WO2009064908A1 - Cbi derivatives subject to reductive activation - Google Patents

Cbi derivatives subject to reductive activation Download PDF

Info

Publication number
WO2009064908A1
WO2009064908A1 PCT/US2008/083433 US2008083433W WO2009064908A1 WO 2009064908 A1 WO2009064908 A1 WO 2009064908A1 US 2008083433 W US2008083433 W US 2008083433W WO 2009064908 A1 WO2009064908 A1 WO 2009064908A1
Authority
WO
WIPO (PCT)
Prior art keywords
chem
cbi
alkyl
group
represented
Prior art date
Application number
PCT/US2008/083433
Other languages
French (fr)
Inventor
Dale Boger
Original Assignee
The Scripps Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Scripps Research Institute filed Critical The Scripps Research Institute
Priority to ES08850632.4T priority Critical patent/ES2441115T3/en
Priority to CA2723883A priority patent/CA2723883C/en
Priority to US12/742,616 priority patent/US8377981B2/en
Priority to EP08850632.4A priority patent/EP2227087B1/en
Publication of WO2009064908A1 publication Critical patent/WO2009064908A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/58[b]- or [c]-condensed
    • C07D209/60Naphtho [b] pyrroles; Hydrogenated naphtho [b] pyrroles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention relates to prodrug anticancer agents and to their use. More particularly, the invention relates to I ⁇ J-acyl 0-amino phenol prodrugs of CBI-TMI and CBI-indole 2 .
  • CC- 1065 the duocarmycins, and yatakemycin constitute exceptionally potent naturally occurring antitumor agents that derive their biological properties through a characteristic sequence- selective DNA alkylation reaction (below) (Chidester, C. G.; et al. J. Am. Chem. Soc. 1981, 103, 7629; Trzupek, J. D.; et al . Nature Chem. Biol. 2006, 2, 79) .
  • the CBI derivatives proved to be four times more stable and, correspondingly, four times more potent than derivatives bearing the CC- 1065 alkylation subunit (7-MeCPI) approaching the stability and potency of duocarmycin SA and yatakemycin derivatives, and they exhibit efficacious in vivo antitumor activity in animal models at doses that reflect this potency (Boger, D. L.; et al . Bioorg. Med. Chem. Lett. 1991, 1, 115; Boger, D. L. ; et al. Bioorg. Med. Chem. 1995, 3, 1429) .
  • a unique feature of this class of molecules including the natural products themselves is the observation that synthetic phenol precursors (e.g., 1) to the final products, entailing a Winstein Ar-3 ' spirocyclization with displacement of an appropriate leaving group, exhibit biological properties typically indistinguishable from the cyclopropane- containing final products (DNA alkylation rate or efficiency, in vitro cytotoxic activity, and in vivo antitumor activity) .
  • This dependable behavior of the precursor phenols has provided the basis on which the development of useful, stable, or safe prodrugs has been conducted (Carzelesin: Aristoff, P. A. Adv. Med. Chem. 1993, 2, 67. KW-2189: Kobayashi, E.; et al . Cancer Res.
  • a contemplated compound of the invention is an N-acyl 0-amino CBI derivative that is represented by Formula I :
  • R 1 is selected from the group of radicals consisting of -C(O) (C 1 -C 6 alkyl) , -C(O)O(Ci- C 10 alkyl) , -C (0) (C 2 -C 6 alkenyl) , -C(O)O(C 2 -C 6 alkenyl) , and -C(0)aryl.
  • R 2 is selected from the group of radicals consisting of hydrogen, -C(O) (Ci-C 6 alkyl), -C(O) 0 (Ci-C 10 alkyl) , -C (0) (C 2 -C 6 alkenyl) , and -C(O)O(C 2 -C 6 alkenyl) .
  • R 1 and R 2 are combined to form a cyclic structure selected from the group consisting of divalent radicals represented as follows:
  • R 3 in Formula I is selected from group consisting of radicals represented as follows:
  • R 4 is selected from group consisting of radicals represented as follows:
  • R 5 , R 6 , R 7 and R 8 in the above structural formulas are each independently selected from the group of radicals consisting of -H, -OH, -0(Ci-C 6 alkyl) , - (Ci-C 6 alkyl) and halogen.
  • R 9 of an above formula is selected from the group of radicals consisting of -H, -C(O)O(C 1 -C 6 alkyl) , -C (0) (Ci-C 6 alkyl) , -C(O)NH 2 , -C(O)NHNH 2 , and -C(O)NHNHC(O)O(Ci-C 6 alkyl) .
  • R 1 is selected from the group of radicals consisting of -C(O) (Ci-C 6 alkyl) and -C(O)O(Ci-Ci 0 alkyl);
  • R 2 is selected from the group of radicals consisting of hydrogen (hydrido; -H) , -C(O) (C 1 -C 6 alkyl), and -C(O)O(C 1 -Ci 0 alkyl); or, alternatively, R 1 and R 2 combine to form a cyclic divalent radical represented by the following structure (phthalyl) :
  • R 3 is selected from the group consisting of the following radicals:
  • Particularly preferred compounds include those with the following structural formulas:
  • a process for treating a proliferative disease such as a cancer or leukemia in a mammal is also contemplated.
  • an effective amount of a compound of Formula I such as one of the five compounds shown immediately above is administered to a mammal in need thereof .
  • the use of a compound of Formula I in the manufacture of a medicament for treating a proliferative disease such as cancer or leukemia is contemplated.
  • Figure 1 illustrates the results of an electrophoresis gel with 8% denaturing PAGE and autoradiography. Thermally- induced strand cleavage of w794 DNA; DNA-agent incubation at 4 0 C for 18 hours, removal of unbound agent by EtOH precipitation, and 30 minutes of thermolysis (100 0 C) followed by 8% denaturing PAGE and autoradiography.
  • Lane 1 control DNA; lanes 2-5, Sanger G, C, A, and T sequencing reactions; lanes 6-8, 2 (IxIO ""4 to IxIO “6 ) ; lanes 9-11, 10 (IxIO “1 to IxIO “3 ) ; lanes 12-14, 4 (IxIO "1 to IxIO “3 ) , lanes 15-17, 9 (IxIO "1 to IxIO “3 ) . All compounds possess the natural IS-configuration.
  • the reductively activated agent 4 was found to alkylate w794 DNA with an identical sequence selectivity as the parent agent CBI-TMI (2) , albeit with a substantially reduced efficiency (1,000- 10, 000-fold) .
  • the O-methyl ether 10 as well as 9 lacking a C4 substituent failed to exhibit significant observable DNA alkylation.
  • a novel set of reductively activated phenol prodrugs of the CC- 1065 and duocarmycin class of compounds is disclosed. These compounds do not require enzymatic release and are illustrative of other phenolic drugs that can benefit from such a designed activation.
  • Alternative and prior efforts at incorporating a reductive activation into the CC-1065 and duocarmycin class includes the Denny disclosures of nitro precursors to aryl amine variants of the phenol precursors (Hay, M. P.; et al . J. Med. Chem. 2003, 46, 5533; Tercel, M.; et al . J. Med. Chem.
  • CBI (1,2,9,9a- tetrahydrocyclopropa [c] benz [e] indol-4-one) and its precursor 1 where R is just the DNA binding portion of the molecule along with its precursor, the 0-amino phenol derivative or prodrug that requires reductive activation by N-O bond cleavage are shown below.
  • the CBI compounds are more accessible than the natural products, yet indistinguishable in their DNA alkylation selectivity (Boger, D. L.; Munk, S. A. J. Am. Chem. Soc. 1992, 114, 5487) .
  • the CBI derivatives proved to be four times more stable and, correspondingly, four times more potent than derivatives bearing the CC- 1065 alkylation subunit (7-MeCPI) approaching the stability and potency of duocarmycin SA and yatakemycin derivatives, and they exhibit efficacious in vivo antitumor activity in animal models at doses that reflect this potency. Consequently, CBI and its derivatives have been the focus of much development as well as the prototype analogues on which new design concepts have been explored, developed, or introduced, including the instant invention. cleavage of weak N-O bond
  • a contemplated compound of the invention is an N-acyl 0-amino CBI derivative that is represented by Formula I :
  • R 1 is selected from the group of radicals consisting of -C(O) (C 1 -C 6 alkyl) , -C(O)O(Ci-Ci 0 alkyl) , -C (0) (C 2 -C 6 alkenyl) , -C(O)O(C 2 -C 6 alkenyl) , and -C(0)aryl.
  • R 2 is selected from the group of radicals consisting of hydrogen, -C(O) (C x -C 6 alkyl), -C(O)O(Ci-Ci 0 alkyl), -C(O) (C 2 -C 6 alkenyl), and -C(O)O(C 2 -C 5 alkenyl) .
  • R 1 and R 2 are combined to form a cyclic structure selected from the group consisting of divalent radicals represented as follows:
  • R 3 in Formula I is selected from group consisting of radicals represented as follows :
  • R 4 is selected from group consisting of radicals represented as follows :
  • R 5 , R 6 , R 7 and R 8 in the above structural formulas are each independently selected from the group of radicals consisting of -H, -OH, -0(Ci-C 6 alkyl) , - (C 1 -C 6 alkyl) and halogen.
  • R 9 of an above formula is selected from the group of radicals consisting of -H, -C(O)O(Ci-C 5 alkyl) , -C (0) (C 1 -C 6 alkyl) , -C(O)NH 2 , -C(O)NHNH 2 , and -C(O)NHNHC(O)O(Ci-C 6 alkyl) .
  • Ci-Cg alkyl denotes a straight or branched chain radical such as a methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, amyl, tert-amyl, hexyl group and the like.
  • C2-Cg alkenyl denotes a radical such as a vinyl, allyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl group and the like, as well as dienes and trienes of straight and branched chains containing up to six carbon atoms and at least one carbon-to-carbon (ethylenic) double bond.
  • halogen includes fluoro, chloro, bromo and iodo, with chloro being preferred.
  • aryl is meant to include a monocyclic or dicyclic aromatic radical containing 5 to 10 atoms in the ring system and zero, one or three atoms other than carbon in the rings .
  • the atoms other than carbon can be selected from oxygen, nitrogen and sulfur.
  • Illustrative aryl radicals include phenyl, 1- and 2-naphthyl, pyridyl, pyrazinyl, pyrimidyl, imidazyl, thiophenyl, furanyl, pyrrolyl, 1, 3 , 5-triaziyl, 1, 2, 4-triazinyl and 1,2,3- triazinyl, quinazolinyl, quinolinyl, their various positional isomers, and the like.
  • a pharmaceutical composition for treating A process for treating a proliferative disease such as a cancer or leukemia in a mammal is also contemplated.
  • Such a composition contains a pharmaceutically effective amount of a before- discussed molecule of Formula I dissolved or dispersed in a pharmaceutically acceptable diluent.
  • a contemplated compound of Formula I can be used in a pharmaceutical composition to treat and preferably kill cancer cells or cells of another proliferative disease such as leukemia in vitro or in vivo in a mammalian subject.
  • an above composition is contacted with the cells to be treated.
  • the cells so treated are maintained in contact with a compound of Formula I until cleared by the body when in vivo, or for various times as desired in an in vitro study.
  • the treatment is generally repeated several times.
  • a mammal to which or whom a compound of Formula I composition is administered can be a primate such as a human, an ape such as a chimpanzee or gorilla, a monkey such as a cynomolgus monkey or a macaque, a laboratory animal such as a rat, mouse or rabbit, a companion animal such as a dog, cat, horse, or a food animal such as a cow or steer, sheep, lamb, pig, goat, llama or the like in need of treatment for a cancerous condition.
