AU2005201115A1 - Quinones for treatment of diseases - Google Patents

Quinones for treatment of diseases Download PDF

Info

Publication number
AU2005201115A1
AU2005201115A1 AU2005201115A AU2005201115A AU2005201115A1 AU 2005201115 A1 AU2005201115 A1 AU 2005201115A1 AU 2005201115 A AU2005201115 A AU 2005201115A AU 2005201115 A AU2005201115 A AU 2005201115A AU 2005201115 A1 AU2005201115 A1 AU 2005201115A1
Authority
AU
Australia
Prior art keywords
alkyl
group
formula
compound
quinone
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2005201115A
Inventor
Andrei V. Blokhin
Benjamin Frydman
Laurence J. Marton
Karen M. Neder
Jerry Shunneng Sun
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.)
CellGate Inc
Original Assignee
SLIL Biomedical Corp
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
Priority claimed from AU46780/00A external-priority patent/AU4678000A/en
Application filed by SLIL Biomedical Corp filed Critical SLIL Biomedical Corp
Priority to AU2005201115A priority Critical patent/AU2005201115A1/en
Publication of AU2005201115A1 publication Critical patent/AU2005201115A1/en
Assigned to CELLGATE, INC. reassignment CELLGATE, INC. Request for Assignment Assignors: SLIL BIOMEDICAL CORPORATION
Abandoned legal-status Critical Current

