WO2011130613A1 - Targeted pyrrolobenzodiazapine conjugates - Google Patents

Targeted pyrrolobenzodiazapine conjugates Download PDF

Info

Publication number
WO2011130613A1
WO2011130613A1 PCT/US2011/032664 US2011032664W WO2011130613A1 WO 2011130613 A1 WO2011130613 A1 WO 2011130613A1 US 2011032664 W US2011032664 W US 2011032664W WO 2011130613 A1 WO2011130613 A1 WO 2011130613A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
conjugate
attachment
mmol
unit
Prior art date
Application number
PCT/US2011/032664
Other languages
French (fr)
Inventor
Philip Wilson Howard
Scott Jeffrey
Patrick Burke
Peter Senter
Original Assignee
Seattle Genetics, Inc.
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
Priority to BR112012026801A priority Critical patent/BR112012026801B8/en
Priority to KR1020167029858A priority patent/KR101772354B1/en
Priority to MX2012011900A priority patent/MX2012011900A/en
Priority to AU2011239522A priority patent/AU2011239522B2/en
Priority to EP21213472.0A priority patent/EP4039280A1/en
Priority to US13/641,219 priority patent/US9242013B2/en
Priority to CA2795349A priority patent/CA2795349C/en
Priority to CN2011800298674A priority patent/CN103068405A/en
Priority to KR1020127028121A priority patent/KR101671360B1/en
Priority to EP11716754.4A priority patent/EP2558127B1/en
Application filed by Seattle Genetics, Inc., Spirogen Limited filed Critical Seattle Genetics, Inc.
Priority to NZ602932A priority patent/NZ602932A/en
Priority to EA201290838A priority patent/EA024118B1/en
Priority to JP2013505171A priority patent/JP5870400B2/en
Publication of WO2011130613A1 publication Critical patent/WO2011130613A1/en
Priority to ZA2012/07317A priority patent/ZA201207317B/en
Priority to IL222269A priority patent/IL222269A/en
Priority to US14/995,944 priority patent/US9592240B2/en
Priority to US15/422,000 priority patent/US20170143846A1/en
Priority to IL252864A priority patent/IL252864B/en
Priority to US15/951,753 priority patent/US20180228916A1/en
Priority to US16/381,448 priority patent/US10561739B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • C07D519/06Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00 containing at least one condensed beta-lactam ring system, provided for by groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00, e.g. a penem or a cepham system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K31/55171,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/44Antibodies bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68035Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a pyrrolobenzodiazepine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6861Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from kidney or bladder cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6867Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of a blood cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6871Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting an enzyme
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6889Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3038Kidney, bladder
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to targeted pyrrolobenzodiazepine (PBD) conjugates, in particular pyrrolobenzodiazepine dimers that are conjugated to a targeting agent via the C2 position of one of the monomers.
  • PBD targeted pyrrolobenzodiazepine
  • PBDs pyrrolobenzodiazepines
  • the biological activity of these molecules can be potentiated by joining two PBD units together through their C8/C'-hydroxyl functionalities via a flexible alkylene linker (Bose, D.S., et al., J. Am. Chem. Soc, 114, 4939-4941 (1992); Thurston, D.E., et al., J. Org.
  • the PBD dimers are thought to form sequence-selective DNA lesions such as the palindromic 5'-Pu-GATC-Py-3' interstrand cross-link (Smellie, M., et al., Biochemistry, 42, 8232-8239 (2003); Martin, C, et al., Biochemistry, 44, 4135-4147) which is thought to be mainly responsible for their biological activity.
  • sequence-selective DNA lesions such as the palindromic 5'-Pu-GATC-Py-3' interstrand cross-link (Smellie, M., et al., Biochemistry, 42, 8232-8239 (2003); Martin, C, et al., Biochemistry, 44, 4135-4147) which is thought to be mainly responsible for their biological activity.
  • a PBD dimer is SG -136):
  • PBD dimers Due to the manner in which these highly potent compounds act in cross-linking DNA, PBD dimers have been made symmetrically, i.e., both monomers of the dimer are the same. This synthetic route provides for straightforward synthesis, either by constructing the PBD dimer moiety simultaneously having already formed the dimer linkage, or by reacting already constructed PBD monomer moieties with the dimer linking group. These synthetic approaches have limited the options for preparation of targeted conjugates containing PBDs. Due to the observed potency of PBD dimers, however, there exists a need for PBD dimers that are conjugatable to targeting agents for use in targeted therapy. Disclosure of the invention
  • the present invention relates to Conjugates comprising dimers of PBDs linked to a targeting agent, wherein a PBD monomer has a substituent in the C2 position that provides an anchor for linking the compound to the targeting agent.
  • the present invention also relates to Conjugates comprising dimers of PBDs conjugated to a targeting agent, wherein the PBD monomers of the dimer are different.
  • One of PBD monomers has a substituent in the C2 position that provides an anchor for linking the compound to the targeting agent.
  • the Conjugates described herein have potent cytotoxic and/or cytostatic activity against cells expressing a target molecule, such as cancer cells or immune cells. These conjugates exhibit good potency with reduced toxicity, as compared with the corresponding PBD dimer free drug compounds.
  • the Conjugates have the following formula I:
  • L is a Ligand unit (i.e., a targeting agent)
  • LU is a Linker unit
  • D is a Drug unit comprising a PBD dimer.
  • the subscript p is an integer of from 1 to 20.
  • the Conjugates comprise a Ligand unit covalently linked to at least one Drug unit by a Linker unit.
  • the Ligand unit described more fully below, is a targeting agent that binds to a target moiety.
  • the Ligand unit can, for example, specifically bind to a cell component (a Cell Binding Agent) or to other target molecules of interest. Accordingly, the present invention also provides methods for the treatment of, for example, various cancers and autoimmune disease.
  • the Ligand unit is a targeting agent that specifically binds to a target molecule.
  • the Ligand unit can be, for example, a protein, polypeptide or peptide, such as an antibody, an antigen-binding fragment of an antibody, or other binding agent, such as an Fc fusion protein.
  • the Conjugates comprise a Conjugate of formula I (supra), wherein the Drug unit comprises a PBD dimer of the following formula II:
  • R 2 is of formula III:
  • A is a C 5 - 7 aryl group
  • X is an activatable group for conjugation to the Linker unit, wherein X is selected from the group comprising: -0-, -S-, -C(0)0-, -C(O)-, -NHC(O)-, and -N(R N )-, wherein R N is selected from the group comprising H, Ci -4 alkyl and (C 2 H 4 0) m CH 3 , where m is 1 to 3, and either:
  • Q 1 is a single bond
  • Q 2 is selected from a single bond and -Z-(CH 2 )n-, where
  • Z is selected from a single bond, O, S and NH and n is from 1 to 3;
  • R 12 is a C 5- io aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, C 1-7 alkyl, C 3-7 heterocyclyl and bis-oxy-C 1-3 alkylene;
  • R 6 and R 9 are independently selected from H, R, OH, OR, SH, SR, NH 2 , NHR, NRR', nitro, Me 3 Sn and halo;
  • R and R' are independently selected from optionally substituted C1-12 alkyl, optionally substituted C 3-20 heterocyclyl and optionally substituted C 5 - 20 aryl groups;
  • R 7 is selected from H, R, OH, OR, SH, SR, NH 2 , NHR, NHRR', nitro, Me 3 Sn and halo; either:
  • R 10 is H, and R 11 is OH or OR A , where R A is C1-4 alkyl;
  • R 10 and R 11 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound;
  • R 10 is H and R 11 is SO z M, where z is 2 or 3 and M is a monovalent
  • R" is a C 3- i 2 alkylene group, which chain may be interrupted by one or more heteroatoms, e.g. O, S, NR N2 (where R N2 is H or Ci -4 alkyl), and/or aromatic rings, e.g. benzene or pyridine;
  • Y and Y' are selected from O, S, or NH;
  • R 6 , R 7 , R 9 are selected from the same groups as R 6 , R 7 and R 9 respectively and R 10 and R 11 are the same as R 10 and R 11 , wherein if R 11 and R 11 are SO z M, M may represent a divalent pharmaceutically acceptable cation.
  • the use of the Conjugate of formula I is provided for the manufacture of a medicament for treating a proliferative disease or autoimmune disease.
  • the use of the Conjugate of formula I is provided for the treatment of a proliferative disease or an autoimmune disease.
  • a Conjugate of formula I to provide a PBD dimer, or a salt or solvate thereof, at a target location.
  • proliferative disease pertains to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo.
  • proliferative conditions include, but are not limited to, benign, pre-malignant, and malignant cellular proliferation, including but not limited to, neoplasms and tumours (e.g., histocytoma, glioma, astrocyoma, osteoma), cancers (e.g. lung cancer, small cell lung cancer, gastrointestinal cancer, bowel cancer, colon cancer, breast carinoma, ovarian carcinoma, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, pancreatic cancer, brain cancer, sarcoma, osteosarcoma, Kaposi's sarcoma, melanoma), leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g.
  • cancers of interest include, but are not limited to, haematological; malignancies such as leukemias and lymphomas, such as non-Hodgkin lymphoma, and subtypes such as DLBCL, marginal zone, mantle zone, and follicular, Hodgkin lymphoma, AML, and other cancers of B or T cell origin.
  • autoimmune disease examples include the following: rheumatoid arthritis, autoimmune demyelinative diseases (e.g., multiple sclerosis, allergic encephalomyelitis), psoriatic arthritis, endocrine ophthalmopathy, uveoretinitis, systemic lupus erythematosus, myasthenia gravis, Graves' disease, glomerulonephritis, autoimmune hepatological disorder, inflammatory bowel disease (e.g., Crohn's disease), anaphylaxis, allergic reaction, Sjogren's syndrome, type I diabetes mellitus, primary biliary cirrhosis, Wegener's granulomatosis, fibromyalgia, polymyositis, dermatomyositis, multiple endocrine failure, Schmidt's syndrome, autoimmune uveitis, Addison's disease, adrenalitis, thyroiditis, Hashimoto's thyroiditis, autoimmune thyroid disease,
  • erythematosus hypoparathyroidism, Dressler's syndrome, autoimmune thrombocytopenia, idiopathic thrombocytopenic purpura, hemolytic anemia, pemphigus vulgaris, pemphigus, dermatitis herpetiformis, alopecia areata, pemphigoid, scleroderma, progressive systemic sclerosis, CREST syndrome (calcinosis, Raynaud's phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), male and female autoimmune infertility, ankylosing spondolytis, ulcerative colitis, mixed connective tissue disease, polyarteritis nedosa, systemic necrotizing vasculitis, atopic dermatitis, atopic rhinitis, Goodpasture's syndrome, Chagas' disease, sarcoidosis, rheumatic fever, asthma, recurrent abortion, anti
  • granulomatosis granulomatosis, Behcet's disease, Caplan's syndrome, Kawasaki's disease, dengue, encephalomyelitis, endocarditis, endomyocardial fibrosis, endophthalmitis, erythema elevatum et diutinum, psoriasis, erythroblastosis fetalis, eosinophilic faciitis, Shulman's syndrome, Felty's syndrome, filariasis, cyclitis, chronic cyclitis, heterochronic cyclitis, Fuch's cyclitis, IgA nephropathy, Henoch-Schonlein purpura, graft versus host disease, transplantation rejection, cardiomyopathy, Eaton-Lambert syndrome, relapsing
  • the autoimmune disease is a disorder of B lymphocytes (e.g., systemic lupus erythematosus, Goodpasture's syndrome, rheumatoid arthritis, and type I diabetes), Th1 -lymphocytes (e.g., rheumatoid arthritis, multiple sclerosis, psoriasis,
  • B lymphocytes e.g., systemic lupus erythematosus, Goodpasture's syndrome, rheumatoid arthritis, and type I diabetes
  • Th1 -lymphocytes e.g., rheumatoid arthritis, multiple sclerosis, psoriasis
  • Th2-lymphocyt.es e.g., atopic dermatitis, systemic lupus erythematosus, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn's syndrome, systemic sclerosis, or chronic graft versus host disease.
  • disorders involving dendritic cells involve disorders of Th1 - lymphocytes or Th2-lymphocytes.
  • the autoimmune disorder is a T cell-mediated immunological disorder.
  • a fourth aspect of the present invention comprises a method of making the Conjugates formula I.
  • the dimeric PBD compounds for use in the present invention are made by different strategies to those previously employed in making symmetrical dimeric PBD compounds.
  • the present inventors have developed a method which involves adding each C2 aryl substituent to a symmetrical PBD dimer core in separate method steps.
  • a sixth aspect of the present invention provides a method of making a
  • Figures 1 to 6 show the effect of conjugates of the present invention in tumours.
  • trade name when a trade name is used herein, reference to the trade name also refers to the product formulation, the generic drug, and the active pharmaceutical ingredient(s) of the trade name product, unless otherwise indicated by context.
  • binding agent and "targeting agent as used herein refer to a molecule, e.g., protein, polypeptide or peptide, that specifically binds to a target molecule.
  • a target molecule e.g., protein, polypeptide or peptide
  • binding agent can include a full length antibody, an antigen binding fragment of a full length antibody, other agent (protein, polypeptide or peptide) that includes an antibody heavy and/or light chain variable region that specifically bind to the target molecule, or an Fc fusion protein comprising an extracellular domain of a protein, peptide polypeptide that binds to the target molecule and that is joined to an Fc region, domain or portion thereof, of an antibody.
  • telomere binding refers to the binding of an antibody or other protein, polypeptide or peptide to a predetermined molecule (e.g., an antigen).
  • the antibody or other molecule binds with an affinity of at least about 1x10 7 M "1 , and binds to the predetermined molecule with an affinity that is at least two-fold greater than its affinity for binding to a non-specific molecule (e.g., BSA, casein) other than the predetermined molecule or a closely-related molecule.
  • a non-specific molecule e.g., BSA, casein
  • Examples of pharmaceutically acceptable monovalent inorganic cations include, but are not limited to, alkali metal ions such as Na + and K + .
  • Examples of pharmaceutically acceptable divalent inorganic cations include, but are not limited to, alkaline earth cations such as Ca 2+ and Mg 2+ .
  • Examples of pharmaceutically acceptable organic cations include, but are not limited to, ammonium ion (i.e. NH 4 + ) and substituted ammonium ions (e.g. NH 3 R + , NH 2 R2 + , NHR 3 + , NR 4 + ).
  • substituted ammonium ions examples include those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine.
  • An example of a common quaternary ammonium ion is N(CH 3 ) 4 + .
  • substituted refers to a parent group which bears one or more substituents.
  • substituted is used herein in the conventional sense and refers to a chemical moiety which is covalently attached to, or if appropriate, fused to, a parent group.
  • substituents are well known, and methods for their formation and introduction into a variety of parent groups are also well known.
  • C-i-12 alkyl refers to a monovalent moiety obtained by removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to 12 carbon atoms, which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated).
  • alkyl includes the sub-classes alkenyl, alkynyl, cycloalkyl, etc., discussed below.
  • saturated alkyl groups include, but are not limited to, methyl (Ci), ethyl (C 2 ), propyl (C 3 ), butyl (C 4 ), pentyl (C 5 ), hexyl (C 6 ) and heptyl (C 7 ).
  • saturated linear alkyl groups include, but are not limited to, methyl (Ci), ethyl (C 2 ), n-propyl (C 3 ), n-butyl (C 4 ), n-pentyl (amyl) (C 5 ), n-hexyl (C 6 ) and n-heptyl (C 7 ).
  • saturated branched alkyl groups include iso-propyl (C 3 ), iso-butyl (C 4 ), sec-butyl (C 4 ), tert-butyl (C 4 ), iso-pentyl (C 5 ), and neo-pentyl (C 5 ).
  • C2-12 Alkenyl The term "C2-12 alkenyl" as used herein, pertains to an alkyl group having one or more carbon-carbon double bonds.
  • C2-12 alkynyl refers to an alkyl group having one or more carbon-carbon triple bonds.
  • unsaturated alkynyl groups include, but are not limited to, ethynyl (-C ⁇ CH) and 2-propynyl (propargyl, -CH 2 -C ⁇ CH).
  • C 3- 12 cycloalkyl The term "C 3- i 2 cycloalkyl" as used herein, pertains to an alkyl group which is also a cyclyl group; that is, a monovalent moiety obtained by removing a hydrogen atom from an alicyclic ring atom of a cyclic hydrocarbon (carbocyclic) compound, which moiety has from 3 to 7 carbon atoms, including from 3 to 7 ring atoms.
  • cycloalkyl groups include, but are not limited to, those derived from:
  • unsaturated monocyclic hydrocarbon compounds cyclopropene (C 3 ), cyclobutene (C 4 ), cyclopentene (C 5 ), cyclohexene (C 6 ), methylcyclopropene (C 4 ), dimethylcyclopropene (C 5 ), methylcyclobutene (C 5 ), dimethylcyclobutene (C 6 ), methylcyclopentene (C 6 ), dimethylcyclopentene (C 7 ) and methylcyclohexene (C 7 ); and
  • C 3- 2o heterocyclyl refers to a monovalent moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound, which moiety has from 3 to 20 ring atoms, of which from 1 to 10 are ring heteroatoms.
  • each ring has from 3 to 7 ring atoms, of which from 1 to 4 are ring heteroatoms.
  • the prefixes e.g. C3-20, C 3-7 , C 5 - 6 , etc.
  • the prefixes denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms.
  • C 5-6 heterocyclyl as used herein, pertains to a heterocyclyl group having 5 or 6 ring atoms.
  • monocyclic heterocyclyl groups include, but are not limited to, those derived from:
  • N-i aziridine (C 3 ), azetidine (C 4 ), pyrrolidine (tetrahydropyrrole) (C 5 ), pyrroline (e.g., 3-pyrroline, 2,5-dihydropyrrole) (C 5 ), 2H-pyrrole or 3H-pyrrole (isopyrrole, isoazole) (C 5 ), piperidine (C 6 ), dihydropyridine (C 6 ), tetrahydropyridine (C 6 ), azepine (C 7 );
  • N 2 imidazolidine (C 5 ), pyrazolidine (diazolidine) (C 5 ), imidazoline (C 5 ), pyrazoline
  • N1O1 tetrahydrooxazole (C 5 ), dihydrooxazole (C 5 ), tetrahydroisoxazole (C 5 ),
  • dihydroisoxazole C 5
  • morpholine C 6
  • tetrahydrooxazine C 6
  • dihydrooxazine C 6
  • oxazine C 6
  • O1S1 oxathiole (C 5 ) and oxathiane (thioxane) (C 6 ); and,
  • OiSi : oxathiazine C 6
  • substituted monocyclic heterocyclyl groups include those derived from saccharides, in cyclic form, for example, furanoses (C 5 ), such as arabinofuranose, lyxofuranose, ribofuranose, and xylofuranse, and pyranoses (C 6 ), such as allopyranose, altropyranose, glucopyranose, mannopyranose, gulopyranose, idopyranose,
  • C5-2 0 aryl refers to a monovalent moiety obtained by removing a hydrogen atom from an aromatic ring atom of an aromatic compound, which moiety has from 3 to 20 ring atoms. Preferably, each ring has from 5 to 7 ring atoms.
  • the prefixes e.g. C3-2 0 , C5-7, C 5 - 6 , etc.
  • C 5-6 aryl as used herein, pertains to an aryl group having 5 or 6 ring atoms.
  • the ring atoms may be all carbon atoms, as in "carboaryl groups".
  • carboaryl groups include, but are not limited to, those derived from benzene (i.e. phenyl) (C 6 ), naphthalene (C1 0 ), azulene (C1 0 ), anthracene (C14), phenanthrene (C14), naphthacene (Ci 8 ), and pyrene (Ci 6 ).
  • aryl groups which comprise fused rings include, but are not limited to, groups derived from indane (e.g. 2,3-dihydro-1 H- indene) (Cg), indene (Cg), isoindene (Cg), tetraline (1 ,2,3,4-tetrahydronaphthalene (C 10 ), acenaphthene (C 12 ), fluorene (C 13 ), phenalene (C 13 ), acephenanthrene (C 15 ), and aceanthrene (C 16 ).
  • the ring atoms may include one or more heteroatoms, as in "heteroaryl groups".
  • monocyclic heteroaryl groups include, but are not limited to, those derived from:
  • Si thiophene (thiole) (C 5 ); N1O1 : oxazole (C 5 ), isoxazole (C 5 ), isoxazine (C 6 );
  • N 2 imidazole (1 ,3-diazole) (C 5 ), pyrazole (1 ,2-diazole) (C 5 ), pyridazine (1 ,2-diazine) (C 6 ), pyrimidine (1 ,3-diazine) (C 6 ) (e.g., cytosine, thymine, uracil), pyrazine (1 ,4-diazine) (C 6 ); N 3 : triazole (C 5 ), triazine (C 6 ); and,
  • heteroaryl which comprise fused rings, include, but are not limited to:
  • Cio (with 2 fused rings) derived from chromene (O1 ), isochromene (O1), chroman (O1), isochroman (O1), benzodioxan (0 2 ), quinoline (N-i ), isoquinoline (N-i), quinolizine (N-i), benzoxazine (N- ⁇ - ⁇ ), benzodiazine (N 2 ), pyridopyridine (N 2 ), quinoxaline (N 2 ), quinazoline (N 2 ), cinnoline (N 2 ), phthalazine (N 2 ), naphthyridine (N 2 ), pteridine (N 4 );
  • Ci 3 (with 3 fused rings) derived from carbazole (N-i), dibenzofuran (O1 ),
  • Ci 4 (with 3 fused rings) derived from acridine (N-i), xanthene (O1), thioxanthene (Si), oxanthrene (0 2 ), phenoxathiin (O1S1 ), phenazine (N 2 ), phenoxazine (N- ⁇ - ⁇ ), phenothiazine (N-I S-I), thianthrene (S 2 ), phenanthridine (N-i ), phenanthroline (N 2 ), phenazine (N 2 ).
  • Halo -F, -CI, -Br, and -I.
  • R is an ether substituent, for example, a Ci_ 7 alkyl group (also referred to as a Ci -7 alkoxy group, discussed below), a C3-20 heterocyclyl group (also referred to as a C3-20 heterocyclyloxy group), or a C 5 -2o aryl group (also referred to as a C 5 -2o aryloxy group), preferably a Ci -7 alkyl group.
  • a Ci_ 7 alkyl group also referred to as a Ci -7 alkoxy group, discussed below
  • C3-20 heterocyclyl group also referred to as a C3-20 heterocyclyloxy group
  • C 5 -2o aryl group also referred to as a C 5 -2o aryloxy group
  • Ci -7 alkoxy groups include, but are not limited to, -OMe (methoxy), -OEt (ethoxy), -O(nPr) (n- propoxy), -O(iPr) (isopropoxy), -O(nBu) (n-butoxy), -O(sBu) (sec-butoxy), -O(iBu)
  • Acetal -CH(OR 1 )(OR 2 ), wherein R 1 and R 2 are independently acetal substituents, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a C1-7 alkyl group, or, in the case of a "cyclic" acetal group, R 1 and R 2 , taken together with the two oxygen atoms to which they are attached, and the carbon atoms to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms.
  • acetal groups include, but are not limited to, -CH(OMe) 2 , -CH(OEt) 2 , and -CH(OMe)(OEt).
  • Hemiacetal -CH(OH)(OR 1 ), wherein R 1 is a hemiacetal substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a Ci -7 alkyl group.
  • R 1 is a hemiacetal substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a Ci -7 alkyl group.
  • hemiacetal groups include, but are not limited to, -CH(OH)(OMe) and - CH(OH)(OEt).
  • Ketal -CR(OR 1 )(OR 2 ), where R 1 and R 2 are as defined for acetals, and R is a ketal substituent other than hydrogen, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5-2 o aryl group, preferably a C 1-7 alkyl group.
  • ketal groups include, but are not limited to, -C(Me)(OMe) 2 , -C(Me)(OEt) 2 , -C(Me)(OMe)(OEt), -C(Et)(OMe) 2 , - C(Et)(OEt) 2 , and -C(Et)(OMe)(OEt).
  • R 1 is as defined for hemiacetals, and R is a hemiketal substituent other than hydrogen, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci -7 alkyl group.
  • hemiacetal groups include, but are not limited to, -C(Me)(OH)(OMe), -C(Et)(OH)(OMe), -C(Me)(OH)(OEt), and -C(Et)(OH)(OEt).
  • Imino (imine): NR, wherein R is an imino substituent, for example, hydrogen, Ci -7 alkyl group, a C3-2 0 heterocyclyl group, or a C5-2 0 aryl group, preferably hydrogen or a Ci -7 alkyl group.
  • R is an acyl substituent, for example, a C 1-7 alkyl group (also referred to as Ci -7 alkylacyl or Ci -7 alkanoyl), a C3-2 0 heterocyclyl group (also referred to as C3-2 0 heterocyclylacyl), or a C 5 -2o aryl group (also referred to as C5-2 0 arylacyl), preferably a Ci -7 alkyl group.
  • a C 1-7 alkyl group also referred to as Ci -7 alkylacyl or Ci -7 alkanoyl
  • C3-2 0 heterocyclyl group also referred to as C3-2 0 heterocyclylacyl
  • C 5 -2o aryl group also referred to as C5-2 0 arylacyl
  • Carboxy (carboxylic acid): -C( 0)OH.
  • Thiocarboxy (thiocarboxylic acid): -C( S)SH.
  • Thiolocarboxy (thiolocarboxylic acid): -C( 0)SH.
  • Thionocarboxy (thionocarboxylic acid): -C( S)OH.
  • Imidic acid: -C( NH)OH.
  • Hydroxamic acid: -C( NOH)OH.
  • Acyloxy (reverse ester): -OC( 0)R, wherein R is an acyloxy substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a Ci -7 alkyl group.
  • R is an acyloxy substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a Ci -7 alkyl group.
  • Oxycarboyloxy: -OC( 0)OR, wherein R is an ester substituent, for example, a Ci -7 alkyl group, a C 3-2 o heterocyclyl group, or a C 5-2 o aryl group, preferably a C 1 -7 alkyl group.
  • R 1 and R 2 are independently amino substituents, for example, hydrogen, a Ci -7 alkyl group (also referred to as Ci -7 alkylamino or di-Ci -7 alkylamino), a C 3-20 heterocyclyl group, or a C 5 - 20 aryl group, preferably H or a Ci -7 alkyl group, or, in the case of a "cyclic" amino group, R 1 and R 2 , taken together with the nitrogen atom to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms.
  • a Ci -7 alkyl group also referred to as Ci -7 alkylamino or di-Ci -7 alkylamino
  • C 3-20 heterocyclyl group or a C 5 - 20 aryl group, preferably H or a Ci -7 alkyl group
  • R 1 and R 2 taken together with the nitrogen atom to which they are attached, form a heterocyclic ring having from 4 to 8 ring atom
  • Amino groups may be primary (-NH 2 ), secondary (-NHR 1 ), or tertiary (-NHR 1 R 2 ), and in cationic form, may be quaternary (- + NR 1 R 2 R 3 ).
  • Examples of amino groups include, but are not limited to, -NH 2 , -N HCH 3 , -NHC(CH 3 ) 2 , -N(CH 3 ) 2 , -N(CH 2 CH 3 ) 2 , and -NHPh.
  • Examples of cyclic amino groups include, but are not limited to, aziridino, azetidino, pyrrolidino, piperidino, piperazino, morpholino, and thiomorpholino.
  • Acylamido (acylamino): -NR 1 C( 0)R 2 , wherein R 1 is an amide substituent, for example, hydrogen, a Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 2 o aryl group, preferably hydrogen or a Ci_ 7 alkyl group, and R 2 is an acyl substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2oaryl group, preferably hydrogen or a C1-7 alkyl group.
  • R 1 and R 2 may together form a cyclic structure, as in for example, succinimidyl, maleimidyl, and phthalimidyl:
  • Ureido -N(R 1 )CONR 2 R 3 wherein R 2 and R 3 are independently amino substituents, as defined for amino groups, and R 1 is a ureido substituent, for example, hydrogen, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-20 aryl group, preferably hydrogen or a C 1-7 alkyl group.
  • R 1 is a ureido substituent, for example, hydrogen, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-20 aryl group, preferably hydrogen or a C 1-7 alkyl group.
  • ureido groups include, but are not limited to, -NHCONH 2 , -
  • Tetrazolyl a five membered aromatic ring having four nitrogen atoms and one carbon atom
  • Imino: NR, wherein R is an imino substituent, for example, for example, hydrogen, a Ci -7 alkyl group, a C 3 - 20 heterocyclyl group, or a C 5 - 2 o aryl group, preferably H or a Ci -7 alkyl group.
  • amidine groups include, but are not limited to,
  • C 1-7 alkylthio groups include, but are not limited to, -SCH 3 and -SCH 2 CH 3 .
  • Disulfide -SS-R, wherein R is a disulfide substituent, for example, a Ci -7 alkyl group, a C 3- 20 heterocyclyl group, or a C 5 - 20 aryl group, preferably a Ci -7 alkyl group (also referred to herein as Ci -7 alkyl disulfide).
  • R is a disulfide substituent, for example, a Ci -7 alkyl group, a C 3- 20 heterocyclyl group, or a C 5 - 20 aryl group, preferably a Ci -7 alkyl group (also referred to herein as Ci -7 alkyl disulfide).
  • Ci -7 alkyl disulfide groups include, but are not limited to, -SSCH 3 and -SSCH 2 CH 3 .
  • Sulfine (sulfinyl, sulfoxide): -S( 0)R, wherein R is a sulfine substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a Ci -7 alkyl group.
  • R is a sulfine substituent, for example, a Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably a Ci -7 alkyl group.
  • R is a sulfinate substituent, for example, a Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 20 aryl group, preferably a Ci -7 alkyl group.
  • Sulfonate (sulfonic acid ester): -S( 0) 2 OR, wherein R is a sulfonate substituent, for example, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-20 aryl group, preferably a
  • R is a sulfonyloxy substituent, for example, a Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 20 aryl group, preferably a Ci -7 alkyl group.
  • R is a sulfate substituent, for example, a Ci -7 alkyl group, a C 3 - 20 heterocyclyl group, or a C 5 - 20 aryl group, preferably a Ci -7 alkyl group.
  • R 1 and R 2 are independently amino substituents, as defined for amino groups.
  • R 1 and R 2 are independently amino substituents, as defined for amino groups.
  • R 1 is an amino substituent, as defined for amino groups.
  • R 1 is an amino substituent, as defined for amino groups
  • R is a sulfonamino substituent, for example, a Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 2 o aryl group, preferably a Ci -7 alkyl group.
  • R 1 is an amino substituent, as defined for amino groups
  • R is a sulfinamino substituent, for example, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-20 aryl group, preferably a C 1-7 alkyl group.
  • phosphino groups include, but are not limited to, -PH 2 , -P(CH 3 ) 2 , -P(CH 2 CH 3 ) 2 , -P(t-Bu) 2 , and -P(Ph) 2 .
  • R is a phosphinyl substituent, for example, a Ci -7 alkyl group, a C 3 - 2 o heterocyclyl group, or a C 5 - 2 o aryl group, preferably a Ci -7 alkyl group or a C 5 - 20 aryl group.
  • R is a phosphonate substituent, for example, -H, a Ci -7 alkyl group, a C 3 - 20 heterocyclyl group, or a C 5 - 2 o aryl group, preferably -H, a Ci -7 alkyl group, or a C 5 - 20 aryl group.
  • Phosphate (phosphonooxy ester): -OP( 0)(OR) 2 , where R is a phosphate substituent, for example, -H, a Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 2 o aryl group, preferably - H, a Ci_7 alkyl group, or a C 5 - 20 aryl group.
  • R is a phosphate substituent, for example, -H, a Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 2 o aryl group, preferably - H, a Ci_7 alkyl group, or a C 5 - 20 aryl group.
  • Phosphorous acid -OP(OH) 2 .
  • Phosphite -OP(OR) 2 , where R is a phosphite substituent, for example, -H, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-20 aryl group, preferably -H, a C 1-7 alkyl group, or a C 5-20 aryl group.
  • phosphite groups include, but are not limited to, -OP(OCH 3 ) 2 , -OP(OCH 2 CH 3 ) 2 , -OP(0-t-Bu) 2 , and -OP(OPh) 2 .
  • Phosphoramidite -OP(OR 1 )-NR 2 2 , where R 1 and R 2 are phosphoramidite substituents, for example, -H, a (optionally substituted) Ci -7 alkyl group, a C 3-20 heterocyclyl group, or a C 5 - 20 aryl group, preferably -H, a Ci -7 alkyl group, or a C 5 - 2 o aryl group. Examples of
  • phosphoramidite groups include, but are not limited to, -OP(OCH 2 CH 3 )-N(CH 3 ) 2 ,
  • substituents for example, -H, a (optionally substituted) Ci -7 alkyl group, a C3-20 heterocyclyl group, or a C 5 -2o aryl group, preferably -H, a Ci -7 alkyl group, or a C 5 -2o aryl group.
  • C 3- 12 alkylene refers to a bidentate moiety obtained by removing two hydrogen atoms, either both from the same carbon atom, or one from each of two different carbon atoms, of a hydrocarbon compound having from 3 to 12 carbon atoms (unless otherwise specified), which may be aliphatic or alicyclic, and which may be saturated, partially unsaturated, or fully unsaturated.
  • alkylene includes the sub-classes alkenylene, alkynylene, cycloalkylene, etc., discussed below.
  • linear saturated C 3- i 2 alkylene groups include, but are not limited to, -(CH 2 ) n - where n is an integer from 3 to 12, for example, -CH 2 CH 2 CH 2 - (propylene),
  • Examples of branched saturated C 3- i 2 alkylene groups include, but are not limited to, -CH(CH 3 )CH 2 -, -CH(CH 3 )CH 2 CH 2 -, -CH(CH 3 )CH 2 CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -,
  • Examples of alicyclic saturated C 3- i2 alkylene groups include, but are not limited to, cyclopentylene (e.g. cyclopent-1 ,3-ylene), and cyclohexylene
  • C3-12 cycloalkylenes examples include, but are not limited to, cyclopentenylene (e.g. 4-cyclopenten-1 ,3-ylene), cyclohexenylene (e.g. 2-cyclohexen-1 ,4-ylene; 3-cyclohexen-1 ,2-ylene; 2,5-cyclohexadien- 1 ,4-ylene).
  • cyclopentenylene e.g. 4-cyclopenten-1 ,3-ylene
  • cyclohexenylene e.g. 2-cyclohexen-1 ,4-ylene; 3-cyclohexen-1 ,2-ylene; 2,5-cyclohexadien- 1 ,4-ylene.
  • Oxygen protecting group refers to a moiety which masks a hydroxy group, and these are well known in the art. A large number of suitable groups are described on pages 23 to 200 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference. Classes of particular interest include silyl ethers (e.g. TMS, TBDMS), substituted methyl ethers (e.g. THP) and esters (e.g. acetate).
  • silyl ethers e.g. TMS, TBDMS
  • substituted methyl ethers e.g. THP
  • esters e.g. acetate
  • Carbamate nitrogen protecting group pertains to a moiety which masks the nitrogen in the imine bond, and these are well known in the art. These groups have the following structure:
  • R' 10 is R as defined above.