  • a primate such as a human, an ape such as a chimpanzee or gorilla, a monkey such as a cynomolgus monkey or a macaque
  • a laboratory animal such as a rat, mouse or rabbit
  • a companion animal such as a dog, cat, horse
  • a food animal such as a cow or steer, sheep, lamb, pig, goat, llama or the like in need of treatment
  • a contemplated composition is administered to a mammal in need of the medication at an proliferative effective dosage level. That level is typically an amount sufficient to provide about 10 to about 100 ⁇ g/kg of body weight to the recipient's plasma or serum, using the molecular weight of the scission-activated duocarmycin-type prodrug Compound 8 itself as the basis for calculation in view of the
  • the amount can vary depending on the recipient and proliferative cell load. Those plasma or serum concentrations can usually be obtained by i.v. administration using a liquid dosage form that contains about 200 mg to about 1000 mg of chimer compound per day. The determination of optimum dosages for a particular situation is within the skill of the art.
  • a compound of Formula I composition is administered repeatedly, on a schedule adapted for a recipient's cancer load and need, as is well known in the art . Typical administrations are given multiple times within a one month time period, usually followed by a rest period and then further administrations and rest periods until the recipient is free of the disease, or longer for prophylactic purposes .
  • an inert, pharmaceutically acceptable carrier or diluent is used for preparing pharmaceutical compositions containing a chimer compound of the invention.
  • the diluent is usually in liquid form.
  • Liquid pharmaceutical compositions include, for example, solutions suitable for parenteral administration.
  • Sterile water solutions of the active chimer or sterile solutions of the active component in solvents comprising water, ethanol, or propylene glycol are examples of liquid compositions suitable for parenteral administration.
  • Sterile solutions can be prepared by dissolving the active component in the desired solvent system, and then passing the resulting solution through a membrane filter to sterilize it or, alternatively, by dissolving the sterile compound in a previously sterilized solvent under sterile conditions .
  • the pharmaceutical composition is in unit dosage form.
  • the composition is divided into unit doses containing appropriate quantities of the active urea.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparation, for example, in vials or ampules.
  • the inactive analogue 9 was also prepared for comparison and by an approach that precludes the presence of such a contaminate phenol because there is no C-4 functionality in the starting material 16.
  • Compound 20 was converted to 21 enlisting a key 5-exo-trig aryl radical-alkene cyclization (Boger, D. L.; et al . Tetrahedron Lett. 1998, 39, 2227) .
  • N-acyl amino phenol prodrugs displayed a useful range of stability, yet were susceptible to cleavage of the critical N-O bond. As might be anticipated, their relative stability followed the order of 4 > 5 > 6 > 7 with 4 and 5 withstanding even long term storage effectively, but with 7 noticeably deteriorating over time.
  • 4 was stable to treatment with BnSH in THF (2-72 hours, 23 0 C) or MeOH (2-72 hours, 23 0 C) , and stable to treatment with BnSH in THF even in the presence of insoluble NaHCO 3 (2 hours, 23 0 C) , but is cleaved to release 2 upon treatment with BnSH in MeOH in the presence of NaHCO 3 (2 hours, 23 0 C) .
  • the stability of 4 was assessed in pH 7.0 phosphate buffer and within the limits of detection (HPLC, UV) , no significant cleavage of the prodrug was observed over the time monitored (72 hours) .
  • the stability of 4 was monitored in human plasma (50 ⁇ g/100 ⁇ L, 10% DMSO) in which it displayed a half-life of 3 hours with release of the free drug 2.
  • the relative potency of the prodrugs when distinguishable, mirrors the expected ease of N-O bond cleavage (e.g. L1210: 7 > 6 > 5 > 4) suggesting fundamental chemical principles can be used to "tune" the reductive free drug release.
  • the potency differences between the free drug 2 and the prodrugs diminish as the hypoxic character of the cell line increases; 4 is 10 -fold less potent than 2 against L1210, but 2 and 4 are essentially equipotent against H460/H596.
  • this reductive activation is not linked to the expression levels of DT-Diaphorase because 2 and 4-7 remain equipotent in the H460 or H596 cell lines, although H596 is 10-fold less sensitive than H460 to seco-CBI-TMI itself.
  • This result illustrates that DT-Diaphorase is not mediating the reductive release of the drug from the 0-amino phenol prodrugs, indicating that their utility is orthogonal to that of mitomycin. Rather, their behavior is consistent with the suggestion that the activation is nonenzymatic and likely is mediated in situ by appropriate nucleophiles .
  • prodrug 8 was essentially equipotent with CBI-indole 2 (3) itself, indicating effective release of the free drug under the conditions of the assay.
  • (+) -CBI-indole 2 Given the efficacy of (+) -CBI-indole 2 in animal tumor models, (Boger, D. L.; Ishizaki, T.; Sakya, S. M.; Munk, S. A.; Kitos, P. A.; Jin, Q.; Besterman, J. M. Bioorg. Med. Chem. Lett. 1991, I 1 115; Boger, D. L.; Yun, W.; Han, N. Bioorg. Med. Chem. 1995, 3, 1429) it was especially interesting to compare 8 with 3 in vivo.
  • the reductively activated agent 4 was found to alkylate w794 DNA with an identical sequence selectivity as the parent agent CBI-TMI (2) , albeit with a substantially reduced efficiency (1,000-10,000 fold) .
  • the 0-methyl ether 10 as well as 9 lacking a C4 substituent failed to exhibit significant observable DNA alkylation.
  • the prodrug 8 was examined for in vivo efficacy alongside the parent drug 3 in a standard antitumor model enlisting L1210 murine leukemia implanted i.p. into DBA/2J mice.
  • This model has been reported to respond well to the parent drugs of related compounds (Li, L. H.; et al . Invest. New Drugs 1991, 9, 137) and is a system that collaborators through the years have used to assess an extensive series of (+) -CBI-indole 2 analogues. Although not published, these latter studies provided the foundation on which examination of 8 is based.
  • the dose range (10-100 ⁇ g/kg) and the dosing schedule (administered three times i.p. on days 1, 5, and 9) found suitable for related parent drugs including (+) -CBI-indole 2 (3) (Boger, D. L.; et al. Bioorg. Med. Chem. Lett. 1991, I 1 115; Boger, D. L.; et al. Bioorg. Med. Chem. 1995, 3, 1429), the prodrug 8 was examined as is shown in the Table below.
  • the efficacy of 8 is extraordinary providing 5/6 long-term survivors at 52 weeks (365 days, T/C >1550) at the optimal dosing examined (100 ⁇ g/kg) . Notably, little distinction between 3 and 8 was observed at days 30-100 except that the prodrug-treated animals appeared healthier, displaying little or no weight loss that was evident with 3 at the highest dosing.
  • the second column is the dose in rag/kg of body weight of the animal that is administered i.p. (into the intraperitoneal cavity) on days 1, 5, and 9.
  • the surviving mice are the number of mice that are still living after 120 days and the experiment was then terminated.
  • the DNA alkylation reactions were performed by treatment of 4.5 ⁇ L of singly 32 P 5' -end- labeled double-stranded w794 DNA (Boger, D. L.; et al . Tetrahedron 1991, 47, 2661) in TE buffer (10 mM Tris, 1 mM EDTA, pH 7.6) with 0.5 ⁇ L of agent in EtOH (at the specified concentration) .
  • TE buffer 10 mM Tris, 1 mM EDTA, pH 7.6
  • EtOH at the specified concentration
  • the DNA was pelleted by centrifugation at 4 0C (13000 rpm, 25 minutes) , dried in a Savant Speed Vac concentrator, and resuspended in 5 ⁇ L of TE buffer (pH 7.6) .
  • Thermal depurination (3 x 10 minutes at 100 "C) was performed and then 2.5 ⁇ L of formamide dye solution was added to the cooled samples .
  • Thermally denatured samples were assayed by gel electrophoresis [8% denaturing gel with 8 M urea in TBE buffer (89 mM Tris-borate, 2 mM EDTA)] followed by autoradiography of the dried gel using Kodak BIOMAX XAR film and a Picker SpectraTM intensifying screen.
  • Methanesulfonyl chloride (59 ⁇ L, 0.607 mmol) was added at 0 0 C. After stirring at 23 0 C for 6 hours, the reaction mixture was diluted with EtOAc (20 mL) , and washed with water (10 mL x 2) , and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. The crude residue was dissolved in DMF (2 mL) and was treated with LiCl (26 mg, 0.607 mmol) .
  • a solution of seco-CBI-TMI (2, 5.0 mg, 0.011 mmol) in THF (0.5 mL) was treated with LiHMDS (1 M in THF, 13 ⁇ L, 0.013 mmol) at -78 0 C, and the resulting mixture was stirred at -78 0 C for 30 minutes.
  • the resulting solution was treated with N- p-tolylsulfonyloxyphthalimide (5.1 mg, 0.016 mmol) .
  • the reaction mixture was stirred at 23 0 C for an additional 60 minutes.
  • the solution was diluted with EtOAc (10 mL) and washed with water (5 mL) , and saturated aqueous NaCl (5 mL) .
  • DBA/2J mice were purchased from Jackson Laboratory (Bar Harbor, ME) and housed in the animal facility at The Scripps Research Institute. L1210 tumor cells, originally isolated from DBA/2 mice, were cultured in DMEM medium containing 5% fetal calf serum. For tumor implantation, DBA/2J mice were i.p. injected with 1 x 10 5 L1210 cells at day 0.
  • Compounds 3 and 8 were formulated with 30% DMSO plus 0.1% glucose solution. Treatment doses of drugs (0, 10, 30, 100 lg/kg wt . of animal) were i.p. injected consecutively on day 1, 5 and 9. The study was performed with six mice per group. Tumor growth in the peritoneal cavity was monitored daily and the death of animals was recorded. If necessary, weights of animals were measured once a week. To date this monitoring of the animals has lasted 365 days.
  • mice per group An analogous study with 10 mice per group was performed at the University of Kansas with the distinction that the compounds were administered in neat DMSO (0, 10, 30, 60, 100 lg/kg wt . of animal) and the study was terminated after 120 days.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A unique class of N-acyl O-amino phenol prodrugs of CBI-TMI and CBI-indole2 were synthesized and shown to be prodrugs, subject to reductive activation by nucleophilic cleavage of a weak N-O bond, effectively releasing the free drug in functional cellular assays for cytotoxic activity approaching or matching the activity of the free drug, yet remain essentially stable to ex vivo DNA alkylation conditions. Most impressively, assessment of the in vivo antitumor activity of a representative O- (acylamino) prodrug, 8, indicate that they approach the potency and exceed the efficacy of the free drug itself (CBI-indole2), indicating that the inactive prodrugs not only effectively release the free drug in vivo, but that they offer additional advantages related to a controlled or targeted release in vivo.