Links

Description

AUSTRALIA
Patents Act 1990 SLIL BIOMEDICAL CORPORATION COMPLETE SPECIFICATION STANDARD PATENT Invention Title: Quinones for treatment of diseases.
The following statement is a full description of this invention including the best method of performing it known to us:-
I
NOVEL QUINONES AS DISEASE
THERAPIES
CROSS-REFERENCE TO RELATED
APPLICATIONS
This application claims priority benefit of co-pending provisional patent application U.S. Serial No. 60/131,842, filed on April 30, 1999. The content of that application is hereby incorporated by reference herein in its entirety.
STATEMENT OF RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED
RESEARCH
Not applicable.
TECHNICAL
FIELD
This invention relates to novel quinones. The invention also relates to conjugates of quinones with various peptides. The invention also relates to various medicinal and industrial uses of these compounds, including the use of these compounds in treating diseases such as cancer.
BACKGROUND OF THE INVENTION The quinones are a large and varied group of natural products found in all major groups of organisms. Quinones are a group of aromatic dioxo compounds derived from benzene or multiple-ring hydrocarbons such as naphthalene, anthracene, etc. They are classified as benzoquinones, naphthoquinones, anthraquinones, etc., on the basis of the.ring system. The C=O groups are generally ortho or para, and form a conjugated system with at least two C=C double bonds; hence the compounds are colored, yellow, orange or red.
Quinones with long isoprenoid side chains, such as plastoquinone, ubiquinone and phytoquinone are involved in the basic life processes of photosynthesis and respiration.
Quinones are biosynthesized from acetate/malonate via shikimic acid. A few quinones are used as laxatives and worming agents, and others are used a pigments in cosmetics, histology and aquarrell paints. Quinones have a variety of medicinal and industrial uses.
Many efficient antineoplastic drugs are either quinones (anthracycline derivatives, mitoxantrone, actinomycin), quinonoid derivatives (quinolones, genistein, bactracyclin), or drugs such as etoposide that can easily be converted to quinones by in vivo oxidation.
Gantchev et al. (1997) Biochem. Biophys. Res. Comm. 237:24-27. The literature on quinone-DNA interactions is replete with references to quinones having the potential to undergo redox cycling with the formation of highly reactive oxygen species that are thought to relate to their cytotoxicity. O'Brien (1991) Chem. Biol. Interactions 80:1-41. It has also been shown that many quinones are efficient modifiers of the enzymatic activity of topoisomerase II, an enzyme essential for cell division.
Quinones are now widely used as anti-cancer, anti-bacterial and anti-malarial drugs, as well as fungicides. The antitumor activities of the quinones were revealed more than two decades ago when the National Cancer Institute published a report in which fifteenhundred synthetic and natural quinones were screened for their anticancer activities.
Driscoll et al. (1974) Cancer Chemot. Reports 4:1-362. Anti-cancer quinones include p- Lapachone, a plant product, which inhibits DNA topoisomerase II and induces cell death with characteristics of apoptosis in human prostate and promyelocytic leukemia cancer cell lines. Human breast and ovary carcinoma showed sensitivity of the cytotoxic effect of Plapachone without signs of apoptosis. Li et al. (1995) Cancer Res. 55:3712-5; and Planchon et al. (1995) Cancer Res. 55:3706-11. 1,2-Naphthoquinone (3,4-b)dihydrofuran inhibits neoplastic cell growth and proliferation of several cancers, such as prostate, breast, colon, brain and lung, including multi-drug resistant types. WO 97/31936. Furanonaphthoquinone derivatives and other naphthoquinones and naphth-[2,3-d]-imidazole-4,9dione compounds are also useful in treating malignant tumors such as those affecting the blood, breast, central nervous system, cervix, colon, kidney, lung, prostate and skin.
WO 97/30022 and JP Patent No. 9235280. Anthraquinone derivatives with telomerase inhibitory activity are also useful in treating leukemia, lung cancer, myeloma, lymphoma, prostate, colon, head and neck, melanoma, hepatocellular carcinoma, bladder, ovarian, breast and gastric cancers. WO 98/25884 and WO 98/25885. Ansamycin benzoquinones are useful in the treatment of primitive neuroectodermal tumors, prostate cancer, melanoma and metastatic Ewing's sarcoma. WO 94/08578.
Quinones also have a number of other medicinal uses. Terpenoid-type quinones are also useful as treatments for diabetes. U.S. Patent No. 5,674,900. Additional quinones can be used to treat cirrhosis and other liver disorders. U.S. Patent Nos. 5,210,239 and 5,385,942. Hydroquinone amines and quinone amines are also useful for treating a number of conditions, including spinal trauma and head injury. U.S. Patent No. 5,120,843.
Degenerative central nervous system diseases, as well as vascular diseases, are treatable with quinones such as Idebenone 2 3 -dimethoxy-5-methyl-6-(10-hydroxydecyl)-1,4benzoquinone] and Rifamycin S. Mordente et al. (1998) Chem. Res. Toxicol. 11:54-63; Rao et al. (1997) Free Radic. Biol. Med. 22:439-46; Cortelli et al. (1997) J. Neurol. Sci. 148:25- 31; and Mahadik et al. (1996) Prostaglandins Leukot. Essent. Fatty Acids 55:45-54.
A
vitamin K analog, 6-cyclo-octylamino-5,8-quinoline quinone shows efficacy for treatment of leprosy and tuberculosis. U.S. Patent No. 4,963,565. Hydroquinone is used to treat skin pigmentation disorders. Clarys et al. (1998) J. Dermatol. 25:412-4. Mitomycin C-related drug indoloquinone E09 has demonstrated cell killing against HL-60 human leukemia cells, H661 human lung cancer cells, rat Walker tumor cells and human HT29 colon carcinoma: cells. Begleiter et al. (1997) Oncol. Res. 9:371- 82 and Bailey et al. (1997) Br. J.
Cancer 76:1596-603: Quinones such as aloin, a C-glycoside derivative of anthraquinone, accelerate ethanol oxidation and may be useful in treating acute alcohol intoxication.
Chung et al. (1996) Biochem. Pharmacol. 52:1461-8 and Nanji et al. (1996) Toxicol. Appl.
Pharmacol. 140:101-7. Quinones capsaicin and resiniferatoxin blocked activation of nuclear transcription factor NF-KB, which is required for viral replication, immune regulation and induction of various inflammatory and growth-regulatory genes. Singh et al.
(1996) J. Immunol. 157:4412-20. Antiretroviral and antiprotozoan naphthoquinones are described in U.S. Patent Nos. 5,780,514 and 5,783,598. Anthraquinones are also useful as laxatives. Ashraf et al. (1994) Aliment. Pharmacol. Ther. 8:329-36; and Muller-Lissner (1993) Pharmacol. 47 (Suppl. 138-45.
A subset.of quinones designated lapachones has been shown to have activity against neoplastic cells, as described in U.S. Patent Nos. 5,969,163, 5,824,700, and 5,763,625.
Antiviral activity (in combination with xanthine) or reverse transcriptase inhibitory activity for p-lapachone is suggested in U.S. Pat. Nos. 5,641,773 and 4,898,870, while antifungal and trypanosidal activity of p-lapachone is suggested in U.S. Pat. Nos. 5,985,331 and 5,912,241.
Quinones can be administered alone or in conjunction with other agents, such as 1,2-dithiole-3-thione. Begleiter et al. (1997). Hydroxyquinone can be used in conjunction with glycol or glyceryl esters of retinoic acid to treat skin disorders. WO 9702030.
Combinational chemotherapy of carboquone, a benzoquinine derivative, and cis-Platinum, diminishes the side effects of the former. Saito (1988) Gan To Kagaku Ryoho 15:549-54.
Quinones also have various additional uses. A few quinones are used as laxatives and worming agents, and others are used a pigments in cosmetics, histology and aquarrell paints. Quinones include 2,5-cyclohexadiene-1,4-dione, which is useful as an oxidizing agent; in photography Patent No. 5,080,998); in manufacturing dyes and hydroquinone; in tanning hides; in strengthening animal fibers; and as a reagent.
In rapidly dividing cells such as tumor cells, cytotoxicity due to quinone administration has been attributed to DNA modification. However the molecular basis for the initiation of quinone cytotoxicity in resting or non-dividing cells has been attributed to the alkylation of essential protein thiol or amine groups and/or the oxidation of essential protein thiols by activated oxygen species and/or GSSG, glutathione disulfide. Oxidative stress arises when the quinone is reduced by reductases to a semiquinone radical which reduces oxygen to superoxide radicals and reforms the quinone. This futile redox cycling and oxygen activation forms cytotoxic levels of hydrogen peroxide and GSSG is retained.
by the cell and causes cytotoxic mixed protein disulfide formation. O'Brien (1991) Chem.
Biol. Interact. 80:1-41.
Conjugation of quinones and glutathione (GSH) are sometimes associated with the process of detoxification. Jeong et al. (1996) Mol. Pharmacol 50:592-8. For example, certain o-quinones contribute to the neurodegenerative processes underlying Parkinson's disease and schizophrenia. Glutathione transferase (GST) M2-2, which conjugates glutathione and o-quinones, prevents these processes. Baez et al. (1997) Biochem. J.
324:25-8. However, in many cases, conjugation with GSH actually leads to quinone bioactivation and: toxicity. For example, the nephrotoxicity of hydroquinone and bromobenzene is mediated via quinone-glutathione conjugates. Jeong et al. (1996) Mol.
Pharmacol. 50:592-8. The formation of GSH conjugates is also involved in the bioactivation of vicinal dihalopropane 1,2-dibromo-3-chloropropane. Hinson et al. (1995) Can. J. Physiol. Pharm. 73:1407-13. Additional examples of GSH conjugation potentiating the toxicity of quinones are described in Fowler et al. (1991) Hum. Exp.
Toxicol. 10:451-9; Mertens et al. (1991) Toxicol. Appl. Pharmacol. 110:45-60; Puckett- Vaughn et al. (1993) Life Sci. 52:1239-47; Dekant (1993) Toxicol. Lett. 67:151-160; Monks et al. (1994) Chem. Res. Toxicol. 7:495-502; Monks (1995) Drug Metab. Rev. 27:93-106; and Eyer (1994) Environ. Health Persp. 102 (Suppl. 6):123-32.
Because of the wide variety of biological processes in which quinones play a C critical role, it would be advantageous to develop novel quinones for various uses, including disease treatment.
All references cited herein are hereby incorporated by reference in their entirety.
SSUMMARY OF THE INVENTION The invention provides novel quinone compounds and methods for use of the 0 quinone compounds in treating diseases.
S 10 In one embodiment, the invention comprises compounds of the formula I0 A-M-B-M 2
-D
wherein A is selected from the group consisting of-O- and -CH 2 wherein Mi is selected from the group consisting of a single bond and CI-Cg alkyl, Ci-Cs branched alkyl,
C
3 -Cg cycloalkyl, and C 3 -Cg cycloaryl; wherein B is selected from the group consisting of
-CH
2 and wherein RI is selected from the group consisting of-H, C,-Cg alkyl, CI-C8 branched alkyl, C 3
-C
8 cycloalkyl, and C 3
-C
8 cycloaryl; wherein M 2 is selected from the group consisting of a single bond and CI-C 8 alkyl, Ci-Cs branched alkyl, C 3
-C
8 cycloalkyl, and C 3 -Cg cycloaryl; wherein D is selected from the group consisting of-H, -OH, -N(R 7 pentoses, hexoses, 2005201115 15 Mar 2005 and
NN
HH
wherein R 4 is selected from the group consisting of Ci-C 8 alkyl, C 1 branched alkyl, C 3
-C
8 cycloalkyl, C 3 CYcloaryl, -N(R 9
)(R
10 and -CN; and wherein R 7
R
8
R
9 and Rio are independently selected from the group consisting of C 1
-C
8 alkyl, C 1
-C
8 branched alkyl, C 3 -Cs cycloalkyl, C 3
-C
8 cycloaryl, and The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds..
In another embodiment, the invention comprises compounds of the formula S 0 O H
O
S----ONa x, K
K
wherein x is an integer between 1 and 2; and each K is independently selected from the group consisting of H, CI-Cs alkyl, CI-Cg alkenyl, CI-C 8 alkanol, CI-C 8 alkoxy,
O
00 0 0' Sand where zero or two, but no more than two, vicinal K's in the molecule represent Ssingle electrons which form a pi bond, thus forming a double bond together with the existing sigma bond between the two adjacent carbons bearing the two vicinal K's.
SThe invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an Sindividual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective 0 compound or compounds.
In another embodiment, the invention comprises compounds of the formula
O
OH 0 ONa
'-R
wherein R is selected from the group consisting of C -C 8 alkyl, CI-C 8 cycloalkyl,
C
3 -Cs cycloaryl,
C,-C
8 branched alkyl. and CI-Cg alkanol. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula wherein Y is selected from the group consisting of-H, -Br, -Cl, and and wherein Gt and G 2 are independently selected from the group consisting of H, CI-Cs alkyl, O and -C(=O)-CHX 3 where n is an integer from 0 to 3 and X is selected from the group consisting ofF, Cl, Br, and I. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective coihipound or compounds.
In another embodiment, the invention comprises compounds of the formula wherein M is selected from the group consisting of-0-, and The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula wherein.x is an integer between 1 and 2; each B is independently- selected from the group consisting of H, Ci-Cs alkyl, C3-C 8 cycloalkyl, C 3
-C
8 cycloaryl, Cl-C 8 alkyl-C 3
-C
8 cycloalkyl, and Ci-C 8 alkyl-C 3 -Cg cycloaryl; and each K is independently selected from the group consisting of H, OH, C -Cg alkyl, CI-C 8 alkenyl, C 1
C
8 alkanol, Ci-Cg alkoxy, and where zero or two, but no more than two, vicinal K's in the molecule represent single electrons which form a pi bond, thus forming a double bond together with the existing sigma bond between the two adjacent carbons bearing the two vicinal K's. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula 0
O
II
0
R
wherein each B is independently selected from the group consisting of H, CI-Cs alkyl, C 3
-C
8 cycloalkyl, C 3
-C
8 cycloaryl, Ci-Cs alkyl-C 3 -Cs cycloalkyl, and
CI-C
8 alkyl-C 3 -Cs cycloaryl; and wherein R is selected from the group consisting of CI-C 8 alkyl and Ci-C 8 alkanol. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease 12 cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula 0 0 i O O-Ms-CN-- H Me -y where Ms is alkyl, y is an integer from 1 to 6, and L is selected from the group consisting of or where K, and K, are independently selected from the group consisting of H, C,-C 8 alkyl, C,-C 8 alkyl-COOH, alkyl-COO-Ci-C, alkyl, alkyl-N(G,G,), and C,-C 8 alkyl-N(G,)-C-C, alkyl-N(GG,); and wherein each of G,,
G
2 and Gs is independently selected from the group consisting of H and C,-C, alkyl. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula
O-GIO--V-M--T
where z is an integer between one and ten; Glo is selected from the group consisting of alkyi; each M is independently selected from the group consisting of alkyl; each V is selected from the group consisting and and T is selected from the group consisting of -COOMs and -CONM 9 MIo, where each of Ms, M 9 and Mio are independently selected from the group consisting of H and alkyl. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula where M 1 2 is selected from the group consisting of C i-Cs alkyl.
In another embodiment, the invention comprises compounds of the formula 0
O
M
1 4 where MI 4 and Ms 1 are independently selected from the group consisting of C -C 8 alkyl.
The invention also comprises the above compounds in combination with a pharmaceuticalyacceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula
OJ
0 where J is selected from the group consisting of Ci-Cg alkyl, CI-C 8 cycloalkyl. C 3 -Cs cycloaryl, and CI-Cs branched alkyl The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention comprises compounds of the formula 0 0
/NH
2 CH3
CH
Rs where Rs is the side chain of a naturally-occuring amino acid. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier.
The invention also comprises use of the above compounds to treat an indication 16 characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
In another embodiment, the invention embraces compounds of the formula S-L-QUIN, where S represents a single amino acid or a peptide of at least two amino acids, L is a .linking group containing at least one carbon, oxygen, or nitrogen atom attached covalently to both S and QUIN, or a nonentity; and QUIN is a quinone, quinone derivative, hydroquinone, or hydroquinone derivative. In a preferred embodiment, S or a portion thereof, S-L or a portion thereof, or both S or a portion thereof and then L or a portion thereof, are cleaved from the quinone-containing remainder of the molecule by an enzyme, such as the enzyme prostate specific antigen. In another preferred embodiment, L is or -NH-(CI-Cs alkyl)-O-. In yet another preferred embodiment, L is
-NH-(C
6
H
4
)CH
2 -Cs alkyl)-0-. A preferred peptide for the S moiety is X-Ser-Lys-Leu-Gln, where X is a protecting group or an amino-terminal capping group, and the side chains of Ser, Lys, and Gln may optionally be protected with protecting groups. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
The invention also embraces compounds of the formula S-L-QUIN, wherein S represents a single amino acid or a peptide of at least two amino acids; L is a linking group containing at least one carbon, oxygen, or nitrogen atom attached covalently to both S and QUIN, or a nonentity; and QUIN is selected from the group consisting of the any of the above-mentioned quinone compounds which have a reactive group capable of being conjugated with an amino or carboxyl group, as well as the compounds 0 0 I O 0 0 0
O
OH
,and The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
The invention also encompasses a method for making the above-described compounds of formula S-L-QUIN, comprising the steps of a) covalently linking L to S, and b) covalently linking L to QUIN. Steps a) and b) can be performed in either order or simultaneously.
The invention also encompasses compounds of the formula
K
D§K
where x is an integer between 1 and 2; W is selected from -OH, -O-C -Cs alkyl, -O-C -C 8 alkyl-NH2, and -O-C -C 8 alkyl-NH-S, wherein S is a single amino acid or a peptide of two or more amino acids; and each K is independently selected from the group consisting of H, OH, C,-Cs alkyl, CI-Cs alkenyl, Ci-Cs alkanol, CI-Cs alkoxy, and where zero or two, but no more than two, vicinal K's in the molecule represent single electrons which form a pi bond, thus forming a double bond together with the existing sigma bond between the two adjacent carbons bearing the two vicinal K's. In a preferred embodiment.
W is -O-C -C alkyl-NH-S, S is a single amino acid or a peptide of two or more ammo acids; and the group -NH- forms an amide bond with the alpha-carboxy group of S when S is a single amino acid. Alternatively, the group -NH- forms an amide bond with the Cterminal alpha-carboxy group of S when S is a peptide of two or more amino acids. A preferred subset of the above compounds are the compounds of the formula W% l0 w- L^
I
where W is selected from -OH, -O-Ci-Cs alkyl, -O-Ci-Cs alkyl-NH 2 and
-O-CI-C
8 alkyl-NH-S, and wherein S is a single amino acid or a peptide of two or more amino acids. In a preferred embodiment, W is -0-C I-C 8 alkyl-NH-S, S is a single amino acid or a peptide of two or more amino acids, and the group -NH- forms an amide bond with the alpha-carboxy group of S when S is a single amino acid. Alternatively, the group -NH- forms an amide bond with the C-terminal alpha-carboxy group of S when S is a peptide of two or more amino acids. The invention also comprises the above compounds in combination with a pharmaceutically acceptable carrier. The invention also comprises use of the above compounds to treat an indication characterized by the proliferation of disease cells in an individual, comprising administering to the individual a therapeutic amount of one or more of the above compounds, optionally together with another therapeutically effective compound or compounds.
The invention also includes all salts, stereoisomers, and tautomers of the foregoing compounds, unless explicitly indicated otherwise.
In another embodiment, the invention comprises any one or more of the foregoing compounds, optionally in combination with another therapeutic compound, combined with a pharmaceutically acceptable excipient or carrier.
The invention also provides methods of treating an indication comprising the step of ,administering to the individual an effective amount of a composition comprising a novel quinone. In one embodiment, the invention comprises a method of treating an indication characterized by the proliferation of disease cells in an individual comprising administering to the individual a therapeutic amount of any of the foregoing compounds. In one method, the indication is cancer. In various embodiments, the cancer affects cells ofthe bladder, blood, brain, breast, colon, digestive tract, lung, ovaries, pancreas, prostate gland, or skin.
In other embodiments, the indication can also include, but is not limited to, Alzheimer's disease, epilepsy, multiple sclerosis, problems associated with tissue grafts and organ transplants, psoriasis, restenosis, stomach ulcers, or tissue overgrowth after-surgery. In other embodiments, the indication is an infection or infestation of parasites, bacteria, fungi or insects.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 depicts Scheme 1, illustrating the synthetic preparation of certain compounds of the invention.
Figure 2 depicts Scheme 2, illustrating the synthetic preparation of additional compounds of the invention.
Figure 3 depicts Scheme 3, illustrating the synthetic preparation of additional compounds of the invention.
Figure 4 depicts Scheme 4, illustrating the synthetic preparation of additional compounds of the invention.
Figure 5 depicts Scheme 5, illustrating the synthetic preparation of additional compounds of the invention.
Figure 6 depicts Scheme 6, illustrating the synthetic preparation of additional compounds of the invention.
Figure 7 depicts Scheme 7, illustrating the synthetic preparation of additional compounds of the invention.
Figure 8 depicts Scheme 8, illustrating the synthetic preparation of additional compounds of the invention.
Figure 9 depicts Scheme 9, illustrating the synthetic preparation of additional compounds of the invention.
Figure 10 depicts Scheme 10, illustrating the synthetic preparation of additional compounds of the invention.
Figure 11 depicts Scheme 11, illustrating the synthetic preparation of additional compounds of the invention.
Figure 12 depicts Scheme 12, illustrating the synthetic preparation of additional compounds of the invention.
Figure 13 depicts Scheme 13, illustrating synthetic preparation of peptides conjugated to certain quinone compounds.
Figure 14 depicts Scheme 14, illustrating additional synthetic preparation of peptides conjugated to certain quinone compounds.
Figure 15 depicts additional synthetic preparation of peptides conjugated to certain quinone compounds, including attachment of a linker group between the quinone and the peptide.
Figure 16 depicts the attachment of doxorubicin to a peptide, including attachment of a linker group between doxorubicin and the peptide.
Figure 17 depicts additional synthetic preparation of peptides conjugated to certain quinone compounds, including attachment of a linker group between the quinone and the peptide.
MODES FOR CARRYING OUT THE INVENTION The present invention encompasses novel quinones and methods of their use. Such methods include treating indications in an individual comprising the step of administering to the individual an effective amount of a novel quinone. The indications include cancer.
In various embodiments, the.cancer affects cells of the bladder, blood, brain, breast, colon, digestive tract, lung, ovaries, pancreas, prostate gland, or skin. In other embodiments, the indication can also include, but is not limited to, Alzheimer's disease, epilepsy, multiple sclerosis, problems associated with tissue grafts and organ transplants, psoriasis, restenosis, stomach ulcers, or tissue overgrowth after surgery. In other embodiments, the indication is an infection or infestation of parasites, bacteria, fungi or insects. The invention also includes industrial uses of these novel quinones, such as uses as pigments or dyes, as laxatives and worming agents, in cosmetics, histology and paint-making, in photography, in tanning hides, in strengthening animal fibers, and as a reagent.
Definitions By a "quinone" is meant any of a group of aromatic dioxo compounds derived from benzene or multiple-ring hydrocarbons such as naphthalene, anthracene, etc. They are classified as benzoquinones, naphthoquinones, anthraquinones,. etc., on the basis of the ring system. The C=O groups are generally ortho orpara, and form a conjugated system with at least two C=C double bonds; hence the compounds are colored, yellow, orange or red.
This type of chromophore is found in many natural and synthetic pigments. Exemplary quinones include 2,5-cyclohexadiene-1,4-dione, which is useful as an oxidizing agent, in photography, in manufacturing dyes and hydroquinone, in tanning hides, in strengthening animal fibers, and as a reagent; and various 1,2-naphthoquinones, which have medicinal uses. Frydman et al. (1997) Cancer Res. 57:620-627. By "hydroquinone" is meant the reduced form of any quinone; for example, the reduced form of 1,4-benzoquinone is 1,4- Sdihydroxybenzene (p-dihydroxybenzene).
An "indication" includes any symptom or the like which points out a suitable Sremedy or treatment or which shows the presence of a disease. As used herein, an "indication" also includes a "disease" itself, where a disease is a condition of an organ, Spart, structure or system of the body in which there is incorrect function resulting from the effect(s) of heredity,-infection, diet and/or environment. The indication can be characterized by proliferation of diseased cells, such as cancer. By "cancer" is meant the abnormal presence of cells which exhibit relatively autonomous growth, so that they 1 10 exhibit an aberrant growth phenotype characterized by a significant loss of cell proliferation control. Cancerous cells can be benign or malignant. In various embodiments, the cancer affects cells of the bladder, blood, brain, breast, colon, digestive tract, lung, ovaries, pancreas, prostate gland, or skin. In other embodiments, the indication can also include, but is not limited to, Alzheimer's disease, epilepsy, multiple sclerosis, problems associated with tissue grafts and organ transplants, psoriasis, restenosis, stomach ulcers, or tissue overgrowth after surgery. In other embodiments, the indication is an infection or infestation of parasites, bacteria, fungi or insects.
