WO2000012507A2 - Pyrrolobenzodiazepines - Google Patents

Pyrrolobenzodiazepines Download PDF

Info

Publication number
WO2000012507A2
WO2000012507A2 PCT/GB1999/002837 GB9902837W WO0012507A2 WO 2000012507 A2 WO2000012507 A2 WO 2000012507A2 GB 9902837 W GB9902837 W GB 9902837W WO 0012507 A2 WO0012507 A2 WO 0012507A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
mmol
group
substituents
Prior art date
Application number
PCT/GB1999/002837
Other languages
French (fr)
Other versions
WO2000012507A8 (en
WO2000012507A3 (en
Inventor
David Edwin Thurston
Philip Wilson Howard
Original Assignee
Spirogen Limited
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 Spirogen Limited filed Critical Spirogen Limited
Priority to DK99941766T priority Critical patent/DK1109811T3/en
Priority to DE69910227T priority patent/DE69910227T2/en
Priority to AT99941766T priority patent/ATE246687T1/en
Priority to JP2000571053A priority patent/JP4669611B2/en
Priority to EP99941766A priority patent/EP1109811B1/en
Priority to AU55261/99A priority patent/AU758398B2/en
Priority to NZ510492A priority patent/NZ510492A/en
Priority to CA002341968A priority patent/CA2341968C/en
Priority to US09/763,814 priority patent/US6562806B1/en
Publication of WO2000012507A2 publication Critical patent/WO2000012507A2/en
Publication of WO2000012507A3 publication Critical patent/WO2000012507A3/en
Publication of WO2000012507A8 publication Critical patent/WO2000012507A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present invention relates to pyrrolobenzodiazepines (PBDs), and is particularly concerned with the use of these compounds as prodrugs for antibody-directed enzyme-prodrug therapy (ADEPT), gene-directed enzyme-prodrug therapy (GDEPT) , photodynamic therapy (PDT) and naturally present enzyme-prodrug therapy (NPEPT) .
  • ADPT antibody-directed enzyme-prodrug therapy
  • GDEPT gene-directed enzyme-prodrug therapy
  • PDT photodynamic therapy
  • NPEPT naturally present enzyme-prodrug therapy
  • PBDs Pyrrolobenzodiazepines
  • PuGPu Purine-Guanine-Purine
  • Family members include abbeymycin (Hochlowski et al., 1987 J. Antibiotics, 40, 145-148), chicamycin (Konishi et al., 1984 J. Antibiotics, 37, 200-206), DC-81 (Japanese Patent 58-180 487; Thurston et al., 1990, Chem. Brit., 26, 767-772; Bose et ai . , 1992 Tetrahedron, 48, 751-758), mazethramycin (Kuminoto et al., 1980 J. Antibiotics, 33, 665-667), neothramycins A and B (Takeuchi et al., 1976 J.
  • PBDs are of the general structure: O 00/12507
  • prodrugs represents a very valuable clinical concept in cancer therapy.
  • a prodrug may be converted into an antitumour agent under the influence of an enzyme that is linked to a monoclonal antibody so that it can bind to a tumour associated antigen.
  • an enzyme that is linked to a monoclonal antibody so that it can bind to a tumour associated antigen.
  • the combination of such a prodrug with such an enzyme monoclonal antibody conjugate represents a very powerful therapeutic strategy.
  • This approach to cancer therapy often referred to as "antibody directed enzyme/prodrug therapy" (ADEPT) is disclosed in WO88/07378.
  • ADPT antibody directed enzyme/prodrug therapy
  • VDEPT virus-directed enzyme prodrug therapy
  • Tumour cells are targeted with a viral vector carrying a gene encoding an enzyme capable of activating a prodrug.
  • the gene may be transcriptionally regulated by tissue specific promoter or enhancer sequences.
  • the viral vector enters tumour cells and expresses the enzyme, thereby converting the prodrug into the active drug within the tumour cells (Huber et al . , Proc. Na tl . Acad. Sci . USA (1991) 88. ⁇ 8039).
  • non-viral methods for the delivery of genes have been used.
  • GDEPT gene-directed enzyme prodrug therapy
  • Photodynamic therapy provides another method which uses prodrugs to deliver desired drugs to specific sites in the human body. Advances in the field of light delivery to internal areas of the body allow delivery to organs and other areas without the need for any extensive surgical procedures.
  • the activation process can be extremely site specific, as the direction of a laser beam can be controlled with great precision, and the beam diameter can be reduced far below that of a single cell, minimising any possible damage to other neighbouring tissue from unwanted activation of the drug.
  • the high energy of ultra-violet light e.g. 350 nm equivalent to 340 kJ/mol
  • a further class of prodrugs is those where the protecting group is removed by an enzyme naturally present at the desired site of action.
  • These enzymes include dopa-decarboxylase, L- ⁇ -glutamyl transpeptidase, and mixed function oxidases and reductase (e.g. DT-diaphrase) .
  • This is method termed "naturally present enzyme- prodrug therapy (NPEPT) in this application.
  • NPEPT enzyme- prodrug therapy
  • One enzyme of particular interest is glutathione transferase (GST) , which forms part of a major cellular defence mechanism based on the use of the tripeptide, glutathione, as a scavenger of toxic electrophiles .
  • GST acts as a catalyst in the reaction between glutathione and its target electrophiles.
  • Prodrugs which make use of this elevated GST level have been made (Satyam et al . , Med. Chem. 1996, 39, 1736-1747). They have a glutathione molecule linked via a 2-sulphonylethyloxycarbonyl linker to a phosphorodiamidate mustard.
  • An alternative type of prodrug has made use of the closely related 2- phenylsulphonylethyloxycarbonyl (Psec) group (Nicolaou et al . ,
  • Such prodrugs showed selectivity between healthy human bone marrow cells and promeocytic and T cell leukemia tumour lines.
  • R 10 is a therapeutically removable nitrogen protecting group
  • R CH 2 , CH 2 -C0 2 R, CH 2 -C0 2 H, CH 2 -S0 2 R, 0-S0 2 -R, C0 2 R, COR and CN;
  • R ⁇ , R 7 and R 9 are independently selected from H, R, OH, OR, halo, amino, nitro, Me 3 Sn; or R 7 and R 8 together form a group
  • X is S, O or NH
  • Ri is either H or R; where R is a lower alkyl group having 1 to 10 carbon atoms, or an aralkyl group (i.e. an alkyl group with one or more aryl substituents ) , preferably of up to 12 carbon atoms, whereof the alkyl group optionally contains one or more carbon-carbon double or triple bonds, which may form part of a conjugated system, or an aryl group, preferably of up to 12 carbon atoms; and is optionally substituted by one or more halo, hydroxy, amino, or nitro groups, and optionally contains one or more hetero atoms, which may form part of, or be, a functional group; and where there is optionally a double bond between Cl and C2 or C2 and C3; and R 8 is selected from H, R, OH, OR, halo, amino, nitro, Me 3 Sn, where R is as defined above, or the compound is a dimer with each monomer being the same or different and being of formula I, where the R 8
  • R is an aryl group, and contains a hetero atom
  • R is a heterocyclic group.
  • the hetero atom may be located anywhere in the alkyl chain, e.g. -0-C 2 H 5 , -CH 2 -S-CH 3 , or may form part of, or be, a functional group, e.g. carbonyl, hydroxy.
  • R is preferably independently selected from a lower alkyl group having 1 to 10 carbon atoms, or an aralkyl group, preferably of up to 12 carbon atoms, or an aryl group, preferably of up to 12 carbon atoms, optionally substituted by one or more halo, hydroxy, amino, or nitro groups. It is more preferred that R groups are independently selected from a lower alkyl group having 1 to 10 carbon atoms optionally substituted by one or more halo, hydroxy, amino, or nitro groups. It is particularly preferred that R groups are unsubstituted straight or branched chain alkyl groups, having 1 to 10, preferably 1 to 6, and more preferably 1 to 4, carbon atoms, e.g. methyl, ethyl, propyl, butyl.
  • R 6 , R 7 , R 8 , and R 9 may preferably be independently selected from R groups with the following structural characteristics:
  • 'electron sink' means a moiety covalently attached to a compound which is capable of reducing electron density in other parts of the compound.
  • electron sinks include cyano, carbonyl and ester groups.
  • 'therapeutically removable nitrogen protecting group means any group which can protect the 10-nitrogen, but which is removable under therapeutic conditions in vivo, that is, removable under conditions which occur or can be caused to occur in vivo and are medically acceptable generally by elimination to produce a N10-C11 imine group or an equivalent, capable of interacting with DNA. The removal of the protecting group should leave the rest of the structure of the PBD unaffected.
  • Suitable removal techniques include applying light, e.g. with a wavelength of 250 to 400, or 550 nm, changing the ambient pH, or cleavage by the action of an enzyme.
  • One particularly suitable enzyme is nitroreductase, although other suitable enzymes include penicillin V/G amidase, ⁇ -lactamase, phosphatase, L- ⁇ -glutamyl transpeptidase, and ⁇ -galactosidase .
  • the action of some of these enzymes is described in Jungheim, L.N. and Shepherd, T.A., Design of Antitumour Prodrugs: Substrates for Antibody Targeted Enzymes, Am. Chem . Soc . Chem . Rev. , 1994, 94: 6, 1553 - 1566.
  • Another particularly suitable enzyme is glutathiffe transferase, as discussed above.
  • R 10 of the formula II is R 10 of the formula II:
  • R (I) is H or R
  • R (II) is one or more optional substituents independently selected from N0 2 , OR, or R, where R is as defined in any of the definitions above; and if two substituents R (II) are on adjacent atoms, they may together be of the formula -O- (CH 2 ) m -0-, where m is 1 or 2.
  • R (II) is preferably N0 2 .
  • therapeutically removable group R 10 is one which is susceptible to nitroreductase, it may be of the formula III:
  • R (III) is one or more optional substituents independently selected from N0 2 , OR or R, where R is as defined in any of the definitions above, and if two substituents R (ni) are on adjacent atoms, they may together be of the formula - O - (CH 2 ) m -0-, where m is 1 or 2.
  • R 10 Another possible therapeutically removable nitrogen protecting group, R 10 , is of the formula XI:
  • R is as defined in any of the definitions above and n is 0 to 3, preferably 0.
  • R is most preferably a phenyl group, substituted or unsubstituted.
  • This protecting group may be removable by the action of glutathione transferase (GST) , which is present at high levels in many human tumour cells (see above) .
  • R 6 and R 9 are H, and further preferred that R 7 and R a are independently selected from H, OH, and OR. It is further preferred that R 2 and R 3 are H.
  • the dimer bridge may be of the formula -O- (CH 2 ) p -0-, where p is from 1 to 12, preferably 3 to 9. Further, R 6 and R 9 are preferably H, and R 7 is preferably independently selected from H, OH, and OR.
  • a product in which XR n is OR may be prepared by direct etherification of compound la.
  • a product in which X is S may be prepared by treatment of compound la with R U SH and a catalyst (generally a Lewis acid such as A1 2 0 3 ) .
  • a product in which X is NH may be prepared by treatment of compound la with an amine R ln NH and a catalyst (generally a Lewis acid such as A1 2 0 3 ) .
  • a third aspect of the present invention provides a method of preparing a compound of formula la as described in the second aspect of the invention, by the oxidation of a compound of formula IVa:
  • a fourth aspect of the present invention provides a method of preparing a compound of formula IVa as described in the third aspect of the invention, by reacting a compound of formula Va:
  • the compound of formula VI is a haloformate of the formula Via:
  • the compound of formula VI is a haloformate of the formula VIb: O 00/12507
  • a fifth aspect of the present invention provides an alternative synthesis of a compound of formula IVa as described in the third aspect of the invention, by reacting a compound of formula VII:
  • a sixth aspect of the present invention provides a method of preparing a compound of formula la as described in the second aspect of the invention, by the unmasking of a compound of formula IVb:
  • a seventh aspect of the present invention provides a method of preparing a compound of formula IVb as described in the sixth aspect of the invention, by reacting a compound of formula Vb :
  • the compound of formula VI is a haloformate of the formula Via:
  • An eighth aspect of the present invention provides an alternative synthesis of a compound of formula IVb as described in the second aspect of the invention, by reacting a compound of formula VII:
  • a ninth aspect of the present invention provides a method of making a compound of formula X:
  • a tenth aspect of the present invention provides a use of a compound of formula I, wherein the therapeutically removable nitrogen protecting group (R 10 )is enzyme labile, in conjunction with an appropriate enzyme in methods of ADEPT or GDEPT therapy. If the enzyme labile group is susceptible to nitroreductase, then compounds of formula I, may be used in conjunction with nitroreductase enzymes (for example, those isolated from E. coli ) in methods of ADEPT and GDEPT therapy.
  • An eleventh aspect of the present invention provides a use of a compound of formula I, wherein the therapeutically removable nitrogen protecting group (R 10 )is photolabile, in conjunction with light of wavelengths between 250 and 400 or 550 nm in methods of PDT.
  • a twelfth aspect of the invention provides a use for a compound of formula I, where the therapeutically removable nitrogen protecting group (R 10 ) is labile by conditions occurring naturally at specific localised sites in the patient in therapy.
  • Suitable compounds of formula I may be those susceptible to a nitroreductase enzyme when used to treat hypoxic tumour cells, or those susceptible to enzymes which are naturally occurring at specific localised sites, such as glutathione transferase.
  • the drug produced by the cleavage of the therapeutically removable nitrogen protecting group may be used for treating cancers or other site-specific diseases where a local increase of toxicity is beneficial to the patient.
  • Cancers that may be treated are solid cancers including ovarian, colonic cancer, renal, breast and bowel CNS, melanoma, as well as leukemias .
  • Such drugs may also be suitable for treating bacterial, viral or parasitic infections by exploiting a unique enzyme produced at the site of the infection which is not natural to the host, or by exploiting an elevation in the amount of an enzyme which does naturally occur in the host.
  • a thirteenth aspect of the present invention is a pharmaceutical composition comprising a compound of formula I as described in the first aspect of the invention.
  • Pharmaceutical compositions according to the present invention, and for use in accordance with the present invention may comprise, in addition to the active ingredient, i.e. a compound of formula I, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous or intravenous.
  • compositions for oral administration may be in tablet, capsule, powder or liquid form.
  • a tablet may comprise a solid carrier or an adjuvant.
  • Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • Capsules may comprise a solid carrier such as gelatin.
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • Suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection.
  • Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
  • a fourteenth aspect of the present invention provides the use of a compound of formula I as described in the first aspect of the invention, to prepare a medicament for the treatment of neoplastic disease or other site-specific diseases where a local increase of toxicity is beneficial to the patient.
  • the compound of formula I may be provided together with a pharmaceutically acceptable carrier or diluent.
  • the preparation of a medicament is described in relation to the thirteenth aspect of the invention.
  • Fig. 1 is a synthesis scheme according to the present invention
  • Figs. 2a & 2b are a synthesis scheme for dimers according to the present invention
  • Fig. 3 is a synthesis scheme showing an alternative cyclisation, for use in the present invention
  • Fig. 4 is a graph illustrating the cytotoxicity results for prodrug compound 7 (see example 1), both before and after activation with nitroreductase/NADH in SW1116 cells and LS174T cells
  • Fig. 5 is a graph illustrating the percentage of compound 11 (see example 3) cleaved by UVA (365 nm) exposure over a 3 hour time course;
  • Fig. 6 is a graph illustrating the in vi tro cytotoxicity (IC S0 ; ⁇ M) of benzyl DC-81 and compound 11, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in DMF at 1 mM initial drug concentration;
  • Fig. 7 is a graph illustrating the in vi tro cytotoxicity (IC S0 ; ⁇ M) of benzyl DC-81 and compound 11, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in DMF at 10 mM initial drug concentration;
  • Fig. 8 is a graph illustrating the in vi tro cytotoxicity (IC 50 ; ⁇ M) of benzyl DC-81 and compound 11, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in methanol at a 1 mM initial drug concentration
  • Fig. 9 is a graph illustrating the in vi tro cytotoxicity (IC 50 ; ⁇ M) of DSB-120 and compound 32, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in methanol at a 1 mM initial drug concentration.
  • a key step in a preferred route to compounds of formula I is a cyclisation to produce the B-ring, involving generation of an aldehyde (or functional equivalent thereof) at what will be the 11-position, and attack thereon by the 10-nitrogen:
  • the masked aldehyde may be an aldehyde precursor, such as an alcohol, -CHOH, in which case the reaction involves oxidation, e.g. by means of TPAP or DMSO (Swern oxidation) .
  • the masked aldehyde compound can be produced by condensing a corresponding 2-substituted pyrrolidine with a 2-nitrobenzoic acid:
  • nitro group can then be reduced to -NH 2 and protected by reaction with a suitable agent, e.g. a chloroformate, which provides the therapeutically removable nitrogen protecting group
  • FIG. 1 A process involving the oxidation-cyclization procedure is illustrated in Figure 1 (an alternative type of cyclisation will be described later with reference to Figure 3) .
  • R xl is other than hydrogen
  • the compound of formula I may be prepared by direct etherification of the alcohol la.
  • X is NH then treatment of the alcohol la with the appropriate amine yields the desired compound of formula I.
  • the uncyclized alcohol (IVa) may be prepared by the addition of a nitrogen protection reagent of formula VI, which is preferably a chloroformate or acid chloride, to the amino alcohol (Va) , generally in solution, generally in the presence of a base such as pyridine (preferably 2 equivalents) at a moderate temperature (e.g. at 0°C) . Under these conditions little or no O-acylation is usually observed.
  • a nitrogen protection reagent of formula VI which is preferably a chloroformate or acid chloride
  • the key amino alcohol (Va) may be prepared by reduction of the corresponding nitro compound (Xla) , by choosing a method which will leave the rest of the molecule intact.
  • Exposure of Xla to hydrazine/Raney nickel avoids the production of tin salts and may result in a higher yield of Va, although this method is less compatible with the range of possible C and A-ring substituents. For instance, if there is C-ring unsaturation (either in the ring itself, or at R 2 or R 3 ) , this technique may be unsuitable.
  • the nitro compound of formula Xla may be prepared by coupling the appropriate o-nitrobenzoyl chloride to a compound of formula IXa, e.g. in the presence of K 2 C0 3 at -25 °C under a N 2 atmosphere.
  • the o-nitrobenzoyl chloride is synthesised from the o-nitro benzoic acid of formula XII - many of these are commercially available, and the synthesis of some examples has been reported by Althuis (Althuis, T.H. and Hess, H-J, 1977, Synthesis and Identification of the Major Metabolites of Prazosin Formed in Dog and Rat,
  • PBD dimers may be synthesized using the strategy developed for the synthesis of the protected PBD monomers ( Figure 2a).
  • Figure 2 also shows a synthesis route where the dimer linkage is of the formula -O- (CH 2 ) n -0- .
  • the step of dimer formation is normally carried out to form a bis (nitro acid) XII' ( Figure 2b).
  • the bis (nitro acid) XII 1 may be obtained by nitrating (e.g. using 70% nitric acid) the bis (carboxylic acid). This can be synthesised by alkylation of two equivalents of the relevant benzoic acid with the appropriate diiodoalkane under basic conditions (Route 2a) . Many benzoic acids are commercially available and others can be synthesised by conventional methods.
  • An alternative synthesis of the bis (nitro acid) involves oxidation of the bis (nitro aldehyde), e.g. with potassium permanganate. This can be obtained in turn by direct nitration of the bis (aldehyde) , e.g. with 70% HN0 3 . Finally, the bis (aldehyde) can be obtained via Mitsunobu etherification of two equivalents of the benzoic aldehyde with the appropriate alkanediol (Route 2b) .
  • the final or penultimate step is an oxidative cyclisation.
  • An alternative, using thioacetal coupling unmasking, is shown in figure 3 (which shows it applied to a dimer, with a dimer linkage of formula -o- (CH 2 ) n -0-) .
  • Mercury- mediated unmasking causes cyclisation to the desired compound (la' ) .
  • the thioacetal compound may be prepared as shown in figure 3: the thioacetal protected C-ring [prepared via a literature method: Langley, D.R. & Thurston, D.E., J. Organi c Chemistry, 52, 91-97 (1987)] is coupled to the bis (nitro carboxylic acid) core using a literature procedure.
  • the resulting nitro compound cannot be reduced by hydrogenation, because of the presence of the thioacetal group, so the tin (II) chloride method is used to afford the bis(amine).
  • This is then N-protected, e.g., by reaction with a chloroformate or acid chloride, such as p- nitrobenzylchloroformate .
  • Acid or palladium-mediated unmasking is the preferred method of unmasking to cause cyclisation to the desired compound of formula la or I' a.
  • the vector for use in GDEPT therapies may be any suitable DNA or RNA vectors.
  • Suitable non-viral vectors include cationic liposomes and polymers.
  • Suitable viral vectors include those which are based upon a retrovirus . Such vectors are widely available in the art. Huber et al . (ibid) report the use of amphotropic retroviruses for the transformation of hepatoma, breast, colon or skin cells. Culver et al . (Science (1992) 256; 1550-1552) also describe the use of retroviral vectors in GDEPT. Such vectors or vectors derived from them may also be used. Other retroviruses may also be used to make vectors suitable for use in the present invention. Such retroviruses include Rous sarcoma virus (RSV) .
  • RSV Rous sarcoma virus
  • Englehardt et al . (Nature Genetics (1993) 4.; 27-34) describe the use of adenovirus-based vectors in the delivery of the cystic fibrosis transmembrane conductance product (CFTR) into cells, and such adenovirus-based vectors may also be used.
  • Vectors utilising adenovirus promoter and other control sequences may be of use in delivering a system according to the invention to cells in the lung, and hence useful in treating lung tumours.
  • vectors based on the Molony murine leukaemia virus are known (Ram, Z et al . , Cancer Research (1993) 53; 83-88; Dalton & Treisman, Cell (1992) 68.; 597-612) .
  • These vectors contain the Murine Leukaemia virus (MLV) enhancer cloned upstream at a ⁇ -globin minimal promoter.
  • MLV Murine Leukaemia virus
  • the ⁇ -globin 5' untranslated region up to the initiation codon ATG is supplied to direct efficient translation of the enzyme.
  • Suitable promoters which may be used in vectors described above, include MLV, CMV, RSV and adenovirus promoters.
  • Preferred adenovirus promoters are the adenovirus early gene promoters . Strong mammalian promoters may also be suitable.
  • An example of such a promoter is the EF-l ⁇ promoter which may be obtained by reference to Mizushima and Nagata ((1990), Nucl . Acids Res. 18; 5322). Variants of such promoters retaining substantially similar transcriptional activities may also be used.
  • promoters include tissue specific promoters, and promoters activated by small molecules, hypoxia or X-rays.
  • nitroreductase is the enzyme of choice for the activation of compounds of formula I
  • the enzyme is a non- mammalian nitroreductase such as a bacterial nitroreductase .
  • An E. coli nitroreductase as disclosed in WO93/08288 is particularly preferred.
  • the enzyme may be modified by standard recombinant DNA techniques, e.g. by cloning the enzyme, determining its gene sequence and altering the gene sequence by methods such as truncation, substitution, deletion or insertion of sequences for example by site-directed mutagenesis. Reference may be made to "Molecular Cloning" by Sambrook et al .
  • the modification made may be any which still leaves the enzyme with the ability to reduce the nitro group in suitable compounds of formula I, but alters other properties of the enzyme, for example its rate of reaction or selectivity.
  • small truncations in the N- and/or C-terminal sequence may occur as a result of the manipulations required to produce a vector in which a nucleic acid sequence encoding the enzyme is linked to the various other vector sequences .
  • an antibody directed against a tumour specific marker is linked to the relevant enzyme, which may be modified as described above.
  • the antibody may be monoclonal or polyclonal.
  • the term "antibody”, unless specified to the contrary, includes fragments of whole antibodies which retain their binding activity for a tumour target antigen. Such fragments include Fv, F(ab') and F(ab') 2 fragments, as well as single chain antibodies.
  • the antibodies and fragments thereof may be humanised antibodies, e.g. as described in EP-A-239400.
  • the antibodies may be produced by conventional hybridoma techniques or, in the case of modified antibodies or fragments, by recombinant DNA technology, e.g. by the expression in a suitable host vector of a DNA construct encoding the modified antibody or fragment operably linked to a promoter.
  • suitable host cells include bacterial (eg. E. coli ) , yeast, insect and mammalian cells.
  • the enzyme may be produced by linking a nucleic acid sequence encoding the enzyme (optionally modified as described above) to the 3' or 5 ' end of the sequence of the construct encoding the antibody or fragment thereof.
  • the activation process in PDT can be highly site specific.
  • the direction of a laser beam can be controlled with great precision, and the beam diameter can be reduced to a width far below that of a single cell. Therefore, it can act upon a very limited area, minimising damage to neighbouring tissue.
  • Ultra-violet light is sufficient to break a range of chemical bonds since the energy spectrum for bond breakage for the majority of organic molecules lies between 250 and 420 kJ/mol, and, for example, 350 nm is equivalent to 340 kJ/mol.
  • a broad range of light-mediated deprotection reactions have been demonstrated including the photochemical deprotection of amino acids, peptides and polysaccharides from their CBZ and o-nitrobenzyl and , 5-dimethoxy-2-nitrobenzyl carbamate forms at wavelengths longer than 350 nm, (Pillai, R. V. N., Photoremovable protecting groups in organic chemistry, Synthesis (1980), 1-26), (Bayley, H., Gasparro, F.
  • tumour types have been identified as potential targets for PDT. They include head and neck tumours, carcinomas of the bronchus, malignant brain tumours, superficial tumours of the bladder and vascular disease, which have all shown promising responses in the clinic, (Regula, J., Mac Roberts, A. J. , Gorchein, A., Buonaccorsi, Thorpe, S. M. , Spencer, G. M. , Hartfield, A. R. . and Bown, S. G., Photosensitisation and photodynamic therapy of oesophageal, duodenal and colorectal tumours using 5-aminoleavulic acid induced protoporphyrin IX-a pilot study, Gut (1995) 36, 67-75).
  • the technique of PDT as discussed above can be used in combination with appropriate compounds of formula I when the therapeutically removable nitrogen protecting group is photolabile.
  • the preferred wavelength of UV light used is 250 to 400 or 550 nm.
  • Compounds of the invention can be used in vi tro or in vivo for a range of applications.
  • a number of vector systems for the expression of nitroreductase in a cell have been developed.
  • the further development of such systems e.g. the development of promoters suitable for specific cell types
  • suitable candidate prodrugs capable of killing cells when activated by nitroreductase e.g. the development of promoters suitable for specific cell types
  • Prodrug compounds of formula I susceptible to nitroreductase may be used in such model systems.
  • the model systems may be in vi tro model systems or in vivo xenograft model systems comprising for example human tumour cells implanted in nude mice.
  • Compounds of formula I susceptible to different enzymes may be used in similar systems which have been appropriately modified.
  • Compounds of formula I which are not activatable by an enzyme may be tested in vi tro with other suitable forms of activation against panels of different tumour cell types to determine efficacy against such tumour cells.
  • the efficacy of compounds of the invention against a range of tumour cell types may be used as points of reference for the development of further antitumour compounds.
  • Compounds of formula I may also be tested in combination with additional anti-cancer compounds to determine potential combination drug systems, for example combinations which are synergistic.
  • Compounds of formula I may also be used in a method of treatment of the human or animal body.
  • Such treatment includes a method of treating the growth of neoplastic cells in a patient with neoplastic disease which comprises administering to a patient in need of treatment compounds of formula I as part of an ADEPT, GDEPT or PDT system or treatment with compounds of formula I alone, where neoplastic diseases include leukaemia and solid tumours such as ovarian, colonic, lung, renal, breast, bowel, CNS and melanomas.
  • the treatment can also be the treatment of other site-specific diseases where local increase in toxicity is beneficial to the patient.
  • treatment includes any measure taken by the physician to alleviate the effect of the tumour on a patient.
  • effective treatment will also include any measures capable of achieving partial remission of the tumour as well as a slowing down in the rate of growth of a tumour including its metastases. Such measures can be effective in prolonging and/or enhancing the quality of life and relieving the symptoms of the disease.
  • the antibody/enzyme conjugate for ADEPT can be administered simultaneously but it is often found preferable, in clinical practice, to administer the enzyme/antibody conjugate before the prodrug, e.g. up to 72 hours or even 1 week before, in order to give the enzyme/antibody conjugate an opportunity to localise in the region of the tumour target.
  • the prodrug is administered, conversion of the prodrug to the cytotoxic agent tends to be confined to the regions where the enzyme/agent conjugate is localised, i.e. the region of the target tumour. In this way, the premature release of the compound produced by the action of the nitroreductase on the prodrugs of the present invention is minimised.
  • the degree of localisation of the enzyme/agent conjugate (in terms of the ratio of localized to freely circulating active conjugate) can be further enhanced using the clearance and/or inactivation systems described in WO89/10140.
  • a component may include an antibody to the enzyme component of the system which is capable of inactivating the enzyme.
  • the second component may be linked to a macromolecule such as dextran, a liposome, albumin, macroglobulin or a blood group O erythrocyte so that the second component is restrained from leaving the vascular compartment.
  • the second component may include a sufficient number of covalently bound galactose residues, or residues of other sugars such as lactose or mannose, so that it can bind the conjugate in plasma but be removed together with the conjugate from plasma by receptors for galactose or other sugars in the liver.
  • the second component should be designed for use and administered such that it will not, to any appreciable extent, enter the extravascular space of the tumour where it could inactivate localised conjugate prior to and during administration of the prodrug.
  • the dose of the prodrug and conjugate will ultimately be at the discretion of the physician, who will take into account such factors as the age, weight and condition of the patient. Suitable doses of prodrug and conjugate are given in Bagshawe et al . Antibody, Immunoconjugates, and Radiopharmaceuticals (1991), 4 . , 915-922.
  • a suitable dose of conjugate may be from 500 to 200,000 enzyme units/m 2 (e.g. 20,000 enzyme units/m 2 ) and a suitable dose of prodrug may be from about 0.1 to 200 mg/Kg, preferably from about 10 to about 100 mg/Kg per patient per day.
  • the ADEPT system when used with nitroreductase also preferably comprises a suitable co-factor for the enzyme. Suitable co- factors may include a riboside or ribotide of nicotinic acid or nicotinamide .
  • the antibody/enzyme conjugate may be administered by any suitable route usually used in ADEPT therapy. This includes parenteral administration of the antibody in a manner and in formulations similar to that described below.
  • the vectors will usually be packaged into viral particles and the particles delivered to the site of the tumour, as described in for example Ram et a 1 . ( supra ) .
  • the viral particles may be modified to include an antibody, fragment thereof (including a single chain) or tumour- directed ligand to enhance targeting of the tumour.
  • the vectors may be packaged into liposomes .
  • the liposomes may be targeted to a particular tumour. This can be achieved by attaching a tumour-directed antibody to the liposome.
  • Viral particles may also be incorporated into liposomes.
  • the particles may be delivered to the tumour by any suitable means at the disposal of the physician.
  • the viral particles will be capable of selectively infecting the tumour cells.
  • selectively infecting it is meant that the viral particles will primarily infect tumour cells and that the proportion of non- tumour cells infected is such that the damage to non-tumour cells by administration of a prodrug will be acceptably low, given the nature of the disease being treated. Ultimately, this will be determined by the physician.
  • Viruses for example isolated from packaging cell lines, may also be administered by regional perfusion or direct intratumoral injection, or direct injection into a body cavity (intracaviterial administration) , for example by intra-peritoneal injection.
  • the exact dosage regime for GDEPT will, of course, need to be determined by individual clinicians for individual patients and this, in turn, will be controlled by the exact nature of the prodrug and the cytotoxic agent to be released from the prodrug. However, some general guidance can be given.
  • Chemotherapy of this type will normally involve parenteral administration of modified virus, and administration by the intravenous route is frequently found to be the most practical.
  • the amount of virus or other vector delivered will be such as to provide a similar cellular concentration of enzyme as in the ADEPT system mentioned above.
  • the vector will be administered to the patient and then the uptake of the vector by transfected or infected (in the case of viral vectors) cells monitored, for example by recovery and analysis of a biopsy sample of targeted tissue. This may be determined by clinical trials which involve administering a range of trial doses to a patient and measuring the degree of infection or transfection of a target cell or tumour.
  • the amount of prodrug required will be similar to or greater than that for ADEPT systems.
  • prodrug In using a GDEPT system the prodrug will usually be administered following administration of the vector encoding an enzyme. Suitable doses of prodrug are from about 0.1 to 200 mg/Kg, preferably from about 10 to about 100 mg/Kg per patient per day.
  • the formulations comprise the compounds, together with one or more acceptable carriers thereof and optionally other therapeutic ingredients, or diluents.
  • the carrier or carriers must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipients thereof, for example, liposomes.
  • Suitable liposomes include, for example, those comprising the positively charged lipid (N [1- (2 , 3-dioleyloxy) propyl] -N, N, N-triethylammonium (DOTMA) , those comprising dioleoylphosphatidylethanolamine (DOPE), and those comprising 3 ⁇ [N- (n'N' -dimethylaminoethane) - carbamoyl] cholesterol (DC-Choi).
  • DOTMA positively charged lipid
  • DOPE dioleoylphosphatidylethanolamine
  • DC-Choi 3 ⁇ [N- (n'N' -dimethylaminoethane) - carbamoyl] cholesterol
  • Formulations suitable for parenteral or intramuscular administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats, bacteriocidal Antibiotics and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs .
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, for example Water for Injection, immediately prior to use.
  • a sterile liquid carrier for example Water for Injection
  • Injection solutions and suspensions may be prepared extemporaneously from sterile powders, granules and tablets of the kind previously described.
  • the formulations may include other agents conventional in the art having regard to the type of formulation in question.
  • sterile pyrogen-free aqueous and non-aqueous solutions are preferred.
  • the doses may be administered sequentially, eg. at hourly, daily, weekly or monthly intervals, or in response to a specific need of a patient.
  • Preferred routes of administration are oral delivery and injection, typically parenteral or intramuscular injection or intratumoural injection.
  • parenteral or intramuscular injection or intratumoural injection may be preferred, e.g. subcutaneous injection or creams and ointments, and such methods of administration are well known.
  • the exact dosage regime will, of course, need to be determined by individual clinicians for individual patients and this, in turn, will be controlled by the exact nature of the compound of formula I, but some general guidance can be given. Typical dosage ranges generally will be those described above which may be administered in single or multiple doses. Other doses may be used according to the condition of the patient and other factors at the discretion of the physician.
  • HRMS Accurate molecular masses
  • PFK perfluorokerosene
  • FAB mass spectra were obtained from a glycerol/thioglycerol/ trifluoroacetic acid (1:1:0.1) matrix with a source temperature of 180°C.
  • Optical rotations at the Na-D line were obtained at ambient temperature using a Perkin-Elmer 141 Polarimeter.
  • Flash chromatography was performed using Aldrich flash chromatography "Silica Gel-60" (E. Merck, 230-400 mesh).
  • Thin-layer chromatography (TLC) was performed using GF 254 silica gel (with fluorescent indicator) on glass plates.
  • Example 1 Synthesis of a nitroreductase-activated benzyl DC-81 prodrug for ADEPT (7) .
  • Method B 4-benzyloxy-3-methoxybenzoic acid (8.5g, 32.9 mmol) was added in small portions over 30 minutes to stirred solution of 70% nitric acid (100 ml) . When addition was complete the reaction mixture was allowed to warm to 15 °C and maintained at that temperature for a further 30 min. The reaction mixture was then poured onto ice and the resultant precipitate was collected by filtration, washed with ice-cold water and dried to afford the nitrated product 3 as a yellow powder.
  • the resulting acid chloride solution was then added dropwise over 30 minutes to a stirred suspension of pyrrolidinemethanol (1.168 g, 11.56 mmol, 1 eq) and K 2 C0 3 (3.675 g, 26.63 mmol, 2.3 eq) in acetonitrile (80 ml) at -25°C under a nitrogen atmosphere.
  • the reaction mixture was stirred at the same temperature for a further 1 hour and then allowed to return to room temperature and quenched with water (200 ml) .
  • Method B Hydrazine hydrate (4.53 g, 90.67 ml, 5 eq) was added dropwise to a solution of the nitro compound 4 (7.0 g, 18.13 mmol) in dry methanol (20 ml) and a catalytic amount of Raney Ni (0.544 g) over antibumping granules whilst a gently reflux was maintained. The mixture was heated at reflux for a further 15 minutes when TLC (5% MeOH/CHCl 3 ) indicated that the reaction had gone to completion. The Ni catalyst was then removed by filtration through Celite and the solvent removed by evaporation in vacuo. The product was further purified by flash chromatography (5% MeOH/CHCl 3 ) to afford the amine 5 as a bright yellow unstable oil which required storage at low temperature.
  • TPAP (23 mg, 10% molar) was then added to the reaction mixture and it was allowed to stir for a further 2 hours when the reaction was found to be complete by TLC (2% MeOH/CHCl 3 ) .
  • the molecular sieve was removed by filtration through Celite and the resulting solution was evaporated in vacuo .
  • the resulting dark oil was subjected to flash chromatography (1% MeOH/CHCl 3 ) to give the photolabile protected PBD (11) .
  • Tributyltinhydride (1.49 g, 5.1 mmol, 1.1 eq) was added to a solution of 18 (1.5 g, 4.62 mmol) in DCM (30 ml) in the presence of water (0.48 g, 27.11 mmol, 6 eq) and a catalytic amount of Pd(PPh 3 ) 2 Cl 2 (0.132 g, 0.189 mmol, 4% molar) and the reaction mixture was allowed to stir for 5 minutes at room temperature.
  • TPAP (9.4 mg, 0.026 mmol, 5% molar) was then added and the mixture was allowed to stir under the same conditions for a further 2.5 hours.
  • the reaction mixture was filtered through Celite and the filtrate evaporated in vacuo .
  • the residue was subjected to flash chromatography (1% MeOH/CHCl 3 ) to obtain the target compound (24).
  • the dimer acid 25 (5 g, 13.3 mmol) was added slowly in small portions over 30 minutes to a stirred solution of 70% HN0 3 (50 ml) at 0°C. After the addition was complete the reaction mixture was allowed to warm to 15 °C and stirring continued for a further 2 hours. The reaction mixture was poured onto ice causing precipitation of the product. The yellow precipitate was collected by filtration, washed with cold water and dried to afford the nitrated dimer acid core (26) as a pale yellow solid. Yield 4.52 g (73%); mp. 241-246°C; IR (cm "1 ) 3500-3200, 2924, 1713, 1604, 1582, 1523, 1459, 1377, 1282, 1218, 1189, 1053; X E-
  • the resulting mixture was allowed to stir at -25°C for a further 1.5 hour when the reaction mixture was diluted with water (100 ml) .
  • the solution was then extracted with chloroform (4 x 100 ml) .
  • the combined organic phase was washed with aq. IM HCl (2 x 75 ml) , water (2 x 75 ml) , brine (100 ml), dried (MgS0 4 ) and evaporated in vacuo .
  • the resulting orange oil was purified by column chromatography (10% MeOH/CHCl 3 ) to afford the pure product 27 as a yellow viscous oil.
  • the reaction mixture was diluted with chloroform (50 ml) and washed with a saturated aqueous solution of CuS0 4 (2 x 50 ml), water (2 x 50 ml), brine (100 ml) and dried (MgS0 4 ) .
  • the organic solvent was removed in vacuo and the pure dimer carbamate 29 was obtained after flash chromatography (7% MeOH/CHCl 3 ) as a bright yellow foam.
  • TPAP (13.4 mg, 0.114 mmol, 0.3 eq) was added in one portion to a solution of the bis carbamate 29 (0.35 g, 0.37 mmol) and NMO (0.134 g, 1.14 mmol, 3.1 eq) in dry DCM/CH 3 CN (9:3 ml) which has been allowed to stir over molecular sieve (0.350 g) for 15 minutes under N 2 and at room temperature. Progress of the reaction was followed by TLC (7% MeOH/CHCl 3 ) . After 2 hours reaction was still incomplete requiring the addition of a further amount of NMO (67 mg, 0.55 mmol, 1.5 eq) and TPAP (6.7 mg, 0.05 mmol, 0.15 eq) .
  • 57 intensity 925 (M + -l, 1), 889 (5), 711 (6), 501 (3), 286 (10), 252 (7), 213 (15), 192 (32), 197 (11), 185 (22), 181 (42), 165 (15), 149 (47), 131 (18), 119 (16), 105 (29), 91 (96), 73 (100), 57 (54) .
  • reaction mixture was then diluted with chloroform (50 ml) and washed with saturated aqueous solution of CuS0 4 (2 x 50 ml), water (2 x 50 ml), brine (100 ml) and dried (MgS0 4 ) . Removal of the solvent under reduced pressure gave a dark yellow oil which was further purified by flash chromatography (7% MeOH/CHCl 3 ) to afford the pure product as a yellow foam.
  • NMO (0.138 g, 1.16 mmol, 3.1 eq) and molecular sieve (350 mg) was added to a stirred solution of the NVOC dimer carbamate 31(0.4 g, 0.38 mmol) in dry DCM and acetonitrile (9:3 ml) under nitrogen. This mixture was allowed to stir for 15 minutes before the addition of TPAP (13.8 mg, 0.116 mmol, 0.3 eq) .
  • Triphenylphosphine oxide was removed by filtration and the filtrate was washed with NaOH (100 ml), dried (MgS0 4 ) , evaporated in vacuo and the remaining opaque oil was purified by flash chromatography (100% CHC1 3 ) to afford a white solid.
  • the dimer aldehyde (39) (5 g, 14.5 mmol) was added in small portions over a period of 30 minutes to HN0 3 (100 ml, 70%) at 0°C. The resulting suspension was stirred for a further 30 minutes at 15 °C when it was poured onto ice-water and the bright yellow precipitate was collected by filtration, washed with cold water and dried to afford the nitrated intermediate.
  • Lithium tetrahydroborate (2.6 g, 0.12 mol) was added portionwise to a solution of N-carbobenzyloxy-L-proline methyl ester 41 (21 g, 0.08 mol) in THF (500 ml) at 0°C. The reaction mixture was allowed to stir at room temperature for 48 hours. The solution was then cooled to 0°C and ice water (150 ml) was added to quench excess lithium tetrahydroborate. The resulting suspension was adjusted to pH 4.0 with aqueous HCl (1.0 N) and extracted with Et 2 0 (250 ml) .
  • Nickel 0.1 g
  • Nickel was removed by filtration through Celite.
  • SW1116 and LS174T are human adenocarcinoma colonic cell lines, which were grown at Charing Cross hospital.
  • the cells at a concentration of 2500 cells/ml, were plated in 96-well microtitre plates, and were incubated at 37°C for 1 hour with different concentrations of the prodrug, in the presence or absence of E. coli nitroreductase-monoclonal antibody conjugate in phosphate buffer saline (available from Sigma) , and NADH (the co-factor necessary for enzyme function) in DMSO.
  • the cells were then washed and incubated for a further 3 days at 37 °C. At the end of this period, the cells were fixed (TCA) and stained. The concentration of the remaining viable cells adhering to the plates was quantified by a sulforhodamine B (SRB) dye.
  • SRB sulforhodamine B
  • the parent drug, benzyl DC-81 (Thurston et al, 1990, Chem. Bri t . , 26, 767-772) , was also evaluated in the same cell lines under the same conditions in order to establish the extent of activation 00/12507
  • Compound 24 was examined in the same cell lines as example 6, and under the same conditions. It exhibited an IC 50 value of 86.2 ⁇ M (average of two experiments) which, after addition of the enzyme conjugate and NADH, dropped to 6.4 ⁇ M, indicating an activation factor of approximately 13.5. In this case, the parent benzyl tomaymycin was unavailable for evaluation as a control. However, assuming that the addition of a benzyl group to the C8-position of the A-ring of tomaymycin does not change the cytotoxicity of the compound significantly (as is the case for DC-81/C8-benzyl DC-81), the cytotoxicity of C8-benzyl tomaymycin should be in the order of 0.01-0. OOl ⁇ M.
  • Example 10 Light-activation of the benzyl DC-81 prodrug (Compound 11)
  • This experiment involved the irradiation of compound 11, see example 3, in DMF at a concentration of 1 mM using a Stratagene UVA Crosslinker (365 nm) .
  • Small aliquots (lOO ⁇ L) were removed at 30 minutes, 1 hour, 2 hours and 3 hours and the cleavage of the photolabile NVOC group was monitored by HPLC, using a Waters C4, 300 angstrom reversed phase column and a mobile phase of 50% methanol/50% water.
  • Compound 11 has a retention time of 11.25 minutes and upon radiation produced a new peak with a retention time of 8.58 minutes.
  • Authentic benzyl DC-81 has an identical retention time of 8.58 minutes.
  • the time course of the deprotection process is shown in figure 5. Complete conversion is achieved by 2 hours of irradiation at 365nm under the conditions employed.
  • An MTT assay was used to evaluate the ability of aliquots of the activated analogues, which were removed at time intervals, to inhibit the growth of chronic human histiocytic leukaemia K562 cells in culture. Following treatment of cells with a range of drug doses, cells were transferred to 96-well microtitre plates, 10 4 cells per well, 8 wells per sample. Plates were incubated at 37°C in a humidified atmosphere containing 5% C0 2 .
  • the assay is based on the ability of viable cells to reduce a yellow soluble tetrazolium salt 3- ( 4 , 5-dimethylthiazolyl ) -2 , 5- diphenyltetrazolium bromide (MTT) , to an insoluble purple formazan precipitate.
  • MTT 5- diphenyltetrazolium bromide
  • DMSO 200 ⁇ L
  • the optical density was then measured at a wavelength of 550 nm on a Titertek Multiscan ELISA plate reader and expressed as a percentage of the control optical density. For each curve, an IC 50 value was read as the dose required to reduce the final optical density to 50% of the control value.
  • the dimer derivative 32 see example 5(b), was found to be slightly cytotoxic before UV irradiation with an IC 50 of 4.5 ⁇ M. Incubation of a solution of compound 32 with cells following 2 hours irradiation reduced the IC 50 to 1.25 ⁇ M (figure 9). This was further reduced to 0.85 ⁇ M following 5 hours irradiation.
  • the parent PBD dimer DSB-120 gave an IC 50 of 0.55 ⁇ M in the same cell line, see Figure 9.
  • Single viable cells were seeded in growth medium (160 ⁇ L) in 96- well microtitre plates and allowed to attach overnight.
  • the test compounds were then dissolved in DMSO (to give 20 mM drug concentrations) immediately prior to adding the cells in quadruplicate wells.
  • the final drug concentrations in the wells ranged from lOO ⁇ M to 2.5nM as follows: 100, 25, 10, 2.5, 1 ⁇ M, 250, 100, 25, 10, 2.5 nM (drugs were diluted in growth medium and then 40 ⁇ L added to the existing well volume of 160 ⁇ L to give final concentrations as above) .
  • the medium was removed and the remaining cells fixed by exposure to 10% trichloroacetic acid on ice for 30 minutes.
  • the wells were then washed 3-4 times with tap water, air dried overnight and treated with 100 ⁇ L of sulphorhodamine B (0.4%) dissolved in 1% acetic acid. Staining was allowed to continue for 10-15 minutes, then the wells were washed 3-4 times with 1% acetic acid, air dried and then added to Tris base (100 ⁇ L of lOmM) . Plates were then shaken and absorbance readings at 540 nM were determined using a plate reader. The IC 50 values were calculated from plots of concentration versus percentage absorbance (compared with 8 untreated wells).
  • RF is the resistance factor, which is the cytotoxicity of the compound in the cisplatin resistant cell line divided by the cytotoxicity in the normal cell line.
  • the Psec-protected prodrug (52) UP2073 was found to be at least 50 times more toxic than the Ptec control.
  • the compound was particularly active against the SKOV3 cell line; this is noteworthy as this cell line is intrinsically resistant to electrophilic cytotoxic agents due to the presence of high levels of glutathione/glutathione transferase.
  • the Psec prodrug UP2073 was actually more active in this cell line than the PBD it was based upon UP2025. Without wishing to be bound by theory, it is possible that the prodrug is protected from glutathione and other biological nucleophiles until it is deprotected close to the site of action.
  • NCI National Cancer Institute
  • NCI National Cancer Institute
  • the NCI has available an in vi tro cytotoxicity screen which consists of approximately 60 human tumour cell lines against which compounds are tested at a minimum of five concentrations each differing 10- fold.
  • a 48 hour continuous exposure protocol is used, where cell viability or growth is estimated with an SRB protein assay.
  • test compound was evaluated against approximately 60 human tumour cell lines.
  • the NCI screening procedures were described in detail by Monks and co-workers (Monks, A et al . , Journal of the National Cancer Institute, 1991, 83, 757) . Briefly, cell suspensions were diluted according to the particular cell type and the expected target cell density (5000-40,000 cells per well based on cell growth characteristics), and added by pipette (100 UL) into 96-well microtitre plates. The cells were allowed a preincubation period of 24 hours at 37°C for stabilisation.
  • test compounds were evaluated at five 10-fold dilutions (10 ⁇ 4 , 10 "5 , 10' 6 , 10" 7 and 10 "8 ⁇ M) .
  • the test compounds were incubated for 48 hours in 5% C0 2 atmosphere and 100% humidity.
  • the cells were then assayed using the sulphorhodamine B assay.
  • a plate reader was used to read the optical densities and a microcomputer processed the readings into LC 50 values, which is the dosage required to kill half of the cells. IC 50 values, the dosage required to inhibit the growth of half the cells, was also measured.
  • Figure 6 & 7 ( ⁇ ) Benzyl DC-81; ( ⁇ ) Compound 11; (•) Compound 11 + UVA 30 mins; (O) Compound 11 + UVA lh; (A) Compound 11 + UVA 2 h.
  • Figure 8 ( ⁇ ) Benzyl DC-81; (D) Compound 11; (•) Compound 11 + UVA 30 mins; (O) Compound 11 + UVA lh; (A) Compound 11 + UVA 2 h
  • Figure 9 ( ⁇ ) DSB-120; ( ⁇ ) Compound 32; (•) Compound 32 + UVA 30 mins; (O) Compound 32 + UVA lh; (A) Compound 32 + UVA 2h; ( ⁇ ) Compound 32 + UVA 5h