Description

CBI DERIVATIVES SUBJECT TO REDUCTIVE ACTIVATION
SPECIFICATION
Cross -Reference To elated Application This application claims priority of provisional applications Serial No. 61/002,978 and Serial No. 60/987,647, both of which were filed on November 13, 2007, and whose disclosures are incorporated by reference .
FIELD OF INVENTION
The invention relates to prodrug anticancer agents and to their use. More particularly, the invention relates to I\J-acyl 0-amino phenol prodrugs of CBI-TMI and CBI-indole2.
BACKGROUND
CC- 1065, the duocarmycins, and yatakemycin constitute exceptionally potent naturally occurring antitumor agents that derive their biological properties through a characteristic sequence- selective DNA alkylation reaction (below) (Chidester, C. G.; et al. J. Am. Chem. Soc. 1981, 103, 7629; Trzupek, J. D.; et al . Nature Chem. Biol. 2006, 2, 79) .
Figure imgf000004_0001
(+)-CC-1065 (+)-Yatakemycin
Figure imgf000004_0002
(+)-Duocarmycin A (+)-Duocarmycin SA
The examination of the natural products, their synthetic unnatural enantiomers, their derivatives, and synthetic analogues have defined fundamental features that control the alkylation selectivity, impact the alkylation efficiency, and are responsible for DNA alkylation catalysis providing a detailed understanding of the relationships between structure, reactivity, and biological activity (Warpehoski, M. A. ; Hurley, L. H. Chem. Res. Toxicol. 1988, 1, 315; Boger, D. L. Chem. Biol. 2004, 11, 1607.) .
One of the most important and widely explored class of analogues is CBI (Boger, D. L.; et al. J. Am. Chem. Soc. 1989, 111, 6461; Boger, D. L.; et al. J. Org. Chem. 1990, 55, 5823) (1,2,9,9a- tetrahydrocyclopropa [c] benz [e] indol-4-one) , being synthetically (Boger, D. L.; et al . J. Am. Chem. Soc. 1989, 111, 6461; Boger, D. L.; et al . J. Org. Chem. 1990, 55, 5823; Boger, D. L.; et al . J. Org. Chem. 1992, 51, 2873; Boger, D. L.; McKie, J. A. J". Org. Chem. 1995, 60, 1271; Drost, K. J.; Cava, M. P. J. Org. Chem. 1991, 56, 2240; Aristoff, P. A.; Johnson, P. D. J. Org. Chem. 1992, 51, 6234; Mohamadi, F.; et al. J. Med. Chem. 1994, 31, 232; Ling, L.; et al . Heterocyclic Commun. 1997, 3, 405; Boger, D. L.; et al. Synlett 1997, 515; Boger, D. L.; et al . Tetrahedron Lett. 1998, 39, 2227; Kastrinsky, D. B . ; Boger, D. L. J. Org. Chem. 2004, 69, 2284) more accessible than the natural products, yet indistinguishable in their DNA alkylation selectivity (Figure 2) (Boger, D. L.; Munk, S. A. J. Am. Chem. Soc. 1992, 114, 5487) .
Moreover, the CBI derivatives proved to be four times more stable and, correspondingly, four times more potent than derivatives bearing the CC- 1065 alkylation subunit (7-MeCPI) approaching the stability and potency of duocarmycin SA and yatakemycin derivatives, and they exhibit efficacious in vivo antitumor activity in animal models at doses that reflect this potency (Boger, D. L.; et al . Bioorg. Med. Chem. Lett. 1991, 1, 115; Boger, D. L. ; et al. Bioorg. Med. Chem. 1995, 3, 1429) . Consequently, CBI and its derivatives have been the focus of much development as well as the prototype analogues on which new design concepts have been explored, developed, or introduced (Boger, D. L.; et al. J. Am. Chem. Soc. 1989, 111, 6461; Tietze, L. F. ; et al. Angew. Chem. Int. Ed. 2006, 45, 6514; Wang, Y.; et al. Bioorg. Med. Chem. 2003, 11, 1569; Jeffrey, S. C; et al . J. Med. Chem. 2005, 48, 1344; Kline, T.; et al . MoI. Pharmaceut. 2004, 1, 9; Hay, M. P.; et al. J. Med. Chem. 2003, 46, 5533; Tercel, M.; et al. J. Med. Chem. 2003, 46, 2132; Gieseg, M. A.; et al. Anti-Cancer Drug Design 1999, 14, 77; Hay, M. P.; et al. Bioorg. Med. Chem. Lett. 1999, 9, 2237; Atwell, G. J.; et al. J. Med. Chem. 1999, 42, 3400; Atwell, G. J.; et al . J. Org. Chem. 1998, 63, 9414; Atwell, G. J.; et al . Bioorg. Med. Chem. Lett. 1997, 7, 1493; Townes, H.; et al . Med. Chem. Res. 2002, 11, 248; Boger, D. L.; Garbaccio, R. M. J. Org. Chem. 1999, 69, 8350) .
A unique feature of this class of molecules including the natural products themselves is the observation that synthetic phenol precursors (e.g., 1) to the final products, entailing a Winstein Ar-3 ' spirocyclization with displacement of an appropriate leaving group, exhibit biological properties typically indistinguishable from the cyclopropane- containing final products (DNA alkylation rate or efficiency, in vitro cytotoxic activity, and in vivo antitumor activity) . This dependable behavior of the precursor phenols has provided the basis on which the development of useful, stable, or safe prodrugs has been conducted (Carzelesin: Aristoff, P. A. Adv. Med. Chem. 1993, 2, 67. KW-2189: Kobayashi, E.; et al . Cancer Res. 1994, 54, 2404; Amishiro, N.; et al . Bioorg. Med. Chem. 2000, 8, 1637; Amishiro, N.; et al. J. Med. Chem. 1999, 42, 669; Nagamura, S.; et al . Chem. Pharm. Bull. 1996, 44, 1723; Nagamura, S.; et al. Chem. Pharm. Bull. 1995, 43. CBI: Boger, D. L. ; et al. Synthesis 1999, 1505) .
One feature limiting the attractiveness of this class of cytotoxic agents is their remarkable potencies (IC50 5-20 pM) creating special requirements for their preparation and handling. In many instances, this has been addressed by the introduction of chemically stable phenol protecting groups that are readily cleaved at the final stage of their preparation or upon in vivo administration. Such protected phenol precursors are intrinsically much less potent, yet readily release an active precursor to the drug upon deprotection. Extensions of this protection and release strategy have been pursued in which the free phenol release in vivo is coupled to features that might facilitate tumor selective delivery or cleavage (Wolkenberg, S. E . ; Boger, D. L. Chem. Rev. 2002, 202, 2477. Reviews on reductive activation: Papadopoulou, M. V. ; Bloomer, W. D. Drugs Future 2004, 29, 807; Jaffar, M. ; Stratford, I. J. Exp. Opin. Ther. Patents 1999, 9, 1371; Patterson, L. H.; Raleigh, S. M. Biomed. Health Res. 1998, 25, 72) . Such inactive prodrugs serve the dual role of providing safer handling intermediates or final products as well as potentially enhancing the therapeutic index of the drug.
As attractive and amenable as this approach is for this class of drugs, a surprisingly small series of such studies have been disclosed (Chari, R. V. J.; et al. Cancer Res. 1995, 55, 4079; Lillo, A. M.; et al. Chem. Biol. 2004, 11, 897; Tietze, L. F. ; et al. Eur. J. Org. Chem. 2002, 10, 1634; Tietze, L. F.; et al. Angew. Chem. Int. Ed. 2002, 41, 759; Tietze, L. F.; et al . ChemBioChem 2001, 2, 758; Tietze, L. F.; et al . Angew. Chem. Int. Ed. 2006, 45, 6574; Wang, Y.; et al . Bioorg. Med. Chem. 2003, 11, 1569; Jeffrey, S. C; et al . J. Med. Chem. 2005, 48, 1344; Kline, T.; et al . MoI. Pharmaceut. 2004, 1, 9; Hay, M. P.; et al . J. Med. Chem. 2003, 46, 5533; Tercel, M.; et al . J. Med. Chem. 2003, 46, 2132; Gieseg, M. A.; et al . Anti-Cancer Drug Design 1999, 14, 77; Hay, M. P.; et al . Bioorg. Med. Chem. Lett.
1999, 9, 2237; Atwell, G. J.; et al . J. Med. Chem.
1999, 42, 3400; Atwell, G. J.; et al . J. Org. Chem.
1998, 63, 9414; Atwell, G. J.; et al . Bioorg. Med.
Chem. Lett. 1997, 7, 1493; Townes, H.; et al . Med.
Chem. Res. 2002, 11, 248; Boger, D. L.; Garbaccio, R.
M. J. Org. Chem. 1999, 69, 8350) .
BRIEF SUMMARY OF THE INVENTION iV-Acyl 0-amino phenol derivatives of CBI- TMI and CBI-indole2 are disclosed herein as prototypical members of a new class of reductively activated prodrugs of the duocarmycin and CC- 1065 class of antitumor agents. The expectation being that hypoxic tumor environments, with their higher reducing capacity, carry an intrinsic higher concentration of "reducing" nucleophiles (e.g., thiols) capable of activating such derivatives (tunable N-O bond cleavage) increasing their sensitivity to the prodrug treatment. Preliminary studies indicate the prodrugs effectively release the free drug in functional cellular assays for cytotoxic activity approaching or matching the activity of the free drug, yet remain essentially stable and unreactive to in vitro DNA alkylation conditions (< 0.1-0.01% free drug release), pH 7.0 phosphate buffer, and exhibit a robust half-life in human plasma ( t^ = 3 hours) . Characterization of a representative 0- (acylamino) prodrug in vivo indicates that they approach the potency and exceed the efficacy of the free drug itself (CBI-indole2) indicating that not only is the free drug effectively released from the inactive prodrug, but that they offer additional advantages related to a controlled or targeted release in vivo.
A contemplated compound of the invention is an N-acyl 0-amino CBI derivative that is represented by Formula I :
Figure imgf000009_0001
In Formula I, R1 is selected from the group of radicals consisting of -C(O) (C1-C6 alkyl) , -C(O)O(Ci- C10 alkyl) , -C (0) (C2-C6 alkenyl) , -C(O)O(C2-C6 alkenyl) , and -C(0)aryl. R2 is selected from the group of radicals consisting of hydrogen, -C(O) (Ci-C6 alkyl), -C(O) 0 (Ci-C10 alkyl) , -C (0) (C2-C6 alkenyl) , and -C(O)O(C2-C6 alkenyl) . In the alternative, R1 and R2 are combined to form a cyclic structure selected from the group consisting of divalent radicals represented as follows:
Figure imgf000009_0002
R3 in Formula I is selected from group consisting of radicals represented as follows:
Figure imgf000009_0003
Figure imgf000010_0001
wherein R4 is selected from group consisting of radicals represented as follows:
Figure imgf000010_0002
R5, R6, R7 and R8 in the above structural formulas are each independently selected from the group of radicals consisting of -H, -OH, -0(Ci-C6 alkyl) , - (Ci-C6 alkyl) and halogen. R9 of an above formula is selected from the group of radicals consisting of -H, -C(O)O(C1-C6 alkyl) , -C (0) (Ci-C6 alkyl) , -C(O)NH2, -C(O)NHNH2, and -C(O)NHNHC(O)O(Ci-C6 alkyl) .
In a preferred compound, R1 is selected from the group of radicals consisting of -C(O) (Ci-C6 alkyl) and -C(O)O(Ci-Ci0 alkyl); R2 is selected from the group of radicals consisting of hydrogen (hydrido; -H) , -C(O) (C1-C6 alkyl), and -C(O)O(C1-Ci0 alkyl); or, alternatively, R1 and R2 combine to form a cyclic divalent radical represented by the following structure (phthalyl) :
Figure imgf000010_0003
R3 is selected from the group consisting of the following radicals:
Figure imgf000011_0001
wherein:
R" is
Figure imgf000011_0002
Particularly preferred compounds include those with the following structural formulas:
Figure imgf000011_0003
Figure imgf000011_0004
A process for treating a proliferative disease such as a cancer or leukemia in a mammal is also contemplated. In accordance with that process, an effective amount of a compound of Formula I such as one of the five compounds shown immediately above is administered to a mammal in need thereof . In yet another aspect, the use of a compound of Formula I in the manufacture of a medicament for treating a proliferative disease such as cancer or leukemia is contemplated.