  • R' 10 is R as defined above.
  • suitable groups are described on pages 503 to 549 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic
  • Hemi-aminal nitrogen protecting group pertains to a group having the following wherein R' 10 is R as defined above.
  • R' 10 is R as defined above.
  • suitable groups are described on pages 633 to 647 as amide protecting groups of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.
  • amide protecting groups of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.
  • the present invention provides Conjugates comprising a PBD dimer connected to a Ligand unit via a Linker Unit.
  • the Linker unit includes a Stretcher unit (A), a Specificity unit (L 1 ), and a Spacer unit (L 2 ).
  • the Linker unit is connected at one end to the Ligand unit and at the other end to the PBD dimer compound.
  • L is the Ligand unit
  • a 1 or 2
  • L 1 - is a Specificity unit
  • s is an integer ranging from 1 to 12,
  • y 0, 1 or 2;
  • -D is an PBD dimer
  • p is from 1 to 20.
  • the drug loading is represented by p, the number of drug molecules per Ligand unit (e.g., an antibody). Drug loading may range from 1 to 20 Drug units (D) per Ligand unit (e.g., Ab or mAb).
  • D Drug units
  • p represents the average drug loading of the Conjugates in the composition, and p ranges from 1 to 20.
  • p is from about 1 to about 8 Drug units per Ligand unit. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is from about 2 to about 8 Drug units per Ligand unit. In some embodiments, p is from about 2 to about 6, 2 to about 5, or 2 to about 4 Drug units per Ligand unit. In some embodiments, p is about 2, about 4, about 6 or about 8 Drug units per Ligand unit.
  • the average number of Drugs units per Ligand unit in a preparation from a conjugation reaction may be characterized by conventional means such as mass spectroscopy, ELISA assay, and HPLC.
  • the quantitative distribution of Conjugates in terms of p may also be determined.
  • separation, purification, and characterization of homogeneous Conjugates, where p is a certain value, from Conjugates with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis.
  • L is the Ligand unit
  • -A 1 - is a Stretcher unit linked to a Stretcher unit (L 2 ),
  • a 1 or 2
  • L 1 - is a Specificity unit linked to a Stretcher unit (L 2 ),
  • s is an integer ranging from 0 to 12,
  • y 0, 1 or 2;
  • -D is a PBD dimer
  • p is from 1 to 20.
  • the Conjugate has the formula: wherein L, A 1 , a, L 1 , s, L 2 , D and p are as described above.
  • the Ligand unit (L) is a Cell Binding Agent (CBA) that specifically binds to a target molecule on the surface of a target cell.
  • CBA Cell Binding Agent
  • CBA is the
  • L 1 is a Specificity unit
  • a 1 is a Stretcher unit connecting L 1 to the Cell Binding Agent
  • L 2 optional.
  • the Ligand unit (L) is a Cell Binding Agent (CBA) that specifically binds to a target molecule on the surface of a target cell.
  • CBA Cell Binding Agent
  • CBA is the Cell Binding Agent
  • L 1 is a Specificity unit
  • a 1 is a Stretcher unit connecting L 1 to the Cell Binding Agent
  • L 2 is a Spacer unit which is a covalent bond or a self-immolative group
  • a is 1 or 2
  • s is 0, 1 or 2
  • y is 0 or 1 or 2.
  • L 1 can be a cleavable Specificity unit, and may be referred to as a "trigger" that when cleaved activates a self-immolative group (or self- immolative groups) L 2 , when a self-immolative group(s) is present.
  • the Specificity unit L 1 is cleaved, or the linkage (i.e., the covalent bond) between L 1 and L 2 is cleaved, the self-immolative group releases the Drug unit (D).
  • the Ligand unit (L) is a Cell Binding Agent (CBA) that specifically binds to a target molecule on the surface of a target cell.
  • CBA Cell Binding Agent
  • An exemplary formula is illustrated below: where the asterisk indicates the point of attachment to the Drug (D), CBA is the Cell Binding Agent, L 1 is a Specificity unit connected to L 2 , A 1 is a Stretcher unit connecting L 2 to the Cell Binding Agent, L 2 is a self-immolative group, and a is 1 or2, s is 1 or 2, and y is 1 or 2.
  • L 1 and L 2 can vary widely. These groups are chosen on the basis of their characteristics, which may be dictated in part, by the conditions at the site to which the conjugate is delivered.
  • the Specificity unit L 1 is cleavable, the structure and/or sequence of L 1 is selected such that it is cleaved by the action of enzymes present at the target site (e.g., the target cell).
  • L 1 units that are cleavable by changes in pH (e.g. acid or base labile), temperature or upon irradiation (e.g. photolabile) may also be used.
  • L 1 units that are cleavable under reducing or oxidising conditions may also find use in the Conjugates.
  • L 1 may comprise one amino acid or a contiguous sequence of amino acids.
  • the amino acid sequence may be the target substrate for an enzyme.
  • L 1 is cleavable by the action of an enzyme.
  • the enzyme is an esterase or a peptidase.
  • L 1 may be cleaved by a lysosomal protease, such as a cathepsin.
  • the enzyme cleaves the bond between L 1 and L 2 , whereby the self-immolative group(s) release the Drug unit.
  • L 1 and L 2 where present, may be connected by a bond selected from:
  • An amino group of L 1 that connects to L 2 may be the N-terminus of an amino acid or may be derived from an amino group of an amino acid side chain, for example a lysine amino acid side chain.
  • a carboxyl group of L 1 that connects to L 2 may be the C-terminus of an amino acid or may be derived from a carboxyl group of an amino acid side chain, for example a glutamic acid amino acid side chain.
  • a hydroxy group of L 1 that connects to L 2 may be derived from a hydroxy group of an amino acid side chain, for example a serine amino acid side chain.
  • n is 0 to 3.
  • the phenylene ring is optionally substituted with one, two or three substituents as described herein.
  • Y is NH
  • n is 0 or 1 .
  • n is 0.
  • the self-immolative group may be referred to as a
  • PABC p-aminobenzylcarbonyl linker
  • the Drug unit i.e., the asymmetric PBD
  • L * is the activated form of the remaining portion of the linker and the released Drug unit is not shown.
  • Each phenylene ring is optionally substituted with one, two or three substituents as described herein. In one embodiment, the phenylene ring having the Y substituent is optionally substituted and the phenylene ring not having the Y substituent is unsubstituted.
  • -C 0)0- and L 2 together form a group selected from:
  • E is O, S or NR
  • D is N, CH, or CR
  • F is N, CH, or CR.
  • D is N.
  • D is CH.
  • E is O or S.
  • F is CH.
  • the covalent bond between L 1 and L 2 is a cathepsin labile (e.g., cleavable) bond.
  • L 1 comprises a dipeptide.
  • the amino acids in the dipeptide may be any combination of natural amino acids and non-natural amino acids.
  • the dipeptide comprises natural amino acids.
  • the linker is a cathepsin labile linker
  • the dipeptide is the site of action for cathepsin-mediated cleavage.
  • the dipeptide then is a recognition site for cathepsin.
  • the group -X X 2 - in dipeptide, - ⁇ -XrX ⁇ CO- is selected from:
  • Cit citrulline.
  • -NH- is the amino group of Xi
  • CO is the carbonyl group of X 2 .
  • the group -XrX 2 - in dipeptide, -NH-XrX 2 -CO-, is selected from:
  • the group -XrX 2 - in dipeptide, -NH-X X 2 -CO-, is -Phe-Lys-, Val-Cit or -Val-Ala-.
  • Other dipeptide combinations of interest include:
  • dipeptide combinations may be used, including those described by Dubowchik et al., which is incorporated herein by reference.
  • the amino acid side chain is chemically protected, where appropriate.
  • the side chain protecting group may be a group as discussed below.
  • Protected amino acid sequences are cleavable by enzymes. For example, a dipeptide sequence comprising a Boc side chain-protected Lys residue is cleavable by cathepsin.
  • Lys Boc, Z-CI, Fmoc, Z;
  • -X 2 - is connected indirectly to the Drug unit.
  • the Spacer unit L 2 is present.
  • the dipeptide is used in combination with a self-immolative group(s) (the Spacer unit).
  • the self-immolative group(s) may be connected to -X 2 -.
  • -X 2 - is connected directly to the self-immolative group.
  • -X 2 - is connected to the group Y of the self-immolative group.
  • the group -X 2 -CO- is connected to Y, where Y is NH.
  • -X is connected directly to A 1 .
  • -X r is connected directly to A 1 .
  • the group NH-X-i- (the amino terminus of X-i) is connected to A 1 .
  • a 1 may comprise the functionality -CO- thereby to form an amide link with -X .
  • the PABC group is connected directly to the Drug unit.
  • the self- immolative group and the dipeptide together form the group -Phe-Lys-PABC-, which is illustrated below:
  • the asterisk indicates the point of attachment to the Drug unit
  • the wavy line indicates the point of attachment to the remaining portion of L 1 or the point of attachment to A 1 .
  • the wavy line indicates the point of attachment to A 1 .
  • the self-immolative group and the dipeptide together form the group -Val-Ala- PABC-, which is illustrated below:
  • the asterisk indicates the point of attachment to the Drug unit
  • the wavy line indicates the point of attachment to A 1
  • Y is a covalent bond or a functional group
  • E is a group that is susceptible to cleavage thereby to activate a self-immolative group.
  • E is selected such that the group is susceptible to cleavage, e.g., by light or by the action of an enzyme.
  • E may be -N0 2 or glucuronic acid (e.g., ⁇ -glucuronic acid).
  • the former may be susceptible to the action of a nitroreductase, the latter to the action of a
  • the group Y may be a covalent bond.
  • the group Y may be a functional group selected from:
  • the group Y is preferably -NH-, -CH 2 -, -0-, and -S-.
  • Y is a covalent bond or a functional group and E is glucuronic acid (e.g., ⁇ -glucuronic acid).
  • Y is preferably a functional group selected from -NH-.
  • L 1 and L 2 together represent:
  • Y is a covalent bond or a functional group and E is glucuronic acid (e.g., ⁇ -glucuronic acid).
  • E is glucuronic acid (e.g., ⁇ -glucuronic acid).
  • Y is preferably a functional group selected from -NH-, -CH 2 -, -0-, and -S-.
  • Y is a functional group as set forth above, the functional group is linked to an amino acid, and the amino acid is linked to the Stretcher unit A 1 .
  • amino acid is ⁇ -alanine. In such an embodiment, the amino acid is equivalently considered part of the Stretcher unit.
  • the Specificity unit L 1 and the Ligand unit are indirectly connected via the Stretcher unit.
  • L 1 and A 1 may be connected by a bond selected from:
  • the group A 1 is:
  • n is 0 to 6. In one embodiment, n is 5.
  • n is 0 to 6. In one embodiment, n is 5.
  • the group A 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the group A 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the group 1 is:
  • n is 0 to 6. In one embodiment, n is 5.
  • the group A 1 is:
  • n is 0 to 6. In one embodiment, n is 5.
  • the group A 1 is:
  • n 0 or 1
  • m 0 to 30.
  • the group A 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • connection between the Ligand unit and A 1 is through a thiol residue of the Ligand unit and a maleimide group of A 1 .
  • connection between the Ligand unit and A 1 is:
  • the S atom is typically derived from the Ligand unit.
  • the maleimide-derived group is replaced with the group:
  • the maleimide-derived group is replaced with a group, which optionally together with a Ligand unit (e.g., a Cell Binding Agent), is selected from:
  • a Ligand unit e.g., a Cell Binding Agent
  • the maleimide-derived group is replaced with a group, which optionally together with the Ligand unit, is s
  • the Stretcher unit A 1 is present, the Specificity unit L 1 is present and Spacer unit L 2 is absent.
  • L 1 and the Drug unit are directly connected via a bond.
  • L 2 is a bond.
  • L 1 and D are preferably connected by a bond selected from:
  • L 1 comprises a dipeptide and one end of the dipeptide is linked to D.
  • the amino acids in the dipeptide may be any combination of natural amino acids and non-natural amino acids.
  • the dipeptide comprises natural amino acids.
  • the linker is a cathepsin labile linker
  • the dipeptide is the site of action for cathepsin-mediated cleavage. The dipeptide then is a recognition site for cathepsin.
  • the group -X X 2 - in dipeptide is selected from:
  • Cit is citrulline.
  • -NH- is the amino group of Xi
  • CO is the carbonyl group of X 2 .
  • the group -X ! -X 2 - in dipeptide is selected from:
  • the group -X ! -X 2 - in dipeptide, -NH-XrX 2 -CO-, is -Phe-Lys- or -Val-Ala-.
  • dipeptide combinations of interest include:
  • L 1 -D is:
  • the wavy line indicates the point of attachment to the remaining portion of L 1 or the point of attachment to A 1 .
  • the wavy line indicates the point of attachment to A 1 .
  • the dipeptide is valine-alanine and L 1 -D is:
  • the dipeptide is phenylalanine-lysine and L 1 -D is:
  • the dipeptide is valine-citrulline. In one embodiment, the groups A 1
  • n 0 to 6. In one embodiment, is 5.
  • the groups A 1 -L 1 are:
  • n is 0 to 6. In one embodiment, n is 5.
  • the groups A 1 -L 1 are:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the groups A 1 -L 1 are:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 7, preferably 3 to 7, most preferably 3 or 7.
  • n is 0 to 6. In one embodiment, n is 5.
  • the groups A 1 -L 1 are:
  • n is 0 to 6. In one embodiment, n is 5.
  • the groups A 1 are identical to each other. In one embodiment, the groups A 1
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the groups A 1 are identical to each other. In one embodiment, the groups A 1
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the groups L- A 1 -L 1 are:
  • n 0 to 6. In one embodiment, n is 5.
  • the group L-A 1 -L 1 are:
  • n 0 to 6. In one embodiment, n is 5.
  • the groups L-A 1 -L 1 are:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the groups L-A 1 -L 1 are:
  • n 0 or 1
  • m 0 to 30.
  • the groups L-A 1 -L 1 are:
  • n is 5.
  • the groups L-A 1 -L 1 are:
  • n is 5.
  • the groups L-A 1 -L 1 are:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the groups L-A 1 are identical to the groups L-A 1
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
  • the Stretcher unit is an acetamide unit, having the formula:
  • Stretcher unit, L 1 or D, and the wavy line indicates the point of attachment to the Ligand unit.
  • Linker-Drug compounds are provided for conjugation to a Ligand unit.
  • the Linker-Drug compounds are designed for connection to a Cell Binding Agent.
  • G 1 is a Stretcher group (A 1 ) to form a connection to a Ligand unit
  • L 1 is a Specificity unit
  • the Drug Linker compound has the formula:
  • G 1 -L 1 -L 2 - where the asterisk indicates the point of attachment to the Drug unit, G 1 is a Stretcher unit (A 1 ) to form a connection to a Ligand unit, L 1 is a Specificity unit, L 2 (a Spacer unit) is a covalent bond or a self-immolative group(s).
  • L 1 and L 2 are as defined above. References to connection to A 1 can be construed here as referring to a connection to G 1 .
  • L 1 comprises an amino acid
  • the side chain of that amino acid may be protected. Any suitable protecting group may be used.
  • the side chain protecting groups are removable with other protecting groups in the compound, where present.
  • the protecting groups may be orthogonal to other protecting groups in the molecule, where present.
  • Suitable protecting groups for amino acid side chains include those groups described in the Novabiochem Catalog 2006/2007. Protecting groups for use in a cathepsin labile linker are also discussed in Dubowchik et al.
  • the group L 1 includes a Lys amino acid residue.
  • the side chain of this amino acid may be protected with a Boc or Alloc protected group.
  • a Boc protecting group is most preferred.
  • the functional group G 1 forms a connecting group upon reaction with a Ligand unit (e.g., a cell binding agent.
  • a Ligand unit e.g., a cell binding agent.
  • the functional group G 1 is or comprises an amino, carboxylic acid, hydroxy, thiol, or maleimide group for reaction with an appropriate group on the Ligand unit.
  • G 1 comprises a maleimide group.
  • the group G 1 is an alkyl maleimide group. This group is suitable for reaction with thiol groups, particularly cysteine thiol groups, present in the cell binding agent, for example present in an antibody.
  • the group G 1 is:
  • n is 0 to 6. In one embodiment, n is 5.
  • the group G 1 is: where the asterisk indicates the point of attachment to L 1 , L 2 or D, and n is 0 to 6. In one embodiment, n is 5.
  • the group G 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 2, preferably 4 to 8, and most preferably 4 or 8.
  • the group G 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, and most preferably 4 or 8.
  • the group G 1 is the group G 1
  • n is 0 to 6. In one embodiment, n is 5.
  • the group G 1 is:
  • n is 0 to 6. In one embodiment, n is 5.
  • the group G 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 2, preferably 4 to 8, and most preferably 4 or 8.
  • the roup G 1 is:
  • n is 0 or 1
  • m is 0 to 30.
  • n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, and most preferably 4 or 8.
  • the maleimide-derived group is replaced with the group:
  • the maleimide group is replaced with a group selected from:
  • L 1 is present, and G 1 is -NH 2 , -NHMe, -COOH, -OH or -SH.
  • G 1 is -NH 2 or -NHMe. Either group may be the N-terminal of an L 1 amino acid sequence.
  • L 1 is present and G 1 is -NH 2 , and L 1 is an amino acid sequence -X X 2 - , as defined above. In one embodiment, L 1 is present and G 1 is COOH. This group may be the C-terminal of an L 1 amino acid sequence.
  • L 1 is present and G 1 is OH.
  • L 1 is present and G 1 is SH.
  • the group G 1 may be convertible from one functional group to another. In one
  • L 1 is present and G 1 is -NH 2 .
  • This group is convertable to another group G 1 comprising a maleimide group.
  • the group -NH 2 may be reacted with an acids or an activated acid (e.g., N-succinimide forms) of those G 1 groups comprising maleimide shown above.
  • the group G 1 may therefore be converted to a functional group that is more appropriate for reaction with a Ligand unit.
  • L 1 is present and G 1 is -NH 2 , -NHMe, -COOH, -OH or -SH.
  • these groups are provided in a chemically protected form.
  • the chemically protected form is therefore a precursor to the linker that is provided with a functional group.
  • G 1 is -NH 2 in a chemically protected form.
  • the group may be protected with a carbamate protecting group.
  • the carbamate protecting group may be selected from the group consisting of:
  • G 1 is -NH 2 , it is protected with an Alloc or Fmoc group.
  • G 1 is -NH 2
  • it is protected with an Fmoc group.
  • the protecting group is the same as the carbamate protecting group of the capping group.
  • the protecting group is not the same as the carbamate protecting group of the capping group.
  • it is preferred that the protecting group is removable under conditions that do not remove the carbamate protecting group of the capping group.
  • the chemical protecting group may be removed to provide a functional group to form a connection to a Ligand unit.
  • this functional group may then be converted to another functional group as described above.
  • the active group is an amine.
  • This amine is preferably the N-terminal amine of a peptide, and may be the N-terminal amine of the preferred dipeptides of the invention.
  • the active group may be reacted to yield the functional group that is intended to form a connection to a Ligand unit.
  • the Linker unit is a precursor to the Linker unit having an active group.
  • the Linker unit comprises the active group, which is protected by way of a protecting group. The protecting group may be removed to provide the Linker unit having an active group.
  • the protecting group may be an amine protecting group, such as those described in Green and Wuts.
  • the protecting group is preferably orthogonal to other protecting groups, where present, in the Linker unit.
  • the protecting group is orthogonal to the capping group.
  • the active group protecting group is removable whilst retaining the capping group.
  • the protecting group and the capping group is removable under the same conditions as those used to remove the capping group.
  • the Linker unit is:
  • the asterisk indicates the point of attachment to the Drug unit
  • the wavy line indicates the point of attachment to the remaining portion of the Linker unit, as applicable or the point of attachment to G 1 .
  • the wavy line indicates the point of attachment to G 1 .
  • the Linker unit is:
  • the Ligand Unit may be of any kind, and include a protein, polypeptide, peptide and a non- peptidic agent that specifically binds to a target molecule.
  • the Ligand unit may be a protein, polypeptide or peptide.
  • the Ligand unit may be a cyclic polypeptide.
  • These Ligand units can include antibodies or a fragment of an antibody that contains at least one target molecule-binding site, lymphokines, hormones, growth factors, or any other cell binding molecule or substance that can specifically bind to a target.
  • Ligand units include those agents described for use in WO 2007/085930, which is incorporated herein.
  • the Ligand unit is a Cell Binding Agent that binds to an extracellular target on a cell.
  • a Cell Binding Agent can be a protein, polypeptide, peptide or a non- peptidic agent.
  • the Cell Binding Agent may be a protein, polypeptide or peptide.
  • the Cell Binding Agent may be a cyclic polypeptide.
  • the Cell Binding Agent also may be antibody or an antigen-binding fragment of an antibody.
  • the present invention provides an antibody-drug conjugate (ADC).
  • the antibody is a monoclonal antibody; chimeric antibody; humanized antibody; fully human antibody; or a single chain antibody.
  • the antibody is a fragment of one of these antibodies having biological activity.
  • fragments include Fab, Fab', F(ab') 2 and Fv fragments.
  • the antibody may be a diabody, a domain antibody (DAB) or a single chain antibody.
  • the antibody is a monoclonal antibody.
  • Antibodies for use in the present invention include those antibodies described in
  • the conjugates are designed to target tumour cells via their cell surface antigens.
  • the antigens may be cell surface antigens which are either over- expressed or expressed at abnormal times or cell types.
  • the target antigen is expressed only on proliferative cells (preferably tumour cells); however this is rarely observed in practice. As a result, target antigens are usually selected on the basis of differential expression between proliferative and healthy tissue.
  • Antibodies have been raised to target specific tumour related antigens including:
  • the Ligand unit is connected to the Linker unit.
  • the Ligand unit is connected to A, where present, of the Linker unit.
  • the connection between the Ligand unit and the Linker unit is through a thioether bond.
  • connection between the Ligand unit and the Linker unit is through a disulfide bond.
  • connection between the Ligand unit and the Linker unit is through an amide bond. In one embodiment, the connection between the Ligand unit and the Linker unit is through an ester bond.
  • connection between the Ligand unit and the Linker is formed between a thiol group of a cysteine residue of the Ligand unit and a maleimide group of the Linker unit.
  • the cysteine residues of the Ligand unit may be available for reaction with the functional group of the Linker unit to form a connection.
  • the thiol groups of the antibody may participate in interchain disulfide bonds. These interchain bonds may be converted to free thiol groups by e.g. treatment of the antibody with DTT prior to reaction with the functional group of the Linker unit.
  • the cysteine residue is an introduced into the heavy or light chain of an antibody. Positions for cysteine insertion by substitution in antibody heavy or light chains include those described in Published U.S. Application No. 2007-0092940 and International Patent Publication WO2008070593, which are incorporated herein. Methods of Treatment
  • the Conjugates of the present invention may be used in a method of therapy. Also provided is a method of treatment, comprising administering to a subject in need of treatment a therapeutically-effective amount of a Conjugate of formula I.
  • a method of treatment comprising administering to a subject in need of treatment a therapeutically-effective amount of a Conjugate of formula I.
  • terapéuticaally effective amount is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom.
  • the actual amount of a Conjugate administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors.
  • the amount of the Conjugate administered ranges from about 0.01 to about 10 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.05 to about 5 mg/kg per dose.
  • the amount of the Conjugate administered ranges from about 0.1 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 4 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.05 to about 3 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 3 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 2 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 1 mg/kg per dose.
  • a conjugate may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • treatments and therapies include, but are not limited to, chemotherapy (the administration of active agents, including, e.g. drugs; surgery; and radiation therapy).
  • compositions according to the present invention may comprise, in addition to the active ingredient, i.e. a
  • Conjugate 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.
  • a capsule may comprise a solid carrier such a 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.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.
  • Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
  • a reference to carboxylic acid (-COOH) also includes the anionic (carboxylate) form (-COO " ), a salt or solvate thereof, as well as conventional protected forms.
  • a reference to an amino group includes the protonated form (-N + HR 1 R 2 ), a salt or solvate of the amino group, for example, a hydrochloride salt, as well as conventional protected forms of an amino group.
  • a reference to a hydroxyl group also includes the anionic form (-0 " ), a salt or solvate thereof, as well as conventional protected forms.
  • a corresponding salt of the active compound for example, a pharmaceutically-acceptable salt.
  • a pharmaceutically-acceptable salt examples are discussed in Berge, et al., J. Pharm. Sci., 66, 1-19 (1977).
  • a salt may be formed with a suitable cation.
  • suitable inorganic cations include, but are not limited to, alkali metal ions such as Na + and K + , alkaline earth cations such as Ca 2+ and Mg 2+ , and other cations such as ⁇ 3 .
  • suitable organic cations include, but are not limited to, ammonium ion (i.e. NH 4 + ) and substituted ammonium ions (e.g. NH 3 R + , NH 2 R2 + , NHR 3 + , NR 4 + ).
  • suitable substituted ammonium ions are those derived from: ethylamine,
  • ethanolamine diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine.
  • amino acids such as lysine and arginine.
  • An example of a common quaternary ammonium ion is N(CH 3 ) 4 + .
  • a salt may be formed with a suitable anion.
  • suitable inorganic anions include, but are not limited to, those derived from the following inorganic acids: hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfurous, nitric, nitrous, phosphoric, and phosphorous.
  • Suitable organic anions include, but are not limited to, those derived from the following organic acids: 2-acetyoxybenzoic, acetic, ascorbic, aspartic, benzoic,
  • suitable polymeric organic anions include, but are not limited to, those derived from the following polymeric acids: tannic acid, carboxymethyl cellulose.
  • solvate is used herein in the conventional sense to refer to a complex of solute (e.g. active Conjugate, salt of active Conjugate) and solvent. If the solvent is water, the solvate may be conveniently referred to as a hydrate, for example, a mono-hydrate, a di-hydrate, a tri-hydrate, etc.
  • the invention includes Conjugate where a solvent adds across the imine bond of the PBD moiety, which is illustrated below for a PBD monomer where the solvent is water or an alcohol (R A OH, where R A is d -4 alkyl):
  • carbinolamine and carbinolamine ether forms of the PBD can be called the carbinolamine and carbinolamine ether forms of the PBD.
  • the balance of these equilibria depend on the conditions in which the compounds are found, as well as the nature of the moiety itself.
  • Certain compounds may exist in one or more particular geometric, optical, enantiomeric, diasteriomeric, epimeric, atropic, stereoisomeric, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans-forms; E- and Z-forms; c-, t-, and r- forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and l-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal- and anticlinal-forms; a- and ⁇ -forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and halfchair- forms; and combinations thereof, hereinafter collectively referred to as "isomers” (or "isomeric forms").
  • isomers are structural (or constitutional) isomers (i.e. isomers which differ in the connections between atoms rather than merely by the position of atoms in space).
  • a reference to a methoxy group, -OCH 3 is not to be construed as a reference to its structural isomer, a hydroxymethyl group, -CH 2 OH.
  • a reference to ortho-chlorophenyl is not to be construed as a reference to its structural isomer, meta- chlorophenyl.
  • a reference to a class of structures may well include structurally isomeric forms falling within that class (e.g. C 1-7 alkyl includes n-propyl and iso-propyl; butyl includes n-, iso-, sec-, and tert-butyl; methoxyphenyl includes ortho-, meta-, and para- methoxyphenyl).
  • C 1-7 alkyl includes n-propyl and iso-propyl
  • butyl includes n-, iso-, sec-, and tert-butyl
  • methoxyphenyl includes ortho-, meta-, and para- methoxyphenyl
  • keto/enol (illustrated below), imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime,
  • H may be in any isotopic form, including 1 H, 2 H (D), and 3 H (T); C may be in any isotopic form, including 12 C, 13 C, and 14 C; O may be in any isotopic form, including 16 0 and 18 0; and the like.
  • a reference to a particular compound or Conjugate includes all such isomeric forms, including (wholly or partially) racemic and other mixtures thereof.
  • Methods for the preparation (e.g. asymmetric synthesis) and separation (e.g. fractional crystallisation and chromatographic means) of such isomeric forms are either known in the art or are readily obtained by adapting the methods taught herein, or known methods, in a known manner.
  • R 2 , R 6 , R 7 , R 9 , R 6' , R 7' , R 9' , R 12 , X, X' and R" are as defined for compounds of formula II
  • Prot N is a nitrogen protecting group for synthesis
  • Prot° is a protected oxygen group for synthesis or an oxo group, by deprotecting the imine bond by standard methods.
  • the compound produced may be in its carbinolamine or carbinolamine ether form depending on the solvents used. For example if Prot N is Alloc and Prot° is an oxygen protecting group for synthesis, then the deprotection is carried using palladium to remove the N10 protecting group, followed by the elimination of the oxygen protecting group for synthesis. If Prot N is Troc and Prot° is an oxygen protecting group for synthesis, then the deprotection is carried out using a Cd/Pb couple to yield the compound of formula (I).
  • Prot N is SEM, or an analogous group, and Prot° is an oxo group
  • the oxo group can be removed by reduction, which leads to a protected carbinolamine intermediate, which can then be treated to remove the SEM protecting group, followed by the elimination of water.
  • the reduction of the compound of Compound formula 2 can be accomplished by, for example, lithium tetraborohydride, whilst a suitable means for removing the SEM protecting group is treatment with silica gel.
  • Compounds of Compound formula 2 can be synthesised from a compound of Compound formula 3
  • R 2 , R 6 , R 7 , R 9 , R 6' , R 7' , R 9' , X, X' and R" are as defined for compounds of Compound formula 2, by coupling an organometallic derivative comprising R 12 , such as an
  • the organoboron derivative may be a boronate or boronic acid.
  • R 12 , R 6 , R 7 , R 9 , R 6' , R 7' , R 9' , X, X' and R" are as defined for compounds of
  • Compound formula 2 by coupling an organometallic derivative comprising R 2 , such as an organoboron derivative.
  • organoboron derivative may be a boronate or boronic acid.
  • R 2 , R 6 , R 7 , R 9 , R 6' , R 7' , R 9' , X, X' and R" are as defined for compounds of Compound formula 2, by coupling about a single equivalent (e.g. 0.9 or 1 to 1.1 or 1.2) of an organometallic derivative, such as an organoboron derivative, comprising R 2 or R 12 .
  • an organometallic derivative such as an organoboron derivative
  • the couplings described above are usually carried out in the presence of a palladium catalyst, for example Pd(PPh 3 ) 4 , Pd(OCOCH 3 ) 2 , PdCI 2 , or Pd 2 (dba) 3 .
  • a palladium catalyst for example Pd(PPh 3 ) 4 , Pd(OCOCH 3 ) 2 , PdCI 2 , or Pd 2 (dba) 3 .
  • the coupling may be carried out under standard conditions, or may also be carried out under microwave conditions.
  • the two coupling steps are usually carried out sequentially. They may be carried out with or without purification between the two steps. If no purification is carried out, then the two steps may be carried out in the same reaction vessel. Purification is usually required after the second coupling step. Purification of the compound from the undesired by-products may be carried out by column chromatography or ion-exchange separation.
  • Nitrogen protecting groups for synthesis are well known in the art.
  • the protecting groups of particular interest are carbamate nitrogen protecting groups and hemi-aminal nitrogen protecting groups.
  • Carbamate nitrogen protecting groups have the following structure:
  • R' 10 is R as defined above.
  • R' 10 is R as defined above.
  • Particularly preferred protecting groups include Troc, Teoc, Fmoc, BOC, Doc, Hoc, TcBOC, 1 -Adoc and 2-Adoc.
  • nitrobenzyloxycarbonyl e.g. 4- nitrobenzyloxycarbonyl
  • 2- (phenylsulphonyl)ethoxycarbonyl e.g. 4- nitrobenzyloxycarbonyl
  • nitrobenzyloxycarbonyl e.g. 4- nitrobenzyloxycarbonyl
  • 2- (phenylsulphonyl)ethoxycarbonyl e.g. 4- nitrobenzyloxycarbonyl
  • 2- (phenylsulphonyl)ethoxycarbonyl e.g. 4- nitrobenzyloxycarbonyl
  • R' 10 is R as defined above.
  • suitable groups are described on pages 633 to 647 as amide protecting groups of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.
  • the groups disclosed herein can be applied to compounds for use in the present invention. Such groups include, but are not limited to, SEM, MOM, MTM, MEM, BOM, nitro or methoxy substituted BOM, and CI 3 CCH 2 OCH 2 -.
  • Protected oxygen group for synthesis include, but are not limited to, SEM, MOM, MTM, MEM, BOM, nitro or methoxy substituted BOM, and CI 3 CCH 2 OCH 2 -.
  • Protected oxygen group for synthesis include, but are not limited to, SEM, MOM, MTM, MEM, BOM, nitro or methoxy substituted BOM, and CI
  • Classes of particular interest include silyl ethers, methyl ethers, alkyl ethers, benzyl ethers, esters, acetates, benzoates, carbonates, and sulfonates.
  • Preferred oxygen protecting groups include acetates, TBS and THP. Further Preferences
  • R 6' , R 7' , R 9' , R 10' , R 11' and Y' are preferably the same as R 6 , R 7 , R 9 , R 10 , R 11 and Y respectively. Dimer link
  • Y and Y' are preferably O.
  • R" is preferably a C 3-7 alkylene group with no substituents. More preferably R" is a C 3 , C 5 or C 7 alkylene.
  • R 9 is preferably H.
  • R 6 is preferably selected from H, OH, OR, SH, NH 2 , nitro and halo, and is more preferably H or halo, and most preferably is H.
  • R 7 is preferably selected from H, OH, OR, SH, SR, NH 2 , NHR, NRR', and halo, and more preferably independently selected from H, OH and OR, where R is preferably selected from optionally substituted C 1-7 alkyl, C 3-10 heterocyclyl and C 5-10 aryl groups. R may be more preferably a C 1-4 alkyl group, which may or may not be substituted.
  • a substituent of interest is a C 5-6 aryl group (e.g. phenyl). Particularly preferred substituents at the 7- positions are OMe and OCH 2 Ph.
  • a in R 2 may be phenyl group or a C 5-7 heteroaryl group, for example furanyl, thiophenyl and pyridyl.
  • A is preferably phenyl.
  • A is preferably thiophenyl, for example, thiophen-2-yl and thiophen-3-yl.
  • Q 2 -X may be on any of the available ring atoms of the C 5-7 aryl group, but is preferably on a ring atom that is not adjacent the bond to the remainder of the compound, i.e. it is preferably ⁇ or ⁇ to the bond to the remainder of the compound. Therefore, where the C 5-7 aryl group (A) is phenyl, the substituent (Q 2 -X) is preferably in the meta- or para- positions, and more preferably is in the para- position.1
  • Q 1 is a single bond.
  • Q 2 is selected from a single bond and -Z-(CH 2 )n-, where Z is selected from a single bond, O, S and NH and is from 1 to 3.
  • Q 2 is a single bond.