By "DNA toposiomerase 11" is meant is the scaffold protein capable of cleaving double-stranded DNA, passing an uncut portion of the DNA between the cut ends, and resealing the cut. DNA topoisomerase II ("topo II") is critical in DNA replication, because it can unknot tangles of DNA that would otherwise form as the long parental strands unwind and daughter strands are synthesized. During cleavage by topo II, the free phosphates on the DNA strands become covalently linked to tyrosine side chains of the enzyme. Staining of metaphase chromosomes with fluorescent antibodies raised against highly purified topo II demonstrates that this enzyme is associated with the chromosome scaffold. Even in interphase chromosomes, which are not as condensed as metaphase chromosomes, the DNA remains associated with topo 1I and hence with the chromosome scaffold. During interphase, proteins, including topo II, are bound to fixed sites in mammalian DNA that are 30-90 kb apart. The binding sites for topo II are called scaffoldassociated regions (SARs), which occur between but not within transcription units. DNA topoisomerase 11 is reviewed and discussed in, for example, Austin et al. (1998) Bioessays 20:215-26; Larsen et al. (1996) Prog. Cell Cycle Res. 2:229-39; Chaly et al. (1996) 23 Chromosome Res. 4:457-66; Kimura et al. (1994) J. Biol. Chem. 269:1173-6; and Roca et al. (1993) J. Biol. Chem. 268:14250-5.
An "individual" is a vertebrate, preferably a mammal, more preferably a human.
Mammals include, but are not limited to, farm animals, sport animals, rodents, primates, and pets.
An "effective amount" or "therapeutic amount" is an amount sufficient to effect beneficial or desired clinical results. An effective amount can be administered in one or more administrations. For purposes of this invention, an effective amount of a quinone is an amount that is sufficient to palliate, ameliorate, stabilize, reverse, slow or delay the progression of the disease state. A therapeutic amount of a quinone of the present invention is an amount sufficient to inhibit proliferation of diseased cells. A quinone is considered to be an.effective agent if it is effective against at least one disease or in at least one application, even if it is not effective against another disease or in another application.
As used herein, "treatment" is an approach for obtaining beneficial or desired clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilization not worsening) of state of disease, prevention of spread metastasis) of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, improvement in quality of enjoyment of life, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment.
"Palliating" a disease means that the extent and/or undesirable clinical manifestations of a disease state are lessened and/or time course of the progression is slowed or lengthened, as compared to not administering quinones of the present invention.
The invention includes all salts of the compounds described herein. Particularly preferred are pharmaceutically acceptable salts. Pharmaceutically acceptable salts are those salts which retain the biological activity of the free acids or bases and which are not biologically or otherwise undesirable. The desired salt may be prepared by methods known to those of skill in the art by treating an amine-containing quinone with an acid, or by treating an acid-containing quinone with a base. Examples of inorganic acids include, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid. Examples of organic acids include, but are not limited to, formic acid, 24 acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, sulfonic acids, and salicylic acid. Examples of bases include, but are not limited to, sodium hydroxide and potassium hydroxide (which yield sodium and potassium salts, respectively), triethylamine, and t-butylamine.
The invention also includes all stereoisomers of the compounds, including diastereomers and enantiomers, as well as mixtures of stereoisomers, including, but not limited to, racemic mixtures. Unless stereochemistry is explicitly indicated in a structure, the structure is intended to embrace all possible stereoisomers of the compound depicted.
The term "alkyl" refers to saturated aliphatic groups including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having up to 12 carbon atoms. Examples of alkyl groups include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and adamantyl. Cyclic groups can consist of one ring, including, but not limited to, groups such as cycloheptyl, or multiple fused rings, including, but not limited to, groups such as adamantyl or norbornyl. Alkyl groups may be unsubstituted, or may be substituted with one or more substituents including, but not limited to, groups such as halogen (fluoro, chloro, bromo, and iodo), alkoxy, acyloxy, amino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde,carboalkoxy and carboxamide. or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group. Examples of substituted alkyl groups include, but are not limited to, -CF3, -CF 2
-CF
3 and other perfluoro and perhalo.groups.
The term "alkenyl" refers to unsaturated aliphatic groups including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having up to 12 carbon atoms, which contain at least one doublebond Examples of alkenyl groups include, but are not limited to, -CH 2 -CH=CH-CH3 and -CH2-CHz-cyclohexenyl, there the ethyl group can be attached to the cyclohexenyl moiety at any available carbon valence. The term "alkynyl" refers to unsaturated aliphatic groups including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having up to 12 carbon atoms, which contain at least one triple bond "Hydrocarbon chain" or "hydrocarbyl" refers to any combination of straight-chain, branched-chain, or cyclic alkyl, alkenyl, or alkynyl groups, and any combination thereof.
"Substituted alkenyl," "substituted alkynyl," and "substituted hydrocarbon chain" or "substituted hydrocarbyl" refer to the respective group substituted with one or more substituents, including, but not limited to, groups such as halogen, alkoxy, acyloxy, amino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group.
"Aryl" or "Ar" refers to an aromatic carbodyclic group having a single ring (including, but not limited to, groups such as phenyl) or multiple condensed rings (including, but not limited to, groups such as naphthyl or anthryl), and includes both unsubstituted and substituted aryl groups. Substituted aryls can be substituted with one or more substituents, including, but not limited to, groups such as alkyl, alkenyl, alkynyl, hydrocarbon chains, halogen, alkoxy, acyloxy, amino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, -carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group.
"Heteroalkyl," "heteroalkenyl," and "heteroalkynyl" refer to alkyl, alkenyl, and alkynyl groups, respectively, that contain the number of carbon atoms specified (or if no number is specified, having up to 12 carbon atoms) which contain one or more heteroatoms as part of the main, branched, or cyclic chains in the group. Heteroatoms include, but are not limited to, N, S, 0, and P; N and O are preferred. Heteroalkyl, heteroalkenyl, and heteroalkynyl groups may be attached to the remainder of the molecule either at a heteroatom (if a valence is available) or at a carbon atom. Examples of heteroalkyl groups include, but are not limited, to, groups such as -0-CH 3
-CH
2
-O-CH
3
-CH
2
-CH
2
-O-CH
3
-S-CH
2
-CH
2
-CH
3
-CH
2
-CH(CH
3
)-S-CH
3
-CH
2
-CH
2
-NH-CH
2
-CH
2 -,1-ethyl-6propyTpiperidino, 2-ethylthiophenyl, and morpholino. Examples of heteroalkenyl groups include, but are not limited to, groups such as -CH=CH-NH-CH(CH 3
)-CH
2 "Heteroaryl" or "HetAr" refers to an aromatic carbocyclic group having a single ring (including, but not limited to, examples such as pyridyl, thiophene, or furyl) or multiple condensed rings (including, but not limited to, examples such as imidazolyl, indolizinyl or benzothienyl) and having at least one hetero atom, including, but not limited to, heteroatoms such as N, 26 0, P, or S, within the ring. Heteroalkyl, heteroalkenyl, heteroalkynyl and heteroaryl groups can be unsubstituted or substituted with one or more substituents, including, but not limited to, groups such as alkyl, alkenyl, alkynyl, benzyl, hydrocarbon chains, halogen, alkoxy, acyloxy, amino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group.
Examples of such substituted heteroalkyl groups include, but are not limited to, piperazine, substituted at a nitrogen or carbon by a phenyl or benzyl group, and attached to the remainder of the molecule by any available valence on a carbon or nitrogen, -NH-SO2 -phenyl,
-NH-(C
O)O-alkyl, -NH-(C=)O-alkyl-aryl, and -NH-(C=O)-alkyl.
The heteroatom(s) as well as the carbon atoms of the group can be substituted. The heteroatom(s) can also be in oxidized form Unless otherwise specified. heteroalkyl, heteroalkenyl, heieroalkynyl, and heteroaryl groups have between one and five heteroatoms and between one and twenty carbon atoms.
The term "alkylaryl" refers to an alkyl group having the number of carbon atoms designated, appended to one, two, or three aryl groups.
The term "alkoxy" as used herein refers to an alkyl, alkenyl, alkynyl, or hydrocarbon chain linked to an oxygen atom and having the number of carbon atoms specified, or if no number is specified, having up to 12 carbon atoms. Examples of alkoxy groups include, but are not limited to, groups such as methoxy, ethoxy, and t-butoxy.
The terms "halo" and "halogen" as used herein refer to Cl, Br, F or I substituents.
"Protecting group' refers to a chemical group that exhibits the following characteristics: 1) reacts selectively with the desired functionality in good yield to give a protected substrate that is stable to the projected reactions for which protection is desired; 2) is selectively removable from the protected substrate to yield the desired functionality; and 3) is removable in good yield by reagents compatible with the other functional group(s) Sp~-ent or generated in such projected reactions. Examples of suitable protecting groups can be found in Greene et al. (1991) Protective Groups in Organic Synthesis, 2nd Ed. (John Wiley Sons, Inc., New York). Preferred amino protecting groups include, but are not limited to, benzyloxycarbonyl (CBz), t-butyloxycarbonyl (Boc), t-butyldimethylsilyl (TBDIMS), 9-fluorenylmethyloxycarbonyl (Fmoc), or suitable photolabile protecting groups such as 6-nitroveratryloxy carbonyl (Nvoc), nitropiperonyl, 27 pyrenylmethoxycarbonyl, nitrobenzyl, dimethyl dimethoxybenzil, 5-bromo-7nitroindolinyl, and the like. Preferred hydroxyl protecting groups include Fmoc, benzyl, tbutyl, TBDIMS, photolabile protecting groups (such as nitroveratryl oxymethyl ether (Nvom)), Mom (methoxy methyl ether), and Mem (methoxy ethoxy methyl ether).
Particularly preferred protecting groups include NPEOC (4-nitrophenethyloxycarbonyl) and NPEOM (4-nitrophenethyloxymethyloxycarbonyl). Amino acid protecting groups are well-known in the field of peptide synthesis, and include groups such as those disclosed in Stewart, J.M. and Young, Solid Phase Peptide Synthesis. 2nd Ed., Pierce Chemical Company: Rockford, IL, 1984; Atherton, E. and Sheppard, Solid Phase Peptide Synthesis: A Practical Approach. IRL Press: New York, 1989; Jones, The Chemical Synthesis of Peptides (International Series of Monographs on Chemistry. No. 23), Clarendon Press: Oxford, 1991; Bodanszky, The.Practice of Peptide Synthesis..
Springer-Verlag: New York, 1984; Bodanszky, Peptide Chemistry: A Practical Textbook. 2nd Ed., Springer-Verlag: New York, 1993; Bodanszky, Principles of Peptide Synthesis. 2nd Ed., Springer-Verlag: New York, 1993; Synthetic Peptides: A User's Guide (Grant, W.H. Freeman: New York, 1992; and Barany, G. and Merrifield, "Solid Phase Peptide Synthesis", Chapter 1 (pp. 1-284) of The Peptides, Vol. 2, Academic Press: New York, 1979. Additional publications include the 97/98 Novabiochem Catalog and Peptide Synthesis Handbook and the Novabiochem Combinatorial Chemistry Catalog (Calbiochem-Novabiochem, San Diego, CA), and the user's manuals and synthesis bulletins for Perkin-Elmer Applied Biosystems (Foster City, CA) peptide synthesizers. Purification methods appropriate for peptides are discussed in the references cited above, and in High-Performance Liquid Chromatography of Peptides and Proteins: Separation. Analysis and Conformation (Mant, C.T. and Hodges, Eds.), CRC Press: Boca Raton, FL, 1991. Materials for use in peptide synthesis, such as protected amino acids, synthesis reagents, solvents, and resin supports, are available commercially from a number of suppliers, iicluding Calbiochem-Novabiochem, San Diego, CA; Advanced Chemtech, Louisville, KY; Bachem Bioscience, Inc., King of Prussia, PA; Sigma Chemical Company, St. Louis, MO; Richelieu Biotechnologies, Inc., Montreal, Quebec, Canada; Peninsula Laboratories, Inc., Belmont, CA; Perkin-Elmer Applied Biosystems, Inc., Foster City, CA; and Peptides International, Louisville, KY.
An "amino-capping group" or "amino-terminal capping group" or"N-terminal An aagnooappn. groupEno capping group" is a group that covalently links to an amino group. Examples of aminocapping groups include, but are not limited to, 4-morpholinocarbonyl, acetyl, and trifluoroacetyl.
Novel quinones While not wishing to be bound The present invention encompasses novel quinones. While not wishing to be bound by any particular theory explaining quinone toxicity, the inventors suggest that the novel quinones can be designed based on the suspected DNA topoisomerase II'poisoning activity of quinones. Alternatively, quinone toxicity may be related to the compound's potential to undergo redox cycling with the, formation of highly reactive oxygen species. O'Brien (1991) Chem. Biol. Interactions 80:1-41. In the next step. the quinone is tested in vitro for efficacy in inhibiting proliferation of diseased cells (such as tumor cells). If it is efficable, the quinone is then tested in animals, such as nude mice with tumor Xenografts.
Simultaneously, toxicity of the compound shouldbe determined. If the quioneis found to efficable and safe, testing can then proceed to human trials.
In vitro testing of novel quinones Novel quinones of the present invention can be tested in vitro by any means known in the art. The quinones can be tested, for example, for toxicity against a chosen cell line, such as a tumor cell line.
In vivo testing of novel quinones Following a showing of efficacy of the novel quinones in vitro, these compounds can be tested in vivo.. Typical tests include, but are not limited to, examinations of the 2 5 can be tes n effects of compound administration on animals, such as nude mice with tumor xenografts.
Methods of administrating quinones The novel quinone compounds of the present invention can be administered to an individual via any route known in the art, inluding, but not limited to, those disclosed herein. Preferably administration of the novel quinones is intravenous. Other methods of administration include but are not limited to, oral, intrarterial, intratumoral, intramuscular, 29 subcutaneous, intraperitoneal, gastrointestinal, and directly to a specific or affected organ.
The novel quinone compounds described herein are administratable in the form of tablets, pills, powder mixtures, capsules, injectables, solutions, suppositories, emulsions, dispersions, food premixes, and in other suitable forms. Additional methods of administration are known in the art. The pharmaceutical dosage form which contains the compounds described herein is conveniently admixed with a non-toxic pharmaceutical organic carrier or a non-toxic pharmaceutical inorganic carrier. Typical pharmaceuticallyacceptable carriers include, for example, mannitol, urea, dextrans, lactose, potato and maize starches, magnesium stearate, talc, vegetable oils, polyalkylene glycols, ethyl cellulose, poly(vinylpyrrolidone), calcium carbonate, ethyl oleate, isopropyl myristate, benzyl benzoate, sodium carbonate, gelatin, potassium carbonate, silicic acid, and other conventionally employed acceptable carriers. The pharmaceutical dosage form can also contain non-toxic auxiliary substances such as emulsifying, preserving, or wetting agents, and the like. A suitable carrier is one which does not cause an intolerable side effect, but which allows the novel quinone compounds to retain its pharmacological activity in the body. Formulations for parenteral and nonparenteral drug delivery are known in the art and are set forth in Remington 's Pharmaceutical Sciences, 18th Edition, Mack Publishing (1990). Solid forms, such as tablets, capsules and powders, can be fabricated using conventional tableting and capsule-filling machinery, which is well known in the art. Solid dosage forms can contain any number of additional non-active ingredients known to the art, including excipients, lubricants, dessicants, binders, colorants, disintegrating agents, dry flow modifiers, preservatives, and the like. Liquid forms for ingestion can be formulated using known liquid carriers, including aqueous and non-aqueous carriers, suspensions, oilin-water and/or water-in-oil emulsions, and the like. Liquid formulations can also contain any number of additional non-active ingredients, including colorants, fragrance, flavorings, viscosity modifiers, preservatives, stabilizers, and the like. For parenteral administration, novel quinone compounds can be administered as injectable dosages of a solution or suspension of the compound in a physiologically acceptable diluent or sterile liquid carrier such as water or oil, with or without additional surfactants or adjuvants. An illustrative list of carrier oils would include animal and vegetable oils (peanut oil, soy bean oil), petroleum-derived oils (mineral oil), and synthetic oils. In general, for injectable unit doses, water, saline, aqueous dextrose and related sugar solutions, and ethanol and glycol
I
solutions such as propylene glycol or polyethylene glycol are preferred liquid carriers. The pharmaceutical unit dosage chosen is preferably fabricated and administered to provide a final concentration of drug at the point of contact with the cancer cell of from I tpM to mM. More preferred is a concentration of from 1 to 100 pM. As with all pharmaceuticals, the optimal effective concentration of novel quinone compounds will need to be determined empirically and will depend on the type and severity of the disease, route of administration, disease progression and health and mass or body area of the patient. Such determinations are within the skill of one in the art.
The following examples areprovided to illustrate, but not limit, the invention.
EXAMPLES
EXAMPLE 1 Synthetic Preparation of Quinone Compounds Preparation of quinones of the invention is described below and depicted in the Figures.
New chemistry was developed in order to construct drugs where the 1,2naphthoquinone moiety is bound to a DNA minor groove binder unit or a DNA intercalator. While not wishing to limit the invention to any particular theory of operation, it is believed that the 1,2-naphthoquinone derivatives "poison" topoisomerase II and transform this essential DNA replication enzyme into a nuclease-type enzyme that cleaves DNA. It is postulated that this modification of topoisomerase II by the 1,2naphthoquinones is very likely due to the alkylation of the thiol residues of the enzyme by the quinones (Michael additions).
Scheme 1 outlines derivatization reactions leading to 1,2-naphthoquinone intermediates. The silver salt of 2-hydroxy-l,4-naphthoquinone was alkylated with the tert-butyl or benzyl esters of 5-bromo-pentanoic acid to give either 1 or 2. The benzyl ester 2 was transformed into the acid 3 by hydrogenolysis. The silver salt was also alkylated with 6-bromohexanol to give 4, or with 1,6-diiodohexane to give 5. The alcohol 4 treated with triphosgene gives 6 (Scheme The acid 3 can be derivatized by reaction with 3- 31 (Baird and Dervan (1996) J. Am. Chem. Soc.
118:6141) in the presence -of o-benzotriazol-1-yl-N,N,N',N'-tetramethyluronium hexafluorophosphate (HBTU) and diisopropylethyl amine (DIEA) to give the amide 7. The silver salt of 2-hydroxy-1,4-naphthoquinone reacted with pivalyl chloride to give 8 (Scheme Acid 3 was condensed with the polypyrrole amide 9 (Baird and Dervan (1996) J. Am. Chem. Soc.. 118:6141) after cleavage of the protecting t-butyl group with TFA. The resulting product 10 is a molecule where the 1,2-naphthoquinone moiety is covalently bound to a DNA minor groove binder (Scheme Alcohol 4 was condensed using the Mitsonobu reaction (triphenylphosphine, diethyl acetylenedicarboxylate) with 4-hydroxybenzonitrile to give 11. Iodide 5 was reacted with the tetrabutyl ammonium salt of phenol to give 12.
The acid 3 was esterified with 3-dimethylaminophenol using dicyclohexylcarbodiimide (DCC) and 4-dimethylamino pyridine (DMAP) and gave 13. By reaction of 5 and the tetrabutylammonium salt of Hoechst 33528 it was possible to obtain 14, where the quinone is covalently bound to the DNA minor groove binder. By esterification of 4 with 6-aminohexanoic acid (used as its BOC derivative and deprotected with TFA) in the presence of DCC and DMAP, it was possible to obtain 15 as its trifluoroacetate (Scheme By condensation of the acid 3 with the N-ethyl diamide 16, the polyamide quinone 17 was prepared (Scheme 4).
A new class of 4-aminoalkyl substituted 1,2-naphthoquinones was obtained following the outline depicted in Scheme 5. A Vilsmeier reaction on 1.2dimethoxynaphthalene gave the formyl derivative 18. Il was converted by reductive amination with n-butylamine into 19. Treatment of 19 with acetyl chloride gave 20, while treatment with trifluoroacetic anhydride gave 21 (Scheme Acylation of 19 with morpholino succinyl chloride gave 22. Cleavage of the 1,2-dimethoxy groups of 19 with boron tribromide gave the quinone 23 which was found to exist in the p-quinonemethine form. Cleavage of the dimethoxy residues of 20 and 21 led to the expected quinones 24 and 25. Cleavage of the methoxy residues of 22 gave the quinone 26 (Scheme The 1,2-naphthoquinone residue was also covalently bound to a porphyrin backbone, since porphyrins are known to concentrate in cancer tissues. By reaction of the iodide 5 with the tetrabutylammonium salt of meso-p-hydroxyphenylporphyrin, the porphyrin quinone 27 was obtained (Scheme 6).
32 By esterification of 4,4 ,4 ,4 -(21H, 23H-porphine-5,10,15,20tetrayl)tetrakis(benzoic acid) with the quinone alcohol 4 in the presence of EDCI dimethyl aminopropyl)-3-ethylcarbodiimide) and DMAP it was possible to prepare the Squinone-porphyrin 28 (Scheme 7).
SSynthesis of 1,2-naphthoquinones bound to DNA intercalators SIt is known that 4-aminoacridine derivatives intercalate in the DNA helix.
Therefore syntheses of 1,2-naphthoquinone residues bound to 4-aminoacridine derivatives Swere designed (Scheme The salt (6-hydroxyhexyl)triphenylphosphonium bromide was prepared by the reaction of 6-bromohexanol with triphenylphosphine in refluxing acetonitrile. Wittig reaction of (6-hydroxyhexyl)triphenylphosphonium bromide with 4acetamidobenzaldehyde produced alkene 29 as a mixture of E and Z isomers. Reduction of the double bond (H7, Pd/C) and acidic hydrolysis (2N HC1, MeOH) afforded 4-(7hydroxyheptyl)-aniline 30. Aniline 30 was reacted with 9-chloroacridine in MeOH in the presence of triethylamine to give alcohol 31. Alcohol 31 was converted to iodide 32 by reaction with methanesulfonyl chloride in pyridine, followed by reaction with sodium iodide in acetone. Reaction of iodide 32 with the silver salt of 2-hydroxy-1,4naphthoquinone afforded quinone 33 as a mixture of ortho- and para-quinone isomers. The ortho- and para-quinone isomers could be separated and purified by column chromatography.
A second approach to these types of compounds is shown in Scheme 9. The isomer 1) mixture 34 was converted to the iodide 35 by reaction with methanesulfonyl chloride in
CH
2
CI
2 in the presence of pyridine, followed by a displacement with sodium iodide in acetone. Reaction of 35 with triphenylphosphine in refluxing acetonitrile afforded the phosphonium salt. A Wittig reaction between the phosphonium salt and naphthaldehyde 18 produced diene 36 (as a mixture of double bond isomers). Reduction with H 2 over Pd/C followed by hydrolysis (2N HC1, MeOH) gave aniline 37. Aniline 37 was reacted with 9chloroacridine in MeOH in the presence of triethylamine to give 38. Cleavage of the methyl ethers with boron tribromide gave quinone 39.
A third synthetic approach to a 1,2-naphthoquinone moiety bound to an aminoacridine intercalator is depicted in Scheme 10.. Aminoacridine was protected with mesitylenesulfonyl chloride to give 41, which was then alkylated with 33 to 42. The latter is brought into reaction with the silver salt of 2-hydroxy-1,4naphthoquinone and the quinone-acridine 43 was thus obtained. Cleavage of the amide group using samarium iodide gave 44, the expected compound.
Synthesis of 1,2-naphthoquinol phosphates In order to obtain 1,2-naphthoquinone derivatives that behave as "pro-drugs" the synthesis of quinol phosphates that can be hydrolyzed by cell phosphatases to liberate the parent quinones was carried out. Scheme 11 outlines the synthesis of the quinol phosphates. The parent 1,2-naphthoquinone 46 was brought into reaction with dibenzylphosphite to give a mixture of the two possible regioisomers 47. By cleavage of the benzyl residues with hydrogen in the presence of 10% Pd on charcoal the mixture of the two possible quinol phosphates 48 was obtained. They were used as such in the biological studies.
Synthesis of 8-hvdroxy-3-lapachone Scheme 12 outlines the synthesis of 55, a phenol derivative of p-lapachone that could be used as a building block for the construction of peptide derivatives of Plapachone. The synthesis.starts with the commercially available ester 49, that is acetylated using a Friedel-Crafts reaction to give 50. Cyclization of 50 in the presence of base and air gave the p-quinone 51. Alkylation of 51 with dimethyl allyl bromide gave a mixture of the C-alkyl derivative 52 and the O-alkyl derivative 53. They were separated and on treatment of 52 with concentrated sulfuric acid, the 8-methoxy-p-lapachone 54 was obtained. O.
Cleavage of the methoxy group with boron tribromide gave the expected o-naphthoquinone Synthesis of 1,2-naphthoquinone bisulfite adducts Bisulfite adducts of 1,2-naphthoquinones were prepared as "pro-drugs." They are stable in aqueous solutions at pH below 7 but liberate the quinone core at pH above 7.
Since biological media are usually above pH 7, the bisulfite adducts led to a slow release of the quinones after administration in an aqueous medium. A list of selected bisulfite adducts is given in Fig. 1. General preparation procedures are given in Experimental.
Sythesis of 1 2-naphthoquilofe peptides I ,2-Naphthoquilofe conjugates of tetra and hexapeptides were prepared to obtain 11prodrug" derivatives that can be cleaved by prostatic PSA. The guidelines followed for the synthesis of the peptides were based on the published results of Isaacs and coworkers S (Denmeade et al. Cancer Res. 1997, 57, 4924), where they define the substrate specificity of PSA (prostate specific antigen). The synthesis of a quinone tetrapeptide is outlined in Scheme 13 for the 3-f -alanyloxy-f3-lapachone (SL-l 1006) conjugate. SL-l 1006 (Quin) was coupled to Boc-Gln with DCC in the presence of I -hydroxybenzotr-iazole to give Boc- Gin-Quin. Removal of the Boc group from Boc-Gln-Quin with TFA in CH 2 CI2 gave TFA-Gln-Quin. Boc-Leu was coupled to TFAKGln-Quin with DCC in the presence of 1 hydroxybenzotriazole to. give.Boc-Leu-Gln-Quin. Removal of the Boc group from Boc- Leu-Gln-Quifl with TFA in CH 2