It is noted that in the structural formulas utilized herein that a wavy line indicates a chemical bond to a depicted atom. It is also noted that to improve readability and minimize seeming duplication, any combination of structural elements described broadly can be present in a specific embodiment unless otherwise stated.
BRIEF DESCRIPTION OF DRAWINGS In the drawings forming a portion of this disclosure,
Figure 1 illustrates the results of an electrophoresis gel with 8% denaturing PAGE and autoradiography. Thermally- induced strand cleavage of w794 DNA; DNA-agent incubation at 4 0C for 18 hours, removal of unbound agent by EtOH precipitation, and 30 minutes of thermolysis (100 0C) followed by 8% denaturing PAGE and autoradiography. Lane 1, control DNA; lanes 2-5, Sanger G, C, A, and T sequencing reactions; lanes 6-8, 2 (IxIO""4 to IxIO"6) ; lanes 9-11, 10 (IxIO"1 to IxIO"3) ; lanes 12-14, 4 (IxIO"1 to IxIO"3) , lanes 15-17, 9 (IxIO"1 to IxIO"3) . All compounds possess the natural IS-configuration. The reductively activated agent 4 was found to alkylate w794 DNA with an identical sequence selectivity as the parent agent CBI-TMI (2) , albeit with a substantially reduced efficiency (1,000- 10, 000-fold) . Similarly, the O-methyl ether 10 as well as 9 lacking a C4 substituent failed to exhibit significant observable DNA alkylation.
DETAILED DESCRIPTION OF THE INVENTION A novel set of reductively activated phenol prodrugs of the CC- 1065 and duocarmycin class of compounds is disclosed. These compounds do not require enzymatic release and are illustrative of other phenolic drugs that can benefit from such a designed activation. Alternative and prior efforts at incorporating a reductive activation into the CC-1065 and duocarmycin class includes the Denny disclosures of nitro precursors to aryl amine variants of the phenol precursors (Hay, M. P.; et al . J. Med. Chem. 2003, 46, 5533; Tercel, M.; et al . J. Med. Chem. 2003, 46, 2132; Gieseg, M. A.; et al . Anti-Cancer Drug Design 1999, 14, 77; Hay, M. P.; et al. Bioorg. Med. Chem. Lett. 1999, 9, 2237; Atwell, G. J.; et al. J. Med. Chem. 1999, 42, 3400; Atwell, G. J.; et al. J. Org. Chem. 1998, 63, 9414; Atwell, G. J.; et al. Bioorg. Med. Chem. Lett. 1997, 7, 1493) , Lee's use of an ester subject to cleavage upon a tethered quinone reduction (Townes, H.; et al . Med. Chem. Res. 2002, 11, 248), and a report of mitomycin- like quinone precursors to a reductively activated o-spirocyclization (versus p-spirocyclization) analogous to those observed with the duocarmycins or its analogues (Boger, D. L.; Garbaccio, R. M. J. Org. Chem. 1999, 69, 8350) . Although the approaches have provided some increase in selectivity resulting from reductive activation, none approach that observed with mitomycin and none effectively or clearly utilize an intrinsic enzyme activity that differentiated normal versus tumor cells. Notably, it may be the ease of the mitomycin hydroquinone reoxidation to the quinone in normal cells that protects them from the effects of the drug, which occurs less readily in hypoxic tumors .
The structure of CBI (1,2,9,9a- tetrahydrocyclopropa [c] benz [e] indol-4-one) and its precursor 1 where R is just the DNA binding portion of the molecule along with its precursor, the 0-amino phenol derivative or prodrug that requires reductive activation by N-O bond cleavage are shown below. The CBI compounds are more accessible than the natural products, yet indistinguishable in their DNA alkylation selectivity (Boger, D. L.; Munk, S. A. J. Am. Chem. Soc. 1992, 114, 5487) . Moreover, the CBI derivatives proved to be four times more stable and, correspondingly, four times more potent than derivatives bearing the CC- 1065 alkylation subunit (7-MeCPI) approaching the stability and potency of duocarmycin SA and yatakemycin derivatives, and they exhibit efficacious in vivo antitumor activity in animal models at doses that reflect this potency. Consequently, CBI and its derivatives have been the focus of much development as well as the prototype analogues on which new design concepts have been explored, developed, or introduced, including the instant invention.
Figure imgf000015_0001
cleavage of weak N-O bond
The approach detailed herein was not designed for enzymatic reductive activation, but rather for activation by cleavage of a weak N-O bond by reducing nucleophiles . The expectation of this approach being that hypoxic tumor cells, with their higher reducing capacity, contain an intrinsically higher concentration of "reducing" nucleophiles
(i.e., thiols) capable of activating such derivatives making them more sensitive to the prodrug treatment
(Wolkenberg, S. E.; Boger, D. L. Chem. Rev. 2002, 102, 2477. Reviews on reductive activation: Papadopoulou, M. V.; Bloomer, W. D. Drugs Future 2004, 29, 807; Jaffar, M. ; Stratford, I. J. Exp. Opin. Ther. Patents 1999, 9, 1371; Patterson, L. H. ; Raleigh, S. M. Biomed. Health Res. 1998, 25, 72) . Moreover, as detailed below, the design lends itself to a rational tuning of the ease of reduction of the derivative allowing empirical experience with the series to guide future design.
A contemplated compound of the invention is an N-acyl 0-amino CBI derivative that is represented by Formula I :
Figure imgf000016_0001
Formula I
In Formula I, R1 is selected from the group of radicals consisting of -C(O) (C1-C6 alkyl) , -C(O)O(Ci-Ci0 alkyl) , -C (0) (C2-C6 alkenyl) , -C(O)O(C2-C6 alkenyl) , and -C(0)aryl. R2 is selected from the group of radicals consisting of hydrogen, -C(O) (Cx-C6 alkyl), -C(O)O(Ci-Ci0 alkyl), -C(O) (C2-C6 alkenyl), and -C(O)O(C2-C5 alkenyl) . In the alternative, R1 and R2 are combined to form a cyclic structure selected from the group consisting of divalent radicals represented as follows:
Figure imgf000016_0002
R3 in Formula I is selected from group consisting of radicals represented as follows :
Figure imgf000016_0003
Figure imgf000016_0004
wherein R4 is selected from group consisting of radicals represented as follows :
Figure imgf000017_0001
R5, R6, R7 and R8 in the above structural formulas are each independently selected from the group of radicals consisting of -H, -OH, -0(Ci-C6 alkyl) , - (C1-C6 alkyl) and halogen. R9 of an above formula is selected from the group of radicals consisting of -H, -C(O)O(Ci-C5 alkyl) , -C (0) (C1-C6 alkyl) , -C(O)NH2, -C(O)NHNH2, and -C(O)NHNHC(O)O(Ci-C6 alkyl) .
In any of the Formulas herein, the term "Ci-Cg alkyl" denotes a straight or branched chain radical such as a methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, amyl, tert-amyl, hexyl group and the like.
The term "C2-Cg alkenyl" denotes a radical such as a vinyl, allyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl group and the like, as well as dienes and trienes of straight and branched chains containing up to six carbon atoms and at least one carbon-to-carbon (ethylenic) double bond.
The term "halogen" includes fluoro, chloro, bromo and iodo, with chloro being preferred.
The term "aryl" is meant to include a monocyclic or dicyclic aromatic radical containing 5 to 10 atoms in the ring system and zero, one or three atoms other than carbon in the rings . The atoms other than carbon can be selected from oxygen, nitrogen and sulfur. Illustrative aryl radicals include phenyl, 1- and 2-naphthyl, pyridyl, pyrazinyl, pyrimidyl, imidazyl, thiophenyl, furanyl, pyrrolyl, 1, 3 , 5-triaziyl, 1, 2, 4-triazinyl and 1,2,3- triazinyl, quinazolinyl, quinolinyl, their various positional isomers, and the like.
Pharmaceutical Compositions and Treatment Methods
A pharmaceutical composition for treating A process for treating a proliferative disease such as a cancer or leukemia in a mammal is also contemplated. Such a composition contains a pharmaceutically effective amount of a before- discussed molecule of Formula I dissolved or dispersed in a pharmaceutically acceptable diluent.
A contemplated compound of Formula I can be used in a pharmaceutical composition to treat and preferably kill cancer cells or cells of another proliferative disease such as leukemia in vitro or in vivo in a mammalian subject. Thus, an above composition is contacted with the cells to be treated. The cells so treated are maintained in contact with a compound of Formula I until cleared by the body when in vivo, or for various times as desired in an in vitro study. The treatment is generally repeated several times.
A mammal to which or whom a compound of Formula I composition is administered can be a primate such as a human, an ape such as a chimpanzee or gorilla, a monkey such as a cynomolgus monkey or a macaque, a laboratory animal such as a rat, mouse or rabbit, a companion animal such as a dog, cat, horse, or a food animal such as a cow or steer, sheep, lamb, pig, goat, llama or the like in need of treatment for a cancerous condition.
A contemplated composition is administered to a mammal in need of the medication at an proliferative effective dosage level. That level is typically an amount sufficient to provide about 10 to about 100 μg/kg of body weight to the recipient's plasma or serum, using the molecular weight of the scission-activated duocarmycin-type prodrug Compound 8 itself as the basis for calculation in view of the
Figure imgf000019_0001
different molecular weights of the other prodrug compounds contemplated herein. The amount can vary depending on the recipient and proliferative cell load. Those plasma or serum concentrations can usually be obtained by i.v. administration using a liquid dosage form that contains about 200 mg to about 1000 mg of chimer compound per day. The determination of optimum dosages for a particular situation is within the skill of the art.
A compound of Formula I composition is administered repeatedly, on a schedule adapted for a recipient's cancer load and need, as is well known in the art . Typical administrations are given multiple times within a one month time period, usually followed by a rest period and then further administrations and rest periods until the recipient is free of the disease, or longer for prophylactic purposes .
For preparing pharmaceutical compositions containing a chimer compound of the invention, an inert, pharmaceutically acceptable carrier or diluent is used. The diluent is usually in liquid form.
Liquid pharmaceutical compositions include, for example, solutions suitable for parenteral administration. Sterile water solutions of the active chimer or sterile solutions of the active component in solvents comprising water, ethanol, or propylene glycol are examples of liquid compositions suitable for parenteral administration.
Sterile solutions can be prepared by dissolving the active component in the desired solvent system, and then passing the resulting solution through a membrane filter to sterilize it or, alternatively, by dissolving the sterile compound in a previously sterilized solvent under sterile conditions .
