-CD154 antibodies and methods of using them
CN105384825B (en) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 A kind of bispecific chimeric antigen receptor and its application based on single domain antibody
WO2017029407A1 (en) 2015-08-20 2017-02-23 Albumedix A/S Albumin variants and conjugates
WO2017053807A2 (en) 2015-09-23 2017-03-30 Genentech, Inc. Optimized variants of anti-vegf antibodies
WO2017053906A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Hiv antibody compositions and methods of use
CN109069622A (en) 2015-09-30 2018-12-21 詹森生物科技公司 Specifically bind the antagonistic antibodies and application method of people CD40
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
KR102146319B1 (en) 2015-10-02 2020-08-25 에프. 호프만-라 로슈 아게 Bispecific antibodies specific for PD1 and TIM3
AU2016329251B2 (en) 2015-10-02 2023-02-02 F. Hoffmann-La Roche Ag Anti-PD1 antibodies and methods of use
CN114014936A (en) 2015-10-02 2022-02-08 豪夫迈·罗氏有限公司 Bispecific anti-human CD 20/human transferrin receptor antibodies and methods of use
MA43354A (en) 2015-10-16 2018-08-22 Genentech Inc CONJUGATE DRUG CONJUGATES WITH CLOUDY DISULPHIDE
MA45326A (en) 2015-10-20 2018-08-29 Genentech Inc CALICHEAMICIN-ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
EP3184547A1 (en) 2015-10-29 2017-06-28 F. Hoffmann-La Roche AG Anti-tpbg antibodies and methods of use
ES2904553T3 (en) 2015-10-30 2022-04-05 Hoffmann La Roche Hinge Modified Antibody Fragments and Preparation Procedures
BR112018008904A2 (en) 2015-11-03 2018-11-27 Janssen Biotech Inc antibodies specifically binding to tim-3 and their uses
JP6998869B2 (en) 2015-11-08 2022-02-04 ジェネンテック, インコーポレイテッド Screening method for multispecific antibody
EP3377103B1 (en) 2015-11-19 2021-03-17 Revitope Limited Functional antibody fragment complementation for a two-components system for redirected killing of unwanted cells
CA3006759A1 (en) 2015-11-30 2017-06-08 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
EP3387013B1 (en) 2015-12-07 2022-06-08 Xencor, Inc. Heterodimeric antibodies that bind cd3 and psma
JP7325186B2 (en) 2015-12-09 2023-08-14 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Type II anti-CD20 antibody for reducing the formation of anti-drug antibodies
EP3178848A1 (en) 2015-12-09 2017-06-14 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody for reducing formation of anti-drug antibodies
PL3390442T3 (en) 2015-12-18 2024-03-18 Chugai Seiyaku Kabushiki Kaisha Anti-c5 antibodies and methods of use
US11414477B2 (en) 2016-01-27 2022-08-16 Medimmune, Llc Methods for preparing antibodies with a defined glycosylation pattern
CA3019952A1 (en) 2016-02-04 2017-08-10 Curis, Inc. Mutant smoothened and methods of using the same
KR20180105155A (en) 2016-02-05 2018-09-27 릭스하스피탈렛 An antibody-drug conjugate targeting uPARAP
CN109196121B (en) 2016-02-29 2022-01-04 基因泰克公司 Methods for treatment and diagnosis of cancer
US11340233B2 (en) 2016-03-07 2022-05-24 Pierre Fabre Medicament Universal method to capture and analyze ADCs for characterization of drug distribution and the drug-to-antibody ratio in biological samples
TW201808978A (en) 2016-03-08 2018-03-16 中央研究院 Methods for modular synthesis of N-glycans and arrays thereof
WO2017172907A1 (en) * 2016-03-29 2017-10-05 Sorrento Therapeutics, Inc. Calicheamicin antibody drug conjugates linking an amidoacetyl group to a sugar moiety on calicheamicin
WO2017180864A1 (en) 2016-04-14 2017-10-19 Genentech, Inc. Anti-rspo3 antibodies and methods of use
MY198114A (en) 2016-04-15 2023-08-04 Macrogenics Inc Novel b7-h3-binding molecules, antibody drug conjugates thereof and methods of use thereof
JP2019515670A (en) 2016-04-15 2019-06-13 ジェネンテック, インコーポレイテッド Methods for monitoring and treating cancer