CI
2 gave TFA-Leu-Gln-Quifl. Boc-Lys(Ne-Cbz) was coupled to TFA-Leu-Gln-Quifl with DCC in the presence of l-hydroxybelzotriazole. to give Boc-Lys(Ns-Cl-Cbz)-Leu-GlnQuin. Removal of the Boc group from Boc-Lys(NS-Cbz)- Leu-Gln-Quin with TFA in CHC1 3 gave TFA.Lys(Ns-Cbz)LeuGln-Quin. Morpholino- Ser(OBn) was coupled to TFA-LyscNs-Cbz)-Leu-Glfl-Quin with DCC in the presence of I1- *hydroxy benzotriazole to give m 0 1 rp 0 hlo-er(OBn)-Lys(N -Cbz)-Leu-Gln-Quifl. The side chain protecting groups were removed by hydrogenolysis to yield morpholino-Ser-Lys- Leu-Gln-Quifl. During the hydrogenolysis, the quinone was reduced. to the hydroquinone, which reoxidized to the quinone on exposure to air.
Morpholino-Ser(OBfl) was prepared from N-Fmoc-Sef(OBn). Esterification of N- Fmoc-Ser(OBn) with isobutylene in the presence of a catalytic amount of H 2 S0 4 afforded N-Fmoc-Ser(OBn)-QtBu. The Fmnoc group was removed with piperidine in CH 2
CI
2 to.
produce Ser(OBn)-OtBu. Reaction of Ser(OBn)-OtBu. with 4-morpholinecarbonyl chloride in pyridine yielded morpholino-Ser(OBfl)-OtBu. Morpholino-Ser(OBn)-OtBu was hydrolyzed with ThA in CH 2
CI
2 to yield morpholine-Ser(OBn).
The synthesis of a tetrapeptide conjugate of 3-leucyloxy-43-lapachone is outlined in Scheme 14.
EXPERIMENTAL
tert-Butyl 5-1(1,2-dihydro-l,2-dioxonaphth-4-yl)oxylvalerate A mixture of tert-butyl 5-bromovalerate (1 g, 4.2 mmol) and the silver salt of 2-hydroxy-1,4naphthoquinone (0.8 g, 3.84 mmol) in benzene (10 mL), was stirred for 24 h at 500 C. The reaction mixture was filtered through celite and the solvent was removed in vacuo. The residue was purified by flash chromatography methanol in chloroform) to give a yellow solid (384 mg, 'H NMR (CDCl 3 8.12 J=7.7 Hz, 1H), 7.89 J=7.7 Hz, 1H), 7.70 J= 6.1 Hz, 1H), 7.59 J=6.4 Hz, 1H), 5.95 1H), 4.17 J=5.9 Hz, 2H), 2.35 J=7.2 Hz, 2H), 1.90-2.05 2H), 1.78-1.90 2H), 1.47 9H).
Benzyl 5-[(1,2-dihydro-1,2-dioxonaphth -4-yl)oxylvalerate A mixture of benzyl bromovalerate (2.27 g, 8.4 mmol) and the silver salt of 2-hydroxy-1,4-naphthoquinone (1.63 g, 5.81 mmol) in benzene (8 mL) was stirred for 48 h at 550 C and filtered through celite. The filtrate was diluted with diethyl ether, extracted with a 20% aqueous solution of NaHSO 3 then basified to:pH 10-11 with Na2CO 3 and extracted with CH 2 zC 2 Yellow solid (1.334 g, 'H NMR (CDC13) 8.12 J=7.5 Hz, 1H), 7.85 J=7.7 Hz, 1H), 7.68 (t, J=7.5, lH), 7.58 J=7.7 Hz, 1H), 7.25-7.50 5H), 5.93 1H), 5.14 2H), 4.15 (t, J=5.7 Hz, 2H), 2.50 J=7.0 Hz, 2H), 1.8-2.2 4H).
5-[(1,2-Dioxo-l-,2-dihydronaphth-4-yl)oxylvaleric Acid Benzyl ester 2 (1.90 g, 5.22 mmol) was hydrogenated at 30 psi with Pd (400 mg) in ethyl acetate (120 mL) for 6 h. The catalyst was removed by filtration through celite, the solvent was evaporated in vacuo and the residue was oxidized with Ag 2 0 (1.45 g, 6.25 mmol) in Et20 by stirring for 10 h.- Following filtration and evaporation of the solvent the product was crystallized from benzene to afford 0.53 g of pure material. The mother liquor was purified by flash chromatography (CH 2 Cl 2 /MeOH 15:1), the product dissolved in CH 2 Cl 2 extracted with aqueous NaHCO3 solution, acidified to pH 1 with 3% HC1 and extracted back with CH 2
CI
2 to give additional 0.25 g of pure material (total yield mp 134-136 0 C; 'H NMR
(CDCI
3 8.12 J=7.0 Hz, I1H), 7.87 J=7.6 Hz, 1KH), 7.70 3=7.5 Hz-, 11-H), 7.59 (t, J=7.4- Hz, 7.27 1K), 4.18 3=5.9 Hz, 2H), 2.51 3=7.0 Hz, 2H), 1.75-2.15 (m, 4H).
I ,2-Dihydro.4-(6-hy droxyhexyloxy) 4 ,2-dioxo-naphthalene A mixture of 6bromohexanol-l (4.5 g, 24.85 mmnol) and the silver salt of 2-hydroxy-1,4-flaphthoquinone_ (6.46 g, 23.01 mmol) in benzene (24 mL) was stirred for 48 h at 600 C. The reaction mixture was worked up as described for 2 and crystallized from hexane to afford a yellow solid (3.18 g, np, 96-98 0 C, 'H NMR (CDC)8.2(,J5Hz 1H), 7.87 J=7.7 Hz, 1K), 7.70 J=7.5 Hz, 1K), 7.58 J=7.5 Hz, 1K), 5.95 IK), 4.15 J=6.3 Hz, 2H), 3.69 J=6.2. Hz, 2H),'-1.9-2-1.97 (in, 2K), 1.3-1 (mn, 7H)~ 1,2-Dihydro-4-(6-iodohexyIoxy) -1,2-dioxonaphthalefle A Mixture of 1,6diiodohexane (10. 14 g, 30 inmol) and the silver salt of 2-hydroxy'-l,4-flaphthoquinone (2.81 g, 10 mm ol) in benzene (60 mL) was stirred for 12 h at room temperature. The reacton mixture was filtered through Celite, concentrated in vacuo, and purified by flash chromatography (hexane/ EtOAc 4:1) to give a yellow solid .(2,19 g, mp 85-870C
'H
NMR (CDCI 3 8.12 (dd, J=6.5, 1.0 Hz, 1H), 7.86 (dd, J=6.9, 0.9 Hz, 7.70 (dt, J=7.6, Hz, IKH), 7.5 8(dt, J=7.5, 1.3 Hz, IlH), 5.95 1 4.15 J=6.3,2H), 3.22 6 9 Hz, 2H), 1.80-2.05 (in, 4H), 1.45-2.10 (in, 4H).
is 164(1,2- Dihydro-7l,2-dioxo naphtfi4-yI)oxyj hexy~jearbonate Pyridine (0.12-ml, mniol) was added to a stirred solution of the alcohol 4 (200 mg, 0.73 mmol) and bis(trichloroinethyl)carbonate (40 mg, 0.134 inmol), in CK 2
CI
2 (5 mL) at 0 0 C. The cooling bath was removed, the reaction mixture was dilute d with CH 2
CI
2 washed with 3%.HCI, brine, dried (Na 2
SO
4 and purified by column chromatography (benzenelEtOAc 4:1, 2:1).
The product was triturated with Et 2 O to afford a yellow. solid (127 mng, mp 7.8-820C (decomp.). MS (LSIMS, 3-NBA) 576 401, 17 5; 'H NMR (CDC1 3 8.09 (dd, 1.6 Hz, 1KH), 7.8 5 (dd, J=7.8, 1.2 Hz, I1H), 7.71 J=6.9 Hz, 1KH), 7.589 3=6.2 Hz, 1KH), 5.94 1H), 4.17 J=6.0 Hz, 2H), 4.15 J=5.6 Hz, 2H),-1.85-2.10 (in, 2K), 1.65-1.85 (in, 2H), 1.40-1.65 (in, 4H).
N-(1-Methyl-5-methyloxycarbonylpyrrol-3-yI)-5-(1 ,2-dihydro-1 ,2-dioxonaphth -4yI)oxylvaleramide A solution of an acid 3 (3 34 mg, 1.22 mmol) in DMF (1.67 mL) was treated with HBTU (462 mg, 1.22 mmol) followed by DlEA (452 mg. 3.5 mmol) and stirred for 5 min. 3 -Amino-1I-methyl- 5-methyloxycarbonylpyrrol hydrochloride (232 mg, 1.22 mmol) and DIEA (3 78 mg, 3 n-mol) were added to the reaction mixture. The latter was stirred for 2 h, diluted with EtAO the precipitate was removed, dissolved. in CHCl 3 washed with 3% HCI, H 2 0, aqueous NaHCO 3
H
2 0 again, dried (MgSO 4 and purified by chromatography on alumina column (CHC1 3 /MeOH 80:1, 50: The product was triturated with Et 2 Q/CHC1 3 to obtain a yellow-red solid (200 mg, mp 122-1230 C (decomp.): MS (LSIMS. _3-NBA) 410 237 (M 4 -173). H NMR (CDCI 3 8. 08 J=7.5 Hz, lH), 7.$6 J=7.3 Hz, 1H). 7.68 J=7.5 Hz. 1H). 7.57 J=7.5 Hz, IH), 7.38 J= 1.8 Hz, IH), 7.34 I 6.65 (d J1=2 Hz, I 5.95 I1H), 4.19 J=5.53 Hz, 2H), 3.88 3H), 3.80 3 2.46 J=6.6 Hz, 1.90-2.15 (in, 4H).
4-(tert-Butylcarbonyloxy)-1,2- dihydro-1,2-dioxonaphthalene A mixture of the silver salt of 2-hydroxy- I ,4-naphthoquinione (842 mg, 3 mmol), and pivaloyl chloride (434 mg, 3.6 mmol) in benzene (5 mL) was stirred for 8 h at room temperature. Th e reaction.
mixture was filtered through Celite, the precipitate washed with EtOAc, and the combined organic solutions were concentrated in vacuo and purified by flash chromatography (EtOAc/hexane. 1: 10, The product was recrystallized from hexane to afford a yellow solid (190 mng, mp 125 -126 0C H NMR (CDC1 3 8.15(dd, J= 7.7, 1.1 Hz, 1H), 7.71 (dt, J1=7.7, 1.5 Hz, I1H), 7.59 (dt, J=1.5, 1.2 Hz, I1H), 7.57 (dd, J= 7.6, 1.1 Hz, I 6.48 1iH), 1 .44 9H1).
N-[3-(Diniethylamino) propyl [13-1[3-113-14-1(1 ,2-dihydro-1 ,2-dioxonaphtb-4yI)oxyl b utyl ca rbonhyla min o I-I-m ethylIpyrro1-5.-yll carbonylam in oI-1-methylpy (10) was prepared from acid 3 (61 mg, 0.222 mmol) and B oc-protected pyrrolylamine 9 (84 mg, 0. 148 mrnol) using the procedure described for 7. After the reaction was completed, the reaction mixture was diluted with Et 2 O, the precipitate was removed, triturated with hot EtOAc and. crystallized from a CHC13I Et 2 O mixture. The product was a yellow solid (30 mg, mp 1 5 9 1 62O (decomp.); 'HNMR (DMSO-d6) 9.90 111), 9.89 9.86 IH), 8.08 (bs, 111I), 7.
97 J= 7.7 Hz, 111), 7.87 J=7.2 Hz, 111), 7.81 J=7.9 Hz, IH), 7.68 3=7.2, 111), 7.24 I11) 7.19 IH), 7.04 111), 6.89 1H1), 6.84 111); 6.06 IH), 4.25
J=
5.8 Hz, 3.85 31H), 3.84 311), 3.80 (s,.311),3.12-3.30 (in, 2H1), 2.25-2.45 (in, 411), 2.19 611), 1.72-2.00 (in, 411), 1.60-1.70 (mn, 2H). ).MS (LSIMS, 3-NBA) 725.2 1,-iyr--6[4-ynpey~xleyoy -l ,2-dioxonaphthalene
A
mixture of 4-hydroxybeflzofitrile (87 mg, 0.73 inmol), naphthoquilofe 4 (200 mg, 0.73 I0 imol), PPh 3 (9rn,07imo)n dixie (10 inL was cooled to10 0 C And treated with DEAD (140 mg, 0.80 inrnol). The reaction mixture was stirred for 10 h, concentrated in vacuc and purified by chromatography EtOAc in benzene) to afford 11 as a yellow *solid (171 mng. NMR
(CDCI
3 8.13 (dd, J=7-3, 1.4 Hz 111). 8.86 (dd. J=7.7, 1.1 Hz, I 7.67 (dt, J=7. 5, 1.5Hz 111), 7.60 (dt, J=7.5, 1.5 Hz, L 7.57 J=8.8 .Hz, 211), 6.93 J=8.9 Hz, 2H),5.96 111), 4.17 Hz, 211), 4.03 J=6.3 Hz,'211), 1.80- 2.05 (mn,4H), 1.58-1.68 (mn,4H).
l1, 2 -Dihydro-4-16-(phelyloxy)hexyloxYl -1,2-dioxo naphthalenle Phenol (28 mng, 0.3 iniol) was treated with tetrabutylammofliuin hydroxide (0.3 mL of 1.0 M solution in methanol) and the reaction mixture was concentrated to dryness in vacuo.
lodonaphtoquinone 5 (115 mg, 0.3 rnrol) in DMF.(3) rnL was added to the tetrabutylaminoniuin salt, stirred. for 48 h and quenched with 1120 (10 miL. The product was extracted with CHCl 3 the extract was washed with 1120, then brine, dried (Na 2
SO
4 and purified by chromatography EtOAc in benzene) to give 12 as a yellow solid m ig, 43 'H1 NMR (CDCI 3 8.13 J=7.4 Hz, 11H), 7.86 J=7.4 Hz, I11), .7.67 J=7.6 Hz, 111), 7.61 J=7.5 Hz, IH),7.15-7.40 (in, 211), 6.85-7.10 (in, 311), 5.96 111), 4.17 (t, Hz, 211), 3.99 3=6.2 Hz), 1.70-2.10 (mn, 411), 1.35-1.70 (mn, 411).
3-Dim ethylanlinop henyl 5-1(1 ,2-dihydro- 1, 2 -dioxonaphth4-yl)oxylvalerate
A
mixture of acid 3 (137 ing, 0.5 inmol), 3-diinethylaIminfophenol (82 mg, 0 .6 inmol),
DCC
(103 mng, 0.5 mmol), and DMAP (12 ing, 0.01 mniol) in THF (2 mL) was stirred for 2 h.
39 The- reaction mixture was concentrated in vacuo, the residue dissolved in benzene, washed with H 2 0 a nd dried (Na 2
SO
4 Column chromatography 10% EtOAc) in benzene gave 13.
as a yellow solid,.(70 mg, 1 H NMR (CDCI 3 8.13, J=73 Hz, 1H), 7.90 J=7.4 Hz, 1H), 7.69 J=6.1 Hz, 7.58 J=7.6 Hz, 1H), 7.22 (dd, J=8.1, 8.1 Hz, IH), 6.30- 6.70 (in, 2H-I), 5.96 1H), 4.21 J=5.6 Hz, 2H), 2.69 Jr=6.5 Hz, 2H), 1.90-2.15 (in, 4H).
2'-[4-j16-(1,2-Dihydro-1 ,2-dioxo-naphth -4-yl)oxyhexylljoxyphenylj-5-(4methylpiperazin-1-yl)-2,5'-bi-1H-benzimidazole Hoechst 33258 (3.0 g, 5 mmol) was dissolved in a hot mixture of isopropanol-water (24 mLI 12 mL) and neutralized with ammnonium hydroxide (3 mL). The precipitate. was filtered, triturated with Et 2 'O and dried in vacuc to obtain the free base of bisbenzimidazole. A 1.0 M solution of BU.4NOH in MeOH (0.6 mL, 0.6 minol) was added to the solution of bisbeazimidazole (1.635 g, 3.85 minol) in MeOH (30 mL), stirred for 15 min and concentrated to dryness in vacuo.
Iodonaphthoquinone 5 (1.485 g, 3.87 n-mol) in DMF (30 mL) was added to the tetrabutyl a mmonium salt and the mixture was stirred for 48 h. The reaction mixture was suspended in H 2 0, the crude product was filtered, washed with H 2 0, dried and purified by flash chromatography (MeOH/CHCl 3 1:9, 1:5) to afford 14 as a yellow solid (790 mg, 'H NMR (CDC1 3 /MeOH-d 4 8.21 1H), 8.09 J=7.6 Hz, 1H), 8.05 J=8.7 Hz, 2H), 7.85- 7.95 7.48-7.75 (in, 4H), 7.14 (bs, 1H), 7.10-6.98 (mn 3H), 4.21 J=6.3 HZ, 2H), 4.08 J=6.2 Hz, 2H), 2.65-2.75 (in, 4H), 2.40 3H), 1.80-2.15 (in, 4H). 1.60-1.75 (in.
4H). MS (LSIMS, 3-NBA) 725.2 Trifluoroacetate of 6-[(1,2-dihydro-1 ,2-dioxonaphth-4-yI)oxy] hexyl 6-aminohexanoate [6-(tert-Butyloxycarbonyl)aminolhexanoic acid (139 mg, 0.6 mmol) was-added into solution of DCC (11 mg, 0.55 mmol) and DMAP (64 mg, 0.52 mmol) in CH 2 C1 2 CLQrnL) at 0 0 C and stirred for 15 min, when naphthoquinone 4 (137 mg, 0.5 iniol) was added. The reaction mixture was stirred for 12 h at room temperature, diluted with CH 2 Cl 2 extracted 3 times with an. aqueous -solution of KHSO 4 then with a NaHCO 3 solution followed by brine, dried (MgSO 4 and finally it was concentrated to, dryness in vacuo and triturated with Et 2 O. The residue was dissolved in CH 2
CI
2 (3 mL), 'EPA (0.5 inL) was added to the solution and the mixture stirred at 0 0 C for Ilh. All volatiles were removed in vacuo and the residue was triturated in Et 2 O to give 15 (100 mg, as a dark yellow oil. HK NMR (ODC1 3 8.90 J=7.6 Hz, 1KH), 7.99 (bs, 3KH), 7.87 J=7.8 Hz1, 11K), 7.71 J=7.6 Hz, 1KH), 7.59 J=7.5 Hz, I 5 .96 IlH), 4.17 J=6.3 Hz, 2H), 4.09 J=62 HIz, 2H), 2.90-3.15 (in, 2.29 J=7.1 Hz, 2H), 1.90-2.10 (mn, 2H), 1.30-1.85 (mn, 12H).
1 ,2-Dihydro-1 ,2-dioxo-4-1-2-1312- (E~thylaminocarbolyl)ethylarninocarbonyll propyl= aminocarbo nyIlethyla minoca rbony 11 butyloxyl naphthalene Acid 3 (137 mg, inmol) was dissolved in DMF (I inL), treated with KBTU (190 mg, 0.5 mmiol) followed by DIEA (260 1 .iL, 1.5 inmol) and stirred for 10 min. N-Ethyl[2-[3-( 2 aminoethylcarbonYlmiflo) propylcarboflylaminflethyllcarboxmide hydrochloride 16 (154 mng, 0.5 inmolY* and DIEA (260 p.L,l .5 mmol) were ddded to the reaction mixture, the latter was stirred for 2 h, and the reaction mixture was diluted with Et 2 O. The product was* filtered and triturated with .CHC1 3 to afford a yellow solid (100 mng, mp 145-170'C (decomp.) H NMR (CDC1 3 MeGH-cii) 8.10 (dd, J=7.6, 1.4 Hz, 11H), 7.92 (dd, J=7.8, 1.2 Hz, IlH), 7.72 (dt, J=7.7, 1.2 H47. 1 7.62 (dt, 7.6, 1.3 Hz, 1KH), 7.3 0-7.50 (mn, 2H), 7.15 (bs, 1K), 5.97 1H), 4.20 J=5.8 Hz, 2K)7 3.35-3..50 (in, 4K), 3.10-3.30 (in, 4K), 3.32- 3.42 (mn, 4H), 2.30 J=6.9 Hz, 2H), 2.19 J=7.4 Hz, 2K), 1.75-2.05 (mn, 4H), 1.78 (t, 3=7.2, 2K), 1.13 J=7.3, 3H). MS (FAB, Nal) 551.2 529 3 ,4-Dimnethoxy-l-na2Phthaldehyde A mixture of l,2-dimethoxynaphthalefle (0.74 g, 4 mmol) and DMF (0.8 mL, 1.0 inmol) in dichlorobenzene (0.8,mL) was stirred with POC1 3 at I 0000 for 2h. The- reaction mixture was cooled to 00) C, quenched with a cold aqueous solution of NaOAc, diluted with H 2 0 and extracted with benzene. The extracts were dried (MgSO4), concentrated and in vacuo and dichlorobenizene was removed by kugelrohi distillation at I I IftI0.5 mm Hg.. Column chromatography (20%EtOAc in hexane) gave the produc t 18 (596 mng, which was used in the following step without further purification. 1 HNMR (CDC3) 10.42 I 9.00-9.15 8.15-8.30 (in, I 7.61 I 7.50-7.65 (in, 2K), 4.12 3H), 4.07 s,3H).
4 -Butylaminomethyl-1,2dimethoxy-naphthalene A suspension Of PtO 2 (40 ing) in EtOK (2 inL) was stirred with.K 2 at 25 psi for- 30 min. Naphthaldehyde 18 (596 mg. 2.8 41 mmol) was dissolved in EtOH and added into the suspension followed by the addition of butylainine (219 mg, 3 mmol).. The. reaction mixture was hydrogenated for 6 h at 50 psi.
The catalyst was filtered through Celite, washed with acetone and the filtrate was concentrated to dryness to give 19 as an oil (665 mg, The product was utilized in the following step without further purification. 'HNMR (CDC1 3 8.16 J=7.5 Hz, 1H), 7.99 J=7.6 Hlz, 2H), 7.40-7.60 (in, 2H), 7.35 1H), 4.19 2H), 4.100 3H), 3.98 3H), 2376 Hz, 2H1), 1.64 (bs, 1H), 1.45-1.60 (in. 2H), 1.30-1.45 (in, 2H), 0.93 J=7.2 H-z,314).
4-NAey--uyaio'tyl-,-iehx-ahhln Triethylamine (350 p1L, 2.5 mmol) was added to a solution of aminonaphthalene 19 (250 mg, 0.9 minol) and AcCi (90 .LL 1.27 mniol) in. CH 2 Cl2 mL) at 0 0 C. The coolin g bath was removed after min, the reaction mixture was stirred for 1 h at room temperature, diluted fivefold with
CH
2
CI
2 washed with an aqueous solution of NaHC0 3 followed by 3% HCl, brine and dried (MgSO 4 The crude product (3 15 mg, 100%) obtained after evaporation of the solvent was used in the following step without further purification. 'H NMR (CDCI 3 8.19,.8.15 (2d, J=7.6, 8.4 Hiz, 1H), 8.97, 7.80 (2d, J=7.9, 8.2 H7, I 73.35-7.58 (in, 2H), 7.16, 7.04 (2s, I 5.05, 4.9.5 (2s, 2H), 4.01, 3.99 (2s, 3H), 3.99, 3.96 (2s, 3H), 3.47, 3. 1 3 (2t, J=7.4, 7.8 Hz, 2H), 2.20, 2.09 (2s, 3H), 1. 15-1.70 (mn, 4H), 0.91, 0.87 (2t, J=7.2, 7.3 Hz, 3H).
4-(N-Butyl-N-trifluoroacetylaminomethyl)- 1,2-dimethoxy-naphthalene Naphthalene 19 (200 mng, 0.73 minol) was acylated with trifluoroacetyl anhydride (2 10 mrg,(1 1 inmol) in the presence of TEA (0.2 mL, 1.5 minol) by raising the temperature during'3 h from -400 to 0 0 C. The reaction mixture was diluted with CH 2 C1 2 washed with aqueous NaHCO 3 3% HCl, brine and finally dried (MgSO 4 The crude product (266 ing, 99%) was used in the following step without further purification.- 'H NMR (CD.C1 3 8.17-8.25 (in, 1H), 7.82 J=7.7 Hz, 1lH), 7.40-7.5 5 2H), 7.16, 7.03 (2s, I1H), 5.11, 5.08 (2s, 2H), 4.01, 4.03 (2s, 3H), 3.98, 3.96 (2s, 3H), 3.40. 3.25 (2t, J=7.5, 7.4 Hz, 2H), 1.45-2.75 (in, 2H), 1.10-1.45 (ni, 2H), 0.89 J=7.4, 3H).
4-[N-Butyl-N- [3-(4-mo rpholin ocarbonyl) ethy Ica rbonyll aminomethyll -1,2diniethoxynaphthalene -3 -(N-Morpholinocarbonyl)propionic acid (139 ing, 0.74 42 Duo)i
H
2 I 5 L a etdt reflux with thionyl chloride (440 mg, 3.7 mmol) for lhadall volatiles were evaporated in vacuo. Tersdewsdsovdi nyru CI-1H 2 C1 2 (3 mL), cooled to 0 0 C and naphthalenle 19 (100 mg 0.3 o) olwdb DMA? (45 mg, 0.37 rruol) and TEA (140piL, I minol) were added into the reaction mixture. After stir ring for I h at room temperature the reaction was quenched with wet EtOAc (10 MnL), washed-with 3 aqueous NaHCO3, brine, and dried (Na 2 SO4).
D Purification by chromatography (1 5%EtOAc in hexane) gave 22 (160 mig, The.
"AI
r) product was used directly in the next step. '1H NMR (CDC13) 8.19, 8.17 (2d, J=7.7, 7.8 Hz, D 7.92, 7.85 (2d, J=.,80 z, 7.38-7.56 (in 41-I), 7.25, 7.17 (2s, 5.05, 5.03 1K 0, (2s, 2H), 4.03, 4.00 (2s, 3H), 3.99, 3.98 (2s, 311), 3.25-3.82 (in, 14H-), 1. 15-1.82 4H), 0.88. 0.5(2,3. 6.7 Hz, 3H); Demethylation of dimethoxyflaphthatenes with boron tribromide. 4-(Butylamilo= methyene).,4-ihyr--hoxy-lonaph thaene A solution of* dimethoxyniaphthalene 19 (30 mg, 0.11 mnmol) in C11 2 C1 2 (2 rnL) was treated with a IlM solutiIon of BBr 3 in CIH2l2 (1.1 mL) at -79 0 C and stirred at this temperature for 2h. The reaction mixture was placed in a freezer at -1 Q0(2 for 3 h. quenched with Et 2 O (1 mL) by stirring for 15 min at room temperature and neutralized with aqueous solution-of NaHC03.
The product was extracted with EtOAc, dried (MgSO4), and the solvent was removed in vacuo. The residue was dissolved in Et 2 O, stirred for 10 h in an open flask and purified by chromatography MeOH in CHC1d 3 Trituration with Et 2 O yielded the product 23 (8 '1H NMR (CDC13) 9.05 (bs, 1HI) 8.31 J8.1 Hz, Il-H), 7.65-7.85.(,
IH),,
7.05-7.65 (mn, 3.20-3.60 (mn, 2H), 1.50-1.85 (in. 2H), 2.25-1.50 (in, 21-),.0.80-1-.10 (in, 3H). HRMS (El) 243.1250. Calcd for C 15
HI
7 N02 243.1259.
4-NAey-N krior ty) 1 ,2-dihydro-1, 2 -dioxonaphthalefe (24) was fepared from dimethoxynaphthalene 20 using the procedure described for 23. The product was purified by chromatography MeOH in CHCI3) followed by trituration with Et 2 O. 'H NMR (CDCl3) 8.1: (dd, J=7.53, 1.2 Hz, 1lH), 7.67 (dd, J=7.7, 1.1 Hz, 11-), 7.50-7.62 (mn, 2H), 6.21 IH), 4.68 3.35 J=8.0 Hz, 2H), 2.25 3H), 1.50-1.75 (mn, 2H), 1.15-1.50 (mn, 0.96 3H). HRMS (El) 285.1383 Calcd for C1 7
HI
9 N03 285.1365.
4-(N-Butylaminoniethyl-N-trifluorocetyl)-1 ,2-dihydro-1 ,2-dioxonaphthalene (25) wa-s obtained from dimethoxynaphthalene 21 using the procedure described for 23. The product was purified by chromatography MeOH in CHCI 3 followed by trituration in Et 2 O. 'H NMR (CDCI 3 8.29 J=7.13 Hz, 1H), 7.40-7.85 (in, 3H), 6.19 1H), 4.73 (s' 2H), 3.35-3.70 2H), 1.50-1.80 (mn, 2H), 1.35-1.80 (mn, 2H), .96 Hz, 3H).
HRMS (El) 339.1106. Calcd for C, 7
H,
6
F
3 N0 3 339.1082.
4-1 [N-B utyl-N-(4-mo rpho lin o4-oxo butyryI)ami no] m ethyl] 12-dihydro- 1,2dioxonaphthalene (26) was obtained from diinethoxynaphthalene 22 using the procedure described for 23. The product was purified by chromatography (25%-40% EtOAc ini hexane) followed by trituration in Et 2 O. 'H NMR (CDCI 3 8.19 J=7.4 Hz,.lH), 7.70 (t, J=6.4 Hz, 1H), 7.59 J=6.5 Hz, iN), 7.50 J=7.9 Hz, IN), 6.33 1H), 4.65 (s,2H), 3.3 5-3.80 (in, 14 1.65- 1.85 (in, 214), 1.25-1.50 (mn, 21-i), 0.96 J=7.2 Hz, 3 H).
meso-Tetral4-16-I(1,2-dihydro-1 ,2-dioxoniaphth -4-yl)oxyjhexyloxyj phenyll porphine A.1I M solution of Bu 4 NOH in MeON (0.212 mL,) was added to a stirred solution of meso-tetra(4-hydroxyphenyl)porphine (36 mg, 0.53,minol) in MeOH (5 mL), stirring was kept for 10 min and the mixture concentrated to dryness in vacuo. Naphthoquinone 5 (8 1 ing, 0.21 inmol) in DMF (2 mL) was added to the porphyrin, the solution stirred for, 48 h and diluted with H 2 0 (20 mL). The product was extracted with CHCI 3 washed with, brine, the solvent was evaporated and the residue was triturated with Et 2 O. Purification by flash chromatography MeOH in CHCI 3 followed by recrystallization from CHCI 3 lEt 2
O
afforded the product as a dark red solid (19.6 mg, 'H NMR (CDC1 3 8.86 (s, 8H), 8.0 1-8.15 (in, 12H), 7.9 J=7.8 Hz, 4H), 7.68 J=6.3'I Hz, 4H), 7.55 J=7.5 Hz, 4H), 7.27 J=7.8 Hz, 8H), 5.98 4H), 4.15-4.30 (in, 16 1.80-2-10 (mn, 16H), 1.6S- 1.80 (in, 16H). Anal. Calcd for C, 08 Hq 4
N
4
O
16 x1l.5 H 2 0: C, 74.87; H, 5.43; N, 3.23. Found: C, 74.62; H, 5.57; N, 3.11.
meso-Tetra[4-[6-[(1,2-dihydro1,2-dioxanaphth.4 yl)oxyhexylloxycarbonyliphenyIlporp hyrin EDCI (518 mng, 2.7 mrnol) was added at 0 0 C to a mixture of ineso-tetra(4-carboxyphenyl)porphyrin (500 mng, 0.63 rnmol), alcohol 44 4 (831 mg, 3 mmol), and DMAP (159 mg, 1.3 mmol) in CH 2
C
2 (10 mL). The solution was stirred for 2 h, the cooling bath was removed and the reaction mixture was left at room Stemperature overnight. It was diluted with CH2C1 2 washed with 2% HC1, H20, aqueous solution ofNaHCO3, H20, 5% aqueous solution ofNaHSO3, H20, dried (Na 2
SO
4 and concentrated in vacuo. The analytical sample was prepared by column chromatography on silica MeOH in CHC1 3 Mp 98-110°C (decomp.) Yield 572 mg, 50%. 'H NMR (CDC13) 8.81 8.45 J=8.2 Hz, 8H), 8.30 J=8.0 Hz, 81), 8.09 J=6.9 Hz, 4H), 7.89 J=7.3 Hz, 4H), 7.70 J=7.1 Hz, 4H), 7.56 J=7.1 Hz, 4H), 5.98 4H), O 4.56 J=6.5 Hz, 8H), 4.21 J=6.1 Hz, 8H), 1.85-2.20 16H), 1.60-1.80 16H). MS (MALDI) 1838 (M 4 1817 Anal. Calcd for Ci 2
H
94
N
4 0 20 x4 H20: C, 71.18; H, 5.40; N, 2.97. Found: C, 71,27; H, 5.24; N, 3.03. N-Acetyl-4-(7-hydroxy-l-heptenyl)-aniline A solution of 5.213 g (28.8 mmol) of 6-, bromohexanol and 7.55 g (28.8 mmol) of triphenylphosphine in 50 mL of CH 3 CN was refluxed for 24 hr. Evaporation of solvent yielded the crude phosphonium salt, which was used directly in the next reaction. The crude phosphonium salt and 4.690 g (28.7 mmol) of 4-acetamidobenzaldehyde were dissolved in a mixture of 150 mL of CH 2 C1 2 and 150 mL of THF. To the cooled solution was added 1.529 g (60.5 mmol) of 95% NaH as a slurry in
CH
2 Ci 2 (i mL). The reaction mixture was stirred in an ice bath for 1 hr, then at room temperature for 19 hr. The mixture was partitioned between 350 mL CH 2 C1 2 and 500 mL IN HCI. The aqueous phase was extracted with CH 2
CI
2 (4 x 100 mL). The CH 2 Cl 2 extracts were combined, dried with MgSO4, and evaporated to dryness. Column chromatography on silica gel eluting first with 1% MeOH in CH 2 C2 and then with 2% MeOH in CH 2
CI
2 afforded4.913 g (69% from 6-bromohexanol) of alkene 29 as a mixture of E and Z isomers: 'H NMR (250 MHz, CDC1 3 TMS) 5 7.5- 7.4 7.3-7.1 4H), 6.4-6.3 (m, 2H), 6.2-6.1 lH), 5.7-5.6 2H), 3.65 J 6.5 Hz, 2H), 3.63 J 6.5 Hz, 2H), 2.4- 2.1 4H), 2.T18(s, 3H), 2.17 3H), 1.7-1.3 12H).
4-(7-Hydroxyheptyl)-aniline To a solution of 4.913 g (19.9 mmol) of N-acetyl-4-( 7 hydroxy- -heptenyl)-aniline 29 in 100 mL of 10% MeOH in CH 2 C1 2 in a Parr bottle were added 490 mg of 10% Pd/C. The bottle was placed on a hydrogenation apparatus and shaken for 4 hr at 25 psi of hydrogen. Removal of catalyst by filtration through a celite pad and evaporation of solvent afforded 5.294 g of alkane: 'H NMR (300 MHz, CDCI 3 TMS) 8 7.80 NH), 7.38 J =8 Hz, 2H), 7.09 J 8 Hz, 2H), 3.61 J 6.6 Hz, 2H), 2.54 (t, J 7.6 Hz, 2H), 2.12 3H), 1.6-1.5 4H), 1.4-1.3 6H).
A solution of the alkane in 40 mL of MeOH was mixed with 190 mL of 2N HCI.
The reaction mixture was refluxed for 23 hr. Then the reaction mixture was added to a cooled mixture of 190 mL 2N NaOH and 200 mL CH 2 C1 2 The aqueous phase was extracted with CH 2
C
2 (4 x 100 mL). The CH 2 C12 extracts were' combined, dried with MgSO 4 and evaporated to dryness, to afford 3.579 g of aniline 30 (87% from alkene): 'H NMR (300 MHz, CDC13, TMS) 5 6.95 J 8.3 Hz, 2H), 6.61 J 8.3 Hz, 2H), 3.60 (t, J 6.6 Hz, 2H), 2.48 J 7.6 Hz, 2H), 1.6-1.5 4H), 1.4-1.3 6H).
N-(9-Acridinyl)-4-(7-hydroxyheptyl)-aniline To a solution of 636.9 mg (3.07 mmol) of 4-(7-hydroxyheptyl)-aniline 30 and 428 pL (3.07 mmol) of Et 3 N in 20 mL of MeOH were added 656.4 mg (3.07 mmol) of 9-chloroacridine. After stirring for 7 hr at room temperature, the solvent was evaporated. Purification by column chromatography on silica gel with 5% MeOH in CHzCI 2 gave 1.079 g of N-(9-acridinyl)-4-(7-hydroxyheptyl)aniline 31: 'H NMR (300 MHz, CDC1 3 TMS) 5 8.0-7.9 4H), 7.63 J 7 Hz, 2H), 7.3-7.2 2H), 7.07 J 8.3 Hz, 2H), 6.85 J 8.3 Hz, 2H), 3.64 J 6.6 Hz, 2H), 2.57 J 7.6 Hz, 2H), 1.7-1.5 4H), 1.4-1.3 6H).
N-(9-acridinyl)-4-(7-iodoheptyl)-aniline To a solution of 604.1 mg (1.57 mmol) of
N-(
9 -acridinyl)-4-(7-hydroxyheptyl)-aniline 31 in 20 mL of pyridine cooled to 0°C was added 200 pL (2.58 mmol) of methanesulfonyl chloride. The reaction mixture was stirred at 0°C for 1 hr 20 min, then partitioned between 180 mL of CH 2 C12 and 75 mL of water.
The aqueous phase was extracted with CH 2 C2 (3 x 30 mL). The CH 2 C12 extracts were _combined, washed with 40 mL of saturated NaCI solution, dried with MgS04,Oand evaporated to dryness.
The sulfonate was dissolved in 20 mL of acetone. To the solution was added 355.0 mg (2.37 mmol) ofNaI, and the mixture was refluxed for 8 hr, then stirred at room temperature for 16hr. The reaction mixture was partitioned between 200 mL of ethyl acetate and 100 mL of water. The organic phase was washed with 5% sodium thiosulfate (3 x 30 mL). All aqueous phases were combined and backextracted with 75 mL of ethyl 46 acetate. Both ethyl acetate phases were combined, dried With MgSO 4 and evaporated to dryness, to afford 600.2 mg of N-(9-acridil)-4(7-iodohepty)aniine 3 2
NMR
(300 MHz, CDC1 3 TMS) 8 8.0-7.9 (in, 4H), 7.66 J 7 Hz, 2H), 7.3-7.2 (mn, 2H), 7.06 J =8 Hz, 2H), 6.!81 J. 8 Hz, 2H), 3.18 J =7 Hz,211), 2.5 7 J =7.6 Hz, 2H), 1.9-L.8 (in, 2H1), 1.7-1.7 (in, 2H), 1.4-1.3 (mn, 6H).
Quinone-anilifloacridifle (33) (SL-11064). To a solution of 1.554 g (3.14 mmol) of N-(9acridinyl)-4-(7-iodohepty)aniline 32 in a mixture of 40 mnL of CHC1 3 and 2 mL of MeOH was added 1.765 g (6.28 mmnol) of silver salt. The reaction mixture was refluxed for 23 hr.
The reaction mixture was diluted with CH 2 Cl 2 filtered, and evaporated to dryness.
Purification and separation of the para- and orthoquinone isomers were accomplished using a series of columns on silica gel using 5% MeOH in CHICI 2 Et 2 O, and 1.0% MeGH in
