CA1341315C - Targeted forms of methyltrithio antitumor agents - Google Patents

Targeted forms of methyltrithio antitumor agents Download PDF

Info

Publication number
CA1341315C
CA1341315C CA000581577A CA581577A CA1341315C CA 1341315 C CA1341315 C CA 1341315C CA 000581577 A CA000581577 A CA 000581577A CA 581577 A CA581577 A CA 581577A CA 1341315 C CA1341315 C CA 1341315C
Authority
CA
Canada
Prior art keywords
formula
alpha
compound
divalent
radical
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA000581577A
Other languages
French (fr)
Inventor
William James Mcgahren
Martin Leon Sassiver
George A. Ellestad
Philip R. Hamann
Lois M. Hinman
Janis Upeslacis
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
American Cyanamid Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by American Cyanamid Co filed Critical American Cyanamid Co
Application granted granted Critical
Publication of CA1341315C publication Critical patent/CA1341315C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This disclosure describes a method for constructing carrier-drug conjugates from the family of methyltrithio antibacterial and antitumor agents.

Description

TARGETED FORMS OF METHYLTRITHIO
ANTITUMOR AGENTS
SUMMARY OF THE INVENTION
The invention describes carrier-drug conjugates of (3 Yl and b the disulfide analogs of the Ocl, Ocz, Oc3, Oc4, (31, z.
components of the LL-E33288 complex as well as the disulfide analogs of BBM-1675, FR-900405, FR-900406, PD 114759, PD
115028, CL-1577A, CL-1577B, CL-1577D, CL-1577E and CL-1724 antitumor antibiotics. The carrier portion of the conjugate is a mono- or polyclonal antibody, their fragments, chemically or genetically manipulated counterparts, growth factors or steroids. The invention includes the method of using the carrier-drug conjugates as well as their process of manufacture. Conjugates of the invention can be used to inhibit growth of tumors in mammals. Commercial packages of pharmaceutically effective amounts of such conjugates together with instructions for such a use are also part of the invention.
DESCRIPTION OF THE DRAWINGS
Figure 1: The ultraviolet spectrum of the antitumor antibiotic designated as LL-E33288Y1i.
Figure 2: The proton magnetic resonance spectrum of the antitumor antibiotic designated as LL-E33288Y1I.
Figure 3: The infrared spectrum of the antitumor antibiotic designated as LL-E33288Y1i.
DETAILED DESCRIPTION OF THE INVENTION
The family of antibacterial and arrtitumor agents, known collectively as the LL-E33288 complex are described and claimed in U.S. Patent 4,970,198 and are used to prepare
-2- 1341315 the disulfur antitumor agents which are starting materials for targeted forms of the antitumor agents of our invention.
U.S. Patent 4,970,198 describes the LL-E33288 complex, the components thereof, namely, LL-E33288a1Sr, LL-E33288a1I, LL-E33288a2Br, LL-E33288a I
, LL-E33288a3Br, LL-E33288a3I, LL-E33288a4$r, LL-E33288~~Br, LL-E33288~ , LL-E33288 H Sr 1 R2 , LL-E33288~2 , LL-E33288~1 r, LL-E33288~1I and LL-E33288d1I, and methods for their production by aerobic fermentation utilizing a new strain of Micromono-spora echinospora ssp calichensi.s or natural or derived mutants thereof. U.5. Patent 4,970,198 also discloses proposed structures for some of the above named components. These proposed structures are reproduced in Table 1.

tB I

_3_ 1341315 Table 1 Proposed Structures for CH3-SSS-W
(wherein W is the substituent attached to CH3-SSS- below) s O
HO~", H ,....- \ N H O
_ CHI O CH~SSS.~ /~,, R'S O C ~CH~
\ ~ o o.... ''~,,~
OH HN~~~ H
ORz CHI O CH~O CO-X
Are ~ Ars ~ C ~O ~ N~.H
R, O O C 1"!~ ~ O O C hip OCH~
C H=
. '/~/~ R 'O 'iw' R2, = N O R'~ ~ ORS R,, = C H~ O
~~ H
Rs OCh4~ CHI 'O "'y OCH~ OH
~O
I
Deslgnatlon R~ R2 R3 R4 Rg Rg R~ X
LL-E33288a21 Ark R2, H H C2H5 ~ I

LL-E33288a31 Ar1 H H R4, ~ I

LL-E33288~i~ Ark R2, H R4, (CH3)2C H I
I

LLE33288y~ Ar1 R2, H R4, C2H5 i I

LLE332885~1 Ar1 R2. H R4, CH3 I

LL-E33288p~8r Ari R2, H R4, (CN3)2CH er LL-E33288y1 Ark R2, H R4, C2H5 Br B r LL-E33288a2B Ar1 R2, H H C2H6 Br r LL-E33288a3B Ark H H Ra, Br r Esperamicin CH3 R2, R3, (CH3)2CH H Ar2 Esperamlcln CH3 R2, R3, (CH3)2C H Ar2 H

Esperamicln CH3 R2, R3, CH3CH2 H Ar2 A1 b Certain other antibiotics are useful in our inven-tion, namely:
1) Esperamicin BBM-1675, a novel class of potent antitumor antibiotics. I. Physico-chemical data and partial structure.
M. Konishi, et. al., J. Antibiotics, 38, 1605 (1985). A new antitumor antibiotic complex, M. Konishi, et. al., U.S.
Patent 4.675,187.
2) New antitumor antibiotics, FR-900405 and FR-900406. I.
Taxonomy of the producing strain. M. Iwami, et. a~., J. Anti-biotics 38, 835 (1985). New antitumor antibiotics FR-900405 and FR-900406.II. Production, isolation, characterization and antitumor activity. S. Kiyoto, et. al., J. Antibiotics, 38, 340 (1985).
3) PD 114759 and PD 115028, novel antitumor antibiotics with phenomenal potency. I. Isolation and characterization. R.H.
Bunge, et. al., J. Antibiotics, 37, 1566 (1984). Biological and biochemical activities of the novel antitumor antibiotic PD 114759 and related derivatives. D.W. Fry et. al., Investigational New Drugs, 4, 3 (1986).
4) New antibiotic complex CL-1577A, CL-1577B produced by Streptomyces sp. ATCC 39363. European Patent Application 0,132,082, A2.
5) CL-1577D and CL-1577E Antibiotic antitumor compounds, their production and use. U.S. Patent 4,539,203.
6) CL-1724 Antibiotic compounds, their production and use.
U.S. Patent 4,554,162.
The complete structures of esperamicins A1. A2' and Alb (the BBM-1675 complex) have been reported, and these are included in Table 1. Tne physical characteristics of the above-named antitumor antibiotics indicate that they all are identical or very similar in structure to the esperamicins, and all contain a methyltrithio functional group.

As can be seen from the structures disclosed above, the a1, a2, a3, a4. ~l' ~2' 71 and a components of the LL-E33288 complex, as well as the BBM-1675, FR-900405, FR-900406, PD 114759, PD 115028, CL-1577A, CL-1577B, CL-1577D, CL-1577E
and CL-1724 antibiotics each contain a methyltrithio group in their structure. The methyltrithio moiety of the above-named antibiotics is subject to displacement by a variety of thiol-containing organic molecules resulting in the formation of a new class of anticancer and antibacterial agents.
It has now been discovered that the displacement of the methyltrithio unit of the compounds listed in Table 1 and as depicted in Scheme I can be used to introduce a spacer (Sp), the judicious choice of which enable the introduction of targeting units into the compounds of the above-named pat-ents and applications.
Q-Sp-SH
CH3SSS-W ~ Q-Sp-SS-W
Cnhcmc T
wherein Sp is a straight or branched-chain divalent (C1-C18) radical, divalent aryl or heteroaryl radicals, divalent (C3-C18) cycloalkyl or heterocycloalkyl radicals, divalent aryl-or heteroaryl-alkyl (C1-C18) radicals, divalent cycloalkyl-or heterocycloalkyl-alkyl (C1-C18) radicals, or divalent (C2-C18) unsaturated alkyl radicals; Q is, or can be subsequently converted to, halogen, amino, alkylamino, carboxyl, carboxal-dehyde, hydroxy, thiol, a-haloacetyloxy, lower alkyldicar-boxyl, -CONHNH2, -NHCONHNH2, -NHCSNHNH2, -ONH2, -CONS, O O F F
-SS
N

-C02N ~ ~ ~ N-.
\O
O F F

C02 N I O _ O
-C02 O F. -C02 O -N02.
F F
fZ~ f~a 110..-_ Cli ~ f-~ --- ~/ Nt~ O
O
fl~S ~O CI13 . \ OCRs and W is ~ ~ O _ 011 tiN-10 ' OF~z C 1-y O
X
. fi, is I \ ~~ or Cty;
f~40 ~ OCty OC11~
JWVN/V' fig is NS O or H;

R~ is or ti;
CH3 0....