Abstract

A compound with formula (I) where R10 is a therapeutically removable nitrogen protecting group; R2 and R3 are independently selected from: H, R, OH, OR, =O, =CH-R, =CH2, CH2-CO2R, CH2-CO2H, CH2-SO2R, O-SO2-R, CO2R, COR and CN; R6, R7 and R9 are independently selected from H, R, OH, OR, halo, amino, nitro, Me3Sn; X is S, O or NH; R11 is either H or R; and where there is optionally a double bond between C1 and C2 or C2 and C3; and R8 is selected from H, R, OH, OR, halo, amino, nitro, Me3Sn, or R7 and R8 together form a group -O-(CH2)p-O-, where p is 1 or 2. Such compounds may be used in methods of ADEPT, GDEPT, NPEPT or PDT.

Description

COMPOUNDS
The present invention relates to pyrrolobenzodiazepines (PBDs), and is particularly concerned with the use of these compounds as prodrugs for antibody-directed enzyme-prodrug therapy (ADEPT), gene-directed enzyme-prodrug therapy (GDEPT) , photodynamic therapy (PDT) and naturally present enzyme-prodrug therapy (NPEPT) .
Background to the invention
Pyrrolobenzodiazepines (PBDs) have the ability to recognise and bond to specific sequences of DNA; the most preferred sequence is PuGPu (Purine-Guanine-Purine) . The first PBD antitumour antibiotic, anthramycin, was discovered in 1965 (Leimgruber et al., 1965 J. Am. Chem. Soc, 87, 5793-5795; Leimgruber et al., 1965 J. Am. Chem. Soc, 87, 5791-5793). Since then, a number of naturally occurring PBDs have been reported, and over 10 synthetic routes have been developed to a variety of analogues (Thurston et al . , 1994 Chem. Rev. 1994, 433-465). Family members include abbeymycin (Hochlowski et al., 1987 J. Antibiotics, 40, 145-148), chicamycin (Konishi et al., 1984 J. Antibiotics, 37, 200-206), DC-81 (Japanese Patent 58-180 487; Thurston et al., 1990, Chem. Brit., 26, 767-772; Bose et ai . , 1992 Tetrahedron, 48, 751-758), mazethramycin (Kuminoto et al., 1980 J. Antibiotics, 33, 665-667), neothramycins A and B (Takeuchi et al., 1976 J. Antibiotics, 29, 93-96), porothramycin (Tsunakawa et al., 1988 J. Antibiotics, 41, 1366-1373), prothracarcin (Shimizu et al, 1982 J. Antibiotics, 29, 2492-2503; Langley and Thurston,
1987 J. Org. Chem., 52, 91-97), sibanomicin (DC-102 ) (Hara et al.,
1988 J. Antibiotics, 41, 702-704; Itoh et al., 1988 J. Antibiotics, 41, 1281-1284), sibiromycin (Leber et al., 1988 J. Am. Chem. Soc, 110, 2992-2993) and tomamycin (Arima et al., 1972 J. Antibiotics, 25, 437-444). PBDs are of the general structure: O 00/12507
Figure imgf000004_0001
They differ in the number, type and position of substituents, in both their aromatic A rings and pyrrolo C rings, and in the degree of saturation of the C ring. In the B-ring there is either an imine (N=C) , a carbinolamine (NH-CH(OH)) or a carbinolamine methyl ether (NH-CH (OMe) ) at the N10-C11 position which is the electrophilic centre responsible for alkylating DNA. All of the known natural products have an ( S) -configuration at the chiral Clla position which provides them with a right-handed twist when viewed from the C ring towards the A ring. This gives them the appropriate three-dimensional shape for isohelicity with the minor groove of B-form DNA, leading to a snug fit at the binding site (Kohn, 1975 In Antibi oti cs III. Springer-Verlag, New York, pp. 3-11 ; Hurley and Needham-VanDevanter, 1986 Ace Chem . Res . , 19, 230-237). Their ability to form an adduct in the minor groove enables them to interfere with DNA processing, hence their use as antitumour agents.
The use of prodrugs represents a very valuable clinical concept in cancer therapy. For example, a prodrug may be converted into an antitumour agent under the influence of an enzyme that is linked to a monoclonal antibody so that it can bind to a tumour associated antigen. The combination of such a prodrug with such an enzyme monoclonal antibody conjugate represents a very powerful therapeutic strategy. This approach to cancer therapy, often referred to as "antibody directed enzyme/prodrug therapy" (ADEPT) is disclosed in WO88/07378.
A further therapeutic approach termed "virus-directed enzyme prodrug therapy" (VDEPT) has been proposed as a method for treating tumour cells in patients using prodrugs. Tumour cells are targeted with a viral vector carrying a gene encoding an enzyme capable of activating a prodrug. The gene may be transcriptionally regulated by tissue specific promoter or enhancer sequences. The viral vector enters tumour cells and expresses the enzyme, thereby converting the prodrug into the active drug within the tumour cells (Huber et al . , Proc. Na tl . Acad. Sci . USA (1991) 88.^ 8039). Alternatively, non-viral methods for the delivery of genes have been used. Such methods include calcium phosphate co-precipitation, microinjection, liposomes, direct DNA uptake, and receptor-mediated DNA transfer. These are reviewed in Morgan & French, Annu. Rev. Biochem., 1993, 62; 191. The term "GDEPT" (gene-directed enzyme prodrug therapy) is used to include both viral and non-viral delivery systems.
Photodynamic therapy (PDT) provides another method which uses prodrugs to deliver desired drugs to specific sites in the human body. Advances in the field of light delivery to internal areas of the body allow delivery to organs and other areas without the need for any extensive surgical procedures. The activation process can be extremely site specific, as the direction of a laser beam can be controlled with great precision, and the beam diameter can be reduced far below that of a single cell, minimising any possible damage to other neighbouring tissue from unwanted activation of the drug. The high energy of ultra-violet light (e.g. 350 nm equivalent to 340 kJ/mol) is sufficient to break a range of chemical bonds, since the bond energy spectrum of the majority of organic molecules lies between 250 and 420 kJ/mol. For example, there has been wide application of the photochemical deprotection of amino acids, peptides and polysaccharides from their o-nitrobenzyl carbamate, CBZ, and 4,5- dimethoxy-2-nitrobenzyl carbamate forms at wavelengths longer than 350 nm.
A further class of prodrugs is those where the protecting group is removed by an enzyme naturally present at the desired site of action. These enzymes include dopa-decarboxylase, L-γ-glutamyl transpeptidase, and mixed function oxidases and reductase (e.g. DT-diaphrase) . This is method termed "naturally present enzyme- prodrug therapy (NPEPT) in this application. One enzyme of particular interest is glutathione transferase (GST) , which forms part of a major cellular defence mechanism based on the use of the tripeptide, glutathione, as a scavenger of toxic electrophiles . GST acts as a catalyst in the reaction between glutathione and its target electrophiles. A consequence of this defence mechanism is the inactivation of electrophilic therapeutic agents. Many human tumour cells exhibit elevated GST levels compared to normal cells and the association of GST with resistance to DNA alkylating agents has been demonstrated by Lewis et al . (Carcinogenesis 1988, 9, 1283-1287), Kuzmich et al . (Journal of Biochemistry 1992, 281 , 219-224), and Tew et al . (Glutathione-S-transferase and anti-cancer drug resistance, in Mechanism of Drug Resistance in Neoplastic Cells; ooley, P. V., Tew, K. D., Eds.; Academic Press: Orlando, FL, 1987; ppl41-159). Chemotherapeutic agents that take advantage of this intrinsic property of cancer cells may prove highly useful in treating refractory cancers.
Prodrugs which make use of this elevated GST level have been made (Satyam et al . , Med. Chem. 1996, 39, 1736-1747). They have a glutathione molecule linked via a 2-sulphonylethyloxycarbonyl linker to a phosphorodiamidate mustard. An alternative type of prodrug has made use of the closely related 2- phenylsulphonylethyloxycarbonyl (Psec) group (Nicolaou et al . ,
Science, 1992, 256, 1172-1178). Such prodrugs showed selectivity between healthy human bone marrow cells and promeocytic and T cell leukemia tumour lines.
Disclosure of the invention A first aspect of the present invention provides a compound with the formula I:
Figure imgf000007_0001
wherein:
R10 is a therapeutically removable nitrogen protecting group;
R2 and R3 are independently selected from: H, R, OH, OR, =0, =CH-
R, =CH2, CH2-C02R, CH2-C02H, CH2-S02R, 0-S02-R, C02R, COR and CN;
Rε, R7 and R9 are independently selected from H, R, OH, OR, halo, amino, nitro, Me3Sn; or R7 and R8 together form a group
-O- (CH2)p-0-, where p is 1 or 2 ;
X is S, O or NH;
Ri is either H or R; where R is a lower alkyl group having 1 to 10 carbon atoms, or an aralkyl group (i.e. an alkyl group with one or more aryl substituents ) , preferably of up to 12 carbon atoms, whereof the alkyl group optionally contains one or more carbon-carbon double or triple bonds, which may form part of a conjugated system, or an aryl group, preferably of up to 12 carbon atoms; and is optionally substituted by one or more halo, hydroxy, amino, or nitro groups, and optionally contains one or more hetero atoms, which may form part of, or be, a functional group; and where there is optionally a double bond between Cl and C2 or C2 and C3; and R8 is selected from H, R, OH, OR, halo, amino, nitro, Me3Sn, where R is as defined above, or the compound is a dimer with each monomer being the same or different and being of formula I, where the R8 groups of the monomers form together a bridge having the formula -T-R' -T- linking the monomers, where R' is an alkylene chain containing from 3 to 12 carbon atoms, which chain may be interrupted by one or more hetero atoms and/or aromatic rings, e.g. benzene or pyridine, and may contain one or more carbon- carbon double or triple bonds, and each T is independently selected from O, S or N.
If R is an aryl group, and contains a hetero atom, then R is a heterocyclic group. If R is an alkyl chain, and contains a hetero atom, the hetero atom may be located anywhere in the alkyl chain, e.g. -0-C2H5, -CH2-S-CH3, or may form part of, or be, a functional group, e.g. carbonyl, hydroxy.
R is preferably independently selected from a lower alkyl group having 1 to 10 carbon atoms, or an aralkyl group, preferably of up to 12 carbon atoms, or an aryl group, preferably of up to 12 carbon atoms, optionally substituted by one or more halo, hydroxy, amino, or nitro groups. It is more preferred that R groups are independently selected from a lower alkyl group having 1 to 10 carbon atoms optionally substituted by one or more halo, hydroxy, amino, or nitro groups. It is particularly preferred that R groups are unsubstituted straight or branched chain alkyl groups, having 1 to 10, preferably 1 to 6, and more preferably 1 to 4, carbon atoms, e.g. methyl, ethyl, propyl, butyl.
Alternatively, R6, R7, R8, and R9 may preferably be independently selected from R groups with the following structural characteristics:
(i) an optionally substituted phenyl group;
(ii) an optionally substituted ethenyl group;
(iii) an ethenyl group conjugated to an electron sink.
The term 'electron sink' means a moiety covalently attached to a compound which is capable of reducing electron density in other parts of the compound. Examples of electron sinks include cyano, carbonyl and ester groups.
The term 'therapeutically removable nitrogen protecting group' means any group which can protect the 10-nitrogen, but which is removable under therapeutic conditions in vivo, that is, removable under conditions which occur or can be caused to occur in vivo and are medically acceptable generally by elimination to produce a N10-C11 imine group or an equivalent, capable of interacting with DNA. The removal of the protecting group should leave the rest of the structure of the PBD unaffected.
Suitable removal techniques include applying light, e.g. with a wavelength of 250 to 400, or 550 nm, changing the ambient pH, or cleavage by the action of an enzyme. One particularly suitable enzyme is nitroreductase, although other suitable enzymes include penicillin V/G amidase, β-lactamase, phosphatase, L-γ-glutamyl transpeptidase, and α-galactosidase . The action of some of these enzymes is described in Jungheim, L.N. and Shepherd, T.A., Design of Antitumour Prodrugs: Substrates for Antibody Targeted Enzymes, Am. Chem . Soc . Chem . Rev. , 1994, 94: 6, 1553 - 1566. Another particularly suitable enzyme is glutathionare transferase, as discussed above.
One possible group is R10 of the formula II:
Figure imgf000009_0001
wherein n is 0 to 3, R(I) is H or R, and R(II) is one or more optional substituents independently selected from N02, OR, or R, where R is as defined in any of the definitions above; and if two substituents R(II) are on adjacent atoms, they may together be of the formula -O- (CH2)m-0-, where m is 1 or 2. R(II) is preferably N02.
If the therapeutically removable group R10 is one which is susceptible to nitroreductase, it may be of the formula III:
Figure imgf000009_0002
wherein n is 0 to 3, RlI> is H or R, and R(III) is one or more optional substituents independently selected from N02, OR or R, where R is as defined in any of the definitions above, and if two substituents R (ni) are on adjacent atoms, they may together be of the formula -O- (CH2)m-0-, where m is 1 or 2.
Another possible therapeutically removable nitrogen protecting group, R10, is of the formula XI:
Figure imgf000010_0001
where R is as defined in any of the definitions above and n is 0 to 3, preferably 0. For this formula, R is most preferably a phenyl group, substituted or unsubstituted. This protecting group may be removable by the action of glutathione transferase (GST) , which is present at high levels in many human tumour cells (see above) .
It is preferred in compounds of formula I that X is O and, independently, that Rlx is H.
If there is a double bond in the C ring, it is preferably between C2 and C3.
Additionally, it is preferred that R6 and R9 are H, and further preferred that R7 and Ra are independently selected from H, OH, and OR. It is further preferred that R2 and R3 are H.
If the compound of formula I is a dimer, the dimer bridge may be of the formula -O- (CH2)p-0-, where p is from 1 to 12, preferably 3 to 9. Further, R6 and R9 are preferably H, and R7 is preferably independently selected from H, OH, and OR.
A second aspect of the present invention provides a method of preparing a compound of formula I as described in the first aspect of the invention wherein XRU ≠ OH, from a corresponding compound la which is a compound of formula I in which XR1 = OH. A product in which XRn is OR may be prepared by direct etherification of compound la. A product in which X is S may be prepared by treatment of compound la with RUSH and a catalyst (generally a Lewis acid such as A1203) . A product in which X is NH may be prepared by treatment of compound la with an amine RlnNH and a catalyst (generally a Lewis acid such as A1203) .
A third aspect of the present invention provides a method of preparing a compound of formula la as described in the second aspect of the invention, by the oxidation of a compound of formula IVa:
Figure imgf000011_0001
wherein the substituents of the compound of formula IVa are the same as for the compound of formula la to be prepared. (For preparation of dimeric compounds, the monomers linked through C8 by -T-R' -T- are both of formula IVa. Similar comments apply to other intermediates in dimer synthesis.) The preferred oxidation method is Swern oxidation.
A fourth aspect of the present invention provides a method of preparing a compound of formula IVa as described in the third aspect of the invention, by reacting a compound of formula Va:
Figure imgf000012_0001
with a compound of formula VI :
Y-R10 (VI) wherein the substituents of the compounds of formulae Va and VI are the same as for the compound of formula IVa to be prepared, and Y is a halogen atom.
If the therapeutically removable nitrogen protecting group is to be of formula II, then it is preferred that the compound of formula VI is a haloformate of the formula Via:
Figure imgf000012_0002
wherein the substituents are as defined for the group of formula II, and Y is a halogen atom.
If the therapeutically removable nitrogen group is to be of formula XI, then it is preferred that the compound of formula VI is a haloformate of the formula VIb: O 00/12507
11
Figure imgf000013_0001
where Y is a halogen atom, and R and n are as defined for formula XI.
A fifth aspect of the present invention provides an alternative synthesis of a compound of formula IVa as described in the third aspect of the invention, by reacting a compound of formula VII:
Figure imgf000013_0002
with a compound of formula VI:
Figure imgf000013_0003
to form a compound of formula VIII:
Figure imgf000013_0004
and then reacting the compound of formula VIII with a compound of formula IXa:
^OH
Figure imgf000013_0005
(e.g. by means of (COCl)2), wherein the substituents for compounds of formulae VI, VII, VIII and IXa are the same as for the compound of formula IVa to be prepared, where Y is a halogen atom. A sixth aspect of the present invention provides a method of preparing a compound of formula la as described in the second aspect of the invention, by the unmasking of a compound of formula IVb:
Figure imgf000014_0001
wherein the substituents of the compound of formula IVb are the same as for the compound of formula la to be prepared, and Q is either S or O and R(rv> are independently selected from Me or Et or may together form -(CH2)q- where q is 2 or 3. (For preparation of dimeric compounds, the monomers linked through C8 by -T-R' -T- are both of formula IVb. Similar comments apply to other intermediates in dimer synthesis.) The preferred unmasking method when Q=S is mercury-mediated unmasking. Unmasking when Q=0 is preferably carried out by the use of acid conditions, e.g. TFA, methanol and water or palladium catalysis.
A seventh aspect of the present invention provides a method of preparing a compound of formula IVb as described in the sixth aspect of the invention, by reacting a compound of formula Vb :
Figure imgf000014_0002
with a compound of formula VI:
Figure imgf000014_0003
wherein the substituents of the compounds of formulae Vb and VI are the same as for the compound of formula IVb to be prepared, and Y is a halogen atom.
If the therapeutically removable nitrogen protecting group is to be of formula II, then it is preferred that the compound of formula VI is a haloformate of the formula Via:
Figure imgf000015_0001
wherein the substituents are as defined for the group of formula II, and Y is a halogen atom.
An eighth aspect of the present invention provides an alternative synthesis of a compound of formula IVb as described in the second aspect of the invention, by reacting a compound of formula VII:
Figure imgf000015_0002
with a compound of formula VI:
Y-R10 (VI ; to form a compound of formula VIII:
Figure imgf000015_0003
and then reacting the compound of formula VIII with a compound of formula IXb:
Figure imgf000015_0004
(e.g. by means of (COCl)2), wherein the substituents for compounds of formulae VI, VII, VIII and IXb are the same as for the compound of formula IVb to be prepared, where Y is a halogen atom.
A ninth aspect of the present invention provides a method of making a compound of formula X:
Figure imgf000016_0001
by cleavage of the therapeutically removable protecting group R10 of a compound of formula I as described in the first aspect of the invention, wherein the substituent groups of the compound of formula X are the same as the substituent groups of the compound I used.
A tenth aspect of the present invention provides a use of a compound of formula I, wherein the therapeutically removable nitrogen protecting group (R10)is enzyme labile, in conjunction with an appropriate enzyme in methods of ADEPT or GDEPT therapy. If the enzyme labile group is susceptible to nitroreductase, then compounds of formula I, may be used in conjunction with nitroreductase enzymes (for example, those isolated from E. coli ) in methods of ADEPT and GDEPT therapy.
An eleventh aspect of the present invention provides a use of a compound of formula I, wherein the therapeutically removable nitrogen protecting group (R10)is photolabile, in conjunction with light of wavelengths between 250 and 400 or 550 nm in methods of PDT.
A twelfth aspect of the invention provides a use for a compound of formula I, where the therapeutically removable nitrogen protecting group (R10) is labile by conditions occurring naturally at specific localised sites in the patient in therapy. Suitable compounds of formula I may be those susceptible to a nitroreductase enzyme when used to treat hypoxic tumour cells, or those susceptible to enzymes which are naturally occurring at specific localised sites, such as glutathione transferase.
The drug produced by the cleavage of the therapeutically removable nitrogen protecting group, in either the tenth or eleventh or twelfth aspect of the invention, may be used for treating cancers or other site-specific diseases where a local increase of toxicity is beneficial to the patient. Cancers that may be treated are solid cancers including ovarian, colonic cancer, renal, breast and bowel CNS, melanoma, as well as leukemias . Such drugs may also be suitable for treating bacterial, viral or parasitic infections by exploiting a unique enzyme produced at the site of the infection which is not natural to the host, or by exploiting an elevation in the amount of an enzyme which does naturally occur in the host.
A thirteenth aspect of the present invention is a pharmaceutical composition comprising a compound of formula I as described in the first aspect of the invention. Pharmaceutical compositions according to the present invention, and for use in accordance with the present invention, may comprise, in addition to the active ingredient, i.e. a compound of formula I, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous or intravenous.
Pharmaceutical compositions for oral administration may be in tablet, capsule, powder or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. Capsules may comprise a solid carrier such as gelatin.
For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
A fourteenth aspect of the present invention provides the use of a compound of formula I as described in the first aspect of the invention, to prepare a medicament for the treatment of neoplastic disease or other site-specific diseases where a local increase of toxicity is beneficial to the patient. The compound of formula I may be provided together with a pharmaceutically acceptable carrier or diluent. The preparation of a medicament is described in relation to the thirteenth aspect of the invention.
Aspects of the invention will now be further described with reference to the accompanying drawings in which:
Fig. 1 is a synthesis scheme according to the present invention; Figs. 2a & 2b are a synthesis scheme for dimers according to the present invention;
Fig. 3 is a synthesis scheme showing an alternative cyclisation, for use in the present invention; Fig. 4 is a graph illustrating the cytotoxicity results for prodrug compound 7 (see example 1), both before and after activation with nitroreductase/NADH in SW1116 cells and LS174T cells; Fig. 5 is a graph illustrating the percentage of compound 11 (see example 3) cleaved by UVA (365 nm) exposure over a 3 hour time course;
Fig. 6 is a graph illustrating the in vi tro cytotoxicity (ICS0; μM) of benzyl DC-81 and compound 11, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in DMF at 1 mM initial drug concentration;
Fig. 7 is a graph illustrating the in vi tro cytotoxicity (ICS0; μM) of benzyl DC-81 and compound 11, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in DMF at 10 mM initial drug concentration;
Fig. 8 is a graph illustrating the in vi tro cytotoxicity (IC50; μM) of benzyl DC-81 and compound 11, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in methanol at a 1 mM initial drug concentration; Fig. 9 is a graph illustrating the in vi tro cytotoxicity (IC50; μM) of DSB-120 and compound 32, with varying irradiation times, against human chronic myeloid leukaemia K562 cells in methanol at a 1 mM initial drug concentration.
Preferred Synthetic Strategies A key step in a preferred route to compounds of formula I is a cyclisation to produce the B-ring, involving generation of an aldehyde (or functional equivalent thereof) at what will be the 11-position, and attack thereon by the 10-nitrogen:
Figure imgf000019_0001
The "masked aldehyde", -CPQ, may be an acetal or thioacetal (e.g. P=Q=SEt or OMe) , which may be cyclic, in which case the cyclisation involves unmasking. Alternatively, the masked aldehyde may be an aldehyde precursor, such as an alcohol, -CHOH, in which case the reaction involves oxidation, e.g. by means of TPAP or DMSO (Swern oxidation) .
The masked aldehyde compound can be produced by condensing a corresponding 2-substituted pyrrolidine with a 2-nitrobenzoic acid:
STEPS
Figure imgf000020_0001
Figure imgf000020_0002
The nitro group can then be reduced to -NH2 and protected by reaction with a suitable agent, e.g. a chloroformate, which provides the therapeutically removable nitrogen protecting group
R10 in the compound of formula
A process involving the oxidation-cyclization procedure is illustrated in Figure 1 (an alternative type of cyclisation will be described later with reference to Figure 3) . If Rxl is other than hydrogen, the compound of formula I, may be prepared by direct etherification of the alcohol la. If X=S, and not O, the alcohol la, or the OR derivative, can be treated with H2S, and a catalyst such as Al203, or by the addition of a thiol, e.g. EtSH. If X is NH then treatment of the alcohol la with the appropriate amine yields the desired compound of formula I.
Exposure of the alcohol (IVa) (in which the pro-10-nitrogen is generally protected as an amide carbamate) to tetrapropylammonium perruthenate (TPAP) /N-methylmorpholine N-oxide (NMO) over A4 sieves results in oxidation accompanied by spontaneous B-ring closure to afford the desired product. The TPAP/NMO oxidation procedure is found to be particularly convenient for small scale reactions while the use of DMSO-based oxidation methods, particularly Swern oxidation, proves superior for larger scale work (e.g. > 1 g) .
The uncyclized alcohol (IVa) may be prepared by the addition of a nitrogen protection reagent of formula VI, which is preferably a chloroformate or acid chloride, to the amino alcohol (Va) , generally in solution, generally in the presence of a base such as pyridine (preferably 2 equivalents) at a moderate temperature (e.g. at 0°C) . Under these conditions little or no O-acylation is usually observed.