CA3021086C (en) 2016-04-15 2023-10-17 Bioatla, Llc Anti-axl antibodies, antibody fragments and their immunoconjugates and uses thereof
ES2850428T3 (en) 2016-04-15 2021-08-30 Hoffmann La Roche Cancer monitoring and treatment procedures
UA123323C2 (en) 2016-05-02 2021-03-17 Ф. Хоффманн-Ля Рош Аг The contorsbody - a single chain target binder
WO2017196847A1 (en) 2016-05-10 2017-11-16 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Variable new antigen receptor (vnar) antibodies and antibody conjugates targeting tumor and viral antigens
EP3455252B1 (en) 2016-05-11 2022-02-23 F. Hoffmann-La Roche AG Modified anti-tenascin antibodies and methods of use
SI3455261T1 (en) 2016-05-13 2023-01-31 Bioatla, Inc. Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
JP7022080B2 (en) 2016-05-27 2022-02-17 ジェネンテック, インコーポレイテッド Biochemical analytical methods for the characterization of site-specific antibody-drug conjugates
EP3464294B1 (en) 2016-05-30 2022-07-13 Toxinvent Oü Antibody-drug-conjugates comprising novel anthracycline-derivatives for cancer treatment
EP3252078A1 (en) 2016-06-02 2017-12-06 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody and anti-cd20/cd3 bispecific antibody for treatment of cancer
TW201902512A (en) 2016-06-02 2019-01-16 瑞士商赫孚孟拉羅股份公司 treatment method
WO2017214182A1 (en) 2016-06-07 2017-12-14 The United States Of America. As Represented By The Secretary, Department Of Health & Human Services Fully human antibody targeting pdi for cancer immunotherapy
WO2017214456A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-cd98 antibodies and antibody drug conjugates
EP3468997B1 (en) 2016-06-08 2023-09-13 Xencor, Inc. Treatment of igg4-related diseases with anti-cd19 antibodies crossbinding to cd32b
WO2017214335A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
CN109563167A (en) 2016-06-08 2019-04-02 艾伯维公司 Anti- B7-H3 antibody and antibody drug conjugates
CA3027103A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
EP3468599A2 (en) 2016-06-08 2019-04-17 AbbVie Inc. Anti-cd98 antibodies and antibody drug conjugates
MX2018015592A (en) 2016-06-14 2019-04-24 Xencor Inc Bispecific checkpoint inhibitor antibodies.
US11617799B2 (en) 2016-06-27 2023-04-04 Tagworks Pharmaceuticals B.V. Cleavable tetrazine used in bio-orthogonal drug activation
CA3029328A1 (en) 2016-06-28 2018-01-04 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
JP2019524706A (en) 2016-07-08 2019-09-05 ジェネンテック, インコーポレイテッド Use of human epididymis protein 4 (HE4) to assess responsiveness of MUC16 positive cancer treatment
CN110088133B (en) 2016-07-29 2023-12-08 朱诺治疗学股份有限公司 Anti-idiotype antibodies and related methods
AU2017305170A1 (en) 2016-08-02 2019-02-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies targeting glypican-2 (GPC2) and use thereof
JP2018058822A (en) 2016-08-03 2018-04-12 ファイザー・インク Heteroaryl sulfone-based conjugation handles, methods for their preparation, and their use in synthesizing antibody drug conjugates
CN116251182A (en) 2016-08-05 2023-06-13 中外制药株式会社 Compositions for preventing or treating IL-8 related diseases
EP3494139B1 (en) 2016-08-05 2022-01-12 F. Hoffmann-La Roche AG Multivalent and multiepitopic anitibodies having agonistic activity and methods of use
JP7213549B2 (en) 2016-08-22 2023-01-27 シーエイチオー ファーマ インコーポレイテッド Antibodies, Binding Fragments, and Methods of Use
WO2018036852A1 (en) 2016-08-25 2018-03-01 F. Hoffmann-La Roche Ag Intermittent dosing of an anti-csf-1r antibody in combination with macrophage activating agent
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
CN109689682B (en) 2016-09-19 2022-11-29 豪夫迈·罗氏有限公司 Complement factor-based affinity chromatography
AU2017339517B2 (en) 2016-10-06 2024-03-14 Foundation Medicine, Inc. Therapeutic and diagnostic methods for cancer
AU2017342559B2 (en) 2016-10-14 2022-03-24 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15/IL-15Ralpha Fc-fusion proteins and PD-1 antibody fragments