13+1315 _ ~a _ Fed is ~Oa ~ or II;
oct~~ oti cE-i~o ~ ,co-t~s or R~ is If or Clyp ~~OCH~
'C[llz R5 is CH3, C2H5, or (CH3)2CH; R8 is OH and R9 is H, or R8 and R9 together is a carbonyl group; X is an iodine or bromine atom, R5 is a hydrogen or the group RCO, wherein R is hydrogen, CH3 or a mono-substituted C6-C11 aryl group.
As long as the product from Scheme I contains at least one functional group which can be converted to, or is directly reactive with a targeting unit (Tu), targeted forms of the antitumor antibiotics of the above-named patents and applications can be generated, as shown in Scheme II below:
Tu-(Y)n Q-Sp-SS-W '-~ ~u-(Z-Sp-SS-w)m (Y)n_m Scheme II
wherein Q, Sp and W are as hereinbefore defined, Tu is a mono-or polyclonal antibody, its fragments, its chemically or genetically manipulated counterparts, growth factors, or steroids; Y is a side-chain amino, carboxyl, or thiol group of a protein, an aldehyde derived from carbohydrate residues, or an amidoalkylthio group; n is an integer of from 1 to 100; Z

is formed from covalent reaction of the groups Q and Y di-rectly or after subsequent reduction and Z is -CONH-, -CONHN=CH-, -CONHNHCH2-, -NHCONHN=CH-, -NHCONHNHCH2-, -NHCSNHN=CH-, -NHCSNHNHCH2-, -ON=CH-, -NH-, -NHCH2-, -N=CH-, -C02-, -NHCH2C02-, -SS-, O 02C_ O I
-S S N -S ~ CH3 -NHCOCH2-CH
N-~ . or (CH2)0 i . CH2 ~ .

and m is 0.1 to 15.
As an example, and with reference to Scheme II, above, the 3-mercaptopropionic acid derivative of E-33288711 (Q=C02H, Sp=-CH2CH2-), when converted to its activated hydroxysuccinimide form (Q=C02Su, Sp=-CH2-CH2-) can be used to react with some of the e-amino groups of lysine residues (e. g., Tu=monoclonal antibody, Y=-NH2 wherein n=50-100 from available lysine residues), at a pH between 7.0 and 9.5 in aqueous buffered solutions at temperatures between 4°C to 40°C to produce targeted forms of the antibiotics attached at random sites along the protein backbone (Tu=monoclonal anti-body, Z=-NHCO-, Sp=-CH2CH2-, m=1-10). Only a fraction of the available lysine residues are substituted in this manner, since high loading is generally not considered compatible with preserving the antibody immunoreactivity. The same randomly-substituted immunoconjugates can also be prepared from the 3-mercaptopropionic acid derivative using other car-boxyl group activating agents such as a variety of carbodi-imides, or the corresponding acyl azide. Alternatively, a 3-mercaptopropionyl hydrazide derivative of E-3328871I

(Q=H2NNHC0-, Sp=-CH2CH2-), when reacted with a periodate-oxidized antibody (Tu=monoclonal antibody, Y=-CHO, n=1-15) as described in U.S. Patent No. 4,671,958 at a pH between 4 and
7, in a buffered aqueous solution at a temperature of between 4°C and 40°C, reacts only at the aldehyde functionality (derived from cleavage of vic-diols of carbohydrate residues situated on the Fc portion of the antibodies) to generate monoclonal antibody conjugates containing the drug substi-tuted at specific sites along the backbone of the protein (Tu=monoclonal antibody, Z=-CH=NNHCO-, Sp=-CH2CH2-, m=0.5-10). In order to block unreacted aldehyde groups on the . antibody and thus avoid crosslinking, as well as stabilize the hydrolytically labile Schiff's base linkages, it is pref-erable (though not essential) to react the latter conjugate first with a compound such as acetyl hydrazide or tyrosine hydrazide, then reduce with sodium cyanoborohydride or sodium borohydride to produce the stabilized constructs of this invention (Tu=monoclonal antibody, Z=-CH2NHNHCO-, Sp=-CH2CH2-, m=0.5-10). Other aldehyde-reactive groups as part of the drug construct are within our invention to generate the prod-ucts of~Scheme II. Such functional groups are preferably, though not limited to, those which react with aldehydes under acidic aqueous conditions. The reactivity of protein lysines under basic conditions is sufficiently great such that their amines compete with the products of Scheme II for available aldehydes of the monoclonal antibody. Alternative aldehyde-reactive groups are, for example, the semicarbazide, the thiosemicarbazide, and the O-substituted hydroxylamine func-tionalities.
Assembly of targeted forms of the compounds listed in Table 1 is not restricted to the sequence outlined in Scheme II. The targeting unit (Tu) can be first modified to contain a thiol group, which is then reacted with the com-pounds of Table 1, in accordance with Scheme III below:

-g_ Tu(Y)n + Q-Sp-S-P ~ Tu-(Z-Sp-SH)m ( Y ) n-m Tu-(Z-Sp-SS-W)m ( Y ) n-m Scheme III
wherein Tu, Y, Q, Sp, W, n, and m are as hereinbefore defined, and P is hydrogen or 2-(pyridylthio), with the proviso that when Y is a thiol derived from a backbone amino acid residue of Tu, Z-Sp taken together is a covalent. bond.
As an example, and with references to Scheme III, above, a monoclonal antibody can be reacted with 3-(2-dithio-pyridyl)propionic acid hydroxysuccinimide ester to modify the protein through lysine residues (Tu=monoclonal antibody, Y=NH2, n=50-100, Q=-C02Su, Sp=-CH2-CH2-, P=2-pyridylthio).
Following reduction with, for example, dithiothreitol, an in-termediate is generated (Tu=monoclonal antibody, Z=-NHCO-, Sp=-CH2CH2-, m=1 to 15) which can be reacted with the com-pounds of Table 1 to generate the subject immunoconjugates.
Similarly, 2-iminothiolane can be reacted with a monoclonal antibody to introduce thiol groups onto the surface of the protein directly, without requiring a reduction step (Tu=monoclonal antibody, Z=-NHCO-, Sp=-(CH2)3-, m=1 to 15), and this intermediate can be reacted with the compounds of Table 1 as before. Alternatively, sulfhydryl groups inherent within the structure of monoclonal antibodies in dimeric form as cystine residues can be used to participate in the reac-tion of Scheme III directly. Such sulfhydryls are tradition-ally exposed by a combination of enzymatic digestion and re-duction of native monoclonal antibodies (Tu=Fab' fragment, Z-Sp=bond, Y=SH), but the use of genetically-altered constructs of monoclonal antibodies containing unpaired cystine residues is likewise contemplated.
A preferred embodiment of this invention is a protein-drug conjugate of the formula:
Tu- ( Z-Sp-SS-W) m ( Y ) n-m prepared from the antitumor antibiotic designated LL-E3328871I (CH3SSS-W) having a) ultraviolet spectrum as shown in Figure I;
b) a proton magnetic resonance spectrum as shown in Figure II;
c) an infrared spectrum as shown in Figure III;
and displacing the dithiomethyl moiety with a compound of formula Q-Sp-SH, wherein Sp is straight or branched-chain divalent (C2-C5) radicals or divalent (C2-C5) arylalkyl or heteroarylalkyl radicals, and Q is carboxyl, lower alkyldi-carboxyl anhydride, -C02Su, -CONHNH2, or _C02 O N02 to produce an intermediate of general formula Q-Sp-SS-W, wherein Q, Sp, and W are as hereinbefore defined, reacting Q-Sp-SS-W with a molecule of the formula Tu-(Y)n wherein Tu is a monoclonal antibody which exhibits preferential reactivity with a human tumor-associated anti-gen, Y is a side-chain amino group on the antibody, or an aldehyde generated by oxidation of the carbohydrate groups of the antibody, and n is an integer of from 1 to 100, to produce a compound of the formula:
Tu-(Z-Sp-SS-W)m (Y) n-m wherein Tu, Y, Sp, W, and n are as hereinbefore defined, and Z is formed from covalent reaction of the groups Q and Y
directly or after subsequent reduction, and Z is -CONH-, CONHN=CH-, -CONHNHCH2-, or (CII2)0 1 ,. CO 2 I-I
and m is 1 to 15.
A number of different monoclonal antibodies (MoAb's) are used to exemplify targeting of the methyltrithio anticancer compounds. MoAb's Lym 1 and Lym 2 recognize dif-ferent antigens on mature B-lymphocytes and their product lymphomas. The production and characteristics of these MoAb's are described by A.L. Epstein, et. al., "Cancer Research" ~, 830 (1987). MoAb 872.3 targets primarily to carcinomas of the breast and colon, through reactivity with z0 pancreatic, ovarian, and lung carcinomas has also been noted.
The ant~.body has been described by T.L. Klug, et. al., "Int.
J. Cancer" 38, 661 (1986). MoAb CT-M-O1, which recognizes primarily breast tmnors is described in Canadian patent application 513,110 filed July 4th, 1986 and MAiC-68 is produced by a sub-clone of the hybridoma which produces CT-M-O1, and recog-nizes both breast and colon carcinomas. Intermediates of tha subject compounds useful for, and conjugates with these anti-bodies, are described in the experimental section. It should not, however, be construed that this patent is limited to or restricted by the aforementioned antibodies. Instead, the methodology is sufficiently general that it can be applied to all antibodies regardless of their class or isotype, their enzymatically-derived fragments, their chemically manipulated and stabilized fragments, as well as their respective chim-eric and humanized counterparts. Nor are the targeting units restricted only to monoclonal antibodies. Other proteins, as well as small molecules for which receptors exist on target tissues, are within the purview of our discovery as targeting entities.
The methods of this invention used to produce mono-clonal antibody conjugates from the compounds of Table 1 yield constructs which retain good immunoreactivity with tar-get cell lines, as determined by the following in vitro assays:
Tarcret Cells All target cells were maintained in RPMI 1640 media supplemented with 5% Fetal Calf Serum (FCS), ITS (Collabora-tive Research, Cat# 40351), streptomycin (50 ~g/ml), peni-cillin (50 units/ml), gentamycin sulfate (50 ~g/ml) and glu-tamine (.03%). The cells were maintained in a humidified 5%
C02 incubator at 37°C.
I. Immunoreactivity Assays Procedure I - Elisa Appropriate target cells were harvested, counted and suspended in Dulbecco's Phosphate Buffered Saline (DPBS) at an optimal concentration for monoclonal antibody (MoAb) being tested. 0.1 ml of cells was aliquoted in each well of a sterile tissue culture polystyrene 96-well plate. The plates were centrifuged for 5 minutes at 1,000 RPM's and the supernatant was flicked off. Plates were air-dried overnight and may be stored at 4°C for up to 3 months.
Non-specific binding sites were blocked by adding 200 ~1 of 1% gelatin in DPBS per well and incubating the plate for 1 hour at 37°C in a humid incubator. (All subse-quent incubations are done under similar.canditions). The plates were washed once with 250 ~cl of 0.05% TWEEN-20*in DPBS
(washing solution) using the automated ELISA washing system from Dynatech (Ultrawash Ii). Samples to be tested were diluted to make a final concentration of 3 ~g/ml MoAb equiva-lents in 0.1% gelatin-DPBS. Six additional threefold serial dilutions were prepared from each 3 ~g/ml sample and 100 ~cl 3b was added to appropriate wells in triplicate. The bottom row of wells only received 100 ~1 of 0.1% gelatin as background.
Plates were incubated for 45 minutes and then washed three *Trade-mark times. Alkaline phosphatase conjugated affinity purified goat anti-mouse immunoglobulins (Cappel Cat# 8611-0231) was diluted 1:125 in 0.1% gelatin and 100 ul was added to each well. Plates were incubated for 45 minutes and then washed three times. 200 ul of p-nitrophenyl phosphate substrate solution (see below) was added to each well. After 45 min-utes at room temperature the reaction was stopped by the addition of 50 ul of 3M NaOH. The absorbance of the contents of each Well was read at 405 nm in the automated spectropho-tometer from Dynatech (EIA Autoreader # EL-310).
Substrate Diethanolamine Buffer (10%Z
- 97 ml diethanolamine 800 ml water 0.2 grams NaN3 100 mg MgCl2'6H20 The reagents were dissolved by continuous stirring and 1M HC1 was added until the pH was 9.8. The total volume was made up to 1 liter with water and filter sterilized with a 0.2 ~ filter. The buffer was stored in the dark at 4°C.
Immediately before use, p-nitrophenyl phosphate (Sigma, Cat# 104-40) was dissolved in the 10% diethanolamine buffer (must be at room temperature) to give a final concentration of 1 mg/ml.
Calculation of O. D. Values The percentage binding of each sample was calculated by the following equation:
A-B
x 100 - % Binding C-B
A = Average O.D. of test sample
8 = Average O.D. of background C = Average O.D. of 3 ~g/ml unmanipulated MoAb control The % binding was plotted on the non-log scale of a semi-log graph and the MoAb concentration was plotted on the log scale. The BD50 (i.e. dose of antibody needed to give 50% binding) of each test sample was derived from the graph and the amount of retention of immunoreactivity was calculated by the following equation:
BD50 of MoAb control x 100 = % Immunoreactivity retained BD50 of test sample Procedure 2 - Indirect RIA
Appropriate amounts of target cells in 0.2 ml of 10% FCS media were aliquoted into 4 ml polystyrene tubes.
Samples to be tested were diluted to a concentration of 2 ug/ml MoAb equivalents in l0% FCS media. Five three-fold serial dilutions were prepared from each 2 ~g/ml sample and 0.2 ml was added to each tube in duplicate. Background sam-ples received only cells and media. Cells were incubated at 4°C for 1 hour, then washed 2 times (all RIA washes were done with a 3 ml volume) with 2% FCS media. 0.05 ml of sheep F(ab')2 anti-mouse IgG [125I] (puPont, Cat# NEX 162-0142) containing approximately 500,000 CPM's was added to each tube;
cells were incubated an additional hour at 4°C, washed once with 2% FCS and twice with PBS. 0.5 ml of PBS was added to each tube, cells were vortexed, transferred to clean tubes and counted for 1 minute in a Packard Gamma 500.
The % binding of each value was determined and graphed like the preceding ELISA equation, except CPM's were substituted for O.D. units and C = Average CPM's of 1 ug/ml unmanipulated MoAb control. The % immunoreactivity retained of each sample was calculated as previously discussed.
Procedure 3 - Direct RIA
Appropriate amounts of target cells in 1 ml of 10%
FCS media were aliquoted into 4 ml polystyrene test tubes, centrifuged and supernatant was discarded. ~ Samples to be tested were diluted to make a concentration of 200 ug/ml MoAb equivalents in 10% FCS media. Five additional five-fold serial dilutions were re ared from each 200 p p ug/ml sample and 0.05 ml was added to each tube in duplicate. 0.05 ml of 1251-MoAb was added to each tube (optimal amount is indivi-dually determined for each MoAb and batch). Positive control samples contained. cells, media and 1251-MoAb. Background samples contained non-specific cells, media and 1251-MoAb.
Cells were incubated for 1 hour at 4°C, washed once with 2%
FCS media, twice with PBS, transferred and counted as previ-ously mentioned.
The % 1251-MoAb binding inhibition of each sample was calculated by the following formula:
A-B
x 100 _ % 1251-MoAb Binding inhibition C-B
A = Average CPM's of sample B = Average CPM's of background C = Average CPM's of positive control The plot and % immunoreactivity retained by each sample was calculated as previously'discussed except the BD50 is actually BID50 (Dose of MoAb needed to give 50% inhibition of the binding of 1251-MoAb).
Notes:
1) Tubes were always vigorously vortexed immediately after the addition of every reagent in the RIA's.
2) An internal control sample equalling 50% of the unmani pulated MoAb control was included in each set of assays to confirm whether each procedure was quantitative in predicting the conjugates' retention of immunoreactivity.
The results from these assays are tabulated below in Table 2.