The key amino alcohol (Va) may be prepared by reduction of the corresponding nitro compound (Xla) , by choosing a method which will leave the rest of the molecule intact. Treatment of Xla with tin (II) chloride in a suitable solvent, e.g. refluxing methanol, generally affords, after the removal of the tin salts, the desired product in high yield.
Exposure of Xla to hydrazine/Raney nickel (or hydrogentation with a catalyst) avoids the production of tin salts and may result in a higher yield of Va, although this method is less compatible with the range of possible C and A-ring substituents. For instance, if there is C-ring unsaturation (either in the ring itself, or at R2 or R3) , this technique may be unsuitable.
The nitro compound of formula Xla may be prepared by coupling the appropriate o-nitrobenzoyl chloride to a compound of formula IXa, e.g. in the presence of K2C03 at -25 °C under a N2 atmosphere. The o-nitrobenzoyl chloride is synthesised from the o-nitro benzoic acid of formula XII - many of these are commercially available, and the synthesis of some examples has been reported by Althuis (Althuis, T.H. and Hess, H-J, 1977, Synthesis and Identification of the Major Metabolites of Prazosin Formed in Dog and Rat,
Journal of Medi cinal Chemistry 20, 1: 146-266) . Compounds of formula IXa can be readily prepared, for example by olefination of the ketone derived from L-trans-hydroxy proline. The ketone intermediate can also be exploited by conversion to the enol triflate for use in palladium mediated coupling reactions such as the Heck, Stille and Suzuki reactions.
Dimer Synthesis (Figure 2)
PBD dimers may be synthesized using the strategy developed for the synthesis of the protected PBD monomers (Figure 2a). Figure 2 also shows a synthesis route where the dimer linkage is of the formula -O- (CH2)n-0- . The step of dimer formation is normally carried out to form a bis (nitro acid) XII' (Figure 2b).
The bis (nitro acid) XII1 may be obtained by nitrating (e.g. using 70% nitric acid) the bis (carboxylic acid). This can be synthesised by alkylation of two equivalents of the relevant benzoic acid with the appropriate diiodoalkane under basic conditions (Route 2a) . Many benzoic acids are commercially available and others can be synthesised by conventional methods.
An alternative synthesis of the bis (nitro acid) involves oxidation of the bis (nitro aldehyde), e.g. with potassium permanganate. This can be obtained in turn by direct nitration of the bis (aldehyde) , e.g. with 70% HN03. Finally, the bis (aldehyde) can be obtained via Mitsunobu etherification of two equivalents of the benzoic aldehyde with the appropriate alkanediol (Route 2b) .
Alternative Cyclisation (Figure 3)
In figures 1 and 2, the final or penultimate step is an oxidative cyclisation. An alternative, using thioacetal coupling unmasking, is shown in figure 3 (which shows it applied to a dimer, with a dimer linkage of formula -o- (CH2)n-0-) . Mercury- mediated unmasking causes cyclisation to the desired compound (la' ) .
The thioacetal compound may be prepared as shown in figure 3: the thioacetal protected C-ring [prepared via a literature method: Langley, D.R. & Thurston, D.E., J. Organi c Chemistry, 52, 91-97 (1987)] is coupled to the bis (nitro carboxylic acid) core using a literature procedure. The resulting nitro compound cannot be reduced by hydrogenation, because of the presence of the thioacetal group, so the tin (II) chloride method is used to afford the bis(amine). This is then N-protected, e.g., by reaction with a chloroformate or acid chloride, such as p- nitrobenzylchloroformate .
An alternative to thioacetal coupling is the use of acetal coupling. The method is the same as that illustrated in figure 3, but with the thioacetal group replaced by an acetal group
(e.g. -CH(0Me)2). Acid or palladium-mediated unmasking is the preferred method of unmasking to cause cyclisation to the desired compound of formula la or I' a.
GDEPT Vector systems
In general, the vector for use in GDEPT therapies may be any suitable DNA or RNA vectors.
Suitable non-viral vectors include cationic liposomes and polymers. Suitable viral vectors include those which are based upon a retrovirus . Such vectors are widely available in the art. Huber et al . (ibid) report the use of amphotropic retroviruses for the transformation of hepatoma, breast, colon or skin cells. Culver et al . (Science (1992) 256; 1550-1552) also describe the use of retroviral vectors in GDEPT. Such vectors or vectors derived from them may also be used. Other retroviruses may also be used to make vectors suitable for use in the present invention. Such retroviruses include Rous sarcoma virus (RSV) .
Englehardt et al . (Nature Genetics (1993) 4.; 27-34) describe the use of adenovirus-based vectors in the delivery of the cystic fibrosis transmembrane conductance product (CFTR) into cells, and such adenovirus-based vectors may also be used. Vectors utilising adenovirus promoter and other control sequences may be of use in delivering a system according to the invention to cells in the lung, and hence useful in treating lung tumours.
Other vector systems including vectors based on the Molony murine leukaemia virus are known (Ram, Z et al . , Cancer Research (1993) 53; 83-88; Dalton & Treisman, Cell (1992) 68.; 597-612) . These vectors contain the Murine Leukaemia virus (MLV) enhancer cloned upstream at a β-globin minimal promoter. The β-globin 5' untranslated region up to the initiation codon ATG is supplied to direct efficient translation of the enzyme.
Suitable promoters which may be used in vectors described above, include MLV, CMV, RSV and adenovirus promoters. Preferred adenovirus promoters are the adenovirus early gene promoters . Strong mammalian promoters may also be suitable. An example of such a promoter is the EF-lα promoter which may be obtained by reference to Mizushima and Nagata ((1990), Nucl . Acids Res. 18; 5322). Variants of such promoters retaining substantially similar transcriptional activities may also be used.
Other suitable promoters include tissue specific promoters, and promoters activated by small molecules, hypoxia or X-rays.
If nitroreductase is the enzyme of choice for the activation of compounds of formula I, then preferably the enzyme is a non- mammalian nitroreductase such as a bacterial nitroreductase . An E. coli nitroreductase as disclosed in WO93/08288 is particularly preferred. The enzyme may be modified by standard recombinant DNA techniques, e.g. by cloning the enzyme, determining its gene sequence and altering the gene sequence by methods such as truncation, substitution, deletion or insertion of sequences for example by site-directed mutagenesis. Reference may be made to "Molecular Cloning" by Sambrook et al . (1989, Cold Spring Harbor) for discussion of standard recombinant DNA techniques. The modification made may be any which still leaves the enzyme with the ability to reduce the nitro group in suitable compounds of formula I, but alters other properties of the enzyme, for example its rate of reaction or selectivity. In addition, small truncations in the N- and/or C-terminal sequence may occur as a result of the manipulations required to produce a vector in which a nucleic acid sequence encoding the enzyme is linked to the various other vector sequences .
For information on the use of penicillin V/G amidase, and β- lactamase in GDEPT, see Jungheim, L.N. and Shepherd, T.A., Design of Antitumour Prodrugs: Substrates for Antibody Targeted Enzymes, Am. Chem. Soc. Chem. Rev., 1994, Vol 94, No. 6, 1553 - 1566.
ADEPT For applications in ADEPT systems, an antibody directed against a tumour specific marker is linked to the relevant enzyme, which may be modified as described above. The antibody may be monoclonal or polyclonal. For the purposes of the present invention, the term "antibody", unless specified to the contrary, includes fragments of whole antibodies which retain their binding activity for a tumour target antigen. Such fragments include Fv, F(ab') and F(ab')2 fragments, as well as single chain antibodies. Furthermore, the antibodies and fragments thereof may be humanised antibodies, e.g. as described in EP-A-239400.
The antibodies may be produced by conventional hybridoma techniques or, in the case of modified antibodies or fragments, by recombinant DNA technology, e.g. by the expression in a suitable host vector of a DNA construct encoding the modified antibody or fragment operably linked to a promoter. Suitable host cells include bacterial (eg. E. coli ) , yeast, insect and mammalian cells. When the antibody is produced by such recombinant techniques the enzyme may be produced by linking a nucleic acid sequence encoding the enzyme (optionally modified as described above) to the 3' or 5 ' end of the sequence of the construct encoding the antibody or fragment thereof.
PPT
The activation process in PDT can be highly site specific. The direction of a laser beam can be controlled with great precision, and the beam diameter can be reduced to a width far below that of a single cell. Therefore, it can act upon a very limited area, minimising damage to neighbouring tissue.
Ultra-violet light is sufficient to break a range of chemical bonds since the energy spectrum for bond breakage for the majority of organic molecules lies between 250 and 420 kJ/mol, and, for example, 350 nm is equivalent to 340 kJ/mol. For example, a broad range of light-mediated deprotection reactions have been demonstrated including the photochemical deprotection of amino acids, peptides and polysaccharides from their CBZ and o-nitrobenzyl and , 5-dimethoxy-2-nitrobenzyl carbamate forms at wavelengths longer than 350 nm, (Pillai, R. V. N., Photoremovable protecting groups in organic chemistry, Synthesis (1980), 1-26), (Bayley, H., Gasparro, F. and Edelson, R. , Photoactivatible drugs, TIPS (1987) 8, 138-143, (Star, W. M., Light delivery and light dosimetry for photodynamic therapy, Laser in Medi cal Sci ence (1990) 5, 107-113. On the other hand, highly reactive and thus cytotoxic species can also result from relatively low energy activations. For example, a reactive excited state of molecular oxygen, the singlet state, differs in only 90 kJ/mol from its ground triplet state. However, this enables sufficient concentrations of the toxic species to be formed by those sensitisers which absorb at wavelengths longer than 600nm, (Carruth, J. A. S., Clinical applications for photodynamic therapy, J Photochem Photobiol (1991) 9, 396-397).
The main limitation of this approach arises from the physics of light itself and its interaction with human tissue. The ability of light to penetrate tissue has been found to be wavelength- dependent. Penetrating ability increases with increasing wavelength but limitations arise due to light scattering and reflection. In biological tissues the scattering coefficient, for example of red light, is much greater than the absorption coefficient, (Carruth, J. A. S., Clinical applications for photodynamic therapy, J Photochem Photobi ol (1991) 9, 396-397), (Kennedy, J. C. And Pottier, R. H, Endogenous protoporphyrin IX, a clinical useful photosensitiser for photodynamic therapy, J Photochem Photobiol (1992) 14, 275-292) . As a result, photons entering the tissue are scattered several times before they are either absorbed or diffused. Although this might be expected to increase the energy delivered to certain areas, internal reflection results in an exponential decrease of energy flux with increasing distance from the tissue-air interface. These limitations have been partially overcome in the treatment of relatively bulky tumours or when deeper penetration is necessary by the use of multiple interstitial optical fibres.
Several tumour types have been identified as potential targets for PDT. They include head and neck tumours, carcinomas of the bronchus, malignant brain tumours, superficial tumours of the bladder and vascular disease, which have all shown promising responses in the clinic, (Regula, J., Mac Roberts, A. J. , Gorchein, A., Buonaccorsi, Thorpe, S. M. , Spencer, G. M. , Hartfield, A. R. . and Bown, S. G., Photosensitisation and photodynamic therapy of oesophageal, duodenal and colorectal tumours using 5-aminoleavulic acid induced protoporphyrin IX-a pilot study, Gut (1995) 36, 67-75).
The technique of PDT as discussed above can be used in combination with appropriate compounds of formula I when the therapeutically removable nitrogen protecting group is photolabile. The preferred wavelength of UV light used is 250 to 400 or 550 nm.
Applications of the invention
Compounds of the invention can be used in vi tro or in vivo for a range of applications. For example, a number of vector systems for the expression of nitroreductase in a cell have been developed. The further development of such systems (e.g. the development of promoters suitable for specific cell types) requires suitable candidate prodrugs capable of killing cells when activated by nitroreductase. Prodrug compounds of formula I susceptible to nitroreductase may be used in such model systems. The model systems may be in vi tro model systems or in vivo xenograft model systems comprising for example human tumour cells implanted in nude mice. Compounds of formula I susceptible to different enzymes may be used in similar systems which have been appropriately modified.
Compounds of formula I which are not activatable by an enzyme may be tested in vi tro with other suitable forms of activation against panels of different tumour cell types to determine efficacy against such tumour cells. The efficacy of compounds of the invention against a range of tumour cell types may be used as points of reference for the development of further antitumour compounds. Compounds of formula I may also be tested in combination with additional anti-cancer compounds to determine potential combination drug systems, for example combinations which are synergistic.
Compounds of formula I may also be used in a method of treatment of the human or animal body. Such treatment includes a method of treating the growth of neoplastic cells in a patient with neoplastic disease which comprises administering to a patient in need of treatment compounds of formula I as part of an ADEPT, GDEPT or PDT system or treatment with compounds of formula I alone, where neoplastic diseases include leukaemia and solid tumours such as ovarian, colonic, lung, renal, breast, bowel, CNS and melanomas. The treatment can also be the treatment of other site-specific diseases where local increase in toxicity is beneficial to the patient.
It will be understood that where treatment of tumours is concerned, treatment includes any measure taken by the physician to alleviate the effect of the tumour on a patient. Thus, although complete remission of the tumour is a desirable goal, effective treatment will also include any measures capable of achieving partial remission of the tumour as well as a slowing down in the rate of growth of a tumour including its metastases. Such measures can be effective in prolonging and/or enhancing the quality of life and relieving the symptoms of the disease.
Therapies
Methods of ADEPT and GDEPT will now be described with reference to nitroreductase, although other enzymes as previously described could be substituted with appropriate modifications to the methods described.
The basis of PDT has been described above, but the information on the administration of products below also applies to this type of therapy. This information is also relevant to therapies where the prodrug is activated by conditions naturally occurring within the body (e.g. hypoxia, elevated level of GST - see discussion of NPEPT above) .
ADEPT therapy
The antibody/enzyme conjugate for ADEPT can be administered simultaneously but it is often found preferable, in clinical practice, to administer the enzyme/antibody conjugate before the prodrug, e.g. up to 72 hours or even 1 week before, in order to give the enzyme/antibody conjugate an opportunity to localise in the region of the tumour target. By operating in this way, when the prodrug is administered, conversion of the prodrug to the cytotoxic agent tends to be confined to the regions where the enzyme/agent conjugate is localised, i.e. the region of the target tumour. In this way, the premature release of the compound produced by the action of the nitroreductase on the prodrugs of the present invention is minimised.
In ADEPT the degree of localisation of the enzyme/agent conjugate (in terms of the ratio of localized to freely circulating active conjugate) can be further enhanced using the clearance and/or inactivation systems described in WO89/10140. This involves, usually following administration of the conjugate and before administration of the prodrug, the administration of a component (a "second component") which is able to bind to part of the conjugate so as to inactivate the enzyme in the blood and/or accelerate the clearance of the conjugate from the blood. Such a component may include an antibody to the enzyme component of the system which is capable of inactivating the enzyme.
The second component may be linked to a macromolecule such as dextran, a liposome, albumin, macroglobulin or a blood group O erythrocyte so that the second component is restrained from leaving the vascular compartment. In addition, or as an alternative, the second component may include a sufficient number of covalently bound galactose residues, or residues of other sugars such as lactose or mannose, so that it can bind the conjugate in plasma but be removed together with the conjugate from plasma by receptors for galactose or other sugars in the liver. The second component should be designed for use and administered such that it will not, to any appreciable extent, enter the extravascular space of the tumour where it could inactivate localised conjugate prior to and during administration of the prodrug.
In ADEPT systems, the dose of the prodrug and conjugate will ultimately be at the discretion of the physician, who will take into account such factors as the age, weight and condition of the patient. Suitable doses of prodrug and conjugate are given in Bagshawe et al . Antibody, Immunoconjugates, and Radiopharmaceuticals (1991), 4., 915-922. A suitable dose of conjugate may be from 500 to 200,000 enzyme units/m2 (e.g. 20,000 enzyme units/m2) and a suitable dose of prodrug may be from about 0.1 to 200 mg/Kg, preferably from about 10 to about 100 mg/Kg per patient per day.
In order to secure maximum concentration of the conjugate at the site of desired treatment, it is normally desirable to space apart administration of the two components by at least 4 hours. The exact regime will be influenced by various factors including the nature of the tumour to be targeted and the nature of the prodrug, but usually there will be an adequate concentration of the conjugate at the site of desired treatment within 48 hours. The ADEPT system when used with nitroreductase also preferably comprises a suitable co-factor for the enzyme. Suitable co- factors may include a riboside or ribotide of nicotinic acid or nicotinamide .
The antibody/enzyme conjugate may be administered by any suitable route usually used in ADEPT therapy. This includes parenteral administration of the antibody in a manner and in formulations similar to that described below.
GDEPT therapy For use of the vectors in therapy, the vectors will usually be packaged into viral particles and the particles delivered to the site of the tumour, as described in for example Ram et a 1 . ( supra ) . The viral particles may be modified to include an antibody, fragment thereof (including a single chain) or tumour- directed ligand to enhance targeting of the tumour.
Alternatively the vectors may be packaged into liposomes . The liposomes may be targeted to a particular tumour. This can be achieved by attaching a tumour-directed antibody to the liposome. Viral particles may also be incorporated into liposomes. The particles may be delivered to the tumour by any suitable means at the disposal of the physician. Preferably, the viral particles will be capable of selectively infecting the tumour cells. By "selectively infecting" it is meant that the viral particles will primarily infect tumour cells and that the proportion of non- tumour cells infected is such that the damage to non-tumour cells by administration of a prodrug will be acceptably low, given the nature of the disease being treated. Ultimately, this will be determined by the physician.
One suitable route of administration is by injection of the particles in a sterile solution. Viruses, for example isolated from packaging cell lines, may also be administered by regional perfusion or direct intratumoral injection, or direct injection into a body cavity (intracaviterial administration) , for example by intra-peritoneal injection. The exact dosage regime for GDEPT will, of course, need to be determined by individual clinicians for individual patients and this, in turn, will be controlled by the exact nature of the prodrug and the cytotoxic agent to be released from the prodrug. However, some general guidance can be given. Chemotherapy of this type will normally involve parenteral administration of modified virus, and administration by the intravenous route is frequently found to be the most practical.
In GDEPT systems the amount of virus or other vector delivered will be such as to provide a similar cellular concentration of enzyme as in the ADEPT system mentioned above. Typically, the vector will be administered to the patient and then the uptake of the vector by transfected or infected (in the case of viral vectors) cells monitored, for example by recovery and analysis of a biopsy sample of targeted tissue. This may be determined by clinical trials which involve administering a range of trial doses to a patient and measuring the degree of infection or transfection of a target cell or tumour. The amount of prodrug required will be similar to or greater than that for ADEPT systems.
In using a GDEPT system the prodrug will usually be administered following administration of the vector encoding an enzyme. Suitable doses of prodrug are from about 0.1 to 200 mg/Kg, preferably from about 10 to about 100 mg/Kg per patient per day.
Administration of prodrugs
While it is possible for the compounds of formula I to be administered alone, it is preferable to present them as pharmaceutical formulations, for use with any of the above methods. The formulations comprise the compounds, together with one or more acceptable carriers thereof and optionally other therapeutic ingredients, or diluents. The carrier or carriers must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipients thereof, for example, liposomes. Suitable liposomes include, for example, those comprising the positively charged lipid (N [1- (2 , 3-dioleyloxy) propyl] -N, N, N-triethylammonium (DOTMA) , those comprising dioleoylphosphatidylethanolamine (DOPE), and those comprising 3β [N- (n'N' -dimethylaminoethane) - carbamoyl] cholesterol (DC-Choi).
Formulations suitable for parenteral or intramuscular administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats, bacteriocidal Antibiotics and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs . The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, for example Water for Injection, immediately prior to use. Injection solutions and suspensions may be prepared extemporaneously from sterile powders, granules and tablets of the kind previously described.
It should be understood that in addition to the ingredients particularly mentioned above, the formulations may include other agents conventional in the art having regard to the type of formulation in question. Of the possible formulations, sterile pyrogen-free aqueous and non-aqueous solutions are preferred.
The doses may be administered sequentially, eg. at hourly, daily, weekly or monthly intervals, or in response to a specific need of a patient. Preferred routes of administration are oral delivery and injection, typically parenteral or intramuscular injection or intratumoural injection. For methods of PDT dermal or topical administration may be preferred, e.g. subcutaneous injection or creams and ointments, and such methods of administration are well known. The exact dosage regime will, of course, need to be determined by individual clinicians for individual patients and this, in turn, will be controlled by the exact nature of the compound of formula I, but some general guidance can be given. Typical dosage ranges generally will be those described above which may be administered in single or multiple doses. Other doses may be used according to the condition of the patient and other factors at the discretion of the physician.
Examples Embodiments of the present invention will now be described in detail by way of example.
General Experimental Methods
Melting points (mp) were determined on a Gallenkamp P1384 digital melting point apparatus and are uncorrected. Infrared (IR) spectra were recorded using a Perkin-Elmer 297 spectrophotometer. 1H- and 13C- NMR spectra were recorded on a Jeol GSX 270 MHZ FT-NMR spectrometer operating at 20°C +/-l°c Chemical shifts are reported in parts per million (δ) downfield from tetramethylsilane (TMS) . Spin multiplicities are described as: s (singlet), bs (broad singlet), d (doublet), dd (doublet of doublets), t (triplet), q (quartet), p (pentuplet) or m (multiplet) . Mass spectra (MS) were recorded using a Jeol JMS-DX 303 GC Mass Spectrometer (El mode: 70eV, source 117-147°C) . Accurate molecular masses (HRMS) were determined by peak matching using perfluorokerosene (PFK) as an internal mass marker, and FAB mass spectra were obtained from a glycerol/thioglycerol/ trifluoroacetic acid (1:1:0.1) matrix with a source temperature of 180°C. Optical rotations at the Na-D line were obtained at ambient temperature using a Perkin-Elmer 141 Polarimeter. Flash chromatography was performed using Aldrich flash chromatography "Silica Gel-60" (E. Merck, 230-400 mesh). Thin-layer chromatography (TLC) was performed using GF254 silica gel (with fluorescent indicator) on glass plates. All solvents and reagents, unless otherwise stated, were supplied by the Aldrich Chemical Company Ltd. and were used as supplied without further purification. Anhydrous solvents were prepared by distillation under a dry nitrogen atmosphere in the presence of an appropriate drying agent, and were stored over 4A molecular sieves or sodium wire. Petroleum ether refers to the fraction boiling at 60-80°C.