CH
2
CI
2 Isolated 108.9 mg of 33 as a dark orange solid.
N-ctl4(-ehnsloy--etnl-nln. To a cooled solution of 500 mg (2.02 mmol) of N-acetyl.4-(7-hydroxy-lI -heptenyl)-anilifle 29 and 0.5 mL (6.18 mmol) of pyridine in 10 rnL Of CH 2 Cl 2 Was added 240 1 ±L 10 minol) of methane-sulfoflyl chloride.
The reaction mixture was stirred at room temperature for 22 hr. The reaction mixture was diluted with CH 2 Cl 2 washed with IN HCI (4 x 50 mL), washed with saturated NaCI solution (50 mL), dried with MgSO4, and evaporated to drynes s. Column chromatography on silica gel with 5% MeOH in CH 2 Cl 2 afforded 416.1 mg of mesylate (mixture of E and Z isomers): 'H NMR (250. MHz, CDC1 3 TMS) 8 7.47 J3 8H4z), 7.43 I 8 Hz), 7.29 J 8 Hz), 7.22 J 8H-z), 6.4-6.3 6.2-6-0 -5.1-5.6 4.23 (t1 6.6 Hz), 4.22 1 6.6 Hz), 2.4-2.3 2.3 -2.1 2.18 2.17 1.9-1.7 1.6-1.4 (mn).
N -Acety4-(7-iodo hepteflyl) aniline To a solution of 2.641 g (8.11 inmol) of N acetyl-4-(7-metharnesulfonyl 11-heptenyl)-aniline in 60 mL of acetone was added 1.832 g.
(12.2 minol) of Nal. The reaction mixture was refluxed for 19 hr. Then, filt ration and evaporation of solvent gave 3.4 10 g (quant) of iodide 34, which was used as is in the next reaction.
Phosphonium iodide A solution of 3.410 g of N-acetyl-4-(7-iodo- 1 -heptenyl)-aniline 34 and 2.143 g (8.1 j7 mmol) of triphenyiphosphine in 70 mL of CH 3 CN was refluxed for 43 hr. Evaporation of solvent and column chromatography on silica gel with 5% MeOH in
CH
2 C1 2 yielded 4.781 g (95% from mesylate) of phosphoniurn iodide 1 -(3,4-Dimethoxy-1-naphthyl)-8-(4-acetamidophenyl)-1 ,7-octadiene To a cooled solution of 3.17 g (5.12 mmol) of phosphoniumn iodide 3.5 and 1.093 g (5.05 mmol) of 3,4dimethoxy-l-naphthaldehyde 18 in 20 rnL of TI-F and 25 ml, of CH 2
CI
2 was added 130 mg (5.14 mmol) of 95% NaH. The reaction mixture was stirred at room temperature for 21 hr. The mixture was partitioned between 200 mL IN HCl and 350 ml, CH 2 Cl 2 The aqueous phase was extracted with CH 2 Cl 2 (6 x 75 mL). The CH 2 C1 2 extracts were combined, dried with MgSO 4 and evaporated to'dryness., Cdlumn. chromatography on silica gel with 1% MeOH in CH 2
CI
2 afforded 1.073. g of diene 36.
1-(3, 4 -Dimethoxy-1-naphthyl)-8-(4-acetamidophenyl)-octane. To a solution of 556.3 mg (1 .29 mmol) of. 1 ,4-dimethoxy- 1 -naphthyl)-8-(4-acetamidophenyl)- I ,7-octadiene 36 in mnL of CH 2
CI
2 in a Parr bottle were -added 55.4 mg of 10% PdIC. The bottle was placed on a hydrogenation apparatus and shaken for 2.5 hr at 32 psi of hydrogen. Removal of catalyst by filtration through a celite pad and evaporation of solvent afforded 554.6 mg of octane: I-H NMR (250 MHz, CDCI 3 TMS) 5 8.14 J1 8 Hz, 111), 7.94 J 8 Hz, INH), 7.5-7.4 (in, IlH), 7.4-7.3 (mn, 3 7.12 (s I 7.11 J 8.2 Hz, 2H), 3.99 (s, 3 3.98 3H), 3.0-2.9 2.6-2.5 (in, 2H), 2:16 3H), 1.8-1.3 (m 12H).
1-(3,4-Diniethoxy-1-naphthyl)-8-(4-a minophenyl)-octane A solution of 554.6 mg.
(1.28 inrol) of 1-( 3 4 -diinethoxy-lI-naphthyl)-8-(4-acetamidophenyl)-octane in 20 mL of MeOH was mixed with 21 rnL of 2N HCL. The reaction mixture was refluxed for 23 hr.
Then the reaction mixture was partitioned between 75 mL of CH 2 Cl 2 and 21 mL of 2N NaOH. The aqueous phase was extracted with CH 2
CI
2 (5 x 40 mL). The CH1 2 Cl 2 extracts *were combined, dried with MgSO 4 and evaporated to dryne ss. Column chromatography on silica gel with 1% MeOH in CH 2
CI
2 gave 374.6 mg of aniline 37: 'HNMR (250 MHz, CDC1 3 TMS) 8 8.14 J 8 Hz, IN), 7.94 J 8 Hz, 1H), 7.47 J 8 Hz, IH), 7.3 7 J 8 8Hz, I 7.12 INH), 6.96 J 8 Hz, 2H), 6.62 J 8 8Hz, 2H), 3.99 (s, 48 3H), 3.97 3K), 3.1-3.0 (in, 2H),'2.5-2.4 (in, 2H), 1.8-1.3 (m 12H).
Naphthylacridine To a solution of 99 mng (2.5"x10 mol) of l-(3,4-dimethoxy-
I-
naphthyl)-8-(4-amTiflophelyl)-octane 37 and 35 mL (2.5 1x10 4 rnol) of Et 3 N in 4 m.L of MeOH were added 54 mg (2.53xl10 4 mol) of 9-chioroacridine. The reaction mixture was stirred at room temperature for 20 hr.;Evaporation of solvent and column chromatography on silica gel with first I MeOH in CH 2
CI
2 and then 3%O/ MeGH in CH 2 C1 2 afforded 118.2 C1 mg of acridine 38: 'H NMR (250 MHz, CDCI 3 TMS) 8 8.14 J =8 Hz, 1H), 7.9 5H), 7.66 (br t, 2H), 7.46 J 8 Hz, I 7.3 7 J 8 Hz, IlH), 7.3 -7.2 (in, 2H), ri 10 7.12 I 7.06 J =8.4 Hz, 2H), 6.82 J 8.4 Hz, 2H), 3.1 -3.0 (in, 2H), 2.6-2.5 (in, (mn, 12H).
Quinone-acridine. (39) (SL-11125). To a solution of 546 mg (9.60xI0 4 mol) of acridine 38 in 15 mnL Of CH 2
CI
2 cooled to -681C was added 9.6 mL of IlM BBr 3 in CH 2
GI
2 After 18.5 hr at -10 0 C, the reaction mixture was cooled to -68"C and 10 mL of Et 2 O were added.
After stirring at room temperature for 30 min, 20 mL of saturated NaHCO 3 Solution were added. The resulting precipitate was collected by fil tration and triturated twice with 50 mL of .CH 2 Cl 2 to give 555.9 mng of quinone 39: 'H NMR (250 MHz, DMSO-d 6 TMS) 5 9.11 8.59 8.14 J =9 Hz), 8.0-7.9 7.82 J =8 Hz), 7.4-7.2 6.98 2.87 (t, J 7 Hz), 2.65 J 7 Hz), 1.7-1.5 1.4-1.3 (mn).
N-(9-acridyI)-mesityleflesulfoflamide To a suspension of 4.00 g (20.6 inmol) of 9aminoacridine 40 in 350 mL of CHC1 3 was added 2.9 mL (20.8 mmol) of Et 3 N and 4.50 g (2.6imol) of mesitylenesulfonyl chloride. The reaction mixture was refluxed for 72 hr.
Then the reaction mixture was filtered and the solvent was evaporated. The material was purified by columrn chromatography on silica gel by eluting first with 1% MeOH in CK 2
CI
2 and then with '5 MeOK in CH 2 CI, to yield 458.4 mg of sulfonamide 41 as an orange solid: 'H NMR (300 MHz, CDCI 3 TMS) 8 9.25 I 8.77 J 8 Hz, 2H), 7.46 J 8 Hz, 2H), 7.21 J 8 Hz, 2H), 7.15 J 8 Hz, 2H), 7.02 2K), 2.78 6H), 2.36 (s, 3K).
N-9aciy)N(- ompeny)meiylnsloaid A solution of 450 mg 49 (1.20 mmol) of N-(9-acridyl)-mesitylenesulfonamide in 20 mL of DMF was placed under an atmosphere of argon and cooled to 0°C. To the cooled solution was added 36 mg (1.42 mmol) of NaH The reaction mixture was stirred at 0°C for 5 min and at room temperature for 1 hr. Then the reaction mixture was cooled to 0°C, and 1.65 mL (12.1 mmol) of 1,5-dibromopentane were added. The reaction mixture was stirred at 70-80 0 C for 23 hr. The reaction mixture was cooled, and quenched with 20 mL of water. The mixture was partitioned between CH 2 C12 and water. The aqueous phase was washed with CH 2
CI
2 (2 x 20 mL). The CH 2 Cl 2 washes were combined with the organic phase, dried with MgSO 4 and evaporated to dryness. The material was purified by column chromatography on silica gel with CH 2 C1 2 to afford 382.2 mg of bromide 42 as an orange oil: 'H NMR (300 MHz, CDC13, TMS) 6 8.25 J 9 Hz, 2H), 7.94 J 9 Hz, 2H), 7.76 J 8 Hz, 2H), 7.45 J 8 Hz, 2H), 6.87 2H), 4.0-3.9 2H), 3.27 J 6.5 Hz, 2H), 2.30 3H), 2.22 6H), 1.8-1.6 4H), 1.4-1.3 2H).
Mesityl-acridine-quinone To a solution of 632.6 mg (1.20 mmol) of N-(9-acridyl)- 42 in 15 mL of benzene was added 338.4 mg (1.20 mmol) of silver salt. The reaction mixture was refluxed for 24 hr. The reaction mixture was diluted with CH 2 C12 and filtered to remove insoluble salts. The solvent was removed and the material was purified by column chromatography on silica gel with to afford 333.1 mg of ortho-quinone 43 as an orange glassy solid: 'H NMR (300 MHz, CDC1 3 TMS) 5 8.24 J 9 Hz, 2H), 8.11 J 8 Hz, 1H), 7.95 J 9 Hz, 2H), 7.8-7.7 3H), 7.7-7.5 2H), 7.5-7.4 2H), 6.86 2H), 5.85 1H), 4.1-4.0 (m, 4H), 2.29 3H), 2.21 6H), 1.9-1.5 4H), 1.5-1.4 2H).
Acridine-quinone (44) (SL-11059). Under an atmosphere of argon, 151.4 mg (2.45x10 4 mol) of mesityl-acridine-quinone 43 was dissolved in 30 mL of 0.1M SmI 2 in THF. Then, 2.2 mL (18.2 mmol) of DMPU were added dropwise. The reaction mixture was refluxed for 24 hr. Filtration to remove a precipitate and evaporation of solvent yielded an orange oil, which was purified by column chromatography on silica gel with 5% MeOH in CH 2 Cl 2 to afford 48.7 mg of acridine-quinone 44 as an orange glassy solid: 'H NMR (300 MHz, DMSO-d 6 TMS) 8 8.54 J 8 Hz, 2H), 7.96 J 7 Hz, 2H), 7.92 J 7 Hz, 114), 7.79 J 8 Hz, 2H), 7.7-7.6 (in, 3H), 7.51 J3 8 Hz, 2H), 6.01 1H), 4.20 J 6 Hz, 2H), 4.13 J 7Hz, 2.1-1.9 (in, 4H), 1. 7-1.6 (mn, 2H).
Synthesis of quinol phosphates: General Procedure To a solution of 500 mg (2.05 inmol) of 4-pentyloxy-l,2-flaphthoquilofe 46 in inL of benzene was added 2.3 mL (25.1 mmol) of dibenzylphosphite. The reaction mixture CI was refluxed under nitrogen for 2.5 hr, after which the benzene was removed. Column chromatography of the residue on silica gel with 1% MeOI- in CH 2 Cl 2 afforded 729.3 mrg of aryldibenzylphosphate 47 (mixture of two regioisomers) as an orange oil: Rf 0.51, 0.66 (1%.MeOH, in CH 2
CI
2 1H NMR-(25,0 MlHz, CDCl 3 TMS) mhajor 8.1 8.0 (br, 7.8 7.3-7.1 6.50 5.3-5.0 (AB of ABX, 8A 5. 16, 8B 5.08, JAB =11.-5 HZ, JAX =8.3 Hz, JBX 8.8 Hz), 4.01 J =6 Hz), 2.0-1. 8 (n,1.6-1.3 0.96 J 7 Hz); 1 3 C NMR (52 MHz, .CDCI 3 TMS) both regioisomers 8 153.4, 144.7, 135.6 (di, J 6.1'Hz, minor regioisomer), 134.8 J3 5.5 Hz, major regioisomer)., 128.7- 127.7 127.2, 123 122.2, 121.4, 119.8, 99.5, 7 1.0 J 4.8 Hz), 68.3 28.8, 22.5.
To a solution of 1.637 g (3.23 inmol) of aryldibenzylphosphate 47 in 40 mL of MeOH was added 150 mg of 10% Pd.C. The reaction mixture was placed under an atmosphere of hydrogen (balloon) and stirred at room temperature for 1 hr. Removal of catalyst by filtration and evaporation of solvent afforded phosphate as a brown oil. The phosphate was dissolved in 6 mL of benzene. Addition of 9 mL of hexane and cooling gave a precipitate. The, precipitate was collected by filtration, washed with benzene/hexane and dried, affording 797.3 mg of Arylphosphate 48 as a gray solid; R 1 0.77 (MeOH); 'H NMR (250 MHz. acetone-d6, TMS) 5 8.13 (di, J 8- Hz, 1H), 7.96 J 8Hz, 7.49 J3 7 Hz, I 7.3 J 7 Hz, I 6.5 9 IlH), 4.1 3 3J 6Hz, I 1.8 (in, 2H),l1.6-.3 (in, 4H), 0.96 J 7 Hz, 3H); 1 3 C NMR (52 MHz, acetone-d 6
TMS)
153.3 J1 1.3 Hz), 145.8 (narrow 129.3 (di, J Hz), 127.4, 123.2, 122.2, 121.6, 120.9, 100.0, 68.7, 29.2, 28.7, 22.7, 13.9.
Ethyl 2?-acetyl-5'-rnethoxyphenylacetate (50) Acetyl chloride (21.3 mL, 300 inmol) was added to a mixture of AIC1 3 (26.7 g, 200 minol) and ethyl 3'-methoxyphenylacetate (49, 28.66 g, 147.6 inmol) in CS 2 (200 mL) at 0 The ice bath was removed and the mixture 51 1 was allowed to warm to 20 'C with HC1 gas bubbling out. After stirring at 20 'C for min, the mixture was refluxed for 30 min. Upon cooling down, the mixture was added ice (200 g) and aqueous 2 N HC1 (400 mL). The resulting mixture was extracted with ethyl acetate (2 x 200 mL). The extracts were washed with water (2 x 100 mL), dried over MgSO 4 and concentrated in vacuo. The residue was crystallized from a mixture of ethyl acetate (20 mL) and hexanes (60 mL) to afford 50 30.60 g, 'H NMR (CDCl 3 7.84 (1H, d, J= 8.6 Hz), 6.86 (1H, dd, J= 8.6, 2.6 Hz), 6.75 (1H, d, J= 2.6 Hz), 4.17 (2H, q, J= 7.1 Hz), 3.92 (2H, 3.86 (3H, 2.55 (3H, 1.28 (3H, t, J= 7.1 Hz); "C NMR (CDC13) 5 199.04 171.44 162.22 137.70 132.97 129.48 118.68 111.84 60.60 55.39 41.17 28.39 14.24 2 -Hydroxy-7-methoxy-1,4-naphthoquinone(51). Sodium ethoxide (10.40 g, 150 mmol) was added to a suspension of 50 30. 45 g, 128.90 mmol) in absolute alcohol (200 mL) at After stirring the mixture for 1 h, air was bubbled in for 20 h. The mixture was concentrated in vacuo. The residue was dissolved in water 500 mL), and extracted with diethyl ether (200 mL). The ether layer was counter-extracted with water (50 mL). The combined aqueous phase was acidified with concentrated HCI (30 mL). The mixture was filtered to afford 51 (14.42 g, 'H NMR (DMSO-d6) 5 11.56 (1H, s, br), 7.89 (1H, d, J= 8.5 Hz), 7.42 (1H, d, J= 2.8 Hz), 7.36 (1H, dd, J= 8.5, 2.8 Hz), 6.10 (1H, 3.92 (3H, "C NMR (DMSO-d6) 5.184.07 181.20 162.92 159.16 132.35 127.82 125.16 120.02 110.85 109.94 55.90 7- Methoxy-lapachol A mixture of K 2
CO
3 (30 mmol) and 51 (10.21 g, 50 mmol) in HMPA (100 mL) was stirred for 30 min, when it became a suspension. Dimethylallyl bromide (8.7 mL, 75 mmol) and KI (4.15 g, 25 mmol) were added, and stirring was continued for 20 h at 20 The mixture was diluted with ice water (600 mL) and concentrated HCI (30 mL), and extracted with ethyl acetate (2 x 200 mL). Some solid was collected by filtration to afford the first portion of 53 (0.628 'H NMR (CDCI 3 6 8.01 (1H, d, J= 8.6 Hz), 7.56 (1H, d, J= 2.7 Hz), 7.20 (lH, dd, J= 8.6, 2.7 Hz), 6.09 (1H, s), 5.49 (1H, t, J= 6.8 Hz), 4.57 (2H, d, J= 6.8 Hz), 3.94 (3H, 1.81 (3H, 1.76 (3H, S).
The ethyl acetate extracts were pooled, extracted with saturated NaHCO 3 (2 x 150 mL), and the resultant aqueous extracts were acidified with concentrated HC1 and filtered to recover 51 (2.10 g, 21%).
The main ethyl acetate extract was concentrated in vacuo. The residue was dissolved in a mixture of 1 N NaOH (500 mL) and diethyl ether (300 mL). After separation, the organic layer was extracted with 1 N NaOH (100 mL) and concentrated in vacuo. The residue was chromatographed on silica gel (10% ethyl acetate in hexanes) to afford a second portion of 53 (3.43 g, 30% total).
The NaOH extracts were acidified by concentrated HCI (50 mL), and extracted with ethyl acetate (2 x 200 mL). The pooled extracts were dried (MgSO4), concentrated in vacuo, and the residue was purified by chromatography on silica gel (10% ethyl acetate in hexanes) to afford 52 (4.39 g, 'H NMR (CDCl3) 5 8.05 (1H, d,J= 8.6 Hz), 7.51 (1H, d, J=2.7 Hz), 7.20 (1H, dd, J= 8.6, 2.7 Hz), 7.18 (OH, 5.20 (1H, tt, J= 6.7, Hz), 3.93 (3H, 3.29 (2H, d, J= 7.2 Hz), 1.79 (3H, 1.68 (3H, 'C NMR
(CDCI
3 5 183.99 181.85 163.28 152.51 133.71 131.18 129.04 126.23 123.28 120.69 119.82 109.82 55.89 25.77 22.60 17.90 8-Methoxy-P-lapachone (54) Concentrated HzS0 4 (25 mL) was added to compound 52 (2.454 g) at 20 After stirring for 20 min, the mixture was diluted with ice water (500 mL). The resulting red precipitate 54 was collected by filtration, washed with water, and dried in vacuo. It was obtained as a red powder (2.36 g, 'H NMR (CDC13) 8 7.72 (1H, d, J= 8.6 Hz), 7.56 (1H, d, J= 2.7 Hz), 7.12 (lH, dd, J= 8.6, 2.7 Hz), 3.90 (3H, S), 2.55 (2H, t, J= 6.7 Hz), 1.84 (2H, t, J= 6.7 Hz), 1.46 (6H, S).
8-Hydroxy--lapachone (55) Boron tribromide (15.0 mL, 1.0 M in CH 2 Cl 2 was added to a solitifbh 6T54 (1.05 g, mmol) in anhydrous CH 2 Cl 2 (40 mL) at 0 After stirring for min, the mixture was allowed to warm to 20 'C and kept stirring for 2 h. Ice water (500 mL) was added, the mixture was extracted with CHC13 (3 x 100 mL), the combined extracts were dried, and concentrated in vacuo. The residue was treated with concentrated H 2 S0 4 (20 mL) at 20 The mixture was diluted with ice water (500 mL) and extracted with CHC13 3 x 100 mL). The combined extracts were reextracted with aqueous 5% NaHSO 3 3 x 150 mL). The aqueous extracts were acidified with concentrated HCI (100 mL), and 53 1 extracted with CHC3 (3 x 150 mL). The extracts were dried and concentrated to afford (270 mg, 'H NMR (CDCI 3 8 9.81 (OH, 7.64 (1H, d, J= 8.5 Hz), 7.49 (1H, d, J= 2.6 Hz), 7.06 (1H, dd, J= 8.5, 2.6 Hz), 2.51 (2H, t, J= 6.6 Hz), 1.84 (2H, t, J= 6.6 Hz), 1.45 (6H, HRMS calcd for C, 5 Hi40 4 258.0892, found 258.0885.
Preparation of 1,2-Naphthoquinone bisulfite adducts General Procedure I. The quinone was dissolved in 10% NaHSO 3 After standing for several hours at room temperature or with cooling, the quinone-bisulfite adduct precipitated. The quinone-bisulfite was collected by filtration and dried. The quinonebisulfite was stablized with addition of 300 its weight of sodium bisulfite.
General Procedure II. The quinone is dissolved in 10% NaHSO 3 in a volume of solution such that there is no more than 300% weight excess of NaHSO 3 (relative to quinone-bisulfite). When the quinone-bisulfite did not precipitate, it was recovered from the solution by evaporation of the water in vacuo. This procedure gives a quinone-bisulfite adduct with a 300% weight excess NaHSO3.
Synthesis of morpholino-Ser-Lys-Leu-Gln-p-Ala-p-Lapachone (Scheme 13) Boc-Gln-p-Ala-P-Lapachone To a solution of 1.000 g (2.437 mmol) of P-Ala-p-Lapachone-TFA salt (SL-11006) and 6 00.3 mg (2.437 mmol) of Boc-Gln in 10 mL of DMF was added 395.3 mg (2.925 mmol) of 1-hydroxybenzotriazole. The mixture was cooled in an ice bath. Then 270 LL C (2.456 mmol) of N-methylmorpholine were added, followed by 553.0 mg (2.680 mmol) of DCC. The reaction mixture was stirred in the ice bath for 30 min and at room temperature for 6.5 hr. The reaction mixture was then diluted with CH 2 C1 2 and filtered. The filtrate was washed with saturated NaHCO 3 (50 mL), with 5% citric acid (3 x 50 mL), witihsaturated NaHCO 3 (2 x 50 mL), with saturated NaCl (50 mL), dried with MgSO 4 and evaporated to dryness. Purification by column chromatography on silica gel with 5% MeOH in CH 2 C2 afforded 692.7 mg of peptide as an orange glassy solid: R/ 0.11 MeOH in
CH
2 C1 2 1 H NMR (250 MHz, acetone-ds, TMS) 5 8.00 (dd, J 7.6, 1.3 Hz, 1H), 7.9-7.7 2H), 7.64 (td, J 7.6, 1.3-Hz, 1H), 7.5-7.4 (br d, NH), 6.9 (br s, NH), 6.2 (br s, NH), 5.2-5.1 1H), 4.1-4.0 1H), 3.5-3.4 2H), 2.7-2.5, 4H), 2.3-2.2 2H), 54
I
1.8 (mn, 2H), 1.53 3H), 1.51 3H), 1.39 9H); NMR (52 MI-z, acetone-d 6
TMS)
8 179.8, 178.8, 175.0, 172.5, 171.6, 160.8, 156.2,1111.1, 135.6, 133.0, 131.6, 131.2, 128.7, 124.8, 80.8, 80.3, 79.2, 70.2, 54.8, 35.6, 34.7, 32.1, 28.4, 24.8, 23.2, 23.1.
Gln-P-Ala-f3-Lap ach one To a solution of 681.9 mg (1.223 mmol) of Boc-Gln-p-Ala-p-Lapachone in 10 mL of CH 2
CI
2 was added 10 mL of TEA. The reaction mixture was- stirred at room temperature for 25-30 min. The solvent was removed in vacuo. Column chromatography on silica gel r) with 10-20% MeOH in CH 2 Cl 2 afforded 578.5 mg of the TEA salt as an orange A~ 10 glassy solid: Rf 0.55 (BuOH/H 2 0/AcOH 0.05 (10% MeOH in CH 2 Cl 2 0.24 MeOH in. CH 2
CI
2 Boc-Leu-Gln-P-Ala-P3-Lapachone To a solution of 650.2 mng 13 8 mmol) of Gln-p-Ala-f3-Lapachone-TFA salt and 263.0mg (1.138 mmol) of Boc-Leu in 4.6 mLof DMF was added 184.5 mg (1.3'65 mmol) of I -hydroxybenzotriazole. The mixture was cooled in an ice, bath. Then 130 .tL 182 mmol) of N-methylmorpholine Were added, followed by 258.4 mg (1.252 mmol) of DCQ.
The reaction mixture was stirred. in the ice bath for 30 min and at room temperature for hr. The reaction mixture was then diluted with CH 2
CI
2 and filtered. The, filtrate was washed With saturated NaHCO 3 (30 mL), with 5% citric acid (4 x.30 mL) with saturated NaHCO 3 (3 x 30 mL), with saturated NaCI. (3 0 mL), dried with MgSO 4 and evaporated to dryness.
Purification by. column chromatography on. silica gel with 5% MeOH in CHCl 2 afforded 396.9mgn (5 of peptide as a yellow-orange glassy solid: Rf 0. 11 MeOH in
CH
2
CI
2 0.45 MeGH in CH 2 Cl 2 0.81 (20% MeOH in CH 2
CI
2 0.78 (BuOHIH 2 O/AcOH 'H NMR (250 MHz, acetone-cl 6 TMS) 6 8.00 J 7.5 Hz, I1H), 7.9-7.174in, 2H), 7.64 J 7.5 Hz, I1H), 7.5 (br d, NH), 6.9 (bn s, NH), 6.3 (bn s, NH), 5.2-5.1 (in, 1H), 4.4-4.2 (in, IH), 4.1-4.0 (in, 1K), 3.6-3.3 (mn, 2H), 2.7-2.5 (in, 4K), 2.3-2.2 (mn, 2H), 2.0-1.8 (mn, 2H), 1.8-1.7 (in, IH), 1.6-1.5 (in; 2H), 1.53 3H), 1.51 3H), .1.39 9H), 1.0-0.9 (mn, 6H4); 3 C NMR (52 MHz, acetone-cl 6 TMS) 8 179.9, 179.0, 175.2, 173.4, 172.0, 171.5, 160.9, 156.8, 13 5.7, 133.1, 131.6,131.2, 128.8, 124.9, 111.2, 80.9, 80.4, 79.5, 70.3, 54.5, 53.5, 41.7, 35.8, 34.8, 32.1, 28.5, 27.8, 25.4,24.9, 23.4, 23.2, 21.9.
I
Leu-Gln-p-Ala-p-Lapachone To a solution of 317.0 mg (4.725x10 4 mol) of Boc-Leu-Gln-P-Ala--Lapachone in 4 mL of CH 2 C1 2 was added 4 mL of TFA. The reaction mixture was stirred at room temperature for 25-30 min. The solvent was removed in vacuo. Column chromatography on silica gel with 20% MeOH in CH 2 C2 afforded 277.3 mg of the TFA salt as an orange glassy solid: R 0.17 (10% MeOH in CH 2 C12), 0.39 (20% MeOH in CH 2 C2), 0.74 (BuOH/H 2 0/AcOH= 5:3:2).
Na-Boc-Lys(NE-Cbz)-Leu-Gln-P-Ala-3-Lapachone To a solution of 277.3 mg (4.050x 104 mol) of Leu-Gln-P-Ala--Lapachone-TFA salt and 1 68.0 mg (4.049x10 4 mol) of Na-Boc-Lys(Ne-Cbz) in 1.6 mL of DMF was added 65.7 mg (4.862x 10 mol) of 1-hydroxybenzotriazole. The mixture was cooled in an ice bath. Then 50 gL (4.548x10 4 mol) of N-methylmorpholine were added, followed by 91.9 mg (4.454x104 mol) of DCC. The reaction mixture was stirred in the ice bath for 30 min and at room temperature for 6.5 hr. The reaction mixture was then diluted with 2 mL of CHC1 3 and filtered. The filtrate was washed with saturated NaHCO 3 (20 mL), with citric acid (4 x 20 mL), with saturated NaHCO 3 (3 x 20 mL), with saturated NaCI (2 x mL), dried with MgSO4, and evaporated to dryness. Purification by column chromatography on silica gel with 10% MeOH in CH 2 C2 afforded 167.5 mg of peptide as anorange glassy solid: R 0.08 MeOH in CH 2 Cl 2 0.44 (10% MeOH in
CH
2
CI
2 'H NMR (250 MHz, DMSO-d 6 TMS) 6 8.0-7.7 6H, quinone-H5, H6, H7, H8, NH's), 7.7-7.6 NH), 7.5-7.4 2H, CI-Cbz), 7.4-7.3 2H, Cl-Cbz), 7.20 (br s, NH), 6.73 (br s, NH), 6.90 (br d, J 7.9 Hz, NH), 5.07 3H), 4.3-4.2 1H), 4.2-4.1 1H), 3.9-3.8 1H), 3.3-3.2 2H), 3.0-2.9 2H), 2.8-2.7 2H), 2.6-2.4 (m, 2H), 2.1-2.0 2H), 1.8-1.3 11H), 1.43 3H), 1.39 3H), 1.36 9H), 0.85 J= Hz, 3H), 0.81 J= 6.6 Hz, 3H); 'C NMR (52 MHz, DMSO-d 6 TMS) 8 178.6, 177.8, 173.5, 173.4, 172.0, 171.7, 171.0, 170.5, 162.2, 155.7, 134.9, 134.5, 132.2, 131.4, 130.9, 129.9, 129.5, 129.2, 127.9, 127.2, 123.7, 79.7, 79.3, 78.0, 68.9, 62.4, 54.2, 52.0, 50.8, 40.7, 35.7, 33.5, 31.2, 30.7, 29.0, 28.1, 27.8, 24.1, 23.9, 23.0, 22.8, 22.7.22.1,21.4.
Lys(NF,-Cbz)-Leu-Gin-jp-Aa-p3-Lapachone To a suspension of 203.1 mng (2.099x10- mol) of Boc-Lys(NE-Cbz)-Leu-G~n-D3- Ala-f3-Lapachone in 2 rnL of CHC1 3 was added 1.7 mL of TEA (material dissolved). The reaction mixture was stirred at room temperature for 20-25 min. The solvent was removed in vacuo. Column chromatography on silica gel with 20% MeGH in CH 2
CI
2 afforded 202.0 mg of the TEA salt as an orange glassy solid: Rf 0.10 (10% MeGH in
CH
2 Cl 2 0.40 (20% MeGH in CH 2 Cl 2
DK
D Morpholino-Ser(OBn)-Lys(Ns.-Cbz)-Leu-Gn-3-Ala-f3-Lapachone
D-
To a solution of 194.8 mg (1.985x 10- mol) of Lys(Nc-Cbz)-Leu-Gln-P-Ala-P- Lapachone-TFA salt and 61.2 mg (1 .985x 10- mol) of morpholino-Ser(OBn) in 1.0 mL of DMF was added 32.2 mg (2.383)x10- mot) of 1-hydroxybenzotriazole. The mixture. was cooled in an ice bath. Then 23 jiL (2.092x 10-4' mol) of N-methylmorpholine were added, followed by 45.1 mg (2.186x1 0 4 Mol) of DCC. The reaction mixture was stirred in the ice bath for 3 5 min and at room temperature for 6 hr. The reaction mixture was then diluted with 2 mL of CH 2
CI
2 and filtered. The filtrate was washed with. 5% citric acid (3 x 20 mL), with saturated NaHCO 3 (3 x 20 mL), with saturated NaCI (20 mL), dried with MgS0 4 and evaporated to dryness. Purification by column chromatography on silica gel with MeOH in CHICl 2 afforded 83.3 mg of peptide as an orange glassy solid: Rj 0.05 MeOHJ in CH1 2
C
2 0.41 (10% MeOH in CH 2
CI
2 'H NMR (250 MHz, acetone-d 6 TMS) 5 8.0-7.7 7H, quinone-HS, H6, H7, H8, NH's), 7.7-7.6 (mn, NH), 7.5-7.2 (in.
lOH, Cl.-Cbz, OBn, NH), 6.75 (br s, 6.60 (br d, J =7.1 Hz, NH), 5.07 3H), 4.49 (s, 2H), 4.4-4.3 (in, IH), 4.3-4.0 (mn, 3H), 3.7-3.6.(in, 2H), 3.6-3.5 (mn, 4H), 3.3-3.2 (in, 6H), 3.0-2.9 (in, 2H4), 2.8-2.7 (m,214), 2.5-2.4 (mn, 2H), 2.1-2.0 (mn, 2H4), 1.8-1.3 (in, 1 1H), 1.43 3H), 1.38 3H), 0.82 J =6.0 Hz, 3H), 0.78 J =6.1 Hz, 3H).
Morpholino-Ser-Lys-Leu-Gln-3-Ala-f3-Lapachone (SL-1 1147) To a solution of 78.3 mg (6.763x10 5 iol) of inorpholino-Ser(OBn)-Lys(NE-Cbz)- Leu-Gln-3-Ala-p-Lapachone in 1.5 mL of MeOWVCH 2
CI
2 1:9 was added 30.6 mg PdIC. Then 0.5 int of MeOH and one drop of HC were added. The reaction mixture was p laced under an atmosphere of H 2 (balloon) and stirred at room temperature for 16 hr.
Removal of catalyst by filtration and evaporation of solvent afforded 64.5 mg of crude quinone-tetrapeptide. The material was purified by prep HPLC to yield 14.4 mg R 0.04 (20% MeOH in CH 2 C1 2 N-Fmoc-Ser(OBn) t-butyl ester Isobutylene was condensed into a 500 mL pressure bottle until the volume was between 30 and 40 mL. A solution of 3.02 g (7.23 mmol) of N-Fmoc-Ser(OBn) in 20 mL of THF was added, followed by 2 mL of concentrated H 2 S0 4 The bottle was securely stoppered arid shaken at room temperature for 24 hr. The reaction mixture was poured into an ice-cold mixture of 150 mL of ethyl acetate and 150 mL of saturated NaHCO 3 The organic phase was washed with water (3 x 50 mL) and dried with MgSO 4 The solvent was removed, and column chromatography on silica gel with CH 2 C2 afforded 2.453 g of t-butyl esteras a colorless oil: 'H NMR (250 MHz, acetone-ds, TMS) 8 7.85 J= 7.5 Hz, 2H), 7.74 J 7.3 Hz, 2H), 7.5-7.3 9H), 6.71 (br d, J 8:6 Hz, NH), 4.55 (ABq, 8 A 4.57, 58 4.52, JAB 12 Hz, 2H), 4.4-4.2 4H), 3.9-3.7 (AB of ABX, 6 A 3.89, 8 B 3.75, JAB 9.5 Hz, JAX 4.6 Hz, JBX 3.6 Hz, 2H); 3 C NMR (52 MHz, acetone-d 6
TMS)
8 170.0, 156.8, 145.0, 144.9, 142.0, 129.0, 128.4, 128.3, 128.2, 127.8, 126.1, 120.7, 81.9, 73.6, 70.9, 67.3, 55.9, 47.9, 28.1.
Ser(OBn) t-butyl ester To a solution of 3.049 g (6.44 mmol) of N-Fmoc-Ser(OBn) t-butyl ester in 50 mL of CH 2
CL
2 was added 3 mL of piperidine. The reaction mixture was stirred at room temperature for 2.3 hr. Removal of solvent and column chromatography on silica gel with MeOH in CH 2 C2 yielded 1.306 g of Ser(OBn) t-butyl ester as a colorless oil: Rf 0.12 MeOH in CH 2
C
2 'H NMR (250 MHz, acetone-ds, TMS) 8 7.4-7.2 4.53 (Abq, SA 4.55, 8 4.52, JAB 12 Hz, 2H), 3.7-3.6 AB of ABX A 3.68, 8 3.61, JAB 12 Hz, JAX 4.9 Hz, JBX 4.4 Hz, 2H), 3.5-3.4 X of ABX, 8 x 3.45, 1H), 1.43 9H); 3 C NMR (52 MHz, acetone-d 6 TMS) 8 173.9, 139.5, 128.9, 128.2, 128.1, 80.7, 73.8, 735, 56.2, 28.1.
Morpholino-Ser(OBn) t-butyl ester To a solution of 140.6 mg (5.59x0O-' mol) of Ser(OBn)-t-butyl ester in 4 mL of pyridine was added 66 p.L (5.66xl 0 4 mol) of 4-morpholinecarbonyl chloride. After stirring for I hr, the reaction mixture was partitioned between 75 mL Of CH 2 C1 2 and 60 mL of water. The organic phase was washed with saturated NaHCO 3 (50 mL), with INKHC (2 x rnL), with saturated NaCi (50 mL), dried with MgSO 4 and evaporated to dryness. The c rude amide was purified by column chromatography on silica gel with ethyl acetate to yield 80.9 mg of amide as a light orange oil: Rf 0.58 (ethyl acetate), 0.60 MeGH in CH 2
CI
2 'H NMR (250 MHz, acetone-d 6 TMS) 5 7.4-7.2 (in, 514), 5.8 (br d, NH4), 4.53 (Abq, 5 A =4.55, 6B 4.52, JAB 12 Hz, 214), 4.5-4.4 (in, X of ABX, 6x =4.47, 111), 3.9-3.6 (in, AB of ABX, 8A =3.86, 8B 3.69, JAB =9.4 Hz. JAX= 4.4 Hz, JBX 3.7 Hz, 2H), 3.63-3.58 (in, 414), 3.4-33 (in, 4H), 1.44. 914); 1 3 C NMR (52 MHz, acetone-d 6 TMS) 5 170.9, 157.9,139.2, 129.0, 128.3, 128.2, 81.5, 73.5, 7.1.3, 67.0, 55.5, 44.9, 28.1.
Morpholino-Ser(OBn) A solution of 80 mg (2..195xl 04 mol) of morpholino-Sor(OBn) t-butyl ester in a mixture of 1.5 mL of CH 2 Cl 2 and 1.5 m L of TFA was stirred at room temperature for min. The solvent was removed in vacuo and the remaining TFA was removed by repeated evaporation with acetone. The residue was triturated with Et 2 O. The mnaterial was then filtered, washed with Dt 2 O, washed with 0.5 mL acetone,. washed again with EtO, and dried to yield 41.8 mg of amino acid as an off-white solid: Rf 0.72 (BuOH/H 2 0/AcOH NMR (250.MHz, acetone-d 6 TMS) 8 7.4-7.3 (in, 5H), 5.9 (br d, NH), 4.6-4.5 (in, 3H, OCH 2 Ph X of ABX), 3.95-3.75 (in, AB of ABX, 6A= 3.90, 813 3.73, JAB =9.6 Hz, JAX 4.9 HZ, JBX 3.9 Hz, 214), 3.6-3.5 (mn, 414), 3.4-3.3 (in, C. NMR (5 2 MHz, DMSO-46, TMS) d 172.4, 157.2, 13 8.2, 128.2, 127.4, 127.4, 72.0, 69.5, 53 43.9.