-16- '~ 3 41 3 1 5 Table 2 Immunoreactivity of MoAb Con~ucrates Non-specific conjugates Immunoreactivitv using the product of ~ of unmodified MoAb example 3 with: Preparation control Lym 1 #1 15 B72.3 #1 70 #2 10 Hydrazide of 3-mercap-topropionic acid di-sulfide analog of E3328871I (example 4) conLgated to Lym 1 #1 100 #2 ° 87 #3 64 #4 80 #5 100 Table 2 i(continued~
Hydrazide of 3-mercapto-propionic acid disulfide Immunoreactivity analog of E3328871I % of unmodified example 4 conjugated to): Preparation MoAb control Lym 2 #1 57 #2 85 #3 39 #4 70 B72.3 #1 100 - #2 90 CT-M-O1 #1 60 MAC-68 #1 40 #2 ° 28 Hydrazide conjugates prepared using the product of example 5 with:
Lym 1 #1 100 #2 100 The monoclonal antibody conjugates of this inven-tion are active as anticancer agents. The assay described below for assessing in vitro cytotoxicity shows the dramatic 30.
preference of the constructs for target cell lines as opposed to non-target cells, and provides a measure of utility of targeted forms of the compounds compared to their non-target-ed counterparts.
Cytotoxicity Assays In Vitro Samples to be tested were diluted to a concentra-tion of 0.2 or 0.02 ug/ml of LL-E3328871I equivalents (starting concentration is dependent on cell line to be tested). Three additional five-fold dilutions were prepared from each original sample dilution and 0.2 ml was added to sterile 15 ml polystyrene tubes. At least one similar conju-gate consisting of LL-E3328871I and an irrelevant MoAb was included in each assay to determine specificity of the rele-vant conjugate. 105 appropriate target cells in 0.2 ml of 10% FCS media were aliquoted into the tubes and vortexed. In addition,~ an identical test was performed utilizing irrelevant cells as targets to further confirm specificity of relevant conjugate. MoAb controls received only equivalent amounts of - MoAb and positive control samples received only 10% FCS media.
Cells were incubated at 37°C for 7 minutes then washed 4 times with 8 ml of 2% FCS media. 0.1 ml of 10% FCS
was added to each tube, cells were vortexed and 0.2 ml was aliquoted to each well of a sterile 96-well polystyrene tis-sue culture plate. ' Plates were incubated for 2 days in a humidified 37°C incubator with 5% C02. One half of the media was re moved and replaced with fresh media containing 2 ~Ci/ml 3H
thymidine (DuPont, NEN, Cat# NET-027). Incubation was con-tinued for 24 hours, cells were frozen, thawed and harvested by a PHD cell harvester (Cambridge Technology, Inc.). Each sample was counted for 1 minute in a Beckman LS 5800 scintil-lation counter on Channel 1.
The % growth inhibition was calculated as follows:
Average CPM of test value x 100 - % Growth Average CPM of media control 100 - % Growth - % Inhibition The % inhibition was plotted on the non-log scale of a semi-log graph and the LL-E3328871I concentration was plotted on the log scale. The IC50 (concentration of LL-E3328871I needed to give 50% inhibition) of each test sample was derived from the graph and the amount of retention of cytotoxicity was calculated by the following equation:
IC50 of LL-E3328871I
x 100 - % Cytotoxicity Retained IC50 of test sample The results from the in vitro cytotoxicity assay are tabulated below in Table 3.
- Table 3 In Vitro Cytotoxicity of MoAb ConZu ads MoAb Preparation Cytotoxicity - % product of %E3328871I Example 1 Non-specific con-jugates prepared using product of Example 3 with:
Lym 1 #1 .9 11.3 872.3 #1 .001 #2 1.4 3.8 % product of %E3328871I Example Hydrazide conju-gates prepared using product of Example 4 with:
Lym 1 #1 80 #2 56 191 #3 40 60 Table 3 ~(continuedl MoAb Preparation Cytotoxicity % product of %E3328871I Example 4 Hydrazide conju-gates prepared using product of Example 4 with:
Lym 1 (#3 Against 0 0 non-tar-geted cells) Lym 2 #1 29 #2 2 100 #3 2 55 B72.3 #1 ~ 0 0 #2 0 0 MAC-68 #1 90 CTM-O1 #1 111 830 %E3328871I
Hydrazide conju-gates prepared using product of Example 5 with:
Lym 1 #1 300 #2 100 The following assay system was used to measure the in vivo activity of the conjugates of this invention.
In vivo tests for antitumor activity on drug-monoclonal antibody conjugates were done using human tumor xenographs in athymic (nude) mice.
Burkitt lymphoma (Raji) and myeloma (HS Sultan) cells were harvested from culture flasks and inoculated subcu-taneously (> 80 x 106 Raji cells or 40 x 106 HS Sultan cells) into test mice. Solid tumors, ovarian carcinomas (CA73, Ovcar-3) and breast carcinoma (MX-1) were propagated in athymic mice, removed, cut into 2 mm3 fragments and implanted - subcutaneously into test mice (5-8 fragments per mouse).
Drugs, monoclonal antibodies and drug-monoclonal antibody conjugates were administered intraperitoneally once each 3 days for 3 or 5 total injections starting on day 2, 3, 4, 6, 7 or 8 days after tumor implantation. Tumor measure-ments (the length and width of the tumor) were made by means of a Fowler ultra CAL II electronic caliper each 7 days for 4 or 5 weeks post tumor implantation. Tumor mass in mg was estimated from the formula:
Length(mm) x Width(mm) Tumor growth inhibition was calculated for each test group on a percent of control [mean mg of treated (T) divided by mean mg of control (C) x 100]. A T/C value < 42%
in groups with > 65% surviving animals is considered neces-sary to demonstrate activity.
The results from this assay appear in Table 4.

Table 4 In Vivo Antitumor Testinc,~ Results Dosag e(mcq) Tumor Size S
T

MoAb Drucr (TIC) %control Hydrazide of 3-mercaptopro- 14.5 0.26 12 5/6 pionic acid disulfide analog of E3328871I conjugated to Lym 2 Hydrazide alone - 0.26 34 4/6 - MoAb Lym 2 alone 14.5 - 32 6/6 Mixture, hydrazide +

MoAb Lym 2 14.5 0.26 20 5/6 ip treatment against human melanoma'cell line H.S. Sultan, 3 injections starting on day 5 after tumor implantation, measurements given made on day 35 post-implantation -, , -23- 1 3 41 3 1 Table 4 [continued) Dosage(mc~ Tumor Size S T

MoAb Drucr (T~) %control Hydrazide of 3-mercapto- 15.5 0.25 39 7/7 propionic acid disulfide analog of E3328871I

conjugated to MAC-68 Hydrazide-alone - 0.25 - 0/6 MoAb MAC-68 alone 31 - 78 6/6 Mixture, hydrazide + 15.5 0.25 - 0/6 MoAb MAC-68 Melphalan (as positive - 10 43 6/6 control) ip treatment against human ovarian cancer line CA73, three injections started 3 days after tumor implantation, measurements given made on day 35 post-implantation '. , -24- 1 3 4 1 3 1 5 Table 4 (continued) Dosacre (mcqZ Tumor Size S T
MoAb Drucx ~(TJC) %control Hydrazide of 3-rnercapto- 8.75 0.25 14 4/6 propionic acid disulfide analog of E3328871I con-jugated to CT-M-O1 Hydrazide alone - 0.25 - 0/6 MoAb CT-M-01 alone 8.75 - 75 5/6 Mixture, hydrazide + 8.75 0.2.5 - 0/6 MoAb CT-M-Ol Vincristine (positive - 1.0 0 4/4 control) ip treatment against human breast cancer cell line MX-1,' three injections started on day 2 following tumor implanta-tinn_ maact~rPmPnts given made on day 35 host-implantation Table 4 (continued) Dosaqe(mca) Tumor Size S
T

MoAb Drua (T/C)%control Hydrazide of 3-mercapto- 6.2 0.125 62 6/6 propionic acid disulfide analog of E3328871I con-jugated to B72.3 Hydrazide alone - 0.125 85 6/6 MoAb B72.3 alone 6.2 - 96 6/6 Mixture, hydrazide + 6.2 0.125 105 5/6 MoAb B72.3 E3328871I - 0.005 141 5/6 (3 treatments) Cis platinum (positive - 3.0 6 6/6 control, 3 treatments) ip treatments against human ovarian cell line OVCAR-3, five injections starting on day 4 after tumor implantation (unless otherwise noted), measurements ven made on gi day Table 4 (continued) Dosaae(mcqZ
Tumor Size S T

MoAb Drua ~T /C)%control Hydrazide of 3-mercapto- 27 0.26 6 3/6 propionic acid disulfide analog of E3328871I con-jugated t o Lym 1 Hydrazid~ alone - 0.26 72 6/6 MoAb Lym 1 alone 27 - 72 6/6 Mixture, hydrazide + 13 0.13 61 4/6 MoAb Lym 1 ip treatm ent against human cell line Raji Burkitt lymphoma TC, three injections started on day'7 after tumor implanta-tion, mea surements given madeon day 28 post -implantation The invention will be further described in conjunc-tion with the following non-limiting examples.
ExamQle 1 3-Mercaptopropionic Acid Disulfide Analoq of To a solution of 90 mg of LL-E3328871I in 90 ml of acetonitrile was added 10.6 mg of 3-mercaptopropionic acid in 1 ml of acetonitrile. The solution was vortexed and then stored at -20°C for 6 days. The solvent was removed in vacuo and the residue chromatographed over 10 ml of silica gel in methylene chloride. The column was developed with 50 ml of methylene chloride, 50 ml of 4% methanol in methylene chloride and finally 100 ml of 8% methanol in methylene chloride.
Evaporation of this last fraction gave a residue which was taken up in ethyl acetate with the aid of a little acetone and added dropwise to an excess of hexane. The precipitate was collected and dried, giving 39 mg of the desired product (FABMS, M+H 1394). Retention time on C18 reverse phase HPLC:
18 minutes with 50% acetonitrile/0.1 M aqueous ammonium chlo-ride. (,~L-E3328871I. 8.0 minutes, ester hydrolysis product:
1.5 minutes) Example 2 Reaction of LL-E3328871I with the p-nitrophenyl ester of 3-mercaptopropionic acid (A) Preparation of p-nitrophenyl ester of 3-mercaptopro-pionic acid Commercial 3-mercaptopropionic.acid in methylene chloride containing a catalytic amount of concentrated sul-furic acid was treated with isobutylene for 20 minutes. The solution was then extracted with 1N sodium bicarbonate solu-tion after which the methylene chloride solution was dried using anhydrous magnesium sulfate. The solution was then evaporated to a colorless mobile liquid which NMR and mass spectral data indicated was the S-t-butylmercaptopropionic acid, t-butyl ester.