  • Q 2 is -Z-(CH 2 ) n --
  • Z may be O or S and n may be 1 or n may be 2.
  • Z may be a single bond and n may be 1.
  • R2 may be -A-CH 2 -X and -A-X.
  • X may be - 0-, -S-, -C(0)0-, -C(O)- and -NH-.
  • X may be -NH-.
  • R 12 may be a C 5-7 aryl group.
  • a C 5-7 aryl group may be a phenyl group or a C 5-7 heteroaryl group, for example furanyl, thiophenyl and pyridyl.
  • R 12 is preferably phenyl.
  • R 12 is preferably thiophenyl, for example, thiophen-2-yl and thiophen-3-yl.
  • R 12 may be a C 8- io aryl, for example a quinolinyl or isoquinolinyl group.
  • the quinolinyl or isoquinolinyl group may be bound to the PBD core through any available ring position.
  • the quinolinyl may be quinolin-2-yl, quinolin-3-yl, quinolin-4yl, quinolin-5-yl, quinolin-6-yl, quinolin-7-yl and quinolin-8-yl. Of these quinolin-3-yl and quinolin-6-yl may be preferred.
  • the isoquinolinyl may be isoquinolin-1-yl, isoquinolin-3-yl, isoquinolin-4yl, isoquinolin-5-yl, isoquinolin-6-yl, isoquinolin-7-yl and isoquinolin-8-yl. Of these isoquinolin- 3-yl and isoquinolin-6-yl may be preferred.
  • R 12 may bear any number of substituent groups. It preferably bears from 1 to 3 substituent groups, with 1 and 2 being more preferred, and singly substituted groups being most preferred. The substituents may be any position.
  • R 12 is C 5-7 aryl group
  • a single substituent is preferably on a ring atom that is not adjacent the bond to the remainder of the compound, i.e. it is preferably ⁇ or ⁇ to the bond to the remainder of the compound. Therefore, where the C 5-7 aryl group is phenyl, the substituent is preferably in the meta- or para- positions, and more preferably is in the para- position.
  • R 12 is a C 8- io aryl group, for example quinolinyl or isoquinolinyl, it may bear any number of substituents at any position of the quinoline or isoquinoline rings. In some embodiments, it bears one, two or three substituents, and these may be on either the proximal and distal rings or both (if more than one substituent).
  • a substituent on R 12 is halo, it is preferably F or CI, more preferably CI.
  • a substituent on R 12 is ether, it may in some embodiments be an alkoxy group, for example, a Ci -7 alkoxy group (e.g. methoxy, ethoxy) or it may in some embodiments be a C 5-7 aryloxy group (e.g. phenoxy, pyridyloxy, furanyloxy). The alkoxy group may itself be further substituted, for example by an amino group (e.g. dimethylamino).
  • a substituent on R 12 is C 1-7 alkyl, it may preferably be a C 1-4 alkyl group (e.g. methyl, ethyl, propyl, butyl).
  • a substituent on R 12 is C 3-7 heterocyclyl, it may in some embodiments be C 6 nitrogen containing heterocyclyl group, e.g. morpholino, thiomorpholino, piperidinyl, piperazinyl. These groups may be bound to the rest of the PBD moiety via the nitrogen atom. These groups may be further substituted, for example, by C- alkyl groups. If the C 6 nitrogen containing heterocyclyl group is piperazinyl, the said further substituent may be on the second nitrogen ring atom. If a substituent on R 12 is bis-oxy-Ci -3 alkylene, this is preferably bis-oxy-methylene or bis- oxy-ethylene.
  • substituents for R 12 include methoxy, ethoxy, fluoro, chloro, cyano, bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thiophenyl.
  • Another particularly preferred substituent for R 12 is dimethylaminopropyloxy.
  • Particularly preferred substituted R 12 groups include, but are not limited to, 4-methoxy- phenyl, 3-methoxyphenyl, 4-ethoxy-phenyl, 3-ethoxy-phenyl, 4-fluoro-phenyl, 4-chloro- phenyl, 3,4-bisoxymethylene-phenyl, 4-methylthiophenyl, 4-cyanophenyl, 4- phenoxyphenyl, quinolin-3-yl and quinolin-6-yl, isoquinolin-3-yl and isoquinolin-6-yl, 2- thienyl, 2-furanyl, methoxynaphthyl, and naphthyl.
  • Another possible substituted R 12 group is 4-nitrophenyl.
  • M and M' are monovalent pharmaceutically acceptable cations, and are more preferably Na + .
  • z is preferably 3.
  • Optical rotations were measured on an ADP 220 polarimeter (Bellingham Stanley Ltd.) and concentrations (c) are given in g/100ml_. Melting points were measured using a digital melting point apparatus (Electrothermal). IR spectra were recorded on a Perkin-Elmer Spectrum 1000 FT IR Spectrometer. 1 H and 13 C NMR spectra were acquired at 300 K using a Bruker Avance NMR spectrometer at 400 and 100 MHz, respectively.
  • Waters Micromass ZQ parameters used were: Capillary (kV), 3.38; Cone (V), 35; Extractor (V), 3.0; Source temperature (°C), 100; Desolvation Temperature (°C), 200; Cone flow rate (L/h), 50; De-solvation flow rate (L/h), 250.
  • HRMS High-resolution mass spectroscopy
  • HRMS High-resolution mass spectroscopy
  • TLC Thin Layer Chromatography
  • Compound 1 b was synthesised as described in WO 00/012508 (compound 210), which is herein incorporated by reference.
  • General LC/MS conditions The HPLC (Waters Alliance 2695) was run using a mobile phase of water (A) (formic acid 0.1 %) and acetonitrile (B) (formic acid 0.1 %). Gradient: initial composition 5% B over 1 .0 min then 5% B to 95% B within 3 min. The composition was held for 0.5 min at 95% B, and then returned to 5% B in 0.3 minutes. Total gradient run time equals 5 min. Flow rate 3.0 mL/min, 400 ⁇ _ was split via a zero dead volume tee piece which passes into the mass spectrometer. Wavelength detection range: 220 to 400 nm. Function type: diode array (535 scans). Column: Phenomenex ® Onyx Monolithic C18 50 x 4.60 mm
  • LC/MS conditions specific for compounds protected by both a Troc and a TBDMs group Chromatographic separation of Troc and TBDMS protected compounds was performed on a Waters Alliance 2895 HPLC system utilizing a Onyx Monoliiic reversed-phase column (3 ⁇ particles, 50 x 4.8 mm) from Phenomenex Corp.
  • Mobile-phase A consisted of 5% acetonitrile - 95 % water containing 0.1 % formic acid
  • mobile phase B consisted of 95% acetonitrile - 5% water containing 0.1 % formic acid.
  • LC/MS conditions specific for compound 33 LC was run on a Waters 2767 sample Manager coupled with a Waters 2996 photodiode array detector and a Waters ZQ single quadruple mass Spectrometer. The column used was Luna Phenyl-Hexyl 150 x 4.60 mm, 5 ⁇ , Part no. 00F-4257-E0 (Phenomenex). The mobile phases employed were:
  • reaction suspension became a solution and the mixture was allowed to stir at room temperature for 16 hours. Conversion to the acid chloride was confirmed by treating a sample of the reaction mixture with MeOH and the resulting bis- methyl ester was observed by LC/MS. The majority of solvent was removed by
  • Method A A suspension of 10% Pd/C (7.5 g, 10% w/w) in DMF (40 mL) was added to a solution of the nitro-ester 2a (75 g, 104 mmol) in DMF (360 mL). The suspension was hydrogenated in a Parr hydrogenation apparatus over 8 hours. Progress of the reaction was monitored by LC/MS (2.12 min (ES+) m/z (relative intensity) 597 ([M + H] + , 100), (ES-) m/z (relative intensity) 595 ([M + H] + , 100) after the hydrogen uptake had stopped.
  • Solid Pd/C was removed by filtration and the filtrate was concentrated by rotary evaporation under vacuum (below 10 mbar) at 40°C to afford a dark oil containing traces of DMF and residual charcoal.
  • the residue was digested in EtOH (500 mL) at 40°C on a water bath (rotary evaporator bath) and the resulting suspension was filtered through celite and washed with ethanol (500 mL) to give a clear filtrate.
  • Hydrazine hydrate (10 mL, 321 mmol) was added to the solution and the reaction mixture was heated at reflux. After 20 minutes the formation of a white precipitate was observed and reflux was allowed to continue for a further 30 minutes. The mixture was allowed to cool down to room temperature and the precipitate was retrieved by filtration, washed with diethyl ether (2 * 1 volume of precipitate) and dried in a vacuum desiccator to provide 3a (50 g, 81 %).
  • Method B A solution of the nitro-ester 2a (6.80 g, 9.44 mmol) in MeOH (300 mL) was added to RaneyTM nickel (4 large spatula ends of a ⁇ 50% slurry in H 2 0) and anti-bumping granules in a 3-neck round bottomed flask. The mixture was heated at reflux and then treated dropwise with a solution of hydrazine hydrate (5.88 mL, 6.05 g, 188 mmol) in MeOH (50 mL) at which point vigorous effervescence was observed. When the addition was complete ( ⁇ 30 minutes) additional RaneyTM nickel was added carefully until effervescence had ceased and the initial yellow colour of the reaction mixture was discharged.
  • Method A A 0.37 M sodium hypochlorite solution (142.5 mL, 52.71 mmol, 2.4 eq) was added dropwise to a vigorously stirred mixture of the diol 6a (18.8 g, 21.96 mmol, 1 eq), TEMPO (0.069 g, 0.44 mmol, 0.02 eq) and 0.5 M potassium bromide solution (8.9 mL, 4.4 mmol, 0.2 eq) in DCM (1 15 mL) at 0°C. The temperature was maintained between 0°C and 5°C by adjusting the rate of addition. The resultant yellow emulsion was stirred at 0°C to 5°C for 1 hour.
  • Sodium hypochlorite solution reagent grade, available at chlorine 10-13%, was used. This was assumed to be 10% (10 g NaCIO in 100 g) and calculated to be 1.34 M in NaCIO. A stock solution was prepared from this by diluting it to 0.37 M with water. This gave a solution of approximately pH 14. The pH was adjusted to 9.3 to 9.4 by the addition of solid NaHC0 3 . An aliquot of this stock was then used so as to give 2.4 mol eq. for the reaction. On addition of the bleach solution an initial increase in temperature was observed. The rate of addition was controlled, to maintain the temperature between 0°C to 5°C. The reaction mixture formed a thick, lemon yellow coloured, emulsion. The oxidation was an adaptation of the procedure described in Thomas Fey et al, J. Org. Chem., 2001 , 66, 8154-8159.
  • Method B Solid TCCA (10.6 g, 45.6 mmol) was added portionwise to a stirred solution of the alcohol 6a (18.05 g, 21 .1 mmol) and TEMPO (123 mg, 0.78 mmol) in anhydrous DCM (700 mL) at 0°C (ice/acetone). The reaction mixture was stirred at 0°C under a nitrogen atmosphere for 15 minutes after which time TLC (EtOAc) and LC/MS [3.57 min (ES+) m/z (relative intensity) 875 ([M + Na] + , 50)] revealed completion of reaction.
  • Method C A solution of anhydrous DMSO (0.72 mL, 0.84 g, 10.5 mmol) in dry DCM (18 mL) was added dropwise over a period of 25 min to a stirred solution of oxalyl chloride (2.63 mL of a 2.0 M solution in DCM, 5.26 mmol) under a nitrogen atmosphere at -60°C (liq N 2 /CHCI 3 ). After stirring at -55°C for 20 minutes, a slurry of the substrate 6a (1 .5 g, 1 .75 mmol) in dry DCM (36 mL) was added dropwise over a period of 30 min to the reaction mixture.
  • Anhydrous 2,6-lutidine (5.15 mL, 4.74 g, 44.2 mmol) was injected in one portion to a vigorously stirred solution of bis-ketone 7a (6.08 g, 7.1 mmol) in dry DCM (180 mL) at - 45°C (dry ice/acetonitrile cooling bath) under a nitrogen atmosphere.
  • Anhydrous triflic anhydride taken from a freshly opened ampoule (7.2 mL, 12.08 g, 42.8 mmol), was injected rapidly dropwise, while maintaining the temperature at -40°C or below.
  • reaction mixture was allowed to stir at -45°C for 1 hour at which point TLC (50/50 v/v n- hexane/EtOAc) revealed the complete consumption of starting material.
  • the cold reaction mixture was immediately diluted with DCM (200 mL) and, with vigorous shaking, washed with water (1 x 100 mL), 5% citric acid solution (1 x 200 mL) saturated NaHC0 3 (200 mL), brine (100 mL) and dried (MgS0 4 ).
  • Solid Pd(PPh 3 ) 4 (10 mg, 8.69 ⁇ ) was added to a stirred solution of the mono-triflate 9 (230 mg, 0.22 mmol) in toluene (3 mL), EtOH (10 mL), with 4-methoxyphenyl boronic acid (43 mg, 0.28 mmol), Na 2 C0 3 (37 mg, 0.35 mmol), in H 2 0 (1 .5 mL) at room temperature.
  • the reaction mixture was allowed to stir under a nitrogen atmosphere for 20 h, at which point the reaction was deemed complete as judged by LC/MS and TLC (EtOAc).
  • Fresh LiBH 4 (183 mg, 8.42 mmol) was added to a stirred solution of the SEM-dilactam 10 (428 mg, 0.42 mmol) in THF (5 mL) and EtOH (5 mL) at room temperature. After 10 minutes, delayed vigorous effervescence was observed requiring the reaction vessel to be placed in an ice bath. After removal of the ice bath the mixture was allowed to stir at room temperature for 1 hour. LC/MS analysis at this point revealed total consumption of starting material with very little mono-reduced product. The reaction mixture was poured onto ice (100 mL) and allowed to warm to room temperature with stirring.
  • Solid Pd(PPh 3 ) 4 (32 mg, 27.7 ⁇ ) was added to a stirred solution of the bis-triflate 8b (1 .04 g, 0.91 mmol) in toluene (10 mL), EtOH (5 mL), with 4-methoxyphenyl boronic acid (0.202 g, 1 .32 mmol), Na 2 C0 3 (0.169 g, 1 .6 mmol), in H 2 0 (5 mL) at 30°C. The reaction mixture was allowed to stir under a nitrogen atmosphere for 20 hours.
  • Fresh LiBH 4 (66 mg, 3.04 mmol) was added to a stirred solution of the SEM-dilactam 12 (428 mg, 0.42 mmol) in THF (3 mL) and EtOH (3 mL) at 0°C (ice bath). The ice bath was removed and the reaction mixture was allowed to reach room temperature (vigorous effervescence). After 2 hours LC/MS analysis indicated the complete consumption of starting material. The reaction mixture was poured onto ice (50 mL) and allowed to warm to room temperature with stirring.
  • the aqueous mixture was extracted with DCM (3 x50 mL) and the combined organic layers washed with H 2 0 (50 mL), brine (50 mL), dried (MgS0 4 ) and concentrated in vacuo.
  • the resulting residue was treated with DCM (2 mL), EtOH (5 mL), H 2 0 (2.5 mL) and silica gel (3.7 g).
  • the viscous mixture was allowed to stir at room temperature for 3 days.
  • the mixture was filtered through a sinter funnel and the silica residue washed with 90% CHCI 3 : 10% MeOH (-250 mL) until UV activity faded completely from the eluent.
  • the organic phase was dried (MgS0 4 ) filtered and evaporated in vacuo to provide the crude material.
  • the crude product was purified by flash chromatography (99.5% CHCIs : 0.5% MeOH to 97.5% CHCI 3 : 2.5% MeOH in 0.5% increments)) to provide the pure C2/C2'aryl PBD dimer 13 (59 mg, 52%).
  • Solid Pd(PPh 3 ) 4 (41 mg, 0.036 mmol) was added to a stirred solution of the bis-triflate 8a (1 g, 0.9 mmol) in toluene (10 mL), EtOH (5 mL), with thien-2-yl boronic acid (149 mg, 1.16 mmol), Na 2 C0 3 (152 mg, 1.43 mmol), in H 2 0 (5 mL).
  • the reaction mixture was allowed to stir under a nitrogen atmosphere overnight at room temperature.
  • the solvent was removed by evaporation in vacuo and the resulting residue partitioned between H 2 0 (100 mL) and EtOAc (100 mL).
  • Solid Pd(PPh 3 ) 4 (7.66 mg, 6.63 ⁇ ) was added to a stirred, cloudy solution of 14 (174 mg, 0.17 mmol), Na 2 C0 3 (28 mg, 0.22 mmol) and 4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaboralan-2-yl)aniline (47 mg, 0.22 mmol) in toluene (2-5 mL), EtOH (1 .25 mL) and H 2 0 (125 mL) at room temperature.
  • reaction mixture was partitioned between chloroform and water, the layers were separated and the aqueous layer extracted with additional chloroform (emulsions). Finally the combined organic phase was washed with brine and dried over magnesium sulphate. The reduced product was dissolved in methanol, chloroform and water and allowed to stir in the presence of silica gel for 72 hours The crude product was subjected to flash column chromatography (methanol/chloroform gradient) to afford the desired imine product (150 mg, 0.21 mmol, 77%) after removal of excess eluent from selected fractions.
  • step (a) The Suzuki coupling procedure described in step (a) was applied to the synthesis of Compound 21.
  • Compound 20 (62.5 mg 0.044 mmol, ) was treated with 1 equivalent of 4- hydroxybenzeneboronic acid (10 mg) at 30°C overnight to afford the desired compound after filtration through a pad of silica gel. (40 mg, 0.029 mmol, 66% yield).
  • the compound was used directly in the subsequent step
  • Cadmium/lead couple (100 mg, Q Dong et al. Tetrahedron Letters vol 36, issue 32, 5681 - 5682, 1995) was added to a solution of 21 (40 mg, 0.029 mmol) in THF (1 mL) and ammonium acetate (1 N, 1 mL) and the reaction mixture was allowed to stir for 1 hour. The reaction mixture was partitioned between chloroform and water, the phases separated and the aqueous phase extracted with chloroform. The combined organic layers were washed with brine and dried over magnesium sulphate. Rotary evaporation under reduced pressure yielded the crude product which was subjected to column chromatography (silica gel, 0 ⁇ 4% MeOH/CHCI 3 ). Removal of excess eluent by rotary evaporation under reduced pressure afforded the desired imine product (17 mg 0.023 mmol 79%).
  • step (a) The Suzuki coupling procedure described in Example 5, step (a), was applied to the synthesis of Compound 24.
  • Compound 21 (62.5 mg, 0.044 mmol ) was treated with 1 equivalent of 4-formylbenzeneboronic acid (10.5 mg) at room temperature overnight to afford the desired compound after filtration through a pad of silica gel (45 mg, 0.033 mmol, 75% yield). The compound was used directly in the subsequent step.
  • step (a) The Suzuki coupling procedure described in Example 5, step (a), was applied to the synthesis of Compound 26, using 3-aminobenzeneboronic acid to afford the desired compound in 41 % yield (230 mg, 0.163 mmol)
  • step (b) The method of Example 7, step (b), was performed to afford the desired product (58 mg, .0.040 mmol, 78%) after filtration through a plug of silica gel (with 1/3 methanol/chloroform) and removal of excess solvent by rotary evaporation under reduced pressure.
  • step (c) The method for Example 7, step (c) was used to deprotect compound 29.
  • Solid 4-aminomethylbenzeneboronic acid hydrochloride (0.1 1 1 g, 0.59 mmol) was added to a solution of 17 (0.394 g, 0.37 mmol), sodium carbonate (175 mg, 1 .654 mmol) and palladium tetrakis triphenylphosphine (28.0 mg, 0.024 mmol) in toluene (10 mL), ethanol (5 mL) and water (5 mL). The reaction mixture was allowed to stir overnight at 30°C. The following day the reaction mixture was heated for a further 3 hours at 70°C. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was washed with water and brine and dried over magnesium sulphate.
  • a catalytic amount of tetrakistriphenylphosphinepalladium (0) (1 1.2 mg) was added to a mixture of the mono triflate 17 (380 mg), the pinnacol ester of 2-aminophenylboronic acid (124 mg) and sodium carbonate (120 mg) in ethanol (5 mL), toluene (5ml_) and water (5 mL).
  • the reaction mixture was allowed to stir over night at room temperature and at 40°C until the reaction was complete (c. 2 hr).
  • the reaction mixture was diluted with ethyl acetate and the organic layer was washed with water and brine. The ethyl acetate solution was dried over magnesium sulphate and filtered under vacuum.
  • K562 human chronic myeloid leukaemia cells were maintained in RPM1 1640 medium supplemented with 10% fetal calf serum and 2 mM glutamine at 37°C in a humidified atmosphere containing 5% C0 2 and were incubated with a specified dose of drug for 1 hour or 96 hours at 37°C in the dark. The incubation was terminated by centrifugation (5 min, 300 g) and the cells were washed once with drug-free medium. Following the appropriate drug treatment, the cells were transferred to 96-well microtiter plates (10 4 cells per well, 8 wells per sample). Plates were then kept in the dark 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-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT, Aldrich-Sigma), to an insoluble purple formazan precipitate.
  • MTT yellow soluble tetrazolium salt
  • 20 ⁇ _ of MTT solution 5 mg/mL in phosphate-buffered saline
  • the plates were then centrifuged for 5 min at 300 g and the bulk of the medium pipetted from the cell pellet leaving 10-20 ⁇ _ per well.
  • DMSO 200 ⁇ _
  • the optical density was then read at a wavelength of 550 nm on a Titertek Multiscan ELISA plate reader, and a dose-response curve was constructed. 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.
  • Compound 13 has an IC 50 of 30 pM in this assay.
  • the A2780 parental cell line was grown in Dulbecco's Modified Eagles' Media (DMEM) containing -10% Foetal Calf Serum (FCS) and -1 % 200mM L-Glutamine solution and grown in Corning Cellbind 75cm 2 flasks.
  • DMEM Dulbecco's Modified Eagles' Media
  • FCS Foetal Calf Serum
  • FCS Foetal Calf Serum
  • a 190 ⁇ cell suspension was added (at 1 x 10 4 ) to each well of columns 2 to 1 1 of a 96 well plate (Nunc 96F flat bottom TC plate). 190 ⁇ of media was added to each well of columns 1 and 12.
  • the media was Dulbecco's Modified Eagles' Media (DMEM) (which included ⁇ 10% Foetal Calf Serum (FCS) and -1 % 200mM L-Glutamine solution).
  • DMEM Dulbecco's Modified Eagles' Media
  • Compound 11 has an IC 50 of 1 1.7 pM in this assay.
  • cytotoxicity of various free drug compounds was tested on a renal cell cancer cell line, 786-0, a Hodgkin lymphoma cell line, L428 and two AML cell lines, HL60 and HEL.
  • test article i.e., free drug
  • 50 ⁇ _ of each dilution was added to the 96-well plates.
  • Resazurin was then added to each well to achieve a 50 ⁇ final concentration, and the plates were incubated for an additional 4 h at 37 °C.
  • the plates were then read for the extent of dye reduction on a Fusion HT plate reader (Packard Instruments, Meridien, CT, USA) with excitation and emission wavelengths of 530 and 590 nm, respectively.
  • the IC 50 value determined in triplicate, is defined here as the concentration that results in a 50% reduction in cell growth relative to untreated controls.
  • the para-aniline compound 11 showed markedly increased activity on these cell lines as compared to the mefa-aniline compound 19 in this assay.
  • the activities of compounds 23, 25, are compared to that of compound 11 on 786-0, Caki-1 , MCF-7, HL-60, THP-1 , H EL, and TF1 cells.
  • Cells were plated in 150 ⁇ iL growth media per well into black-sided clear-bottom 96-well plates (Costar, Corning) and allowed to settle for 1 hour in the biological cabinet before placing in the incubator at 37°C, 5% C0 2 .
  • 4X concentration of drug stocks were prepared, and then titrated as 10-fold serial dilutions producing 8-point dose curves and added at 50 ⁇ per well in duplicate. Cells were then incubated for 48 hours at 37°C, 5% C0 2 .
  • Cytotoxicity was measure by incubating with 100 ⁇ _ Cell Titer Glo (Promega) solution for 1 hour, and then luminescence was measured on a Fusion HT plate reader (Perkin Elmer). Data was processed with Excel (Microsoft) and Graph Pad (Prism) to produce dose response curves and IC50 values were generated and data collected.
  • the acidic mobile phase consisted of acetonitrile and water both containing either 0.05% trifluoroacetic acid or 0.1 % formic acid (denoted for each compound).
  • LC-MS was performed on a ZMD Micromass mass spectrometer interfaced to an HP Agilent 1 100 HPLC instrument equipped with a C12 Phenomenex Synergi 2.0 x 150 mm, 4 ⁇ , 80 A reverse phase column.
  • Preparative HPLC was carried out on a Varian ProStar 210 solvent delivery system configured with a Varian ProStar 330 PDA detector. Products were purified over a C12 Phenomenex Synergi 10.0 x 250 mm, 4 ⁇ , 80 A reverse phase column eluting with 0.1 % formic acid in water (solvent A) and 0.1 % formic acid in acetonitrile (solvent B).
  • the purification method consisted of the following gradient of solvent A to solvent B: 90:10 from 0 to 5 min; 90:10 to 10:90 from 5 min to 80 min; followed by isocratic 10:90 for 5 min.
  • the flow rate was 4.6 mL/min with monitoring at 254 nm.
  • NMR spectral data were collected on a Varian Mercury 400 MHz spectrometer. Coupling constants (J) are reported in hertz.
  • PBD dimer 57 linked directly through maleimidocaproyl spacer (Scheme 14): PBD dimer 57 is coupled to maleimidocaproic acid 39 employing the chemistry described in Scheme 2.
  • tert-butyl 2-(2-aminoacetamido)acetate (69): To a mixture of the glycine ferf-butyl ester hydrogen chloride salt (70) (484 mg, 2.9 mmol) in dichloromethane (25 mL) was added Fmoc-Gly-OH (71 ) (0.861 mg, 2.99 mmol), DIPEA (756 mg, 4.35 mmol) and HATU (1 .3 g, 3.5 mmol). The reaction mixture was stirred at an ambient temperature for 16 h and then poured into ethyl acetate and was washed with water (3X) and brine (1X). The organic phase was dried over MgS04, filtered and concentrated under reduced pressure.

Abstract

Provided are Conjugate comprising PBDs conjugated to a targeting agent and methods of using such PBDs.

Description

TARGETED PYRROLOBENZODIAZEPINE CONJUGATES
The present invention relates to targeted pyrrolobenzodiazepine (PBD) conjugates, in particular pyrrolobenzodiazepine dimers that are conjugated to a targeting agent via the C2 position of one of the monomers.
Background to the invention
Some pyrrolobenzodiazepines (PBDs) have the ability to recognise and bond to specific sequences of DNA; the preferred sequence is PuGPu. The first PBD antitumour antibiotic, anthramycin, was discovered in 1965 (Leimgruber, et al., J. Am. Chem. Soc, 87, 5793-
5795 (1965); Leimgruber, et al., J. Am. Chem. Soc, 87, 5791-5793 (1965)). 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., Chem. Rev. 1994, 433-465 (1994)). Family members include abbeymycin (Hochlowski, et al., J. Antibiotics, 40, 145- 148 (1987)), chicamycin (Konishi, et al., J. Antibiotics, 37, 200-206 (1984)), DC-81
(Japanese Patent 58-180 487; Thurston, et al., Chem. Brit, 26, 767-772 (1990); Bose, et al., Tetrahedron, 48, 751-758 (1992)), mazethramycin (Kuminoto, et al., J. Antibiotics, 33, 665-667 (1980)), neothramycins A and B (Takeuchi, et al., J. Antibiotics, 29, 93-96 (1976)), porothramycin (Tsunakawa, et al., J. Antibiotics, 41 , 1366-1373 (1988)), prothracarcin (Shimizu, et al, J. Antibiotics, 29, 2492-2503 (1982); Langley and Thurston, J. Org. Chem., 52, 91 -97 (1987)), sibanomicin (DC-102)(Hara, et al., J. Antibiotics, 41 , 702-704 (1988); Itoh, et al., J. Antibiotics, 41 , 1281-1284 (1988)), sibiromycin (Leber, et al., J. Am. Chem. Soc, 110, 2992-2993 (1988)) and tomamycin (Arima, et al., J. Antibiotics, 25, 437-444 (1972)). PBDs are of the general structure:
Figure imgf000002_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-C1 1 position, which is the electrophilic centre responsible for alkylating DNA. All of the known natural products have an (S)-configuration at the chiral C1 1 a 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, In Antibiotics III. Springer-Verlag, New York, pp. 3-1 1 (1975); Hurley and Needham- VanDevanter, Acc. Chem. Res., 19, 230-237 (1986)). The ability of PBDs to form an adduct in the minor groove enables them to interfere with DNA processing, hence their use as antitumour agents.
The biological activity of these molecules can be potentiated by joining two PBD units together through their C8/C'-hydroxyl functionalities via a flexible alkylene linker (Bose, D.S., et al., J. Am. Chem. Soc, 114, 4939-4941 (1992); Thurston, D.E., et al., J. Org.
Chem., 61 , 8141 -8147 (1996)). The PBD dimers are thought to form sequence-selective DNA lesions such as the palindromic 5'-Pu-GATC-Py-3' interstrand cross-link (Smellie, M., et al., Biochemistry, 42, 8232-8239 (2003); Martin, C, et al., Biochemistry, 44, 4135-4147) which is thought to be mainly responsible for their biological activity. One example of a PBD dimer is SG -136):
Figure imgf000003_0001
(Gregson, S., et al., J. Med. Chem., 44, 737-748 (2001 ); Alley, M.C., et al., Cancer Research, 64, 6700-6706 (2004); Hartley, J.A., et al., Cancer Research, 64, 6693-6699 (2004)).
Due to the manner in which these highly potent compounds act in cross-linking DNA, PBD dimers have been made symmetrically, i.e., both monomers of the dimer are the same. This synthetic route provides for straightforward synthesis, either by constructing the PBD dimer moiety simultaneously having already formed the dimer linkage, or by reacting already constructed PBD monomer moieties with the dimer linking group. These synthetic approaches have limited the options for preparation of targeted conjugates containing PBDs. Due to the observed potency of PBD dimers, however, there exists a need for PBD dimers that are conjugatable to targeting agents for use in targeted therapy. Disclosure of the invention
The present invention relates to Conjugates comprising dimers of PBDs linked to a targeting agent, wherein a PBD monomer has a substituent in the C2 position that provides an anchor for linking the compound to the targeting agent. The present invention also relates to Conjugates comprising dimers of PBDs conjugated to a targeting agent, wherein the PBD monomers of the dimer are different. One of PBD monomers has a substituent in the C2 position that provides an anchor for linking the compound to the targeting agent. The Conjugates described herein have potent cytotoxic and/or cytostatic activity against cells expressing a target molecule, such as cancer cells or immune cells. These conjugates exhibit good potency with reduced toxicity, as compared with the corresponding PBD dimer free drug compounds.
In some embodiments, the Conjugates have the following formula I:
L - (LU-D)p (I)
wherein L is a Ligand unit (i.e., a targeting agent), LU is a Linker unit and D is a Drug unit comprising a PBD dimer. The subscript p is an integer of from 1 to 20. Accordingly, the Conjugates comprise a Ligand unit covalently linked to at least one Drug unit by a Linker unit. The Ligand unit, described more fully below, is a targeting agent that binds to a target moiety. The Ligand unit can, for example, specifically bind to a cell component (a Cell Binding Agent) or to other target molecules of interest. Accordingly, the present invention also provides methods for the treatment of, for example, various cancers and autoimmune disease. These methods encompass the use of the Conjugates wherein the Ligand unit is a targeting agent that specifically binds to a target molecule. The Ligand unit can be, for example, a protein, polypeptide or peptide, such as an antibody, an antigen-binding fragment of an antibody, or other binding agent, such as an Fc fusion protein.
In a first aspect, the Conjugates comprise a Conjugate of formula I (supra), wherein the Drug unit comprises a PBD dimer of the following formula II:
Figure imgf000004_0001
wherein:
R2 is of formula III:
III where A is a C5-7 aryl group, X is an activatable group for conjugation to the Linker unit, wherein X is selected from the group comprising: -0-, -S-, -C(0)0-, -C(O)-, -NHC(O)-, and -N(RN)-, wherein RN is selected from the group comprising H, Ci-4 alkyl and (C2H40)mCH3, where m is 1 to 3, and either:
(i) Q1 is a single bond, and Q2 is selected from a single bond and -Z-(CH2)n-, where
Z is selected from a single bond, O, S and NH and n is from 1 to 3; or
(ii) Q1 is -CH=CH-, and Q2 is a single bond;
R12 is a C5-io aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, C1-7 alkyl, C3-7 heterocyclyl and bis-oxy-C1-3 alkylene;
R6 and R9 are independently selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR', nitro, Me3Sn and halo;
where R and R' are independently selected from optionally substituted C1-12 alkyl, optionally substituted C3-20 heterocyclyl and optionally substituted C5-20 aryl groups;
R7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NHRR', nitro, Me3Sn and halo; either:
(a) R10 is H, and R11 is OH or ORA, where RA is C1-4 alkyl;
(b) R10 and R11 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or
(c) R10 is H and R11 is SOzM, where z is 2 or 3 and M is a monovalent
pharmaceutically acceptable cation;
R" is a C3-i2 alkylene group, which chain may be interrupted by one or more heteroatoms, e.g. O, S, NRN2 (where RN2 is H or Ci-4 alkyl), and/or aromatic rings, e.g. benzene or pyridine;
Y and Y' are selected from O, S, or NH;
R6 , R7 , R9 are selected from the same groups as R6, R7 and R9 respectively and R10 and R11 are the same as R10 and R11, wherein if R11 and R11 are SOzM, M may represent a divalent pharmaceutically acceptable cation. In a second aspect, the use of the Conjugate of formula I is provided for the manufacture of a medicament for treating a proliferative disease or autoimmune disease. In a related third aspect, the use of the Conjugate of formula I is provided for the treatment of a proliferative disease or an autoimmune disease. In another aspect there is provided the use of a Conjugate of formula I to provide a PBD dimer, or a salt or solvate thereof, at a target location.
One of ordinary skill in the art is readily able to determine whether or not a candidate conjugate treats a proliferative condition for any particular cell type. For example, assays which may conveniently be used to assess the activity offered by a particular compound are described in the examples below.
The term "proliferative disease" pertains to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo.