KR102617264B1 (en) 2016-10-19 2023-12-29 인벤라 인코포레이티드 antibody structure
EP3532091A2 (en) 2016-10-29 2019-09-04 H. Hoffnabb-La Roche Ag Anti-mic antibidies and methods of use
NZ752394A (en) 2016-11-14 2021-07-30 Hangzhou Dac Biotech Co Ltd Conjugation linkers, cell binding molecule-drug conjugates containing the likers, methods of making and uses such conjugates with the linkers
AU2017361081A1 (en) 2016-11-15 2019-05-23 Genentech, Inc. Dosing for treatment with anti-CD20/anti-CD3 bispecific antibodies
TW201829463A (en) 2016-11-18 2018-08-16 瑞士商赫孚孟拉羅股份公司 Anti-hla-g antibodies and use thereof
AU2017361887B2 (en) 2016-11-21 2019-08-15 Cureab Gmbh Anti-GP73 antibodies and immunoconjugates
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
US11236171B2 (en) 2016-12-21 2022-02-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies specific for FLT3 and uses thereof
EA201991204A1 (en) 2016-12-22 2019-12-30 Университа Дельи Студи Манья Греча Катандзаро MONOCLONAL ANTIBODY AGAINST UNIQUE Sialoglycosylated Tumor-Associated Epitope CD43
JP7304287B2 (en) 2016-12-22 2023-07-06 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Treatment of tumors with anti-CSF-1R antibodies in combination with anti-PD-L1 antibodies after unsuccessful anti-PD-L1/PD1 therapy
US20190345186A1 (en) * 2017-01-24 2019-11-14 Pfizer Inc. Calicheamicin deratives and antibody drug conjugates thereof
AR110873A1 (en) 2017-02-10 2019-05-08 Genentech Inc ANTIBODIES AGAINST TRIPTASE, COMPOSITIONS OF THESE AND USES OF THESE
TW201837467A (en) 2017-03-01 2018-10-16 美商建南德克公司 Diagnostic and therapeutic methods for cancer
US20220135670A1 (en) 2017-04-27 2022-05-05 Tesaro, Inc. Antibody agents directed against lymphocyte activation gene-3 (lag-3) and uses thereof
US11389480B2 (en) 2017-05-19 2022-07-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibody targeting TNFR2 for cancer immunotherapy
AR111963A1 (en) 2017-05-26 2019-09-04 Univ California METHOD AND MOLECULES
CA3066754A1 (en) 2017-06-22 2018-12-27 Mersana Therapeutics, Inc. Methods of producing drug-carrying polymer scaffolds and protein-polymer-drug conjugates
WO2019005208A1 (en) 2017-06-30 2019-01-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human mesothelin antibodies and uses in cancer therapy
JP2020529832A (en) 2017-06-30 2020-10-15 ゼンコア インコーポレイテッド Targeted heterodimer Fc fusion protein containing IL-15 / IL-15Rα and antigen binding domain
WO2019006280A1 (en) 2017-06-30 2019-01-03 Lentigen Technology, Inc. Human monoclonal antibodies specific for cd33 and methods of their use
US20190048073A1 (en) 2017-07-20 2019-02-14 Pfizer Inc. Anti-gd3 antibodies and antibody-drug conjugates
JP2020527351A (en) 2017-07-21 2020-09-10 ジェネンテック, インコーポレイテッド Cancer treatment and diagnosis
JP7232190B2 (en) 2017-10-20 2023-03-02 中外製薬株式会社 Methods for Measuring Uptake of Molecules into Cells
CA3078676A1 (en) 2017-10-30 2019-05-09 F. Hoffmann-La Roche Ag Method for in vivo generation of multispecific antibodies from monospecific antibodies
PL3704146T3 (en) 2017-11-01 2022-03-07 F. Hoffmann-La Roche Ag Trifab-contorsbody
CA3080904A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
EP3704150A1 (en) 2017-11-01 2020-09-09 F. Hoffmann-La Roche AG The compbody - a multivalent target binder
JP2021502066A (en) 2017-11-06 2021-01-28 ジェネンテック, インコーポレイテッド Cancer diagnosis and therapy
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
EP3706793A1 (en) 2017-11-08 2020-09-16 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-pd-1 sequences
WO2019125732A1 (en) 2017-12-19 2019-06-27 Xencor, Inc. Engineered il-2 fc fusion proteins
WO2019131988A1 (en) 2017-12-28 2019-07-04 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP3740505A1 (en) 2018-01-16 2020-11-25 Lakepharma Inc. Bispecific antibody that binds cd3 and another target