An aliquot of this ester was refluxed with 6N hy-drochloric acid in dioxane for 2.5 hours. The solvent was evaporated, ethyl acetate was added and this solution was extracted with sodium carbonate. The sodium carbonate ex-tract was treated with 6N hydrochloric acid until the pH of the suspension was 2Ø The suspension was then extracted with ethyl acetate, the extract dried over anhydrous magne-sium sulfate and the solvent evaporated to a colorless liquid which 1H-NMR -and mass spectral data indicated was S-t-butyl-mercaptopropionic acid.
This compound was converted to the p-nitrophenyl ester by treatment with equimolar amounts of g-nitrophenol and dicyclohexylcarbodiimide in tetrahydrofuran for 4 hours.
The dicyclohexyl urea by-product was removed by filtration and the filtrate was evaporated to an oil which was purified by passage over neutral silica gel using the solvent system hexane:methylene chloride (50:50). 'The pure p-nitrophenyl ester derivative was a faintly yellow, mobile oil.
The free mercaptan was unmasked by the following procedure. The S-t-butylmercaptopropionic acid p-nitrophenyl ester was dissolved in trifluoroacetic acid and a slight molar excess (10%) of mercuric acetate was added. The mixture was stirred for 30 minutes, then the trifluoroacetic acid was evaporated and the residue taken up in dimethylformamide.
This solution was treated with hydrogen sulfide gas for 15 minutes, then the black mercuric sulfide was filtered off and the filtrate evaporated under reduced pressure to eliminate up to 99% of the dimethylformamide. The. resultant slightly brownish mobile liquid was purified over neutral silica gel using hexane:methylene chloride (50:50). The major component was shown by 1H -NMR to contain a small amount of the t-butyl mercapto derivative. Analytical HPLC over two Perkin-Elmer Pecosphere~ClB columns in tandem [4.6 x 33 mm and 4.6 x 83 mm] using a gradient system of 37.5/62.5 to 47.5/52.5 of acetonitrile and O.1M ammonium acetate buffer at pH 6.5 (acetic acid) over a 12 minute span indicated that the prod-uct was 88% of the p-nitrophenyl ester of 3-mercaptopropionic *Trade-mark acid and 10% of the less polar S-t-butylmercaptopropionic acid p-nitrophenyl ester. There was also a small amount of free p-nitrophenol present.
(B) Reaction of p-nitrophenyl ester of 3-mercaptopro-pionic acid with LL-E3328871I
A 100 mg portion of LL-E3328871I was dissolved in 50 ml of acetonitrile. To this was added a solution of 25.7 mg _of p-nitrophenyl ester of 3-mercaptopropionic acid in 1 ml of acetonitrile. The reaction was left at -20oC for 48 hours. HPLC indicated the reaction was complete. The solu-tion was evaporated to dryness and the residue taken up in 4-5 ml of ethyl acetate using sonication to effect solution.
The mixture was filtered and the filtrate dripped into 45 ml of stirred hexane. The resultant faintly yellow solid was collected and dried under reduced pressure, giving 93 mg of the p,-nitrophenyl ester of propionip acid derivative of LL-E3328871I as established by 1H NMR. By FABMS the [M+H] ion appeared at M/Z=1515.
Example 3 N_~YdroxYsucc ~,nimidyl 3-mercaptopropionate disulfide analog of LL-E3328871I
To a solution of 5 mg of the 3-mercaptopropionic acid disulfide analog of LL-E3328871I from Example 1 in 0.5 ml of tetrahydrofuran was added 0.45 mg of N-hydroxysuc-cinimide in 0.1 ml of tetrahydrofuran and then 1.8 mg of dicyclohexylcarbodiimide in 0.2 ml of tetrahydrofuran. The reaction was allowed to stir at room temperature for 4 hours and was then quenched with a large excess of hexanes. The solid was isolated by filtration and dissolved in ethyl ace-tate. The resulting solution was washed three times with brine, dried with magnesium sulfate, and evaporated to 5 mg of the desired product as a tan powder which was used without further purification. Retention time on reverse phase C18 HPLC: 15 minutes with 40% acetonitrile/0.1 M aqueous ammonium chloride (starting material: 6.0 minutes).

x341315 Example 4 3-Mercaptopropionyl hydrazide disulfide analog of LL-E3328871I
To 5.4 ml (3 eq) of anhydrous hydrazine in 100 ml of refluxing tetrahydrofuran under argon was added dropwise
9.2 ml (83 mmol) of methyl 3-mercaptopropionate in 50 ml tetrahydrofuran over 2 hours. The solution was refluxed an additional two hours, evaporated, and then diluted and evapo-rated twice from 300 ml of toluene. The product was applied lg to a plug of silica gel with 5% ethyl acetate/chloroform and eluted from the plug with 20% methanol/chloroform. The re-' sultant 3-mercaptopropionyl hydrazide was a faintly pink oil which solidified when cooled but melted at room temperature.
To 50 mg of LL-E3328871I in 50 ml of acetonitrile at -15°C was added 6.6 mg of 3-mercaptopropionyl hydrazide in 1 ml tetrahydrofuran. One equivalent of triethylamine and/or one equivalent of acetic acid was added as catalyst. The _ reaction was allowed to stir at 0°C for one hour and the sol-vent was then evaporated. The residue was chromatographed on silica gel with a 10-15% methanol in chloroform gradient to yield 26 mg of the desired product. Retention time on reverse phase C18 HPLC: 5.0 minutes in 41% acetonitrile/0.1 M
aqueous ammonium chloride.
Example 5 N- ~j~(4-Methyl-coumarin-7-yl)amino]acetyl)cysteine hydrazide disulfide analog of LL-E3328871I
A mixture of 1.0 g (5.7 mmol) of 4-methyl-7-amino-coumarin, 3.0 ml of ethyl bromoacetate (5 eq), 90 mg (0.1 eq) of sodium iodide, and 30 ml dimethylformamide was heated under argon at 80°C for 5 hours. The mixture was cooled, diluted with ethyl ether, washed three times with 50% brine, dried with magnesium sulfate, and evaporated to dryness. The crude product was dissolved in chloroform containing 1% ethyl ace-tate and filtered through a plug of silica gel. Recrystal-lization from diethyl ether containing a trace of chloroform yielded pure ethyl N-[(4-methyl-coumarin-7-yl)amino]acetate.