Preferably, the pharmaceutical composition is in unit dosage form. In such form, the composition is divided into unit doses containing appropriate quantities of the active urea. The unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparation, for example, in vials or ampules.
Chemistry Synthesis
A range of methods for direct conversion of a precursor phenol to the corresponding 0-amino phenol were examined (O-amidation) and several routes to the final compounds were explored. It was anticipated that this might best be conducted on a seco-N-Boc-CBI derivative lacking the capabilities of spirocyclization (e.g., 11) . However, the lability of the resulting N-acyl 0-amino phenol derivatives to subsequent chemical transformations proved significant and this approach proved less viable than a surprisingly effective direct O-amidation reaction of seco-CBI-TMI or seco-CBI- indole2.
Schemes IA and IB, below, show the synthesis of the N-acyl 0-amino phenols directly from the precursors 2 and 3. Thus, low temperature phenol deprotonation of 2 (3 equiv of LiHMDS, 0 0C, ether- dioxane) followed by treatment with the amidating reagents TsONHBoc (Greek, C; et al . Bull. Soc. Chim. Fr. 1994, 131, 429) or TsONPhth (Neumann, U. ; Gϋtschow, M. J. Biol. Chem. 1994, 269, 21561) provided 4 and 8 directly in good conversions. Competitive spirocyclization of 2 to CBI-TMI itself was observed if the deprotonation was carried out at higher reaction temperatures or in more polar solvents . It diminished as the solvent polarity was reduced (glyme > THF > dioxane-ether > ether, insoluble) and was less prominent with LiHMDS versus NaHMDS .
In most instances, recovered starting phenol was present in the crude reaction product and was chromatographically close enough to the N-acyl 0-amino phenols that special precautions were taken to ensure its removal. This entailed exposure of the product mixture to conditions that promote deliberate spirocyclization of the seco phenol derivatives [saturated aqueous NaHCO3-THF (1:1), 23 0C, 2 hours (h) ] and subsequent chromatographic separation of the much more polar CBI-TMI or CBI-indole2. N-Acetylation of 4 (Ac2O, cat. DMAP, CH2Cl2, 23 0C, 12 h, 81%) provided 6 and subsequent Boc deprotection (TFA-CH2Cl2 (1:1), 23 0C, 3 hours, 88%) afforded 5. In an analogous manner, seco-CBI-indole2 (3) was directly converted to 8 (45%) upon LiHMDS deprotonation (3 equiv of LiHMDS, ether-dioxane , 0 0C, 30 minutes) and subsequent O-amidation with TsONHBoc (Greek, C. ; et al. Bull. Soc. Chim. Fr. 1994, 131, 429) .
Scheme IA
H Ac
Figure imgf000022_0001
Scheme IB
Figure imgf000023_0001
For comparison purposes, two analogues of seco- CBI-TMI were prepared that are incapable of spirocyclization to CBI-TMI itself. The first incorporates the C4 phenol protected as its methyl ether (10) and second contains no C4 substituent (9) . The former was prepared from 11 (Kastrinsky, D. B . ; Boger, D. L. J. Org. Chem. 2004, 69, 2284) by phenol O-methylation, primary alcohol OTBS deprotection and subsequent conversion to the primary chloride 14, followed by N-Boc deprotection and coupling with 5 , 6 , 7-trimethoxyindole-2-carboxylic acid (15) to provide 10. See, Scheme 2, below.
Scheme 2
Figure imgf000024_0001
K2CO3, MeI 11, R = H MsCI1 LiCI 13, X = OH 100% 12, R = Me 93% 14, X = CI
ChiralCel OD resolution
Figure imgf000024_0002
Throughout this sequence and as a result of the multiple purifications, the chances of residual, contaminant phenol (2) being present in the final product 10 are remote. Nonetheless, because even trace quantities of 2 can be misleadingly detected in the subsequent biological evaluations (e.g., 0.01%), the inactive analogue 9 was also prepared for comparison and by an approach that precludes the presence of such a contaminate phenol because there is no C-4 functionality in the starting material 16.
Thus, following a route analogous to that used for CBI itself (Boger, D. L.; et al . J. Org. Chem. 1992, 57, 2873; Boger, D. L.; McKie, J. A. J. Org. Chem. 1995, 60, 1271; Drost, K. J.; Cava, M. P. J. Org. Chem. 1991, 56, 2240; Aristoff, P. A.; Johnson, P. D. J. Org. Chem. 1992, 57, 6234; Mohamadi, F.; et al . J. Med. Chem. 1994, 37, 232; Ling, L.; et al . Heterocyclic Commun. 1997, 3, 405; Boger, D. L.; et al . Synlett 1997, 515; Boger, D. L. ; et al. Tetrahedron Lett. 1998, 39, 2227; Kastrinsky, D. B.; Boger, D. L. J. Org. Chem. 2004, 69, 2284), 20 was prepared from 16 and converted to 21 enlisting a key 5-exo-trig aryl radical-alkene cyclization (Boger, D. L.; et al . Tetrahedron Lett. 1998, 39, 2227) . See, Scheme 3, below, that also illustrates the synthesis of the analog of CBI-TMI, 9.
Scheme 3
Figure imgf000025_0001
DPPA, 16, R = CO2H (BoC)2O, DMAF 18, R = H t-BuOH 17, R = NHBoc K2CO3, MeOH 19, R = Boc
74% 86%
Figure imgf000025_0002
Compound 20 was converted to 21 enlisting a key 5-exo-trig aryl radical-alkene cyclization (Boger, D. L.; et al . Tetrahedron Lett. 1998, 39, 2227) . The product 21, like 14 (α = 1.19), was chromatographically resolved on a semipreparative ChiralCel OD column (α = 1.42) providing each enantiomer, and 21 was coupled with 5,6,7- tritnethoxyindole-2-carboxylic acid (15) upon N-Boc deprotection to provide 9.
Stability and Reactivity of the N-Acyl O-Amino Phenol Derivatives Clear from efforts directed at their preparation, the N-acyl amino phenol prodrugs displayed a useful range of stability, yet were susceptible to cleavage of the critical N-O bond. As might be anticipated, their relative stability followed the order of 4 > 5 > 6 > 7 with 4 and 5 withstanding even long term storage effectively, but with 7 noticeably deteriorating over time. Derivatives 4 and 6, as well as 7, proved surprisingly robust to acidic conditions (TFA-CH2Cl2, 4 N HCl-EtOAc) , and stable to mild base treatment in nonpolar, aprotic solvents (Et3N or DMAP, CH2Cl2) , but exhibited a diminished stability as the solvent polarity increases: stable to NaHCO3 in THF or THF- H2O, but cleaved in NaHCO3/DMF-H2O or H2O and DBU/CH3CN. Similarly, 4 proved stable in MeOH, but 2 was released slowly upon treatment with NaHCO3 or Na2CO3 in MeOH (2 hours, 23 0C) . Most pertinent to the potential source of cleavage under physiological conditions, 4 was stable to treatment with BnSH in THF (2-72 hours, 23 0C) or MeOH (2-72 hours, 23 0C) , and stable to treatment with BnSH in THF even in the presence of insoluble NaHCO3 (2 hours, 23 0C) , but is cleaved to release 2 upon treatment with BnSH in MeOH in the presence of NaHCO3 (2 hours, 23 0C) . Significantly, the stability of 4 was assessed in pH 7.0 phosphate buffer and within the limits of detection (HPLC, UV) , no significant cleavage of the prodrug was observed over the time monitored (72 hours) . The stability of 4 was monitored in human plasma (50 μg/100 μL, 10% DMSO) in which it displayed a half-life of 3 hours with release of the free drug 2.
Biological Properties Cytotoxic Activity
The 0-amino phenol derivatives bearing the N-O prodrug linkages and the various W-acyl substituents were assayed for cytotoxic activity alongside the parent drugs CBI-TMI (2) (Boger, D. L. ; Yun, W. J. Am. Chem. Soc. 1994, 116, 7996) and mitomycin C (Boger, D. L.; et al . Bioorg. Med. Chem. Lett. 1991, 2, 115; Boger, D. L.; et al . Bioorg. Med. Chem. 1995, 3, 1429) as well as the two control standards 9 and 10 incapable of free phenol release. Three cell lines were examined including a standard L1210 cell line (mouse leukemia) as well as the mitomycin-sensitive (H460, expresses high levels of DT-Diaphorase) and resistant (H596, lacks DT-Diaphorase) non small cell lung cancer (NSCLC) cell lines, with results shown in the Table below.
Natural enantiomer series
Figure imgf000028_0001
IC50 (nM)
Compel, R
L1210 H460 H596 mitomycin C 40 20 5000
9, H >100 >100 >100
10, OMe 50 >100 >100
2, OH 0.04 0.5 5
4, ONHBoc 0.5 1 6
5, ONHAc 0.3 0.7 7
6, ON(Ac)BoC 0.2 0.6 5
7, ONPhth 0.06 0.5 5
Several important trends emerged from these studies. First, the natural enantiomer control standards 9 and 10, incapable of free phenol release, were inactive against all three cell lines (IC50 >100 nM) being >10, 000-fold less active than the free drug 2 (seco-CBI-TMI) . In sharp contrast, the natural enantiomers of the 0-amino phenol prodrugs exhibited potent cytotoxic activity approaching that of the free drug itself (1-0.1 times the activity of 2) indicating its successful release under the assay conditions .
Even more significantly, the relative potency of the prodrugs, when distinguishable, mirrors the expected ease of N-O bond cleavage (e.g. L1210: 7 > 6 > 5 > 4) suggesting fundamental chemical principles can be used to "tune" the reductive free drug release. Provocatively, the potency differences between the free drug 2 and the prodrugs diminish as the hypoxic character of the cell line increases; 4 is 10 -fold less potent than 2 against L1210, but 2 and 4 are essentially equipotent against H460/H596.
More significantly and unlike mitomycin C, this reductive activation is not linked to the expression levels of DT-Diaphorase because 2 and 4-7 remain equipotent in the H460 or H596 cell lines, although H596 is 10-fold less sensitive than H460 to seco-CBI-TMI itself. This result illustrates that DT-Diaphorase is not mediating the reductive release of the drug from the 0-amino phenol prodrugs, indicating that their utility is orthogonal to that of mitomycin. Rather, their behavior is consistent with the suggestion that the activation is nonenzymatic and likely is mediated in situ by appropriate nucleophiles .
Analogous trends are also observed with the CBI-TMI unnatural enantiomers albeit at concentrations that are approximately 100 to 1000- fold higher than that of the natural enantiomers as is seen in the Table below.
Unnatural enantiomer series
Figure imgf000030_0001
IC50CnM)
Compd, R L1210 H460 H596 mitomycin C 40 20 5000
9, H 900 5500 > 10000
1O1 OMe 800 5000 > 10000
2, OH 5 50 300
4, ONHBoc 160 900 6400
5, ONHAc 100 700 6300
6, ON(Ac)BoC 70 600 6300
7, ONPhth 60 600 6000
Especially interesting and exciting was the behavior of the CBI-indole2 prodrug. For this CBI analogue, only the NHBoc derivative was examined because it was the most stable of the W-acyl 0-amino phenol prodrugs examined as is seen from the data below.
Natural enantiomer series
Figure imgf000030_0002
IC50 (nM)
Compd, R L1210 H460 H596 mitomycin C 40 20 5000 3, OH 0.03 0.2 2
8, ONHBoc 0.05 0.3 4
Figure imgf000031_0001
IC50 CnM)
Compd, R L1210 H460 H596 mitomycin C 40 20 5000 3, OH 0.7 6 40
8, ONHBoc 2 10 60