AU2019218959A1 (en) 2018-02-08 2020-09-03 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
WO2019165434A1 (en) 2018-02-26 2019-08-29 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
US20200040103A1 (en) 2018-03-14 2020-02-06 Genentech, Inc. Anti-klk5 antibodies and methods of use
SG11202008796VA (en) 2018-03-14 2020-10-29 The United States Of America As Represented By The Secretary Anti-cd33 chimeric antigen receptors and their uses
CA3093729A1 (en) 2018-03-15 2019-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies having cross-reactivity to zika virus and methods of use
WO2019192432A1 (en) 2018-04-02 2019-10-10 上海博威生物医药有限公司 Lymphocyte activation gene-3 (lag-3) binding antibody and use thereof
TW202011029A (en) 2018-04-04 2020-03-16 美商建南德克公司 Methods for detecting and quantifying FGF21
AU2019247415A1 (en) 2018-04-04 2020-10-22 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
AU2019256529A1 (en) 2018-04-18 2020-11-26 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and TIM-3 antigen binding domains
AR114789A1 (en) 2018-04-18 2020-10-14 Hoffmann La Roche ANTI-HLA-G ANTIBODIES AND THE USE OF THEM
CA3097593A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof
DK3788032T3 (en) 2018-05-04 2024-04-15 Tagworks Pharmaceuticals B V COMPOUNDS COMPRISING A LINKER TO INCREASE TRANSCYCLOOCTEN STABILITY
CA3099419A1 (en) 2018-05-04 2019-11-07 Tagworks Pharmaceuticals B.V. Tetrazines for high click conjugation yield in vivo and high click release yield
ES2955511T3 (en) 2018-05-14 2023-12-04 Werewolf Therapeutics Inc Activatable interleukin 2 polypeptides and methods of use thereof
JP2021524756A (en) 2018-05-14 2021-09-16 ウェアウルフ セラピューティクス, インコーポレイテッド Activateable cytokine polypeptides and how to use them
EP3802609A2 (en) 2018-05-24 2021-04-14 Janssen Biotech, Inc. Psma binding agents and uses thereof
EP3802580A1 (en) 2018-06-05 2021-04-14 King's College London Btnl3/8 targeting constructs for delivery of payloads to the gastrointestinal system
WO2020010079A2 (en) 2018-07-02 2020-01-09 Amgen Inc. Anti-steap1 antigen-binding protein
EP3820903A1 (en) 2018-07-12 2021-05-19 The United States of America, as represented by the Secretary, Department of Health and Human Services Affinity matured cd22-specific monoclonal antibody and uses thereof
WO2020033430A1 (en) 2018-08-08 2020-02-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services High affinity monoclonal antibodies targeting glypican-2 and uses thereof
WO2020032230A1 (en) 2018-08-10 2020-02-13 中外製薬株式会社 Anti-cd137 antigen-binding molecule and utilization thereof
EP4268831A2 (en) 2018-09-12 2023-11-01 Fred Hutchinson Cancer Center Reducing cd33 expression to selectively protect therapeutic cells
AU2019342099A1 (en) 2018-09-19 2021-04-08 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
MX2021003382A (en) 2018-09-26 2021-05-27 Jiangsu Hengrui Medicine Co Ligand-drug conjugate of exatecan analogue, preparation method therefor and application thereof.
JP2022502088A (en) 2018-09-27 2022-01-11 エクシリオ デベロップメント, インコーポレイテッド Masked cytokine polypeptide
JP2022503959A (en) 2018-10-03 2022-01-12 ゼンコア インコーポレイテッド IL-12 heterodimer FC-fusion protein
MX2021004348A (en) 2018-10-18 2021-05-28 Genentech Inc Diagnostic and therapeutic methods for sarcomatoid kidney cancer.
CN113365664A (en) 2018-10-29 2021-09-07 梅尔莎纳医疗公司 Cysteine engineered antibody-drug conjugates with peptide-containing linkers
CA3118816A1 (en) 2018-11-07 2020-05-14 Crispr Therapeutics Ag Anti-cd33 immune cell cancer therapy
EP3883610A4 (en) 2018-11-20 2022-11-02 Cornell University Macrocyclic complexes of radionuclides and their use in radiotherapy of cancer
MX2021006573A (en) 2018-12-06 2021-07-15 Genentech Inc Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody.