Examples of proliferative conditions include, but are not limited to, benign, pre-malignant, and malignant cellular proliferation, including but not limited to, neoplasms and tumours (e.g., histocytoma, glioma, astrocyoma, osteoma), cancers (e.g. lung cancer, small cell lung cancer, gastrointestinal cancer, bowel cancer, colon cancer, breast carinoma, ovarian carcinoma, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, pancreatic cancer, brain cancer, sarcoma, osteosarcoma, Kaposi's sarcoma, melanoma), leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g. of connective tissues), and atherosclerosis. Other cancers of interest include, but are not limited to, haematological; malignancies such as leukemias and lymphomas, such as non-Hodgkin lymphoma, and subtypes such as DLBCL, marginal zone, mantle zone, and follicular, Hodgkin lymphoma, AML, and other cancers of B or T cell origin. Examples of autoimmune disease include the following: rheumatoid arthritis, autoimmune demyelinative diseases (e.g., multiple sclerosis, allergic encephalomyelitis), psoriatic arthritis, endocrine ophthalmopathy, uveoretinitis, systemic lupus erythematosus, myasthenia gravis, Graves' disease, glomerulonephritis, autoimmune hepatological disorder, inflammatory bowel disease (e.g., Crohn's disease), anaphylaxis, allergic reaction, Sjogren's syndrome, type I diabetes mellitus, primary biliary cirrhosis, Wegener's granulomatosis, fibromyalgia, polymyositis, dermatomyositis, multiple endocrine failure, Schmidt's syndrome, autoimmune uveitis, Addison's disease, adrenalitis, thyroiditis, Hashimoto's thyroiditis, autoimmune thyroid disease, pernicious anemia, gastric atrophy, chronic hepatitis, lupoid hepatitis, atherosclerosis, subacute cutaneous lupus
erythematosus, hypoparathyroidism, Dressler's syndrome, autoimmune thrombocytopenia, idiopathic thrombocytopenic purpura, hemolytic anemia, pemphigus vulgaris, pemphigus, dermatitis herpetiformis, alopecia areata, pemphigoid, scleroderma, progressive systemic sclerosis, CREST syndrome (calcinosis, Raynaud's phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), male and female autoimmune infertility, ankylosing spondolytis, ulcerative colitis, mixed connective tissue disease, polyarteritis nedosa, systemic necrotizing vasculitis, atopic dermatitis, atopic rhinitis, Goodpasture's syndrome, Chagas' disease, sarcoidosis, rheumatic fever, asthma, recurrent abortion, anti- phospholipid syndrome, farmer's lung, erythema multiforme, post cardiotomy syndrome, Cushing's syndrome, autoimmune chronic active hepatitis, bird-fancier's lung, toxic epidermal necrolysis, Alport's syndrome, alveolitis, allergic alveolitis, fibrosing alveolitis, interstitial lung disease, erythema nodosum, pyoderma gangrenosum, transfusion reaction, Takayasu's arteritis, polymyalgia rheumatica, temporal arteritis, schistosomiasis, giant cell arteritis, ascariasis, aspergillosis, Sampter's syndrome, eczema, lymphomatoid
granulomatosis, Behcet's disease, Caplan's syndrome, Kawasaki's disease, dengue, encephalomyelitis, endocarditis, endomyocardial fibrosis, endophthalmitis, erythema elevatum et diutinum, psoriasis, erythroblastosis fetalis, eosinophilic faciitis, Shulman's syndrome, Felty's syndrome, filariasis, cyclitis, chronic cyclitis, heterochronic cyclitis, Fuch's cyclitis, IgA nephropathy, Henoch-Schonlein purpura, graft versus host disease, transplantation rejection, cardiomyopathy, Eaton-Lambert syndrome, relapsing
polychondritis, cryoglobulinemia, Waldenstrom's macroglobulemia, Evan's syndrome, and autoimmune gonadal failure.
In some embodiments, the autoimmune disease is a disorder of B lymphocytes (e.g., systemic lupus erythematosus, Goodpasture's syndrome, rheumatoid arthritis, and type I diabetes), Th1 -lymphocytes (e.g., rheumatoid arthritis, multiple sclerosis, psoriasis,
Sjogren's syndrome, Hashimoto's thyroiditis, Graves' disease, primary biliary cirrhosis, Wegener's granulomatosis, tuberculosis, or graft versus host disease), or Th2-lymphocyt.es (e.g., atopic dermatitis, systemic lupus erythematosus, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn's syndrome, systemic sclerosis, or chronic graft versus host disease). Generally, disorders involving dendritic cells involve disorders of Th1 - lymphocytes or Th2-lymphocytes. In some embodiments, the autoimmune disorder is a T cell-mediated immunological disorder.
In a fourth aspect of the present invention comprises a method of making the Conjugates formula I. The dimeric PBD compounds for use in the present invention are made by different strategies to those previously employed in making symmetrical dimeric PBD compounds. In particular, the present inventors have developed a method which involves adding each C2 aryl substituent to a symmetrical PBD dimer core in separate method steps.
Accordingly, a sixth aspect of the present invention provides a method of making a
Conjugate of formula I, comprising at least one of the method steps described herein.
Brief Description of the Figures
Figures 1 to 6 show the effect of conjugates of the present invention in tumours.
Definitions
When a trade name is used herein, reference to the trade name also refers to the product formulation, the generic drug, and the active pharmaceutical ingredient(s) of the trade name product, unless otherwise indicated by context.
Binding Agent and Targeting Agent
The terms "binding agent" and "targeting agent as used herein refer to a molecule, e.g., protein, polypeptide or peptide, that specifically binds to a target molecule. Examples can include a full length antibody, an antigen binding fragment of a full length antibody, other agent (protein, polypeptide or peptide) that includes an antibody heavy and/or light chain variable region that specifically bind to the target molecule, or an Fc fusion protein comprising an extracellular domain of a protein, peptide polypeptide that binds to the target molecule and that is joined to an Fc region, domain or portion thereof, of an antibody.
Specifically Binds
The terms "specifically binds" and "specific binding" refer to the binding of an antibody or other protein, polypeptide or peptide to a predetermined molecule (e.g., an antigen).
Typically, the antibody or other molecule binds with an affinity of at least about 1x107 M"1, and binds to the predetermined molecule with an affinity that is at least two-fold greater than its affinity for binding to a non-specific molecule (e.g., BSA, casein) other than the predetermined molecule or a closely-related molecule. Pharmaceutically acceptable cations
Examples of pharmaceutically acceptable monovalent and divalent cations are discussed in Berge, et al., J. Pharm. Sci., 66, 1 -19 (1977), which is incorporated herein by reference. The pharmaceutically acceptable cation may be inorganic or organic.
Examples of pharmaceutically acceptable monovalent inorganic cations include, but are not limited to, alkali metal ions such as Na+ and K+. Examples of pharmaceutically acceptable divalent inorganic cations include, but are not limited to, alkaline earth cations such as Ca2+ and Mg2+. Examples of pharmaceutically acceptable organic cations include, but are not limited to, ammonium ion (i.e. NH4 +) and substituted ammonium ions (e.g. NH3R+, NH2R2+, NHR3 +, NR4 +). Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH3)4 +. Substituents
The phrase "optionally substituted" as used herein, pertains to a parent group which may be unsubstituted or which may be substituted.
Unless otherwise specified, the term "substituted" as used herein, pertains to a parent group which bears one or more substituents. The term "substituent" is used herein in the conventional sense and refers to a chemical moiety which is covalently attached to, or if appropriate, fused to, a parent group. A wide variety of substituents are well known, and methods for their formation and introduction into a variety of parent groups are also well known.
Examples of substituents are described in more detail below. C-i-12 alkyl: The term
Figure imgf000009_0001
alkyl" as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to 12 carbon atoms, which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated). Thus, the term "alkyl" includes the sub-classes alkenyl, alkynyl, cycloalkyl, etc., discussed below. Examples of saturated alkyl groups include, but are not limited to, methyl (Ci), ethyl (C2), propyl (C3), butyl (C4), pentyl (C5), hexyl (C6) and heptyl (C7).
Examples of saturated linear alkyl groups include, but are not limited to, methyl (Ci), ethyl (C2), n-propyl (C3), n-butyl (C4), n-pentyl (amyl) (C5), n-hexyl (C6) and n-heptyl (C7).
Examples of saturated branched alkyl groups include iso-propyl (C3), iso-butyl (C4), sec-butyl (C4), tert-butyl (C4), iso-pentyl (C5), and neo-pentyl (C5). C2-12 Alkenyl: The term "C2-12 alkenyl" as used herein, pertains to an alkyl group having one or more carbon-carbon double bonds.
Examples of unsaturated alkenyl groups include, but are not limited to, ethenyl (vinyl, - CH=CH2), 1 -propenyl (-CH=CH-CH3), 2-propenyl (allyl, -CH-CH=CH2), isopropenyl (1- methylvinyl, -C(CH3)=CH2), butenyl (C4), pentenyl (C5), and hexenyl (C6).
C2-12 alkynyl: The term "C2-12 alkynyl" as used herein, pertains to an alkyl group having one or more carbon-carbon triple bonds. Examples of unsaturated alkynyl groups include, but are not limited to, ethynyl (-C≡CH) and 2-propynyl (propargyl, -CH2-C≡CH).
C3-12 cycloalkyl: The term "C3-i2 cycloalkyl" as used herein, pertains to an alkyl group which is also a cyclyl group; that is, a monovalent moiety obtained by removing a hydrogen atom from an alicyclic ring atom of a cyclic hydrocarbon (carbocyclic) compound, which moiety has from 3 to 7 carbon atoms, including from 3 to 7 ring atoms.
Examples of cycloalkyl groups include, but are not limited to, those derived from:
saturated monocyclic hydrocarbon compounds:
cyclopropane (C3), cyclobutane (C4), cyclopentane (C5), cyclohexane (C6), cycloheptane (C7), methylcyclopropane (C4), dimethylcyclopropane (C5), methylcyclobutane (C5), dimethylcyclobutane (C6), methylcyclopentane (C6), dimethylcyclopentane (C7) and methylcyclohexane (C7);
unsaturated monocyclic hydrocarbon compounds: cyclopropene (C3), cyclobutene (C4), cyclopentene (C5), cyclohexene (C6), methylcyclopropene (C4), dimethylcyclopropene (C5), methylcyclobutene (C5), dimethylcyclobutene (C6), methylcyclopentene (C6), dimethylcyclopentene (C7) and methylcyclohexene (C7); and
saturated polycyclic hydrocarbon compounds:
norcarane (C7), norpinane (C7), norbornane (C7).
C3-2o heterocyclyl: The term "C3-2o heterocyclyl" as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound, which moiety has from 3 to 20 ring atoms, of which from 1 to 10 are ring heteroatoms. Preferably, each ring has from 3 to 7 ring atoms, of which from 1 to 4 are ring heteroatoms.
In this context, the prefixes (e.g. C3-20, C3-7, C5-6, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term "C5-6heterocyclyl", as used herein, pertains to a heterocyclyl group having 5 or 6 ring atoms.
Examples of monocyclic heterocyclyl groups include, but are not limited to, those derived from:
N-i : aziridine (C3), azetidine (C4), pyrrolidine (tetrahydropyrrole) (C5), pyrroline (e.g., 3-pyrroline, 2,5-dihydropyrrole) (C5), 2H-pyrrole or 3H-pyrrole (isopyrrole, isoazole) (C5), piperidine (C6), dihydropyridine (C6), tetrahydropyridine (C6), azepine (C7);
O1 : oxirane (C3), oxetane (C4), oxolane (tetrahydrofuran) (C5), oxole (dihydrofuran) (C5), oxane (tetrahydropyran) (C6), dihydropyran (C6), pyran (C6), oxepin (C7);
S-i : thiirane (C3), thietane (C4), thiolane (tetrahydrothiophene) (C5), thiane
(tetrahydrothiopyran) (C6), thiepane (C7);
02: dioxolane (C5), dioxane (C6), and dioxepane (C7);
03: trioxane (C6);
N2: imidazolidine (C5), pyrazolidine (diazolidine) (C5), imidazoline (C5), pyrazoline
(dihydropyrazole) (C5), piperazine (C6);
N1O1 : tetrahydrooxazole (C5), dihydrooxazole (C5), tetrahydroisoxazole (C5),
dihydroisoxazole (C5), morpholine (C6), tetrahydrooxazine (C6), dihydrooxazine (C6), oxazine (C6);
thiazoline (C5), thiazolidine (C5), thiomorpholine (C6); N2Oi : oxadiazine (C6);
O1S1 : oxathiole (C5) and oxathiane (thioxane) (C6); and,
N"|OiSi : oxathiazine (C6). Examples of substituted monocyclic heterocyclyl groups include those derived from saccharides, in cyclic form, for example, furanoses (C5), such as arabinofuranose, lyxofuranose, ribofuranose, and xylofuranse, and pyranoses (C6), such as allopyranose, altropyranose, glucopyranose, mannopyranose, gulopyranose, idopyranose,
galactopyranose, and talopyranose.
C5-20 aryl: The term "C5-20 aryl", as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from an aromatic ring atom of an aromatic compound, which moiety has from 3 to 20 ring atoms. Preferably, each ring has from 5 to 7 ring atoms. In this context, the prefixes (e.g. C3-20, C5-7, C5-6, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term "C5-6 aryl" as used herein, pertains to an aryl group having 5 or 6 ring atoms.
The ring atoms may be all carbon atoms, as in "carboaryl groups".
Examples of carboaryl groups include, but are not limited to, those derived from benzene (i.e. phenyl) (C6), naphthalene (C10), azulene (C10), anthracene (C14), phenanthrene (C14), naphthacene (Ci8), and pyrene (Ci6).
Examples of aryl groups which comprise fused rings, at least one of which is an aromatic ring, include, but are not limited to, groups derived from indane (e.g. 2,3-dihydro-1 H- indene) (Cg), indene (Cg), isoindene (Cg), tetraline (1 ,2,3,4-tetrahydronaphthalene (C10), acenaphthene (C12), fluorene (C13), phenalene (C13), acephenanthrene (C15), and aceanthrene (C16). Alternatively, the ring atoms may include one or more heteroatoms, as in "heteroaryl groups". Examples of monocyclic heteroaryl groups include, but are not limited to, those derived from:
Ni : pyrrole (azole) (C5), pyridine (azine) (C6);
O1 : furan (oxole) (C5);
Si : thiophene (thiole) (C5); N1O1 : oxazole (C5), isoxazole (C5), isoxazine (C6);
N2Oi : oxadiazole (furazan) (C5);
N3O1 : oxatriazole (C5);
N-\S-\ \ thiazole (C5), isothiazole (C5);
N2: imidazole (1 ,3-diazole) (C5), pyrazole (1 ,2-diazole) (C5), pyridazine (1 ,2-diazine) (C6), pyrimidine (1 ,3-diazine) (C6) (e.g., cytosine, thymine, uracil), pyrazine (1 ,4-diazine) (C6); N3: triazole (C5), triazine (C6); and,
N4: tetrazole (C5). Examples of heteroaryl which comprise fused rings, include, but are not limited to:
Cg (with 2 fused rings) derived from benzofuran (O-i), isobenzofuran (O-i ), indole (N-i ), isoindole (N-i ), indolizine (N-i), indoline (N-i), isoindoline (N-i), purine (N4) (e.g., adenine, guanine), benzimidazole (N2), indazole (N2), benzoxazole (N-ιΟ-ι), benzisoxazole (N-I O-I ), benzodioxole (02), benzofurazan (N2Oi ), benzotriazole (N3), benzothiofuran (Si), benzothiazole (N-IS-I), benzothiadiazole (N2S);
Cio (with 2 fused rings) derived from chromene (O1 ), isochromene (O1), chroman (O1), isochroman (O1), benzodioxan (02), quinoline (N-i ), isoquinoline (N-i), quinolizine (N-i), benzoxazine (N-ιΟ-ι), benzodiazine (N2), pyridopyridine (N2), quinoxaline (N2), quinazoline (N2), cinnoline (N2), phthalazine (N2), naphthyridine (N2), pteridine (N4);
C11 (with 2 fused rings) derived from benzodiazepine (N2);
Ci3 (with 3 fused rings) derived from carbazole (N-i), dibenzofuran (O1 ),
dibenzothiophene (Si), carboline (N2), perimidine (N2), pyridoindole (N2); and,
Ci4 (with 3 fused rings) derived from acridine (N-i), xanthene (O1), thioxanthene (Si), oxanthrene (02), phenoxathiin (O1S1 ), phenazine (N2), phenoxazine (N-ιΟ-ι), phenothiazine (N-I S-I), thianthrene (S2), phenanthridine (N-i ), phenanthroline (N2), phenazine (N2).
The above groups, whether alone or part of another substituent, may themselves optionally be substituted with one or more groups selected from themselves and the additional substituents listed below.
Halo: -F, -CI, -Br, and -I.
Hydroxy: -OH. Ether: -OR, wherein R is an ether substituent, for example, a Ci_7 alkyl group (also referred to as a Ci-7 alkoxy group, discussed below), a C3-20 heterocyclyl group (also referred to as a C3-20 heterocyclyloxy group), or a C5-2o aryl group (also referred to as a C5-2o aryloxy group), preferably a Ci-7alkyl group.
Alkoxy: -OR, wherein R is an alkyl group, for example, a Ci-7 alkyl group. Examples of Ci-7 alkoxy groups include, but are not limited to, -OMe (methoxy), -OEt (ethoxy), -O(nPr) (n- propoxy), -O(iPr) (isopropoxy), -O(nBu) (n-butoxy), -O(sBu) (sec-butoxy), -O(iBu)
(isobutoxy), and -O(tBu) (tert-butoxy).
Acetal: -CH(OR1)(OR2), wherein R1 and R2 are independently acetal substituents, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a C1-7 alkyl group, or, in the case of a "cyclic" acetal group, R1 and R2, taken together with the two oxygen atoms to which they are attached, and the carbon atoms to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Examples of acetal groups include, but are not limited to, -CH(OMe)2, -CH(OEt)2, and -CH(OMe)(OEt).
Hemiacetal: -CH(OH)(OR1), wherein R1 is a hemiacetal substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group. Examples of hemiacetal groups include, but are not limited to, -CH(OH)(OMe) and - CH(OH)(OEt).
Ketal: -CR(OR1)(OR2), where R1 and R2 are as defined for acetals, and R is a ketal substituent other than hydrogen, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a C1-7 alkyl group. Examples ketal groups include, but are not limited to, -C(Me)(OMe)2, -C(Me)(OEt)2, -C(Me)(OMe)(OEt), -C(Et)(OMe)2, - C(Et)(OEt)2, and -C(Et)(OMe)(OEt).
Hemiketal: -CR(OH)(OR1), where R1 is as defined for hemiacetals, and R is a hemiketal substituent other than hydrogen, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci-7 alkyl group. Examples of hemiacetal groups include, but are not limited to, -C(Me)(OH)(OMe), -C(Et)(OH)(OMe), -C(Me)(OH)(OEt), and -C(Et)(OH)(OEt).
Oxo (keto, -one): =0. Thione (thioketone): =S.
Imino (imine): =NR, wherein R is an imino substituent, for example, hydrogen, Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably hydrogen or a Ci-7 alkyl group. Examples of ester groups include, but are not limited to, =NH, =NMe, =NEt, and =NPh.
Formyl (carbaldehyde, carboxaldehyde): -C(=0)H.
Acyl (keto): -C(=0)R, wherein R is an acyl substituent, for example, a C1-7 alkyl group (also referred to as Ci-7 alkylacyl or Ci-7 alkanoyl), a C3-20 heterocyclyl group (also referred to as C3-20 heterocyclylacyl), or a C5-2o aryl group (also referred to as C5-20 arylacyl), preferably a Ci-7 alkyl group. Examples of acyl groups include, but are not limited to, -C(=0)CH3 (acetyl), -C(=0)CH2CH3 (propionyl), -C(=0)C(CH3)3 (t-butyryl), and -C(=0)Ph (benzoyl, phenone).
Carboxy (carboxylic acid): -C(=0)OH. Thiocarboxy (thiocarboxylic acid): -C(=S)SH. Thiolocarboxy (thiolocarboxylic acid): -C(=0)SH. Thionocarboxy (thionocarboxylic acid): -C(=S)OH. Imidic acid: -C(=NH)OH. Hydroxamic acid: -C(=NOH)OH.
Ester (carboxylate, carboxylic acid ester, oxycarbonyl): -C(=0)OR, wherein R is an ester substituent, for example, a Ci-7 alkyl group, a C3-2o heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group. Examples of ester groups include, but are not limited to, -C(=0)OCH3, -C(=0)OCH2CH3, -C(=0)OC(CH3)3, and -C(=0)OPh. Acyloxy (reverse ester): -OC(=0)R, wherein R is an acyloxy substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group. Examples of acyloxy groups include, but are not limited to, -OC(=0)CH3 (acetoxy), -OC(=0)CH2CH3, -OC(=0)C(CH3)3, -OC(=0)Ph, and -OC(=0)CH2Ph.
Oxycarboyloxy: -OC(=0)OR, wherein R is an ester substituent, for example, a Ci-7 alkyl group, a C3-2o heterocyclyl group, or a C5-2o aryl group, preferably a C1 -7 alkyl group.
Examples of ester groups include, but are not limited to, -OC(=0)OCH3,
-OC(=0)OCH2CH3, -OC(=0)OC(CH3)3, and -OC(=0)OPh.
Amino: -N R1 R2, wherein R1 and R2 are independently amino substituents, for example, hydrogen, a Ci-7 alkyl group (also referred to as Ci-7 alkylamino or di-Ci-7 alkylamino), a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably H or a Ci-7 alkyl group, or, in the case of a "cyclic" amino group, R1 and R2, taken together with the nitrogen atom to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Amino groups may be primary (-NH2), secondary (-NHR1), or tertiary (-NHR1 R2), and in cationic form, may be quaternary (-+NR1 R2R3). Examples of amino groups include, but are not limited to, -NH2, -N HCH3, -NHC(CH3)2, -N(CH3)2, -N(CH2CH3)2, and -NHPh. Examples of cyclic amino groups include, but are not limited to, aziridino, azetidino, pyrrolidino, piperidino, piperazino, morpholino, and thiomorpholino.
Amido (carbamoyl, carbamyl, aminocarbonyl, carboxamide): -C(=0)NR1 R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=0)NH2, -C(=0)NHCH3, -C(=0)N(CH3)2,
-C(=0)NHCH2CH3, and -C(=0)N(CH2CH3)2, as well as amido groups in which R1 and R2, together with the nitrogen atom to which they are attached, form a heterocyclic structure as in, for example, piperidinocarbonyl, morpholinocarbonyl, thiomorpholinocarbonyl, and piperazinocarbonyl. Thioamido (thiocarbamyl): -C(=S)NR1 R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=S)NH2, -C(=S)NHCH3, -C(=S)N(CH3)2, and -C(=S)NHCH2CH3.
Acylamido (acylamino): -NR1C(=0)R2, wherein R1 is an amide substituent, for example, hydrogen, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably hydrogen or a Ci_7 alkyl group, and R2 is an acyl substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2oaryl group, preferably hydrogen or a C1-7 alkyl group. Examples of acylamide groups include, but are not limited to, -NHC(=0)CH3 ,
-NHC(=0)CH2CH3, and -NHC(=0)Ph. R1 and R2 may together form a cyclic structure, as in for example, succinimidyl, maleimidyl, and phthalimidyl:
Figure imgf000017_0001
nidyl maleimidyl phthalimidyl
Aminocarbonyloxy: -OC(=0)NR1R2, wherein R1 and R2 are independently amino
substituents, as defined for amino groups. Examples of aminocarbonyloxy groups include, but are not limited to, -OC(=0)NH2, -OC(=0)NHMe, -OC(=0)NMe2, and -OC(=0)NEt2.
Ureido: -N(R1)CONR2R3 wherein R2 and R3 are independently amino substituents, as defined for amino groups, and R1 is a ureido substituent, for example, hydrogen, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably hydrogen or a C1-7 alkyl group. Examples of ureido groups include, but are not limited to, -NHCONH2, -
NHCONHMe, -NHCONHEt, -NHCONMe2, -NHCONEt2, -NMeCONH2, -NMeCONHMe, -NMeCONHEt, -NMeCONMe2, and -NMeCONEt2.
Guanidino: -NH-C(=NH)NH2.
Tetrazolyl: a five membered aromatic ring having four nitrogen atoms and one carbon atom,
Figure imgf000017_0002
Imino: =NR, wherein R is an imino substituent, for example, for example, hydrogen, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably H or a Ci-7alkyl group. Examples of imino groups include, but are not limited to, =NH, =NMe, and =NEt. Amidine (amidino): -C(=NR)NR2, wherein each R is an amidine substituent, for example, hydrogen, a Ci_7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably H or a C-i-7 alkyl group. Examples of amidine groups include, but are not limited to,
-C(=NH)NH2, -C(=NH)NMe2, and -C(=NMe)NMe2.
Nitro: -N02. Nitroso: -NO. Azido: -N3.
Cyano (nitrile, carbonitrile): -CN. Isocyano: -NC.
Cyanato: -OCN. Isocyanato: -NCO. Thiocyano (thiocyanato): -SCN.
Isothiocyano (isothiocyanato): -NCS. Sulfhydryl (thiol, mercapto): -SH.
Thioether (sulfide): -SR, wherein R is a thioether substituent, for example, a C1-7 alkyl group (also referred to as a C1-7alkylthio group), a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a C1-7 alkyl group. Examples of C1-7 alkylthio groups include, but are not limited to, -SCH3 and -SCH2CH3.
Disulfide: -SS-R, wherein R is a disulfide substituent, for example, a Ci-7 alkyl group, a C3- 20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci-7 alkyl group (also referred to herein as Ci-7 alkyl disulfide). Examples of Ci-7 alkyl disulfide groups include, but are not limited to, -SSCH3 and -SSCH2CH3. Sulfine (sulfinyl, sulfoxide): -S(=0)R, wherein R is a sulfine substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group. Examples of sulfine groups include, but are not limited to, -S(=0)CH3 and -S(=0)CH2CH3. Sulfone (sulfonyl): -S(=0)2R, wherein R is a sulfone substituent, for example, a Ci-7 alkyl group, a C3-2o heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group, including, for example, a fluorinated or perfluorinated C1-7 alkyl group. Examples of sulfone groups include, but are not limited to, -S(=0)2CH3 (methanesulfonyl, mesyl), -S(=0)2CF3 (triflyl), -S(=0)2CH2CH3 (esyl), -S(=0)2C4F9 (nonaflyl), -S(=0)2CH2CF3 (tresyl),
-S(=0)2CH2CH2NH2 (tauryl), -S(=0)2Ph (phenylsulfonyl, besyl), 4-methylphenylsulfonyl (tosyl), 4-chlorophenylsulfonyl (closyl), 4-bromophenylsulfonyl (brosyl), 4-nitrophenyl (nosyl), 2-naphthalenesulfonate (napsyl), and 5-dimethylamino-naphthalen-1 -ylsulfonate (dansyl). Sulfinic acid (sulfino): -S(=0)OH, -S02H. Sulfonic acid (sulfo): -S(=0)2OH, -S03H.
Sulfinate (sulfinic acid ester): -S(=0)OR; wherein R is a sulfinate substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci-7 alkyl group. Examples of sulfinate groups include, but are not limited to, -S(=0)OCH3
(methoxysulfinyl; methyl sulfinate) and -S(=0)OCH2CH3 (ethoxysulfinyl; ethyl sulfinate).
Sulfonate (sulfonic acid ester): -S(=0)2OR, wherein R is a sulfonate substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a
C1-7 alkyl group. Examples of sulfonate groups include, but are not limited to, -S(=0)2OCH3 (methoxysulfonyl; methyl sulfonate) and -S(=0)2OCH2CH3 (ethoxysulfonyl; ethyl sulfonate).
Sulfinyloxy: -OS(=0)R, wherein R is a sulfinyloxy substituent, for example, a C1 -7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci-7 alkyl group.
Examples of sulfinyloxy groups include, but are not limited to, -OS(=0)CH3 and
-OS(=0)CH2CH3.
Sulfonyloxy: -OS(=0)2R, wherein R is a sulfonyloxy substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci-7 alkyl group. Examples of sulfonyloxy groups include, but are not limited to, -OS(=0)2CH3 (mesylate) and -OS(=0)2CH2CH3 (esylate).
Sulfate: -OS(=0)2OR; wherein R is a sulfate substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a Ci-7 alkyl group. Examples of sulfate groups include, but are not limited to, -OS(=0)2OCH3 and -SO(=0)2OCH2CH3.
Sulfamyl (sulfamoyl; sulfinic acid amide; sulfinamide): -S(=0)NR1 R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of sulfamyl groups include, but are not limited to, -S(=0)NH2, -S(=0)NH(CH3), -S(=0)N(CH3)2,
-S(=0)NH(CH2CH3), -S(=0)N(CH2CH3)2, and -S(=0)NHPh.
Sulfonamido (sulfinamoyl; sulfonic acid amide; sulfonamide): -S(=0)2NR1R2, wherein R1 and R2 are independently amino substituents, as defined for amino groups. Examples of sulfonamido groups include, but are not limited to, -S(=0)2NH2, -S(=0)2NH(CH3),
-S(=0)2N(CH3)2, -S(=0)2NH(CH2CH3), -S(=0)2N(CH2CH3)2, and -S(=0)2NHPh.
Sulfamino: -NR1S(=0)2OH, wherein R1 is an amino substituent, as defined for amino groups. Examples of sulfamino groups include, but are not limited to, -NHS(=0)2OH and -N(CH3)S(=0)2OH.
Sulfonamino: -NR1S(=0)2R, wherein R1 is an amino substituent, as defined for amino groups, and R is a sulfonamino substituent, for example, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group. Examples of sulfonamino groups include, but are not limited to, -NHS(=0)2CH3 and -N(CH3)S(=0)2C6H5.
Sulfinamino: -NR1S(=0)R, wherein R1 is an amino substituent, as defined for amino groups, and R is a sulfinamino substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a C1-7 alkyl group. Examples of sulfinamino groups include, but are not limited to, -NHS(=0)CH3 and -N(CH3)S(=0)C6H5.
Phosphino (phosphine): -PR2, wherein R is a phosphino substituent, for example, -H, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably -H, a Ci-7 alkyl group, or a C5-20 aryl group. Examples of phosphino groups include, but are not limited to, -PH2, -P(CH3)2, -P(CH2CH3)2, -P(t-Bu)2, and -P(Ph)2. Phospho: -P(=0)2.
Phosphinyl (phosphine oxide): -P(=0)R2, wherein R is a phosphinyl substituent, for example, a Ci-7 alkyl group, a C3-2o heterocyclyl group, or a C5-2o aryl group, preferably a Ci-7 alkyl group or a C5-20 aryl group. Examples of phosphinyl groups include, but are not limited to, -P(=0)(CH3)2, -P(=0)(CH2CH3)2, -P(=0)(t-Bu)2, and -P(=0)(Ph)2.
Phosphonic acid (phosphono): -P(=0)(OH)2.
Phosphonate (phosphono ester): -P(=0)(OR)2, where R is a phosphonate substituent, for example, -H, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably -H, a Ci-7 alkyl group, or a C5-20 aryl group. Examples of phosphonate groups include, but are not limited to, -P(=0)(OCH3)2, -P(=0)(OCH2CH3)2, -P(=0)(0-t-Bu)2, and -P(=0)(OPh)2.
Phosphoric acid (phosphonooxy): -OP(=0)(OH)2.
Phosphate (phosphonooxy ester): -OP(=0)(OR)2, where R is a phosphate substituent, for example, -H, a Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably - H, a Ci_7 alkyl group, or a C5-20 aryl group. Examples of phosphate groups include, but are not limited to, -OP(=0)(OCH3)2, -OP(=0)(OCH2CH3)2, -OP(=0)(0-t-Bu)2, and
-OP(=0)(OPh)2.
Phosphorous acid: -OP(OH)2.
Phosphite: -OP(OR)2, where R is a phosphite substituent, for example, -H, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably -H, a C1-7 alkyl group, or a C5-20 aryl group. Examples of phosphite groups include, but are not limited to, -OP(OCH3)2, -OP(OCH2CH3)2, -OP(0-t-Bu)2, and -OP(OPh)2.
Phosphoramidite: -OP(OR1)-NR2 2, where R1 and R2 are phosphoramidite substituents, for example, -H, a (optionally substituted) Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably -H, a Ci-7 alkyl group, or a C5-2o aryl group. Examples of
phosphoramidite groups include, but are not limited to, -OP(OCH2CH3)-N(CH3)2,
-OP(OCH2CH3)-N(i-Pr)2, and -OP(OCH2CH2CN)-N(i-Pr)2. Phosphoramidate: -OP(=0)(OR1)-NR2 2, where R1 and R2 are phosphoramidate
substituents, for example, -H, a (optionally substituted) Ci-7 alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably -H, a Ci-7 alkyl group, or a C5-2o aryl group.
Examples of phosphoramidate groups include, but are not limited to, -OP(=0)(OCH2CH3)- N(CH3)2, -OP(=0)(OCH2CH3)-N(i-Pr)2, and -OP(=0)(OCH2CH2CN)-N(i-Pr)2.
Alkylene
C3-12 alkylene: The term "C3-i2 alkylene", as used herein, pertains to a bidentate moiety obtained by removing two hydrogen atoms, either both from the same carbon atom, or one from each of two different carbon atoms, of a hydrocarbon compound having from 3 to 12 carbon atoms (unless otherwise specified), which may be aliphatic or alicyclic, and which may be saturated, partially unsaturated, or fully unsaturated. Thus, the term "alkylene" includes the sub-classes alkenylene, alkynylene, cycloalkylene, etc., discussed below.
Examples of linear saturated C3-i2 alkylene groups include, but are not limited to, -(CH2)n- where n is an integer from 3 to 12, for example, -CH2CH2CH2- (propylene),
-CH2CH2CH2CH2- (butylene), -CH2CH2CH2CH2CH2- (pentylene) and -CH2CH2CH2CH- 2CH2CH2CH2- (heptylene).
Examples of branched saturated C3-i2 alkylene groups include, but are not limited to, -CH(CH3)CH2-, -CH(CH3)CH2CH2-, -CH(CH3)CH2CH2CH2-, -CH2CH(CH3)CH2-,
-CH2CH(CH3)CH2CH2-, -CH(CH2CH3)-, -CH(CH2CH3)CH2-, and -CH2CH(CH2CH3)CH2-. Examples of linear partially unsaturated C3-12 alkylene groups (C3-12 alkenylene, and alkynylene groups) include, but are not limited to, -CH=CH-CH2-, -CH2-CH=CH2-,
-CH=CH-CH2-CH2-, -CH=CH-CH2-CH2-CH2-, -CH=CH-CH=CH-, -CH=CH-CH=CH-CH2-, - CH=CH-CH=CH-CH2-CH2-, -CH=CH-CH2-CH=CH-, -CH=CH-CH2-CH2-CH=CH-, and -CH2- C≡C-CH2-.
Examples of branched partially unsaturated C3-i2 alkylene groups (C3-i2 alkenylene and alkynylene groups) include, but are not limited to, -C(CH3)=CH-, -C(CH3)=CH-CH2-, -CH=CH-CH(CH3)- and -C≡C-CH(CH3)-. Examples of alicyclic saturated C3-i2 alkylene groups (C3-12 cycloalkylenes) include, but are not limited to, cyclopentylene (e.g. cyclopent-1 ,3-ylene), and cyclohexylene
(e.g. cyclohex-1 ,4-ylene).