In each cell line examined, the prodrug 8 was essentially equipotent with CBI-indole2 (3) itself, indicating effective release of the free drug under the conditions of the assay. In addition prodrug 8 proved to be exceptionally potent, being 100-1000 times more active than mitomycin C (IC50 = 30-200 pM vs 20-40 nM) and it remained remarkably active against the mitomycin-resistant H596 cell line (IC50 = 4 nM vs 5 μM) . Even the unnatural enantiomer of prodrug 8, which was found to be 10-100 fold less active than the natural enantiomer, proved to be more active than mitomycin C. Given the efficacy of (+) -CBI-indole2 in animal tumor models, (Boger, D. L.; Ishizaki, T.; Sakya, S. M.; Munk, S. A.; Kitos, P. A.; Jin, Q.; Besterman, J. M. Bioorg. Med. Chem. Lett. 1991, I1 115; Boger, D. L.; Yun, W.; Han, N. Bioorg. Med. Chem. 1995, 3, 1429) it was especially interesting to compare 8 with 3 in vivo.
DNA Alkylation Selectivity and Efficiency
The DNA alkylation properties of 4 were examined alongside the parent drug CBI-TMI (2) , and the two control standards 9 and 10 (incapable of spirocyclization) within w794 duplex DNA (Boger, D. L.; et al. Tetrahedron 1991, 47, 2661) for which results for an extensive series of duocarmycin analogues have been reported. The sites of DNA alkylation and its efficiency were directly assessed by thermally- induced singly 5' end-labeled duplex DNA strand cleavage following incubation with the agents (Figure 8, natural enantiomers examined) .
The reductively activated agent 4 was found to alkylate w794 DNA with an identical sequence selectivity as the parent agent CBI-TMI (2) , albeit with a substantially reduced efficiency (1,000-10,000 fold) . Similarly, the 0-methyl ether 10 as well as 9 lacking a C4 substituent failed to exhibit significant observable DNA alkylation. In fact, 9 showed no appreciable DNA alkylation even under forcing conditions (37 0C, 18 hours, data not shown) , whereas the potentially more reactive 0-methyl ether 10 (via assisted phenonium ion formation) displayed perhaps a trace amount of DNA alkylation (<0.01% that of 2) that could be attributed to either its direct, but much less facile, DNA alkylation or contaminant free phenol present in the synthetic sample of 10.
With detection of DNA alkylation by the prodrug 4 at the level observed (0.1-0.01% of 2), one cannot distinguish whether this is due to direct alkylation by 4 itself, trace release of 2 from 4 under the DNA incubation conditions {in situ N-O cleavage) , or attributable to trace contaminate 2 in the synthetic samples of 4. What the results do indicate is that 4 is incapable of significant DNA alkylation in its own right (requires N-O bond cleavage) , and that 4 is essentially stable to the DNA alkylation conditions examined requiring deliberate N-O bond cleavage to initiate effective DNA alkylation. These observations are consistent with the stability of 4 observed in pH 7.0 phosphate buffer. Significantly, the results then suggest that the in vitro cytotoxic activity of 4, and by analogy that of the related 0-amino phenol prodrugs that all approach that of the parent drug CBI-TMI (2) , is derived from in situ intracellular cleavage of the N- 0 bond and productive release of the active drug under the cell culture conditions.
In Vivo Antitumor Activity
The prodrug 8 was examined for in vivo efficacy alongside the parent drug 3 in a standard antitumor model enlisting L1210 murine leukemia implanted i.p. into DBA/2J mice. This model has been reported to respond well to the parent drugs of related compounds (Li, L. H.; et al . Invest. New Drugs 1991, 9, 137) and is a system that collaborators through the years have used to assess an extensive series of (+) -CBI-indole2 analogues. Although not published, these latter studies provided the foundation on which examination of 8 is based.
With use the dose range (10-100 μg/kg) and the dosing schedule (administered three times i.p. on days 1, 5, and 9) found suitable for related parent drugs including (+) -CBI-indole2 (3) (Boger, D. L.; et al. Bioorg. Med. Chem. Lett. 1991, I1 115; Boger, D. L.; et al. Bioorg. Med. Chem. 1995, 3, 1429), the prodrug 8 was examined as is shown in the Table below.
Figure imgf000034_0001
Compound Dose Mean Survival Treated/Control Surviving μg/kg Period (MSP) (MSP x IOO) Mice
(days) none 0 20 100 0/6
8 10 25 120 0/6
8 30 >145 >730 2/6
8 100 >310 >1550 5/6
3 10 34 170 0/6
3 30 >115 >580 1/6
3 100 125 625 0/6
The dose at which a maximal response was observed for 8 corresponded closely to that of
(+) -CBI-indole2 (3) whereas its efficacy was significantly improved. This result indicates that the prodrug 8 (a) efficiently and effectively releases the free drug 3 in the in vivo model
(reductive activation) , and (b) that either the rate of release or the site of release enhances the efficacy of the drug. Moreover, the efficacy of 8 is extraordinary providing 5/6 long-term survivors at 52 weeks (365 days, T/C >1550) at the optimal dosing examined (100 μg/kg) . Notably, little distinction between 3 and 8 was observed at days 30-100 except that the prodrug-treated animals appeared healthier, displaying little or no weight loss that was evident with 3 at the highest dosing.
With the prolonged management of the treated animals herein that exceeded the time frame typically allotted for such an in vivo antitumor assessment, it was observed that the surviving mice at day 90 treated with the free drug 3, but not the prodrug 8, eventually expired due to drug administration related complications. (This appears to arise from damage to the intraperitoneal cavity or its organs that originate with the bolus drug administration.) Although these administration effects would likely be capable of being managed with an optimized dosing schedule, this distinction between 3 and 8 in the long-term cures (>90 days) suggests the prodrug 8 offers significant advantages over the free drug administration.
It is also worth noting that these compounds are extraordinarily potent, requiring less than 1 mg of sample to conduct the entire in vivo antitumor testing, suggesting that clinical supplies of such agents could easily be supplied by chemical synthesis .
Confirming these observations, an analogous antitumor assessment was carried out independently at a second site utilizing a slightly different and harsher protocol for drug administration (neat DMSO vs 30% DMSO in 0.1% glucose) . Although this assessment was terminated after 120 days, it similarly indicates that administration of the prodrug 8 is significantly less toxic than free drug 3, and that it is comparable or superior in terms of reducing deaths due to the disease, tumor counts, and tumor volume as seen from the Table below. Again, 7/10 long-term survivors were observed with prodrug 8 at day 120 at the optimal dosing (60 μg/kg) .
Figure imgf000036_0001
Compound Dose Mean Survival Treated/Control Surviving μg/kg Period (MSP) (MSP x IOO) Mice (days) none 0 22 100 0/10
8 10 >46 >210 2/10
8 30 >51 >232 2/10
8 60 >93 >425 7/10
8 100 >63 >288 3/10
3 10 >60 >271 3/10
3 30 >65 >295 3/10
3 60 >71 >324 3/10
3 100 11 52 0/10
In the above Table, the second column is the dose in rag/kg of body weight of the animal that is administered i.p. (into the intraperitoneal cavity) on days 1, 5, and 9. The surviving mice are the number of mice that are still living after 120 days and the experiment was then terminated.
Experimental DNA alkylation selectivity and efficiency
The DNA alkylation reactions were performed by treatment of 4.5 μL of singly 32P 5' -end- labeled double-stranded w794 DNA (Boger, D. L.; et al . Tetrahedron 1991, 47, 2661) in TE buffer (10 mM Tris, 1 mM EDTA, pH 7.6) with 0.5 μL of agent in EtOH (at the specified concentration) . The samples were incubated for 18 h at 4 °C. Unbound agent was removed by EtOH precipitation of DNA (0.5 μL of 3.0 M sodium acetate and 12.5 μL of cold absolute EtOH) and the solutions were stored at -78 0C for 1 hour or longer. The DNA was pelleted by centrifugation at 4 0C (13000 rpm, 25 minutes) , dried in a Savant Speed Vac concentrator, and resuspended in 5 μL of TE buffer (pH 7.6) . Thermal depurination (3 x 10 minutes at 100 "C) was performed and then 2.5 μL of formamide dye solution was added to the cooled samples . Thermally denatured samples were assayed by gel electrophoresis [8% denaturing gel with 8 M urea in TBE buffer (89 mM Tris-borate, 2 mM EDTA)] followed by autoradiography of the dried gel using Kodak BIOMAX XAR film and a Picker Spectra™ intensifying screen.
Figure imgf000037_0001
16 74% 17
A solution of 2-naphthoic acid (16, 1.5 g,
8.7 mmol) in t-BuOH (50 mL) and toluene (50 mL) was treated with Et3N (1.44 mL, 10 mmol), 3 A molecular sieves (10 g) and diphenyl phosphorylazide (2.1 mL, 10 mmol) . The reaction mixture was warmed at reflux for 24 h and then cooled to 23 °C. The solid was filtered off through Celite and the solvent was removed in vacuo. The residue was dissolved in EtOAc (75 mL) , and the organic phase was washed with 1 N aqueous HCl (50 mL x 2) , saturated aqueous NaHCO3 (50 mL x 2), dried over anhydrous sodium sulfate, and concentrated. Chromatography (SiO2, 10% EtOAc/hexane) afforded 17 as a pale yellow solid (1.56 g, 74%) : ESI-TOF HRMS m/z 266.1150 (M+Na+, Ci5H17NO2 requires 266.1151) .
Figure imgf000038_0001
Compound 17 (1.5 g, 6.2 mmol) was treated with 4 N HCl-EtOAc (50 mL) for 1 hour before the solvent was removed to yield a white powder. The crude HCl salt (790 mg, 5.5 mmol) , and TsOH (170 mg, 1.1 mmol) in THF (50 mL) cooled to 0 0C was treated with NBS (982 mg, 5.5 mmol) in THF (30 mL) , and the solution was allowed to warm to 23 0C. After stirring for 5 hours, the reaction mixture was washed with saturated aqueous NaHCO3 (30 mL x 2) . The organic layer was dried over anhydrous sodium sulfate and was concentrated. Chromatography (SiO2, 10% EtOAc/hexane) afforded 18 (863 mg, 59% for two Steps) : ESI-TOF HRMS m/z 221.9910 (M+H+, C10H8BrN requires 221.9913) .
Figure imgf000038_0002
18 86%
A solution of 18 (800 mg, 3.6 mmol) in CH2Cl2 was treated with Et3N (496 μL, 3.6 mmol) , DMAP (36 mg, 0.36 mmol) , and BOc2O (830 mg, 3.8 mmol) and the reaction mixture was stirred at 55 0C for 36 hours. The reaction mixture was cooled to 23 0C and washed with aqueous 1 N HCl (30 mL x 2) , and saturated aqueous NaHCO3 (30 mL x 2) . The organic layer was dried over anhydrous sodium sulfate, and concentrated. Chromatography (SiO2, 10% EtOAc/hexanes) provided the product as a white solid (1.25 g, 83%) : ESI-TOF HRMS m/z 444.0780 (M+Na+, C2oH24BrN04 requires 444.0781) .
Figure imgf000039_0001
19
A solution of the product above (516 mg, 1.18 mmol) in MeOH (20 mL) was treated with K2CO3 (490 mg, 3.6 mmol), and the resulting mixture was warmed at reflux for 1.5 hours. The reaction mixture was allowed to cool to 23 0C and filtered through Celite to remove solid residue. The solvent was removed to yield 19 as a white solid (448 mg, quant.), which was sufficiently pure to use for next step without further purification: ESI-TOF HRMS m/z 344.0250 (M+Na+, Ci5H15BrNO2 requires 344.0257) .