Example 1: Synthesis of a nitroreductase-activated benzyl DC-81 prodrug for ADEPT (7) .
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000035_0003
Synthesis of 4-Benzyloxy-3-methoxybenzoic acid (2)
A solution of benzyl chloride (24.6 ml, 209 mmol, 1.1 eq.) in THF (100 ml) was added dropwise at 0°C over 15 min, to a mechanically stirred solution of 4-hydroxy-3-methoxybenzoic acid (vanillic acid, 1) (30 g, 179 mmol) in THF (90 ml) and 2.0M aq. NaOH (225 ml) . The mixture was allowed to warm to room temperature and then heated under reflux for 48 hours. After cooling, the mixture was washed with hexane (2 x 100 ml) and the THF was removed in vacuo . The remaining aqueous phase was acidified to pH 1 with cone. HC1. The resulting precipitate was collected by filtration, washed with water and dried to afford 4-benzyloxy-3- methoxybenzoic acid (2) as a pale amorphous solid.
Yield (after recrystallisation from EtOAc) 31g (67%); mp 171-172 *C; IR (cm"1) 3700-3200, 2820-3000, 2210, 2140, 1670, 1600, 1580, 1510, 1450, 1430, 1410, 1380, 1340, 1300, 1265, 1220, 1180, 1130- 1110, 1030, 1010; XH NMR (CDCl3+DMSO-ds) δ 7.60 (d, J = 2 Hz, 1H), 7.55 (d, J = 2 Hz ,1H), 7.30-7.44 (m, 5H), 6.90 (d, J = 8.4 Hz, 1H), 5.19 (s, 2H), 3.91 (s, 3H) ; 13C NMR (CDCl3+DMSO-d δ 168.3, 151.7, 148.8, 136.3, 128.5, 128.0, 127.2, 123.7, 123.5, 112.5, 112.2, 70.6, 55.9; MS (El) (m/z, relative intensity) 258 (M+, 20), 91 (100), 79 (3), 65 (10), 51 (3); EI-HRMS m/z 258.0949 (calc'd f°r C15H1A0A m/z 258.0892).
Synthesis of 4-Benzyloxy-5-methoxy-2-nitrobenzoic acid (3)
Method A : A freshly prepared mixture of SnCl4 (5 g, 19.5 mmol) and fuming nitric acid (1.67g, 26.5 mmol) was added dropwise over 5 minutes to a mechanically stirred solution of 2 (4.35 g, 17 mmol) in DCM at -25°C (dry ice/carbon tetrachloride) . The mixture was maintained at the same temperature for a further 15 min, quenched with water (150 ml), and allowed to warm to room temperature After the organic layer was separated, the aqueous layer was extracted with EtOAc (2 x 75 ml) . The combined organic phase was dried (MgSO and evaporated in vacuo to afford a light brown gum which was recrystallised to form 3 as pale yellow needles. Yield = 4.16 g (82%)
Method B : 4-benzyloxy-3-methoxybenzoic acid (8.5g, 32.9 mmol) was added in small portions over 30 minutes to stirred solution of 70% nitric acid (100 ml) . When addition was complete the reaction mixture was allowed to warm to 15 °C and maintained at that temperature for a further 30 min. The reaction mixture was then poured onto ice and the resultant precipitate was collected by filtration, washed with ice-cold water and dried to afford the nitrated product 3 as a yellow powder.
Yield (after recrystallisation from EtOAc/hexane) 7.8 g (78%); mp 182-185°C; IR (cm"1) 3400-3200, 2820-2930, 1670, 1600, 1580, 1550, 1510, 1450, 1410, 1400, 1370, 1350, 1330, 1265, 1210, 1180, 1160, 1055, 1005; 1H-NMR (CDCl3+DMSO-d6) δ 7.36-7.45 (m, 6H), 7.16 (s, 1H), 5.19 (s, 2H), 3.95 (s, 3H); 13C-NMR (CDCl3+DMSO-de) δ 167.3, 152.7, 149.0, 140.9, 135.2, 128.4, 127.6, 127.3, 111.1, 71.2,
56.5; MS (El) (m/z, relative intensity) 303 (M+, 64), 286 (36), 273 (5), 259 (5), 181 (7), 123 (8), 105 (13), 91 (100), 77 (8), 65 (63), 51 (23); EI-HRMS m/z 303.0824 (calc'd for C15H13N06 m/z 303.0743) .
Synthesis of (2S) -N- (4-benzyloxy-5-methoxy-2-nitrobenzoyl) pyrrolidine-methanol (4)
A catalytic amount of DMF (3 drops) was added to a solution of 4- benzyloxy-5-methoxy-2-nitrobenzoic acid (3) (3.5 g, 11.55 mmol) and oxalyl chloride (1.75 g, 13.56 mmol, 1.2 eq) in dry acetonitrile (30 ml) . The solution was allowed to stir under nitrogen overnight. The resulting acid chloride solution was then added dropwise over 30 minutes to a stirred suspension of pyrrolidinemethanol (1.168 g, 11.56 mmol, 1 eq) and K2C03 (3.675 g, 26.63 mmol, 2.3 eq) in acetonitrile (80 ml) at -25°C under a nitrogen atmosphere. The reaction mixture was stirred at the same temperature for a further 1 hour and then allowed to return to room temperature and quenched with water (200 ml) . The solution was then extracted with chloroform (4 x 100 ml) and the combined organic phase was washed with IM HC1 (2 x 50 ml) , water (2 x 75 ml), brine (2 x 50 ml) and water (100 ml), dried (MgSO and the solvent was evaporated in vacuo to afford a light yellow oil. The product was further purified by means of flash chromatography (5% MeOH/CHCl3) to afford (4) as a pale yellow oil which slowly crystallised on standing.
Yield 3.92 g (88%); [ ]20 D: -62.3° (c 0.45, CHC13) ; IR (cm"1) 3500- 3250, 2860, 2910, 1600, 1580, 1520, 1455, 1430, 1370, 1330, 1275, 1220, 1210, 1185, 1150, 1110, 1060, 1025, 1000; *H-NMR (CDC13) δ 7.76 (s, 1H), 7.34-7.47 (m, 5H) , 6.83 (s, 1H) , 5.21 (s, 2H), 4.38-4.40 (bs, 1H) , 3.98 (s, 3H) , 3.80-3.95 (m, 2H), 3.15-3.20 (m, 2H), 2.17-2.21 (m, 2H) , 1.69-1.89 (m, 2H) ; 13C-NMR (CDC13) δ 155.0, 148.1, 136.9, 135.2, 128.8, 128.5, 127.9, 127.6, 109.2,
109.1, 71.3, 66.1, 61.5, 56.8, 49.5, 28.4, 24.4; MS (El) (m/z, relative intensity) 386 (M+, 4) , 368 (6), 355 (39) , 286 (90) , 121 (4) , 91 (100) , 65 (4) ; EI-HRMS m/z 386.1531 (calc'd for C20H22N2O6 m/z 386.1478) .
Synthesis of (2S) -N- (2-amino-4-benzyloxy-5-methoxybenzoyl) pyrrolidine-methanol (5)
Method A : A solution of the nitro compound (4) (1. 4 g, 3.62 mmol) and SnCl2.2H20 (4.58 g, 20.08 g, 5.5 eq) in methanol (70 ml) was heated at reflux for 45 min. The solvent was removed by evaporation in vacuo and the resulting brown oil was diluted with EtOAc (150 ml), treated with sat. aq. NaHC03 (150 ml) and allowed to stir under N2 overnight. The resulting suspension was filtered through Celite, the organic phase was then separated and washed with brine (2 x 100 ml) , dried (MgS04) and the excess solvent was finally evaporated in vacuo. The residual light brown oil was further purified by column chromatography (5% MeOH/CHCl3) to afford the amine (5) as a bright yellow oil. Yield = 0.82 g (62%) .
Method B : Hydrazine hydrate (4.53 g, 90.67 ml, 5 eq) was added dropwise to a solution of the nitro compound 4 (7.0 g, 18.13 mmol) in dry methanol (20 ml) and a catalytic amount of Raney Ni (0.544 g) over antibumping granules whilst a gently reflux was maintained. The mixture was heated at reflux for a further 15 minutes when TLC (5% MeOH/CHCl3) indicated that the reaction had gone to completion. The Ni catalyst was then removed by filtration through Celite and the solvent removed by evaporation in vacuo. The product was further purified by flash chromatography (5% MeOH/CHCl3) to afford the amine 5 as a bright yellow unstable oil which required storage at low temperature.
Yield 5.2 g (81%); [α]2°D : -15.4° (c 0.13, CHC13) ; H-NMR (CDC13) δ 7.28-7.42 (m, 5H) , 6.76 (s, 1H) , 6.26 (s, 1H) , 5.09 (s, 2H) , 4.37-4.42 (bs, 1H) , 3.78 (s, 3H) , 3.58-3.77 (m, 4H) , 2.04-2.10 (m, 2H), 1.69-1.92 (m, 2H) ; 13C-NMR (CDC13) δ 171.8, 151.0, 141.2, 136.5, 128.6, 127.6, 127.1, 112.9, 102.9, 70.6, 66.8, 60.9, 57.1, 51.0, 28.5, 24.9; IR (cm"1) 3500-3100, 2950, 1680, 1620, 1590, 1510, 1450, 1425, 1400, 1330, 1260, 1230, 1170, 1165, 1130, 1100, 1080, 1055, 1030; MS (El) (m/z, relative intensity) 356 (M+, 100), 256 (68), 237 (14), 226 (4), 164 (6), 138 (15), 100 (8), 91 (96), 84 (22), 65 (8); EI-HRMS m/z 356.1785 (calc'd for C20H24N2O4 m/z 356.1736) .
Synthesis of (2S) -N- [2- (p-nitrobenzyloxy) carboxamido-4-benzyloxy- 5-methoxybenzoyl] pyrrolidinemethanol (6) A solution of 4-nitrobenzyl chloroformate (0.6 g, 2.8 mmol, 1 eq) in dry DCM (15 ml) was added dropwise over 20 minutes to a solution of the amine (5) (1 g, 2.8 mmol) and pyridine (0.44 g, 5.6 mmol, 2 eq) in dry DCM (20 ml) at 0°C under N2. After the addition was complete the solution was allowed to stir under for a further 1.5 hours. The reaction mixture was then washed with sat. aq. CuS04 (2 x 100 ml), water (150 ml), brine (100 ml), dried (MgS04) and the excess solvent removed under vacuum. The resulting oil was subjected to flash chromatography (3% MeOH/CHCl3) to afford the protected amine (6) as a pale yellow oil.
Yield 1.16 g (77%); IR (cm"1) 3500-3100, 2950, 1680, 1620, 1590, 1510, 1450, 1425, 1400, 1330, 1260, 1230, 1170, 1165, 1130, 1100, 1080, 1055, 1030; ^-NMR (CDC13, rotamers) δ 9.04 (s, 1H) , 8.20 (d, J=2Hz, 2H), 7.81 (s, 1H) , 7.28-7.55 (m, 5H) , 6.86 (s, 1H) , 5.14-5.24 (bs, 4H) , 4.39 (bs, 1H) , 3.82 (s, 3H) , 3.55-3.89 (m, 4H), 2.04-2.14 (m, 2H) , 1.70-1.87 (m, 2H) ; 13C-NMR (CDC13) δ 170.7, 153.2, 150.3, 147.5, 143.6, 136.1, 128.7-127.7, 123.7, 111.5, 106.0, 70.6, 65.2, 60.9, 56.5, 51.7, 28.2, 25.1; MS (FAB) (m/z, relative intensity) 535 (MH+, 4), 435 (2), 356 (3), 256 (9), 192 (2), 185 (4), 166 (3), 136 (13), 120 (5), 102 (26), 91 (100) , 84 (5) . Synthesis of (llaS) -8-benzyloxy-7-methoxy-l ,2 , 3, 10 , 11 , 11a- hexahydro-ll-hydroxy-10- (p-nitrobenzyloxy) carboxy-5H-pyrrolo [2 , 1- c] [1, 4] -benzodiazepine-5-one (7)
A solution of the carbamate 6 (0.4 g, 0.75 mmol), NMO (0.131 g, 1.12 mmol, 1.5 eq) over 4A molecular sieve (0.375 g) in a solvent mixture of dry DCM:CH3CN (9:3 ml) was allowed to stir at room temperature under nitrogen for 15 min. A portion of TPAP (13 mg) was then added and the solution was allowed to stir for a further 2 hours. TLC (2% MeOH/CHCl3) indicated that reaction incomplete and a further amount of NMO (65 mg, 0.845 mmol, 0.75 eq) and TPAP (6.5 mg) was added. After 15 min, TLC indicated a complete loss of starting material. The molecular sieve was removed by filtration through Celite and the solvent was removed under reduced pressure. The black residue was purified by column chromatography (1% MeOH/CHCl3 followed by EtOAc : Petroleum ether, 95:5) to afford the cyclised final product 7.
Yield 0.124 (31%); IR (cm"1) 3600-3200, 2820-3000, 1710, 1600, 1510, 1450, 1430, 1400, 1370, 1350, 1305, 1270, 1220, 1120-1100, 1050, 1030, 1010; ^- MR (CDC13, rotamers) δ 8.10 (d, J = 8.43 Hz, 2H) , 7.27-7.53 (m, 6H) , 7.20 (d, J = 8.4 Hz, 2H) , 6.70 (s, 1H) , 5.62 (d, J = 10.2 Hz ,1H), 5.07 (d, J = 10.7 Hz, 4H) , 3.94 (s, 3H) , 3.52-3.72 (m, 3H) , 2.04-2.10 (m, 4H) ; 13C-NMR (CDC13) δ 166.83, 149.26, 142.85, 136.03, 128.79-127.17, 126.38, 114.58, 112.53, 111.00, 105.92, 86.23, 71.12, 66.21, 59.90, 56.21, 47.32, 46.42, 28.68, 23.05; MS (FAB) (m/z, relative intensity) 535 (M+
+2, 2), 353 (6), 337 (10), 286 (5), 256 (3) , 241 (2), 228 (2), 192 (3) , 136 (7) , 91 (100); EI-HRMS m/z 533.1813 (calc'd for C28H27N308 m/z 533.1798) . Example 2. Synthesis of a Benzyl DC-81 prodrug for the PGA ADEPT 191
Figure imgf000041_0001
Synthesis of (2S) -N- [2-phenyl acetamido-4-benzyloxy-5- methoxybenzoyll pyrrolidine methanol (8) A catalytic amount DMF ( 4 drops) was added to a solution of phenyl acetic acid (0.42 g, 3.3 mmol, 1.2 eq) and oxalyl chloride (0.51 g, 3.96 mmol, 1.4 eq) in dry acetonitrile (10 ml) and the reaction mixture was allowed to stir overnight under nitrogen. The resulting acid chloride was added dropwise over 20 minutes to a mixture of the amine (5) (1 g, 2.8 mmol) and K2C03 (0.97 g,
7.02 mmol, 2.5 eq) in dry acetonitrile (60 ml) at -25°C under N2. The reaction mixture was allowed to stir for a further 2 hours at -25°C and then allowed to warm to room temperature. The reaction mixture was diluted with water (200 ml) and it was extracted with CHC13 (4 x 100 ml) . The combined organic phase was washed with IM HC1 (2 x 50 ml), water (2 x 75 ml) and brine (100 ml) and dried over MgS04. The solvent was removed under vacuum and the residual oil was further purified by flash chromatography (5% MeOH/CHCl3) to afford the phenylacetamide (8) as a pale oil.
Yield 0.91 g (68%); IR (cm"1) 3400, 2093, 1660, 1613, 1519, 1495, 1454, 1435, 1401, 1345, 1262, 1218, 1175, 1101, 1028, 1003, 969; 1H-NMR (CDC13, rotamers) δ 9.27 (s, 1H) , 7.90 (s, 1H) , 7.26-7.48 (m, 10H) , 6.77 (s, 1H) , 5.11 (s, 2H) , 4.19-4.29 (bs, 1H) , 3.78 (s, 3H) , 3.65 (s, 2H), 3.35-3.60 (m, 4H), 1.81-2.08 (m, 2H) , 1.62-1.81 (m, 2H); "C-NMR (CDCl3) δ 169.8, 149.9, 145.0, 136.2, 134.5, 129.3-127.3, 110.9, 107.8, 70.6, 66.2, 61.2, 56.5, 51.2, 44.8, 28.3, 24.9; MS (El) (m/z, relative intensity) 474 (M+, 7), 374 (44), 284 (7), 256 (9), 228 (3), 166 (9), 105 (14), 102 (25), 91 (100), 84 (5); EI-HRMS m/z 474.2142 (calc'd for C28H30N2O5 m/z 474.2155) .
Synthesis of (llaS) -8-benzyloxy-7-methoxy-l ,2 , 3, 11 , 11a- pentahydro-ll-hydroxy-10-phenylacetyl-5H-pyrrolo [2 , 1-c] [1,4]- benzodiazepine-5-one (9)
A solution of the phenylacetamide (8) (0.5 g, 1.05 mmol) and NMO (0.184 g, 1.57 mmol, 1.5 eq) over molecular sieve (0.525 g) in a solvent system consisting of dry DCM and CH3CN (9:3 ml) was allowed to stir for 15 minutes at room temperature under nitrogen. TPAP (19 mg, 5% molar) was then added and the mixture was stirred for 1 hour when TLC (5% MeOH/CHCl3) indicated complete consumption of starting material. The reaction mixture was filtered and evaporated in vacuo . The product was further purified by flash chromatography (2% MeOH/CHCl3) to afford the protected PBD 9 as an opaque oil.
Yield 0.15 g (30%); [α]20 D: +1325° (c 0.04, CHC13) ; IR (cm"1) 3400, 2956, 2927, 2094, 1631, 1553, 1514, 1454, 1433, 1407, 1379, 1352, 1278, 1219, 1202, 1181, 1136, 1067, 1009, 752; XH-NMR (CDC13, rotamers) δ 7.19-7.38 (m, 11H) , 6.47 (s, 1H) , 5.76 (d, J = 10.1 Hz, 1H), 4.87 (d, J = 12.1 Hz, 2H) , 3.95 (s, 3H) , 3.42-3.67 (m, 3H) , 2.62 (s, 2H), 1.98-2.11 (m, 4H) ; "C-NMR (CDC13) δ 173.56, 166.54, 149.74, 135.81, 134.84, 130.94-126.56, 114.77, 111.45, 84.38, 70.60, 59.87, 56.28, 46.41, 28.54, 22.67; MS (El) (m/z, relative intensity) 472 (M+, 40), 374 (25), 352 (15), 326 (6), 284 (7), 136 (5), 91 (100), 70 (17); EI-HRMS m/z 472.1944 (calc'd for C28H28N205 m/z 472.1998).
Example 3. Synthesis of a photolabile Benzyl DC-81 prodrug (11)
Figure imgf000043_0001
Synthesis of (2S) -N- [2- (2 ' -nitro-4 ' , 5 ' - dimethoxybenzyloxy) carboxamido- -benzyloxy-5-methoxybenzoyl ] pyrrolidinemethanol (10)
A solution of 4 , 5-dimethoxy-2-nitrobenzyl chloroformate (NVOC-Cl) (0.77 g, 2.8 mmol, 1 eq) in dry DCM (10 ml) was added dropwise over 15 minutes to a solution of the amine (5) (1 g, 2.8 mmol) and pyridine (0.44 g, 5.6 mmol, 2 eq) in dry DCM at 0"c under N2. The reaction mixture was then stirred at 0°C for a further 2.5 hours when TLC (3% MeOH/CHCl3) indicated completion of the reaction. The solution was then washed with aq. sat. CuS04 (2 x 75 ml), water (2 x 100 ml), brine (150 ml) and dried over MgS04. The excess solvent was removed under reduced pressure to give the crude product. Further purification by column chromatography (5% MeOH/CHCl3) afforded 10 as a yellow oil.
Yield 1.45 g (87%); [α]20 D: -175.5° (c 0.225, CHC13) ; IR (cm"1) 4330, 4253, 3428, 2924, 2854, 1711, 1626, 1513, 1463, 1377, 1266, 1216, 1173, 761, 722; XH-NMR (CDC13, rotamers) δ 8.90 (s, 1H) , 7.82 (s, 1H), 7.73 (s, 1H) , 7.26-7.47 (m, 6H) , 6.85 (s, 1H) , 5.58 (d, J = 15 Hz, 2H), 5.16 (s, 2H) , 4.29-4.39 (bs, 1H) , 3.95-3.99 (s, 6H), 3.84 (s, 3H) , 3.49-3.72 (m, 4H) , 1.86-2.17 (m, 2H) , 1.72-1.86 (m, 2H) ; 13C-NMR (CDCl3) δ 153.7, 153.2, 150.4, 148.1, 144.5, 139.6, 136.1, 128.5, 128.1, 127.8, 127.6, 111.5, 110.8, 109.9, 108.1, 70.7, 66.4, 63.7, 62.7, 61.0, 56.7-56.4, 51.6, 28.3; MS (FAB) (m/z, relative intensity) 596 (M++l, 16), 356 (6), 256 (19), 196 (95), 181 (21), 166 (20), 149 (14), 102 (32), 91 ( 100 ) , 87 ( 10 ) , 73 ( 44 ) , 61 ( 22 ) , 57 ( 31 ) .
Synthesis of (llaS) -8-benzyloxy-7-methoxy-l ,2 , 3, 10, 11 , 11a- hexahydro-11-hydroxy-10- (2 ' -nitro-4 ' , 5 ' -dime hoxybenzyloxy) carboxy-5H-pyrrolo [2 , 1-c] [1, 4 ] -benzodiazepine-5-one (11) A solution of the NVOC protected amine, 10 (0.4 g, 0.67 mmol) and NMO (0.236 g, 20.17 mmol, 3 eq) over molecular sieve (0.335 g) in a mixture of dry DCM:CH3CN (9:3 ml) was allowed stir for 15 minutes at room temperature under N2. TPAP (23 mg, 10% molar) was then added to the reaction mixture and it was allowed to stir for a further 2 hours when the reaction was found to be complete by TLC (2% MeOH/CHCl3) . The molecular sieve was removed by filtration through Celite and the resulting solution was evaporated in vacuo . The resulting dark oil was subjected to flash chromatography (1% MeOH/CHCl3) to give the photolabile protected PBD (11) .
Yield 0.21 g (49%); [α]20 D : 212.5° (c 0.08, CHC13) ; IR (cm"1) 4329, 4258, 3405, 2925, 2361, 1713, 1620, 1602, 1579, 1514, 1463, 1377, 1342, 1277, 1219, 1103, 1065, 1012, 968, 870, 791, 722; E- NMR (CDC13, rotamers) δ 7.66-7.71 (s, 1H) , 7.11-7.39 (m, 6H) , 6.80-6.92 (s, 1H) , 6.32 (s, 1H), 5.14-5.67 (m, 5H), 3.85-4.15 (m, 9H), 3.49-3.67 (m, 3H) , 2.01-2.15 (m, 4H) ; 13C-NMR (CDC13) δ 170.41, 166.73, 194.97, 149.03, 135.92, 128.67-126.30, 124.52, 114.64, 112.24, 110.87, 109.82, 108.92, 107.97, 86.02, 71.51, 66.38, 65.03, 60.40, 56.48-56.19, 46.67, 28.70, 26.41, 23.65, 21.05; IR (cm"1) 3600-3100, 3020, 2400, 2105, 1765, 1640, 1525, 1430, 1385, 1350, 1310, 1280, 1215, 1170, 1110, 1045; MS (FAB) (m/z, relative intensity) 594 (MH+, 9), 353 (5), 337 (4), 282 (6), 256 (4), 241 (8), 196 (100), 180 (15), 166 (16), 151 (13), 136 (6), 123 (5), 105 (17), 91 (98); EI-HRMS m/z 593.2010 (calc'd for C30H31N3O10 m/z 593.2042). Example 4. Synthesis of a Benzyl tomaymycin prodrug for nitroreductase ADEPT (147)
HOOC HOOC HOOC HOOC
H
Figure imgf000045_0001
12 13 14 15
MeOOC
Figure imgf000045_0003
Figure imgf000045_0002
16 17 18
Figure imgf000045_0004
19 20
Figure imgf000045_0005
23 24 Synthesis of (2S, 4S) -N- (Allyloxycarbonyl) -4-hydroxypyrrolidine- 2-carboxylic acid (13)
A solution of allyl chloroformate (33.17 g, 275 mmol, 1.2 eq) in THF (30 ml) was added dropwise over 30 minutes to a suspension of trans-4-hydroxy proline (30 g, 229 mmol) in THF (150 ml) and water (150 ml) at pH 9 (adjusted with 4M NaOH) and at 0°C. The reaction mixture was allowed to stir at 0°C and pH 9 for a further 1 hour. The aqueous layer was separated and saturated with NaCl, washed with EtOAc (4 x 100 ml) and the pH of the aqueous phase was adjusted to 2 with cone. HCl . The resulting oil was extracted with EtOAc(3 x 100 ml). The combined organic phase was dried (MgSO and evaporated in vacuo to afford the Alloc protected hydroxy proline (13) , as a thick transparent oil which was used for the next stage without further purification.
Yield 41 g (84%); [α]2°D : -236° (c 0.25, CHCl3) ; IR (cm"1) 3429,
2522, 2340, 2258, 2130, 1688, 1437, 1415, 1345, 1277, 1222, 1177, 1133, 1085, 1047, 1025, 994, 827, 768; XH-NMR (CDC13, rotamers ) δ 5.80-5.99 (m, 1H) , 5.13-5.34 (m, 2H) , 4.52-4.63 (m, 2H) , 4.37- 4.44 (m, 2H), 3.53-3.60 (m, 2H) , 2.00-2.36 (m, 2H) ; 13C-NMR (CDC13) δ 174.7, 174.5, 154.8, 154.5, 132.8, 132.7, 117.1, 116.7, 69.2, 68.5, 65.7, 65.6, 57.9, 57.6, 54.9, 54.4, 38.9, 38.2; MS (El) (m/z, relative intensity) 215 (M+, 14), 170 (100), 130 (95), 126 (34), 108 (35), 68 (51), 56 (21); EI-HRMS m/z 215.0759 (calc'd for C9H13N05 m/z 215.0743).
Synthesis of (2S) -N- (Allyloxycarbonyl) -4-oxopyrrolidine-2- carboxylic acid (14)
The alloc protected hydroxy proline (13) (18 g, 83.72 mmol) was dissolved in acetone (1260 ml). The Jone's reagent
[Cr03: 26.6g/H2S04: 21.3 ml and the solution was made up to 100 ml with water] (87 ml) was added over 10 min. The resulting mixture was stirred for a further 45 minutes when the excess oxidant was quenched with methanol (15 ml). The green chromium salts were removed by filtration through Celite and the filtrate was diluted in CHC13 (1000 ml) . The combined organic phase was washed with brine several times (5 x 500 ml) and the solvent was evaporated under reduced pressure, to yield the ketone 14 which was used without any purification.
Yield 14.68 (82%); IR (cπT1) 3471, 3020, 2932, 2610, 2041, 1761, 1691, 1649, 1547, 1411, 1343, 1266, 1195, 1164, 1128, 972, 939, 877, 759; ^-NMR (CDC13, rotamers) δ 9.20 (bs, 1H) , 5.86-6.00 (m, 1H), 5.21-5.35 (m, 2H) , 4.87-4.91 (m, 2H) , 4.87-4.91 (m, 1H) , 3.87-3.99 (s, 2H), 2.68-2.95 (m, 2H); 13C-NMR (CDC13) δ 207.6, 207.1, 175.8, 175.3, 155.3, 154.2, 131.9, 118.4, 118.2, 67.0, 66.8, 55.8, 52.5, 52.3, 41.0, 40.3; MS (El) (m/z, relative intensity) = 213 (M\ 19), 168 (81), 152 (8), 128 (100), 112
(15), 100 (37), 96 (36), 68 (14), 58 (13), 56 (37); EI-HRMS m/z 213.0641 (calc'd for C9HX1N05 m/z 213.0637).
Synthesis of (2S) - (E, Z) -N- (Allyloxycarbonyl) -4- ethylidenepyrrolidine-2-carboxylic acid (15) A mixture of NaH (60% suspension) (4 g, 100 mmol, 4 eq) and ethyl triphenylphosphonium bromide (37 g, 100 mmol, 4 eq) in freshly distilled THF (250 ml) was refluxed for 4 hours under nitrogen. A solution of the ketone 14 (5.325 g, 25 mmol) in freshly distilled THF (50 ml) was added dropwise over 40 minutes and the reaction mixture was refluxed overnight under nitrogen. It was then cooled and poured into a 5%w/v aq. solution of NaHC03 (500 ml) and stirred for 10 min. The mixture was then washed with EtOAc (2 x 250 ml). The aqueous layer was acidified to pH 3 with dil. HC1 (vigorous effervescence was observed) . The emulsion was extracted with EtOAc (4 x 200 ml) and the combined organic phase was washed with brine (250 ml) and dried (MgS04) . The solvent was then evaporated in vacuo to give an orange oil which was used in the next stage without further purification. Crude yield 3.81 g (68%)
Synthesis of Methyl- (2S) - (E, Z) -N- (Allyloxycarbonyl) -4- ethylidenepyrrolidine-2-carboxylate (16)
Oxalyl chloride and DMF (4 drops) was added to a solution of the Wittig product 15 (3.5 g, 16 mmol) in dry toluene (15 ml) and it was stirred overnight at room temperature and under N2. Freshly distilled methanol (20 ml) was then added and the mixture was stirred for a further 4 hours when TLC (EtOAc : Petroleum ether, 1:1) revealed complete loss of starting material. The solvent was then evaporated in vacuo . The resulting oil was dissolved in EtOAc (60 ml) and the solution was washed with sat. aq. NaHC03 (4 x 25 ml), brine (50 ml), dried (MgSO and the solvent was removed under reduced pressure to afford the esterified Wittig product (16) .
Yield 3.48 g (94%); [α]20 D : -377.7° (c 0.045, CHC13) ; IR (cm"1) 4214, 3452, 3019, 2955, 2862, 2400, 1746, 1704, 1649, 1437, 1410, 1343, 1310, 1274, 1215, 1179, 1120, 1072, 1029, 991, 932, 755; XH-NMR (CDC13, rotamers) δ 5.83-6.00 (m, 1H) , 5.16-5.48 (m, 3H) , 4.56-4.68 (m, 3H), 4.05-4.14 (s, 2H), 3.87-3.99 (m, 3H), 2.53- 2.97 (m, 2H), 1.60-1.63 (m, 3H); 13C-NMR (CDC13) (E/Z isomers) δ 172.9, 154.8, 134.1, 134.0, 133.1, 133.0, 132.8, 132.7, 118.2,
118.1, 117.8, 117.7, 117.5, 117.4, 117.1, 66.1, 66.0, 65.9, 58.8,
58.6, 58.4, 52.3, 51.1, 50.6, 48.3, 47.7, 36.6, 35.7, 32.6, 31.7,
14.7, 14.6, 14.4; MS (El) (m/z, relative intensity) 239 (M+, 4), 180 (70), 154 (100), 136 (55), 94 (33), 80 (10), 67 (19), 59 (8); EI-HRMS m/z 239.1030 (calc'd for C12H17N04 m/z 239.1158).
Synthesis of (2S) - (E,Z) -N- (Allyloxycarbonyl) -4- ethylidenepyrrolidinemethanol (17)
A solution of the Alloc protected C-Ring ester (16) (2.5 g, 10.46 mmol) was dissolved in freshly distilled THF (50 ml) and stirred under nitrogen at 0°C. Lithium borohydride (0.4 g, 17.95 mmol, 1.7 eq) was added slowly and in small portions to that solution (effervescence was observed) . The mixture was allowed to warm to room temperature and stirred for a further 4 hours when the reaction was complete as it was indicated by TLC (EtOAc : Petroleum ether, 1:1) . It was then cooled back down to 0°C and the reaction was stopped by the addition of water (40 ml) dropwise over 15 min. The mixture was neutralised with 2M HC1 (40 ml) which was added very slowly and vigorous effervescence occurred. It was then extracted with EtOAc (3 x 100 ml), the combined organic phase was washed with water (150 ml) and brine (100 ml), dried (MgSO and the solvent was removed under reduced pressure to afford an oil which was further purified by flash chromatography (EtOAc: Petroleum ether, 60:40) to give the pure product 17.
Yield 1.57 g (71%); IR (cm"1) 3425, 2947, 2864, 2085, 1678, 1547, 1536, 1411, 1349, 1308, 1236, 1203, 1114, 1047, 975, 931, 883,
851, 769; XH-NMR (CDC13, rotamers ) δ 5.87-6.01 (m, 1H) , 5.19-5.40 (m, 3H), 4.59-4.61 (s, 2H) , 3.64-4.15 (m, 5H) , 2.37-2.80 (m, 2H), 1.62-1.64 (m, 3H) ; "C-NMR (CDC13) (E/Z isomers) δ 156.3, 154.5, 133.1, 132.8, 131.8, 131.7, 130.9, 130.7, 128.7, 128.6, 125.5, 117.5-115.9, 66.1, 65.9, 65.7, 64.6, 60.4, 59.9, 57.9,
57.3, 51.6, 51.3, 48.3, 48.1, 34.7, 34.4, 34.2, 34.1, 30.6, 30.3, 29.9, 29.5, 25.6, 14.5, 14.4, 14.2.
Synthesis of (2S) - (E, Z) -N- (Allyloxycarbonyl) -4-ethylidene-O- ( tez-fc-butyldimethylsilyl) -pyrrolidinemethanol (18) Tert-butyldimethylsilyl chloride (1.11 g, 7.37 mmol, 1.2 eq) was added to a solution of the hydroxy compound 17 (1.3 g, 6.36 mmol) and imidazole (1.05 g, 15.41 mmol, 2.5 eq) in dry DMF (3 ml) and the reaction mixture was allowed to stir overnight under N2. The reaction mixture was then saturated with water (200 ml) and extracted with EtOAc (3x100 ml) . The organic layer was washed with water (100 ml), brine (150 ml), dried (MgSO and excess solvent was evaporated in vacuo to give a light brown oil which was subjected to flash chromatography (EtOAc : Petroleum ether, 50:50) to afford the pure silyl ether (18).
Yield 1.54 g (75%); IR (cm"1) 3416, 2093, 1642, 1406, 1253, 1189, 1104, 775; ^- MR (CDC13, rotamers) δ 5.89-5.95 (m, 1H) , 5.18-5.34 (m, 3H),4.62 (s, 2H) , 3.53-4.08 (m, 5H) , 2.49-2.70 (m, 2H) , 1.43- 1.59 (m, 3H) , 0.91 (bs, 9H) , 0.02-.003 (bs, 6H) ; "C-NMR (CDC13) (E/Z isomers) δ 162.5, 154.4, 136.2, 135.1, 133.1, 130.9, 128.8, 117.4, 117.1, 116.7, 116.1, 65.9, 65.7, 65.5, 63.9, 63.6, 63.1,
58.6, 58.2, 58.1, 51.2, 48.3, 48.1, 36.5, 34.6, 33.9, 31.8, 31.4,
30.4, 30.3, 29.7, 29. 2, 29.1, 22.7, 18.1, 18.0, 14.5, 14.4, 14.1, -3.6, -5.4; MS (El) (m/z , relative intensity) 325 (M+, 6), 310 (3), 268 (100), 256 (6), 240 (18), 224 (7), 182 (22), 180 (38), 168 (10 ), 154 (5), 136 (24), 115 (8), 94 (6), 75 (13), 73 (21), 57 (17); EI-HRMS m/z 325.2003 [ (M-isobutyl) m/z 268.1233] (calc'd for C17H31N03 m/z 325.2073 [(M-isobutyl) /z 268.1369).
Synthesis of (2S) - (E, Z) -4-ethylidene-O- ( te:rfc-butyldimethylsilyl) - pyrrolidinemethanol (19)
Tributyltinhydride (1.49 g, 5.1 mmol, 1.1 eq) was added to a solution of 18 (1.5 g, 4.62 mmol) in DCM (30 ml) in the presence of water (0.48 g, 27.11 mmol, 6 eq) and a catalytic amount of Pd(PPh3)2Cl2 (0.132 g, 0.189 mmol, 4% molar) and the reaction mixture was allowed to stir for 5 minutes at room temperature.