Svnthesis of Morpholino-Ser-Lvs-Leu-Gln-Leu-B-Lapachone (Scheme 14) Boc-Leu-P3-Lapachone A solution of 2.820 g-(12.20 inmol) of Boc-Leu and 1.976 g (12.19 inmol) of 1,1carbonyldiiinidazole in 33 mL of DMF was stirred at room temperature for 20 min. To the solution was added 2.100 g (8.130 inmol) of 3-hydroxy-J3-lapachone followed by 1.6 mL (10.70 mmol) of DBU. After stirring at room temperature for 5 hr, the reaction mixture was partitioned between 200 mL of water and 200 mL of CHC13. The aqueous phase was washed with CHCI 3 (4 x 50 mL). The CHC13 extracts were combined, dried with MgSO 4 and evaporated to dryness. Column chromatography on silica gel with 2% MeOH in
CH
2 C1 2 afforded 2.038 g of quinone as an orange glassy solid (and mixture of two diastereomers): R 0.45 MeOH in CH 2 Cl 2 'H NMR (250 MHz, acetone-d 6 TMS) 8.1-8.0 1H), 8.0-7.9 1H), 7.9-7.8 1H), 7.7-7.6 1H), 6.34 (br d, NH), 5.2-5.1 1H), 4.2-4.1 1H), 2.9-2.8 1H), 2.7-2.5 1H), 1.8-1.6 3H), 1.56 1.53 3H), 1.52 1.5H), 1.34 4.5H), 1.33 4.5H), 0.91 J 7.0 Hz, 1.5H), 0.88 J 6.7 Hz, 1.5H), 0.84 J =6.3 Hz, 1.5H), 0.82 J 6.1 Hz, Leu-p-Lapachone To a solution of 2.017 mg (4.277 mmol) of Boc-Leu--Lapachone in 20 mL of
CH
2 C2 was added 20 mL of TFA. The reaction mixture was stirred at room temperature for 30 min. The solvent was removed in vacuo. Column chromatography on silica gel with 20% MeOH in CH 2 Cl 2 afforded 2.507 g (quant.) of the TFA salt as an orange glassy solid: Rf 0.52 (10% MeOH in CH 2 C12), 0.82 (20% MeOH in CH 2 C2); 'H NMR (250 MHz, DMSO-d 6 TMS) 5 8.6-8.5 (br s, NH), 8.0-7.9 1H), 7.9-7.8 2H), 7.7-7.6 1H), 5.3-5.2 1H), 4.1-4.0 1H), 2.8-2.5 2H), 1.8-1.5 3H), 1.52 1.5H), 1.49 (s, 1.43 3H), 0.83 J 6.0 Hz, 3H), 0.66 (brt, 3H); "C NMR (52 MHz, DMSOdo, TMS) 8 178.7, 177.8, 169.2, 169.1, 160.0, 159.7, 135.1, 135.1, 131.5, 131.4, 131.1, 131.0, 129.8, 129.8, 127.9, 123.9, 123.8, 109.6, 109.3, 79.4, 79.1, 71.1, 70.9, 50.6, 50.4, 39.0, 24.0, 23.9,22.9, 22.3, 22.1, 22.0, 21.8, 21.7, 21.1.
Boc-Gln-Leu-p-Lapachone To a solution of 2.235 g (3.895 mmol) of Leu-p-Lapachone-TFA salt and 959.1 mg (3.894 mmol) of Boc-Gln in 15.6 mL of DMF was added 631.4 mg (4.673 mmol) of 1hydroxybenzotriazole. The mixture was cooled in an ice bath. Then 760 pL (6.912 mmol) of N-methylmorpholine were added, followed by 883.9 mg (4.284 mmol) of DCC. The reaction mixture was stirred in the ice bath for 30 min and at room temperature for 5.8 hr.
The reaction mixture was then diluted with 8 mL of CH 2 Cl 2 and filtered. The filtrate was washed with 5% citric acid (3 x 50 mL), with saturated NaHCO 3 (3 x 50 mL), with saturated NaCI (50 mL), dried with MgSO 4 and evaporated to dryness. Purification by column chromatography on silica gel with 5% MeOH in CH 2 C12 afforded 1.555 g of peptide as an orange glassy solid: Rf 0.19 MeOH in CH 2 Cl 2 0.09 MeOH in CHC13), 0.37 (10% MeOH in CHC13); 'H NMR (250 MHz, DMSO-d 6 TMS) 5 8.24 (br d, J 7 Hz, NH), 8.17 (br d, J 7 Hz, NH), 8.0-7.9 1H), 7.8-7.7 2H), 7.7-7.6 1H), 7.22 (br s, NH), 6.83 (br d, J 8 Hz, NH), 6.76 (br s, NH), 5.1-5.0 1H), 4.3-4.1 (m, IH), 3.9-3.8 1H), 2.8-2.6 1H), 2.6-2.4 1H), 2.1-2.0 2H), 1.8-1.4 1.47 1.5H), 1.43 1.5H), 1.42 1.5H), 1.40 1.5H), 1.36 9H), 0.86 J 6.3 Hz, 1.5H), 0.79 J= 6.2 Hz, 1.5H), 0.73 (br t, 3H); 'C NMR (52-MHz, DMSO-d 6
TMS)
178.7, 177.8, 177.7, 173.7, 172.0, 171.7, 171.5, 159.9, 159.7, 155.1, 135.1, 135.0, 131.5, 131.4, 131.0, 130.9, 129.8, 129.7, 127.9, 127.8, 123.8, 109.8, 109.6, 79.5, 79.3,77.9, 69.6, 69.4, 53.7, 53.6, 50.5, 50.4, 31.4, 28.1, 27.6, 27.4, 24.2, 24.1,24.0, 22.6, 22.5, 22.1, 21.9, 21.6 21.2.
GIn-Leu-p-Lapachone To a solution of 1.519 g (2.533 mmol) of Boc-Gln-Leu-p-Lapachone in 12 mL of
CH
2 C2 was added 11 mL of TFA. The reaction mixture was stirred at room temperature for 30 min. The solvent was removed in vacuo. Column chromatography on silica gel with MeOH in CH 2 C2 afforded 1.976 mg (quant) of the TFA salt as an orange glassy solid; 'H NMR (250 MHz, DMSO-d 6 TMS) 8 8.97 (br d, J 6.5 Hz, NH), 8.90 (br d, J 7.0 Hz, NH), 8.30 (br s, NH), 8.0-7.9 1H), 7.9-7.8 2H), 7.7-7.6 1H), 7.45 (br s, NH), 6.98 (br s, NH), 5.2-5.1 1H), 4.3-4.2 1H), 3.9-3.8 1H), 2.8-2.7 1H), 2.5-2.4 1H). 2.2-2.1 2H), 2.0-1.8 2H), 1.7-1.5 3H), 1.49 1.5H), 1.44 1.42 1.5H), 1.41 1.5H), 0.87 J 6.3 Hz, 1.5H), 0.81 J =6.3 Hz, 1.5H), 0.75 (d, J 5.8 Hz, 1.5H), 0.73 J= 5.8 Hz, 1.5H); 13C NMR (52 MHz, DMSO-d 6 TMS) 178.7, 177.8, 177.8, 173.5, 171.3, 171.1, 168.7, 168.7, 159.9, 159.8, 135.1, 131.5, 131.4, 131.1, 131.0, 129.9, 129.8, 128.0, 123.8, 109.7, 109.5, 79.5, 79.3, 69.9,69.8, 51.7, 51.6, 50.8, 50.8,303, 26.8, 24.2, 24.1, 22.7, 22.5. 22.2, 22.0, 21.9, 21.6, 21.2.
Boc-Leu-Gln-Leu-p-Lapachone To a solution of 1.949 g (max 2.533 mmol) of Gln-Leu-p-Lapachone-TFA salt and 585.7 mg (2.533 mmol) of Boc-Leu in 10 mL of DMF was added 410.6 mg (3.038 mmol) of 1-hydroxybenzotriazole. The mixture was cooled in an ice bath. Then 685 p.L (6.230 mmol) of N-methylmorpholine were added, followed by 574.7 mg (2.785 mmol) of DCC.
The reaction mixture was stirred in the ice -bath for 3 0 min and at room temperature. for hr. The reaction mixture was then diluted with CHC1 3 and filtered. The filtrate was washed with 5% citric acid (5 x 50 mL), with saturated NaHCO 3 (4 x 70 mL), with saturated NaCI min), dried with MgSO 4 and evaporated to dryness. Purification by column chromatography on silica gel with 5% MeGH in CHC1 3 afforded 1.221 g from Boc- Gln-Leu-p-Lapachone)of peptide as an orange glassy solid: Rf 0.09 MeGH in
CHCI
3 0.29 MeOH in CHCl 3 NMR (250 MI-z, DMSO-d 6 TMS) 58.36 (br d, NH), 8.3 0 br d, 8.0-7.9 (in, I 7.9-7.7 (mn, 2H), 7.7-7.6 (in, I 7.19 (br s, NH), 6.90 (br s, NH), 6.75 (brdNH), 5.1-5.0(in, 1H), 4.3-4.1 2H), 4.0-3.9(in, 11), 2.8--2.7 (in, 1H), 2.5-2.4 (mn, 111), 2.1-2.0 (in, 2H), 1.8-1.4 (in, 8H), 1.47 1.5H), 1.43 1.41 1.5H), 1.40 1.514), 1.37 4.514) 1.35 4.5H), 0.9-0.8 (in, 7.5H), 0.78 (d,J= 6.2 Hz, 1.5H), 0.73 .J =5.5 Hz, 1.5H), 0.71 J 5.3 Hz, 1.5H); 3 C NMR (52 MHz, DMSO-d 6 TMS) 8 178.7, 177.8, 177.7, 173.6, 173.6, 172.3, 171.5, 171.4, 171.3, 159.9, 159.7, 155.2, 135.0, 131.5,.13 1.4, 131.0, 130.9, 129.8, 129.8, 127.9, 127.9, 123.8, 109.7,.
109.6, 79.5, 79.3, 78.0, 69.6, 69.5, 52.8, 51.4, 50.5, 50.5, 40.7, 31.2, 28.1,~ 24.2, 24.1, 22.9,- 22.6, 22.5,.22.1,.22.0, 21.9, 21.6, 21.4, 21.2.
Leu'Gln-Leu-j3-Lapachone To a solution of 1. 196 g (1.678 mmol) of Boc-Leu-Gn-Leu-13-Lapachone in 8 mnL of CH 2 Cl 2 was added 8 mL of TFA. The reaction mixture was stirred at room temperature for 30 min. The solvent was removed in vacuo. Column chromatography on silica gel with MeGH in CHC1 3 afforded 1.430 g (quant) of the TEA salt as an orange glassy solid: JRj =0.04 (10% MeOH in CHC1 3 0. 10 (15% MeOH in CHC1 3 0.19 (20% MeOH in CHCI 3 'H NMR (250 MHz, DMSO-d 6 TMS) 5 8.46 (br d, J 6.6 Hz, NH), 8.41 (br d, J =7.2 Hz, NH), 8.0-7.9 (mn, I1H), 7.9-7.8 (in, 2H1), 7.7-7.6 (mn, I 7.26 (br s, NH), 6.77 (br s, NH), 5.1-5.0 (mn, LH), 4.3-4.1 (mn, 2H1), 3.5-3.4 (mn, 1H), 2.8-2.7 (mn, IH), 2.5-2.4 (in;1H), 0.9-0.8 (in, 7.5H), 0.78 J= 6.1 Hz. 1.5H1), 0.174 J= 5.9 Hz, 1.5 0.72 (di J Hz, 1.5H1); 3 C NMR (52 MHz, DMSO-d 6 TMS) 5 178.7, 177.8, 177.8, 173.6, 171.6, 171.4.,171.2, 159.9, 159.8, 135.1, 131.5, 131.4, 131.1, 131.0, 129.9, 129.8, 127.9, 123.9, 109.8, 109.6, 79.6, 79.3, 69.6, 69.5, 51.9-51.6, 51.6, 50.5, 42.3-41.8, 31.2, 28.2, 28.0, 24.2, 24.1, 23.7, 22.8, 22.7, 22.6, 22.1, 21.9, 21.8, 21.6, 21.3, 21.2.
Nc-Boc-Lys(Ne-Cl-Cbz)-Leu-Gln-Leu-p3-Lapachone To a solution of 1.400 g (max 1.643 mmol) of Leu-Gln-Leu--Lapachone-TFA salt and 681.6 mg (1.643 mmol) of No-Boc-Lys(NE-Cl-Cbz) in 6.6 mL of DMF was added 266.3 mg (1.971 mmol) of 1-hydroxybenzotriazole. The mixture was cooled in an ice bath.
Then 380 iL (3.456 mmol) of N-methylmorpholine were added, followed by 372.9 mg (1.807 mmol) of DCC. The reaction mixture was stirred in the ice bath for 30 min and at room temperature for 5.5 hr. The reaction mixture was then diluted with CHC1 3 and filtered. The filtrate was washed with 5% citric acid (4 x 50 mL), with saturated NaHCO 3 (4 x 50 mL), with saturated NaCl (65 mL), dried with MgSO 4 and evaporated to dryness.
Purification by column chromatography on silica gel with 5% MeOH in CHCt 3 afforded 897.4 mg of peptide as an orange glassy solid: R 0.10 MeOH in CHCI 3
'H
NMR (250 MHz, DMSO-d 6 TMS) 5 8.31 (br d, J 7 Hz, NH). 8.25 (br d, J 7 Hz, NH), 8.0-7.9 2H (1 quinone-H 1 7.8-7.7 3H (2 quinone-H 1 7.7-7.6 (m, 1H (quinone-H)), 7.5-7.4 2H), 7.4-7.3 3H (2 Cl-Ph-H 1 7.19 (br s, NH), 6.90 (br d, J 8 Hz, NH), 6.77 (br s, NH), 5.1-5.0 4H), 4.3-4.1 3H), 3.9-3.8 (m, 1H), 3.0-2.9 2H), 2.8-2.7 1H), 2.5-2.4 1H), 2.1-2.0 2H), 1.9-1.4 14H), 1.47 1.5H), 1.42 1.5H), 1.41 1.5H), 1.40 1.5H), 1.37 9H), 0.9-0.8 0.77 J 6.2 Hz, 1.5H), 0.73 J 5.7 Hz, 1.5H), 0.70 J= 5.6 Hz, 1.5H); 1C NMR (52 MHz, DMSO-d 6 TMS) 8 178.7, 177.8, 177.7, 173.6, 171.8, 171.6,171.4, 171.3, 159.9, 159.7, 155.7, 155.3, 135.0, 134.5, 132.2, 131.5, 131.4, 131.0, 130.9, 129.8, 129.8, 129.5, 129.1, 127.9, 127.8, 127.2, 123.8, 109.7, 109.6, 79.5. 79.3, 78.0, 69.6, 69.5, 62.4, 54.3, 51.6, 50.7, 50.5, 50.4, 41.0,40.1, 31.3, 29.0, 28.1, 27.9, 27.7, 24.2, 24.1, 24.0, 23.9, 23.0, 22.7, 22.6, 22.5, 22.1, 22.0, 21.9, 21.6, 21.5, 21.2.
Lys(Ne-CI-Cbz)-Leu-Gln-Leu-p-Lapachone To a solution of 1.196 g (1.678 mmol) of Boc-Lys(Ne-Cl-Cbz)-Leu-Gln-Leu-p- Lapachone in 6 mL of CH 2 C1 2 was added 5 mL of TFA. The reaction mixture was stirred at room temperature for 30 min. The solvent was removed in vacuo. Column chromatography on silica gel with 15% MeOH in CHC1 3 afforded 568.9 mg of the TFA salt as an orange glassy solid: Rf 0.09 (10% MeOH in CHC1 3 0.23 (15% MeOH in CHC1 3 0.38 (20% MeOH in CHC1 3 'H NMR (250 MHz, DMSO-d 6 TMS) 8 8.28 (br d, J 7 Hz, NH), 8.23 (br d, J 7 Hz, NH), 8.1-8.0 NH), 8.0-7.9 2H (1 quinone-H 1 7.8-7.7 (in, 2H4), 7.7-7.6 (mn, 1W, 7.5-7.4 (in, 2H), 7.4-7.3 (mn, 3H (2 Cl-Ph-H 1NH)), 7.23 (br s, NH), 6.78 (br s, NH), 5.1-5.0 (mn, 4H), 4.3-4.1 (in, 4H), 3.0-2.9 (mn, 2H), 2.8-2.7 (mn, 1H), 2.5-2.4 (mn, 1H), 2.1-2.0 (mn, 2H), 1.9-1.4 (in, 14H), 1.47 1.5H), 1.42 (s, 1.41 1.5H), 1.39 1.5H4), 0.9-0O.8 (mn, 7.5H), 0.77 J 6.2 Hz, 1.5H), 0.73 (d, J 5.8 Hz, 1.5H), 0.71 J 5.6 Hz, 1.5K); 3 C NMR (52 MHz, DMSO-d 6 TMS) 8 178.7,.177.8, 177.7, 173.7, 171.8, 171.6, 171.4, 171.3, 159.9, 159.7, 155.7, 135.0, 134.6, 132.2, 131.5, 131.4, 131.0, 130.9, 129.9, 129.8, 129.5,-129.2, 127.9, 127.8, 127.2, 123.8, 109.7, 109.6, 79.5, 79.3, 69.6, 69.4, 62.4, 54.4, 51.7, 50.6, 50.5, 50.4, 41.1, 31.2, 29.2, 27.6, 27.5, 24.2, 24.2, 24.1, 23.0, 22.6, 22.5, 22.4, 22.0, 21.9, 21.6, 21.2.
Morpholino-Ser(OBn)-Lys(Ns,-Cl-Cbz)-Leu-Gln-Leu-p-Lapachone To a solution of 544.9 mg (5.323x104 mol) of Lys(Ns-Cl-Cbz)-Leu-Gln-Leu-P3- Lapachone-TFA salt and 164.2 ing 5.325x10 4 mol) of morpholino-Ser(OBn) in 2.15 mL of DMF was added 86.2 mg (6.3 79x10-4 mol) of I1-hydroxybenzotriazole. The mixture was cooled in an ice bath. Then 59 j tL (5.366xl 04'mol) of N-inethylmorpholine were added, followed by 120.7 mg (5.850xl 04 nol) of DCC. The reaction mixture was stirred in the ice bath for 3 0 min and at room temperature for 5.5 hr. The reaction mixture was then diluted with CHC1 3 and filtered. The filtrate was washed with 5% citric acid (4 x 30 mL), with *saturated NaH.CO 3 (4 x 30 mL), with saturated NaCl (30 mL), dried with MgSO 4 and evaporated to dryness. Purification by columrn chromatography on silica gel with 7%, MeOR in CHC1 3 afforded 515.8 mng (8 1 of peptide as an orange glassy solid:.Rf -0.17 MeOK in CHCl 3 0.36 (10% MeOH in CHCI 3 'H.NMR (250 MHz, DMSO-d 6 TMS) 6 8.22 (br d, J =7 Hz, NH), 8.18 (br d, J 7 Hz, NH), 8.0-7.9 (in, 2H (1 quin one-H I 7.9-7.7 (in, 3H (2 quinone-H I 7.7-7.6 (mn, 1H), 7.5-7.4 (mn, 2H), 7.4-7.2 (i,8H (2 Cl-Ph-H 5 Ph-H I 7.20 (br s, NH), 6.78 (br s, NH), 6.60 (br d, J =7 Hz, NH), 5.1-5.0 (in, 4.50 2H), 4.4-4.3 (in 1K), 4.3-4.1 (in, 4H), 3.7-3.6 (in, 2H), 3.6-3.5 3.3-3.2 (mn, 4H), 3.0-2.9 (in, 2H), 2.8-2.6 (in, I1K), 2.5-2T4:Cri5i, 1K), 2.1-2.0, (in, 2H), 1.9-1.4 (in, 14K), 1.46 1.5H4), 1.42 1.5 1.41 1.5H), 1.39 0.9-0.7 (nm, 9H), 0.72 J 5.4 Hz, 1.5H), 0.70 J 5.3 Hz, 1.5K); 1 3 C NMR (52 MHz, DMSO-d 6 TMS) 8 178.7, 177.8, 177.7, 173.6, 171.6, 171.5, 171.4, 171.3, 171.3, 170.8, 170.8, 159.9, 159.7, 157.3, 1552-, 138.2, 135.0, 134.5, 132.2, 131.5, 131.4, 131.0, 130.9, 129.9, 129.8, 129.5, 129.1, 128.1, 127.9, 127.8, 127.4, 127.3, 127.2, 123.8, 109.8, 109.6, 79.5, 79.3, 71.9, 69.6, 69.5, 65.8, 62.4, 54.6, 52.7, 51.7, 51.0, 50.5, 50.4, 43.9, 31.3, 31.3, 29.0, 27.8, 27.7, 24.2, 24.2, 24.1,24.0, 22.9, 22.5, 22.5, 22.0, 21.8, 21.6, 21.4, 21.2.
Morpholino-Ser-Lys-Leu-Gln-Leu-p-Lapachone (SL-11154) To a solution of 486.8 mg (4.057x10 4 mol) of morpholino-Ser(OBn)-Lys(Ne-C1- Cbz)-Leu-Gln-Leu-p-Lapachone in 9 mL of MeOH/CHC1 3 1:9 was added 180.5 mg Pd/C. Then two drops of HC1 were added. The reaction mixture was placed under an atmosphere of H 2 (balloon) and stirred at room temperature for 15.5 hr. Removal of catalyst by filtration and evaporation of solvent afforded a light brown solid. The material was dissolved in 12 mL ofMeOH/CHCl 3 1:9, and stirred at room temperature for 1 hr while bubbling air through the solution. Evaporation of solvent afforded an orange glassy solid.
Column chromatography on silica gel with 20-30% MeOH in CHCI 3 yielded 52.8 mg of material as an orange solid. The material was further purified by prep HPLC: Rf 0.06 (20% MeOH in CHC13).
Synthesis Of Morpholine-Ser-Lys-Leu-Gln-NHCH 2
CH
2 O-P-Lapachone (SI-11173) (see Fig. 8-(N-Boc-(2-Aminoethoxy))-P-Lapachone To a solution of 507.1 mg (2.263 mmol) of N-boc-2-bromethylamine and 562.3 mg (2.177 mmol) of 8-hydroxy-p-Lapachone in 18 mL of DMF was added 727 mg (4.786 mmol) of CsF, followed by 2.2 mL of a solution of IM TBAF in THF. The reaction mixture was stirred under N 2 at room temperature for 48 hr. Then the reaction mixture was partitioned between 100 mL of CHC1 3 and 75 mL of water plus 10 mL of 5% citric acid.
The aqueous phase was extracted with CHC1 3 (5 x 40 mL). The CHC13 extracts were combined, dried with MgSO 4 and evaporated. Column chromotography on silica gel with 5% MeOH in CHC13 afforded 305.8 mg of quinone as a red-orange glassy solid; Rf 0.49(5% MeOH in CHC13); 'H NMR (250 MHz, DMSO-d 6 TMS) 6 7.68 J=8.6 Hz, 1H), 7.35 J=2.7 Hz, 1H), 7.28 (dd, J=8.6, 2.7 Hz, 1H), 7.05 (br t, NH), 4.08 J=5.6 Hz, 2H), 3.4-3.3 2H), 2.37 J=6.5 Hz, 2H), 1.81 J=6.5 Hz, 2H), 1.41 6H), 1.39 (s, 9H), 3C NMR (52 MHz, DMSO-d 6 TMS) 6 179.0, 177.8, 161.3, 160.3, 131.4, 125.6, 124.7, 120.5, 113.3, 110.3, 78.9, 77.8, 67.0, 30.8, 28.1, 26.2, 15.7.
8-(2-Aminoethoxy)-f-Lapachone (SL-11168) To a solution of 219.8 mg (5.474xl 04 mol) of 8-(N-Boc-(2aminoethyoxy))-p3-lapachone in 6 mL of CHC1 3 was added 6 mL of TFA. The reaction mixtur e was stirred at room temperature for 20 min. The solvent was removed in vacuo. Column chromatography on silica gel with 20% MeOH in CHC1 3 afforded 210.7 mg of quinione (as the trifluoroacetate salt) as a red glassy solid: Rf= 0. 13 (10% MeOHJ in CHCI 3 'H NMR (25 0 MHz, DMSO-d 6 TMS) 5 8.1-8.0 (v br s, NH), .7.74 J=8.6 Hz, I1H), 7.45 J=2.6 Hz, I 7.34 (dd, J=8.6, 2.6 Hz, I 4.4-4.2 (in, 2H4), 3_3-3.2 (in, 2H), 2.3 8 6.5 Hz, 2H), 1.82 J=6.5 Hz, 2H), 1.42 6H).
Morphdlino-Ser(OBn)-Lys(Ne-Cbz)-Leu-Gn-NHCH 2 CHOj3..Lapachone To a solution of 210.7 mg (5.072xl104 mol) of NH2CH 2
CH
2 O-13-lapachone-TFA salt and 4 119mg(5.72x0 4 mol) of morpholino-Ser(OBn)-Lys(Ns..Cbz).Leu*Glnin mL of DMF was added 82.4 mg (6.098xlO0-4 mol) of I1-hydroxybenzotriazole. The mixture was cooled in an ice bath. Then 56 (5.0 9 3 x 104 mol) of N-inethylmorpholine were added, followed by 115.1 mng (5.578x10 4 mol) of DCC. The reaction mixture was stirred in the ice bath for 45 min and at room temperature for 5 hr.. The reaction mixture was then filtered and the filtrate diluted with CHC1 3 The filtrate was washed with 5% citric acid (4 x 30 inL), with saturated NaHCO 3 (3 x 40 with saturated NaCI (40 inL), dried with MgSO 4 and evaporated to dryness. Purification by column chromatography on silica gel with 5% MeGH in CHC1 3 afforded 139.6 mng of peptide as a red-orange glassy solid: Rr 0.07 MeGH in CHCI 3 0.3 3 (10% MeGH in CHC1 3 'H NMR (250 MHz, DMSO-d 6 TMS) 5 8.00 (br d, J=6 Hz, 7.85 (br d, J=8 Hz, NH), 7.82 (br d, J=7 Hz, NH), 7.68 J:=8.6 Hz, IH (quinone)), 7.4-7.2 (in, 12H (2 quinione 10 7.2-7.1. NH), 6.75 (br s, NH), 6.60 (br d, J=7 Hz, NH), 4.99 2H), 4.48 2H), 4.4-4.3 (in, 1H), 4.3-4.0 (in, 5H), 3.7-3.6 (in, 2H), 3.6-3.4 (in, 6H), 3.3 5-3.25 (in, 4H), 3.0-2.9 (in, 2H), 2.4- 2.3 (in, 2H), 2.1-2.0 (in, 2H), 1.9-1.4 (in, 13H), 1.40 6H), 0.8.1 J=6.4j Hz, 3H), 0.77 J=6.3 H-z, 3H).
Morpholiino-Ser-Lys-Leu-Gln4VHCH,CH 2 0-j3Lapachone To a solution of 133. 1 mng (1.215xl04 inol) of inorpholino-Ser(OBn)-Lys(Np,-Cbz)-Leu.
Gln-NHCH 2
CH
2 O-p3-lapachone in 45 mL. MeOH plus 5 mL. CHC1 3 was added 57.9 mng of Pd/C. Then two drops of HC1 were added. The reaction mixture was placed under an.
atmosphere of H 2 (balloon) and stirred at room temperature for 23 hr. Removal of catalyst by filtration and evaporation of solvent afforded a reddish-brown solid. The material was dissolved in 20 mL of MeOH and stirred at room temperature for 21 hr while bubbling air n through the solution. Evaporation of solvent afforded 107.0 mg of a dark red glassy solid.
The material was purified by prep HPLC to yield 55.1 mg Synthesis of Morpholino-Ser-Lys-Leu-Gln-PABC-DOX (see Fig. 16) D Morpholino-Ser(OAloc) was prepared from Ser(OtBu)-OtBu. Reaction of n Ser(OtBu)-OtBu with 4-morpholinecarbonyl chloride in pyridine yielded morpholino- Ser(OtBu)-OtBu. Morpholino-Ser(OtBu)-OtBu was hydrolyzed with TFA to yield morpholino-Ser. Esterification of morpholino-Ser with isobutylene in the presence of a catalytic amount of H 2 SO4 afforded morpholino-Ser-OtBu. Reaction of morpholino-Ser- OtBu with allyl 1-benzotriazolyl carbonate yielded morpholino-Ser(OAloc)-OtBu.
Morpholino-Ser(OAloc)-OtBu was hydrolyzed with TFA in CHCb3 to yield morpholino-Ser(OAloc).
Preparation of the tetrapeptide was accomplished using standard procedures. Fmoc- Leu was coupled to Gln-OtBu with DCC in the presence of l-hydroxybenzotriazole (HOBt) to give Fmoc-Leu-Gln-OtBu. Removal of the Fmoc group from Fmoc-Leu-Gln- OtBu with piperidine in CH 2
CI
2 /DMF produced Leu-Gln-OtBu. Fmoc-Lys(NE-Aloc) was coupled to Leu-Gln-OtBu with DCC in the presence of HOBt to give Fmoc-Lys(Ne-Aloc)- Leu-Gln-OtBu. Removal of the MFmoc group from Fmoc-Lys(NE-Aloc)-Leu-Gln-OtBu with piperidine in DMF produced Lys(Ns-Aloc)-Leu-Gln-OtBu. Morpholino-Ser(OAloc) was coupled to Lys(Ne-Aloc)-Leu-Gln-OtBu with DCC in the presence of HOBt to give morpholino-Ser(OAloc)-Lys(Ns-Aloc)-Leu-Gln-OtBu. Hydrolysis of morpholino- Ser(OAloc)-Lys(Ne-Aloc)-Leu-Gln-OtBu with TFA in CHC1 3 would give the tetrapeptide morpholino-Ser(OAloc)-Lys(Ns-Aloc)-Leu-Gln-OH. The tetrapeptide is condeinsed with PABC-DOX as described elsewhere. De Groot et al. (1999)1 J Med. Chem.
42:5277-83. The amino acid side chains are deprotected as described. De Groot et al.
(1999) 1 Med. Chem. 42:5277-83. Morpholino-Ser-Lys-Leu-Gln-PABC-DOX has been used as a substrate of the enzyme PSA as shown in Figure 16.
Synthesis of MopoioSrLsLuGnPB-HHC2--aahn (see Fig. 17) Morpholino-Ser(OAloc) was prepared from SER(OtBu)-OtBu. Reaction of Ser(OtBu)-OtBu with 4 -moi-pholineacarbonyl chloride in pyridine yielded morpholino- Ser(OtBu)-OtBu. Morpholino-Ser(OtBu)-OtBu was hydrolyzed with TFA to yield rnorpholino-Ser. Esterification of morpholino-Ser with isobutylene in the presence of a catalytic amount of H 2 S0 4 afforded morpholino-Ser-OtBu. Reaction of mor-pholino-Ser- OtBu with allyl I1-benzotriazolyl carbonate yielded morpholino-Ser(OAloc)-OtBu.
Morpholino-Ser(OAloc)-OtBu was hydrolyzed with TEA in CHC 13 1) to yield nmorpholino-Ser(OAloc).
Preparation of the tetrapeptide was accomplished using standard procedures. Fmoc- Leu was coupled to Gln-OtBu with DCC in the presence of 1-hydroxybenzotriazole (HOBt) to give Fmoc-Leu-Gln-OtBu. Removal of the Fmoc group from Fnoc,-Leu-Gln- OtBu with piperidine in CH 2
CI
2 /DMF produced Leu-Gln-OtBu. Fmoc-Lys(NE-Aloc) was coupled to Leu-Gln-OtBu with DCC in the presence of HOBt to give Fmoc-Lys(Ns-Aloc)- Leu-Gln-OtBu. Removal of the Fmoc group from Fmoc-Lys(NE-Aloc)-Leu-Gn.OtBu with piperidine in DMF produced Lys(Ne-Aloc)-Leu-Gln-OtBu. Morpholino-Ser(OAloc) was coupled to Lys(NE-Aloc)-Leu-Gln-OtBu with DCC in the presence. of HOBt to give morpholio-Ser(OAloc)-LyEAloc)-Leu-GlnOtBu. Hydrolysis. of morpholino- Ser(OAloc)-Lys(Ns-Aloc)LeuGnOtu with TFA in CHCl 3 would give the tetrapeptide morpholino-Ser(OAloc)..Lysq E-Aloc)-Leu-Gln-OH. The tetrapeptide is condensed with PABC-NHCH 2
CH
2 O0f3-lapachone in an analogous manner as the condensation of the tetrapeptide with doxorubicin, described in De Groot et al. (1999) J Med. Chemn. 42:5277-83; the amino acid side chains are deprotected using the procedure described in that reference. Morpholino-Ser-Lys-Leu-Gln-PAB
C-NHCH
2
CH
2 O7lapachone is used as a substrate of the enzyme PSA as shown in Figure 17.
EXAMPLE 2 CELL CULTURE AND DRUG TESTING PROTOCOL Cell Culture: The human lung adenocarcinoma cell line, A549, and human prostatic cancer cell line, DUPRO, were a gift from Dr. M. Eileen Dolan, University of Chicago, Department of Medicine. A549 was grown in Ham's F-12K medium (Fisher Scientific, Itasca, IL) supplemented with 10% fetal bovine serum and 2 mM L-glutamine.
DUPRO was grown in RPMI-1640 supplemented with 10% fetal bovine serum. The human colon carcinoma cell line, HT29, and the human breast carcinoma cell line, MCF7, were obtained from the American Type Culture Collection, Rockville, MD. HT29 cells were grown in McCoy's 5A medium (Gibco, BRL, Gaithersburg, MD) supplemented with 10% fetal bovine serum. MCF7 cells were grown in Richter's Improved Modified Eagle's medium supplemented with 10% fetal bovine serum and2.2 g/L sodium bicarbonate. The human prostate adenocarcinoma cell lines, LNCAP, PC-3 and DU145, were gifts from Dr.
George Wilding, University of Wisconsin Comprehensive Cancer Center and the Department of Medicine, and were grown in Dulbecco's Modified Eagle's medium supplemented with a 5% fetal bovine serum. The malignant glioma cell line, U251MG NCI was obtained from the brain tumor tissue bank at the University of California, San Francisco Department ofNeurosurgery, and was grown in Dulbecco's Modified Eagle's medium supplemented wth 10% fetal bovine serum. DUPRO, A549 and MCF7 cells were grown in 100 units/mL penicillin and 100 ug/mL streptomycin. HT29 and U251MG NCI cells were grown in 50 pg/mL gentamycin. LNCAP, PC-3 and DU145 cells were maintained in 1% antibiotic antimycotic solution (Sigma, St. Louis. MO). All cell cultures were maintained at 37°C in 5% C0 2 /95% humidified air.
MTT assay. Exponentially growing monolayer cells were plated in 96 well plates at a density of 500 cells/well and allowed to grow for 24 h. Serially diluted drug solutions were added such that the final drug concentrations in the treatment media were between 0 and 35 gM. Cells were incubated with drug at either 4 hr or 72 hr. After 4 hr and 72 hr treatment, drugs were removed, fresh media (without) drug (100 uL) was added.and cells were incubated for 6 days. After six days, 25 pL of a Dulbecco's phosphate-buffered saline solution containing 5 mg/mL of MTT (Thiazolyl blue) (Sigma) was added to each well and incubated for 4h at 37 0 C. Then 100 gL of lysis buffer (20% sodium dodecyl sulfate, 50% N,N-dimethylformamide and 0.8% acetic acid, pH 4.7) was added to each 69 well and incubated for an additional 22 h. A microplate reader (E max, Molecular Devices, Sunnyvale, CA) set at 570 nm was used to determine the optical density. Results were plotted as a ratio of the optical density in drug treated wells to the optical density in wells treated with vehicle alone. Plotting and estimation of ID 50 values were accomplished with manufacturer supplied software.
SL- 0 2.4 11087 0
N
SL- 0>31.25 11088 0 SL- 0 11.03 11089 H
H
SL- 0 4.2 11095 0 N Br SL- 0 3.6 11096 0 N
NJ)
SL- >12 11106 >31.2 .11106
OCH
2 Ph) 2
H
SL- 04.3 >31.25 17.2 11107 (bH)
H
SL- 0>3 1.25 >27.9 22.9 11112 I O (OH) 2
H
0
H
SL- 27.9 >31.25 29.2 I11113 O"PI OH) 2
HO
SL- 6.4 13.1 3.9 11120 OJ-
O)
.0 v-v Table 2 IDso (giM) Values of Quinones in Various Cultured Human Tumor Cell Lines Determined by the MTT Assay No. Structures of Quinones IDso (p4M) of different Tumor cells Lung Colon Breast Brain A549 HT-29 MCF7 U251-
MG
SL- 0 17.23 20.02 11051 0 O H
SL-M
e 26.88 11052 CO O 0 Me SL- 7.39 2.8 11053 0 0 SL- >31.25 >31.25 11054 O 0 0 SL- 0 HN >31.25 >31.25 >31.25 >31.25 11056 0 x= Me
(CH
2 )nCO(-X-) 3
NH(CH
2 3 NMe 2 n=4
SL- -o 11080 0 'O1~a 0 SL- 071 o 11081 I 0' ONa 0 SL- 11082 0 'Or'a 0' SL- 11083. I -W0 ONa 0
SL-H
11084 -W 0 Oi~a S-f 00 11085 0 0H SL- 19.8 6.05 11087 z
C)
7 2' SL- 0 11148 c O
NHTFA
NH
2
.TFA
Table 3 IDso pM) Value(s) of Non-Quinone Structure in A Cultured Human Prostate Tumor Cell Line Determined by the MTT Assay No. Structures of Compound IDso (pM) of different prostate cells Table 4
ID
50 Values of Selected Non-Quinone Compounds in Various Cultured Human Tumor Cell Lines Determined by the MTT Assay i No. Structures of Non-Quinone Compounds ID 50 of different Tumor cells Lung Colon Breast -Brain A549 HT-29 MCF7 U251-
MG
SL- HN\ >31.25 >31.25 >31.25 >31.25 11055x= 7 1N Me N CO(-X- 3 NH(C 2 3 NMe 2 Me) H SL- HN >31.25 11058 02X=
\N
Me.
/N CO(-X-) 2 NMeCH 2
CONH-
2 Me SL- N>31.25 11063 N N~
N'
HeH Me 1) (9 Table 5 lists additional quinones and quinone derivatives which are useful in the invention, either as therapeutics or, in the case of quinones which are not already covalently linked to or derivatized with peptides, as therapeutics in conjunction with peptides.
PCTUSOO/1 1538 Table No. Name and/lor Structure P-1apachone 0 0 SL-1 1001 0 3-hydroxy-p-1apachone SL- 11002 7 0
OH
3-malonyloxy-I3-1apachone SL-I 1003 0 00 0 0 3 -(pyrrolidinosuccinoy)-3-lapachofle 0 SL- 11004 0 0 X 00 3 -(morpholinosuccinoyl)-p-1apachofle SL-1 1005 0 00 SL-1 1006 3-(P-alanyloxy)- P-lapachone hydrobromide 0 0 0 N 72 3 -ethyl- D-alanyloxy)- j3-lapachone hydrobromide 0 SL- 11007 0
H
3 ,3-dinordunnione 0 SL-11008 0 0 *3-nordunnione 0 SL-1 100.9 0i 0 0 0 SL-11010 0 .0 naphthoquinonefiiran- 0 SL-11011 0 0' j3-norlapachone 0 SL-11012 c 0 4-pentoxy-1I,2-naphthoquinone 0 SL-11013 0 4-isobutoxy- I ,2-naphthoquinone 0 SL- 110 14 .0 4-isoamyloxy- 1 ,2-naphthoquinone 0 SL-1 1015 0 0 4-isopropoxy- I ,2-naphthoquinone 0 0 SL-11016 4-(2butenytoxy)- -1 ,2-naphthoquinone 0 SL-11017 7
I SL-11152
/NHVTFA
0 Hc- 0 In SL-11153 N 7 0l 0 NH 2 H SL-1 1154 0 00 0 H 0 0 SL-1 1168 H N
*TFA
SL-111N 0 00 SL-1 1173 NN
H
0 0
H
SL-1 1185 0 0 SL- 11187 0 91
I
K)
03 Although the foregoing invention has been described in some detail by way of
S
illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the appended claims.
0