Examples of alicyclic partially unsaturated C3-i2 alkylene groups (C3-12 cycloalkylenes) include, but are not limited to, cyclopentenylene (e.g. 4-cyclopenten-1 ,3-ylene), cyclohexenylene (e.g. 2-cyclohexen-1 ,4-ylene; 3-cyclohexen-1 ,2-ylene; 2,5-cyclohexadien- 1 ,4-ylene).
Oxygen protecting group: the term "oxygen protecting group" refers to a moiety which masks a hydroxy group, and these are well known in the art. A large number of suitable groups are described on pages 23 to 200 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference. Classes of particular interest include silyl ethers (e.g. TMS, TBDMS), substituted methyl ethers (e.g. THP) and esters (e.g. acetate).
Carbamate nitrogen protecting group: the term "carbamate nitrogen protecting group" pertains to a moiety which masks the nitrogen in the imine bond, and these are well known in the art. These groups have the following structure:
Figure imgf000023_0001
*
wherein R'10 is R as defined above. A large number of suitable groups are described on pages 503 to 549 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic
Synthesis, 3rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.
Hemi-aminal nitrogen protecting group: the term "hemi-aminal nitrogen protecting group" pertains to a group having the following
Figure imgf000023_0002
wherein R'10 is R as defined above. A large number of suitable groups are described on pages 633 to 647 as amide protecting groups of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference. Detailed Description of the Invention
The present invention provides Conjugates comprising a PBD dimer connected to a Ligand unit via a Linker Unit. In one embodiment, the Linker unit includes a Stretcher unit (A), a Specificity unit (L1), and a Spacer unit (L2). The Linker unit is connected at one end to the Ligand unit and at the other end to the PBD dimer compound.
In one aspect, such a Conjugate is shown below in formula la: L- (A1 a-L1 s-L2y-D)p (la)
wherein:
L is the Ligand unit; and
-A1a-L1 s-L2y- is a Linker unit (LU), wherein:
-A1- is a Stretcher unit,
a is 1 or 2,
L1 - is a Specificity unit,
s is an integer ranging from 1 to 12,
-L2- is a Spacer unit,
y is 0, 1 or 2;
-D is an PBD dimer; and
p is from 1 to 20.
The drug loading is represented by p, the number of drug molecules per Ligand unit (e.g., an antibody). Drug loading may range from 1 to 20 Drug units (D) per Ligand unit (e.g., Ab or mAb). For compositions, p represents the average drug loading of the Conjugates in the composition, and p ranges from 1 to 20.
In some embodiments, p is from about 1 to about 8 Drug units per Ligand unit. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is from about 2 to about 8 Drug units per Ligand unit. In some embodiments, p is from about 2 to about 6, 2 to about 5, or 2 to about 4 Drug units per Ligand unit. In some embodiments, p is about 2, about 4, about 6 or about 8 Drug units per Ligand unit.
The average number of Drugs units per Ligand unit in a preparation from a conjugation reaction may be characterized by conventional means such as mass spectroscopy, ELISA assay, and HPLC. The quantitative distribution of Conjugates in terms of p may also be determined. In some instances, separation, purification, and characterization of homogeneous Conjugates, where p is a certain value, from Conjugates with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis.
In another aspect, such a Conjugate is shown below in formula lb:
Figure imgf000025_0001
I
L - (A1 a- L2 y -D)p (lb)
Also illustrated as:
L - (A1 a- L2 y (- L1s) -D)p (lb) wherein:
L is the Ligand unit; and
-A1 a-L1s(L2y)- is a Linker unit (LU), wherein:
-A1- is a Stretcher unit linked to a Stretcher unit (L2),
a is 1 or 2,
L1 - is a Specificity unit linked to a Stretcher unit (L2),
s is an integer ranging from 0 to 12,
-L2- is a Spacer unit,
y is 0, 1 or 2;
-D is a PBD dimer; and
p is from 1 to 20.
Preferences
The following preferences may apply to all aspects of the invention as described above, or may relate to a single aspect. The preferences may be combined together in any combination.
In one embodiment, the Conjugate has the formula:
Figure imgf000025_0002
wherein L, A1, a, L1 , s, L2, D and p are as described above. In one embodiment, the Ligand unit (L) is a Cell Binding Agent (CBA) that specifically binds to a target molecule on the surface of a target cell. An exemplary formula is illustrated below:
Figure imgf000026_0001
where the asterisk indicates the point of attachment to the Drug unit (D), CBA is the
Cell Binding Agent, L1 is a Specificity unit, A1 is a Stretcher unit connecting L1 to the Cell Binding Agent, L2 is a Spacer unit, which is a covalent bond, a self-immolative group or together with -OC(=0)- forms a self-immolative group, and L2 optional.
In another embodiment, the Ligand unit (L) is a Cell Binding Agent (CBA) that specifically binds to a target molecule on the surface of a target cell. An exemplary formula is illustrated below:
Figure imgf000026_0002
where the asterisk indicates the point of attachment to the Drug unit (D), CBA is the Cell Binding Agent, L1 is a Specificity unit, A1 is a Stretcher unit connecting L1 to the Cell Binding Agent, L2 is a Spacer unit which is a covalent bond or a self-immolative group, and a is 1 or 2, s is 0, 1 or 2, and y is 0 or 1 or 2.
In the embodiments illustrated above, L1 can be a cleavable Specificity unit, and may be referred to as a "trigger" that when cleaved activates a self-immolative group (or self- immolative groups) L2, when a self-immolative group(s) is present. When the Specificity unit L1 is cleaved, or the linkage (i.e., the covalent bond) between L1 and L2 is cleaved, the self-immolative group releases the Drug unit (D).
In another embodiment, the Ligand unit (L) is a Cell Binding Agent (CBA) that specifically binds to a target molecule on the surface of a target cell. An exemplary formula is illustrated below:
Figure imgf000026_0003
where the asterisk indicates the point of attachment to the Drug (D), CBA is the Cell Binding Agent, L1 is a Specificity unit connected to L2, A1 is a Stretcher unit connecting L2 to the Cell Binding Agent, L2 is a self-immolative group, and a is 1 or2, s is 1 or 2, and y is 1 or 2.
In the various embodiments discussed herein, the nature of L1 and L2 can vary widely. These groups are chosen on the basis of their characteristics, which may be dictated in part, by the conditions at the site to which the conjugate is delivered. Where the Specificity unit L1 is cleavable, the structure and/or sequence of L1 is selected such that it is cleaved by the action of enzymes present at the target site (e.g., the target cell). L1 units that are cleavable by changes in pH (e.g. acid or base labile), temperature or upon irradiation (e.g. photolabile) may also be used. L1 units that are cleavable under reducing or oxidising conditions may also find use in the Conjugates.
In some embodiments, L1 may comprise one amino acid or a contiguous sequence of amino acids. The amino acid sequence may be the target substrate for an enzyme.
In one embodiment, L1 is cleavable by the action of an enzyme. In one embodiment, the enzyme is an esterase or a peptidase. For example, L1 may be cleaved by a lysosomal protease, such as a cathepsin. In one embodiment, L2 is present and together with -C(=0)0- forms a self-immolative group or self-immolative groups. In some embodiments, -C(=0)0- also is a self-immolative group.
In one embodiment, where L1 is cleavable by the action of an enzyme and L2 is present, the enzyme cleaves the bond between L1 and L2, whereby the self-immolative group(s) release the Drug unit.
L1 and L2, where present, may be connected by a bond selected from:
-C(=0)NH-,
-C(=0)0-,
-NHC(=0)-,
-OC(=0)-,
-OC(=0)0-,
-NHC(=0)0-,
-OC(=0)NH-,
-NHC(=0)NH, and
-O- (a glycosidic bond).
An amino group of L1 that connects to L2 may be the N-terminus of an amino acid or may be derived from an amino group of an amino acid side chain, for example a lysine amino acid side chain. A carboxyl group of L1 that connects to L2 may be the C-terminus of an amino acid or may be derived from a carboxyl group of an amino acid side chain, for example a glutamic acid amino acid side chain.
A hydroxy group of L1 that connects to L2 may be derived from a hydroxy group of an amino acid side chain, for example a serine amino acid side chain.
In one embodiment, -C(=0 2 together form the group:
Figure imgf000028_0001
where the asterisk indicates the point of attachment to the Drug unit, the wavy line indicates the point of attachment to the L1, Y is -N(H)-, -0-, -C(=0)N(H)- or -C(=0)0-, and n is 0 to 3. The phenylene ring is optionally substituted with one, two or three substituents as described herein.
In one embodiment, Y is NH.
In one embodiment, n is 0 or 1 . Preferably, n is 0.
Where Y is NH and n is 0, the self-immolative group may be referred to as a
p-aminobenzylcarbonyl linker (PABC).
The self-immolative group will allow for release of the Drug unit (i.e., the asymmetric PBD) when a remote site in the linker is activated, proceeding along the lines shown below (for n=0):
Figure imgf000028_0002
where the asterisk indicates the attachment to the Drug, L* is the activated form of the remaining portion of the linker and the released Drug unit is not shown. These groups have the advantage of separating the site of activation from the Drug. In another embodiment, -C(=0)0- and L2 together form a group selected from:
Figure imgf000029_0001
where the asterisk, the wavy line, Y, and n are as defined above. Each phenylene ring is optionally substituted with one, two or three substituents as described herein. In one embodiment, the phenylene ring having the Y substituent is optionally substituted and the phenylene ring not having the Y substituent is unsubstituted.
In another embodiment, -C =0)0- and L2 together form a group selected from:
Figure imgf000029_0002
where the asterisk, the wavy line, Y, and n are as defined above, E is O, S or NR, D is N, CH, or CR, and F is N, CH, or CR.
In one embodiment, D is N.
In one embodiment, D is CH.
In one embodiment, E is O or S.
In one embodiment, F is CH.
In a preferred embodiment, the covalent bond between L1 and L2 is a cathepsin labile (e.g., cleavable) bond.
In one embodiment, L1 comprises a dipeptide. The amino acids in the dipeptide may be any combination of natural amino acids and non-natural amino acids. In some
embodiments, the dipeptide comprises natural amino acids. Where the linker is a cathepsin labile linker, the dipeptide is the site of action for cathepsin-mediated cleavage. The dipeptide then is a recognition site for cathepsin. In one embodiment, the group -X X2- in dipeptide, -ΝΗ-XrX^CO-, is selected from:
-Phe-Lys-,
-Val-Ala-,
-Val-Lys-,
-Ala-Lys-,
-Val-Cit-,
-Phe-Cit-,
-Leu-Cit-,
-lle-Cit-,
-Phe-Arg-, and
-Trp-Cit-;
where Cit is citrulline. In such a dipeptide, -NH- is the amino group of Xi, and CO is the carbonyl group of X2. Preferably, the group -XrX2- in dipeptide, -NH-XrX2-CO-, is selected from:
-Phe-Lys-,
-Val-Ala-,
-Val-Lys-,
-Ala-Lys-, and
-Val-Cit-.
Most preferably, the group -XrX2- in dipeptide, -NH-X X2-CO-, is -Phe-Lys-, Val-Cit or -Val-Ala-. Other dipeptide combinations of interest include:
-Gly-Gly-,
-Pro-Pro-, and
-Val-Glu-.
Other dipeptide combinations may be used, including those described by Dubowchik et al., which is incorporated herein by reference.
In one embodiment, the amino acid side chain is chemically protected, where appropriate. The side chain protecting group may be a group as discussed below. Protected amino acid sequences are cleavable by enzymes. For example, a dipeptide sequence comprising a Boc side chain-protected Lys residue is cleavable by cathepsin.
Protecting groups for the side chains of amino acids are well known in the art and are described in the Novabiochem Catalog. Additional protecting group strategies are set out in Protective groups in Organic Synthesis, Greene and Wuts. Possible side chain protecting groups are shown below for those amino acids having reactive side chain functionality:
Arg: Z, Mtr, Tos;
Asn: Trt, Xan;
Asp: Bzl, t-Bu;
Cys: Acm, Bzl, Bzl-OMe, Bzl-Me, Trt;
Glu: Bzl, t-Bu;
Gin: Trt, Xan;
His: Boc, Dnp, Tos, Trt;
Lys: Boc, Z-CI, Fmoc, Z;
Ser: Bzl, TBDMS, TBDPS;
Thr: Bz;
Trp: Boc;
Tyr: Bzl, Z, Z-Br.
In one embodiment, -X2- is connected indirectly to the Drug unit. In such an embodiment, the Spacer unit L2 is present.
In one embodiment, the dipeptide is used in combination with a self-immolative group(s) (the Spacer unit). The self-immolative group(s) may be connected to -X2-.
Where a self-immolative group is present, -X2- is connected directly to the self-immolative group. In one embodiment, -X2- is connected to the group Y of the self-immolative group. Preferably the group -X2-CO- is connected to Y, where Y is NH.
-X is connected directly to A1. In one embodiment, -Xr is connected directly to A1. Preferably the group NH-X-i- (the amino terminus of X-i) is connected to A1. A1 may comprise the functionality -CO- thereby to form an amide link with -X . In one embodiment, L1 and L2 together with -OC(=0)- comprise the group -X X2-PABC-. The PABC group is connected directly to the Drug unit. In one example, the self- immolative group and the dipeptide together form the group -Phe-Lys-PABC-, which is illustrated below:
Figure imgf000032_0001
where the asterisk indicates the point of attachment to the Drug unit, and the wavy line indicates the point of attachment to the remaining portion of L1 or the point of attachment to A1. Preferably, the wavy line indicates the point of attachment to A1.
Alternatively, the self-immolative group and the dipeptide together form the group -Val-Ala- PABC-, which is illustrated below:
Figure imgf000032_0002
where the asterisk and the wavy line are as defined above. In another embodiment, L1 and L2 together with -OC(=0)- represent:
Figure imgf000032_0003
where the asterisk indicates the point of attachment to the Drug unit, the wavy line indicates the point of attachment to A1, Y is a covalent bond or a functional group, and E is a group that is susceptible to cleavage thereby to activate a self-immolative group.
E is selected such that the group is susceptible to cleavage, e.g., by light or by the action of an enzyme. E may be -N02 or glucuronic acid (e.g., β-glucuronic acid). The former may be susceptible to the action of a nitroreductase, the latter to the action of a
β-glucuronidase.
The group Y may be a covalent bond.
The group Y may be a functional group selected from:
-C(=0)- -NH- -O-
-C(=0)NH-,
-C(=0)0-,
-NHC(=0)-,
-OC(=0)-,
-OC(=0)0-,
-NHC(=0)0-,
-OC(=0)NH-,
-NHC(=0)NH-,
-NHC(=0)NH,
-C(=0)NHC(=0)-,
S02, and
-S-.
The group Y is preferably -NH-, -CH2-, -0-, and -S-.
In some embodiments, L1 and L2 together with -OC(=0)- represent:
Figure imgf000033_0001
where the asterisk indicates the point of attachment to the Drug unit, the wavy line indicates the point of attachment to A, Y is a covalent bond or a functional group and E is glucuronic acid (e.g., β-glucuronic acid). Y is preferably a functional group selected from -NH-.
In some embodiments, L1 and L2 together represent:
Figure imgf000033_0002
where the asterisk indicates the point of attachment to the remainder of L2 or the Drug unit, the wavy line indicates the point of attachment to A1, Y is a covalent bond or a functional group and E is glucuronic acid (e.g., β-glucuronic acid). Y is preferably a functional group selected from -NH-, -CH2-, -0-, and -S-. In some further embodiments, Y is a functional group as set forth above, the functional group is linked to an amino acid, and the amino acid is linked to the Stretcher unit A1. In some embodiments, amino acid is β-alanine. In such an embodiment, the amino acid is equivalently considered part of the Stretcher unit.
The Specificity unit L1 and the Ligand unit are indirectly connected via the Stretcher unit.
L1 and A1 may be connected by a bond selected from:
-C(=0)NH-,
-C(=0)0-,
-NHC(=0)-,
-OC(=0)-,
-OC(=0)0-,
-NHC(=0)0-,
-OC(=0)NH-, and
-NHC(=0)NH-.
In one embodiment, the group A1 is:
Figure imgf000034_0001
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group A1
Figure imgf000034_0002
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group A1 is:
Figure imgf000034_0003
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the group A1 is:
Figure imgf000035_0001
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the group 1 is:
Figure imgf000035_0002
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group A1 is:
Figure imgf000035_0003
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group A1 is:
Figure imgf000035_0005
Figure imgf000035_0004
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8. In one embodiment, the group A1 is:
Figure imgf000036_0001
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the connection between the Ligand unit and A1 is through a thiol residue of the Ligand unit and a maleimide group of A1.
In one embodiment, the connection between the Ligand unit and A1 is:
Figure imgf000036_0002
where the asterisk indicates the point of attachment to the remaining portion of A1 , L1, L2 or D, and the wavy line indicates the point of attachment to the remaining portion of the Ligand unit. In this embodiment, the S atom is typically derived from the Ligand unit.
In each of the embodiments above, an alternative functionality may be used in place of the malemide-derived group shown below:
Figure imgf000036_0003
where the wavy line indicates the point of attachment to the Ligand unit as before, and the asterisk indicates the bond to the remaining portion of the A1 group, or to L1, L2 or D.
In one embodiment, the maleimide-derived group is replaced with the group:
Figure imgf000036_0004
where the wavy line indicates point of attachment to the Ligand unit, and the asterisk indicates the bond to the remaining portion of the A1 group , or to L1, L2 or D.
In one embodiment, the maleimide-derived group is replaced with a group, which optionally together with a Ligand unit (e.g., a Cell Binding Agent), is selected from:
-C(=0)NH-,
-C(=0)0-,
-NHC(=0)-,
-OC(=0)-,
-OC(=0)0-,
-NHC(=0)0-,
-OC(=0)NH-,
-NHC(=0)NH-,
-NHC(=0)NH,
-C(=0)NHC(=0)-,
-S-,
-S-S-,
-CH2C(=0)- -C(=0)CH2-,
=N-NH-, and
-NH-N=.
In one embodiment, the maleimide-derived group is replaced with a group, which optionally together with the Ligand unit, is s
Figure imgf000037_0001
where the wavy line indicates either the point of attachment to the Ligand unit or the bond to the remaining portion of the A1 group, and the asterisk indicates the other of the point of attachment to the Ligand unit or the bond to the remaining portion of the A1 group. Other groups suitable for connecting L1 to the Cell Binding Agent are described in
WO 2005/082023.
In one embodiment, the Stretcher unit A1 is present, the Specificity unit L1 is present and Spacer unit L2 is absent. Thus, L1 and the Drug unit are directly connected via a bond. Equivalently in this embodiment, L2 is a bond.
L1 and D may be connected by a bond selected from: -C(=0)NH-,
-C(=0)0-,
-NHC(=0)-,
-OC(=0)-,
-OC(=0)0-,
-NHC(=0)0-,
-OC(=0)NH-, and
-NHC(=0)NH-. In one embodiment, L1 and D are preferably connected by a bond selected from:
-C(=0)NH-, and
-NHC(=0)-.
In one embodiment, L1 comprises a dipeptide and one end of the dipeptide is linked to D. As described above, the amino acids in the dipeptide may be any combination of natural amino acids and non-natural amino acids. In some embodiments, the dipeptide comprises natural amino acids. Where the linker is a cathepsin labile linker, the dipeptide is the site of action for cathepsin-mediated cleavage. The dipeptide then is a recognition site for cathepsin.
In one embodiment, the group -X X2- in dipeptide,
Figure imgf000038_0001
is selected from:
-Phe-Lys-,
-Val-Ala-,
-Val-Lys-,
-Ala-Lys-,
-Val-Cit-,
-Phe-Cit-,
-Leu-Cit-,
-lle-Cit-,
-Phe-Arg-, and
-Trp-Cit-;
where Cit is citrulline. In such a dipeptide, -NH- is the amino group of Xi, and CO is the carbonyl group of X2. Preferably, the group -X!-X2- in dipeptide,
Figure imgf000038_0002
is selected from:
-Phe-Lys-,
-Val-Ala-,
-Val-Lys-,
-Ala-Lys-, and
-Val-Cit-. Most preferably, the group -X!-X2- in dipeptide, -NH-XrX2-CO-, is -Phe-Lys- or -Val-Ala-.
Other dipeptide combinations of interest include:
-Gly-Gly-,
-Pro-Pro-, and
-Val-Glu-.
Other dipeptide combinations may be used, including those described above. In one embodiment, L1-D is:
/ -NH-X Xz-CO-NH- * where
Figure imgf000039_0001
is the dipeptide, -NH- is part of the Drug unit, the asterisk indicates the point of attachment to the remainder of the Drug unit, and the wavy line indicates the point of attachment to the remaining portion of L1 or the point of attachment to A1. Preferably, the wavy line indicates the point of attachment to A1.
In one embodiment, the dipeptide is valine-alanine and L1-D is:
Figure imgf000039_0002
where the asterisk, -NH- and the wavy line are as defined above. In one embodiment, the dipeptide is phenylalanine-lysine and L1-D is:
Figure imgf000039_0003
where the asterisk, -NH- and the wavy line are as defined above. In one embodiment, the dipeptide is valine-citrulline. In one embodiment, the groups A1
Figure imgf000040_0001
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, is 5.
In one embodiment, the groups A1-L1 are:
Figure imgf000040_0002
where the asterisk indicates the point of attachment to D, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the groups A1-L1 are:
Figure imgf000040_0003
where the asterisk indicates the point of attachment to D, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the groups A1-L1 are:
Figure imgf000040_0004
where the asterisk indicates the point of attachment to D, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 7, preferably 3 to 7, most preferably 3 or 7. In one embodiment, the groups A1
Figure imgf000041_0001
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the groups A1-L1 are:
Figure imgf000041_0002
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the groups A1
Figure imgf000041_0005
Figure imgf000041_0003
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the groups A1
Figure imgf000041_0004
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the groups L- A1-L1 are:
Figure imgf000042_0001
where the asterisk indicates the point of attachment to D, S is a sulphur group of the Ligand unit, the wavy line indicates the point of attachment to the rest of the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group L-A1-L1 are:
Figure imgf000042_0002
where the asterisk indicates the point of attachment to D, S is a sulphur group of the Ligand unit, the wavy line indicates the point of attachment to the remainder of the Ligand unit, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the groups L-A1-L1 are:
Figure imgf000042_0003
where the asterisk indicates the point of attachment to D, S is a sulphur group of the Ligand unit, the wavy line indicates the point of attachment to the remainder of the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the groups L-A1-L1 are:
Figure imgf000042_0004
where the asterisk indicates the point of attachment to D, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 7, preferably 4 to 8, most preferably 4 or 8. In one embodiment, the groups L-A1-L1 are:
Figure imgf000043_0001
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the remainder of the Ligand unit, and n is 0 to 6.
embodiment, n is 5.
In one embodiment, the groups L-A1-L1 are:
Figure imgf000043_0002
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the remainder of the Ligand unit, and n is 0 to 6.
embodiment, n is 5.
In one embodiment, the groups L-A1-L1 are:
Figure imgf000043_0003
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the remainder of the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the groups L-A1
Figure imgf000044_0001
where the asterisk indicates the point of attachment to L2 or D, the wavy line indicates the point of attachment to the remainder of the Ligand unit, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, most preferably 4 or 8.
In one embodiment, the Stretcher unit is an acetamide unit, having the formula:
Figure imgf000044_0002
where the asterisk indicates the point of attachment to the remainder of the
Stretcher unit, L1 or D, and the wavy line indicates the point of attachment to the Ligand unit.
In other embodiments, Linker-Drug compounds are provided for conjugation to a Ligand unit. In one embodiment, the Linker-Drug compounds are designed for connection to a Cell Binding Agent.
In one embodiment, the Drug Linker formula:
Figure imgf000044_0003
where the asterisk indicates the point of attachment to the Drug unit, G1 is a Stretcher group (A1) to form a connection to a Ligand unit, L1 is a Specificity unit, L2 (a Spacer unit) is a covalent bond or together with -OC(=0)- forms a self-immolative group(s).
In another embodiment, the Drug Linker compound has the formula:
G1-L1-L2- . where the asterisk indicates the point of attachment to the Drug unit, G1 is a Stretcher unit (A1) to form a connection to a Ligand unit, L1 is a Specificity unit, L2 (a Spacer unit) is a covalent bond or a self-immolative group(s).
L1 and L2 are as defined above. References to connection to A1 can be construed here as referring to a connection to G1. In one embodiment, where L1 comprises an amino acid, the side chain of that amino acid may be protected. Any suitable protecting group may be used. In one embodiment, the side chain protecting groups are removable with other protecting groups in the compound, where present. In other embodiments, the protecting groups may be orthogonal to other protecting groups in the molecule, where present.
Suitable protecting groups for amino acid side chains include those groups described in the Novabiochem Catalog 2006/2007. Protecting groups for use in a cathepsin labile linker are also discussed in Dubowchik et al.
In certain embodiments of the invention, the group L1 includes a Lys amino acid residue. The side chain of this amino acid may be protected with a Boc or Alloc protected group. A Boc protecting group is most preferred.
The functional group G1 forms a connecting group upon reaction with a Ligand unit (e.g., a cell binding agent.
In one embodiment, the functional group G1 is or comprises an amino, carboxylic acid, hydroxy, thiol, or maleimide group for reaction with an appropriate group on the Ligand unit. In a preferred embodiment, G1 comprises a maleimide group.
In one embodiment, the group G1 is an alkyl maleimide group. This group is suitable for reaction with thiol groups, particularly cysteine thiol groups, present in the cell binding agent, for example present in an antibody.
In one embodiment, the group G1 is:
Figure imgf000045_0001
where the asterisk indicates the point of attachment to L1, L2 or D, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group G1 is:
Figure imgf000045_0002
where the asterisk indicates the point of attachment to L1, L2 or D, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group G1 is:
Figure imgf000046_0001
where the asterisk indicates the point of attachment to L1, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 2, preferably 4 to 8, and most preferably 4 or 8.
In one embodiment, the group G1 is:
Figure imgf000046_0002
where the asterisk indicates the point of attachment to L1, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, and most preferably 4 or 8.
In one embodiment, the group G1
Figure imgf000046_0003
where the asterisk indicates the point of attachment to L1, L2 or D, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group G1 is:
Figure imgf000046_0004
where the asterisk indicates the point of attachment to L1, L2 or D, and n is 0 to 6. In one embodiment, n is 5.
In one embodiment, the group G1 is:
where the asterisk indicates the point of attachment to L1, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 2, preferably 4 to 8, and most preferably 4 or 8.
In one embodiment, the roup G1 is:
Figure imgf000047_0005
Figure imgf000047_0002
where the asterisk indicates the point of attachment to L1, n is 0 or 1 , and m is 0 to 30. In a preferred embodiment, n is 1 and m is 0 to 10, 1 to 8, preferably 4 to 8, and most preferably 4 or 8.
In each of the embodiments above, an alternative functionality may be used in place of the malemide group shown below:
Figure imgf000047_0003
where the asterisk indicates the bond to the remaining portion of the G
In one embodiment, the maleimide-derived group is replaced with the group:
Figure imgf000047_0004
where the asterisk indicates the bond to the remaining portion of the G group.
In one embodiment, the maleimide group is replaced with a group selected from:
-C(=0)OH,
-OH,
-NH2,
-SH, -C(=0)CH2X, where X is CI, Br or I,
-CHO, -C≡CH, and
-N3 (azide).
In one embodiment, L1 is present, and G1 is -NH2, -NHMe, -COOH, -OH or -SH.
In one embodiment, where L1 is present, G1 is -NH2 or -NHMe. Either group may be the N-terminal of an L1 amino acid sequence.
In one embodiment, L1 is present and G1 is -NH2, and L1 is an amino acid sequence -X X2- , as defined above. In one embodiment, L1 is present and G1 is COOH. This group may be the C-terminal of an L1 amino acid sequence.
In one embodiment, L1 is present and G1 is OH.
In one embodiment, L1 is present and G1 is SH.
The group G1 may be convertible from one functional group to another. In one
embodiment, L1 is present and G1 is -NH2. This group is convertable to another group G1 comprising a maleimide group. For example, the group -NH2 may be reacted with an acids or an activated acid (e.g., N-succinimide forms) of those G1 groups comprising maleimide shown above.
The group G1 may therefore be converted to a functional group that is more appropriate for reaction with a Ligand unit. As noted above, in one embodiment, L1 is present and G1 is -NH2, -NHMe, -COOH, -OH or -SH. In a further embodiment, these groups are provided in a chemically protected form. The chemically protected form is therefore a precursor to the linker that is provided with a functional group. In one embodiment, G1 is -NH2 in a chemically protected form. The group may be protected with a carbamate protecting group. The carbamate protecting group may be selected from the group consisting of:
Alloc, Fmoc, Boc, Troc, Teoc, Cbz and PNZ.
Preferably, where G1 is -NH2, it is protected with an Alloc or Fmoc group.
In one embodiment, where G1 is -NH2, it is protected with an Fmoc group. In one embodiment, the protecting group is the same as the carbamate protecting group of the capping group. In one embodiment, the protecting group is not the same as the carbamate protecting group of the capping group. In this embodiment, it is preferred that the protecting group is removable under conditions that do not remove the carbamate protecting group of the capping group. The chemical protecting group may be removed to provide a functional group to form a connection to a Ligand unit. Optionally, this functional group may then be converted to another functional group as described above.
In one embodiment, the active group is an amine. This amine is preferably the N-terminal amine of a peptide, and may be the N-terminal amine of the preferred dipeptides of the invention.
The active group may be reacted to yield the functional group that is intended to form a connection to a Ligand unit.
In other embodiments, the Linker unit is a precursor to the Linker unit having an active group. In this embodiment, the Linker unit comprises the active group, which is protected by way of a protecting group. The protecting group may be removed to provide the Linker unit having an active group.
Where the active group is an amine, the protecting group may be an amine protecting group, such as those described in Green and Wuts.
The protecting group is preferably orthogonal to other protecting groups, where present, in the Linker unit.
In one embodiment, the protecting group is orthogonal to the capping group. Thus, the active group protecting group is removable whilst retaining the capping group. In other embodiments, the protecting group and the capping group is removable under the same conditions as those used to remove the capping group.
In one embodiment, the Linker unit is:
Figure imgf000050_0001
where the asterisk indicates the point of attachment to the Drug unit, and the wavy line indicates the point of attachment to the remaining portion of the Linker unit, as applicable or the point of attachment to G1. Preferably, the wavy line indicates the point of attachment to G1.
In one embodiment, the Linker unit is:
Figure imgf000050_0002
where the asterisk and the wavy line are as defined above.
Other functional groups suitable for use in forming a connection between L1 and the Cell Binding Agent are described in WO 2005/082023.
Ligand Unit
The Ligand Unit may be of any kind, and include a protein, polypeptide, peptide and a non- peptidic agent that specifically binds to a target molecule. In some embodiments, the Ligand unit may be a protein, polypeptide or peptide. In some embodiments, the Ligand unit may be a cyclic polypeptide. These Ligand units can include antibodies or a fragment of an antibody that contains at least one target molecule-binding site, lymphokines, hormones, growth factors, or any other cell binding molecule or substance that can specifically bind to a target.
Examples of Ligand units include those agents described for use in WO 2007/085930, which is incorporated herein.
In some embodiments, the Ligand unit is a Cell Binding Agent that binds to an extracellular target on a cell. Such a Cell Binding Agent can be a protein, polypeptide, peptide or a non- peptidic agent. In some embodiments, the Cell Binding Agent may be a protein, polypeptide or peptide. In some embodiments, the Cell Binding Agent may be a cyclic polypeptide. The Cell Binding Agent also may be antibody or an antigen-binding fragment of an antibody. Thus, in one embodiment, the present invention provides an antibody-drug conjugate (ADC). In one embodiment the antibody is a monoclonal antibody; chimeric antibody; humanized antibody; fully human antibody; or a single chain antibody. One embodiment the antibody is a fragment of one of these antibodies having biological activity. Examples of such fragments include Fab, Fab', F(ab')2 and Fv fragments. The antibody may be a diabody, a domain antibody (DAB) or a single chain antibody.
In one embodiment, the antibody is a monoclonal antibody.
Antibodies for use in the present invention include those antibodies described in
WO 2005/082023 which is incorporated herein. Particularly preferred are those antibodies for tumour-associated antigens. Examples of those antigens known in the art include, but are not limited to, those tumour-associated antigens set out in WO 2005/082023. See, for instance, pages 41 -55. In some embodiments, the conjugates are designed to target tumour cells via their cell surface antigens. The antigens may be cell surface antigens which are either over- expressed or expressed at abnormal times or cell types. Preferably, the target antigen is expressed only on proliferative cells (preferably tumour cells); however this is rarely observed in practice. As a result, target antigens are usually selected on the basis of differential expression between proliferative and healthy tissue.
Antibodies have been raised to target specific tumour related antigens including:
Cripto, CD19, CD20, CD22, CD30, CD33, Glycoprotein NMB, CanAg, Her2 (ErbB2/Neu), CD56 (NCAM), CD70, CD79, CD138, PSCA, PSMA (prostate specific membrane antigen), BCMA, E-selectin, EphB2, Melanotransferin, Mud 6 and TMEFF2.
The Ligand unit is connected to the Linker unit. In one embodiment, the Ligand unit is connected to A, where present, of the Linker unit. In one embodiment, the connection between the Ligand unit and the Linker unit is through a thioether bond.
In one embodiment, the connection between the Ligand unit and the Linker unit is through a disulfide bond.
In one embodiment, the connection between the Ligand unit and the Linker unit is through an amide bond. In one embodiment, the connection between the Ligand unit and the Linker unit is through an ester bond.
In one embodiment, the connection between the Ligand unit and the Linker is formed between a thiol group of a cysteine residue of the Ligand unit and a maleimide group of the Linker unit.
The cysteine residues of the Ligand unit may be available for reaction with the functional group of the Linker unit to form a connection. In other embodiments, for example where the Ligand unit is an antibody, the thiol groups of the antibody may participate in interchain disulfide bonds. These interchain bonds may be converted to free thiol groups by e.g. treatment of the antibody with DTT prior to reaction with the functional group of the Linker unit. In some embodiments, the cysteine residue is an introduced into the heavy or light chain of an antibody. Positions for cysteine insertion by substitution in antibody heavy or light chains include those described in Published U.S. Application No. 2007-0092940 and International Patent Publication WO2008070593, which are incorporated herein. Methods of Treatment
The Conjugates of the present invention may be used in a method of therapy. Also provided is a method of treatment, comprising administering to a subject in need of treatment a therapeutically-effective amount of a Conjugate of formula I. The term
"therapeutically effective amount" is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The actual amount of a Conjugate administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors. In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 10 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.05 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 4 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.05 to about 3 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 3 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 2 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 1 mg/kg per dose.
A conjugate may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. Examples of treatments and therapies include, but are not limited to, chemotherapy (the administration of active agents, including, e.g. drugs; surgery; and radiation therapy).