The reaction mixture was diluted with DCM (40 ml) , dried (MgSO and excess solvent removed under vacuum. The pure amine 19 was isolated by flash chromatography (EtOAc : Petroleum ether, 50:50) to afford a light brown oil.
Yield 0.67 g (60%); IR (cm"1) 4329, 4257, 3426, 2923, 2728, 2672, 2360, 2341, 2035, 1777, 1713, 1641, 1512, 1463, 1377, 1302, 1242, 1169, 1019, 890, 760, 721; 1H-NMR (CDC13) δ 5.33-5.37 (m, 1H) , 4.06 (s, 1H), 3.28-3.75 (m, 4H) , 2.17-2.47 (m, 2H) , 1.54-1.63 (m, 3H) , 0.89 (bs, 9H) , 0.06-.007 (bs, 6H).
Synthesis of (2S) - (E, Z) -N- (4-benzyloxy-5-methoxy-2-nitrobenzoyl) - 2-ethylidene-O- ( e_rt-butyldimethylsilyl) -pyrrolidinemethanol (20)
A catalytic amount of DMF (4 drops) was added to a solution of 4- benzyloxy-5-methoxy-2-nitrobenzoic acid (3) (2.5 g, 8.25 mmol) and oxalyl chloride (1.25 g, 9.84 mmol, 1.2 eq) in dry THF (10 ml) was allowed to stir overnight at room temperature. The resulting chloride was added dropwise over 20 minutes to a solution of the deprotected C-ring 19 (1.98 g, 8.25 mmol, 1 eq) , TEA (1.75 g, 17.35 mmol, 2.1 eq) and water (0.6 ml) at 0°C, under nitrogen. The reaction mixture was allowed to stir for a further 2.5 hours, at which point TLC (EtOAc: Petroleum ether, 40:60) indicated that the reaction was complete. The organic solvent was then removed under reduced pressure and the residue was partitioned between EtOAc (150 ml) and water (150 ml) and the layer separated. The aqueous layer was washed with EtOAc (100 ml) and the combined organic phase was washed with sat. NH4C1 (100 ml), brine (150 ml), dried (MgSO and evaporated in vacuo to afford, after flash chromatography (EtOAc: Petroleum ether, 70:30), the coupled product 20.
Yield 2.85 g (66%); [α]20 D: -11.7° (c 0.305, CHC13) ; IR (cm"1) 4328, 3424, 2960, 2855, 2359, 2332, 2084, 1709, 1641, 1548, 1526, 1462, 1377, 1278, 1215, 1107, 1058, 836, 761, 722; XH-NMR (CDC13) δ 7.37-7.47 (m, 6H) , 6.77 (s, 1H), 5.33-5.37 (m, 1H), 5.22 (s, 2H) , 4.43-4.59 (bs, 1H), 3.95 (s, 3H), 3.59-3.86 (m, 4H), 2.47- 2.75 (m, 2H), 1.60-1.67 (m, 3H) , 0.89 (bs, 9H) , 0.087-0.09 (bs, 6H) ; 13C-NMR (CDCl3) (E/Z isomers) δ 154.8, 135.3, 128.9, 127.6, 109.2, 71.3, 58.0, 57.6, 56.6, 30.3, 29.7, 25.7, 14.6, 14.4, - 5.4, -5.6; MS (Cl) (m/z, relative intensity) 527 (MH+, 25), 418 (82), 388 (6), 359 (9), 328 (7), 304 (25), 279 (45), 258 (13), 240 (35), 219 (11), 184 (10), 161 (51), 147 (21), 133 (41), 113 (21), 107 (100), 91 (25), 81 (8), 73 (40); EI-HRMS m/z 526.2642 (calc'd for C28H38N206Si m/z 526.2499).
Synthesis of (2S) - (E,Z) -N- (4-benzyloxy-5-methoxy-2-nitrobenzoyl) - 2-ethylidene pyrro-lidine methanol (21)
A solution of TBAF (IM solution in THF, 1.75 ml, 1.75 mmol, 1.2 eq) was added dropwise over 15 minutes at 0°C under N2 to a solution of the TBDMS protected intermediate 20 (0.75 g, 1.42 mmol) in dry THF (30 ml) . The reaction mixture was allowed to warm to room temperature and to stir for a further 30 minutes when TLC (EtOAc) indicated complete consumption of the starting material. The reaction mixture was diluted with sat. aq. NH4C1 (150 ml) and extracted with EtOAc (3 x 100 ml) . The organic phase was washed with brine (150 ml), dried (MgSO and evaporated under reduced pressure to afford the deprotected product 21 which was used for the next step without further purification.
Yield 0.64 g (109%, some TBDMSF associated with the product; Yields 0.57 g) ; IR (cm'1) 4214, 3406, 3020, 2958, 2925, 2854, 2400, 2085, 1631, 1581, 1523, 1463, 1453, 1433, 1378, 1335, 1278, 1215, 1053, 870, 754; XH-NMR (CDC13) δ 7.37-7.47 (m, 6H), 6.77 (s, IH), 5.33-5.37 (m, IH) , 5.22 (s, 2H) , 4.43-4.59 (bs, IH), 3.95 (s, 3H), 3.59-3.86 (m, 4H), 2.47-2.75 (m, 2H), 1.60-1.67 (m, 3H), 0.89 (bs, 9H) , 0.087-.009 (bs, 6H).
(2S) - (E, Z) -N- (2-amino-4-benzyloxy-5-methoxybenzoyl) -2- ethylidenepyrrolidine methanol (22)
A solution of the nitro compound 21 (0.64 g, 1.38 mmol, assumes quantitative yield in previous step) in MeOH (30 ml) and SnCl2.2H20 (1.58 g, 7 mmol, 5 eq) was heated to reflux for 40 minutes when TLC (5% MeOH/CHCl3) indicated the reaction was complete. The excess solvent was evaporated in vacuo and the residual oil was partitioned between EtOAc (100 ml) and sat. aq. NaHC03 (100 ml) and allowed to stir overnight under N2. The organic layer was then separated and washed with brine (150 ml), dried (MgSO and evaporated in vacuo . The residue was purified by flash chromatography (5% MeOH/CHCl3) to give the pure amine intermediate 22 as a bright yellow oil.
Yield 0.32 g (61%); IR (cm"1) 4329, 4257, 3427, 2923, 2727, 2360, 2341, 2036, 1712, 1624, 1513, 1463, 1377, 1264, 1215, 1172, 1120, 1002, 872, 761, 722; XH-NMR (CDC13) δ 7.35-7.51 (m, 5H) , 6.87 (s, IH) , 6.27 (s, IH), 5.28-5.42 (m, IH) , 5.17 (s, 2H) , 4.43-4.59 (bs, IH), 3.84 (s, 3H), 3.59-3.78 (m, 4H) , 2.25-2.75 (m, 2H) , 1.60 (d, J = 6.8Hz, 3H); 13C-NMR (CDCl3) (E/Z isomers) δ 150.9, 141.5, 136.6, 134.5, 128.8, 127.9, 127.1, 117.7, 117.3, 112.6, 103.0, 70.6, 66.1, 60.4, 59.8, 57.2, 34.3, 30.0, 29.4, 21.0, 14.6, 14.3, 13.6.
Synthesis of (2S) - (E,Z) -N- [2- (p-nitrobenzyloxy) carboxamido-4- benzyloxy-5-methoxy benzoyl] -2-ethylidenepyrrolidine methanol (23)
The freshly made amine 22 (0.5 g, 1.31 mmol) and dry pyridine (0.207 g, 2.62 mmol, 2eq) was dissolved in dry DCM (30 ml) at 0°C under N2. A solution of 4-nitrobenzyl chloroformate (0.282 g, 1.31 mmol, 1 eq) was added to the amine solution dropwise over 20 O 00/12507
51 min. The reaction mixture was allowed to stir under the same conditions for 2 hours at which time TLC (5% MeOH/CHCl3) revealed complete consumption of the starting material. The mixture was then dissolved in DCM (100 ml) and washed with sat. aq. CuS04 (2 x 75 ml), water (2 x 100 ml), brine (150 ml) and dried (MgS04) . The organic solvent was evaporated in vacuo and after purification by flash chromatography (3% MeOH/CHCl3) the p-nitrobenzyl carbamate was isolated.
Yield 0.56 g (76.3%); [α]20 D : +204.6° (c 0.22, CHC13) ; IR (cm"1) 4329, 4257, 3426, 2924, 2853, 2093, 1710, 1628, 1524, 1462, 1406, 1377, 13460, 1269, 1219, 1175, 1118, 722; 1H-NMR (CDC13, rotamers) δ 8.81 (bs, IH), 8.19 (d, J = 8.6 Hz, 2H) , 7.80 (s, IH) , 7.31- 7.61 (m, 7H) , 6.85 (s, IH) , 5.29-5.35 (m, IH) , 5.24 (s, 2H) , 5.15 (s, 2H), 4.08-4.13 (bs, IH) , 3.83 (s, 3H), 3.66-3.86 (m, 4H), 2.39-2.75 (m, 2H) , 1.51 (d, J = 6.6 Hz, 3H) ; 13C-NMR (CDC13) (E/Z isomers) δ 153.2, 150.3, 147.6, 143.6, 136.1, 134.1, 128.7, 128.4, 128.1, 127.7, 126.9, 123.7, 117.8, 111.2, 70.7, 65.2, 63.8, 56.6, 29.7, 14.6, 14.4; MS (FAB) (m/z, relative intensity) 562 (MH+, 15), 279 (77), 256 (15), 240 (35), 231 (37), 136 (13), 128 (25), 106 (14), 91 (100), 73 (11), 57 (11); EI-HRMS m/z 561.2150 (calc'd for C30H31N3O8 m/z 561.2111).
Synthesis of (llaS) -8-benzyloxy-7-methoxy-l, 3, 10, 11 , 11a- tetrahydro-2-ethylidene-ll-hydroxy-10- (p-nitrobenzyloxy) carboxy- 5H-pyrrolo [2 , 1-c] [1, 4] -benzodiazepine-5-one (24) A solution of the uncyclised carbamate 23 (0.3 g, 0.535 mmol), NMO (94 mg, 0.803 mmol, 1.5 eq) and mol. sieve (0.267 g) in a mixture of dry (DCM :CH3CN, 9 : 3 ml) was allowed to stir at room temperature under N2 for 15 min. TPAP (9.4 mg, 0.026 mmol, 5% molar) was then added and the mixture was allowed to stir under the same conditions for a further 2.5 hours. The reaction mixture was filtered through Celite and the filtrate evaporated in vacuo . The residue was subjected to flash chromatography (1% MeOH/CHCl3) to obtain the target compound (24). O 00/12507
52 Example 5. Synthesis of PBD Dimer Prodrugs
Example 5(a) - Synthesis of 1, 1' - \ [ (Propane-1 , 3- diyl) dioxylbis I \2- (4-nitrobenzyloxycarboxamido) Ibis T (llaS) -7- methoxy-1 ,2,3, lla-tetrahydro-ll-hvdroxy-5H-pyrrolo I"! , 2- c] ri , 4 Ibenzodiazepine -5-one (30)
Figure imgf000054_0001
30
Synthesis of 1', 3' -Bis ( -carboxy-2-methoxyphenoxy) propane (25)
A solution of diiodopropane (4.2 g, 14.2 mmol) was dissolved in THF (30 ml) was added dropwise to a vigorously stirred solution of vanillic acid (1) ( 4-hydroxy-3-methoxybenzoic acid) (5 g, 29. mmol, 2.1 eq) and aq. 0.5 M NaOH (70 ml) in THF (50 ml). The reaction mixture was allowed to reflux for 48 hours and the organic solvent was removed under reduced pressure. The remaining aqueous phase was washed with Petroleum ether 40-60 (3x100 ml) and it was then acidified with cone. HCl to pH 2 until no further precipitation was observed. The precipitate was collected filtration, washed with water, dried to afford the dimer acid (25) as white crystals.
Yield 7.63 g (68%); ^-NMR (CDCl3+DMSO-d6) δ 7.64 (dd, J, = 8.3 Hz, J2 = 8.3 Hz, 2H) , 7.54 (s, 2H) , 6.96 (d, J = 8.4 Hz, 2H) , 4.29 (t, J = 6.1 Hz, 4H) 3.88 (s, 6H), 2.38 (t, J = 6.2 Hz, 2H); 13C-NMR (CDCl3+DMSO-d5) δ 173.1, 168.1, 151.9, 148.6, 123.5, 112.4, 111.45, 65.21, 55.8, 28.8; IR (cm"1) = 3600-3100, 2925, 2855, 1713, 1680, 1597, 1516, 1459, 1377, 1344, 1309, 1275, 1223, 1178, 1133, 1045, 1021; MS (El) (m/z, relative intensity) 376 (M+, 34), 208 (31), 168 (43), 152 (15), 101 (100), 69 (87); EI- HRMS m/z 376.1136 (calc'd for C19H20O8 m/z 376.1158)
Synthesis of 1', 3' -Bis (4-carboxy-2-rnethoxy-5- nitrophenoxy) propane (26)
The dimer acid 25 (5 g, 13.3 mmol) was added slowly in small portions over 30 minutes to a stirred solution of 70% HN03 (50 ml) at 0°C. After the addition was complete the reaction mixture was allowed to warm to 15 °C and stirring continued for a further 2 hours. The reaction mixture was poured onto ice causing precipitation of the product. The yellow precipitate was collected by filtration, washed with cold water and dried to afford the nitrated dimer acid core (26) as a pale yellow solid. Yield 4.52 g (73%); mp. 241-246°C; IR (cm"1) 3500-3200, 2924, 1713, 1604, 1582, 1523, 1459, 1377, 1282, 1218, 1189, 1053; XE-
NMR (CDCl3+DMSO-de) δ 7.44 (s, 2H) , 7.16 (s, 2H) , 4.32 (t, J = 6.5 Hz, 4H), 3.94 (s, 6H) , 2.44 (t, J = 6.4Hz, 2H) ; 13C-NMR (CDCl3+DMSO-d6) δ 172.3, 167.11, 152.5, 149.2, 141.1, 132.1, 128.6, 122.7, 111.2, 108.3, 65.2, 56.4, 33.9; MS (El) (m/z, relative intensity) 467 (MH+, 1), 436 (5), 423 (10), 376 (2), 256 (6), 210 (4), 183 (10), 164 (10), 153 (3), 91 (9), 77 (4), 51 (13), 44 (100); EI-HRMS m/z 466.0876 (calc'd for C19H18N2012 466.0858) . O 00/12507
54
Synthesis of 1, 1' - [ [ (Propane-1 , 3-diyl) dioxy]bis [ (2-nitro-5- methoxy-1 , 4-phenylene) carbon yl]bis [pyrrolidinemethanol] (27)
A solution of the nitrated dimer core 26 (1 g, 2.14 mmol) in a mixture of dry CH3CN/THF (30:5 ml) was treated overnight with oxalyl chloride (0.64 g, 5.14 mmol, 2.4 eq) and DMF (5 drops). The resulting acid chloride was then added dropwise over 30 minutes to a suspension of pyrrolidinemethanol (0.432 g, 4.28 mmol, 2 eq) and K2C03 (1.2 g, 8.56 mmol, 4 eq) in dry acetonitrile (80 ml) at -25°C, under nitrogen. The resulting mixture was allowed to stir at -25°C for a further 1.5 hour when the reaction mixture was diluted with water (100 ml) . The solution was then extracted with chloroform (4 x 100 ml) . The combined organic phase was washed with aq. IM HCl (2 x 75 ml) , water (2 x 75 ml) , brine (100 ml), dried (MgS04) and evaporated in vacuo . The resulting orange oil was purified by column chromatography (10% MeOH/CHCl3) to afford the pure product 27 as a yellow viscous oil.
Yield 0.88 g (65%); [α] 20 D +246.1° (c 0.067, CHCl3) ; IR (cm"1) 4329, 4257, 3370, 2916, 2728, 1787, 1713, 1614, 1574, 1513, 1462, 1377, 1337, 1274, 1215, 1058, 868, 823, 749, 723; XH-NMR (CDC13) δ 7.73 (s, 2H), 6.80 (s, 2H) , 4.31-4.36 (m, 6H) , 3.94 (s, 6H) , 3.74-3.87 (m, 4H) , 3.17 (t, J = 6 Hz, 4H) , 2.44 (t, J = 5.9 Hz), 2.12-2.22 (m, 4H) , 1.70-1.92 (m, 4H) ; 13C-NMR (CDCl3) δ 154.8, 148.3, 137.0, 128.0, 109.1, 108.4, 66.0, 65.6, 61.4, 56.7, 49.5, 28.6, 24.4; MS (FAB) (m/z, relative intensity) 633 (M++l, 52),
449 (12), 236 (11), 219 (17), 206 (12), 196 (15), 191 (22), 178 (20), 166 (20), 151 (34), 135 (23), 128 (11), 122 (33), 115 (11), 102 (47) , 91 (100) , 84 (45) .
Synthesis of 1, 1' -[[ (Propane-1, 3-diyl) dioxy]bis [ (2-amino-5- me hoxy-1 , 4-phenylene) carbonyl] is [pyrrolidinemethanol] (28)
Hydrazine hydrate (0.87 g, 1.74 mmol, 10 eq) was added dropwise to a solution of the nitro intermediate (27) (1 g, 1.58 mmol) over Raney Nickel (0.25 g) in gently refluxing methanol (20 ml). (Anti-bumping granules were used to ensure even boiling) . The reaction mixture was heated at reflux for a further 15 minutes at O 00/12507
55 which time TLC (10% MeOH/CHCl3) indicated that the reaction had gone to completion. The catalyst was then removed by filtration (causation! pyrophoric Ni) and the filtrate was evaporated in vacuo to give a dark brown oil which was purified by flash chromatography (7.5% MeOH/CHCl3) to afford the coupled dimer amine (28) as a bright yellow oil.
Yield = 0.632 g (70 %); IR (cm"1) 3100-3600, 2925, 1611, 1460, 1377, 1275, 1216, 1175; XH-NMR (CDCl3) δ 6.74 (s, 2H) , 6.33 (s, 2H), 4.31-4.42 (m, 2H) , 4.19-4.23 (m, 4H) , 3.76 (s, 6H) , 3.50- 3.66 (m, 8H), 2.27-2.40 (m, 2H) , 2.02-2.15 (m, 4H) , 1.71-1.87 (m, 4H); 13C-NMR (CDC13) δ 151.1, 141.0, 112.8, 102.3, 65.1, 60.8, 57.1, 50.9, 28.7, 28.4; MS (FAB) (m/z, relative intensity) 574 (M++2, 44), 503 (3), 473 (30), 389 (11), 371 (31), 219 (14), 206 (50), 198 (13), 192 (27), 180 (30), 166 (18), 149 (28), 137 (23), 128 (17), 102 (43), 93 (68), 84 (57), 70 (33), 57 (35).
Synthesis of 1, 1' -[[ (Propane-1, 3-diyl) dioxy]bis [ [2- (4- nitrobenzyloxycarboxamido) -5-methoxy-l, -phenylene] carbonyl]bis- pyrrolidinemethanol (29)
A solution of 4-nitrobenzyl chloroformate (0.347 g, 1.61 mmol, 2.3 eq) in dry DCM (10 ml) was added dropwise over 20 min, under nitrogen to a freshly prepared solution of dimer amine 28(0.4 g, 0.7 mmol) in dry DCM (15 ml) and pyridine (0.193 g, 2.45 mmol, 3.5 eq) at 0°C. The resulting reaction mixture was allowed to stir for a further 1.5 hours at 0°C at which time TLC (10% MeOH/CHCl3) indicated complete consumption of starting material. The reaction mixture was diluted with chloroform (50 ml) and washed with a saturated aqueous solution of CuS04 (2 x 50 ml), water (2 x 50 ml), brine (100 ml) and dried (MgS04) . The organic solvent was removed in vacuo and the pure dimer carbamate 29 was obtained after flash chromatography (7% MeOH/CHCl3) as a bright yellow foam.
Yield 0.53 g (81%); IR (cm'1) 3424, 2088, 1641, 1502, 1247; XH-NMR (CDC13) δ 9.02 (bs, 2H), 8.21 (d, J = 8.8 Hz, 4H) , 7.72 (s, 2H) , 7.56 (d, J = 8.8 Hz, 4H) , 6.82 (s, 2H), 5.26 (s, 4H), 4.28-4.38 O 00/12507
56
(m, 2H), 4.26 (t, J = 6Hz, 4H) , 3.79 (s, 6H) , 3.42-3.79 (m, 8H) , 2.27-2.42 (m, 2H) , 2.11-2.18 (m, 4H) , 1.71-1.88 (m, 4H) ; 13C-NMR (CDC13) δ 170.7, 153.2, 150.6, 147.6, 143.6, 128.5, 126.9, 123.9, 111.5, 105.9, 65.3, 63.8, 61.0, 56.6, 51.6, 30.3, 28.4; MS (FAB) (m/z, relative intensity) 932 (M++2, 3), 753 (5), 551 (5), 249 (7), 232 (13), 222 (8), 206 (23), 192 (32), 179 (27), 166 (46), 149 (28), 136 (66), 120 (22), 106 (45), 102 (100), 91 (80), 84 (48), 73 (71), 57 (47).
Synthesis of 1, 1' -[[ (Propane-1, 3-diyl) dioxy]bis [ [2- (4- nitrobenzyloxycarboxamido) ]bis [ (llaS) -7-methoxy-1 , 2 , 3, 11a- tetrahydro-ll-hydroxy-5H-pyrrolo [1,2-c] [1, 4]benzodiazepine -5-one (30)
TPAP (13.4 mg, 0.114 mmol, 0.3 eq) was added in one portion to a solution of the bis carbamate 29 (0.35 g, 0.37 mmol) and NMO (0.134 g, 1.14 mmol, 3.1 eq) in dry DCM/CH3CN (9:3 ml) which has been allowed to stir over molecular sieve (0.350 g) for 15 minutes under N2 and at room temperature. Progress of the reaction was followed by TLC (7% MeOH/CHCl3) . After 2 hours reaction was still incomplete requiring the addition of a further amount of NMO (67 mg, 0.55 mmol, 1.5 eq) and TPAP (6.7 mg, 0.05 mmol, 0.15 eq) . After stirring for a further 30 minutes TLC revealed the complete consumption of starting material. The reaction mixture was filtered through Celite and the filtrate evaporated in vacuo . The resulting black residue was subjected to column chromatography (1.5% MeOH/CHCl3) to afford the product as an opaque pale yellow oil.
Yield 0.143 (42%); IR (cm"1) 3500-3000, 2933, 2253, 1728, 1599, 1523, 1465, 1431, 1409, 1348, 1270, 1206, 1174, 1111, 1060; E- NMR (CDC13, rotamers) δ 8.18 (d, J = 8.8 Hz, 4H) , 7.74 (s, 2H) , 7.48 (d, J = 8.8 Hz, 4H) , 6.71 (s, 2H) , 5.65 (d, J=10 Hz, 2H) ,
5.26 (s, 4H), 4.29 (t, J = 6Hz, 4H) , 4.07-4.16 (m, 2H) , 3.83 (s, 6H), 3.42-3.75 (m, 8H) , 2.25-2.32 (m, 2H) , 2.10-2.22 (m, 4H) , 1.68-1.75 (m, 4H) ; 13C-NMR (CDC13) δ 166.9, 153.2, 147.6, 143.8, 142.8, 128.2, 123.9, 113.9, 110.9, 108.3, 86.2, 69.1, 66.3, 65.6, 60.0, 56.4, 46.4, 29.7, 28.7, 23.1, 14.8; MS (FAB) (m/z, relative O 00/12507
57 intensity) 925 (M+-l, 1), 889 (5), 711 (6), 501 (3), 286 (10), 252 (7), 213 (15), 192 (32), 197 (11), 185 (22), 181 (42), 165 (15), 149 (47), 131 (18), 119 (16), 105 (29), 91 (96), 73 (100), 57 (54) .
Example 5(b) - Synthesis of 1, 1' - f \ (Propane-1 , 3- diyl) dioxy]bis [ [2- (2 , 3-dimethoxy-5-nitrobenzyloxy carboxamido) Ibis \ (llaS) -7-methoxy-l ,2,3, lla-tetrahydro-11- hydroxy-5H-pyrrolo r ,2-c] [1 , 4 ]benzodiazepine -5-one (32)
Figure imgf000059_0001
Figure imgf000059_0002
32
Synthesis of 1, 1' -[[ (Propane-1, 3-diyl) dioxy]bis [ [2- (2 ,3- dimethoxy-5-nitrobenzyloxy carboxamido) -5-methoxy-l, 4- phenylene] carbonyl]bis-pyrrolidinemethanol (31)
A solution of 2-nitro-4 , 5-dimethoxybenzyl chloroformate (0.443 g, 1.61 mmol, 2.3 eq) in dry DCM (10 ml) was added dropwise over 20 minutes to a stirred solution of the amino alcohol 28 (0.4 g, 0.7 mmol) and pyridine (0.193 g, 2.54 mmol, 3.5 eq) in dry DCM (15 ml) at 0°C, under N2. TLC (10% MeOH/CHCl3) revealed the reaction had gone to completion after 2 hours . The reaction mixture was then diluted with chloroform (50 ml) and washed with saturated aqueous solution of CuS04 (2 x 50 ml), water (2 x 50 ml), brine (100 ml) and dried (MgS04) . Removal of the solvent under reduced pressure gave a dark yellow oil which was further purified by flash chromatography (7% MeOH/CHCl3) to afford the pure product as a yellow foam.
Yield 0.564 (77%); [α]20 D-43.3° (c 0.485, CHC13) ; IR (cm"1) 4214, 3416, 3020, 2973, 2940, 2613, 2400, 2254, 2075, 1727, 1618, 1585, 1522, 1465, 1438, 1408, 1331, 1278, 1216, 1174, 1118, 1071, 1030, 987, 909, 874, 850, 754; 1H-NMR (CDC13, rotamers) δ 8.93 (bs, 2H), 7.71 (s, 2H), 7.69 (s, 2H) , 7.10 (s, 2H) , 6.82 (s, 2H) , 5.60 (m, 4H), 4.34 (m, 2H) , 4.26 (t, J = 6Hz, 4H) , 3.99 (s, 12H) , 3.94 (s, 6H), 3.51-3.79 (m, 8H) , 2.26-2.36 (m, 2H) , 2.05-2.17 (m, 4H) , 1.80-1.88 (m, 4H) ; 13C-NMR (CDC13) δ 170.6, 153.7, 150.45, 148.1, 139.5, 132.9, 127.8, 111.2, 110.4, 109.9, 108.1, 65.4, 63.8, 62.4, 60.8, 56.4, 53.5, 51.4, 50.6, 28.2, 25.0, 21.0, 14.2; MS (FAB) (m/z, relative intensity) 1053 (M++2, 7), 814 (6), 416 (3), 306 (13), 292 (5), 280 (27), 246 (46), 197 (81), 186 (38), 180 (25), 166 (37), 151 (19), 102 (23), 93 (100), 84 (10), 75 (37), 57 (42) .
Synthesis of 1, 1' -[[ (Propane-1, 3-diyl) dioxy]bis [ [2- (2,3- dimethoxy-5-nitrobenzyloxy carboxamido) ]bis [ (llaS) -7-methoxy- 1,2,3, lla-tetrahydro-ll-hydroxy-5H-pyrrolo [1,2- c] [l,4]benzodiazepine -5-one (32)
NMO (0.138 g, 1.16 mmol, 3.1 eq) and molecular sieve (350 mg) was added to a stirred solution of the NVOC dimer carbamate 31(0.4 g, 0.38 mmol) in dry DCM and acetonitrile (9:3 ml) under nitrogen. This mixture was allowed to stir for 15 minutes before the addition of TPAP (13.8 mg, 0.116 mmol, 0.3 eq) . Further stirring of the reaction mixture for 2 hours at room temperature was followed by the addition of an additional amount of TPAP (6.9 mg, 0.058 mmol, 0.15 eq) and NMO (69 mg, 1.58 mmol, 1.5 eq) to drive the reaction to completion [TLC (5% MeOH/CHCl3) ] after a further 45 minute period of vigorous stirring. The organic solvent was evaporated in vacuo to afford a black residue which was further purified by column chromatography (1% MeOH/CHCl3) to afford the pure final product as a dark yellow oil. O 00/12507
59
Yield 0.162 (39 %); [α] 20 D -121.5° (c 1.07, CHC13);IR (cm"1) 4329, 4258, 3426, 2926, 2854, 2728, 2360, 2341, 2046, 1712, 1620, 1583, 1523, 1464, 1408, 1330, 1278, 1219, 1172, 1149, 1071, 1030, 987, 871, 796, 759; 1H- R (CDC13, rotamers) δ 7.71 (s, 2H) , 7.62 (s, 2H), 7.10 (s, 2H), 6.43 (s, 2H) , 5.48-5.59 (m, 6H), 4.29-4.36 (m, 2H), 4.24-4.28 (t, J = 6Hz, 4H) , 3.95 (s, 12H) , 3.88 (s, 6H), 3.47-3.53 (m, 8H) , 2.32-2.43 (m, 2H) , 2.07-2.17 (m, 4H) , 1.67- 1.92 (m, 4H); 1C-NMR (CDCl3) δ 170.4, 153.8, 150.2, 148.9, 139.6, 130.9, 128.8, 127.6, 111.5, 110.6, 108.1, 86.0, 65.4, 65.0, 63.8, 60.9, 56.4, 46.4, 31.9, 28.2, 25.9, 22.7, 14.1.
O 00/12507
60
Example 5(c) - Synthesis of 1, 1' - [ \ (Propane-1 , 3- diyl) dioxylbis [ \2- (phenylacetamide) Ibis [ (llaS) -7-methoxy-
1,2,3, lla-tetrahydro-ll-hydroxy-5H-pyrrolo \X .2- c] ri , 1benzodiazepine-5-one (34)
Figure imgf000062_0001
33
Figure imgf000062_0002
34
Synthesis of 1, 1' -[[ (Propane-1 , 3-diyl) dioxy]bis [ [2-
(phenylacetamide) -5-methoxy-l , 4-phenylene] carbonyl]bis- pyrrolidinemethanol [33]
A catalytic quantity of DMF (4 drops) was added to a stirred solution of phenyl acetic acid (0.2 g, 1.47 mmol, 2.1 eq) and oxalyl chloride (0.224 g, 1.76 g, 2.5 eq) in dry acetonitrile
(10 ml) and the reaction mixture was allowed to stir overnight at room temperature under N2. The solution of the resulting acid chloride was added dropwise over 30 minutes to a stirred solution of the amine 28 (0.4 g, 0.7 mmol) in dry acetonitrile (40 ml) over K2C03 (0.406 g, 2.94 mmol, 4.2 eq) at -25°C, under N2 and the reaction mixture was allowed to stir for a further 1.5 hours. The reaction mixture was diluted in chloroform (50 ml) and washed O 00/12507
61 with IM HCl (2 x 50 ml), water (2 x 75 ml), brine (100 ml), dried (MgS04) . Excess solvent was evaporated in vacuo to give a dark yellow oil which, after flash chromatography (10% MeOH/CHCl3) , afforded the pure phenylacetamide protected dimer amino alcohol as a pale yellow oil.
Yield 0.344 (64%); XH-NMR (CDC13, rotamers) δ 9.31 (bs, 2H) , 7.59 (s, 2H), 7.35 (s, 10H), 6.68 (s, 2H) , 4.20-4.28 (m, 6H), 4.14- 4.18 (m, 4H), 3.71 (s, 6H) , 3.24-3.56 (m, 8H) , 2.27-2.39 (m, 2H) , 2.02-2.17 (m, 4H) , 1.64-1.81 (m, 4H) ; 13C-NMR (CDC13) δ 169.7, 150.1, 134.7, 130.2, 129.4, 128.9, 127.3, 110.5, 108.2, 65.3, 61.1, 56.3, 50.8, 44.7, 29.7, 28.3, 24.7.
Synthesis of 1, 1' -[[ (Propane-1 , 3-diyl) dioxy]bis [ [2- (phenylacetamide) ]bis [ (llaS) -7-methoxy-l , 2 , 3, lla-tetrahydro-11- hydroxy-5H-pyrrolo [1,2-c] [1 , 4]benzodiazepine-5-one (34) NMO (0.141 g, 1.2 mmol, 3.1 eq) and mol. sieve (350 mg) was added to a stirred solution of the phenylacetamide protected dimer carbamate 33(0.3 g, 0.38 mmol) in dry DCM and acetonitrile (9:3 ml) under nitrogen. This mixture was allowed to stir for 20 minutes before the addition of TPAP (14.1 mg, 0.12 mmol, 0.31 eq) . Further stirring of the reaction mixture for 1.5 hours at room temperature was followed by the addition of an additional amount of TPAP (7 mg, 0.06 mmol, 0.15 eq) and NMO (70 mg, 0.6 mmol, 1.5 eq) to drive the reaction to completion [TLC (5% MeOH/CHCl3) ] after a further 45 minute period of vigorous stirring. The organic solvent was evaporated in vacuo to afford a black residue which was further purified by column chromatography (1% MeOH/CHCl3) to afford the pure final product as a dark yellow oil.
Yield 0.138 g (47.5%); [α]20 D+132° (c 0.265, CHC13) ; IR (cm"1) 3412, 2959, 2924, 2854, 2094, 1642, 1462, 1377; 1H-NMR (CDCl3, rotamers) δ 7.28-7.39 (s, 10H) , 7.15 (s, 2H) , 6.46 (s, 2H) , 5.75- 5.78 (d, J = 10 Hz, 2H), 4.10-4.35 (m, 10H) , 3.89 (s, 6H) , 3.35- 3.73 (m, 8H), 2.27-2.32 (m, 2H) , 1.68-2.05 (m, 8H) ; 13C-NMR (CDC13) δ 169.9, 153.2, 147.6, 143.8, 142.8, 128.2, 123.9, 113.9, O 00/12507
62
110.9, 108.3, 86.5, 69.1, 66.3, 65.6, 60.0, 56.4, 46.4, 29.7, 28.7, 23.1, 14.8.
Example 5(d) - Alternative Synthesis of 1, 1' -[ f (Propane-1 , 3- diyl) dioxy]bis f f2- (4-nitrobenzyloxycarboxamido) ]bis T (llaS) -7- me hoxy-1 ,2,3, lla-tetrahvdro-ll-hydroxy-5ff-pyrrolo [1 ,2- c] [1 , 4 Ibenzodiazepine -5-one (30)
Figure imgf000064_0001
Synthesis of 1 , 1' -[[ (propane-1 , 3-diyl) dioxy]bis [ (2-nitro-5- methoxyl , 4-phenylene) carbonyl] ]bis [pyrrolidine-2- carboxaldehydediethyldi hioacetal] (35) 3-4 drops of DMF was added to a stirred suspension of the dimer nitro acid 26 (0.25 g, 1.01 mmol) and oxalyl chloride (0.3 g, 2.33 mmol, 2.3 eq) in dry THF volume and the reaction mixture was allowed to stir overnight under nitrogen. The resulting acid chloride was added dropwise, over 20 min, to a stirred solution of (2S) -pyrrolidine-2-carbaldehyde diethyl thioacetal (0.416 g, 2.02 mmol, 2 eq) and triethylamine (0.41 g, 4.04 mmol, 4 eq) at 0°C, under N2. The reaction mixture was allowed to warm to room temperature and stirring was continued for a further 1.5 hours. Excess THF was then removed and the residue was diluted with water (5 ml) and extracted with EtOAc (15 ml) . The pH of the O 00/12507
63 aqueous phase was adjusted to pH 3 with two drops of cone. HCl and subsequently extracted with EtOAc (3x10 ml) . The combined organic phase was washed with water (2 x 10 ml), brine (15 ml), dried (MgS04) and the organic solvent was evaporated in vacuo to afford a dark red oil. Purification by flash chromatography
(EtOAc: Petroleum ether 40-60, 1:1) afforded the pure product 35.
Yield 0.558 g (66%); ÷H-NMR (CDCl3) δ 7.72 (s, 2H) , 6.83 (s, 2H) , 4.89 (d, J = 4.0 Hz, 2H) , 4.64-4.70 (m, 2H) , 4.30-4.40 (m, 4H) , 3.91 (s, 6H), 3.15-3.30 (m, 4H) , 2.59-2.76 (m, 8H) , 1.70-2.38 (m, 8H) , 1.30-1.38 (m, 12H) .
Synthesis of 1 , 1' -[[ (propane-1, 3-diyl) dioxy]bis [ (2-amino-5- methoxyl , 4-phenylene) carbonyl] ]bis [pyrrolidine-2- carboxaldehydediethyldi hioacetal ] (36)
A solution of the nitro thioacetal 35(0.4 g, 0.47 mmol) and SnCl2-2H20 (1.4 g, 6.22 mmol, 13.2 eq) in methanol (7 ml) was heated at reflux for 40 minutes at which time TLC (EtOAc: Petroleum ether 40-60, 1:1) indicated the reaction was complete. After the reaction mixture was allowed to return to room temperature the pH was adjusted to pH 8 by the addition of sat. aq. NaHC03. The resulting suspension was diluted with EtOAc (100 ml) and it was allowed to stir overnight under nitrogen. The organic phase was separated and washed with brine (50 ml) , dried (MgS04) and the solvent was removed under reduced pressure to give a dark yellow gum which was further purified by flash chromatography (EtOAc: Petroleum ether 40-60, 3:7) to afford the pure dimer amino thioacetal as a bright yellow oil.
Yield 0.281 (56%); XH-NMR (CDC13) δ 6.82 (s, 2H) , 6.29 (s, 2H) , 5.30 (d, J = 4.0 Hz, 2H), 4.69-4.87 (m, 2H) , 4.08-4.21 (m, 4H) , 3.76 (s, 6H), 3.60-3.66 (m, 4H) , 2.65-2.73 (m, 8H) , 1.80-2.35 (m, 8H) , 1.22-1.31 (m, 12H) . 00/12507
64