EP3898667A2 (en) 2018-12-20 2021-10-27 F. Hoffmann-La Roche AG Modified antibody fcs and methods of use
WO2020127968A1 (en) 2018-12-20 2020-06-25 Marino Stephen F Protein-drug conjugate comprising a monomeric form of proteinase 3
EP3902833A2 (en) 2018-12-26 2021-11-03 City of Hope Activatable masked anti-ctla4 binding proteins
WO2020146182A1 (en) 2019-01-08 2020-07-16 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cross-species single domain antibodies targeting mesothelin for treating solid tumors
WO2020154405A2 (en) 2019-01-22 2020-07-30 Genentech, Inc. Immunoglobulin a antibodies and methods of production and use
CA3125033A1 (en) 2019-01-22 2020-07-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services High affinity monoclonal antibodies targeting glypican-1 and methods of use
JPWO2020153467A1 (en) 2019-01-24 2021-12-02 中外製薬株式会社 New cancer antigens and antibodies against those antigens
PE20212198A1 (en) 2019-01-29 2021-11-16 Juno Therapeutics Inc ANTIBODIES AND CHIMERIC RECEPTORS OF SPECIFIC ANTIGENS TO ORPHAN RECEPTOR 1, RECEPTOR TYROSINE KINASE TYPE (ROR1)
CN113710706A (en) 2019-02-27 2021-11-26 豪夫迈·罗氏有限公司 Administration for anti-TIGIT antibody and anti-CD 20 antibody or anti-CD 38 antibody treatment
WO2020180726A1 (en) 2019-03-01 2020-09-10 Xencor, Inc. Heterodimeric antibodies that bind enpp3 and cd3
MA55296A (en) 2019-03-14 2022-03-23 Hoffmann La Roche CANCER TREATMENT WITH BISPECIFIC ANTIBODIES TO HER2XCD3 IN COMBINATION WITH AN ANTI-HER2 MAB
CA3133155A1 (en) 2019-03-19 2020-09-24 Fundacio Privada Institut D'investigacio Oncologica De Vall Hebron Combination therapy for the treatment for cancer
CN114364703A (en) 2019-04-19 2022-04-15 豪夫迈·罗氏有限公司 Anti-merk antibodies and methods of use thereof
CN113747944A (en) 2019-04-19 2021-12-03 詹森生物科技公司 Methods of treating prostate cancer with anti-PSMA/CD 3 antibodies
JP2022536602A (en) 2019-05-14 2022-08-18 ジェネンテック, インコーポレイテッド Methods of using anti-CD79B immunoconjugates to treat follicular lymphoma
JP2022532217A (en) 2019-05-14 2022-07-13 ウェアウルフ セラピューティクス, インコーポレイテッド Separation part and how to use it
CA3137649A1 (en) 2019-05-15 2020-11-19 Chugai Seiyaku Kabushiki Kaisha An antigen-binding molecule, a pharmaceutical composition, and a method
CA3144535A1 (en) 2019-06-03 2020-12-10 The University Of Chicago Methods and compositions for treating cancer with collagen binding drug carriers
CA3140802A1 (en) * 2019-06-12 2020-12-17 Ontario Institute For Cancer Research (Oicr) Unsaturated heterocycloalkyl and heteroaromatic acyl hydrazone linkers, methods and uses thereof
IL289094A (en) 2019-06-17 2022-02-01 Tagworks Pharmaceuticals B V Tetrazines for high click release speed and yield
EP3983363B1 (en) 2019-06-17 2024-04-10 Tagworks Pharmaceuticals B.V. Compounds for fast and efficient click release
CN113950485A (en) 2019-07-10 2022-01-18 中外制药株式会社 Claudin-6 binding molecules and uses thereof
JPWO2021010326A1 (en) 2019-07-12 2021-01-21
WO2021030780A1 (en) 2019-08-14 2021-02-18 Codiak Biosciences, Inc. Extracellular vesicle-aso constructs targeting cebp/beta
CA3147366A1 (en) 2019-08-14 2021-02-18 Adam T. BOUTIN Extracellular vesicles with stat3-antisense oligonucleotides
JP2022544935A (en) 2019-08-14 2022-10-24 コディアック バイオサイエンシーズ, インコーポレイテッド Extracellular vesicles - NLRP3 antagonists
MX2022001767A (en) 2019-08-14 2022-06-09 Codiak Biosciences Inc Extracellular vesicle-aso constructs targeting stat6.