. ' -31- 1 3 41 3 1 5 To 1.96 g (7.5 mmol) of the above ester in 15 ml of methanol and 15 ml of tetrahydrofuran was added 10 ml of 1N
aqueous sodium hydroxide. After 30 minutes, 4 ml of 10%
aqueous hydrochloric acid was added. The organic solvents were evaporated and the resultant crystalline product was filtered and washed with cold ethanol and then ether. This material was dissolved in 20 ml of tetrahydrofuran and 4 ml of dimethylformamide. Dicyclohexylcarbonyldiimidazole (1.3 g, 2.2 eq) was added and the reaction allowed to stir for 15 minutes. Cysteine ethyl ester hydrochloride (1.6 g, 2.5 eq) and triethylamine (1.2 ml) were then added. After a further three hours, the reaction was diluted with ethyl ether con-taining 5% methylene chloride and washed once with 10% aqueous hydrochloric acid and twice with brine. After drying with magnesium sulfate and evaporating the solvents, the crude product was crystallized by dissolving in chloroform con-taining a minimal amount of ethanol and then adding an excess of ether. The crystals were filtered and dried to give pure N-[[(4-methyl-coumarin-7-yl)amino]acetyl]cysteine ethyl ester.
A mixture of 5 ml of chloroform, 20 ml of methanol, and 0.4~m1 of hydrazine hydrate were heated to reflux under argon. To this was added 550 mg of N-[[(4-methyl-coumarin-7-yl)amino]acetyl]cysteine ethyl ester. After refluxing for 9 hours the mixture was cooled and the solid product was fil-tered and washed with chloroform and then ethyl ether. The crude product (which contained thiol and disulfide) was dis-solved in dimethylformamide containing dithiothreitol and triethyl amine. After 30 minutes the product was precipitated with excess ethyl ether and collected by filtration. This material was purified further by recrystallization from de-gassed acetonitrile containing dithiothreitol and a trace of triethyl amine to give pure N-[[(4-methyl-coumarin-7-yl)-amino]acetyl]cysteine hydrazide.
To 12 mg of 70% pure LL-E3328871I in 12 ml aceto-nitrile at 0°C was added 4 mg of N-[[(4-methyl-coumarin-7-yl)amino]acetyl]cysteine hydrazide in 1.2 ml dimethylform-amide. After stirring overnight another 2 mg of N-[[(4-methyl-coumarin-7-yl)amino]acetyl]cysteine hydrazide in 0.6 ml dimethylformamide was added. The reaction was stirred for 3 days at 0°C and filtered. The acetonitrile was evapo-rated and the resultant dimethylformamide solution was diluted with an excess of 1:1 hexanes/ether. The product was isolated by filtration and further purified by chromatography on silica gel with a 15-20% gradient of methanol in chloroform to yield 3 mg of the desired product. Retention time on reverse phase C18 HPLC: 3.5 minutes using 45% acetonitrile/0.1 M aqueous ammonium chloride.
Example 6 3-Mercaptopropionyl hYdrazide disulfide analocr of LL-E33288a3I
To 10 mg of LL-E33288a3I in 9 ml of acetonitrile at -15°C was added 6.6 mg of 3-mercaptopropionyl hydrazide in 1 ml acetonitrile. One equivalent of triethylamine and/or one equivalent of acetic acid were added as a catalyst. The.
reaction was allowed to stir at 0°C for one hour and the sol-vent was then evaporated. The residue was chromatographed on silica gel with a 10-15% methanol in chloroform gradient to give the desired product. Retention time on reverse phase C18 HPLC: 3.5 minutes in the system 45% acetonitrile/0.1 M
aqueous ammonium chloride.
Example 7 Non-specific con~ju_qation to proteins The hydroxysuccinimide ester described in Example 3 was covalently attached to antibodies under slightly alkaline conditions. The following is a general procedure used to make the antibody conjugates listed in Table 5. Antibody at a concentration of 3-5 mg/ml in phosphate buffer containing O.1M sodium chloride, pH 7.5 was reacted with a 5-20-fold molar excess of the product from Example 3 with stirring, at room temperature for from 1-4 hours. The conjugated protein was desalted chromatographically and aggregated protein was separated from monomeric material by gel filtration HPLC.
Monomeric fractions were pooled and concentrated.

'. -33-Table 5 Non-specific coniuqates prepared usinc~the product of Example 3 MoAb Drug Loading Lym 1 5.2 B72.3 6.0 B72.3 2.9 Example 8 Site-specific conjugate preparation The general method for attaching hydrazide deriva-tives of drugs to oxidized antibodies is described in T. J.
McKearn, et al., in U.S. Patent No. 4,671,958. The procedure has been applied to preparing antibody conjugates from the products of Examples 4 and 5 with specific modifications as described below. The products from'these reactions and their characteristics are summarized in Table 6.
(A) Antibody Oxidation Antibody at a concentration of 5 to
10 mg/ml was dialyzed overnight against a 200 fold volume of 50mM sodium acetate buffer, pH 5.5 containing O.1M sodium chloride (Buffer A). After dialysis, the MoAb was oxidized with lSmM to 200mM periodic acid in 0.2M sodium acetate. The oxidation was allowed to proceed in the dark, with stirring, at 4°C for 45 minutes after which time the oxidized MoAb was desalted on a >5 bed volume Sephadex G-25 column. The degree of oxidation of the antibody was assessed by reaction with p-nitrophenylhydrazine and comparing absorbance of the pro-tein at 280mm vs. p-nitrophenylhydrazine at 395mm.
(B) Druq Hydrazide Conjugation The oxidized MoAb was react-ed with 25 to 200-fold molar excess of drug hydrazide. The hydrazides were dissolved into dimethylformamide and added to the aqueous solution of MoAb. To avoid precipitation of MoAb, the final volume of dimethylformamide added did not exceed lOx of the total reaction volume. Reaction was allowed to proceed for 3 hours at room temperature, with stirring. To prevent crosslinking of unreacted aldehydes and subsequent ' -34-aggregation, a blocking agent, acetyl hydrazide was added in 100-fold molar excess three hours after addition of the drug hydrazide. To stabilize the Schiff's base linkage between aldehyde and drug hydrazide (a hydrazone), the product gen-erally was reduced to an alkyl hydrazine by the addition of lOmM sodium cyanoborohydride, allowing the reaction to pro-ceed for one more hour (total conjugation time - 4 hours).
The conjugate was chromatographically desalted and exhaus-tively dialyzed (minimum time 48 hours) into pH 6.5 phosphate buffer for storage and testing.
Conjugates were analyzed for the presence of aggre-- gates by gel filtration HPLC and for free drug by reverse phase HPLC. Drug loading was determined spectroscopically using the extinction coefficients of both the antibody and the drug to estimate molar concentrations of drug in conju-gates.

Table 6 Hydrazide conZuqates prepared from the product of Example 4 MoAb Preparation Druq Loadin~/M

Lym 1 #1 1.4 #2 2.4 #3 1.0 #4 6.7 #5 3.3 Lym 2 #1 2.9 #2 1.9 #3 2.0 #4 2.8 B72.3 #1 ' 2.3 #2 ' 1.3 CTM-O1 3.1 MAC-68 1.7 Hvdrazide Conjugates prepared from the product of Example Lym 1 #1 0.15 #2 0.76

Claims (13)