Claims (20)

1. A c6mpound of the formula: A-M 1 -B-M 2 -D wherein A is selected from the group consisting of and -CH 2 wherein M 1 is selected from the group consisting of a single bond and C I-Cs, alkyl, C 1 -C8 branched alkyl, C 3 -C 8 cycloalkyl, and C 3 -Cs cycloaryl; wherein B is selected from the group consisting of -CH 2 and wherein R, is selected from the group consisting of C 1 -Cg alkyl, C 1 -Cs branched alkyl, C 3 -C 8 cycloalkyl, and C 3 -Cs cycloaryl; wherein M- is selected from the group consisting of a single bond and C 1 -C 8 alkyl,, C 1 -C 8 branched alkyl, C 3 -Cg cycloalkyl, and C 3 cycloaryl; wherein D is sele ctePd from the group consisting of -OH, -N(R 7 8 pentoses, hexoses, 2005201115 15 Mar 2005 and NN HH U(3) wherein 4 is selected from the group consisting of C I-C 8 alkyl, C -Cs branched alkyl, C 3 -C 8 cycloalIkyl, C 3 -C 8 cycloaryl, -N(R 9 )(R 10 and -CN; and wherein R 7 Ra, R 9 and RI() are independently selected from the group consisting of C,-C 8 alkyl, C 1 -Cg branched alkyl, C 3 -C cycloalkyl, C 3 -C 8 cycloaryl, and N
2. A compound according to the formula wherein x is an integer between 1 and 2; and each K is independently selected from the group consisting of H. Ci-C 8 alkyl, S 10 CI-C 8 alkenyl, Ci-C 8 alkanol, Ci-C 8 alkoxy, O 0 SO and where zero or two, but no more than two, vicinal K's in the molecule represent single electrons which form a pi bond, thus forming a double bond together with the existing sigma bond between the two adjacent carbons bearing the two vicinal K's.
3. A compound of the formula O S-ONa 0 'R y wherein R is selected from the group consisting of C -Cs alkyl, Ci-Cg cycloalkyl, C 3 -C 8 cycloaryl, Ci-Cs branched alkyl, and CI-Cs alkanol.
4. A compound of the formula 0 NG2 G, wherein Y is selected from the group consisting of -Br, -Cl, and and wherein G and G2 are independently selected from the group consisting of H, CI-C 8 alkyl, 0 and -C(=0)-CHnX3 where n is an intcger from 0 to 3 and X is selected from the group consisting of F, Cl, Br, and I. A compound of the formula wherein M is selected'from the group consisting of and
6. A compound of the formula wherein x is an integer between 1 and 2; each B is independently selected from the group consisting of H, Ci-Cg alkyl, C 3 -C8 cycloalkyl, C 3 -C 8 cycloaryl, Ci-C 8 alkyl-C 3 -C cycloalkyl, and CI-Cs alkyl-C 3 -C 8 cycloaryl; and each K is independently selected from the group consisting of H. OH, CI-Cs alkyl, CI-C 8 alkenyl, C -C 8 alkanol, C -Cs alkoxy, and where zero or two, but no more than two, vicinal K's in the molecule represent single electrons which form a pi bond, thus forming a double bond together with the existing sigma bond between the two adjacent carbons bearing the two vicinal K's.
7. A compound of the formula 0 O O/P (OB)2 O^^E; wherein each B is independently selected from the group consisting of H, CI-Cs alkyl, C 3 -Cs cycloalkyl, C 3 -Cs cycloaryl, Ci-Cs alkyl-C 3 -Cs cycloalkyl, and C -Cs alkyl-C 3 -Cs cycloaryl; and wherein R is selected from the group consisting of Ci-Cs alkyl and Ci-Cs alkanol.
8. A compound of the formula where M, is alkyl, y is an integer from 1 to 6, and L is selected from the group consisting of-O-K, or -N(K,K2); where K, and K, are independently selected from the group consisting of H, C,-Cs alkyl, Ci-C, alkyl-COOH, alkyl-COO-C,-C, alkyl, C,-C 8 alkyl-N(G,G,), and C,-C, alkyl-N(G3)-C-Cs alkyl-N(G 4 Gs); and wherein each of G 4 and G 5 is independently selected from the group consisting of H and alkyl.
9. A compound of the formula: where z is an integer between one and ten; Go 1 is selected from the group consisting of C,-C 8 alkyl; each M is independently selected from the group consisting of C,-C 8 alkyl; V is selected from the group consisting and and T is selected from the group consisting of-COOM 8 and -CONM 9 Mlo, where each of Ms, M 9 and Mio are independently selected from the group consisting of H and C alkyl. A compound of the formula O-M 1 2, where M 1 2 is selected from the group consisting of C -C 8 alkyl.
11. A compound of the formula n O o where M 1 4 and Mis are independently selected from the group consisting of CI-C 8 alkyl. Ct
12. A compound of the formula O J where J is selected from the group consisting of C-Cs alkyl, Ci-C 8 cycloalkyl, C3-Cs cycloaryl, and C -C 8 branched alkyl.
13. A compound of the formula NH 2 (9 CH 3 where Rs is the side chain of a naturally-occuring amino acid, attached in S or R configuration.
14. A compound of the formula S-L-QUIN wherein S represents a single amino acid or a peptide of at least two amino acids; L is a linking group containing at least one carbon, oxygen, or nitrogen atom attached covalently to both S and QUIN, or a nonentity; and QUIN is a quinone, quinone derivative, hydroquinone, or hydroquinone derivative. A compound according to claim 14, wherein S or a portion thereof, S-L or a portion thereof, or both S or a portion thereof and then L or a portion thereof, are cleaved from the quinone-containing remainder of the molecule by an enzyme.
16. A compound according to claim 15, wherein the enzyme is prostate specific antigen.
17. A compound according to claim 14, wherein L is or -NH-(CI-C 8 alkyl)-O-.
18. A compound according to claim 14, wherein L is -NH-(C 6 H 4 )CH 2 -0=ONH-)-NH-(C -Cg alkyl)-O-.
19. A compound according to claim 14, wherein S is X-Ser-Lys-Leu-Gln, wherein X is a protecting group or an amino-terminal capping group, and the side chains of Ser, Lys, and Gin may optionally be protected with protecting groups. A compound of the formula S-L-QUIN wherein S represents a single amino acid or a peptide of at least two amino acids; L is a linking group containing at least one carbon, oxygen, or nitrogen atom attached covalently to both S and QUIN, or a nonentity; and QUIN is selected from the group consisting of the quinone compounds of claim 13 and the compounds O0 0 0 H 2 N C OH ,and 0 O NH 2
21. A method for making a compound according to claim 14, comprising the steps of a) covalently linking L to S, and b) covalently linking L to QUIN, wherein steps a) and b) can be performed in either order or simultaneously.
22. A compound of the formula K wherein x is an integer between 1 and 2; W is selected from -OH, -O-Ci-Cs alkyl, -O-Ci-Cs alkyl-NH 2 and -O-CI-C 8 alkyl-NH-S, wherein S is a single amino acid or a peptide of two or more amino acids; and each K is independently selected from the group consisting of H, OH, Ci-C8 alkyl, CI-Cg alkenyl, CI-Cs alkanol, Ci-C 8 alkoxy, and where zero or two, but no more than two, vicinal K's in the molecule represent single electrons which form a pi bond, thus forming a double bond together with the existing.sigma bond between the two adjacent carbons bearing the two vicinal K's.
23. A compound according to claim 22 of the formula O 0 wherein W is. selected from -OH, -O-Ci-Cs alkyl, -O-Ci-Cg alkyl-NH2, and -O-C -C 8 alkyl-NH-S, and wherein S is a single amino acid or a peptide of two or more amino acids.
24. A compound according to claim 22, wherein W is -O-Ci-Cg alkyl-NH-S, wherein S is a single amino acid or a peptide of two or more amino acids; wherein the group -NH- forms an amide bond with the alpha-carboxy group of S when S .is a single amino acid and the group -NH- forms an amide bond with the C-terminal alpha-carboxy group of S when S is a peptide of two or more amino acids. A compound according to claim 23, wherein W is -O-CI-Cs alkyl-NH-S, wherein S is a single amino acid or a peptide of two or more amino acids; wherein the 107 group -NH- forms an amide bond with the alpha-carboxy group of S when S is a single amino acid and the group -NH- forms an amide bond with the C-terminal alpha-carboxy group of S when S is a peptide of two or more amino acids. Dated this fifteenth day of March 2005 SLIL Biomedical Corporation Patent Attorneysfor'the Applicant: FBRICE CO. O3
AU2005201115A 1999-04-30 2005-03-15 Quinones for treatment of diseases Abandoned AU2005201115A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2005201115A AU2005201115A1 (en) 1999-04-30 2005-03-15 Quinones for treatment of diseases