CA3150807A1 (en) 2019-09-04 2021-03-11 Y-Biologics Inc. Anti-vsig4 antibody or antigen binding fragment and uses thereof
CR20220127A (en) 2019-09-27 2022-05-27 Genentech Inc Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
CA3154710A1 (en) 2019-10-04 2021-04-08 TAE Life Sciences Antibody compositions comprising fc mutations and site-specific conjugation properties
JP7413519B2 (en) 2019-10-18 2024-01-15 ジェネンテック, インコーポレイテッド Methods of using anti-CD79B immunoconjugates to treat diffuse large B-cell lymphoma
EP4031250A1 (en) 2019-10-22 2022-07-27 The United States of America, as represented by the Secretary, Department of Health and Human Services High affinity nanobodies targeting b7h3 (cd276) for treating multiple solid tumors
US20220389103A1 (en) 2019-11-06 2022-12-08 Genentech, Inc. Diagnostic and therapeutic methods for treatment of hematologic cancers
EP4247957A1 (en) 2019-11-22 2023-09-27 MedImmune Limited Fusion proteins comprising an e2 ubiquitin or ubiquitin-like conjugating domain and a targeting domain for specific protein degradation
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
JP2023504740A (en) 2019-12-06 2023-02-06 ジュノー セラピューティクス インコーポレイテッド Anti-idiotypic antibodies against BCMA target binding domains and related compositions and methods
AU2020402752A1 (en) 2019-12-12 2022-06-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibody-drug conjugates specific for CD276 and uses thereof
PE20221511A1 (en) 2019-12-13 2022-10-04 Genentech Inc ANTI-LY6G6D ANTIBODIES AND METHODS OF USE
US20230058982A1 (en) 2019-12-27 2023-02-23 Chugai Seiyaku Kabushiki Kaisha Anti-ctla-4 antibody and use thereof
CN110818795B (en) 2020-01-10 2020-04-24 上海复宏汉霖生物技术股份有限公司 anti-TIGIT antibodies and methods of use
JP2023511163A (en) 2020-01-22 2023-03-16 上海森輝医薬有限公司 Drug conjugate of eribulin derivative, preparation method thereof and pharmaceutical application thereof
WO2021194481A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
WO2022050954A1 (en) 2020-09-04 2022-03-10 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
TW202144395A (en) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 Anti-CD137 antigen-binding molecule for use in cancer treatment
WO2021183849A1 (en) 2020-03-13 2021-09-16 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
EP4121163A1 (en) 2020-03-19 2023-01-25 Genentech, Inc. Isoform-selective anti-tgf-beta antibodies and methods of use
CN115103858A (en) 2020-03-25 2022-09-23 江苏恒瑞医药股份有限公司 Preparation method of antibody drug conjugate
BR112022019073A2 (en) 2020-03-25 2022-11-08 Jiangsu Hengrui Pharmaceuticals Co Ltd PHARMACEUTICAL COMPOSITION INCLUDING ANTIBODY-DRUG CONJUGATE AND USE THEREOF
CN115698717A (en) 2020-04-03 2023-02-03 基因泰克公司 Methods of treatment and diagnosis of cancer
TW202206111A (en) 2020-04-24 2022-02-16 美商建南德克公司 Methods of using anti-cd79b immunoconjugates
EP4143345A1 (en) 2020-04-28 2023-03-08 Genentech, Inc. Methods and compositions for non-small cell lung cancer immunotherapy
WO2021231976A1 (en) 2020-05-14 2021-11-18 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (psma) and cd3
US20230220068A1 (en) 2020-06-05 2023-07-13 Codiak Biosciences, Inc. Anti-transferrin extracellular vesicles
MX2022015651A (en) 2020-06-11 2023-01-16 Genentech Inc Nanolipoprotein-polypeptide conjugates and compositions, systems, and methods using same.
EP4165415A1 (en) 2020-06-12 2023-04-19 Genentech, Inc. Methods and compositions for cancer immunotherapy
EP4168118A1 (en) 2020-06-18 2023-04-26 Genentech, Inc. Treatment with anti-tigit antibodies and pd-1 axis binding antagonists
WO2022015726A1 (en) 2020-07-13 2022-01-20 Genentech, Inc. Cell-based methods for predicting polypeptide immunogenicity
WO2022025184A1 (en) 2020-07-29 2022-02-03 中外製薬株式会社 Method for measuring pharmacokinetics of drug labeled with non-radioactive substance
EP4192868A1 (en) 2020-08-05 2023-06-14 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
CA3188426A1 (en) 2020-08-07 2022-02-10 Yichin Liu Flt3 ligand fusion proteins and methods of use
JP2023537683A (en) 2020-08-07 2023-09-05 ジェネンテック, インコーポレイテッド T cell-based methods for predicting polypeptide immunogenicity
JP2023541456A (en) 2020-09-14 2023-10-02 ブイオーアール バイオファーマ インコーポレーテッド Chimeric antigen receptor for cancer treatment