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
Conjugate 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. A capsule may comprise a solid carrier such a 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 Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
Includes Other Forms
Unless otherwise specified, included in the above are the well known ionic, salt, solvate, and protected forms of these substituents. For example, a reference to carboxylic acid (-COOH) also includes the anionic (carboxylate) form (-COO"), a salt or solvate thereof, as well as conventional protected forms. Similarly, a reference to an amino group includes the protonated form (-N+HR1R2), a salt or solvate of the amino group, for example, a hydrochloride salt, as well as conventional protected forms of an amino group. Similarly, a reference to a hydroxyl group also includes the anionic form (-0"), a salt or solvate thereof, as well as conventional protected forms.
Salts
It may be convenient or desirable to prepare, purify, and/or handle a corresponding salt of the active compound (the Conjugate), for example, a pharmaceutically-acceptable salt. Examples of pharmaceutically acceptable salts are discussed in Berge, et al., J. Pharm. Sci., 66, 1-19 (1977).
For example, if the compound is anionic, or has a functional group which may be anionic (e.g. -COOH may be -COO"), then a salt may be formed with a suitable cation. Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such as Na+ and K+, alkaline earth cations such as Ca2+ and Mg2+, and other cations such as Α 3. Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e. NH4 +) and substituted ammonium ions (e.g. NH3R+, NH2R2+, NHR3 +, NR4 +). Examples of some suitable substituted ammonium ions are those derived from: ethylamine,
diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine,
ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH3)4 +.
If the Conjugate is cationic, or has a functional group which may be cationic (e.g. -NH2 may be -NH3 +), then a salt may be formed with a suitable anion. Examples of suitable inorganic anions include, but are not limited to, those derived from the following inorganic acids: hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfurous, nitric, nitrous, phosphoric, and phosphorous.
Examples of suitable organic anions include, but are not limited to, those derived from the following organic acids: 2-acetyoxybenzoic, acetic, ascorbic, aspartic, benzoic,
camphorsulfonic, cinnamic, citric, edetic, ethanedisulfonic, ethanesulfonic, fumaric, glucheptonic, gluconic, glutamic, glycolic, hydroxymaleic, hydroxynaphthalene carboxylic, isethionic, lactic, lactobionic, lauric, maleic, malic, methanesulfonic, mucic, oleic, oxalic, palmitic, pamoic, pantothenic, phenylacetic, phenylsulfonic, propionic, pyruvic, salicylic, stearic, succinic, sulfanilic, tartaric, toluenesulfonic, and valeric. Examples of suitable polymeric organic anions include, but are not limited to, those derived from the following polymeric acids: tannic acid, carboxymethyl cellulose.
Solvates
It may be convenient or desirable to prepare, purify, and/or handle a corresponding solvate of the Conjugate(s). The term "solvate" is used herein in the conventional sense to refer to a complex of solute (e.g. active Conjugate, salt of active Conjugate) and solvent. If the solvent is water, the solvate may be conveniently referred to as a hydrate, for example, a mono-hydrate, a di-hydrate, a tri-hydrate, etc.
Carbinolamines
The invention includes Conjugate where a solvent adds across the imine bond of the PBD moiety, which is illustrated below for a PBD monomer where the solvent is water or an alcohol (RAOH, where RA is d-4 alkyl):
Figure imgf000055_0001
These forms can be called the carbinolamine and carbinolamine ether forms of the PBD. The balance of these equilibria depend on the conditions in which the compounds are found, as well as the nature of the moiety itself.
These particular compounds may be isolated in solid form, for example, by lyophilisation. Isomers
Certain compounds may exist in one or more particular geometric, optical, enantiomeric, diasteriomeric, epimeric, atropic, stereoisomeric, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans-forms; E- and Z-forms; c-, t-, and r- forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and l-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal- and anticlinal-forms; a- and β-forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and halfchair- forms; and combinations thereof, hereinafter collectively referred to as "isomers" (or "isomeric forms").
Note that, except as discussed below for tautomeric forms, specifically excluded from the term "isomers", as used herein, are structural (or constitutional) isomers (i.e. isomers which differ in the connections between atoms rather than merely by the position of atoms in space). For example, a reference to a methoxy group, -OCH3, is not to be construed as a reference to its structural isomer, a hydroxymethyl group, -CH2OH. Similarly, a reference to ortho-chlorophenyl is not to be construed as a reference to its structural isomer, meta- chlorophenyl. However, a reference to a class of structures may well include structurally isomeric forms falling within that class (e.g. C1-7 alkyl includes n-propyl and iso-propyl; butyl includes n-, iso-, sec-, and tert-butyl; methoxyphenyl includes ortho-, meta-, and para- methoxyphenyl).
The above exclusion does not pertain to tautomeric forms, for example, keto-, enol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol (illustrated below), imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime,
thioketone/enethiol, N-nitroso/hyroxyazo, and nitro/aci-nitro.
Figure imgf000056_0001
keto enol enolate
Note that specifically included in the term "isomer" are compounds with one or more isotopic substitutions. For example, H may be in any isotopic form, including 1H, 2H (D), and 3H (T); C may be in any isotopic form, including 12C, 13C, and 14C; O may be in any isotopic form, including 160 and 180; and the like.
Unless otherwise specified, a reference to a particular compound or Conjugate includes all such isomeric forms, including (wholly or partially) racemic and other mixtures thereof. Methods for the preparation (e.g. asymmetric synthesis) and separation (e.g. fractional crystallisation and chromatographic means) of such isomeric forms are either known in the art or are readily obtained by adapting the methods taught herein, or known methods, in a known manner. General synthetic routes
The synthesis of PBD dimer compounds is extensively discussed in the following references, which discussions are incorporated herein by reference:
a) WO 00/12508 (pages 14 to 30);
b) WO 2005/023814 (pages 3 to 10);
c) WO 2004/043963 (pages 28 to 29); and
d) WO 2005/085251 (pages 30 to 39).
Synthesis route
The Conjugates of the present invention, where R10 and R11 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound, can be synthesised from a co
Formula 2
Figure imgf000057_0001
where R2, R6, R7, R9, R6', R7', R9', R12, X, X' and R" are as defined for compounds of formula II, ProtN is a nitrogen protecting group for synthesis and Prot° is a protected oxygen group for synthesis or an oxo group, by deprotecting the imine bond by standard methods.
The compound produced may be in its carbinolamine or carbinolamine ether form depending on the solvents used. For example if ProtN is Alloc and Prot° is an oxygen protecting group for synthesis, then the deprotection is carried using palladium to remove the N10 protecting group, followed by the elimination of the oxygen protecting group for synthesis. If ProtN is Troc and Prot° is an oxygen protecting group for synthesis, then the deprotection is carried out using a Cd/Pb couple to yield the compound of formula (I). If ProtN is SEM, or an analogous group, and Prot° is an oxo group, then the oxo group can be removed by reduction, which leads to a protected carbinolamine intermediate, which can then be treated to remove the SEM protecting group, followed by the elimination of water. The reduction of the compound of Compound formula 2 can be accomplished by, for example, lithium tetraborohydride, whilst a suitable means for removing the SEM protecting group is treatment with silica gel. Compounds of Compound formula 2 can be synthesised from a compound of Compound formula 3
Formula 3a
Figure imgf000058_0001
where R2, R6, R7, R9, R6', R7', R9', X, X' and R" are as defined for compounds of Compound formula 2, by coupling an organometallic derivative comprising R12, such as an
organoboron derivative. The organoboron derivative may be a boronate or boronic acid.
Compounds of Compound formula 2 can be synthesised from a compound of Compound formula 3
Formula 3b
Figure imgf000058_0002
where R12, R6, R7, R9, R6', R7', R9', X, X' and R" are as defined for compounds of
Compound formula 2, by coupling an organometallic derivative comprising R2, such as an organoboron derivative. The organoboron derivative may be a boronate or boronic acid.
Compounds of Compound formulae 3a and 3b can be synthesised from a compound of formula 4:
Formula 4
Figure imgf000058_0003
where R2, R6, R7, R9, R6', R7', R9', X, X' and R" are as defined for compounds of Compound formula 2, by coupling about a single equivalent (e.g. 0.9 or 1 to 1.1 or 1.2) of an organometallic derivative, such as an organoboron derivative, comprising R2 or R12.
The couplings described above are usually carried out in the presence of a palladium catalyst, for example Pd(PPh3)4, Pd(OCOCH3)2, PdCI2, or Pd2(dba)3. The coupling may be carried out under standard conditions, or may also be carried out under microwave conditions.
The two coupling steps are usually carried out sequentially. They may be carried out with or without purification between the two steps. If no purification is carried out, then the two steps may be carried out in the same reaction vessel. Purification is usually required after the second coupling step. Purification of the compound from the undesired by-products may be carried out by column chromatography or ion-exchange separation.
The synthesis of compounds of Compound formula 4 where Prot° is an oxo group and ProtN is SEM are described in detail in WO 00/12508, which is incorporated herein by reference. In particular, reference is made to scheme 7 on page 24, where the above compound is designated as intermediate P. This method of synthesis is also described in WO 2004/043963.
The synthesis of compounds of Compound formula 4 where Prot° is a protected oxygen group for synthesis are described in WO 2005/085251 , which synthesis is herein incorporated by reference.
Compounds of formula I where R10 and R10 are H and R11 and R11 are SOzM, can be synthesised from compounds of formula I where R10 and R11 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound, by the addition of the appropriate bisulphite salt or sulphinate salt, followed by an appropriate purification step. Further methods are described in GB 2 053 894, which is herein incorporated by reference.
Nitrogen protecting groups for synthesis
Nitrogen protecting groups for synthesis are well known in the art. In the present invention, the protecting groups of particular interest are carbamate nitrogen protecting groups and hemi-aminal nitrogen protecting groups.
Carbamate nitrogen protecting groups have the following structure:
.0 wherein R'10 is R as defined above. A large number of suitable groups are described on pages 503 to 549 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic
Synthesis, 3rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.
Particularly preferred protecting groups include Troc, Teoc, Fmoc, BOC, Doc, Hoc, TcBOC, 1 -Adoc and 2-Adoc.
Other possible groups are nitrobenzyloxycarbonyl (e.g. 4- nitrobenzyloxycarbonyl) and 2- (phenylsulphonyl)ethoxycarbonyl.
Those protecting groups which can be removed with palladium catalysis are not preferred, e.g. Alloc. aminal nitrogen protecting groups llowing structure
Figure imgf000060_0001
*
wherein R'10 is R as defined above. A large number of suitable groups are described on pages 633 to 647 as amide protecting groups of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference. The groups disclosed herein can be applied to compounds for use in the present invention. Such groups include, but are not limited to, SEM, MOM, MTM, MEM, BOM, nitro or methoxy substituted BOM, and CI3CCH2OCH2-. Protected oxygen group for synthesis
Protected oxygen group for synthesis are well known in the art. A large number of suitable oxygen protecting groups are described on pages 23 to 200 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.
Classes of particular interest include silyl ethers, methyl ethers, alkyl ethers, benzyl ethers, esters, acetates, benzoates, carbonates, and sulfonates.
Preferred oxygen protecting groups include acetates, TBS and THP. Further Preferences
The following preferences may apply to all aspects of the invention as described above, or may relate to a single aspect. The preferences may be combined together in any combination.
In some embodiments, R6', R7', R9', R10', R11' and Y' are preferably the same as R6, R7, R9, R10, R11 and Y respectively. Dimer link
Y and Y' are preferably O.
R" is preferably a C3-7 alkylene group with no substituents. More preferably R" is a C3, C5 or C7 alkylene.
R6 to R9
R9 is preferably H.
R6 is preferably selected from H, OH, OR, SH, NH2, nitro and halo, and is more preferably H or halo, and most preferably is H.
R7 is preferably selected from H, OH, OR, SH, SR, NH2, NHR, NRR', and halo, and more preferably independently selected from H, OH and OR, where R is preferably selected from optionally substituted C1-7 alkyl, C3-10 heterocyclyl and C5-10 aryl groups. R may be more preferably a C1-4 alkyl group, which may or may not be substituted. A substituent of interest is a C5-6 aryl group (e.g. phenyl). Particularly preferred substituents at the 7- positions are OMe and OCH2Ph.
These preferences apply to R9 , R6 and R7 respectively.
R2
A in R2 may be phenyl group or a C5-7 heteroaryl group, for example furanyl, thiophenyl and pyridyl. In some embodiments, A is preferably phenyl. In other embodiments, A is preferably thiophenyl, for example, thiophen-2-yl and thiophen-3-yl. X is a group selected from the list comprising: -0-, -S-, -C(0)0-, -C(O)-, -NH(C=0)- and -N(RN)-, wherein RN is selected from the group comprising H and Ci-4 alkyl. X may preferably be: -0-, -S-, -C(0)0-, -NH(C=0)- or -NH-, and may more preferably be: -0-, -S-, or -NH-, and most preferably is -NH-.
Q2-X may be on any of the available ring atoms of the C5-7 aryl group, but is preferably on a ring atom that is not adjacent the bond to the remainder of the compound, i.e. it is preferably β or γ to the bond to the remainder of the compound. Therefore, where the C5-7 aryl group (A) is phenyl, the substituent (Q2-X) is preferably in the meta- or para- positions, and more preferably is in the para- position.1
In some embodiments, Q1 is a single bond. In these embodiments, Q2 is selected from a single bond and -Z-(CH2)n-, where Z is selected from a single bond, O, S and NH and is from 1 to 3. In some of these embodiments, Q2 is a single bond. In other embodiments, Q2 is -Z-(CH2)n-- In these embodiments, Z may be O or S and n may be 1 or n may be 2. In other of these embodiments, Z may be a single bond and n may be 1.
In other embodiments, Q1 is -CH=CH-. In some embodiments, R2 may be -A-CH2-X and -A-X. In these embodiments, X may be - 0-, -S-, -C(0)0-, -C(O)- and -NH-. In particularly preferred embodiments, X may be -NH-.
R12
R12 may be a C5-7 aryl group. A C5-7 aryl group may be a phenyl group or a C5-7 heteroaryl group, for example furanyl, thiophenyl and pyridyl. In some embodiments, R12 is preferably phenyl. In other embodiments, R12 is preferably thiophenyl, for example, thiophen-2-yl and thiophen-3-yl.
R12 may be a C8-io aryl, for example a quinolinyl or isoquinolinyl group. The quinolinyl or isoquinolinyl group may be bound to the PBD core through any available ring position. For example, the quinolinyl may be quinolin-2-yl, quinolin-3-yl, quinolin-4yl, quinolin-5-yl, quinolin-6-yl, quinolin-7-yl and quinolin-8-yl. Of these quinolin-3-yl and quinolin-6-yl may be preferred. The isoquinolinyl may be isoquinolin-1-yl, isoquinolin-3-yl, isoquinolin-4yl, isoquinolin-5-yl, isoquinolin-6-yl, isoquinolin-7-yl and isoquinolin-8-yl. Of these isoquinolin- 3-yl and isoquinolin-6-yl may be preferred. R12 may bear any number of substituent groups. It preferably bears from 1 to 3 substituent groups, with 1 and 2 being more preferred, and singly substituted groups being most preferred. The substituents may be any position.
Where R12 is C5-7 aryl group, a single substituent is preferably on a ring atom that is not adjacent the bond to the remainder of the compound, i.e. it is preferably β or γ to the bond to the remainder of the compound. Therefore, where the C5-7 aryl group is phenyl, the substituent is preferably in the meta- or para- positions, and more preferably is in the para- position.
Where R12 is a C8-io aryl group, for example quinolinyl or isoquinolinyl, it may bear any number of substituents at any position of the quinoline or isoquinoline rings. In some embodiments, it bears one, two or three substituents, and these may be on either the proximal and distal rings or both (if more than one substituent).
R12 substituents
If a substituent on R12 is halo, it is preferably F or CI, more preferably CI. If a substituent on R12 is ether, it may in some embodiments be an alkoxy group, for example, a Ci-7 alkoxy group (e.g. methoxy, ethoxy) or it may in some embodiments be a C5-7 aryloxy group (e.g. phenoxy, pyridyloxy, furanyloxy). The alkoxy group may itself be further substituted, for example by an amino group (e.g. dimethylamino). If a substituent on R12 is C1-7 alkyl, it may preferably be a C1-4 alkyl group (e.g. methyl, ethyl, propyl, butyl).
If a substituent on R12 is C3-7 heterocyclyl, it may in some embodiments be C6 nitrogen containing heterocyclyl group, e.g. morpholino, thiomorpholino, piperidinyl, piperazinyl. These groups may be bound to the rest of the PBD moiety via the nitrogen atom. These groups may be further substituted, for example, by C- alkyl groups. If the C6 nitrogen containing heterocyclyl group is piperazinyl, the said further substituent may be on the second nitrogen ring atom. If a substituent on R12 is bis-oxy-Ci-3 alkylene, this is preferably bis-oxy-methylene or bis- oxy-ethylene.
Particularly preferred substituents for R12 include methoxy, ethoxy, fluoro, chloro, cyano, bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl-thiophenyl. Another particularly preferred substituent for R12 is dimethylaminopropyloxy.
R12 groups
Particularly preferred substituted R12 groups include, but are not limited to, 4-methoxy- phenyl, 3-methoxyphenyl, 4-ethoxy-phenyl, 3-ethoxy-phenyl, 4-fluoro-phenyl, 4-chloro- phenyl, 3,4-bisoxymethylene-phenyl, 4-methylthiophenyl, 4-cyanophenyl, 4- phenoxyphenyl, quinolin-3-yl and quinolin-6-yl, isoquinolin-3-yl and isoquinolin-6-yl, 2- thienyl, 2-furanyl, methoxynaphthyl, and naphthyl. Another possible substituted R12 group is 4-nitrophenyl.
M and z
It is preferred that M and M' are monovalent pharmaceutically acceptable cations, and are more preferably Na+. z is preferably 3.
Examples
General Experimental Methods
Optical rotations were measured on an ADP 220 polarimeter (Bellingham Stanley Ltd.) and concentrations (c) are given in g/100ml_. Melting points were measured using a digital melting point apparatus (Electrothermal). IR spectra were recorded on a Perkin-Elmer Spectrum 1000 FT IR Spectrometer. 1 H and 13C NMR spectra were acquired at 300 K using a Bruker Avance NMR spectrometer at 400 and 100 MHz, respectively. Chemical shifts are reported relative to TMS (δ = 0.0 ppm), and signals are designated as s (singlet), d (doublet), t (triplet), dt (double triplet), dd (doublet of doublets), ddd (double doublet of doublets) or m (multiplet), with coupling constants given in Hertz (Hz). Mass spectroscopy (MS) data were collected using a Waters Micromass ZQ instrument coupled to a Waters 2695 HPLC with a Waters 2996 PDA. Waters Micromass ZQ parameters used were: Capillary (kV), 3.38; Cone (V), 35; Extractor (V), 3.0; Source temperature (°C), 100; Desolvation Temperature (°C), 200; Cone flow rate (L/h), 50; De-solvation flow rate (L/h), 250. High-resolution mass spectroscopy (HRMS) data were recorded on a Waters Micromass QTOF Global in positive W-mode using metal-coated borosilicate glass tips to introduce the samples into the instrument. Thin Layer Chromatography (TLC) was performed on silica gel aluminium plates (Merck 60, F254), and flash chromatography utilised silica gel (Merck 60, 230-400 mesh ASTM). Except for the HOBt (NovaBiochem) and solid-supported reagents (Argonaut), all other chemicals and solvents were purchased from Sigma-Aldrich 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 40-60°C.
Compound 1 b was synthesised as described in WO 00/012508 (compound 210), which is herein incorporated by reference. General LC/MS conditions: The HPLC (Waters Alliance 2695) was run using a mobile phase of water (A) (formic acid 0.1 %) and acetonitrile (B) (formic acid 0.1 %). Gradient: initial composition 5% B over 1 .0 min then 5% B to 95% B within 3 min. The composition was held for 0.5 min at 95% B, and then returned to 5% B in 0.3 minutes. Total gradient run time equals 5 min. Flow rate 3.0 mL/min, 400μΙ_ was split via a zero dead volume tee piece which passes into the mass spectrometer. Wavelength detection range: 220 to 400 nm. Function type: diode array (535 scans). Column: Phenomenex® Onyx Monolithic C18 50 x 4.60 mm
LC/MS conditions specific for compounds protected by both a Troc and a TBDMs group: Chromatographic separation of Troc and TBDMS protected compounds was performed on a Waters Alliance 2895 HPLC system utilizing a Onyx Monoliiic reversed-phase column (3 μπι particles, 50 x 4.8 mm) from Phenomenex Corp. Mobile-phase A consisted of 5% acetonitrile - 95 % water containing 0.1 % formic acid, and mobile phase B consisted of 95% acetonitrile - 5% water containing 0.1 % formic acid. After 1 min at 5% B, the proportion of B was raised to 95% B over the next 2.5 min and maintained at 95% B for a further 1 min, before returning to 95% A in 10 s and re-equilibration for a further 50 sec, giving a total run time of 5.0 min. The flow rate was maintained at 3.0 mL/min. LC/MS conditions specific for compound 33: LC was run on a Waters 2767 sample Manager coupled with a Waters 2996 photodiode array detector and a Waters ZQ single quadruple mass Spectrometer. The column used was Luna Phenyl-Hexyl 150 x 4.60 mm, 5μιη, Part no. 00F-4257-E0 (Phenomenex). The mobile phases employed were:
Mobile phase A: 100% of HPLC grade water (0.05% triethylamine), pH=7
Mobile phase B: 20% of HPLC grade water and 80% of HPLC grade acetonitrile (0.05% triethylamine), pH=7
The gradients used were:
Time Flow Rate %A %B
(min) (ml/min)
Initial 1 .50 90 10
1 .0 1 .50 90 10
16.0 1 .50 64 36
30.0 1 .50 5 95
31.0 1 .50 90 10
32.0 1 .50 90 10
Mass Spectrometry was carried out in positive ion mode and SIR (selective ion monitor) and the ion monitored was m/z = 727.2.
Synthesis of key intermediates
Figure imgf000067_0001
Figure imgf000067_0002
(a) 1 , 1 '-[[(Propane-1 ,3-diyl)dioxy]bis[(5-methoxy-2-nitro-1 ,4- phenylene)carbonyl]]bis[(2S,4R)-methyl-4-hydroxypyrrolidine-2-carboxylate] (2a) Method A: A catalytic amount of DMF (2 drops) was added to a stirred solution of the nitro-acid 1 a (1 .0 g, 2.15 mmol) and oxalyl chloride (0.95 mL, 1 .36 g, 10.7 mmol) in dry THF (20 mL). The reaction mixture was allowed to stir for 16 hours at room temperature and the solvent was removed by evaporation in vacuo. The resulting residue was re- dissolved in dry THF (20 mL) and the acid chloride solution was added dropwise to a stirred mixture of (2S,4R)-methyl-4-hydroxypyrrolidine-2-carboxylate hydrochloride (859 mg, 4.73 mmol) and TEA (6.6 mL, 4.79 g, 47.3 mmol) in THF (10 mL) at -30°C (dry ice/ethylene glycol) under a nitrogen atmosphere. The reaction mixture was allowed to warm to room temperature and stirred for a further 3 hours after which time TLC (95:5 v/v CHCIs/MeOH) and LC/MS (2.45 min (ES+) m/z (relative intensity) 721 {[M + H]+ , 20)) revealed formation of product. Excess THF was removed by rotary evaporation and the resulting residue was dissolved in DCM (50 mL). The organic layer was washed with 1 N HCI (2 x 15 mL), saturated NaHC03 (2 x 15 mL), H20 (20 mL), brine (30 mL) and dried (MgS04). Filtration and evaporation of the solvent gave the crude product as a dark coloured oil. Purification by flash chromatography (gradient elution: 100% CHCI3 to 96:4 v/v CHCI3/MeOH) isolated the pure amide 2a as an orange coloured glass (840 mg, 54%).
Method B: Oxalyl chloride (9.75 mL, 14.2 g, 1 1 1 mmol) was added to a stirred suspension of the nitro-acid 1 a (17.3 g, 37.1 mmol) and DMF (2 mL) in anhydrous DCM (200 mL).
Following initial effervescence the reaction suspension became a solution and the mixture was allowed to stir at room temperature for 16 hours. Conversion to the acid chloride was confirmed by treating a sample of the reaction mixture with MeOH and the resulting bis- methyl ester was observed by LC/MS. The majority of solvent was removed by
evaporation in vacuo, the resulting concentrated solution was re-dissolved in a minimum amount of dry DCM and triturated with diethyl ether. The resulting yellow precipitate was collected by filtration, washed with cold diethyl ether and dried for 1 hour in a vacuum oven at 40°C. The solid acid chloride was added portionwise over a period of 25 minutes to a stirred suspension of (2S,4R)-methyl-4-hydroxypyrrolidine-2-carboxylate hydrochloride (15.2 g, 84.0 mmol) and TEA (25.7 mL, 18.7 g, 185 mmol) in DCM (150 mL) at -40°C (dry ice/CH3CN). Immediately, the reaction was complete as judged by LC/MS (2.47 min (ES+) m/z (relative intensity) 721 {[M + H]+ , 100)). The mixture was diluted with DCM (150 mL) and washed with 1 N HCI (300 mL), saturated NaHC03 (300 mL), brine (300 mL), filtered (through a phase separator) and the solvent evaporated in vacuo to give the pure product 2a as an orange solid (21 .8 g, 82%).
Analytical Data: [a]22 D = -46.1 ° (c = 0.47, CHCI3); 1 H NMR (400 MHz, CDCI3) (rotamers) δ 7.63 (s, 2H), 6.82 (s, 2H), 4.79-4.72 (m, 2H), 4.49-4.28 (m, 6H), 3.96 (s, 6H), 3.79 (s, 6H), 3.46-3.38 (m, 2H), 3.02 (d, 2H , J = 1 1 .1 Hz), 2.48-2.30 (m, 4H), 2.29-2.04 (m, 4H); 13C NMR (100 MHz, CDCI3) (rotamers) δ 172.4, 166.7, 154.6, 148.4, 137.2, 127.0, 109.7, 108.2, 69.7, 65.1 , 57.4, 57.0, 56.7, 52.4, 37.8, 29.0; IR (ATR, CHCI3) 3410 (br), 3010, 2953, 1741 , 1622, 1577, 1519, 1455, 1429, 1334, 1274, 121 1 , 1 177, 1072, 1050, 1008, 871 cm"1 ; MS (ES+) m/z (relative intensity) 721 ([M + Hf, 47), 388 (80); HRMS [M + H]+ theoretical C3i H36N40i6 m/z 721 .2199, found (ES+) m/z 721 .2227.
(a) 1, 1 '-[[(Pentane-1 ,5-diyl)dioxy]bis[(5-methoxy-2-nitro-1 ,4- phenylene)carbonyl]]bis[(2S,4R)-methyl-4-hydroxypyrrolidine-2-carboxylate] (2b)
Preparation from 1 b according to Method B gave the pure product as an orange foam (75.5 g, 82%). Analytical Data: (ES+) m/z (relative intensity) 749 {[M + H]+ , 100).
(b) 1, 1 '-[[(Propane-1,3-diyl)dioxy]bis(11aS,2R)-2-(hydroxy)-7-methoxy-1,2,3, 10, 11, 11a- hexahydro-5H-pyrrolo[2, 1 -c][1 ,4]-benzodiazepin-5, 11 -dione] (3a)
Method A: A suspension of 10% Pd/C (7.5 g, 10% w/w) in DMF (40 mL) was added to a solution of the nitro-ester 2a (75 g, 104 mmol) in DMF (360 mL). The suspension was hydrogenated in a Parr hydrogenation apparatus over 8 hours. Progress of the reaction was monitored by LC/MS (2.12 min (ES+) m/z (relative intensity) 597 ([M + H]+ , 100), (ES-) m/z (relative intensity) 595 ([M + H]+ , 100) after the hydrogen uptake had stopped. Solid Pd/C was removed by filtration and the filtrate was concentrated by rotary evaporation under vacuum (below 10 mbar) at 40°C to afford a dark oil containing traces of DMF and residual charcoal. The residue was digested in EtOH (500 mL) at 40°C on a water bath (rotary evaporator bath) and the resulting suspension was filtered through celite and washed with ethanol (500 mL) to give a clear filtrate. Hydrazine hydrate (10 mL, 321 mmol) was added to the solution and the reaction mixture was heated at reflux. After 20 minutes the formation of a white precipitate was observed and reflux was allowed to continue for a further 30 minutes. The mixture was allowed to cool down to room temperature and the precipitate was retrieved by filtration, washed with diethyl ether (2*1 volume of precipitate) and dried in a vacuum desiccator to provide 3a (50 g, 81 %).
Method B: A solution of the nitro-ester 2a (6.80 g, 9.44 mmol) in MeOH (300 mL) was added to Raney™ nickel (4 large spatula ends of a ~ 50% slurry in H20) and anti-bumping granules in a 3-neck round bottomed flask. The mixture was heated at reflux and then treated dropwise with a solution of hydrazine hydrate (5.88 mL, 6.05 g, 188 mmol) in MeOH (50 mL) at which point vigorous effervescence was observed. When the addition was complete (~ 30 minutes) additional Raney™ nickel was added carefully until effervescence had ceased and the initial yellow colour of the reaction mixture was discharged. The mixture was heated at reflux for a further 30 minutes at which point the reaction was deemed complete by TLC (90:10 v/v CHCI3/MeOH) and LC/MS (2.12 min (ES+) m/z (relative intensity) 597 ([M + H]+ , 100)). The reaction mixture was allowed to cool to around 40°C and then excess nickel removed by filtration through a sinter funnel without vacuum suction. The filtrate was reduced in volume by evaporation in vacuo at which point a colourless precipitate formed which was collected by filtration and dried in a vacuum desiccator to provide 3a (5.40 g, 96%). Analytical Data: [a]27 D = +404° (c = 0.10, DMF); 1H NMR (400 MHz, DMSO-d6) δ 10.2 (s, 2H, NH), 7.26 (s, 2H), 6.73 (s, 2H), 5.1 1 (d, 2H, J = 3.98 Hz, OH), 4.32-4.27 (m, 2H), 4.19- 4.07 (m, 6H), 3.78 (s, 6H), 3.62 (dd, 2H, J = 12.1 , 3.60 Hz), 3.43 (dd, 2H, J = 12.0, 4.72 Hz), 2.67-2.57 (m, 2H), 2.26 (p, 2H, J = 5.90 Hz), 1 .99-1.89 (m, 2H); 13C NMR (100 MHz, DMSO-d6) 8 169.1 , 164.0, 149.9, 144.5, 129.8, 1 17.1 , 1 1 1 .3, 104.5, 54.8, 54.4, 53.1 , 33.5, 27.5; IR (ATR, neat) 3438, 1680, 1654, 1610, 1605, 1516, 1490, 1434, 1379, 1263, 1234, 1216, 1 177, 1 156, 1 1 15, 1089, 1038, 1018, 952, 870 cm"1; MS (ES+) m/z (relative intensity) 619 {[M + Na]+ , 10), 597 {[M + H]+ , 52), 445 (12), 326 (1 1 ); HRMS [M + H]+ theoretical C29H32N4O10 m/z 597.2191 , found (ES+) m/z 597.2205.
(b) 1, 1 '-[[(Pentane-1,5-diyl)dioxy]bis(11aS,2R)-2-(hydroxy)-7-methoxy-1,2,3, 10, 11, 11a- hexahydro-5H-pyrrolo[2, 1-c][1,4]-benzodiazepin-5, 11-dione] (3b)
Preparation from 2b according to Method A gave the product as a white solid (22.1 g, 86%).
Analytical Data: MS (ES") m/z (relative intensity) 623.3 ([M - H]" , 100);
(c) 1, 1 '-[[(Propane-1,3-diyl)dioxy]bis(11aS, 2R)-2-(tert-butyldimethylsilyloxy)- 7-methoxy- 1,2,3, 10, 11, 11 a-hexahydro-5H-pyrrolo[2, 1-c][1,4]-benzodiazepin-5, 11-dione] (4a)
TBSCI (317 mg, 2.1 mmol) and imidazole (342 mg, 5.03 mmol) were added to a cloudy solution of the tetralactam 3a (250 mg, 0.42 mmol) in anhydrous DMF (6 mL). The mixture was allowed to stir under a nitrogen atmosphere for 3 hours after which time the reaction was deemed complete as judged by LC/MS (3.90 min (ES+) m/z (relative intensity) 825 ([M + H]+ , 100)). The reaction mixture was poured onto ice (~ 25 mL) and allowed to warm to room temperature with stirring. The resulting white precipitate was collected by vacuum filtration, washed with H20, diethyl ether and dried in the vacuum desiccator to provide pure 4a (252 mg, 73%).
Analytical Data: [a]23 D = +234° (c = 0.41 , CHCI3); 1 H NMR (400 MHz, CDCI3) δ 8.65 (s, 2H, NH), 7.44 (s, 2H), 6.54 (s, 2H), 4.50 (p, 2H, J = 5.38 Hz), 4.21-4.10 (m, 6H), 3.87 (s, 6H), 3.73-3.63 (m, 4H), 2.85-2.79 (m, 2H), 2.36-2.29 (m, 2H), 2.07-1.99 (m, 2H), 0.86 (s, 18H), 0.08 (s, 12H); 13C NMR (100 MHz, CDCI3) δ 170.4, 165.7, 151.4, 146.6, 129.7, 1 18.9, 1 12.8, 105.3, 69.2, 65.4, 56.3, 55.7, 54.2, 35.2, 28.7, 25.7, 18.0, -4.82 and -4.86; IR (ATR, CHCI3) 3235, 2955, 2926, 2855, 1698, 1695, 1603, 1518, 1491 , 1446, 1380, 1356, 1251 , 1220, 1 120, 1099, 1033 cm"1; MS (ES+) m/z (relative intensity) 825 ([M + H]+ , 62), 721 (14), 440 (38); HRMS [M + H]+ theoretical C4i H6oN4OioSi2 m/z 825.3921 , found (ES+) m/z 825.3948.
(c) 1, 1 '-[[(Pentane- 1, 5-diyl)dioxy]bis(11aS, 2R)-2-(tert-butyldimethylsilyloxy)-7-methoxy- 1,2,3, 10, 11, 11 a-hexahydro-5H-pyrrolo[2, 1-c][1,4]-benzodiazepin-5, 11-dione] (4b)
Preparation from 3b according to the above method gave the product as a white solid (27.3 g, 93%).
Analytical Data: MS (ES+) m/z (relative intensity) 853.8 {[M + H]+ , 100), (ES") m/z (relative intensity) 851.6 ([M - H]-, 100.