Synthesis of 1 , 1' -[[ (propane-1, 3-diyl) dioxy]bis [ (2- (4- nitrobenzyloxycarbonylamine-5-methoxyl , 4-phenylene) carbonyl] ]bis [pyrrolidine-2-carboxaldehydediethyldithioacetal ]
(37) A solution of 4-nitrobenzyl chloroformate (0.100 g, 0.46 mmol, 2 eq) in dry DCM (10 ml) was added dropwise to a solution of the bis amine 36(0.18 g, 0.23 mmol) and pyridine (0.101 g, 1.28 mmol, 5.5 eq) in dry DCM (15 ml) over 20 minutes at 0°C, under nitrogen. The reaction solution was allowed to stir at 0°C for a further 1.5 hours (TLC: 7% MeOH/CHCl3) , after which time it was allowed to warm to room temperature and diluted with chloroform (50 ml) . The organic phase was washed with sat. aq. CuS04 (2 x 50 ml), water (75 ml), brine (75 ml), dried (MgS04) and the excess solvent evaporated in vacuo to give a dark yellow oil. After purification by flash chromatography (7% MeOH/CHCl3) the pure product was obtained as a dark yellow oil.
Yield 0.179 (68%); [α]2°D+100° (c 0.232 CHCl3) ; IR (cm"1) 4214, 3426, 3020, 2400, 2085, 1658, 1609, 1525, 1466, 1452, 1408, 1348, 1268, 1216, 1174, 1112, 1052, 1015, 908, 753; *H-NMR (CDC13, rotomers) δ 9.18 (bs, 2H) , 8.19-8.24 (d, J = 8.8 Hz, 4H) , 6.82 (s, 2H), 6.29 (s, 2H), 5.30 (d, J = 4.0 Hz, 2H, ) , 4.69-4.87 (m, 2H) , 4.08-4.21 (m, 4H) , 3.76 (s, 6H) , 3.60-3.66 (m, 4H) , 2.65- 2.73 (m, 8H), 1.80-2.35 (m, 8H) , 1.22-1.31 (m, 12H) .
Synthesis of 1, 1' -[[ (Propane-1, 3-diyl) dioxy]bis [ [2- (4- nitrobenzyloxycarboxamido) ]bis [ (llaS) -7-methoxy-1,2 , 3, 11a- tetrahydro-ll-hydroxy-5ff-pyrrolo [1,2-c] [1 , 4 ]benzodiazepine -5-one (30)
Mercury chloride (1.6 g, 5.88 mmol, 4.5 eq) was added to a slowly stirred solution of the N-protected amino thioacetal 37(0.15 g, 0.13 mmol) and CaC03 (65 mg, 0.65 mmol, 5 eq) in acetonitrile/water (4:1, 5 ml) and stirring continued at room temperature for 24 hours. The reaction mixture was diluted with EtOAc (20 ml) and filtered through Celite. The filtrate was washed with sat. aq. NaHC03 (2 x 15 ml), brine (20 ml), dried (MgS04) and evaporated under reduced pressure to afford a yellow O 00/12507
65 oil which was further purified by column chromatography (0 to 2% MeOH/CHCl3) to afford the dimer prodrug compound 30 (Yield = 67.3 mg (56%) ) .
Example 5(e) - Alternative synthesis of intermediate (26) in Examples 5(a) to 5(d)
Figure imgf000067_0001
Figure imgf000067_0002
Synthesis of 1', 3' -Bis (4-carbonyl-2-methoxyphenoxy) propane (39)
A solution of diethyl azodicarboxylate (4.57 g, 26 3 mmol, 1 eq) in dry THF (15 ml) was added dropwise over 15 minutes to a solution of vanillin (38) (10.4 g, 68.3 mmol, 2.6 eq) , 1,3- propanediol (2 g, 26.3 mmol, 1 eq) and triphenylphosphine (6.87 g, 26.3 mmol, 1 eq) in dry THF (50 ml) under nitrogen. The reaction mixture was allowed to stir overnight and diluted with chloroform (70 ml), washed with IM NaOH (2 x 75 ml). The organic solvent was removed by rotary evaporation and the residue was triturated with toluene (150 ml) for 24 hours.
Triphenylphosphine oxide was removed by filtration and the filtrate was washed with NaOH (100 ml), dried (MgS04) , evaporated in vacuo and the remaining opaque oil was purified by flash chromatography (100% CHC13) to afford a white solid.
Yield = 6 . 27 ( 70% ) ; XH-NMR ( CDCl3+DMSO-ds) δ 9 . 84 ( s , 2H ) , 7 . 62- 00/12507
66
7.66 (d, J = 8.3Hz, 2H), 7.38 (s, 2H) , 7.02-7.05 (d, J = 8.4 Hz, 2H), 4.24-4.28 (t, J = 6.1 Hz, 4H) , 3.91 (s, 6H) , 2.05-2.12 (t, J = 6.2 Hz, 2H); "C-NMR (CDCl3+DMS0-d5) δ 190.8, 154.0, 149.6, 131.9, 129.7, 128.5, 126.7, 11.5, 109.1, 66.6, 61.4, 58.8, 55.9, 31.8, 14.5; MS (FAB) (m/z, relative intensity) 344 (M+, 2), 210
(54), 152 (100), 123 (9) , 109 (14) , 81 (11) , 65 (10) , 51 (11) .
Synthesis of 1', 3' -Bis (4-carbonyl-2-methoxy-5- nitrσphenoxy) propane (40)
The dimer aldehyde (39) (5 g, 14.5 mmol) was added in small portions over a period of 30 minutes to HN03 (100 ml, 70%) at 0°C. The resulting suspension was stirred for a further 30 minutes at 15 °C when it was poured onto ice-water and the bright yellow precipitate was collected by filtration, washed with cold water and dried to afford the nitrated intermediate.
Yield 4.63 g (74%); *H-NMR (CDCl3+DMSO-ds) δ 10.41 (s, 2H) , 7.67 (s, 2H), 7.37 (s, 2H), 4.41-4.45 (t, J = 6.1 Hz, 4H) , 3.99 (s, 6H), 2.87-2.92 (t, J = 6.2 Hz, 2H) ; 13C-NMR (CDCl3+DMS0-d6) δ 187.7, 172.2, 153.4, 151.5, 143.6, 132.0, 128.6, 125.6, 110.3, 109.9, 108.4, 65.8, 56.6, 33.8; MS (El) (m/z, relative intensity) 434 (M+, 1), 277 (33), 269 (37), 214 (13), 197 (21), 167 (100), 152 (21), 122 (20), 111 (15), 96 (10), 79 (13), 72 (32).
Synthesis of 1', 3' -Bis (4-carboxy-2-methoxy-5- nitrophenoxy) propane (26)
Hot aqueous KMn04 (10% w/v, 200 ml) was added dropwise to a solution of the aldehyde (40) (4 g, 9.21 mmol) in acetone (300 ml) over 15 min. The resulting mixture was stirred for 40 minutes and the insoluble material was removed by filtration through celite. The pad was washed with hot water and the combined filtrate concentrated in vacuo. The remaining aqueous phase was acidified with cone. HCl to afford a pale yellow precipitate which was collected by filtration, washed with water and dried to give the dimer acid (26) (Yield 3.08 g (71.8%)). 00/12507
67
Example 6 Synthesis of a Psec-protected PBD (52) and related Ptec-protected PBD (55a/b) (for comparison)
.OH
MeO-. .O e
Cbz— -* Cbz — N J >~ Cbz— j Cbz— N
41 42 43 44
Figure imgf000069_0001
51 52
Synthesis of Secondary Amine 45
Lithium tetrahydroborate (2.6 g, 0.12 mol) was added portionwise to a solution of N-carbobenzyloxy-L-proline methyl ester 41 (21 g, 0.08 mol) in THF (500 ml) at 0°C. The reaction mixture was allowed to stir at room temperature for 48 hours. The solution was then cooled to 0°C and ice water (150 ml) was added to quench excess lithium tetrahydroborate. The resulting suspension was adjusted to pH 4.0 with aqueous HCl (1.0 N) and extracted with Et20 (250 ml) . The organic phase was separated and washed with H20 (3 x 100 ml), brine (2 x 100 ml), dried (MgS04) and concentrated to give alcohol 42 as a pale yellow oil (18.6 g, 99%). XHNMR (270 MHz, CDC13) d 2.1-1.77 (m, 4H); 3.76- 3.35 (m, 4H); 4.1-3.77 (m, IH); 5.14 (2 x s, 2H) ; 7.38-7.28 (m, 5H) . CIMS 236 (M+)
A solution of triethylamine (32 ml, 0.23 mol) and S03.pyridine complex (37 g, 0.23 mol) in DMSO (210 ml) was added to a solution of alcohol 42 (18 g, 0.077 mol) in CH2C12 (250 ml) at -10°C, under a nitrogen atmosphere. The reaction mixture was allowed to warm to room temperature and stirred for 30 minutes and then poured into ice water (200 ml) and extracted with Et20. The organic phase was washed with aqueous HCl (1.0 N, 3 x 150 ml), H20 (3 x 150 ml) , brine (2 x 150 ml) , dried (MgS04) and concentrated to give a yellow oil. The crude material was purified by flash column chromatography (silica gel, EtOAc) to give aldehyde 43 as a colourless oil (12.6 g, 71%). 1H NMR (270 MHz, CDC13) δ 2.16- 1.8 (m, 4H); 3.66-3.5 (m, 2H) ; 4.33-4.17 (m, IH) ; 5.22-5.13 (m, 2H); 7.37-7.3 (m, 5H) ; 9.59 (2 x s, IH) . CIMS 234 (M+ + 1).
Thionyl chloride (5.5 ml) was added to a solution of aldehyde 43 (11 g, 0. 047 mol) and trimethyl orthoformate (36 ml, 0.33 mol) in MeOH (55 ml) at 0°C. The reaction mixture was heated at 60°C for 2 hours. The solution was allowed to cool to room temperature, and treated with excess solid Na2C03 and diluted with Et20 (60 ml). The suspension was filtered to remove insoluble inorganics and resultant filtrate was concentrated in vacuo and then redissolved in EtOAc. The organic solution was washed with saturated aqueous NaHC03 (3 x 50 ml), brine (2 x 50 ml), dried (MgS04) and concentrated to give acetal 44 as a yellow liquid (12.5 g, 95%). XH NMR (270 MHz, CDC13) δ 2.16-1.7 (m, 4H) ; 3.64- 3.33 (m, 8H); 4.02-3.91 (br. m, IH) ; 4.4 and 4.6 (2 x br . s, IH) ; 5.17-5.1 (m, 2H); 7.47-7.28 (m, 5H) .
A solution of acetal 44 (5.8 g, 0.02 mol) in EtOH (50 ml) was allowed to stir for 16 hours at room temperature over Raney 00/12507
69 nickel (0.2 g) , in order to remove the trace amounts of sulphur impurities prior to hydrogenation. Nickel was removed by filtration through Celite.
10% Palladium on carbon (580 mg) was added to the alcoholic solution which was subjected to hydrogenation under pressure (c. 50 psi). After 16 h, the reaction mixture was filtered through Celite and the pad was washed with EtOAc, the combined organic solutions were concentrated to give secondary amine 45 as a pale green liquid (2.9 g, 100%). H NMR (270 MHz, CDC13) δ 1.93-1.59 (m, 4H); 3.1-2.92 (m, 2H) ; 3.4-3.3 (d, J = 6.9 Hz, IH) ; 3.41 (2 x s, 6H); 3.53 (br.s, IH) ; 4.2 (d, J= 6.8 Hz, IH) .
Synthesis of Amine 48
A solution of acetal 45 (1 g, 6.9 mmol), 4 , 5-dimethoxy 2- nitrobenzoic acid 46 (1.6 g, 6.9 mmol), TBTU (2.2 g, 6.9 mmol) and DIPEA (1.2 ml, 6.9 mmol) in DMF (30 ml) were stirred at room temperature for 16 hours. The reaction mixture was concentrated in vacuo and the crude material was extracted with EtOAc and washed with saturated aqueous NaHC03 (3 x 30 ml), HCl (1.0 N, 3 x 30 ml), H20 (3 x 30 ml), brine (3 x 30 ml), dried (MgS04) and concentrated to give a yellow semi solid. The crude material was purified by flash column chromatography (silica gel, 2:1 EtOAc : petroleum ether 40-60) to give the nitro compound 47 as a pale cream solid (1.28 g, 51%). *H NMR (270 MHz, CDC13) δ 2.26-1.67 (m, 4H) ; 3.19-3.06 (m, 2H) ; 3.59 and 3.57 (2 x s, 6H) ; 3.98 (s, 6H); 4.45-4.4 (m, IH) ; 4.95-4.94 (d, J" = 2.6 Hz, IH) ; 6.76 (s, IH); 7.71 (s, IH) ; CIMS 355 (M+ + 1).
10% Palladium on carbon (130 mg) was added to a solution of the nitro compound 47 (1.28 g, 3.6 mmol) in EtOH (50 ml), which was subjected to hydrogenation under pressure (c. 50 psi) . After 20 hours, the reaction mixture was filtered through Celite and the pad was washed with EtOAc, the combined organic solutions were concentrated to give secondary amine 48 as a pale oil (1.26 g, 98%). XH NMR (270 MHz, CDC13) δ 2.18-1.65 (m, 4H) ; 3.57-3.47 (m, 8H); 3.85 and 3.8 (2 x s, 6H) ; 4.42-4.38 (m, IH) ; 4.74-4.7 (m, 00/12507
70
IH); 6.3 (s, IH); 6.77 (s, IH) ; 1C NMR (68.7 MHz, CDC13) δ 24.03, 25.07, 50.56, 55.8, 56.24, 56.86, 57.65, 58.82, 101.07, 105.01, 111.75, 112.52, 141.07, 151.81, 169.76; EIMS 324 (M+) .
Synthesis of Psec-protected PBD 52 Pyridine (55 μl , 0.67 mmol) was added to a solution of 2-
(phenylsulfonyl) ethanol 49 (375 mg, 2.01 mmol) and triphosgene (200 mg, 0.67 mmol) in CH2C12 (10 ml) at 0°C. The reaction mixture was stirred at room temperature for 16 hours. Pyridine (150 μl , 1.85 mmol) and crude chloroformate 50 were added to a solution of amine 48 (0.5 g, 1.5 mmol) in CH2C12 (20 ml) at 0°C. The reaction mixture was allowed to stir at room temperature for 3 hours. Then the solution was concentrated in vacuo and extracted with CH2C12. The organic phase was washed with 10% citric acid (2 x 20 ml), H20 (20 ml), brine (20 ml), dried (MgS04) and concentrated to give a yellow oil. The crude material was purified by flash column chromatography (silica gel, EtOAc) to give carbamate 51, as a colourless oil (770 mg, 93%). XH NMR (270 MHz, CDC13) δ 2.17-1.72 (m, 6H) , 3.55-3.34 (m, 10H) , 3.92 and 3.84 (2 x s, 6H) , 4.5-4.4 (m, 2H) , 4.75 (br.s, IH) , 6.82 (s, IH), 7.97-7.56 (m, 6H) , 8.89 (br.s, IH) ; 13C NMR (68.7 MHz,
CDC13) δ 14.19, 21.04, 23.88, 55.31, 56.12, 56.31, 56.39, 57.52, 58.25, 58.94, 60.38, 103.91, 104.74, 111.15, 127.97, 128.09, 129.43, 133.98, 134.05, 139.29, 143.75, 151.09, 152.72, 168.83; FABMS 536 (M +) .
Trans-bis (acetonitrile) palladium (II) chloride (107 mg, 0.41 mmol) was added to a solution of carbamate 51 in anhydrous acetone (14 ml). The reaction mixture was stirred at room temperature for 16 hours. The solution was concentrated in vacuo to give a brown foam. The crude material was purified by flash column chromatography (silica gel, EtOAc) to give methyl ether 52, as an orange oil (690 mg, 85%) .
XH NMR (270 MHz, CDC13) δ 2.12-1.78 (m, 6H), 3.22-3.17 (m, IH), 3.7-3.4 (m, 7H), 3.98 and 3.94 (2 x s, 6H) , 4.69-4.65 (m, IH) , 5.46-5.43 (d, J = 9.3 Hz, IH) , 7.03 (s, IH), 7.22 (s,lH), 7.87- 7.53 (m, 5H) ; 13C NMR (68.7 MHz, CDC13) δ 14.19, 21.06, 23.21, „Λ n„ 00/12507
71
28.98, 46.33, 54.74, 56.15, 56.39, 56.45, 58.58, 60.09, 60.39, 93.33, 110.18, 113.32, 126.24, 128.07, 128.27, 129.49, 134.13, 138.59, 148.77, 151.15, 155.65, 167.36; EIMS 504 (M +) .
Synthesis of Ptec-PBD 55a/b
Figure imgf000073_0001
Pyridine (65 μl, 0.77 mmol) was added to a solution of 2-
(phenylthio) ethanol 53 (355 mg, 2.3 mmol) and triphosgene (230 mg, 0.77 mmol) in CH2C12 (10 ml) at 0°C. The reaction mixture was stirred at room temperature for 16 hours. Pyridine (150 μl, 1.85 mmol) and crude chloroformate 54 were added to a solution of amine 47 (0.5 g, 1.5 mmol) in CH2C12 (20 ml) at 0°C. The reaction mixture was allowed to stir at room temperature for 3 hours. Then the solution was concentrated in vacuo and extracted with CH2Cl2. The organic phase was washed with HCl (1.0 N, 2 x 20 ml), H20 (20 ml), brine (20 ml), dried (MgS04) and concentrated to give a brown oil. The crude material was purified by flash column chromatography (silica gel, 2:1 EtOAc : petroleum ether 40 - 60) to give a mixture of carbinolamine 55a and methyl ether 55b, as a yellow gum (510 mg, 66%) . 1H NMR (270 MHz, CDC13) δ 2.1-1.99 (m, 4H); 3.15-2.98 (m, 2H) ,
3.71-3.44 ( , 4H) , 3.93 and 3.89 (2 x s, 6H) , 4.38-4.07 (m, IH) , 5.45-5.41 (d, J = 9.3 Hz,lH), 5.65-5.61 (d, J = 9.5 Hz, IH) , 6.76 (s, IH), 7.29-7.19 (m, 6H); 13C NMR (68.7 MHz, CDC13) δ 23.05, 23.21, 28.7, 28.98, 29.36, 29.68, 32.72, 32.99, 46.36, 56.13, 56.19, 56.59, 59.9, 60.13, 63.91, 64.19, 86.07, 93.37, 110.36, 112.55, 112.85, 125.67, 126.69, 128.3, 129.15, 129.72, 129.97, 134.85, 148.36, 150.83, 156.02, 167.05; EIMS 458 (M+, 55a), 472 (M+, 55b) .
Example 7: Nitroreductase-activation of benzyl DC-81 prodrug (Compound 7)
Compound 7, synthesized according to example 1, was evaluated in two different cell lines, namely S 1116 and LS174T. Both SW1116 and LS174T are human adenocarcinoma colonic cell lines, which were grown at Charing Cross hospital. The cells, at a concentration of 2500 cells/ml, were plated in 96-well microtitre plates, and were incubated at 37°C for 1 hour with different concentrations of the prodrug, in the presence or absence of E. coli nitroreductase-monoclonal antibody conjugate in phosphate buffer saline (available from Sigma) , and NADH (the co-factor necessary for enzyme function) in DMSO. The cells were then washed and incubated for a further 3 days at 37 °C. At the end of this period, the cells were fixed (TCA) and stained. The concentration of the remaining viable cells adhering to the plates was quantified by a sulforhodamine B (SRB) dye. The control sets of cells treated with the prodrug alone were used in order to evaluate the cytotoxicity of the compound prior to enzymatic activation.
The results are illustrated in Figure 4. The prodrug alone was found to be essentially non-toxic in SW1116 cells even at concentrations of up to 500μM. A slight toxicity was observed in the LS174T cell line at concentrations higher than lOOμM. In the presence of the enzyme and co-factor, the IC50 of compound 7 was established as 1-5 M in both cell lines.
The parent drug, benzyl DC-81 (Thurston et al, 1990, Chem. Bri t . , 26, 767-772) , was also evaluated in the same cell lines under the same conditions in order to establish the extent of activation 00/12507
73 achieved by the prodrug/enzyme system. A difference was observed between the IC50 value of the parent drug (ICS0 = 0.008 μM) and the nitroreductase-activated prodrug (IC50 = 1-5 μM) of 20-100 fold.
Example 8: Nitroreductase-activation of DSB-120 prodrug (Compound 30)
Compound 30, see examples 5(a) and 5(e), was evaluated in the LS174T cell line, under the same conditions as were used in example 6. The IC50 value was found to be 215.3 μM (average of two measurements) which reduced to 13.7μM after the addition of the enzyme and co-factor, representing a 15-16 fold activation factor. The cytotoxicity (IC50) of the parent agent DSB-120 in this cell line was found to be 0.0005μM, indicating less efficient activation of the dimer prodrug compared to compound 7.
Example 9: Nitroreductase-activation of benzyl tomaymycin prodrug (Compound 24)
Compound 24, see example 4, was examined in the same cell lines as example 6, and under the same conditions. It exhibited an IC50 value of 86.2μM (average of two experiments) which, after addition of the enzyme conjugate and NADH, dropped to 6.4μM, indicating an activation factor of approximately 13.5. In this case, the parent benzyl tomaymycin was unavailable for evaluation as a control. However, assuming that the addition of a benzyl group to the C8-position of the A-ring of tomaymycin does not change the cytotoxicity of the compound significantly (as is the case for DC-81/C8-benzyl DC-81), the cytotoxicity of C8-benzyl tomaymycin should be in the order of 0.01-0. OOlμM.
Example 10: Light-activation of the benzyl DC-81 prodrug (Compound 11) This experiment involved the irradiation of compound 11, see example 3, in DMF at a concentration of 1 mM using a Stratagene UVA Crosslinker (365 nm) . Small aliquots (lOOμL) were removed at 30 minutes, 1 hour, 2 hours and 3 hours and the cleavage of the photolabile NVOC group was monitored by HPLC, using a Waters C4, 300 angstrom reversed phase column and a mobile phase of 50% methanol/50% water. Compound 11 has a retention time of 11.25 minutes and upon radiation produced a new peak with a retention time of 8.58 minutes. Authentic benzyl DC-81 has an identical retention time of 8.58 minutes. The time course of the deprotection process is shown in figure 5. Complete conversion is achieved by 2 hours of irradiation at 365nm under the conditions employed.
An MTT assay was used to evaluate the ability of aliquots of the activated analogues, which were removed at time intervals, to inhibit the growth of chronic human histiocytic leukaemia K562 cells in culture. Following treatment of cells with a range of drug doses, cells were transferred to 96-well microtitre plates, 104 cells per well, 8 wells per sample. Plates were incubated at 37°C in a humidified atmosphere containing 5% C02. The assay is based on the ability of viable cells to reduce a yellow soluble tetrazolium salt 3- ( 4 , 5-dimethylthiazolyl ) -2 , 5- diphenyltetrazolium bromide (MTT) , to an insoluble purple formazan precipitate. Following incubation of the plates for 4 days (to allow control cells to increase 10-fold in number) , 20 μL of a 5 mg/mL solution of MTT in phosphate-buffered saline was added to each well and the plates were incubated for a further 5 hours. The plates were then centrifuged for 5 minutes at 300g and the bulk of the medium was removed from the cell pellet. DMSO (200 μL) was added to each well, and the samples agitated to ensure complete mixing. The optical density was then measured at a wavelength of 550 nm on a Titertek Multiscan ELISA plate reader and expressed as a percentage of the control optical density. For each curve, an IC50 value was read as the dose required to reduce the final optical density to 50% of the control value.
The PBD prodrug compound 11 investigated was itself shown to have negligible cytotoxicity (ICE0= 47.5 μM; Figure 6). Following irradiation, significantly increased cytotoxicity was observed (figure 6) with an IC50 value of 0.6 μM achieved after 1 hour of irradiation. The IC50 value of authentic benzyl DC-81 under O 00/12507
75 similar conditions was 0.5 μM.
Figure imgf000077_0001
* comparative example
irradiation was in DMF at lmM initial drug concentration
The efficiency of photoinduced cleavage was reduced at the higher concentration of lO M of prodrug in DMF, when 2 hours irradiation gave an ICS0 value of 1.75 μM, see Figure 7.
10
Figure imgf000077_0002
* comparative example
+ irradiation was in DMF at lOmM initial drug concentration
It was thought that the high concentration of prodrug might be preventing efficient absorption of UV light. Furthermore, changing the solvent from DMF to methanol resulted in a less efficient conversion, see Figure 8.
20
Figure imgf000077_0003
comparative example
irradiation was in DMF at lmM initial drug concentration O 00/12507
76
Example 11: Light-activation of the dimer DSB-120 prodrug (Compound 32)
The dimer derivative 32, see example 5(b), was found to be slightly cytotoxic before UV irradiation with an IC50 of 4.5 μM. Incubation of a solution of compound 32 with cells following 2 hours irradiation reduced the IC50 to 1.25 μM (figure 9). This was further reduced to 0.85 μM following 5 hours irradiation. The parent PBD dimer DSB-120 gave an IC50 of 0.55 μM in the same cell line, see Figure 9.
10
Figure imgf000078_0001
* Comparative example
+ irradiation was in DMF at lmM initial drug concentration
For these experiments, the HPLC data (not shown) suggested that almost complete conversion of the prodrug 32 (retention time = 5.18 minutes) to a product with retention time = 3.64 minutes occurred by 2 hours irradiation, with complete conversion by 5 hours. Authentic DSB-120 eluted with a retention time of 3.64 minutes.
Example 12 : Comparison of Biological Activity of Psec and Ptec protected PBDs (Compounds 52 and 55)
Compounds 52 (UP 2073) and the comparative compound 55b (UP2090) were evaluated for their cytotoxic activity in ovarian cell lines by Dr Lloyd R. Kelland' s group at The Institute of Cancer Research, Sutton, UK. The five cell lines investigated were SKOV-3, A2780/A2780cisR and CHl/CHlcisR (cisR denotes that the cell line is resistant to cisplatin) . 00/12507
77
Single viable cells were seeded in growth medium (160 μL) in 96- well microtitre plates and allowed to attach overnight. The test compounds were then dissolved in DMSO (to give 20 mM drug concentrations) immediately prior to adding the cells in quadruplicate wells. The final drug concentrations in the wells ranged from lOOμM to 2.5nM as follows: 100, 25, 10, 2.5, 1 μM, 250, 100, 25, 10, 2.5 nM (drugs were diluted in growth medium and then 40 μL added to the existing well volume of 160 μL to give final concentrations as above) . After 96 hours, the medium was removed and the remaining cells fixed by exposure to 10% trichloroacetic acid on ice for 30 minutes. The wells were then washed 3-4 times with tap water, air dried overnight and treated with 100 μL of sulphorhodamine B (0.4%) dissolved in 1% acetic acid. Staining was allowed to continue for 10-15 minutes, then the wells were washed 3-4 times with 1% acetic acid, air dried and then added to Tris base (100 μL of lOmM) . Plates were then shaken and absorbance readings at 540 nM were determined using a plate reader. The IC50 values were calculated from plots of concentration versus percentage absorbance (compared with 8 untreated wells).
The assay was also carried out using compound 56 (UP2025):
Figure imgf000079_0001
56
which is the unprotected version of compound 52. O 00/12507
The results are shown below:
Figure imgf000080_0001
RF is the resistance factor, which is the cytotoxicity of the compound in the cisplatin resistant cell line divided by the cytotoxicity in the normal cell line.
The Ptec protected PBD (55b), UP2090, was essentially inactive in against the ovarian cell lines, as expected as the Ptec protecting group lacks the acidic protons thought to be required to trigger the fragmentation of the protecting group when exposed to GST. However, the Psec-protected prodrug (52) UP2073 was found to be at least 50 times more toxic than the Ptec control. The compound was particularly active against the SKOV3 cell line; this is noteworthy as this cell line is intrinsically resistant to electrophilic cytotoxic agents due to the presence of high levels of glutathione/glutathione transferase. Interestingly, the Psec prodrug UP2073 was actually more active in this cell line than the PBD it was based upon UP2025. Without wishing to be bound by theory, it is possible that the prodrug is protected from glutathione and other biological nucleophiles until it is deprotected close to the site of action.
UP2073 also underwent screening carried out by The National Cancer Institute (NCI), Bethesda, Maryland USA. The NCI has available an in vi tro cytotoxicity screen which consists of approximately 60 human tumour cell lines against which compounds are tested at a minimum of five concentrations each differing 10- fold. A 48 hour continuous exposure protocol is used, where cell viability or growth is estimated with an SRB protein assay. O 00/12507
79
Method
The test compound was evaluated against approximately 60 human tumour cell lines. The NCI screening procedures were described in detail by Monks and co-workers (Monks, A et al . , Journal of the National Cancer Institute, 1991, 83, 757) . Briefly, cell suspensions were diluted according to the particular cell type and the expected target cell density (5000-40,000 cells per well based on cell growth characteristics), and added by pipette (100 UL) into 96-well microtitre plates. The cells were allowed a preincubation period of 24 hours at 37°C for stabilisation.
Dilutions at twice the intended test concentration were added at time zero in 100 μL aliquots to the wells. The test compounds were evaluated at five 10-fold dilutions (10~4, 10"5, 10'6, 10"7 and 10"8 μM) . The test compounds were incubated for 48 hours in 5% C02 atmosphere and 100% humidity. The cells were then assayed using the sulphorhodamine B assay. A plate reader was used to read the optical densities and a microcomputer processed the readings into LC50 values, which is the dosage required to kill half of the cells. IC50 values, the dosage required to inhibit the growth of half the cells, was also measured.
UP2073 did well in the screening showing activity against cell lines in the lung, colon, CNS, melanoma, renal and breast tumour cell line panels. Interestingly, analysis of the LC50 data across 53 cell lines suggested some correlation to glutathione transferase activity. Selected results are shown below:
Figure imgf000081_0001
Key to Figures
Figure 4 SW1116 Compound 7; (•) Compound 7 + enzyme + NADH O 00/12507
80 LS174T - (A) Compound 7; (T) Compound 7 + enzyme + NADH
Figure 6 & 7: (■) Benzyl DC-81; (□) Compound 11; (•) Compound 11 + UVA 30 mins; (O) Compound 11 + UVA lh; (A) Compound 11 + UVA 2 h.
Figure 8: (■) Benzyl DC-81; (D) Compound 11; (•) Compound 11 + UVA 30 mins; (O) Compound 11 + UVA lh; (A) Compound 11 + UVA 2 h
Figure 9: (■) DSB-120; (□) Compound 32; (•) Compound 32 + UVA 30 mins; (O) Compound 32 + UVA lh; (A) Compound 32 + UVA 2h; (Δ) Compound 32 + UVA 5h