CN116406291A (en) 2020-10-05 2023-07-07 基因泰克公司 Administration of treatment with anti-FCRH 5/anti-CD 3 bispecific antibodies
WO2022093745A1 (en) 2020-10-26 2022-05-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Single domain antibodies targeting sars coronavirus spike protein and uses thereof
JP2023548111A (en) 2020-10-27 2023-11-15 ブイオーアール バイオファーマ インコーポレーテッド Compositions and methods for treating hematopoietic malignancies
US20220162329A1 (en) 2020-11-04 2022-05-26 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
EP4240766A2 (en) 2020-11-04 2023-09-13 Genentech, Inc. Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
JP2023548064A (en) 2020-11-04 2023-11-15 ジェネンテック, インコーポレイテッド Administration for treatment with anti-CD20/anti-CD3 bispecific antibody and anti-CD79B antibody drug conjugate
UY39610A (en) 2021-01-20 2022-08-31 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
BR112023015097A2 (en) 2021-01-28 2023-10-03 Janssen Biotech Inc PSMA-BINDING PROTEINS AND USES THEREOF
EP4301781A1 (en) 2021-03-01 2024-01-10 Xilio Development, Inc. Combination of masked ctla4 and pd1/pdl1 antibodies for treating cancer
TW202317612A (en) 2021-03-01 2023-05-01 美商艾希利歐發展股份有限公司 Combination of ctla4 and pd1/pdl1 antibodies for treating cancer
WO2022184853A1 (en) 2021-03-03 2022-09-09 Pierre Fabre Medicament Anti-vsig4 antibody or antigen binding fragment and uses thereof
EP4305067A1 (en) 2021-03-09 2024-01-17 Xencor, Inc. Heterodimeric antibodies that bind cd3 and cldn6
WO2022192586A1 (en) 2021-03-10 2022-09-15 Xencor, Inc. Heterodimeric antibodies that bind cd3 and gpc3
AR125344A1 (en) 2021-04-15 2023-07-05 Chugai Pharmaceutical Co Ltd ANTI-C1S ANTIBODY
WO2022232612A1 (en) 2021-04-29 2022-11-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Lassa virus-specific nanobodies and methods of their use
EP4172212A1 (en) 2021-04-30 2023-05-03 Pierre Fabre Medicament New stable anti-vista antibody
EP4330282A1 (en) 2021-04-30 2024-03-06 F. Hoffmann-La Roche AG Dosing for combination treatment with anti-cd20/anti-cd3 bispecific antibody and anti-cd79b antibody drug conjugate
AU2021443863A1 (en) 2021-04-30 2023-10-26 F. Hoffmann-La Roche Ag Dosing for treatment with anti-cd20/anti-cd3 bispecific antibody
IL308351A (en) 2021-05-12 2024-01-01 Genentech Inc Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2022244838A1 (en) 2021-05-19 2022-11-24 中外製薬株式会社 Method for predicting in vivo pharmacokinetics of molecule
BR112023024804A2 (en) 2021-05-28 2024-02-15 Glaxosmithkline Ip Dev Ltd COMBINATION THERAPIES TO TREAT CANCER
TW202306994A (en) 2021-06-04 2023-02-16 日商中外製藥股份有限公司 Anti-ddr2 antibodies and uses thereof
CA3216228A1 (en) 2021-06-09 2022-12-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cross species single domain antibodies targeting pd-l1 for treating solid tumors
JP7472405B2 (en) 2021-06-25 2024-04-22 中外製薬株式会社 Anti-CTLA-4 antibody
WO2022270612A1 (en) 2021-06-25 2022-12-29 中外製薬株式会社 Use of anti-ctla-4 antibody
US11807685B2 (en) 2021-08-05 2023-11-07 The Uab Research Foundation Anti-CD47 antibody and uses thereof
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023017159A1 (en) 2021-08-13 2023-02-16 Ludwig-Maximilians-Universität München Anti-csf1r car expressing lymphocytes for targeted tumor therapy
CA3230774A1 (en) 2021-09-06 2023-03-09 Veraxa Biotech Gmbh Novel aminoacyl-trna synthetase variants for genetic code expansion in eukaryotes
TW202321308A (en) 2021-09-30 2023-06-01 美商建南德克公司 Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
CA3233953A1 (en) 2021-10-05 2023-04-13 Matthew Bruce Combination therapies for treating cancer
WO2023094525A1 (en) 2021-11-25 2023-06-01 Veraxa Biotech Gmbh Improved antibody-payload conjugates (apcs) prepared by site-specific conjugation utilizing genetic code expansion
EP4186529A1 (en) 2021-11-25 2023-05-31 Veraxa Biotech GmbH Improved antibody-payload conjugates (apcs) prepared by site-specific conjugation utilizing genetic code expansion
WO2023104941A1 (en) 2021-12-08 2023-06-15 European Molecular Biology Laboratory Hydrophilic tetrazine-functionalized payloads for preparation of targeting conjugates
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023158305A1 (en) 2022-02-15 2023-08-24 Tagworks Pharmaceuticals B.V. Masked il12 protein