THE EMBODIMENTS OF THE INVENTION IN WHICH AN EXCLUSIVE
PROPERTY OR PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. A carrier-drug conjugate of the formula prepared from a compound of formula CH3SSS-W wherein CH3SSS-W
is an antitumor antibiotic designated as LL-E33288.alpha.1Br, .alpha.1I, .alpha.2Br, .alpha.2I, .alpha.3Br, .alpha.3I, .alpha.4Br, .beta.1Br, .beta.1I, .beta.2Br, .beta.2I, .gamma.1Br, .gamma.1I, 61I, BBM-1675, FR-900405, FR-900406, PD 114759, PD 115028, CL-1577A, CL-1577B, CL-1577D, CL-1577E, or CL-1724 -36a- R5 is CH3, C2H5, or (CH3)2CH; R8 is OH and R9 is H, or R8 and R9 together is a carbonyl group; X is an iodine or bromine atom, R5 is a hydrogen or the group RCO, wherein R is hydrogen, CH3 or a mono-substituted C6-C11 aryl group comprising:
reacting CH3SSS-W with a compound of general formula Q-Sp-SH, wherein Sp is a straight or branched-chain divalent (C1-C18) radical, divalent aryl or heteroaryl radical, divalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent aryl- or heteroaryl-alkyl (C1-C18) radicals, divalent cycloalkyl- or heterocycloalkyl-alkyl (C1-C18) radical or divalent (C2-C18) unsaturated alkyl radical, and Q is, or can be subsequently converted to, halogen, amino, alkylamino, carboxyl, carboxaldehyde, hydroxy, thiol, .alpha.-haloacetyloxy, lower alkyldicarboxyl, -CONHNH2, -NHCONHNH2, -NHCSNHNH2, -ONH2, -CON3, to produce an intermediate of formula Q-Sp-SS-W, wherein Q, Sp, and W are as hereinbefore defined, reacting Q-Sp-SS-W with a molecule of the formula Tu-(Y)n wherein Tu is defined as a mono- or polyclonal anti-body, its fragments, its chemically or genetically manipulat-ed counterparts, growth factors, or steroids, Y is a side-chain amino, carboxy, or thiol group of a protein, an alde-hyde derived from carbohydrate residues, or an amidoalkylthio group; and n is an integer of from 1 to 100, to produce a compound of the formula:
wherein Tu, Y, Sp, W, and n are as hereinbefore defined, and Z is formed from covalent reaction of the groups Q and Y
directly or after subsequent reduction, and Z is -CONH-, -CONHN=CH-, -CONHNHCH2-, -NHCONHN=CH-, -NHCONHNHCH2-, -NHCSNHN=CH-, -NHCSNHNHCH2-, -ON=CH-, -NH-, -NHCH2-, -N=CH-, -CO2-, -NHCH2CO2-, -SS-, and m is 0.1 to 15.
2. A protein-drug conjugate of the formula prepared from the antitumor antibiotic designated LL-E33288.UPSILON.1I
(CH3SSS-W) wherein W is as defined in claim 1 having (a) ultraviolet spectrum as shown in Figure 1;
(b) a proton magnetic resonance spectrum as shown in Figure 2; and (c) an infrared spectrum as shown in Figure 3;
comprising:
displacing the diothiomethyl moiety with a compound of formula Q-Sp-SH, wherein Sp is straight or branched-chain divalent (C2-C5) radicals or divalent aryl- or heteroaryl-alkyl (C2-C5) radicals, and Q is, or can be subsequently converted to, carboxyl, lower alkyldicarboxylanhydride, -CONHNH2, or to produce an intermediate of general formula Q-Sp-SS-W, wherein Q, Sp, and W are as hereinbefore defined, reacting Q-Sp-SS-W with a molecule of the formula Tu-(Y)n wherein Tu is a monoclonal antibody which exhibits preferential reactivity with a human tumor-associated antigen, Y is a side-chain amino group on the antibody, or an aldehyde generated by oxidation of the carbohydrate groups of the antibody, and n is an integer of from 1 to 100, to produce a compound of the formula:
wherein Tu, Y, Sp, W, and n are as hereinbefore defined, and Z
is formed from covalent reaction of the groups Q and Y directly or after subsequent reduction, and Z is -CONH-, -CONHN=CH-, -CONHNHCH2-, OR~~~
and m is 0.1 to 15.
3. A carrier-drug conjugate according to claim 1 wherein CH3SSSW is LL-E33288~1I, Q is the 4-nitrophenyl ester of a carboxyl group, Sp is -CH2CH2-, Tu is a monoclonal antibody which is CT-M-01, Y is -NH2, Z is -CONH-, and m is 0.5 to 15.
4. A carrier-drug conjugate according to claim 1 wherein CH3SSSW is LL-E33288~1I, Q is the hydroxysuccinimide ester of a carboxyl group, Sp is -CH2CH2-, Tu is a monoclonal antibody which is MAC-68, Y is -NH2, Z is -CONH-, and m is 0.5 to 15.
5. A carrier-drug conjugate according to claim 1 wherein CH3SSSW is LL-E33288~1I, Q is -CONHNH2, Sp is -CH2CH2-, Tu is a monoclonal antibody which is Lym 1, Y is -CHO, Z is -CONHNHCH2-, and m is 0.1 to 10.
6. A carrier-drug conjugate according to claim 1 wherein CH3SSSW is LL-E33288.UPSILON.1I, Sp is Tu is a monoclonal antibody which is B72.3, Y is -CHO, Z is -CONHNHCH2-, and m is 0.1 to 10.
7. A carrier-drug conjugate according to claim 1 wherein CH3SSSW is LL-E33288.UPSILON.1I, Sp is Tu is a monoclonal antibody which is Lym 2, Y is -CHO, Z is -CONHNHCH2-, and m is 0.1 to 10.
8. A process for preparing a targeted derivative of a compound of formula CH3SSS-W wherein CH3SSS-W is an antitumor antibiotic LL-E33288.alpha.1Br, .alpha.1I, .alpha.2Br, .alpha.2I, .alpha.3Br, .alpha.3I, .alpha.4Br, .beta.1Br, .beta.1I, .beta.2Br, .beta.2I,.gamma.1Br, .gamma.1I, .sigma.1I, BBM-1675, FR-900405, FR-900406, PD 114759, PD 115028, CL-1577A, CL-1577B, CL-1577D, CL-1577E, or CL-1724 and W is as defined in claim 1 comprising:
reacting CH3SSS-W with a compound of formula Q-Sp-SH, wherein Sp is a straight or branched-chain divalent (C1-C18) radical, divalent aryl or heteroaryl radical, divalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent aryl- or heteroaryl-alkyl (C1-C18) radicals, divalent cycloalkyl- or heterocycloalkyl-alkyl (C1-C18) radical or divalent (C2-C18) unsaturated alkyl radical, and Q is halogen, amino, alkylamino, carboxyl, carboxaldehyde, hydroxy, lower alkyldicarboxyl anhydride, -CONHNH2, -NHCONHNH2, -NHCSNHNH2, -ONH2, or in acetonitrile in the presence of one equivalent of triethylamine or one equivalent of acetic acid at -10° to -30°C for 1-48 hours, isolating the intermediate of formula Q-Sp-SS-W, wherein Q, Sp, and W are as hereinbefore defined, then reacting the compound of formula Q-Sp-SS-W, wherein Sp and W are as hereinbefore defined and Q is halogen, amino, alkylamino, carboxyl, carboxaldehyde, hydroxy or lower alkyldicarboxylic anhydride with a molecule of the formula Tu-(Y)n wherein Tu is a mono- or polyclonal antibody, its fragments, its chemically or genetically manipulated counterparts, growth factors, or steroids; Y is a side-chain amino or carboxyl functionality; n is 1-100, in aqueous buffer at a pH of between 6.5 and 9, at 4° to 40°C either directly or in the presence of a water-soluble carbodiimide, to generate the compound wherein Tu, Sp, W, n, and Y are as hereinbefore defined, m is 1-15 and Z is formed from covalent react ion of the groups Q
and Y and is -CONH-, -NH-, -N=CH-, or -CO2-or reacting the compound of formula Q-Sp-SS-W, wherein Sp and W are as hereinbefore defined and Q is a carboxylic acid, with N-hydroxysuccinimide, 2, 3, 5, F, -tetrafluorophenol, pentafluorophenol, or 4-nitrophenol in the presence of a carbodiimide activating agent to generate a compound of formula Q-Sp-SS-W wherein Sp and W are as hereinbefore defined and Q is and reacting the resulting compound with a molecule of formula Tu(Y)n, where Tu and n are as hereinbefore defined, and Y is a side-chain amino group, in an aqueous buffered solution at a pH between 6.5 and 9, at a temperature of between 4° and 40°C, inclusive, to generate compounds of the formula:

wherein Tu, Sp, Y, and n are as hereinbefore defined, m is 1-15, and Z is formed from covalent reaction between Q and Y
and is defined as -CONH-, or reacting a compound of formula Q-Sp-SS-W, wherein Sp and W are as hereinbefore defined and Q is -CONHNH2 with nitrous acid in aqueous actonitrile to generate a compound of formula Q-Sp-SS-W, wherein Sp and W are as hereinabove defined and Q is -CON3, then reacting Q-Sp-SS-W with a compound of formula Tu-(Y)n, wherein, Tu, Y and N are as hereinabove defined to produce a compound of the formula wherein Tu, Z, Sp, W, m, Y, and n are as hereinabove defined;
or reacting a compound of formula Q-Sp-SS-W wherein Sp and W are as hereinbefore defined and Q is hydroxy, with an alpha-haloacetic anhydride to produce a compound wherein Q is .alpha.-haloacetyloxy, and reacting the a-haloacetyloxy-Sp-SS-W or a compound of formula Q-Sp-SS-W, wherein Sp and W are as here-inbefore defined and Q is with a molecule of the formula Tu-(Y)n wherein Tu is as here-inbefore defined, Y is a side-chain thiol of a protein, or an amidoalkylthio group introduced on an amine of Tu using 3-(2-dithiopyridyl)propionic acid hydroxy-succinimide ester followed by reduction with dithiothreitol, or an amidoalkylthio group introduced on an amine of Tu using 2-iminothiolane, and n is 1-10, under aque-ous buffered conditions at a pH between 4.5 and 7, at a tem-perature between 4° and 40°C, inclusive, to produce a com-pound of formula:

wherein Tu, Sp, W, and n are as hereinbefore defined, and Z
is formed from covalent reaction of the groups Q and Y and Z
is -SS-, and m is 0.1 to 10;
or reacting a compound of the formula Q-Sp-SS-W where-in Sp and W are as hereinbefore defined and Q is -NH2, -CONHNH2, -NHCONHNH2, -NHCSNHNH2, or -ONH2 with a molecule of formula Tu-(Y)n wherein Tu is as hereinbefore defined, Y is an aldehyde generated from carbohydrate residues on Tu by oxidation in the presence of an alkaline earth periodate, in an aqueous buffer at a pH between 4.0 and 6.5, at 4° to 40°C, inclusive, and n is 1 to 20 to generate a compound of formu-la:

wherein Tu, Sp, W, Y, and n are as hereinbefore defined and Z
is formed from the covalent reaction of Q and Y and is -ON=CH-, -N=CH-, -CONHN=CH-, -NHCONHN=CH-, or -NHCSNHN=CH-, and m is 0.1 to 15; or treating the compound immediately here-inabove of formula:

wherein Tu, Z, Sp, W, Y, n, and m are as immediately herein-above defined with acetylhydrazine or tyrosine hydrazine in an aqueous buffer at a pH between 4.0 and 6.5, at 4° to 40°C, inclusive, to generate a compound of formula:

wherein Tu, Z, Sp, W, n, and m are as immediately hereinabove defined and Y is -CH=NNHCOCH3 or and reacting this compound with sodium cyanoborohydride or sodium borohydride, in an aqueous buffer at a pH of 4.0 to 6.5, at a temperature of 4o to 40°C, inclusive, to generate a compound of formula:

wherein Tu, Sp, W, m, and n are as hereinabove defined, Z is -NH-CH2-, -CONHNHCH2-, -NHCONHNHCH2-, or -NHCSNHNHCH2-, and Y
is -CH2NHNHCOCH3 or
9. Use of an oncolytic amount of a product prepared from a compound of general formula CH3SSS-W wherein CH3SSS-W is an antitumor antibiotic designated as LL-E33288.alpha.1Br, .alpha.1I, .alpha.2Br, .alpha.2I, .alpha.3Br, .alpha.3I, .alpha.4Br, .beta.1Br, .beta.1I, .beta.2Br, .beta.2I, .gamma.1Br, .gamma.1I, .delta.1I, BBM-1675, FR-900405, FR-900406, PD 114759, PD 115028, CL-1577A, CL-1577B, CL-1577D, CL-1577E, or CL-1724, wherein W is as defined in claim 1, comprising:
reacting CH3SSS-W with a compound of general formula Q-Sp-SH, wherein Sp is a straight or branched-chain divalent (C1-C18) radical, divalent aryl or heteroaryl radical, divalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent aryl-or heteroaryl-alkyl (C1-C18) radical, divalent cycloalkyl- or heterocycloalkyl-alkyl (C1-C18) radical or divalent (C2-C18) unsaturated alkyl radical, and Q is, or can be subsequently converted to, halogen, amino, alkylamino, carboxyl, carboxaldehyde, hydroxy, thiol, .alpha.-haloacetyloxy, lower alkyldicarboxyl, -CONHNH2, -NHCONHNH2, -NHCSNHNH2, -ONH2, -CON3, to produce an intermediate of formula Q-Sp-SS-W, wherein Q, Sp, and W are as hereinbefore defined, reacting Q-Sp-SS-W with a molecule of the formula Tu-(Y)n wherein Tu is defined as a mono- or polyclonal antibody, its fragments, its chemically or genetically manipu-lated counterparts, growth factors, or steroids; Y is a side-chain amino, carboxy, or thiol group of a protein, an alde-hyde derived from carbohydrate residues, ar en amidoalkylthio group; and n is an integer of from 1 to 100, to produce a compound of the formula:

wherein Tu, Y, Sp, W, and n are as hereinbefore defined, and Z is formed from covalent reaction of the groups Q and Y
directly or after subsequent reduction, and Z is -CONH-, -CONHN=CH-, -CONHNHCH2-, -NHCONHN=CH-, -NHCONHNHCH2-, -NHCSNNN=CH-, -NHCSNHNHCH2-, -ON=CH-, -NH-, -NHCH2-, -N=CH-, -CO2-, -NHCH2CO2-, -SS-, and m is 0.1 to 15 to inhibit growth of a tumor in a mammal.
10. Use of an oncolytic effective amount of a protein-drug conjugate of the formula prepared from the antitumor antibiotic designated LL-E33288.UPSILON.1I
(CH3SSS-W) wherein W is as defined in claim 1 having (a) ultraviolet spectrum as shown in Figure 1;
(b) a proton magnetic resonance spectrum as shown in Figure 2; and (c) an infrared spectrum as shown in Figure 3;
comprising displacing the dithiomethyl moiety with a compound of formula Q-Sp-SH, wherein Sp is straight or branched-chain divalent (C2-C5) radicals or divalent aryl- or heteroarylalkyl (C2-C5) radicals, and Q is, or can be subsequently converted to, carboxyl, lower alkyldicarboxylanhydride, -CONHNH2, or to produce an intermediate of general formula Q-Sp-SS-W, wherein Q, Sp, and W are as hereinbefore defined, reacting Q-Sp-SS-W with a molecule of the formula Tu-(.UPSILON.)n wherein Tu is a monoclonal antibody which exhibits preferential reactivity with a human tumor-associated antigen, .UPSILON. is a side-chain amino group on the antibody, or an aldehyde generated by oxidation of the carbohydrate groups of the antibody, and n is an integer of from 1 to 100, to produce a compound of the formula:

wherein Tu, Y, Sp, W, and n are as hereinbefore defined, and Z is formed from covalent reaction of the groups Q and Y
directly or after subsequent reduction, and Z is -CONH-, -CONHN=CH-, -CONHNHCH2-, or and m is 0.1.to 15 to deliver a compound to an antigenic site in a mammal.
11. A carrier-drug conjugate of the formula wherein SS-W has the meaning ascribed to it in a compound of formula CH3SSS-W wherein CH3SSS-W is an antitumor antibiotic designated as LL-E33288.alpha.1Br, .alpha.1I, .alpha.2Br, .alpha.2I, .alpha.3Br, .alpha.3I, .alpha.4Br, .beta.1Br, .beta.1I, .beta.2Br, .beta.2I, .gamma.1Br, .gamma.1I, .delta.1I, BBM-1675, FR-900405, FR-900406, PD 114759, PD 115028, CL-1577A, CL-1577B, CL-1577D, CL-1577E, or CL-1724, Sp is a straight or branched-chain divalent (C1-C18) radical, divalent aryl or heteroaryl radical, divalent (C3-C18) cycloalkyl or heterocycloalkyl radical, divalent aryl- or heteroaryl-alkyl (C1-C18) radicals, divalent cycloalkyl- or heterocycloalkyl-alkyl (C1-C18) radical or divalent (C2-C18) unsaturated alkyl radical and W
is as defined in claim 1, Tu is defined as a mono- or polyclonal antibody its fragments, its chemically or genetically manipulated counterparts, growth factors, or steroids; .gamma. is a side-chain amino, carboxy, or thiol group of a protein, an aldehyde derived from carbohydrate residues, or an amidoalkylthio group; and n is an integer of from 1 to 100, Z is -CONH-, -CONHN=CH-, -CONHNHCH2-, -NHCONHN=CH-, -NHCONHNHCH2-, -NHCSNHN=CH-, -NHCSNHNHCH2-, -ON=CH-, -NH-, -NHCH2-, -N=CH-, -CO2-, -NHCH2CO2-, -SS-, and m is 0.1 to 15.
12. Use of a pharmaceutically effective amount of a carrier-drug conjugate according to any one of claims 3 to 7 to prepare a conjugate to inhibit growth of a tumor in a mammal.
13. A commercial package comprising a pharmaceutically effective amount of a conjugate prepared from a carrier-drug conjugate according to any one of claims 3 to 7 together with instructions for use thereof to inhibit growth of a tumor in a mammal.
CA000581577A 1987-10-30 1988-10-28 Targeted forms of methyltrithio antitumor agents Expired - Fee Related CA1341315C (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US11494087A 1987-10-30 1987-10-30
US07/114,940 1987-10-30
US24624888A 1988-09-21 1988-09-21
US246,248 1988-09-21

Publications (1)

Publication Number Publication Date
CA1341315C true CA1341315C (en) 2001-11-06

Family

ID=26812695

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000581577A Expired - Fee Related CA1341315C (en) 1987-10-30 1988-10-28 Targeted forms of methyltrithio antitumor agents

Country Status (1)

Country Link
CA (1) CA1341315C (en)

Similar Documents

Publication Publication Date Title
US5053394A (en) Targeted forms of methyltrithio antitumor agents
US5770701A (en) Process for preparing targeted forms of methyltrithio antitumor agents
EP0313873B1 (en) Targeted forms of methyltrithio antitumor agents
JP3234980B2 (en) Preparation of conjugates containing novel bifunctional compounds
US5824805A (en) Branched hydrazone linkers
US4980457A (en) Cytotoxic conjugates which can be used in therapy and process for their preparation
PL172824B1 (en) Novel thioether conjugates
US5006652A (en) Intermediates for antibody-vinca drug conjugates
CZ298024B6 (en) Processes for preparing monomeric conjugates of calicheamicin carrier and derivative
EP0392384B1 (en) Targeted forms of methyltrithio antitumor agents
US5241078A (en) Coupling agents and sterically hindered disulfide linked conjugates prepared therefrom
CA1341315C (en) Targeted forms of methyltrithio antitumor agents
US4670563A (en) Imidazolides as intermediates for the synthesis of cytotoxic conjugates
CA1314245C (en) Process for the preparation of conjugates in which a monovalent carboxylic ionophore is associated by means of a covalent bond with a macromolecule, which are useful as immunotoxin potentiators
CA2014459C (en) Targeted forms of methyltrithio antitumor agents
KR0159767B1 (en) Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group, targeted forms and preparation thereof

Legal Events

Date Code Title Description
MKLA Lapsed
MKLA Lapsed

Effective date: 20121106