Claims

O 00/1250781CLAIMS
1. A compound with the formula I:
Figure imgf000083_0001
wherein:
R10 is a therapeutically removable nitrogen protecting group; R2 and R3 are independently selected from: H, R, OH, OR, =0, =CH-R, =CH2, CH2-C02R, CH2-C02H, CH2-S02R, 0-S02-R, C02R, COR and CN; R6, R7 and R9 are independently selected from H, R, OH, OR, halo, amino, nitro, Me3Sn; or R7 and R8 together form a group -O- (CH2)p-0-, where p is 1 or 2. X is S, O or NH;
RX1 is either H or R; where R is a lower alkyl group having 1 to 10 carbon atoms, or an aralkyl group, of up to 12 carbon atoms, whereof the alkyl group optionally contains one or more carbon-carbon double or triple bonds, which may form part of a conjugated system, or an aryl group, of up to 12 carbon atoms; and is optionally substituted by one or more halo, hydroxy, amino, or nitro groups, and optionally contains one or more hetero atoms, which may form part of, or be, a functional group; and where there is optionally a double bond between Cl and C2 or C2 and C3; and R8 is selected from H, R, OH, OR, halo, amino, nitro, Me3Sn, where R is as defined above, or the compound is a dimer with each monomer being the same or different and being of formula I, where the R8 groups of the monomers form together a bridge having the formula -T-R' -T- linking the monomers, where R' is an alkylene chain containing from 3 to 12 carbon atoms, which chain may be interrupted by one or more hetero atoms and/or aromatic rings, and may contain one or more carbon-carbon double or triple bonds, and each T is independently selected from O, S or N.
2. A compound according to claim 1, wherein R10 is of the formula II:
Figure imgf000084_0001
wherein n is 0 to 3, R(I) is H or R, and R(II) is one or more optional substituents independently selected from N02, OR, or R, where R is as defined in claim 1, and if two substituents R(II) are on adjacent atoms, they may together be of the formula -0-(CH2) m-0- where m is 1 or 2.
3. A compound according to either claim 1 or claim 2, wherein R10 is a photolabile protecting group.
4. A compound according to claim 3, wherein R10 is cleavable by light with a wavelength of 250 to 550 nm.
5. A compound according to either claim 1 or claim 2 wherein R10 is an enzyme labile group.
6. A compound according to claim 5, wherein R10 is a nitroreductase labile group.
7. A compound according to claim 5, wherein R10 is a penicillin V/G amidase labile group.
8. A compound according to claim 5, wherein R10 is a L-╬│- glutamyl transpeptidase labile group.
9. A compound according to claim 5, wherein R10 is a glutathione transferase labile group.
10. A compound according to claim 6, wherein R10 is of the formula III: O 00/12507
Figure imgf000085_0001
wherein n is 0 to 3, R(I) is H or R, and R( is one or more optional substituents independently selected from N020, OR, or R, where R is as defined in claim 1, and if two substituents R(III) are on adjacent atoms, they may together be of the formula -0-(CH2)m-0- where m is 1 or 2.
11. A compound according to claim 9, wherein R10 is of the formula XI:
Figure imgf000085_0002
wherein R is as defined in claim 1, and n is 0 to 3.
12. A compound according to claim 11, wherein R in the group formula XI is a substituted or unsubstituted phenyl group.
13. A compound according to any one of the preceding claims, wherein X is O.
14. A compound according to any one of the preceding claims, wherein Ru is H.
15. A compound according to any one of the preceding claims, wherein R6 and R9 are H.
16. A compound according to claim 15, wherein R7 and Re are independently selected from H, OH, and OR, where R is as defined in claim 1. O 00/12507
84
17. A compound according to any one of the preceding claims, wherein the compound is a C8 dimer, wherein R6 and R9 are H, and R7 is independently selected from H, OH, and OR, where R is as defined in claim 1.
18. A compound according to any one of the preceding claims, wherein the compound is a C8 dimer, and R' is -O- (CH2)p-0-, where p is from 1 to 12.
19. A method of preparing a compound of formula I as defined in any one of claims 1 to 18 wherein XR1X Γëá OH from a compound of formula la:
Figure imgf000086_0001
wherein the substituents of the compound of formula la are the same as for the compound of formula I to be prepared.
20. A method of preparing a compound of formula la as defined in claim 19 by the oxidation of a compound of formula IVa:
Figure imgf000086_0002
wherein the substituents of the compound of formula IVa are the same as for the compound of formula la to be prepared.
21. A method according to claim 20, wherein the oxidation is a Swern oxidation.
22. A method of preparing a compound of formula IVa as defined in claim 20, by reacting a compound of formula Va:
Figure imgf000087_0001
with a compound of formula VI:
Y-R10 (VI) wherein the substituents of the compounds of formulae Va and VI are the same as for the compound of formula IVa to be prepared, and Y is a halogen atom.
23. A method of preparing a compound of formula IVa according to claim 20, wherein the compound of formula VI is a haloformate of the formula Y-A, where A is a group of formula II as defined in claim 2.
24. A method of making a compound of formula IVa as defined in claim 20, by reacting a compound of formula VII:
Figure imgf000087_0002
with a compound of formula VI:
Figure imgf000087_0003
to form a compound of formula VIII:
Figure imgf000087_0004
and then reacting the compound of formula VIII with a compound of formula IXa:
^OH
Figure imgf000088_0001
via the formation of an acid chloride, wherein the substituents for compounds of formulae VI, VII, VIII and IXa are the same as for the compound of formula Va to be prepared, and where Y is a halogen atom.
25. A method of preparing a compound of formula la as defined in claim 19 by the unmasking of a compound of formula IVb:
Figure imgf000088_0002
wherein the substituents of the compound of formula IVb are the same as for the compound of formula la to be prepared, Q is either O or S and R(IV) is Me or Et, or together form -(CH2)q- where q is 2 or 3.
26. A method according to claim 25, wherein Q is S, R(1V> is Et and the unmasking is mercury-mediated unmasking.
27. A method according to claim 25, wherein Q is O, R(1V) is Me and the unmasking is palladium-mediated unmasking.
28. A method of preparing a compound of formula IVb as defined in claim 25, by reacting a compound of formula Vb: O 00/12507
87
Figure imgf000089_0001
with a compound of formula VI:
Figure imgf000089_0002
wherein the substituents of the compounds of formulae Vb and VI are the same as for the compound of formula IVb to be prepared, and Y is a halogen atom.
29. A method of preparing a compound of formula IVb according to claim 28, wherein the compound of formula VI is a haloformate of the formula Y-A, where A is a group of formula II as defined in claim 2.
30. A method of making a compound of formula IVb as defined in claim 25, by reacting a compound of formula VII:
Figure imgf000089_0003
with a compound of formula VI : Y-R10 (v╬╣; to form a compound of formula VIII:
Figure imgf000089_0004
and then reacting the compound of formula VIII with a compound of O 00/12507
formula IXb:
Figure imgf000090_0001
via the formation of an acid chloride, wherein the substituents for compounds of formulae VI, VII, VIII and IXb are the same as for the compound of formula Vb to be prepared, and where Y is a halogen atom.
31. A method of making a compound of formula X:
Figure imgf000090_0002
by the cleavage of the therapeutically removable protecting group R10 of a compound of formula I as defined in any one of claims 1 to 18, wherein the substituent groups of the compound of formula X are the same as the substituent groups of compound I used.
32. The use of a compound of formula I as defined in any one of claims 5 to 8 or 10 in conjunction with an appropriate enzyme in methods of ADEPT or GDEPT therapy.
33. The use of compounds of formula I as defined in any one of claims 2 to 4 in conjunction with light of wavelengths between 250 and 550 nm in methods of PDT.
34. The use of compounds of formula I as defined in claim 9 or 11 in methods of NPEPT. O 00/12507
89
35. A pharmaceutical composition comprising a compound of formula I as defined in any one of claims 1 to 18 and a pharmaceutically acceptable carrier or diluent.
36. The use of a compound of formula I as defined in any one of claims 1 to 18 to prepare a medicament for the treatment of a neoplastic disease.
37. The use of a compound of formula I as defined in any one of claims 1 to 18 to prepare a medicament for the treatment of a bacterial, viral or parasitic infection.
PCT/GB1999/002837 1998-08-27 1999-08-27 Pyrrolobenzodiazepines WO2000012507A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
DK99941766T DK1109811T3 (en) 1998-08-27 1999-08-27 pyrrolobenzodiazepines
DE69910227T DE69910227T2 (en) 1998-08-27 1999-08-27 PYRROLOBENZODIAZEPINE
AT99941766T ATE246687T1 (en) 1998-08-27 1999-08-27 PYRROLOBENZODIAZEPINES
JP2000571053A JP4669611B2 (en) 1998-08-27 1999-08-27 Compound
EP99941766A EP1109811B1 (en) 1998-08-27 1999-08-27 Pyrrolobenzodiazepines
AU55261/99A AU758398B2 (en) 1998-08-27 1999-08-27 Pyrrolobenzodiazepines
NZ510492A NZ510492A (en) 1998-08-27 1999-08-27 5H-pyrrolo[1,2-c][1,4]benzodiazepin-5-one derivatives useful for treating neoplastic diseases
CA002341968A CA2341968C (en) 1998-08-27 1999-08-27 Pyrrolobenzodiazepine compounds
US09/763,814 US6562806B1 (en) 1998-08-27 1999-08-27 Pyrrolobenzodiazepines

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9818731.3 1998-08-27
GBGB9818731.3A GB9818731D0 (en) 1998-08-27 1998-08-27 Compounds

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09/763,814 A-371-Of-International US6562806B1 (en) 1998-08-27 1999-08-27 Pyrrolobenzodiazepines
US10/379,049 Continuation US20030195196A1 (en) 1998-08-27 2003-03-04 Pyrrolobenzodiazepines

Publications (3)

Publication Number Publication Date
WO2000012507A2 true WO2000012507A2 (en) 2000-03-09
WO2000012507A3 WO2000012507A3 (en) 2000-08-31
WO2000012507A8 WO2000012507A8 (en) 2000-10-19

Family

ID=10837953

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1999/002837 WO2000012507A2 (en) 1998-08-27 1999-08-27 Pyrrolobenzodiazepines

Country Status (12)

Country Link
US (2) US6562806B1 (en)
EP (1) EP1109811B1 (en)
JP (1) JP4669611B2 (en)
AT (1) ATE246687T1 (en)
CA (1) CA2341968C (en)
DE (1) DE69910227T2 (en)
DK (1) DK1109811T3 (en)
ES (1) ES2205872T3 (en)
GB (1) GB9818731D0 (en)
NZ (1) NZ510492A (en)
PT (1) PT1109811E (en)
WO (1) WO2000012507A2 (en)

Cited By (102)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2825705A1 (en) * 2001-06-08 2002-12-13 Aventis Pharma Sa New benzo or heterocyclo fused aza-bicycloalkane derivatives, are antibacterial agents effective against Gram positive and Gram negative bacteria e.g. Staphylococcus aureus and Escherichia coli
WO2005023814A1 (en) * 2003-09-11 2005-03-17 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
WO2005085251A1 (en) 2004-03-01 2005-09-15 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4]benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
WO2005085260A1 (en) * 2004-03-09 2005-09-15 Spirogen Limited Pyrrolobenzodiazepines
WO2005121080A1 (en) * 2004-06-02 2005-12-22 Eli Lilly And Company Histamine h3 receptor agents, preparation and therapeutic uses
WO2005085259A3 (en) * 2004-03-01 2006-01-05 Spirogen Ltd Pyrrolobenzodiazepines as key intermediates in the synthesis of dimeric cytotoxic pyrrolobenzodiazepines
US7049311B1 (en) 1998-08-27 2006-05-23 Spirogen Limited Pyrrolbenzodiazepines
US7365071B2 (en) 2002-09-05 2008-04-29 Branislav Musicki Heterocyclic compounds, preparation process and intermediates, and use as medicaments, in particular as β-lactamase inhibitors and antibacterials
US7407951B2 (en) 2002-11-14 2008-08-05 Spirogen Limited Pyrrolobenzodiazepines
US7612087B2 (en) 2002-01-28 2009-11-03 Novexel Heterocyclic compounds as inhibitors of beta-lactamases
US7612062B2 (en) 2005-04-21 2009-11-03 Spirogen Limited Pyrrolobenzodiazepines
US7704924B2 (en) 1998-08-27 2010-04-27 Spirogen Limited Library of compounds comprising pyrrolobenzodiazepine moieties
WO2011023883A1 (en) 2009-08-25 2011-03-03 Sanofi-Aventis Conjugates of pyrrolo[1,4]benzodiazepine dimers as anticancer agents
WO2011039724A1 (en) 2009-10-02 2011-04-07 Sanofi-Aventis Antibodies that specifically bind to the epha2 receptor
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
WO2011128650A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Intermediates useful for the synthesis of pyrrolobenzodiazepines
EP2393362A1 (en) * 2009-02-05 2011-12-14 Immunogen, Inc. Novel benzodiazepine derivatives
WO2012014147A1 (en) 2010-07-26 2012-02-02 Sanofi Anticancer derivatives, preparation thereof and therapeutic use thereof
US8163736B2 (en) 2006-01-25 2012-04-24 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives
US8404678B2 (en) 2007-07-19 2013-03-26 Sanofi Cytotoxic agents comprising new tomaymycin derivatives and their therapeutic use
WO2013053872A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Synthesis method and intermediates useful in the preparation of pyrrolobenzodiazepines
WO2013055987A1 (en) * 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US8460667B2 (en) 2006-07-18 2013-06-11 Sanofi EPHA2 receptor antagonist antibodies
US8501934B2 (en) 2008-07-22 2013-08-06 Spirogen Sarl Pyrrolobenzodiazepines
US8592576B2 (en) 2008-10-17 2013-11-26 Spirogen Sarl Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
WO2013177481A1 (en) 2012-05-25 2013-11-28 Immunogen, Inc. Benzodiazepines and conjugates thereof
WO2014018625A1 (en) 2012-07-25 2014-01-30 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
US8697688B2 (en) 2010-04-15 2014-04-15 Seattle Genetics Inc. Pyrrolobenzodiazepines used to treat proliferative diseases
WO2014057122A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
WO2014057120A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2014057119A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2014057117A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2014057113A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine - anti-psma antibody conjugates
US8765740B2 (en) 2011-02-15 2014-07-01 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
WO2015157592A1 (en) 2014-04-11 2015-10-15 Medimmune, Llc Bispecific her2 antibodies
WO2015179658A2 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
US9242013B2 (en) 2010-04-15 2016-01-26 Seattle Genetics Inc. Targeted pyrrolobenzodiazapine conjugates
WO2016040868A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040856A2 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016044396A1 (en) 2014-09-17 2016-03-24 Genentech, Inc. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
WO2016054315A1 (en) 2014-10-01 2016-04-07 Medimmune, Llc Method of conjugating a polypeptide
US9321774B2 (en) 2012-04-30 2016-04-26 Medimmune Limited Pyrrolobenzodiazepines
US9376440B2 (en) 2012-04-30 2016-06-28 Medimmune Limited Pyrrolobenzodiazepines as antiproliferative agents
WO2016166304A1 (en) 2015-04-15 2016-10-20 Van Berkel Patricius Hendrikus Cornelis Site-specific antibody-drug conjugates
EP3095797A1 (en) 2012-02-24 2016-11-23 Stemcentrx, Inc. Anti dll3 antibodies and methods of use thereof
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2017059289A1 (en) 2015-10-02 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
AU2015224492B2 (en) * 2009-02-05 2017-04-20 Immunogen, Inc. Novel benzodiazepine derivatives
US9707301B2 (en) 2011-10-14 2017-07-18 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US9713647B2 (en) 2011-10-14 2017-07-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
WO2017132298A1 (en) 2016-01-27 2017-08-03 Medimmune, Llc Methods for preparing antibodies with a defined glycosylation pattern
US9745303B2 (en) 2012-10-12 2017-08-29 Medimmune Limited Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
WO2017180813A1 (en) 2016-04-15 2017-10-19 Macrogenics, Inc. Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
WO2017194568A1 (en) 2016-05-11 2017-11-16 Sanofi Treatment regimen using anti-muc1 maytansinoid immunoconjugate antibody for the treatment of tumors
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2018027204A1 (en) 2016-08-05 2018-02-08 Genentech, Inc. Multivalent and multiepitopic anitibodies having agonistic activity and methods of use
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
US9956298B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2018091724A1 (en) 2016-11-21 2018-05-24 Cureab Gmbh Anti-gp73 antibodies and immunoconjugates
EP3338793A1 (en) 2013-08-28 2018-06-27 AbbVie Stemcentrx LLC Novel sez6 modulators and methods of use
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10017580B2 (en) 2014-04-15 2018-07-10 ADC Therpeutics S.A. Humanized anti-Tn-MUC1 antibodies and their conjugates
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10059768B2 (en) 2014-09-12 2018-08-28 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10179820B2 (en) 2014-09-12 2019-01-15 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
EP3498735A1 (en) 2006-10-19 2019-06-19 Sanofi Novel anti-cd38 antibodies for the treatment of leukemia
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10544223B2 (en) 2017-04-20 2020-01-28 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
US10624972B2 (en) 2015-03-13 2020-04-21 Endocyte, Inc. Conjugates for treating diseases
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2020245283A1 (en) 2019-06-07 2020-12-10 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
EP3763740A1 (en) 2011-01-26 2021-01-13 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
US10899775B2 (en) 2015-07-21 2021-01-26 Immunogen, Inc. Methods of preparing cytotoxic benzodiazepine derivatives
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11318211B2 (en) 2017-06-14 2022-05-03 Adc Therapeutics Sa Dosage regimes for the administration of an anti-CD19 ADC
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
US11583590B2 (en) 2017-09-29 2023-02-21 Daiichi Sankyo Company, Limited Antibody-pyrrolobenzodiazepine derivative conjugate and method of use thereof for treating a tumor
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
EP4223319A2 (en) 2017-05-26 2023-08-09 MedImmune, LLC Method and molecules
US11807685B2 (en) 2021-08-05 2023-11-07 The Uab Research Foundation Anti-CD47 antibody and uses thereof
WO2023215737A1 (en) 2022-05-03 2023-11-09 Genentech, Inc. Anti-ly6e antibodies, immunoconjugates, and uses thereof