TW202346365A (en) 2022-03-23 2023-12-01 瑞士商赫孚孟拉羅股份公司 Combination treatment of an anti-cd20/anti-cd3 bispecific antibody and chemotherapy
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
US20230406930A1 (en) 2022-04-13 2023-12-21 Genentech, Inc. Pharmaceutical compositions of therapeutic proteins and methods of use
WO2023198727A1 (en) 2022-04-13 2023-10-19 F. Hoffmann-La Roche Ag Pharmaceutical compositions of anti-cd20/anti-cd3 bispecific antibodies and methods of use
US20230346862A1 (en) 2022-05-02 2023-11-02 Athanor Biosciences, Inc. Cancer eradicating - bio-nanoparticles (ce-bnp)
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024073751A1 (en) 2022-09-29 2024-04-04 Vor Biopharma Inc. Methods and compositions for gene modification and enrichment
WO2024080872A1 (en) 2022-10-12 2024-04-18 Tagworks Pharmaceuticals B.V. Strained bicyclononenes

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5053394A (en) * 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
EP0329184A3 (en) * 1988-02-19 1990-05-23 Neorx Corporation Antimers and antimeric conjugation
US5028697A (en) * 1988-08-08 1991-07-02 Eli Lilly And Company Cytotoxic antibody conjugates of hydrazide derivatized methotrexate analogs via simple organic linkers
US5144012A (en) * 1988-08-08 1992-09-01 Eli Lilly And Company Cytotoxic drug conjugates
US5006652A (en) * 1988-08-08 1991-04-09 Eli Lilly And Company Intermediates for antibody-vinca drug conjugates
US5094849A (en) * 1988-08-08 1992-03-10 Eli Lilly And Company Cytotoxic antibody conjugates of hydrazide derivatized vinca analogs via simple organic linkers
US5045451A (en) * 1988-10-26 1991-09-03 Board Of Regents Methods for screening antibodies for use as immunotoxins
IL93733A (en) * 1989-04-14 1996-01-19 American Cyanamid Co Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group and targeted forms thereof
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
GB9120467D0 (en) * 1991-09-26 1991-11-06 Celltech Ltd Anti-hmfg antibodies and process for their production
US5773001A (en) * 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis

Also Published As

Publication number Publication date
NO952206D0 (en) 1995-06-02
AU697280B2 (en) 1998-10-01
US5773001A (en) 1998-06-30
TW394778B (en) 2000-06-21
IL113984A (en) 2000-09-28
KR960000234A (en) 1996-01-25
BR1100990A (en) 2002-04-02
IL113984A0 (en) 1995-10-31
NO952206L (en) 1995-12-04
NZ536440A (en) 2007-08-31
CY2334B1 (en) 2004-02-06
DE69528016T2 (en) 2003-01-02
ATE223234T1 (en) 2002-09-15
PT689845E (en) 2002-12-31
US5767285A (en) 1998-06-16
SI0689845T1 (en) 2002-12-31
JP2005139200A (en) 2005-06-02
NZ272274A (en) 1997-11-24
DE69528016D1 (en) 2002-10-10
EP0689845B1 (en) 2002-09-04
US5877296A (en) 1999-03-02
ES2181752T3 (en) 2003-03-01
FI117690B (en) 2007-01-31
EP0689845A2 (en) 1996-01-03
NZ328762A (en) 2000-08-25
KR100408376B1 (en) 2004-03-09
JPH0848637A (en) 1996-02-20
JP3650165B2 (en) 2005-05-18
FI952720A0 (en) 1995-06-02
FI952720A (en) 1995-12-04
US5739116A (en) 1998-04-14
ZA954570B (en) 1996-05-22
NO313617B1 (en) 2002-11-04
AU2044995A (en) 1995-12-14
BR1101078A (en) 1999-10-26
DK0689845T3 (en) 2003-01-06
CA2150785A1 (en) 1995-12-04
EP0689845A3 (en) 1999-08-25

Similar Documents

Publication Publication Date Title
CA2150785C (en) Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
EP0837698B1 (en) Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
CA1303524C (en) Drug-monoclonal antibody conjugates
DK175174B1 (en) Anthracycline immunoconjugates with a novel linker and methods for their preparation
CN105636591B (en) Ligand-cytotoxic drug conjugate, preparation method and application thereof
JPH09500125A (en) Prodrugs of protein tyrosine kinase inhibitors
HU210147B (en) Process for producing antibody-drug conjugates and pharmaceutical compositions containing them
FI95204B (en) Process for the preparation of targeted derivatives of methyltrithio compounds
US20080139493A1 (en) Bi- or Tetra-Guanidino-Biphenyl Compounds as Small Molecule Carriers
CN113444111A (en) Novel benzodiazepine derivative and use thereof
IL132974A (en) Compounds useful in the synthesis of conjugates of methyltrithio anti-tumour agents
KR20220108783A (en) Compositions and Methods Related to Molecular Conjugation
WO1992014758A1 (en) Conjugate molecules

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20150601