Figure imgf000039_0002
A solution of 19 (980 mg, 3 mmol) in DMF (20 mL) was treated with NaH (60%, 304 mg, 7.5 mmol) and Bu4NI (11 mg, 0.3 mmol) at 0 0C. After stirring for 15 minutes, 1, 3-dichloropropene (0.8 mL, 9 mmol) was added, and the resulting mixture was warmed to 23 0C and stirred for another 4 hours. The reaction mixture was diluted with EtOAc (50 mL) and washed with saturated aqueous NH4Cl (30 mL x 2) . The organic layer was dried over anhydrous sodium sulfate and concentrated. The crude product 20 was used for the next step without further purification.
A solution of crude 20 (1.0 g, 2.52 mmol) and AIBN (41 mg, 0.25 mmol) in degassed toluene (40 mL) was treated with Bu3SnH (0.75 mL, 2.77 mmol) . The resulting solution was purged with N2 gas for 10 minutes and then warmed at reflux overnight (about 18 hours) . The solvent was removed and the crude product was purified by chromatography (SiO2, 10% EtOAc/hexanes) to yield racemic 21 as a white solid (780 mg, 97%) . The two enantiomers were separated by chromatography (semiprep 2 x 25 cm Chiral OD column, 10% iPrOH/hexanes, flow rate = 0.5 mL/min, tR = 35.5 min (natural), 25.0 min (unnatural), α = 1.42) : ESI- TOF HRMS m/z 340.1076 (M+H+, C18H20ClNO2 requires 340.1075) . 1S-21: [α] 23 D -0.38 (c 0.18, CH3OH), natural enantiomer; li?-21: [α] 23 D +0.46 (c 0.13, CH3OH), unnatural enantiomer.
Figure imgf000040_0001
A sample of 21 (13 mg, 41 μmol) was treated with 4 N HCl-EtOAc (3 mL) for 30 min before the solvent was removed by a stream of N2. The resulting crude HCl salt, 5, 6, 7-trimethoxyindol-2-carboxylic acid (15, 10.3 mg, 41 μmol) and EDCI (24 mg, 0.12 mmol) were dissolved in DMF (3 mL) , and the resulting solution was stirred at 23 0C for 3 hours. The reaction mixture was diluted with EtOAc (15 mL) and washed with aqueous 1 N HCl (5 mL x 2) , and saturated aqueous NaHCO3 (5 mL x 2) . The organic layer was dried over anhydrous sodium sulfate, and concentrated. PTLC (SiO2, 50% EtOAc/hexanes) gave 9 as a white solid (13.6 mg, 74%) : ESI-TOF HRMS m/z 451.1420 (M+H+, C25H23ClN2O4 requires 451.1419) . 1S-9: [α] 23 D -0.26 (c 0.46, THF), natural enantiomer; 1R-9: [α] 23 D +0.27 (c 0.73, THF), unnatural enantiomer.
Figure imgf000041_0001
A solution of 11 (Kastrinsky, D. B.; Boger, D. L. J. Org. Chem. 2004, 69, 2284) (50 mg, 0.116 mmol) , and methyl iodide (14.5 μL, 0.233 mmol) in acetone (12 mL) was treated with K2CO3 (48 mg, 0.349 mmol) at 23 0C, and the resulting mixture was stirred at 23 °C for 3 hours. The reaction was diluted with water (10 mL) and extracted with EtOAc (15 mL x 2) . The combined organic layers were washed with water (15 mL x 2) , saturated aqueous NaCl (15 mL) and dried over anhydrous sodium sulfate. The solvent was removed and the crude product 12 was sufficiently pure for use without further purification (55 mg, quant . ) .
Figure imgf000042_0001
A solution of 12 (51 mg, 0.115 mmol) in THF (5 mL) was treated with Bu4NF (1 M in THF, 575 μL, 0.575 mmol) at 23 0C. After stirring at 23 0C for 1 hour, the reaction mixture was diluted with EtOAc (20 mL) and washed with water (10 mL) , and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated to afford pure 13 (40 mg, quant.) . The above crude compound 13 (40 mg, 0.121 mmol) was dissolved in pyridine (2 mL) . Methanesulfonyl chloride (59 μL, 0.607 mmol) was added at 0 0C. After stirring at 23 0C for 6 hours, the reaction mixture was diluted with EtOAc (20 mL) , and washed with water (10 mL x 2) , and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. The crude residue was dissolved in DMF (2 mL) and was treated with LiCl (26 mg, 0.607 mmol) . After stirring at 23 0C for 3 days, the reaction mixture was diluted with EtOAc (20 mL) and washed with water (10 mL) , saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. Chromatography (SiO2, 20% EtOAc/hexanes) afforded 14 (37.5 mg, 93% for two steps) . The two enantiomers were separated by chromatography (semiprep 2 x 25 cm Chiral OD column, 10% iPrOH/hexanes, flow rate = 0.5 mL/min, tR = 14.4 min (natural), 12.1 min (unnatural), α = 1.19) : IS- 14: [α] 23 D -0.43 (c 0.28, THF), natural enantiomer; Ii?- 14: [α] 23 D +0.45 (c 0.53, THF) , unnatural enantiomer .
Figure imgf000043_0001
A sample of 14 (6.1 mg, 17 μmol) was treated with 4 N HCl-EtOAc (0.6 mL) for 30 minutes before the solvent was removed by a stream of N2. The resulting crude HCl salt, 5, 6, 7-trimethoxyindol-2- carboxylic acid (15, 4.8 mg, 19 μmol) and EDCI (10.1 mg, 0.05 mmol) were dissolved in DMF (0.15 mL) and the resulting solution was stirred at 23 °C for 3 hours. EtOAc (10 mL) was added to the reaction mixture and the resulting solution was washed with aqueous 1 N HCl (5 mL x 2) , saturated aqueous NaHCO3 (5 mL x 2) , dried over anhydrous sodium sulfate and concentrated. PTLC (SiO2, 50% EtOAc/hexanes) gave 10 as a white solid (5.5 mg, 65%) : ESI-TOF HRMS m/z 481.1521 (M+H+, C26H25ClN2O5 requires 481.1525) . lS-10:
23 αj D -0.50 (c 0.31, THF), natural enantiomer; li?-10:
[α] D +0.86 (c 0.14, THF), unnatural enantiomer.
Figure imgf000043_0002
A solution of seco-CBI-TMI (Boger, D.L. ; Yun, W. J. Am. Chem. Soc. 1994, 116, 7996) (2, 30 mg, 0.064 mmol) in ether-dioxane (1:1, 3 mL) was treated with LiHMDS (1 M in THF, 193 μL, 0.193 mmol) at 0 0C, and the resulting mixture was stirred at 0 0C for 30 minutes. The resulting solution was treated with t-butyl-N-tosyloxycarbamate (55 mg, 0.193 mmol) . The reaction mixture was allowed to warm to 23 0C and stirred for an additional 4 hours. The solution was diluted with EtOAc (20 mL) and washed with water (10 mL) , and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. PTLC (SiO2, 50% EtOAc/hexanes) afforded crude product (31.2 mg) . To ensure the complete removal of any 2, the product (12 mg) was dissolved in THF (6 mL) and saturated aqueous NaHCO3 (6 mL) was added. After stirring at 23 0C for 2 hours to promote spirocyclization of any residual 2 to the much more polar and easily separable CBI-TMI, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (10 mL) and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. PTLC (SiO2, 20% EtOAc/hexanes) afforded 4 (6.6 mg, 46%) as a pale yellow solid: ESI-TOF HRMS m/z 582.2000 (M+H+, C30H32ClN3O7 requires 582.2001) . 1S-4: [α] 23 D -0.39 (c 0.31, THF) , natural enantiomer; 1R-4: [α] 23 D +0.68 (c 0.44, THF) , unnatural enantiomer.
Figure imgf000044_0001
A solution of 4 (3.4 mg, 0.00584 mmol) in CH2Cl2 (0.34 mL) was treated with acetic anhydride (2.7 μL, 0.0292 mmol), Et3N (4.1 μL, 0.0292 mmol) and DMAP (cat) . After the resulting mixture was stirred at 23 0C for 12 hours, the solvent was removed and the residue was purified by PTLC (SiO2, 50% EtOAc/hexanes) to afford 6 (2.9 mg, 81%) : ESI-TOF HRMS m/z 642.2102 (M+H+, C32H34ClN3O8 requires 642.2107) . 1S-6: [α] 23 D -0.43 (c 0.23, THF), natural enantiomer; Ii?- 6: [α] 23 D +0.54 (c 0.52, THF), unnatural enantiomer.
Figure imgf000045_0001
A solution of 6 (3.1 mg, 0.0053 mmol) in CH2Cl2 (1 mL) was treated with TFA (1 mL) at 23 0C for 3 hours. The solvent and excess TFA were removed and the residue was purified by PTLC (SiO2, 50% EtOAc/hexanes) to afford 5 (2.3 mg, 88%) : ESI-TOF HRMS m/z 522.1431 (M-H", C27H26ClN3O6 requires 522.1437) . 1S-5: [α]23 D -1.2 (c 0.10, THF), natural enantiomer; 1R-5: [α] 23 D +0.76 (c 0.21, THF), unnatural enantiomer.
Figure imgf000046_0001
A solution of seco-CBI-TMI (2, 5.0 mg, 0.011 mmol) in THF (0.5 mL) was treated with LiHMDS (1 M in THF, 13 μL, 0.013 mmol) at -78 0C, and the resulting mixture was stirred at -78 0C for 30 minutes. The resulting solution was treated with N- p-tolylsulfonyloxyphthalimide (5.1 mg, 0.016 mmol) . The reaction mixture was stirred at 23 0C for an additional 60 minutes. The solution was diluted with EtOAc (10 mL) and washed with water (5 mL) , and saturated aqueous NaCl (5 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. PTLC (SiO2, 50% EtOAc/hexanes) afforded 7 (4.6 mg, 70%) as a pale yellow solid: 1S-7: [α] 23 D -0.42 (c 0.28, THF), natural enantiomer; Ii?-7: [α] 23 D +0.53 (c 0.36, THF), unnatural enantiomer.
Figure imgf000046_0002
A solution of seco-CBI-indole2 (Boger, D. L.; Yun, W.; Han, N. Bioorg. Med. Chem. 1995, 3, 1429) (3, 16.5 mg, 0.031 mmol) in THF (1.5 mL) was treated with LiHMDS (1 M in THF, 93 μh, 0.093 mmol) at 0 0C and the mixture was stirred at 0 0C for 30 minutes. The resulting solution was treated with t- butyl-N-tosyloxycarbamate (26.6 mg, 0.093 mmol), and the reaction mixture was allowed to warm to 23 0C and stirred for an additional 4 hours. The solution was diluted with EtOAc (20 mL) and washed with water (10 mL) , and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. PTLC (SiO2, 50% THF/hexanes) afforded 8 (12.0 mg) . The product (12 mg) was dissolved in THF (6 mL) and treated with saturated aqueous NaHCO3 (6 mL) to promote the spirocyclization of any residual 3. After stirring at 23 0C for 2 hours, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (10 mL) and saturated aqueous NaCl (10 mL) . The organic layer was dried over anhydrous sodium sulfate and concentrated. PTLC (SiO2, 20% THF/hexanes) afforded 8 (9.0 mg, 45%) : ESI-TOF HRMS m/z 650.2150 (M+H+, C36H32ClN5O5 requires 650.2165) . 1S-8: [α] 23 D +2.1 (c 0.50, THF), natural enantiomer; IiR-8: [α] 23 D - 2.0 ( c 0.89, THF), unnatural enantiomer.
In Vivo Antitumor Activity DBA/2J mice were purchased from Jackson Laboratory (Bar Harbor, ME) and housed in the animal facility at The Scripps Research Institute. L1210 tumor cells, originally isolated from DBA/2 mice, were cultured in DMEM medium containing 5% fetal calf serum. For tumor implantation, DBA/2J mice were i.p. injected with 1 x 105 L1210 cells at day 0.
Compounds 3 and 8 were formulated with 30% DMSO plus 0.1% glucose solution. Treatment doses of drugs (0, 10, 30, 100 lg/kg wt . of animal) were i.p. injected consecutively on day 1, 5 and 9. The study was performed with six mice per group. Tumor growth in the peritoneal cavity was monitored daily and the death of animals was recorded. If necessary, weights of animals were measured once a week. To date this monitoring of the animals has lasted 365 days.
An analogous study with 10 mice per group was performed at the University of Kansas with the distinction that the compounds were administered in neat DMSO (0, 10, 30, 60, 100 lg/kg wt . of animal) and the study was terminated after 120 days.
Each of the patents, patent applications and articles cited herein is incorporated by reference. The use of the article "a" or "an" is intended to include one or more.
The foregoing description and the examples are intended as illustrative and are not to be taken as limiting. Still other variations within the spirit and scope of this invention are possible and will readily present themselves to those skilled in the art .