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9818731D0 (en) * 1998-08-27 1998-10-21 Univ Portsmouth Compounds
GB9818732D0 (en) * 1998-08-27 1998-10-21 Univ Portsmouth Collection of compounds
WO2004080284A2 (en) * 2003-03-07 2004-09-23 Board Of Regents, The University Of Texas System Antibody-targeted photodynamic therapy
GB0404574D0 (en) * 2004-03-01 2004-04-07 Spirogen Ltd Amino acids
FR2869231B1 (en) * 2004-04-27 2008-03-14 Sod Conseils Rech Applic THERAPEUTIC COMPOSITION CONTAINING AT LEAST ONE PYRROLOBENZODIAZEPINE DERIVATIVE AND FLUDARABINE
GB0410725D0 (en) * 2004-05-13 2004-06-16 Spirogen Ltd Pyrrolobenzodiazepine therapeutic agents
JP2008525429A (en) * 2004-12-27 2008-07-17 カウンシル オブ サイエンティフィック アンド インダストリアル リサーチ Pyrrolo [2,1-C] [1,4] benzodiazepine-anthraquinone conjugate useful as an antitumor agent
JO2768B1 (en) * 2005-07-29 2014-03-15 تيبوتيك فارماسيوتيكالز ليمتد Macrocylic Inhibitors Hepatitis C Virus
US8637664B2 (en) * 2005-10-05 2014-01-28 Spirogen Sarl Alkyl 4- [4- (5-oxo-2,3,5, 11a-tetrahydo-5H-pyrrolo [2, 1-c] [1,4] benzodiazepine-8-yloxy)-butyrylamino]-1H-pyrrole-2-carboxylate derivatives and related compounds for the treatment of a proliferative disease
GB0619325D0 (en) * 2006-09-30 2006-11-08 Univ Strathclyde New compounds
EP2188297B1 (en) * 2007-08-01 2012-02-01 Council of Scientific & Industrial Research Pyrrolo [2,1-c][1, 4] benzodiazepine-glycoside prodrugs useful as a selective anti tumor agent
JP5187534B2 (en) * 2010-02-12 2013-04-24 有機合成薬品工業株式会社 Process for producing N-tert-butoxycarbonyl-4-formylpiperidine
UA110640C2 (en) * 2011-05-05 2016-01-25 Янссен Фармацевтика Нв Antifungal 5,6-dihydro-4h-pyrrolo-[l,2-a][l,4]benzodiazepines and 6h-pyrrolo[1,2-a][l,4]benzodiazepines substituted with heterocyclic derivatives
KR101860174B1 (en) 2011-09-20 2018-05-21 메디뮨 리미티드 Pyrrolobenzodiazepines as unsymmetrical dimeric pbd compounds for inclusion in targeted conjugates
WO2013053873A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines
EP2751110B1 (en) 2011-10-14 2017-04-19 MedImmune Limited Asymmetrical bis-(5H-Pyrrolo[2,1-c][1,4]benzodiazepin-5-one) derivatives for the treatment of proliferative and autoimmune diseases
CN110256469B (en) 2012-10-12 2022-05-17 麦迪穆有限责任公司 Pyrrolobenzodiazepines and conjugates thereof
ES2701076T3 (en) 2012-11-24 2019-02-20 Hangzhou Dac Biotech Co Ltd Hydrophilic linkers and their uses for the conjugation of drugs to molecules that bind to cells
CN110452242A (en) 2012-12-21 2019-11-15 麦迪穆有限责任公司 Pyrrolobenzodiazepines Zhuo and its conjugate
JP6527466B2 (en) 2012-12-21 2019-06-05 メドイミューン・リミテッドMedImmune Limited Asymmetric pyrrolobenzodiazepine dimers for use in the treatment of proliferative and autoimmune diseases
AU2014318545A1 (en) 2013-09-12 2016-03-24 Halozyme, Inc. Modified anti-epidermal growth factor receptor antibodies and methods of use thereof
CN114262344A (en) 2014-02-28 2022-04-01 杭州多禧生物科技有限公司 Charged linkers and their use in conjugation reactions
NZ739830A (en) 2015-07-12 2021-12-24 Hangzhou Dac Biotech Co Ltd Bridge linkers for conjugation of cell-binding molecules
US9839687B2 (en) 2015-07-15 2017-12-12 Suzhou M-Conj Biotech Co., Ltd. Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
KR20220147721A (en) 2016-11-14 2022-11-03 항저우 디에이씨 바이오테크 씨오, 엘티디 Conjugation linkers, cell binding molecule-drug conjugates containing the likers, methods of making and uses such conjugates with the linkers
US20210070845A1 (en) 2017-12-15 2021-03-11 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
JP2022513482A (en) 2018-12-21 2022-02-08 アビディティー バイオサイエンシーズ,インク. Anti-transferrin receptor antibody and its use
CN116063520A (en) 2019-01-30 2023-05-05 真和制药有限公司 anti-GAL 3 antibodies and uses thereof
CN115666589A (en) 2020-03-19 2023-01-31 艾维迪提生物科学公司 Compositions and methods for treating facioscapulohumeral muscular dystrophy
CN115697418A (en) 2020-03-27 2023-02-03 艾维迪提生物科学公司 Compositions and methods for treating muscular dystrophy
CA3231330A1 (en) 2021-09-16 2023-03-23 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3523941A (en) * 1967-03-06 1970-08-11 Hoffmann La Roche Benzodiazepine compounds and process for their preparation

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3524849A (en) 1967-10-27 1970-08-18 Hoffmann La Roche Process for the preparation of pyrrolo-benzodiazepine acrylamides and intermediates useful therein
FR2027356A1 (en) 1968-12-30 1970-09-25 Fujisawa Pharmaceutical Co Benzodiazepinone antibiotics
JPS585916B2 (en) * 1977-12-27 1983-02-02 株式会社ミドリ十字 New benzodiazepine compounds
JPS5615289A (en) * 1979-07-17 1981-02-14 Green Cross Corp:The Novel benzodiazepinnbased compound 3
JPS58180487A (en) 1982-04-16 1983-10-21 Kyowa Hakko Kogyo Co Ltd Antibiotic dc-81 and its preparation
FR2586683B1 (en) 1985-08-29 1988-07-01 Centre Nat Rech Scient NOVEL NEOTHRAMYCIN DERIVATIVES, THEIR PREPARATION PROCESS AND THEIR APPLICATION AS MEDICAMENTS
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
GB8705477D0 (en) 1987-03-09 1987-04-15 Carlton Med Prod Drug delivery systems
GB8809616D0 (en) 1988-04-22 1988-05-25 Cancer Res Campaign Tech Further improvements relating to drug delivery systems
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
FR2676230B1 (en) 1991-05-07 1993-08-27 Centre Nat Rech Scient NOVEL PYRROLO [1,4] -BENZODIAZEPINES DERIVATIVES, PROCESS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THEM.
EP0540263A1 (en) 1991-10-23 1993-05-05 Cancer Research Campaign Technology Limited Bacterial nitroreductase for the reduction of CB 1954 and analogues thereof to a cytotoxic form
GB9205051D0 (en) 1992-03-09 1992-04-22 Cancer Res Campaign Tech Pyrrolobenzodiazepine derivatives,their preparation,and compositions containing them
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
GB9818732D0 (en) * 1998-08-27 1998-10-21 Univ Portsmouth Collection of compounds
AU757510C (en) * 1998-08-27 2003-09-11 Medimmune Limited Pyrrolobenzodiazepines
GB9818731D0 (en) * 1998-08-27 1998-10-21 Univ Portsmouth Compounds
GB9818730D0 (en) * 1998-08-27 1998-10-21 Univ Portsmouth Collections of compounds
US6660856B2 (en) * 2002-03-08 2003-12-09 Kaohsiung Medical University Synthesis of pyrrolo[2,1-c][1,4]benzodiazepine analogues

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3523941A (en) * 1967-03-06 1970-08-11 Hoffmann La Roche Benzodiazepine compounds and process for their preparation

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BARALDI P G ET AL: "Design, synthesis and biological activity of a pyrrolo [2,1-cÜ[1,4Übenzodiazepine (PBD)-distamycin hybrid" BIOORGANIC & MEDICINAL CHEMISTRY LETTERS,GB,OXFORD, vol. 8, no. 21, 3 November 1998 (1998-11-03), pages 3019-3024, XP004141867 ISSN: 0960-894X *
GUIOTTO A ET AL: "Synthesis of novel C7-aryl substituted pyrrolo[2,1-cÜ[1,4Übenzodiaze pines (PBDs) via Pro-N10-troc protection and suzuki coupling" BIOORGANIC & MEDICINAL CHEMISTRY LETTERS,GB,OXFORD, vol. 8, no. 21, 3 November 1998 (1998-11-03), pages 3017-3018, XP004141866 ISSN: 0960-894X *
S.J. GREGSON ET AL.: "Synthesis of a novel C2/C2'-exo unsaturated pyrrolobenzodiazepine cross-linking agent with remarkable DNA binding affinity and cytotoxicity" CHEMICAL COMMUNICATIONS.,1999, pages 797-98, XP002136001 ROYAL SOCIETY OF CHEMISTRY., GB ISSN: 1359-7345 *
T. FUKUYAMA ET A.: "Total synthesis of (+)-porothramycin B" TETRAHEDRON LETTERS., vol. 34, no. 16, 1993, pages 2577-2580, XP002135999 ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM., NL ISSN: 0040-4039 *
T. NAGASAKA ET AL.: "Stereoselective synthesis of tilivalline" JOURNAL OF ORGANIC CHEMISTRY., vol. 63, no. 20, 1998, pages 6797-6801, XP002136000 AMERICAN CHEMICAL SOCIETY. EASTON., US ISSN: 0022-3263 *
T. NAGASAKA ET AL.: "Stereoselective synthesis of tilivalline" TETRAHEDRON LETTERS., vol. 30, no. 14, 1989, pages 1871-72, XP002135998 ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM., NL ISSN: 0040-4039 *
WILSON S C ET AL: "Design and Synthesis of a Novel Epoxide-Containing Pyrrolo[2,1-cÜ[1,4Übenzodiazepine (PBD) via a New Cyclization Procedure" TETRAHEDRON LETTERS,NL,ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, vol. 36, no. 35, 28 August 1995 (1995-08-28), pages 6333-6336, XP004027383 ISSN: 0040-4039 *

Cited By (205)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7265105B2 (en) 1998-08-27 2007-09-04 Spirogen Limited Pyrrolobenzodiazepines
US7704924B2 (en) 1998-08-27 2010-04-27 Spirogen Limited Library of compounds comprising pyrrolobenzodiazepine moieties
US7049311B1 (en) 1998-08-27 2006-05-23 Spirogen Limited Pyrrolbenzodiazepines
US7067511B2 (en) 1998-08-27 2006-06-27 Spirogen Limited Pyrrolobenzodiazepines
WO2002100860A2 (en) * 2001-06-08 2002-12-19 Aventis Pharma S.A. Heterocyclic compounds, method for preparing same and use thereof as medicines, in particular as anti-bacterial agents
WO2002100860A3 (en) * 2001-06-08 2003-11-20 Aventis Pharma Sa Heterocyclic compounds, method for preparing same and use thereof as medicines, in particular as anti-bacterial agents
FR2825705A1 (en) * 2001-06-08 2002-12-13 Aventis Pharma Sa New benzo or heterocyclo fused aza-bicycloalkane derivatives, are antibacterial agents effective against Gram positive and Gram negative bacteria e.g. Staphylococcus aureus and Escherichia coli
US8148540B2 (en) 2001-06-08 2012-04-03 Astrazeneca Heterocyclic compounds, their preparation and their use as medicaments, in particular as anti-bacterial agents
EP1798231A3 (en) * 2001-06-08 2008-02-27 Novexel Intermediate products for the preparation of antibacterial compounds
US7612087B2 (en) 2002-01-28 2009-11-03 Novexel Heterocyclic compounds as inhibitors of beta-lactamases
US7365071B2 (en) 2002-09-05 2008-04-29 Branislav Musicki Heterocyclic compounds, preparation process and intermediates, and use as medicaments, in particular as β-lactamase inhibitors and antibacterials
US7407951B2 (en) 2002-11-14 2008-08-05 Spirogen Limited Pyrrolobenzodiazepines
US7429658B2 (en) 2003-09-11 2008-09-30 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
WO2005023814A1 (en) * 2003-09-11 2005-03-17 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
WO2005085259A3 (en) * 2004-03-01 2006-01-05 Spirogen Ltd Pyrrolobenzodiazepines as key intermediates in the synthesis of dimeric cytotoxic pyrrolobenzodiazepines
EP2270010A1 (en) 2004-03-01 2011-01-05 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4]benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepins
US7741319B2 (en) 2004-03-01 2010-06-22 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
US7557099B2 (en) 2004-03-01 2009-07-07 Spirogen Limited Pyrrolobenzodiazepines as key intermediates in the synthesis of dimeric cytotoxic pyrrolobenzodiazepines
WO2005085251A1 (en) 2004-03-01 2005-09-15 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4]benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
WO2005085260A1 (en) * 2004-03-09 2005-09-15 Spirogen Limited Pyrrolobenzodiazepines
US7528126B2 (en) 2004-03-09 2009-05-05 Spirogen Limited Pyrrolobenzodiazepines
WO2005121080A1 (en) * 2004-06-02 2005-12-22 Eli Lilly And Company Histamine h3 receptor agents, preparation and therapeutic uses
US7696234B2 (en) 2004-06-02 2010-04-13 Eli Lilly And Company Histamine H3 receptor agents, preparation and therapeutic uses
CN1956952B (en) * 2004-06-02 2013-12-11 伊莱利利公司 Histamine H3 receptor agents, preparation and therapeutic uses
AU2005252178B2 (en) * 2004-06-02 2011-04-21 Eli Lilly And Company Histamine H3 receptor agents, preparation and therapeutic uses
US7612062B2 (en) 2005-04-21 2009-11-03 Spirogen Limited Pyrrolobenzodiazepines
US8633185B2 (en) 2005-04-21 2014-01-21 Spirogen Sarl Pyrrolobenzodiazepines
US8163736B2 (en) 2006-01-25 2012-04-24 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives
USRE47123E1 (en) 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
US8460667B2 (en) 2006-07-18 2013-06-11 Sanofi EPHA2 receptor antagonist antibodies
EP3498735A1 (en) 2006-10-19 2019-06-19 Sanofi Novel anti-cd38 antibodies for the treatment of leukemia
EP3909980A1 (en) 2006-10-19 2021-11-17 Sanofi Novel anti-cd38 antibodies for the treatment of cancer
US8404678B2 (en) 2007-07-19 2013-03-26 Sanofi Cytotoxic agents comprising new tomaymycin derivatives and their therapeutic use
US8501934B2 (en) 2008-07-22 2013-08-06 Spirogen Sarl Pyrrolobenzodiazepines
US8940733B2 (en) 2008-10-17 2015-01-27 Spirogen Sarl Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
US9624227B2 (en) 2008-10-17 2017-04-18 Medimmune Limited Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
US8592576B2 (en) 2008-10-17 2013-11-26 Spirogen Sarl Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
US10208127B2 (en) 2009-02-05 2019-02-19 Immunogen, Inc. Benzodiazepine derivatives
US9550787B2 (en) 2009-02-05 2017-01-24 Immunogen, Inc. Benzodiazepine derivatives
AU2015224492B2 (en) * 2009-02-05 2017-04-20 Immunogen, Inc. Novel benzodiazepine derivatives
EP3360879A1 (en) * 2009-02-05 2018-08-15 ImmunoGen, Inc. Benzodiazepine derivatives as cytotoxic agents
CN108727407A (en) * 2009-02-05 2018-11-02 伊缪诺金公司 Novel benzodiazepine * derivatives
EP2393362A4 (en) * 2009-02-05 2013-07-24 Immunogen Inc Novel benzodiazepine derivatives
EP3100745A1 (en) * 2009-02-05 2016-12-07 Immunogen, Inc. Novel benzodiazepine derivatives
US9265841B2 (en) 2009-02-05 2016-02-23 Immunogen, Inc. Benzodiazepine derivatives
AU2010210646B2 (en) * 2009-02-05 2015-10-29 Immunogen, Inc. Novel benzodiazepine derivatives
EP2393362A1 (en) * 2009-02-05 2011-12-14 Immunogen, Inc. Novel benzodiazepine derivatives
US10947315B2 (en) 2009-02-05 2021-03-16 Immunogen, Inc. Benzodiazepine derivatives
US11505617B2 (en) 2009-02-05 2022-11-22 Immunogen, Inc. Benzodiazepine derivatives
AU2019202435B2 (en) * 2009-02-05 2021-04-01 Immunogen, Inc. Novel benzodiazepine derivatives
CN108727407B (en) * 2009-02-05 2022-01-28 伊缪诺金公司 Novel benzodiazepine derivatives
US8809320B2 (en) 2009-02-05 2014-08-19 Immunogen, Inc. Benzodiazepine derivatives
US8802667B2 (en) 2009-02-05 2014-08-12 Immunogen, Inc. Benzodiazepine derivatives
WO2011023883A1 (en) 2009-08-25 2011-03-03 Sanofi-Aventis Conjugates of pyrrolo[1,4]benzodiazepine dimers as anticancer agents
US8481042B2 (en) 2009-08-25 2013-07-09 Sanofi Conjugates of pyrrolo[1,4]benzodiazepine dimers as anticancer agents
US9676864B2 (en) 2009-10-02 2017-06-13 Sanofi Antibodies that specifically bind to the EphA2 receptor
US8668910B2 (en) 2009-10-02 2014-03-11 Sanofi Antibodies that specifically bind to the EphA2 receptor
WO2011039724A1 (en) 2009-10-02 2011-04-07 Sanofi-Aventis Antibodies that specifically bind to the epha2 receptor
AU2011239507B2 (en) * 2010-04-15 2015-04-09 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9732084B2 (en) 2010-04-15 2017-08-15 Medimmune Limited Pyrrolobenzodiazepines used to treat proliferative diseases
CN102933236B (en) * 2010-04-15 2014-10-08 斯皮罗根有限公司 Pyrrolobenzodiazepines and conjugates thereof
KR101738203B1 (en) 2010-04-15 2017-05-19 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
US8697688B2 (en) 2010-04-15 2014-04-15 Seattle Genetics Inc. Pyrrolobenzodiazepines used to treat proliferative diseases
WO2011128650A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Intermediates useful for the synthesis of pyrrolobenzodiazepines
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
US9242013B2 (en) 2010-04-15 2016-01-26 Seattle Genetics Inc. Targeted pyrrolobenzodiazapine conjugates
US8829184B2 (en) 2010-04-15 2014-09-09 Spirogen Sarl Intermediates useful for the synthesis of pyrrolobenzodiazepines
US10561739B2 (en) 2010-04-15 2020-02-18 Seattle Genetics Inc. Targeted pyrrolobenzodiazapine conjugates
CN102933236A (en) * 2010-04-15 2013-02-13 斯皮罗根发展有限公司 Pyrrolobenzodiazepines and conjugates thereof
EA024730B1 (en) * 2010-04-15 2016-10-31 Медимьюн Лимитед Pyrrolobenzodiazepine compounds, conjugates thereof, pharmaceutical compositions comprising said conjugates, and use of said conjugates
WO2012014147A1 (en) 2010-07-26 2012-02-02 Sanofi Anticancer derivatives, preparation thereof and therapeutic use thereof
US9056914B2 (en) 2010-07-26 2015-06-16 Sanofi Anticancer derivatives, preparation thereof and therapeutic use thereof
EP3763740A1 (en) 2011-01-26 2021-01-13 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
US10179818B2 (en) 2011-02-15 2019-01-15 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US8889669B2 (en) 2011-02-15 2014-11-18 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9868791B2 (en) 2011-02-15 2018-01-16 Immunogen, Inc. Methods of preparation of conjugates
US9840564B2 (en) 2011-02-15 2017-12-12 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US8765740B2 (en) 2011-02-15 2014-07-01 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
USRE49918E1 (en) 2011-02-15 2024-04-16 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9353127B2 (en) 2011-02-15 2016-05-31 Immunogen, Inc. Methods of preparation of conjugates
US10570212B2 (en) 2011-02-15 2020-02-25 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9434748B2 (en) 2011-02-15 2016-09-06 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9534000B2 (en) 2011-02-15 2017-01-03 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and methods of preparation
US10364294B2 (en) 2011-02-15 2019-07-30 Immunogen, Inc. Methods of preparation of conjugates
US9169272B2 (en) 2011-02-15 2015-10-27 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9707301B2 (en) 2011-10-14 2017-07-18 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US11135303B2 (en) 2011-10-14 2021-10-05 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2013053872A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Synthesis method and intermediates useful in the preparation of pyrrolobenzodiazepines
US10329352B2 (en) 2011-10-14 2019-06-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
AU2012322607B2 (en) * 2011-10-14 2017-02-16 Genentech, Inc. Pyrrolobenzodiazepines and conjugates thereof
US9713647B2 (en) 2011-10-14 2017-07-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US9102704B2 (en) 2011-10-14 2015-08-11 Spirogen Sarl Synthesis method and intermediates useful in the preparation of pyrrolobenzodiazepines
WO2013055987A1 (en) * 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
KR101877598B1 (en) * 2011-10-14 2018-07-11 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
US10328084B2 (en) 2011-10-14 2019-06-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
EA026827B1 (en) * 2011-10-14 2017-05-31 Медимьюн Лимитед Pyrrolobenzodiazepines and conjugates thereof
EP3095797A1 (en) 2012-02-24 2016-11-23 Stemcentrx, Inc. Anti dll3 antibodies and methods of use thereof
US9376440B2 (en) 2012-04-30 2016-06-28 Medimmune Limited Pyrrolobenzodiazepines as antiproliferative agents
US9321774B2 (en) 2012-04-30 2016-04-26 Medimmune Limited Pyrrolobenzodiazepines
WO2013177481A1 (en) 2012-05-25 2013-11-28 Immunogen, Inc. Benzodiazepines and conjugates thereof
EP3381943A1 (en) 2012-07-25 2018-10-03 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
EP4063391A1 (en) 2012-07-25 2022-09-28 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
WO2014018625A1 (en) 2012-07-25 2014-01-30 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
US9745303B2 (en) 2012-10-12 2017-08-29 Medimmune Limited Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
WO2014057117A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2014057122A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
US11779650B2 (en) 2012-10-12 2023-10-10 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11771775B2 (en) 2012-10-12 2023-10-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11701430B2 (en) 2012-10-12 2023-07-18 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11690918B2 (en) 2012-10-12 2023-07-04 Medimmune Limited Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US10722594B2 (en) 2012-10-12 2020-07-28 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US10780181B2 (en) 2012-10-12 2020-09-22 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2014057120A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10799596B2 (en) 2012-10-12 2020-10-13 Adc Therapeutics S.A. Pyrrolobenzodiazepine-antibody conjugates
US10994023B2 (en) 2012-10-12 2021-05-04 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2014057113A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine - anti-psma antibody conjugates
US10646584B2 (en) 2012-10-12 2020-05-12 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2014057119A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
US10576164B2 (en) 2013-03-13 2020-03-03 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9821074B2 (en) 2013-03-13 2017-11-21 Genentech, Inc. Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
US9649390B2 (en) 2013-03-13 2017-05-16 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10150813B2 (en) 2013-03-14 2018-12-11 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US11230600B2 (en) 2013-03-14 2022-01-25 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
EP3299391A1 (en) 2013-03-14 2018-03-28 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
EP3338793A1 (en) 2013-08-28 2018-06-27 AbbVie Stemcentrx LLC Novel sez6 modulators and methods of use
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9956298B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
EP3461845A1 (en) 2013-12-13 2019-04-03 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
WO2015157592A1 (en) 2014-04-11 2015-10-15 Medimmune, Llc Bispecific her2 antibodies
US10017580B2 (en) 2014-04-15 2018-07-10 ADC Therpeutics S.A. Humanized anti-Tn-MUC1 antibodies and their conjugates
WO2015179658A2 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10188746B2 (en) 2014-09-10 2019-01-29 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11286302B2 (en) 2014-09-12 2022-03-29 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040856A2 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016040868A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
US10556966B2 (en) 2014-09-12 2020-02-11 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
US10059768B2 (en) 2014-09-12 2018-08-28 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10179820B2 (en) 2014-09-12 2019-01-15 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
EP3782654A1 (en) 2014-09-12 2021-02-24 Genentech, Inc. Anti-her2 antibodies and immunoconjugates
EP3693391A1 (en) 2014-09-12 2020-08-12 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
WO2016044396A1 (en) 2014-09-17 2016-03-24 Genentech, Inc. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
WO2016054315A1 (en) 2014-10-01 2016-04-07 Medimmune, Llc Method of conjugating a polypeptide
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US10624972B2 (en) 2015-03-13 2020-04-21 Endocyte, Inc. Conjugates for treating diseases
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
WO2016166304A1 (en) 2015-04-15 2016-10-20 Van Berkel Patricius Hendrikus Cornelis Site-specific antibody-drug conjugates
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US10899775B2 (en) 2015-07-21 2021-01-26 Immunogen, Inc. Methods of preparing cytotoxic benzodiazepine derivatives
WO2017059289A1 (en) 2015-10-02 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US10632196B2 (en) 2015-10-02 2020-04-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US10639373B2 (en) 2015-10-02 2020-05-05 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US10058613B2 (en) 2015-10-02 2018-08-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
WO2017132298A1 (en) 2016-01-27 2017-08-03 Medimmune, Llc Methods for preparing antibodies with a defined glycosylation pattern
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
WO2017180813A1 (en) 2016-04-15 2017-10-19 Macrogenics, Inc. Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11591400B2 (en) 2016-04-15 2023-02-28 Macrogenics, Inc. B7-H3 directed antibody drug conjugates
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
WO2017194568A1 (en) 2016-05-11 2017-11-16 Sanofi Treatment regimen using anti-muc1 maytansinoid immunoconjugate antibody for the treatment of tumors
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2018027204A1 (en) 2016-08-05 2018-02-08 Genentech, Inc. Multivalent and multiepitopic anitibodies having agonistic activity and methods of use
US11046776B2 (en) 2016-08-05 2021-06-29 Genentech, Inc. Multivalent and multiepitopic antibodies having agonistic activity and methods of use
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
EP4015532A1 (en) 2016-11-21 2022-06-22 cureab GmbH Anti-gp73 antibodies and immunoconjugates
WO2018091724A1 (en) 2016-11-21 2018-05-24 Cureab Gmbh Anti-gp73 antibodies and immunoconjugates
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11813335B2 (en) 2017-02-08 2023-11-14 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10544223B2 (en) 2017-04-20 2020-01-28 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
EP4223319A2 (en) 2017-05-26 2023-08-09 MedImmune, LLC Method and molecules
US11938192B2 (en) 2017-06-14 2024-03-26 Medimmune Limited Dosage regimes for the administration of an anti-CD19 ADC
US11318211B2 (en) 2017-06-14 2022-05-03 Adc Therapeutics Sa Dosage regimes for the administration of an anti-CD19 ADC
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11628223B2 (en) 2017-09-29 2023-04-18 Daiichi Sankyo Company, Limited Antibody-drug conjugates comprising substituted benzo[e]pyrrolo[1,2-α][1,4]diazepines
US11583590B2 (en) 2017-09-29 2023-02-21 Daiichi Sankyo Company, Limited Antibody-pyrrolobenzodiazepine derivative conjugate and method of use thereof for treating a tumor
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
US11484606B2 (en) 2019-06-07 2022-11-01 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2020245283A1 (en) 2019-06-07 2020-12-10 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11807685B2 (en) 2021-08-05 2023-11-07 The Uab Research Foundation Anti-CD47 antibody and uses thereof
WO2023215737A1 (en) 2022-05-03 2023-11-09 Genentech, Inc. Anti-ly6e antibodies, immunoconjugates, and uses thereof

Also Published As

Publication number Publication date
EP1109811A2 (en) 2001-06-27
CA2341968A1 (en) 2000-03-09
DE69910227D1 (en) 2003-09-11
JP2002525284A (en) 2002-08-13
ATE246687T1 (en) 2003-08-15
US20030195196A1 (en) 2003-10-16
NZ510492A (en) 2003-08-29
GB9818731D0 (en) 1998-10-21
JP4669611B2 (en) 2011-04-13
US6562806B1 (en) 2003-05-13
EP1109811B1 (en) 2003-08-06
CA2341968C (en) 2009-04-07
DK1109811T3 (en) 2003-11-24
DE69910227T2 (en) 2004-06-17
PT1109811E (en) 2003-12-31
ES2205872T3 (en) 2004-05-01
WO2000012507A8 (en) 2000-10-19
WO2000012507A3 (en) 2000-08-31

Similar Documents

Publication Publication Date Title
EP1109811B1 (en) Pyrrolobenzodiazepines
US7067511B2 (en) Pyrrolobenzodiazepines
KR101671360B1 (en) Targeted pyrrolobenzodiazepine conjugates
US6130237A (en) Condensed N-aclyindoles as antitumor agents
KR101687054B1 (en) Pyrrolobenzodiazepines used to treat proliferative diseases
CA2885340C (en) Pyrrolobenzodiazepines and conjugates thereof
AU721037B2 (en) Condensed N-acylindoles as antitumor agents
US20100113425A1 (en) Pyrrolobenzodiazepines
AU758398B2 (en) Pyrrolobenzodiazepines

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

AK Designated states

Kind code of ref document: C1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

CFP Corrected version of a pamphlet front page

Free format text: REVISED TITLE RECEIVED BY THE INTERNATIONAL BUREAU AFTER COMPLETION OF THE TECHNICAL PREPARATIONS FOR INTERNATIONAL PUBLICATION

ENP Entry into the national phase

Ref document number: 2341968

Country of ref document: CA

Ref country code: CA

Ref document number: 2341968

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 09763814

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1999941766

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 510492

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 55261/99

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1999941766

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 55261/99

Country of ref document: AU

WWG Wipo information: grant in national office

Ref document number: 1999941766

Country of ref document: EP