(d) 1, 1 '-[[(Propane-1,3-diyl)dioxy]bis(11aS,2R)-2-(tert-butyldimethylsilyloxy)-7-methoxy-10- ((2-(trimethylsilyl)ethoxy)methyl)-1,2,3, 10, 11, 11a-hexahydro-5H-pyrrolo[2, 1-c][1,4]- benzodiazepin-5, 11-dione] (5a)
A solution of n-BuLi (4.17 mL of a 1.6 M solution in hexane, 6.67 mmol) in anhydrous THF (10 mL) was added dropwise to a stirred suspension of the tetralactam 4a (2.20 g, 2.67 mmol) in anhydrous THF (30 mL) at -30°C (dry ice/ethylene glycol) under a nitrogen atmosphere. The reaction mixture was allowed to stir at this temperature for 1 hour (now a reddish orange colour) at which point a solution of SEMCI (1 .18 mL, 1 .1 1 g, 6.67 mmol) in anhydrous THF (10 mL) was added dropwise. The reaction mixture was allowed to slowly warm to room temperature and was stirred for 16 hours under a nitrogen atmosphere. The reaction was deemed complete as judged by TLC (EtOAc) and LC/MS (4.77 min (ES+) m/z (relative intensity) 1085 ([M + H]+ , 100)). The THF was removed by evaporation in vacuo and the resulting residue dissolved in EtOAc (60 mL), washed with H20 (20 mL), brine (20 mL), dried (MgS04) filtered and evaporated in vacuo to provide the crude product.
Purification by flash chromatography (80:20 v/v Hexane/EtOAc) gave the pure N10-SEM- protected tetralactam 5a as an oil (2.37 g, 82%).
Analytical Data: [a]23 D = +163° (c = 0.41 , CHCI3); 1 H NMR (400 MHz, CDCI3) δ 7.33 (s, 2H), 7.22 (s, 2H), 5.47 (d, 2H, J = 9.98 Hz), 4.68 (d, 2H, J = 9.99 Hz), 4.57 (p, 2H, J = 5.77 Hz), 4.29-4.19 (m, 6H), 3.89 (s, 6H), 3.79-3.51 (m, 8H), 2.87-2.81 (m, 2H), 2.41 (p, 2H, J = 5.81 Hz), 2.03-1 .90 (m, 2H), 1 .02-0.81 (m, 22H), 0.09 (s, 12H), 0.01 (s, 18H); 13C NMR (100 MHz, CDCI3) 8 170.0, 165.7, 151 .2, 147.5, 133.8, 121.8, 1 1 1.6, 106.9, 78.1 , 69.6, 67.1 , 65.5, 56.6, 56.3, 53.7, 35.6, 30.0, 25.8, 18.4, 18.1 , -1 .24, -4.73; IR (ATR, CHCI3) 2951 , 1685, 1640, 1606, 1517, 1462, 1433, 1360, 1247, 1 127, 1065 cm"1 ; MS (ES+) m/z (relative intensity) 1 1 13 {[M + Na]+ , 48), 1085 {[M + H]+ , 100), 1009 (5), 813 (6); HRMS [M + Hf theoretical C53H88N4O12S14 m/z 1085.5548, found (ES+) m/z 1085.5542.
(d) 1, 1 '-[[(Pentane 1, 5-diyl)dioxy]bis(11aS, 2R)-2-(tert-butyldimethylsilyloxy)- 7-methoxy- 10- ((2-(trimethylsilyl)ethoxy)methyl)-1,2,3, 10, 11, 11 a-hexahydro-5H-pyrrolo[2, 1-c][1,4]- benzodiazepin-5, 11-dione] (5b)
Preparation from 4b according to the above method gave the product as a pale orange foam (46.9 g, 100%), used without further purification. Analytical Data: MS (ES+) m/z (relative intensity) 1 1 14 ([M + H]+ , 90), (ES") m/z (relative intensity) 1 158 ([M + 2Na]" , 100).
(e) 1, 1 '-[[(Propane-1,3-diyl)dioxy]bis(11aS,2R)-2-hydroxy-7-methoxy-10-((2- (trimethylsilyl)ethoxy)methyl)-1,2,3, 10, 11, 11 a-hexahydro-5H-pyrrolo[2, 1-c][1,4]- benzodiazepin-5, 11-dione] (6a)
A solution of TBAF (5.24 mL of a 1.0 M solution in THF, 5.24 mmol) was added to a stirred solution of the bis-silyl ether 5a (2.58 g, 2.38 mmol) in THF (40 mL) at room temperature. After stirring for 3.5 hours, analysis of the reaction mixture by TLC (95:5 v/v CHCI3/MeOH) revealed completion of reaction. The reaction mixture was poured into a solution of saturated NH4CI (100 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (60 mL), dried (MgS04), filtered and evaporated in vacuo to provide the crude product. Purification by flash chromatography (gradient elution: 100% CHCI3 to 96:4 v/v CHCI3/MeOH) gave the pure tetralactam 6a as a white foam (1 .78 g, 87%).
Analytical Data: [a]23 D = +202° (c = 0.34, CHCI3); 1 H NMR (400 MHz, CDCI3) δ 7.28 (s, 2H), 7.20 (s, 2H), 5.44 (d, 2H, J = 10.0 Hz), 4.72 (d, 2H, J = 10.0 Hz), 4.61 -4.58 (m, 2H), 4.25 (t, 4H, J = 5.83 Hz), 4.20-4.16 (m, 2H), 3.91 -3.85 (m, 8H), 3.77-3.54 (m, 6H), 3.01 (br s, 2H, OH), 2.96-2.90 (m, 2H), 2.38 (p, 2H, J = 5.77 Hz), 2.1 1-2.05 (m, 2H), 1.00-0.91 (m, 4H), 0.00 (s, 18H); 13C NMR (100 MHz, CDCI3) δ 169.5, 165.9, 151.3, 147.4, 133.7, 121 .5, 1 1 1 .6, 106.9, 79.4, 69.3, 67.2, 65.2, 56.5, 56.2, 54.1 , 35.2, 29.1 , 18.4, -1.23; IR (ATR, CHCI3) 2956, 1684, 1625, 1604, 1518, 1464, 1434, 1361 , 1238, 1058, 1021 cm"1; MS (ES+) m/z (relative intensity) 885 ([M + 29]+ , 70), 857 ([M + H]+ , 100), 71 1 (8), 448 (17); HRMS [M + H]+ theoretical C4i H6oN4Oi2Si2 m/z 857.3819, found (ES+) m/z 857.3826. (e) 1, 1 '-[[(Pentane-1,5-diyl)dioxy]bis(11aS,2R)-2-hydroxy-7-methoxy-10-((2- (trimethylsilyl)ethoxy)methyl)-1,2,3, 10, 11, 11 a-hexahydro-5H-pyrrolo[2, 1-c][1,4]- benzodiazepin-5, 11-dione] (6b)
Preparation from 5b according to the above method gave the product as a white foam (15.02 g).
Analytical Data: MS (ES+) m/z (relative intensity) 886 ([M + H]+ , 10), 739.6 (100), (ES") m/z (relative intensity) 884 ([M - H]" , 40). (f) 1, 1 '-[[(Propane-1,3-diyl)dioxy]bis[(11aS)-11-sulpho-7-methoxy-2-oxo-10-((2- (trimethylsilyl)ethoxy)methyl) 1,2,3, 10, 11, 11a-hexahydro-5H-pyrrolo[2, 1- c][1 ,4]benzodiazepin-5, 11 -dione]] (7a)
Method A: A 0.37 M sodium hypochlorite solution (142.5 mL, 52.71 mmol, 2.4 eq) was added dropwise to a vigorously stirred mixture of the diol 6a (18.8 g, 21.96 mmol, 1 eq), TEMPO (0.069 g, 0.44 mmol, 0.02 eq) and 0.5 M potassium bromide solution (8.9 mL, 4.4 mmol, 0.2 eq) in DCM (1 15 mL) at 0°C. The temperature was maintained between 0°C and 5°C by adjusting the rate of addition. The resultant yellow emulsion was stirred at 0°C to 5°C for 1 hour. TLC (EtOAc) and LC/MS [3.53 min. (ES+) m/z (relative intensity) 875 ([M + Na]+ , 50), (ES-) m/z (relative intensity) 852 ([M - H]" , 100)] indicated that reaction was complete.
The reaction mixture was filtered, the organic layer separated and the aqueous layer was backwashed with DCM (x 2). The combined organic portions were washed with brine (x 1 ), dried (MgS04) and evaporated to give a yellow foam. Purification by flash column chromatography (gradient elution 35/65 v/v n-hexane/EtOAC, 30/70 to 25/75 v/v n- hexane/EtOAC) afforded the bis-ketone 7a as a white foam (14.1 g, 75%).
Sodium hypochlorite solution, reagent grade, available at chlorine 10-13%, was used. This was assumed to be 10% (10 g NaCIO in 100 g) and calculated to be 1.34 M in NaCIO. A stock solution was prepared from this by diluting it to 0.37 M with water. This gave a solution of approximately pH 14. The pH was adjusted to 9.3 to 9.4 by the addition of solid NaHC03. An aliquot of this stock was then used so as to give 2.4 mol eq. for the reaction. On addition of the bleach solution an initial increase in temperature was observed. The rate of addition was controlled, to maintain the temperature between 0°C to 5°C. The reaction mixture formed a thick, lemon yellow coloured, emulsion. The oxidation was an adaptation of the procedure described in Thomas Fey et al, J. Org. Chem., 2001 , 66, 8154-8159.
Method B: Solid TCCA (10.6 g, 45.6 mmol) was added portionwise to a stirred solution of the alcohol 6a (18.05 g, 21 .1 mmol) and TEMPO (123 mg, 0.78 mmol) in anhydrous DCM (700 mL) at 0°C (ice/acetone). The reaction mixture was stirred at 0°C under a nitrogen atmosphere for 15 minutes after which time TLC (EtOAc) and LC/MS [3.57 min (ES+) m/z (relative intensity) 875 ([M + Na]+ , 50)] revealed completion of reaction. The reaction mixture was filtered through celite and the filtrate was washed with saturated aqueous NaHC03 (400mL), brine (400mL), dried (MgS04), filtered and evaporated in vacuo to provide the crude product. Purification by flash column chromatography (80:20 v/v EtOAc/Hexane) afforded the bis-ketone 7a as a foam (1 1 .7 g, 65%).
Method C: A solution of anhydrous DMSO (0.72 mL, 0.84 g, 10.5 mmol) in dry DCM (18 mL) was added dropwise over a period of 25 min to a stirred solution of oxalyl chloride (2.63 mL of a 2.0 M solution in DCM, 5.26 mmol) under a nitrogen atmosphere at -60°C (liq N2/CHCI3). After stirring at -55°C for 20 minutes, a slurry of the substrate 6a (1 .5 g, 1 .75 mmol) in dry DCM (36 mL) was added dropwise over a period of 30 min to the reaction mixture. After stirring for a further 50 minutes at -55°C, a solution of TEA (3.42 mL, 2.49 g; 24.6 mmol) in dry DCM (18 mL) was added dropwise over a period of 20 min to the reaction mixture. The stirred reaction mixture was allowed to warm to room temperature (~ 1 .5 h) and then diluted with DCM (50 mL). The organic solution was washed with 1 N HCI (2 x 25 mL), H20 (30 mL), brine (30 mL) and dried (MgS04). Filtration and evaporation of the solvent in vacuo afforded the crude product which was purified by flash column chromatography (80:20 v/v EtOAc/Hexane) to afford bis-ketone 7a as a foam (835 mg, 56%)
Analytical Data: [a]20 D = +291 ° (c = 0.26, CHCI3); 1H NMR (400 MHz, CDCI3) δ 7.32 (s, 2H), 7.25 (s, 2H), 5.50 (d, 2H, J = 10.1 Hz), 4.75 (d, 2H, J = 10.1 Hz), 4.60 (dd, 2H, J = 9.85, 3.07 Hz), 4.31-4.18 (m, 6H), 3.89-3.84 (m, 8H), 3.78-3.62 (m, 4H), 3.55 (dd, 2H, J = 19.2, 2.85 Hz), 2.76 (dd, 2H, J = 19.2, 9.90 Hz), 2.42 (p, 2H, J = 5.77 Hz), 0.98-0.91 (m, 4H), 0.00 (s, 18H); 13C NMR (100 MHz, CDCI3) δ 206.8, 168.8, 165.9, 151.8, 148.0, 133.9, 120.9, 1 1 1 .6, 107.2, 78.2, 67.3, 65.6, 56.3, 54.9, 52.4, 37.4, 29.0, 18.4, -1.24; IR (ATR, CHCIs) 2957, 1763, 1685, 1644, 1606, 1516, 1457, 1434, 1360, 1247, 1209, 1098, 1066, 1023 cm"1; MS (ES+) m/z (relative intensity) 881 {[M + 29]+ , 38), 853 {[M + H]+ , 100), 707 (8), 542 (12); HRMS [M + Hf theoretical C4iH56N4012Si2 m/z 853.3506, found (ES+) m/z 853.3502.
(f) 1, 1 '-[[(Pentane-1,5-diyl)dioxy]bis[(11aS)-11-sulpho-7-methoxy-2-oxo-10-((2- (trimethylsilyl)ethoxy)methyl) 1,2,3, 10, 11, 11a-hexahydro-5H-pyrrolo[2, 1- c][1 ,4]benzodiazepin-5, 11-dione]] (7b)
Preparation from 6b according to Method C gave the product as a white foam (10.5 g, 76%).
Analytical Data: MS (ES+) m/z (relative intensity) 882 ([M + H]+ , 30), 735 (100), (ES") m/z (relative intensity) 925 ([M + 45]" , 100), 880 ([M - H]" , 70).
(9) 1 > 1 '-[[(Propane-1 ,3-diyl)dioxy]bis(11 aS)-7-methoxy-2-[[(trifluoromethyl)sulfonyl]oxy]-10- ((2-(trimethylsilyl)ethoxy)methyl)-1, 10, 11, 11 a-tetrahydro-5H-pyrrolo[2, 1-c][1,4]- benzodiazepin-5, 11-dione] (8a)
Anhydrous 2,6-lutidine (5.15 mL, 4.74 g, 44.2 mmol) was injected in one portion to a vigorously stirred solution of bis-ketone 7a (6.08 g, 7.1 mmol) in dry DCM (180 mL) at - 45°C (dry ice/acetonitrile cooling bath) under a nitrogen atmosphere. Anhydrous triflic anhydride, taken from a freshly opened ampoule (7.2 mL, 12.08 g, 42.8 mmol), was injected rapidly dropwise, while maintaining the temperature at -40°C or below. The reaction mixture was allowed to stir at -45°C for 1 hour at which point TLC (50/50 v/v n- hexane/EtOAc) revealed the complete consumption of starting material. The cold reaction mixture was immediately diluted with DCM (200 mL) and, with vigorous shaking, washed with water (1 x 100 mL), 5% citric acid solution (1 x 200 mL) saturated NaHC03 (200 mL), brine (100 mL) and dried (MgS04). Filtration and evaporation of the solvent in vacuo afforded the crude product which was purified by flash column chromatography (gradient elution: 90:10 v/v n-hexane/EtOAc to 70:30 v/v n-hexane/EtOAc) to afford bis-enol triflate 8a as a yellow foam (5.5 g, 70%).
Analytical Data: [a]24 D = +271 ° (c = 0.18, CHCI3); 1 H NMR (400 MHz, CDCI3) δ 7.33 (s, 2H), 7.26 (s, 2H), 7.14 (t, 2H, J = 1 .97 Hz), 5.51 (d, 2H, J = 10.1 Hz), 4.76 (d, 2H, J = 10.1 Hz), 4.62 (dd, 2H, J = 1 1 .0, 3.69 Hz), 4.32-4.23 (m, 4H), 3.94-3.90 (m, 8H), 3.81 -3.64 (m, 4H), 3.16 (ddd, 2H, J = 16.3, 1 1.0, 2.36 Hz), 2.43 (p, 2H, J = 5.85 Hz), 1.23-0.92 (m, 4H), 0.02 (s, 18H); 13C NMR (100 MHz, CDCI3) δ 167.1 , 162.7, 151.9, 148.0, 138.4, 133.6, 120.2, 1 18.8, 1 1 1 .9, 107.4, 78.6, 67.5, 65.6, 56.7, 56.3, 30.8, 29.0, 18.4, -1.25; IR (ATR, CHCI3) 2958, 1690, 1646, 1605, 1517, 1456, 1428, 1360, 1327, 1207, 1 136, 1096, 1060, 1022, 938, 913 cm"1 ; MS (ES+) m/z (relative intensity) 1 144 ([M + 28]+ , 100), 1 1 17 ([M + H]+ , 48), 1041 (40), 578 (8); HRMS [M + H]+ theoretical C43H54N4016Si2S2F6 m/z
1 1 17.2491 , found (ES+) m/z 1 1 17.2465. (g) 1, 1 '-[[(Pentane-1,5-diyl)dioxy]bis(11aS)-7-methoxy-2-[[(trifluoromethyl)sulfonyl]oxy]-10- ((2-(trimethylsilyl)ethoxy)methyl)-1, 10, 11, 11 a-tetrahydro-5H-pyrrolo[2, 1-c][1,4]- benzodiazepin-5, 11-dione] (8b)
Preparation from 7b according to the above method gave the bis-enol triflate as a pale yellow foam (6.14 g, 82%).
Analytical Data: (ES+) m/z (relative intensity) 1 146 ([M + H]+ , 85).
Example 1
Figure imgf000076_0001
(a) (S)-2-(4-aminophenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(trifluoromethyte^ 11- dioxo-10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo[2, 1- c][1, 4]benzodiazepin-8-yloxy)propoxy)-10-( ( 2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2, 1- c] [1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione (9)
Solid Pd(PPh3)4 (20.18 mg, 17.46 mmol) was added to a stirred solution of the triflate 8a
(975 mg, 0.87 mmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboralane-2-yl)aniiine (172 mg, 0.79 mmol) and Na2C03 (138 mg, 3.98 mol) in toluene (13 mL) EtOH (6.5 mL) and H20 (6.5 mL). The dark solution was allowed to stir under a nitrogen atmosphere for 24 hours, after which time analysis by TLC (EtOAc) and LC/MS revealed the formation of the desired mono-coupled product and as well as the presence of unreacted starting material. The solvent was removed by rotary evaporation under reduced pressure and the resulting residue partitioned between H20 (100 mL) and EtOAc (100 mL), after eventual separation of the layers the aqueous phase was extracted again with EtOAc (2 x 25 mL). The combined organic layers were washed with H20 (50 mL), brine (60 mL), dried (MgS04), filtered and evaporated in vacuo to provide the crude Suzuki product. The crude Suzuki product was subjected to flash chromatography (40%EtOAc/60% Hexane→70% EtOAc, 30% Hexane). Removal of the excess eluent by rotary evaporation under reduced pressure afforded the desired product 9 (399 mg) in 43% yield.
1H-NMR: (CDCIs, 400 MHz) δ 7.40 (s, 1 H), 7.33 (s, 1 H), 7.27 (bs, 3H), 7.24 (d, 2H, J = 8.5 Hz), 7.15 (t, 1 H, J = 2.0 Hz), 6.66 (d, 2H, J = 8.5 Hz), 5.52 (d, 2H, J = 10.0 Hz), 4.77 (d, 1 H, J = 10.0 Hz), 4.76 (d, 1 H, J = 10.0 Hz), 4.62 ( dd, 1 H, J = 3.7, 1 1 .0 Hz), 4.58 (dd, 1 H, J = 3.4, 10.6 Hz), 4.29 (t, 4H, J = 5.6 Hz), 4.00-3.85 (m, 8H), 3.80 - 3.60 (m, 4H), 3.16 (ddd, 1 H, J = 2.4, 1 1 .0, 16.3 Hz), 3.1 1 (ddd, 1 H, J = 2.2, 10.5, 16.1 Hz), 2.43 (p, 2H, J = 5.9 Hz), 1 .1-0.9 (m, 4H), 0.2 (s, 18H). 13C-NMR: (CDCI3, 100 MHz) δ 169.8, 168.3, 164.0, 162.7, 153.3, 152.6, 149.28, 149.0, 147.6, 139.6, 134.8, 134.5, 127.9 (methine), 127.5, 125.1 , 123.21 , 121.5, 120.5 (methine), 120.1 (methine), 1 16.4 (methine), 1 13.2 (methine), 108.7 (methine), 79.8 (methylene), 79.6 (methylene), 68.7 (methylene), 68.5 (methylene), 67.0 (methylene), 66.8 (methylene), 58.8 (methine), 58.0 (methine), 57.6 (methoxy), 32.8 (methylene), 32.0 (methylene), 30.3 (methylene), 19.7 (methylene), 0.25 (methyl).
(b) (S)-2-(4-aminophenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-methoxyphenyl)-5, 11-dioxo- 10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo[2, 1- c][1, 4]benzodiazepin-8-yloxy)propoxy)-10-( ( 2-(trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2, 1- c] [1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione (10)
Solid Pd(PPh3)4 (10 mg, 8.69 μιηοΙ) was added to a stirred solution of the mono-triflate 9 (230 mg, 0.22 mmol) in toluene (3 mL), EtOH (10 mL), with 4-methoxyphenyl boronic acid (43 mg, 0.28 mmol), Na2C03 (37 mg, 0.35 mmol), in H20 (1 .5 mL) at room temperature. The reaction mixture was allowed to stir under a nitrogen atmosphere for 20 h, at which point the reaction was deemed complete as judged by LC/MS and TLC (EtOAc). The solvent was removed by rotary evaporation under reduced pressure in vacuo and the resulting residue partitioned between EtOAc (75 mL) and H20 (75 mL). The aqueous phase was extracted with EtOAc (3 x 30 mL) and the combined organic layers washed with H20 (30 mL), brine (40 mL), dried (MgS04), filtered and evaporated to provide the crude product. The crude product was purified by flash chromatography (60% Hexane: 40% EtOAc→ 80% EtOAc: 20% Hexane) to provide the pure dimer as an orange foam.
Removal of the excess eluent under reduced pressure afforded the desired product 10 (434 mg) in 74% yield.
1 H-NMR: (CDCIs, 400 MHz) δ 7.38 (s, 2H), 7.34 (d, 2H, J = 8.8 Hz), 7.30 (bs, 1 H), 7.26- 7.24 (m, 3H), 7.22 (d, 2H, J = 8.5 Hz), 6.86 (d, 2H , J = 8.8 Hz), 6.63 (d, 2H, J = 8.5 Hz), 5.50 (d, 2H, J = 10.0 Hz), 4.75 (d, 1 H, J = 10.0 Hz), 4.74 (d, 1 H, J = 10.0 Hz), 4.56 (td, 2 H, J = 3.3, 10.1 Hz), 4.27 (t, 2H, J = 5.7 Hz), 4.00-3.85 (m, 8H), 3.80 (s, 3H), 3.77-3.60 (m, 4H), 3.20-3.00 (m, 2H), 2.42 ( p, 2H, J = 5.7 Hz), 0.96 (t, 4H, J = 8.3 Hz), 0.00 (s, 18H). 13C-NMR: (CDCI3, 100 MHz) δ 169.8, 169.7, 162.9, 162.7, 160.6, 152.7, 152.6, 149.0, 147.5, 134.8, 127.8 (methine), 127.4, 126.8, 125.1 , 123.1 , 123.0, 121 .5 (methine), 120.4 (methine), 1 16.4 (methine), 1 15.5 (methine), 1 13.1 (methine), 108.6 (methine), 79.6 (methylene), 68.5 (methylene), 66.9 (methylene), 58.8 (methine), 57.6 (methoxy), 56.7 (methoxy), 32.8 (methylene), 30.3 (methylene), 19.7 (methylene), 0.0 (methyl).
(c) (S)-2-(4-aminophenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo- 5, 11a-dihydro-1H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-8-yloxy)propoxy)-1H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-5(11 aH)-one (11)
Fresh LiBH4 (183 mg, 8.42 mmol) was added to a stirred solution of the SEM-dilactam 10 (428 mg, 0.42 mmol) in THF (5 mL) and EtOH (5 mL) at room temperature. After 10 minutes, delayed vigorous effervescence was observed requiring the reaction vessel to be placed in an ice bath. After removal of the ice bath the mixture was allowed to stir at room temperature for 1 hour. LC/MS analysis at this point revealed total consumption of starting material with very little mono-reduced product. The reaction mixture was poured onto ice (100 mL) and allowed to warm to room temperature with stirring. The aqueous mixture was extracted with DCM (3 x30 mL) and the combined organic layers washed with H20 (20 mL), brine (30 mL) and concentrated in vacuo. The resulting residue was treated with DCM (5 mL), EtOH (14 mL), H20 (7 mL) and silica gel (10 g). The viscous mixture was allowed to stir at room temperature for 3 days. The mixture was filtered slowly through a sinter funnel and the silica residue washed with 90% CHCI3: 10% MeOH (-250 mL) until UV activity faded completely from the eluent. The organic phase was washed with H20 (50 mL), brine 60 mL), dried (MgS04) filtered and evaporated in vacuo to provide the crude material. The crude product was purified by flash chromatography (97% CHCI3 : 3% MeOH) to provide the pure C2/C2'aryl PBD dimer 1 1 (185 mg) 61 % yield.
1 H-NMR: (CDCI3, 400 MHz) δ 7.88 (d, 1 H, J = 4.0 Hz), 7.87 (d, 1 H, J = 4.0 Hz), 7.52 (s, 2H), 7.39 (bs, 1 H), 7.37-7.28 (m, 3H), 7.20 (d, 2H, J = 8.5 Hz), 6.89 (d, 2H, J = 8.8 Hz),
6.87 (s, 1 H), 6.86 (s, 1 H), 6.67 (d, 2H, J = 8.5 Hz), 4.40-4.20 (m, 6H), 3.94 (s, 6H), 3.82 (s, 3H), 3.61 -3.50 (m, 2H), 3.40-3.30 (m, 2H), 2.47-2.40 (m, 2H). 13C-NMR: (CDCI3, 100 MHz) δ 162.5 (imine methine), 161 .3, 161 .1 , 159.3, 156.0, 151 .1 , 148.1 , 146.2, 140.3, 126.2 (methine), 123.2, 122.0, 120.5 (methine), 1 19.4, 1 15.2 (methine), 1 14.3 (methine), 1 1 1 .9 (methine), 1 1 1 .2 (methine), 65.5 (methylene), 56.2 (methoxy), 55.4 (methoxy), 53.9 (methine), 35.6 (methylene), 28.9 (methylene).
Example 2
Figure imgf000079_0001
(a) (S)-2-(4-aminophenyl)-7-methoxy-8-(5-((S)-7-methoxy-2-(4-methoxyphenyl)-5, 11-dioxo- 10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo[2, 1- c][1, 4]benzodiazepin-8-yloxy)pentyloxy)- 10-((2-(trimethylsilyl)ethoxy)methyl)-1H- pyrrolo[2, 1-c][1 ,4]benzodiazepine-5, 11(101-1, 11aH)-dione (12)
Solid Pd(PPh3)4 (32 mg, 27.7 μιηοΙ) was added to a stirred solution of the bis-triflate 8b (1 .04 g, 0.91 mmol) in toluene (10 mL), EtOH (5 mL), with 4-methoxyphenyl boronic acid (0.202 g, 1 .32 mmol), Na2C03 (0.169 g, 1 .6 mmol), in H20 (5 mL) at 30°C. The reaction mixture was allowed to stir under a nitrogen atmosphere for 20 hours. Additional solid 4- (4,4,5,5-tetramethyl-1 ,3,2-dioxaboralan-2-yl)aniline (0.203 g, 0.93 mmol) and Na2C03 (0.056 g, 0.53 mmol) were added followed by solid Pd(PPh3)4 (10 mg, 8.6 μιηοΙ). The reaction mixture was allowed to stir under a nitrogen atmosphere for a further 20 hours. LC/MS indicated the formation of desired product. EtOAc (100 mL) and H20 (100 mL) were added, the aqueous was separated and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with H20 (100 mL), brine (100 mL), dried (MgS04), filtered and evaporated to provide a dark brown oil. The oil was dissolved in DCM and loaded onto a 10 g SCX-2 cartridge pre-equilibrated with DCM (1 vol). The cartridge was washed with DCM (3 vol), MeOH (3 vol) and the crude product eluted with 2M NH3 in MeOH (2 vol). Flash chromatography (50% n-hexane: 50% EtOAc→ 20% n-hexane: 80% EtOAc) provided the pure dimer 12 as a yellow foam (0.16 g, 34%).
Analytical Data: [a]23 D = +388° (c = 0.22, CHCI3); 1H-NMR: (CDCI3, 400 MHz) δ 7.39 (s, 2H), 7.35 (d, 2H, J = 12.8 Hz), 7.32 (bs, 1 H), 7.26-7.23 (m, 5H), 6.89 (d, 2H, J = 8.8 Hz), 6.66 (d, 2H, J = 8.5 Hz), 5.55 (d, 2H, J = 10.0 Hz), 4.73 (d, 1 H, J = 10.0 Hz), 4.72 (d, 1 H, J = 10.0 Hz), 4.62 (td, 2 H, J = 3.2, 10.4 Hz), 4.15 - 4.05 (m, 4H), 4.00-3.85 (m, 8H), 3.82 (s, 3H), 3.77-3.63 (m, 4H), 3.20-3.05 (m, 2H), 2.05 - 1.95 ( m, 4H), 1.75 - 1.67 (m, 2H) 1 .01 - 0.95 (m, 4H), 0.03 (s, 18H); MS (ES+) m/z (relative intensity) 1047 {[M + H]+ , 45).
(b) (S)-2-(4-aminophenyl)-7-methoxy-8-(5-((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo- 5, 11a-dihydro-1H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-8-yloxy)pentyloxy)-1H-pyrrolo[2, 1- c][1 ,4]benzodiazepine-5( 11aH)-one (13)
Fresh LiBH4 (66 mg, 3.04 mmol) was added to a stirred solution of the SEM-dilactam 12 (428 mg, 0.42 mmol) in THF (3 mL) and EtOH (3 mL) at 0°C (ice bath). The ice bath was removed and the reaction mixture was allowed to reach room temperature (vigorous effervescence). After 2 hours LC/MS analysis indicated the complete consumption of starting material. The reaction mixture was poured onto ice (50 mL) and allowed to warm to room temperature with stirring. The aqueous mixture was extracted with DCM (3 x50 mL) and the combined organic layers washed with H20 (50 mL), brine (50 mL), dried (MgS04) and concentrated in vacuo. The resulting residue was treated with DCM (2 mL), EtOH (5 mL), H20 (2.5 mL) and silica gel (3.7 g). The viscous mixture was allowed to stir at room temperature for 3 days. The mixture was filtered through a sinter funnel and the silica residue washed with 90% CHCI3: 10% MeOH (-250 mL) until UV activity faded completely from the eluent. The organic phase was dried (MgS04) filtered and evaporated in vacuo to provide the crude material. The crude product was purified by flash chromatography (99.5% CHCIs : 0.5% MeOH to 97.5% CHCI3 : 2.5% MeOH in 0.5% increments)) to provide the pure C2/C2'aryl PBD dimer 13 (59 mg, 52%).
Analytical Data: [a]28 D = +760° (c = 0.14, CHCI3); 1H NMR (400 MHz, CDCI3) δ 7.89 (d, 1 H, J = 4.0 Hz), 7.87 (d, 1 H, J = 4.0 Hz), 7.52 (s, 2H), 7.39 (bs, 1 H), 7.37-7.28 (m, 3H), 7.22 (d, 2H, J = 8.4 Hz), 6.91 (d, 2H, J = 8.8 Hz), 6.815 (s, 1 H), 6.81 (s, 1 H), 6.68 (d, 2H, J = 8.4 Hz), 4.45 - 4.35 (m, 2H), 4.2-4.0 (m, 4H), 3.94 (s, 6H), 3.85 - 3.7 (s, 3H), 3.65 - 3.50 (m, 2H), 3.45 - 3.3 (m, 2H), 2.05 - 1.9 (m, 4H), 1.75 - 1.65 (m, 2H); MS (ES+) (relative intensity) 754.6 ([M + H]+ , 100), (ES") (relative intensity) 752.5 ([M - H]" , 100).
Figure imgf000081_0001
(a)(S)-2-(thien-2-yl)-7-methoxy-8-(3-((S)-7-methoxy-2-(trifluorometh
dioxo-10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo[2, 1- c][1, 4]benzodiazepin-8-yloxy)propyloxy)-10-(( 2-(trimethylsilyl)ethoxy)methyl)- 1 H- pyrrolo[2, 1-c][1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione ( 14)
Solid Pd(PPh3)4 (41 mg, 0.036 mmol) was added to a stirred solution of the bis-triflate 8a (1 g, 0.9 mmol) in toluene (10 mL), EtOH (5 mL), with thien-2-yl boronic acid (149 mg, 1.16 mmol), Na2C03 (152 mg, 1.43 mmol), in H20 (5 mL). The reaction mixture was allowed to stir under a nitrogen atmosphere overnight at room temperature. The solvent was removed by evaporation in vacuo and the resulting residue partitioned between H20 (100 mL) and EtOAc (100 mL). The aqueous layer was extracted with EtOAc (2 x 30 mL) and the combined organic layers washed with H20 (50 mL), brine (50 mL) dried (MgS04), filtered and evaporated in vacuo to provide the crude product which was purified by flash chromatography (80 hexane: 20 EtOAc→ 50 hexane: 50 EtOAc) to provide the dimer 14 (188 mg, 20 %) yield
Analytical data: LC-MS RT 4.27 mins, 1051 (M + H); 1H-NMR (400 MHZ, CDCI3) δ 7.36 (s, 1 H), 7.31 (bs, 1 H), 7.27 (bs, 1 H), 7.26-7.23 (m, 2H), 7.22-7.17 (m, 1 H), 7.12 (bs, 1 H), 7.02-6.96 (m, 2H), 5.50 (d, J = 10.0 Hz, 2H), 7.75 (d, J = 10.0 Hz, 2H), 4.65-4.55 (m, 2H), 4.37-4.13 (m, 4H), 4.00-3.85 (m, 8H), 3.8-3.6 (m, 4H), 3.20-3.10 (m, 2H), 2.50-2.35 (m, 2H), 1 .0-0.9 (m, 4H), 0 (s, 18H). (b) (S)-2-(thien-2-yl)-7-methoxy-8-(3-((S)-7-methoxy-2-( trifluoromethanesulfonyloxy)-5, 11- dioxo-10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo[2, 1- c][1, 4]benzodiazepin-8-yloxy)pentyloxy)- 10-((2-(trimethylsilyl)ethoxy)methyl)-1H- pyrrolo[2, 1-c] [1 ,4]benzodiazepine-5, 11 (10H, 11aH)-dione (15)
Solid Pd(PPh3)4 (7.66 mg, 6.63 μιηοΙ) was added to a stirred, cloudy solution of 14 (174 mg, 0.17 mmol), Na2C03 (28 mg, 0.22 mmol) and 4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaboralan-2-yl)aniline (47 mg, 0.22 mmol) in toluene (2-5 mL), EtOH (1 .25 mL) and H20 (125 mL) at room temperature. The reaction mixture was allowed to stir under a N2 atmosphere for 24 hours at which point the reaction was deemed complete by LC/MS major peak (@ 3.97 min, FW= 1016, M+Na) and TLC (EtOAc). The solvent was removed by evaporation in vacuo and the resulting residue partitioned between EtOAc (60 mL) and H20 (30 mL). The layers were separated and the organic phase was washed with H20) (20 mL), brine (30 mL) dried (MgS04) filtered and evaporated in vacuo to provide the crude product 123 mg, 75 % yield.