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60131842 1999-04-30
AU46780/00A AU4678000A (en) 1999-04-30 2000-04-27 Quinones for treatment of diseases
AU2005201115A AU2005201115A1 (en) 1999-04-30 2005-03-15 Quinones for treatment of diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU46780/00A Division AU4678000A (en) 1999-04-30 2000-04-27 Quinones for treatment of diseases

Publications (1)

Publication Number Publication Date
AU2005201115A1 true AU2005201115A1 (en) 2005-06-23

Family

ID=34682585

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005201115A Abandoned AU2005201115A1 (en) 1999-04-30 2005-03-15 Quinones for treatment of diseases

Country Status (1)

Country Link
AU (1) AU2005201115A1 (en)

Similar Documents

Publication Publication Date Title
US6809176B2 (en) Quinones as disease therapies
EP1185493B1 (en) Quinones for treatment of diseases
US7279502B2 (en) Polyamine analog conjugates and quinone conjugates as therapies for cancers and prostate diseases
US5880131A (en) High molecular weight polymer-based prodrugs
EP1177197B1 (en) Polyamines and their use in therapy
JP2002543163A5 (en)
JP2004513136A (en) Novel polyamine analog-amino acid conjugate useful as an anticancer agent
US6649587B1 (en) Polyamine analog conjugates and quinone conjugates as therapies for cancers and prostate diseases
EP1574507A2 (en) Furo- and pyrano-naphthoquinones and their use in the treatment of cancer
Liu et al. A novel anti-cancer stem cells compound optimized from the natural symplostatin 4 scaffold inhibits Wnt/β-catenin signaling pathway
AU2005201115A1 (en) Quinones for treatment of diseases
AU2009200157B2 (en) Novel polyamine analog conjugates and quinone conjugates as therapies for cancers and prostate diseases

Legal Events

Date Code Title Description
PC1 Assignment before grant (sect. 113)

Owner name: CELLGATE, INC.

Free format text: FORMER APPLICANT(S): SLIL BIOMEDICAL CORPORATION

MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application