Claims

WHAT IS CLAIMED IS:
1. An N-acyl O-amino CBI derivative represented by Formula I :
Figure imgf000049_0001
Formula I
wherein :
R1 is selected from the group of radicals consisting of -C(O) (C1-C6 alkyl) , -C(O)O(Ci-C10 alkyl) , -C(O) (C2-C6 alkenyl) , -C(O)O(C2-C6 alkenyl) , and -C(0)aryl;
R2 is selected from the group of radicals consisting of hydrogen, -C(O) (C1-C6 alkyl), -C(O)O(C1-C10 alkyl), -C(O) (C2-C6 alkenyl), and -C(O)O(C2-C6 alkenyl); or, alternatively,
R1 and R2 are combined to form a cyclic structure selected from the group consisting of divalent radicals represented as follows:
Figure imgf000049_0002
R3 is selected from group consisting of radicals represented as follows:
Figure imgf000050_0001
Figure imgf000050_0002
wherein:
R4 is selected from group consisting of radicals represented as follows:
Figure imgf000050_0004
R5, R6, R7 and R8 are each independently selected from the group of radicals consisting of -H, -OH, -0(Ci-C5 alkyl) , -(C1-C6 alkyl) and halogen; and
R9 is selected from the group of radicals consisting of -H, -C(O)O(C1-C6 alkyl), -C(O) (C1-C6 alkyl) , -C(O)NH2, -C(O)NHNH2, and -C(O)NHNHC(O)O(C1-C6 alkyl) .
2. An N-acyl 0-amino CBI derivative according to claim 1, wherein R1 is selected from the group of radicals consisting of -C(O) (C1-C6 alkyl) and -C(O)O(C1-C10 alkyl); R2 is selected from the group of radicals consisting of hydrogen, -C(O) (C1-C6 alkyl), and -C(O)O(C1-Ci0 alkyl) ; or, alternatively, R1 and R2 can combine to form a cyclic divalent radical represented by the following structure:
Figure imgf000051_0001
R3 is selected from the group consisting of the following radicals:
Figure imgf000051_0002
wherein:
R4 is represented by the following structure:
Figure imgf000051_0003
3. An N-acyl 0-amino CBI derivative according to claim 2 represented by the following structure:
Figure imgf000051_0004
4. An N-acyl 0-amino CBI derivative according to claim 2 represented by the following structure :
Figure imgf000052_0001
5. An N-acyl 0-amino CBI derivative according to claim 2 represented by the following structure:
Figure imgf000052_0002
6. An N-acyl 0-amino CBI derivative according to claim 2 represented by the following structure :
Figure imgf000052_0003
7. An N-acyl 0-amino CBI derivative according to claim 2 represented by the following structure :
Figure imgf000053_0001
8. A process for treating a proliferative disease in a mammal comprising the step of administering an effective amount of a compound of claim 1.
PCT/US2008/083433 2007-11-13 2008-11-13 Cbi derivatives subject to reductive activation WO2009064908A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
ES08850632.4T ES2441115T3 (en) 2007-11-13 2008-11-13 CBI derivatives subject to reductive activation
CA2723883A CA2723883C (en) 2007-11-13 2008-11-13 Cbi derivatives subject to reductive activation
US12/742,616 US8377981B2 (en) 2007-11-13 2008-11-13 CBI derivatives subject to reductive activation
EP08850632.4A EP2227087B1 (en) 2007-11-13 2008-11-13 Cbi derivatives subject to reductive activation

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US98764707P 2007-11-13 2007-11-13
US297807P 2007-11-13 2007-11-13
US60/987,647 2007-11-13
US61/002,978 2007-11-13

Publications (1)

Publication Number Publication Date
WO2009064908A1 true WO2009064908A1 (en) 2009-05-22

Family

ID=40639127

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2008/083433 WO2009064908A1 (en) 2007-11-13 2008-11-13 Cbi derivatives subject to reductive activation
PCT/US2008/083439 WO2009064913A1 (en) 2007-11-13 2008-11-13 Chimer containing a targeting portion linked to a scission-activated duocarmycin-type prodrug

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2008/083439 WO2009064913A1 (en) 2007-11-13 2008-11-13 Chimer containing a targeting portion linked to a scission-activated duocarmycin-type prodrug

Country Status (5)

Country Link
US (1) US8377981B2 (en)
EP (1) EP2227087B1 (en)
CA (1) CA2723883C (en)
ES (1) ES2441115T3 (en)
WO (2) WO2009064908A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011133039A2 (en) 2010-04-21 2011-10-27 Syntarga B.V. Novel conjugates of cc-1065 analogs and bifunctional linkers
US8889868B2 (en) 2008-11-03 2014-11-18 Syntarga Bv CC-1065 analogs and their conjugates
US9421278B2 (en) 2014-01-10 2016-08-23 Synthon Biopharmaceuticals B.V. Duocarmycin ADCS showing improved in vivo antitumor activity
US9427480B2 (en) 2014-01-10 2016-08-30 Synthon Biopharmaceuticals B.V. Duocarmycin ADCs for use in treatment of endometrial cancer
US9901567B2 (en) 2007-08-01 2018-02-27 Syntarga B.V. Substituted CC-1065 analogs and their conjugates
US10266606B2 (en) 2014-01-10 2019-04-23 Synthon Biopharmaceuticals B.V. Method for purifying Cys-linked antibody-drug conjugates

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2868447C (en) * 2012-03-30 2020-09-15 The Scripps Research Institute Cyclic prodrugs of duocarmycin analogs
AU2014241731B2 (en) * 2013-03-28 2017-08-31 The Scripps Research Institute Cyclic N-acyl O-amino phenol CBI derivative

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040002528A1 (en) * 1995-10-03 2004-01-01 The Scripps Research Institute CBI analogs of CC-1065 and the duocarmycins
US20050026987A1 (en) * 2003-05-13 2005-02-03 The Scripps Research Institute CBI analogues of the duocarmycins and CC-1065

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5502037A (en) * 1993-07-09 1996-03-26 Neuromed Technologies, Inc. Pro-cytotoxic drug conjugates for anticancer therapy
US6756397B2 (en) * 2002-04-05 2004-06-29 Immunogen, Inc. Prodrugs of CC-1065 analogs

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040002528A1 (en) * 1995-10-03 2004-01-01 The Scripps Research Institute CBI analogs of CC-1065 and the duocarmycins
US20050032860A1 (en) * 1995-10-03 2005-02-10 The Scripps Research Institute CBI analogs of CC-1065 and the duocarmycins
US20050026987A1 (en) * 2003-05-13 2005-02-03 The Scripps Research Institute CBI analogues of the duocarmycins and CC-1065

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2227087A4 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9901567B2 (en) 2007-08-01 2018-02-27 Syntarga B.V. Substituted CC-1065 analogs and their conjugates
US9815784B2 (en) 2008-11-03 2017-11-14 Syntarga B.V. CC-1065 analogs and their conjugates
US8889868B2 (en) 2008-11-03 2014-11-18 Syntarga Bv CC-1065 analogs and their conjugates
EP3056203A1 (en) 2010-04-21 2016-08-17 Syntarga B.V. Conjugates of cc-1065 analogs and bifunctional linkers
EP3108886A2 (en) 2010-04-21 2016-12-28 Syntarga B.V. Conjugates of cc-1065 analogs and bifunctional linkers
US9629924B2 (en) 2010-04-21 2017-04-25 Syntarga Bv Conjugates of CC-1065 analogs and bifunctional linkers
WO2011133039A2 (en) 2010-04-21 2011-10-27 Syntarga B.V. Novel conjugates of cc-1065 analogs and bifunctional linkers
JP2013525347A (en) * 2010-04-21 2013-06-20 シンタルガ・ビーブイ Novel complex and bifunctional linker of CC-1065 analogue
US11052155B2 (en) 2010-04-21 2021-07-06 Syntarga Bv Conjugates of CC-1065 analogs and bifunctional linkers
US9421278B2 (en) 2014-01-10 2016-08-23 Synthon Biopharmaceuticals B.V. Duocarmycin ADCS showing improved in vivo antitumor activity
US9427480B2 (en) 2014-01-10 2016-08-30 Synthon Biopharmaceuticals B.V. Duocarmycin ADCs for use in treatment of endometrial cancer
US10092659B2 (en) 2014-01-10 2018-10-09 Synthon Biopharmaceuticals B.V. Duocarmycin ADCs for use in treatment of endometrial cancer
US10266606B2 (en) 2014-01-10 2019-04-23 Synthon Biopharmaceuticals B.V. Method for purifying Cys-linked antibody-drug conjugates
US10603387B2 (en) 2014-01-10 2020-03-31 Synthon Biopharmaceuticals B.V. Duocarmycin ADCs showing improved in vivo antitumor activity
US11382982B2 (en) 2014-01-10 2022-07-12 Byondis B.V. Duocarmycin ADCs showing improved in vivo antitumor activity

Also Published As

Publication number Publication date
WO2009064913A1 (en) 2009-05-22
CA2723883C (en) 2014-10-28
ES2441115T3 (en) 2014-01-31
EP2227087A4 (en) 2011-10-12
CA2723883A1 (en) 2009-05-22
US8377981B2 (en) 2013-02-19
EP2227087B1 (en) 2013-10-09
EP2227087A1 (en) 2010-09-15
US20110112163A1 (en) 2011-05-12

Similar Documents

Publication Publication Date Title
EP2227087B1 (en) Cbi derivatives subject to reductive activation
US7268143B2 (en) Isoquinoline derivatives and methods of use thereof
USRE42152E1 (en) Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity
CA2829939C (en) Tricyclic gyrase inhibitors
US5597831A (en) 6-[X-(2-hydroxyethyl) aminoalkyl]-5,11-dioxo-5,6-dihydro-11-H-indeno[1,2-c]isoquinolines and their use as antineoplastic agents
JPH06503095A (en) Tricyclic polyhydroxy tyrosine kinase inhibitor
CN1880306A (en) Substituted indeno[1,2-c]isoquinoline derivatives and methods of use thereof
JP4862120B2 (en) Benz-indole and benzo-quinoline derivatives as prodrugs for tumor therapy
JPH0755923B2 (en) Pharmaceutical composition
US20050222418A1 (en) Novel tyloindicines and related processes, pharmaceutical compositions and methods
AU2013239962B2 (en) Cyclic prodrugs of duocarmycin analogs
US7999101B2 (en) Cationic lipids for the transfection of nucleic acids
US7919500B2 (en) Bis-carbazole DNA intercalating agents for antitumor therapy
EP2978760B1 (en) Cyclic n-acyl o-amino phenol cbi derivative
US6998413B1 (en) Treatment of neoplasms with yujungamycins
CZ232695A3 (en) Indolinesulfonamides and substituted indolesulfonamide derivatives, process of their preparation and anti-tumor preparations
CN113831346A (en) Multi-target anti-tumor small molecule and derivative, preparation method, pharmaceutical composition and application thereof
CA2167504A1 (en) 1,2-benzoquinones

Legal Events

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

Ref document number: 08850632

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12742616

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008850632

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2723883

Country of ref document: CA