Analytical data: LC-MS RT 3.98 mins, 100 % area, 994 (M + H); 1H-NMR (400 MHZ, CDCIs) δ 7.40 (d, J = 5.3 Hz, 2H), 7.30 (t, J =1 .70 Hz, 1 H), 7.29-7.27 (m, 3H), 7.25 (d, J = 8.5 Hz, 2H), 7.21 (dd, J = 1 .4, 4.73 Hz, 1 H), 7.03-6.97 (m, 2H), 6.66 (d, J = 8.5 Hz, 2H), 5.52 (d, J = 10.0 Hz, 2H), 4.78 ( d, J = 10.0 Hz, 1 H), 4.77 (d, J = 10.0 Hz, 1 H), 4.62 (dd, J = 3.4, 10.5 Hz, 1 H), 4.59 (dd, J = 3.40, 10.6 Hz, 1 H), 4.30 (t, J = 5.85 Hz, 4H), 3.85-4.03 (m, 8H), 3.84-3.64 (m, 6H), 3.18 (ddd, J = 2.2, 10.5, 16.0 Hz, 1 H), 3.1 1 (ddd, J = 2.2, 10.5, 16.0 Hz, 1 H), 2.44 (p, J = 5.85 Hz, 2H), 0.98 (t, J = 1.5 Hz, 4H), 0 (s, 18H).
(c) (S)-2-(thien-2-yl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-aminoph 11a- dihydro-1 H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-8-yloxy)propyloxy)-1 H-pyrrolo[2, 1- c][1, 4]benzodiazepine-5( 11aH)-one (16) Fresh LiBH4 (47 mg, 2,22 mmol) was added to a stirred solution of the SEM-dilactam 15 (1 10 mg, 0.1 1 mmol) in dry THF (3 mL) and EtOH (3 mL) at 0°C (ice bath). The ice bath was removed and the reaction mixture stirred under a N2 atmosphere for 1 hour. Analysis of the reaction by LC/MS analysis revealed significant formation of the desired product (Pk @ 2.57 min) (1=69.32), FW= 702, M+H) and half-imine. The reaction mixture was allowed to stir for a further 1 hour after which time no further reaction progress was observed by LC/MS. The reaction mixture was poured onto ice, stirred and allowed to warm to room temperature. Following partition between DCM (50 mL) and water (50 mL), the aqueous phase was extracted with DCM (3 x 20 mL). The combined organic layers were washed with H20 (50 mL), brine (50 mL) and the solvent removed by evaporation in vacuo under reduced pressure.
The resulting residue was dissolved in DCM (5 mL), EtOH (15 mL) and H20 (7 mL) then treated with silica gel (5 g). The reaction was allowed to stir at room temperature for 48 h. The silica was removed by filtration through a sinter funnel and the residue rinsed with 90:10 CHCI3: MeOH (100 mL). H20 (50 mL) was added to the filtrate and the layers were separated (after shaking). The aqueous layer was extracted with CHCI3 (2 x 30 mL) and H20 (50 mL), brine (50 mL), dried (MgS04) filtered and evaporated in vacuo to provide the crude product. Flash chromatography (CHCI3→ 98% CHCI3: 2% MeOH) afforded the product (41 mg, 53%).
Anayltical data: LC-MS RT 2.55 mins, 702 (M
Figure imgf000083_0001
(a) (S)-2-(4-methoxyphenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-
(trifluoromethylsulphonyl)-5, 11-dioxo-10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a- tetrahydro-1H-pyrrolo[2, 1-c][1,4]benzodiazepin-8-yloxy)propyloxy)-10-((2- (trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione (17)
Solid 4-methoxybenzeneboronic acid (0.388 g, 2.55 mmol) was added to a solution of the SEM protected bis triflate (8a)(3.0 g, 2.69 mmol), sodium carbonate (426 mg, 4.02 mmol) and palladium tetrakis triphenylphosphine (0.08 mmol) in toluene (54.8 mL), ethanol (27 mL) and water (27 mL). The reaction mixture was allowed to stir at room temperature for 3 hours. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was washed with water and brine and dried over magnesium sulphate.
Excess solvent was removed by rotary evaporation under reduced pressure and the resulting residue was subjected to flash column chromatography (silica gel; gradient elution EtOAc/hexane 30/70→35/65→40/60→45/55) to remove unreacted b/s-triflate (0.6 g). Removal of excess eluent from selected fractions afforded the 4-methoxyphenyl coupled product (1.27 g, 1.18 mmol, 41 %).
LC-MS RT 4.30 mins, 1076 (M + H); 1H-NMR (400 MHZ, CDCI3) δ 7.41 (s, 1 H), 7.39 (d, J = 8.8 Hz, 2H), 7.35 (s, 1 H), 7.34 (bs, 1 H), 7.29 (s, 1 H), 7.16 (t, J = 1 .9 Hz, 1 H), 6.90 (d, J = 8.8 Hz, 2H), 5.53 (d, J = 10.0 Hz, 2H), 4.79 (d, J = 10.0 Hz, 1 H), 4.78 (d, J = 10.0 Hz, 1 H), 4.66 - 4.60 (m, 2H), 4.30 (t, J = 5.7 Hz, 4H), 4.0 - 3.94 (m, 2H), 3.93 (s, 3H), 3.92 (s, 3H), 3.84 (s, 3H), 3.83 - 3.60 (m, 4H), 3.22 - 3.10 (m, 2H), 2.45 (t, J = 5.9 Hz, 2H), 1 .05 - 0.94 (m, 4H), 0 (s, 18H).
(b) (S)-2-(3-aminophenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-methoxyphenyl)-5, 11-dioxo- 10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo[2, 1- c][1, 4]benzodiazepin-8-yloxy)propyloxy)-10-( ( 2-(trimethylsilyl)ethoxy)methyl)-1H- pyrrolo[2, 1-c] [1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione (18)
Solid 3-aminobenzeneboronic acid (0.143 g, 0.92 mmol) was added to a solution of the mono triflate (17)(0.619 g, 0.58 mmol), sodium carbonate (195 mg, 1 .84 mmol) and palladium tetrakis triphenylphosphine (26.6mg, 0.023 mmol) in toluene (10 mL), ethanol (5 mL) and water (5 mL). The reaction mixture was allowed to stir at room temperature for overnight at 30°C. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was washed with water and brine and dried over magnesium sulphate. Excess solvent was removed by rotary evaporation under reduced pressure and the resulting residue was subjected to flash column chromatography (silica gel; gradient elution EtOAc/hexane 70/30→85/15).. Removal of excess eluent from selected fractions afforded the desired product (0.502 g, 0.49 mmol, 85%). LC-MS RT 4.02 mins, 1019 (M + H); 1H-NMR (400 MHZ, CDCI3) δ 7.38 - 7.35 (m, 4H), 7.33 (bs, 1 H), 7.30 (bs, 1 H), 7.25 (s, 2H), 7.10 (t, J = 7.8 Hz, 1 H), 6.88 - 6.80 (m, 3H), 6.72 (bs, 1 H), 6.57 (dd, J = 7.9, 1.8 Hz, 1 H), 5.50 (d, J = 10.0 Hz, 2H), 4.75 (d, 10.0 Hz, 2H), 4.58 (dd, J = 10.6, 3.3 Hz, 2H), 4.27 (t, J = 5.8 Hz, 4H), 3.95 - 3.91 (m, 2H), 3.90 (s, 6H), 3.80 (s, 3H), 3.77 - 3.60 (m. 6H), 3.15 - 3.05 (m, 2H), 2.41 (p, J = 5.8 Hz, 2H), 0.95 (t, = 8.25 Hz, 4H), 0 (s, 18H).
(c) (S)-2-(3-aminophenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo- 5, 11a-dihydro-1H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-8-yloxy)propyloxy)-1H-pyrrolo[2, 1-c] [1 ,4]benzodiazepine-5( 11aH)-one (19)
A solution of superhydride (0.56 ml_, 0.56 mmol, 1 .0 M in THF) was added dropwise to a solution of the SEM dilactam (18)(0.271 g, 0.27 mmol) in dry THF (10 mL) at -78°C under a nitrogen atmosphere. After 1 hr a further aliquot of superhydride solution (0.13 ml, 0.13 mmol) was added and the reaction mixture was allowed to stir for another 0.5 hr, at which time LC-MS indicated that reduction was complete. The reaction mixture was diluted with water and allowed to warm to room temperature. The reaction mixture was partitioned between chloroform and water, the layers were separated and the aqueous layer extracted with additional chloroform (emulsions). Finally the combined organic phase was washed with brine and dried over magnesium sulphate. The reduced product was dissolved in methanol, chloroform and water and allowed to stir in the presence of silica gel for 72 hours The crude product was subjected to flash column chromatography (methanol/chloroform gradient) to afford the desired imine product (150 mg, 0.21 mmol, 77%) after removal of excess eluent from selected fractions.
LC-MS RT 2.63 mins, 97 % area, 726 (M + H); 1H-NMR (400 MHZ, CDCI3) δ 7.85 (d, J = 3.9 Hz, 1 H), 7.84 (d, J = 3.9 Hz, 1 H), 7.50 (s, 1 H), 7.49 (s, 1 H), 7.42 (s, 1 H), 7.36 (s, 1 H), 7.32 (d, J = 7.3 Hz, 2H), 7.1 1 (t, (d, J = 7.8 Hz, 1 H), 6.90-6.80 (m, 4H), 6.77 (d, J = 7.9 Hz, 1 H), 4.40-4.20 (m, 6H), 3.92 (s, 6H), 3.80 (s, 3H), 3.60-3.27 (m, 6H), 2.48-2.29 (m,2H)
Figure imgf000086_0001
(a) (11 S, 11aS)-2,2,2-trichloroethyl 11-(tert-butyldimethylsilyloxy)-8-(5-((11S, 11aS)-11-(tert- butyldimethylsilyloxy)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-10-((2,2,2- trichloroethoxy)carbonyl)-5, 10, 11, 11 a-tetrahydro— 1H-pyrrolo [2, 1-c][1,4] benzodiazepin-8- yloxy)pentyloxy)-7-methoxy-5-oxo-2-(trifluoromethylsulfonyloxy)-11, 11a-dihydro- pyrrolo[2, 1-c][1,4]benzodiazepine-10(5H)-carboxylate 21
Solid 4-methoxybenzeneboronic acid (59 mg, 0.39 mmol) was added to a solution of the Troc protected bis triflate (Compound 44, WO 2006/1 1 1759) (600 mg, 0.41 mmol), sodium carbonate (65 mg, 0.61 mmoml) and palladium tetrakis triphenylphosphine (0.012 mmol) in toluene (10.8 ml_), ethanol (5.4 mL) and water (5.4 ml_). The reaction mixture was allowed to stir at room temperature overnight. The reaction mixture was then partitioned between ethylacetate and water. The organic layer was washed with water and brine and dried over magnesium sulphate. Excess solvent was removed by rotary evaporation under reduced pressure and the resulting residue was subjected to flash column chromatography (silica gel; gradient elution EtOAc/hexane 20/80→30/70→40/60→60/40) to remove unreacted b/s-triflate. Removal of excess eluent from selected fractions afforded the 4- methoxyphenyl coupled product (261 mg, 0.18 mmol, 46%).
LC-MS RT 4.17 mins, 1427 (M + H); 1H-NMR (400 MHZ, CDCI3) δ 7.38 (s, 1 H), 7.33 (s,
1 H), 7.31 (s, 1 H), 7.30 (s, 1 H), 7.25 (s, 1 H), 7.20 (bs, 1 H), 6.92 (d, J = 8.6 Hz, 2H), 6.77 (d, J = 8.7 Hz, 2H), 6.0 - 5.90 (m, 2H), 5.25 (d, J = 12.0 Hz, 1 H), 5.24 (d, J = 12.0 Hz, 1 H), 4.24 (d, J = 12.0 Hz, 1 H), 4.22 (d, J = 12.0 Hz, 1 H), 4.18-4.08 (m, 2H), 4.07 - 3.89 (m, 10H), 3.81 (s, 3H), 3.44 - 3.25 (m, 2H), 2.85 (d, J = 16.6 Hz, 2H), 2.05 - 1 .90 (m, 4H), 1.76 - 1 .64 (m, 2H), 0.93 (s, 9H), 0.90 (s, 9H), 0.30 (s, 6H), 0.26 (s, 6H).
(b) (11 S, 11aS)-2,2,2-trichloroethyl 11-(tert-butyldimethylsilyloxy)-8-(5-((11S, 11aS)-11-(tert- butyldimethylsilyloxy)-2-(4-hydroxyphenyl)-7-methoxy-5-oxo-10-((2,2,2- trichloroethoxy)carbonyl)-5, 10, 11, 11a-tetrahydro-1H-pyrrolo [2, 1 -c][1 ,4]benzodiazepin-8- yloxy)pentyloxy)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-11 , 11 a-dihydro-1 H-pyrrolo[2, 1- c][1, ^benzodiazepine- 10( 5H)-carboxylate 22
The Suzuki coupling procedure described in step (a) was applied to the synthesis of Compound 21. Compound 20 (62.5 mg 0.044 mmol, ) was treated with 1 equivalent of 4- hydroxybenzeneboronic acid (10 mg) at 30°C overnight to afford the desired compound after filtration through a pad of silica gel. (40 mg, 0.029 mmol, 66% yield). The compound was used directly in the subsequent step
LC-MS RT 4.27 mins, 1371 (M + H) (c) (S)-2-(4-hydroxyphenyl)-7-methoxy-8-(5-((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo- 5, 11a-dihydro- 1H-pyrrolo[2, 1 -c][1 ,4]benzodiazepin-8-yloxy)pentyloxy)-1H- pyrrolo[2, 1- c][1 ,4]benzodiazepine-5( 11aH)-one 23
Cadmium/lead couple (100 mg, Q Dong et al. Tetrahedron Letters vol 36, issue 32, 5681 - 5682, 1995) was added to a solution of 21 (40 mg, 0.029 mmol) in THF (1 mL) and ammonium acetate (1 N, 1 mL) and the reaction mixture was allowed to stir for 1 hour. The reaction mixture was partitioned between chloroform and water, the phases separated and the aqueous phase extracted with chloroform. The combined organic layers were washed with brine and dried over magnesium sulphate. Rotary evaporation under reduced pressure yielded the crude product which was subjected to column chromatography (silica gel, 0→ 4% MeOH/CHCI3). Removal of excess eluent by rotary evaporation under reduced pressure afforded the desired imine product (17 mg 0.023 mmol 79%).
LC-MS RT 2.20 mins, 755 (M + H); 1 H-NMR (400 MHZ, CDCI3) δ 7.89 (d, J = 3.94 Hz, 1 H), 7.89 (d, J = 4.00 Hz, 1 H), 7.53 (s, 1 H), 7.52 (s, 1 H), 7.38 (d, J = 8.7 Hz, 2H), 7.33 (d, J = 8.6 Hz, 2H), 7.28 (s, 1 H), 6.90 (d, J = 8.7 Hz, 2H), 6.84 (d, J = 8.6 Hz, 2H), 6.82 (s, 1 H), 6.81 (s, 1 H), 5.68 (bs, 1 H), 4.50 - 4.30 (m, 2H), 4.22 - 4.00 (m, 4H), 3.93 (s, 6H), 3.82 (s, 3H), 3.69 - 3.45 (m, 2H), 3.44 - 3.28 (m, 2H), 2.64 - 1 .88 (m, 4H), 1 .77 - 1 .62 (m, 2H).
Figure imgf000088_0001
(a) (11S, 11aS)-2,2,2-trichloroethyl 11-(tert-butyldimethylsilyloxy)-8-(5-((11 S, 11 aS)-11 -(tert- butyldimethylsilyloxy)-2-(4-formylphenyl)- 7-methoxy-5-oxo- 10-((2, 2, 2- trichloroethoxy)carbonyl)-5, 10, 11, 11 a-tetrahydro-1 H- pyrrolo[2, 1-c][1,4] benzodiazepin-8- yloxy)pentyloxy)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-11, 11 a-dihydro-1 H-pyrrolo[2, 1- c][1 ,4]benzodiazepine-10(5H)-carboxylate 24
The Suzuki coupling procedure described in Example 5, step (a), was applied to the synthesis of Compound 24. Compound 21 (62.5 mg, 0.044 mmol ) was treated with 1 equivalent of 4-formylbenzeneboronic acid (10.5 mg) at room temperature overnight to afford the desired compound after filtration through a pad of silica gel (45 mg, 0.033 mmol, 75% yield). The compound was used directly in the subsequent step.
LC-MS RT 4.42 mins, 1383 (M + H)
(b) 4-((S)-7-methoxy-8-(5-((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5, 11 a-dihydro-1 H- pyrrolo[2, 1 -c][1 ,4]benzodiazepin-8-yloxy)pentyloxy)-5-oxo-5, 11 a-dihydro-1 H-pyrrolo[2, 1- c][1,4]benzodiazepine-2-yl)benzaldehyde 25
Compound 24 was deprotected by the method described in Example 5, step (c), to yield the desired compound (18 mg, 0.023 mmol, 79%).
LC-MS RT 3.18 mins, 768 (M + H); 1 H-NMR (400 MHZ, CDCI3) δ 9.98 (s, 1 H), 7.91 (d, J = 3.90 Hz, 1 H), 7.90 - 7.80 (m, 3H), 7.68 (s, 1 H), 7.60 - 7.45 (m, 4H), 7.39 (s, 1 H), 7.33 (d, J = 8.7 Hz, 1 H), 6.90 (d, J = 8.7 Hz, 2H), 6.83 (s, 1 H), 6.82 (s, 1 H), 4.55 - 4.44 (m, 1 H), 4.43 - 4.36 (m, 1 H), 4.23 - 4.00 (m, 4H), 3.95 (s, 3H), 3.94 (s, 3H), 3.82 (s, 3H), 3.66 - 3.51 (m, 2H), 3.50 - 3.34 (m, 2H), 2.05 - 1.87 (m, 4H), 1.76 - 164 (m, 2H).
Figure imgf000089_0001
(a) (11S, 11aS)-2,2,2-trichloroethyl 2-(3-aminophenyl)-11-(tert-butyldimethylsilyloxy)-8-(5- ((11 S, 11aS)-11-(tert-butyldimethylsilyloxy)-7-methoxy-5-oxo-10-((2,2,2- trichloroethoxy)carbonyl)-2-(trifluoromethylsulphonyloxy)-5, 10, 11, 11 a-tetrahydro-1 H- pyrrolo[2, 1 -c][1 ,4]benzodiazepin-8-yloxy)pentyloxy)-7-methoxy-5-oxo-11 , 11 a-dihydro-1 H- pyrrolo[2, 1-c][1,4]benzodiazepine-10(5H)-carboxylate 26
The Suzuki coupling procedure described in Example 5, step (a), was applied to the synthesis of Compound 26, using 3-aminobenzeneboronic acid to afford the desired compound in 41 % yield (230 mg, 0.163 mmol)
LC-MS RT 4.28 mins, 141 1 (M + H); 1H-NMR (400 MHZ, CDCI3) δ 7.44 (bs, 1 H), 7.29 (s, 1 H), 7.25 (s, 1 H), 7.20 (s, 1 H), 7.16 (t, J = 7.9 Hz, 1 H), 6.84 - 6.73 (m, 3H), 6.70 (bs, 1 H),
6.62 (dd, J = 7.9, 1 .7 Hz, 1 H), 6.66 - 6.58 (m, 2H), 5.25 (d, J = 12.0 Hz, 1 H), 5.24 (d, J = 12.0 Hz, 1 H), 4.24 (d, J = 12.0 Hz, 1 H), 4.22 (d, J = 12.0 Hz, 1 H), 4.17 - 4.07 (m, 2H), 4.08 - 3.89 (m, 10H), 3.43 - 3.28 (m, 2H), 2.85 (d, J = 1.65 Hz, 2H), 2.07 - 1.90 (m, 4H), 1.78 -
1 .63 (m, 2H), 0.94 (s, 9H), 0.90 (s, 9H), 0.30 (s, 6H), 0.27 (s, 6H).
(b) (11S, 11aS)-2,2,2-trichloroethyl 2-(3-aminophenyl)-11-(tert-butyldimethylsilyloxy)-8-(5- ((11S, 11aS)-11-(tert-butyldimethylsilyloxy)-2-(4-(3-(dimethylamino)propoxy)phenyl)-7- methoxy-5-oxo-10-((2,2,2-trichloroethoxy)carbonyl)-5, 10, 11, 11 a-tetrahydro-1 H-pyrrolo[2, 1- c][1,4]benzodiazepin-8-yloxy)pentyloxy)-7-methoxy-5-oxo-11, 11 a-dihydro-1 H-pyrrolo[2, 1- c][1,4]benzodiazepine-10(5H)-carboxylate 27
Solid 4-[3-(dimethylamino)propoxybenzeneboronic acid pinacol ester (25 mg, 0.082 mmol) was added to a solution of 26 (73 mg, 0.052 mmol mmol), sodium carbonate (18 mg, 0.17 mmol) and palladium tetrakis triphenylphosphine (3 mg) in toluene (1 mL), ethanol (0.5 mL) and water (0.5 mL). The reaction mixture was allowed to stir at room temperature over night. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was washed with water and brine and dried over magnesium sulphate.
Excess solvent was removed by rotary evaporation under reduced pressure and the resulting residue was eluted through a plug of silica gel with chloroform/methanol. Removal of excess eluent from selected fractions afforded the 4-methoxyphenyl coupled product (50 mg, 0.035 mmol, 67%).
LC-MS RT 4.12 mins, 1440 (M + H) (c) (S)-2-(3-aminophenyl)-8-(5-((S)-2-(4-(3-(dimethylamino)propoxy)phenyl)-7-m
oxo-5, 11a-dihydro-1H-pyrrolo[2, 1-c][1 ,4]benzodiazepine-8-yloxy)pentyloxy)-7-methoxy-1H- pyrrolo[2, 1 -c][1 ,4]benzodiazepine-5( 11aH)-one 28
Compound 27 was deprotected by the method described in Example 5, step (c), to yield the desired compound. The reaction mixture was partitioned between DCM and aqueous sodium hydrogen carbonate (emulsion) and the crude product purified by gradient column chromatography on silica gel (5% methanol chloroform→35% methanol/chloroform) to afford the desired unsymmetrical PBD imine (50 mg, 0.018 mmol, 58%)
LC-MS RT 2.55 mins, 826 (M + H) ; 1H-NMR (400 MHZ, CDCI3) δ 7.92 - 7.82 (m, 2H), 7.52 (bs, 2H), 7.45 (bs, 1 H), 7.39 (bs, 1 H), 7.31 (d, J = 8.6 Hz, 2H), 7.14 (t, J = 7.8 Hz, 1 H), 6.89 (d, J = 8.6 Hz, 2H), 6.85 - 6.75 (m, 3H), 6.72 (bs, 1 H), 6.60 (d, J = 8.0 Hz, 1 H), 4.46 - 4.33 (m, 2H), 4.21 - 3.98 (m, 6H), 3.94 (s, 6H), 3.63 - 3.50 (m, 2H), 3.43 - 3.29 (m, 2H), 2.64 - 2.48 (m, 2H), 2.34 (s, 6H), 2.10 - 1 .89 (m, 6H), 1 .57 (m, 2H).
Figure imgf000091_0001
(a) (11S, 11aS)-2,2,2-trichloroethyl 2-(3-aminophenyl)-11-(tert-butyldimethylsilyloxy)-8-(5- ((11 S, 11aS)-11-(tert-butyldimethylsilyloxy)-7-methoxy-2-(4-(4-methylpiperazin-1-yl)phenyl)- 5-oxo-10-((2,2,2-trichloroethoxy)carbonyl)-5, 10, 11, 11 a-tetrahydro-1 H-pyrrolo[2, 1- c][1,4]benzodiazepin-8-yloxy)pentyloxy)-7-methoxy-5-oxo-11, 11 a-dihydro-1 H-pyrrolo[2, 1- c][1, 4]benzodiazepine- 10(5H)-carboxylate 29
The method of Example 7, step (b), was performed to afford the desired product (58 mg, .0.040 mmol, 78%) after filtration through a plug of silica gel (with 1/3 methanol/chloroform) and removal of excess solvent by rotary evaporation under reduced pressure.
LC-MS RT 4.08 mins, 1439 (M + H)
(b) (S)-2-(3-aminophenyl)-7-methoxy-8-(5-((S)-7-methoxy-2-(4-(4-methylpi
yl)phenyl)-5-oxo-5, 11 a-dihydro-1 H-pyrrolo[2, 1 -c][1 ,4]benzodiazepin-8-yloxy)pentyloxy)-1 H- pyrrolo[2, 1 -c][1 ,4]benzodiazepine-5( 11aH)-one 30
The method for Example 7, step (c) was used to deprotect compound 29. The crude product was purified by silica gel gradient chromatography (2% methanol chloroform→35% methanol/chloroform) to afford the desired unsymmetrical PBD imine (18 mg, 0.022 mmol, 59%) LC-MS RT 2.52 mins, 823 (M + H) ; 1H-NMR (400 MHZ, CDCI3) δ 7.80 (d, J = 3.8Hz, 2H), 7.45 (s, 2H), 7.38 (s, 1 H), 7.30 (s, 1 H), 7.23 (d, J = 8.6Hz, 2H), 7.07 (t, J = 7.8 Hz, 1 H), 6.83 (d, J = 8.6 Hz, 2H), 6.79-6.89 (m, 3H), 6.65 (s, 1 H), 6.54 (d, J = 7.9 Hz, 1 H), 4.40-4.24 (m, 2H), 4.15-3.93 (m, 4H), 3.87 (s, 6H), 3.56-3.42 (m, 2H), 3.37-3.23 (m, 2H), 3.22-3.08 (m, 4H), 2.61 -2.41 (m, 4H), 2.29 (s, 3H), 1 .98-1 .80 (m, 4H), 1.67-1 .54 (m, 2H).
Figure imgf000092_0001
Figure imgf000092_0002
(a) (S)-2-(4-(aminomethyl)phenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-methoxyph
5, 11-dioxo-10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11 a-tetrahydro- H-pyrrolo[2, 1 - c][1 ,4]berzod\azep\n-8-yloxy)propyloxy)-10-((2-(trimethylsilyl)ethoxy)methyl)-1 H- pyrrolo[2, 1-c][1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione 31
Solid 4-aminomethylbenzeneboronic acid hydrochloride (0.1 1 1 g, 0.59 mmol) was added to a solution of 17 (0.394 g, 0.37 mmol), sodium carbonate (175 mg, 1 .654 mmol) and palladium tetrakis triphenylphosphine (28.0 mg, 0.024 mmol) in toluene (10 mL), ethanol (5 mL) and water (5 mL). The reaction mixture was allowed to stir overnight at 30°C. The following day the reaction mixture was heated for a further 3 hours at 70°C. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was washed with water and brine and dried over magnesium sulphate. Excess solvent was removed by rotary evaporation under reduced pressure and the resulting residue was subjected to flash column chromatography (silica gel; gradient elution EtOAc/hexane 2/98→15/85). Removal of excess eluent from selected fractions afforded the desired product (0.230 mg, 0.22 mmol, 61 %). LC-MS RT 3.63 mins, 1034 (M + 2H); 1 H-NMR (400 MHz, DMSO d6) δ 1 1.7 (s, 2H), 7.52 (d, J = 8.2 Hz, 2H), 7.48 (d, J = 8.7 Hz, 2H), 7.40 (s, 1 H), 7.50 (d, J = 8.1 Hz, 2H),7.38-7.19 (m, 5H) 6.93 (d, J = 8.7 Hz, 2H), 5.40 (d, J = 2.13 Hz, 1 H), 5.38 (d, J = 2.12 Hz, 1 H), 5.32 (d, J = 10.6 Hz, 2H), 5.25 (d, J = 10.6 Hz, 2H), 4.87-4.72 (m, 2H), 4.35-4.15 (m, 4H), 3.85 (s, 6H), 3.79 (s, 3H), 3.73-3.56 (m, 2H), 3.55-3.39 (m, 4H), 3.22-3.02 (m, 2H), 2.39-2.23 (m, 2H), 0.94-0.67 (m, 4H), -0.06 (s, 18H).
(b) (S)-2-(4-(aminomethyl)phenyl)-7-methoxy-8-(3-((S)-7-methoxy-2-(4-m
oxo-5, 11a-dihydro-1H-pyrrolo[2, 1-c][1 ,4]benzodiazepine-8-yloxy)propy!oxy)-1H-pyrrolo[2, 1- c][1 ,4]benzodiazepine-5( 11aH)-one 32
Compound 31 was deprotected following the method of Example 1 , step (c). The crude product was purified by gradient column chromatography (5/95→30/70 MeOH/CHCI3) to afford the product as a mixture of imine and carbinolamine methyl ethers.
LC-MS RT 2.58 mins, 740 (M + H).
Figure imgf000093_0001
(S) -2-(4-aminophenyl)- 7-methoxy- 11 (S)-sulpho-8-(3-((S)- 7-methoxy- 11 (S)-sulpho-2-(4- methoxyphenyl)-5-oxo-5, 10, 11, 11a -tetrahydro-1 H-pyrrolo[2, 1-c][1 ,4]benzodiazepin-8- yloxy)propyloxy)-1 H-pyrrolo[2, 1-c][1,4]benzodiazepine-5(11aH)-one disodium salt 33
Sodium bisulphite (8.5 mg, 3.1 eq) was added to a stirred suspension of bis-imine 1 1 (20 mg, 0.036 mmol) in isopropanol (4 mL) and water (2 ml_). The reaction mixture was allowed to stir vigorously and eventually became clear (c. 1 hour). The reaction mixture was transferred to a funnel and filtered through a cotton wall (and then washed with 2 mL water). The filtrate was flash frozen (liquid and to bath) and lyophilized to afford the desired product 33 in quantitative yield.
LC-MS RT 1 1 .77 mins, 727.2 (M + H) (Mass of parent compound, bisulphite adducts unstable in mass spectrometer); 1 H-NMR (400 MHz, CDCI3) δ 7.66-7.55 (m, 5H), 7.43 (s, 1 H), 7.39 (d, J = 8.66 Hz, 2H), 7.06 (m, 2H), 6.93 (d, J = 8.84 Hz, 2H), 6.54 (m, 2H), 5.29- 5.21 (m, 2H), 4.32-4.28 (m, 2H), 4.14-4.20 (m, 4H), 3.96-3.83 (m, 2H), 3.77 (s, 3H), 3.73 (m, 6H), 3.52-3.43 (m, 2H), 3.30-3.08 (m, 2H), 2.24-2.21 (m, 2H).
Figure imgf000094_0001
(a) (S)-2-(2-aminophenyl)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxy 11- dioxo-10-((2-(trimethylsilyl)ethoxy)methyl)-5, 10, 11, 11a-tetrahydro-pyrrolo[2, 1- c][1,4]benzodiazepin-8-yl)oxy)propoxy)-10-((2-(trimethylsilyl)ethoxy)methyl)-pyrrolo[2 c][1 ,4]benzodiazepine-5, 11(1 OH, 11aH)-dione (103)
A catalytic amount of tetrakistriphenylphosphinepalladium (0) (1 1.2 mg) was added to a mixture of the mono triflate 17 (380 mg), the pinnacol ester of 2-aminophenylboronic acid (124 mg) and sodium carbonate (120 mg) in ethanol (5 mL), toluene (5ml_) and water (5 mL). The reaction mixture was allowed to stir over night at room temperature and at 40°C until the reaction was complete (c. 2 hr). The reaction mixture was diluted with ethyl acetate and the organic layer was washed with water and brine. The ethyl acetate solution was dried over magnesium sulphate and filtered under vacuum. Removal of ethyl acetate by rotary evaporation under reduced pressure afforded the crude product which was subjected to flash chromatography (silica gel, ethyl acetate/hexane). Pure fractions were collected and combined. Removal of excess eluent by rotary evaporation under reduced pressure afforded the pure product 103 (330 mg, 86% yield). LC/MS RT: 4.17 min, ES+1018.48. (b) (S)-2-(2-aminophenyl)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-metho
5, 11a-dihydro-pyrrolo[2, 1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-pyrrolo[2, 1- c][1,4]benzodiazepin-5(11aH)-one (104)
A solution of Superhydride in dry tetrahydrofuran (1.0 M, 4.4 eq.) was added to a solution of the 2-analino compound 103 (300 mg) in dry tetrahydrofuran (5 mL) at -78°C under an inert atmosphere. As reduction was proceeding slowly an aliquot of lithium borohydride (20 eq.) was added and the reaction mixture was allowed to return to room temperature.
Water/ice was added to the reaction mixture to quench unreacted hydrides and the reaction was diluted with dichloromethane. The organic layer was washed sequentially with water (twice), citric acid and brine. Excess dichloromethane was removed by rotary evaporation under reduced pressure and the residue was redissolve in ethanol and water and treated with silica gel for 96 hours. The reaction mixture was vacuum filtered and the filtrate evaporated to dryness. The residue was subjected to flash column chromatography (silica gel, gradient chloroform/methanol). Pure fractions were collected and combined and excess eluent was removed by rotary evaporation under educed pressure to afford the pure product 104 (30 mg, 14% yield). LC/MS RT: 2.90 min, ES+726.09.
Example 12: Determination of In Vitro Cytotoxicity of Representative PBD
Compounds
K562 assay
K562 human chronic myeloid leukaemia cells were maintained in RPM1 1640 medium supplemented with 10% fetal calf serum and 2 mM glutamine at 37°C in a humidified atmosphere containing 5% C02 and were incubated with a specified dose of drug for 1 hour or 96 hours at 37°C in the dark. The incubation was terminated by centrifugation (5 min, 300 g) and the cells were washed once with drug-free medium. Following the appropriate drug treatment, the cells were transferred to 96-well microtiter plates (104 cells per well, 8 wells per sample). Plates were then kept in the dark 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-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT, Aldrich-Sigma), to an insoluble purple formazan precipitate. Following incubation of the plates for 4 days (to allow control cells to increase in number by approximately 10 fold), 20 μΙ_ of MTT solution (5 mg/mL in phosphate-buffered saline) was added to each well and the plates further incubated for 5 h. The plates were then centrifuged for 5 min at 300 g and the bulk of the medium pipetted from the cell pellet leaving 10-20 μΙ_ per well. DMSO (200 μΙ_) was added to each well and the samples agitated to ensure complete mixing. The optical density was then read at a wavelength of 550 nm on a Titertek Multiscan ELISA plate reader, and a dose-response curve was constructed. For each curve, an IC50 value was read as the dose required to reduce the final optical density to 50% of the control value. Compound 13 has an IC50 of 30 pM in this assay. A2780 assay
The A2780 parental cell line was grown in Dulbecco's Modified Eagles' Media (DMEM) containing -10% Foetal Calf Serum (FCS) and -1 % 200mM L-Glutamine solution and grown in Corning Cellbind 75cm2 flasks.
A 190 μΙ cell suspension was added (at 1 x 104) to each well of columns 2 to 1 1 of a 96 well plate (Nunc 96F flat bottom TC plate). 190μΙ of media was added to each well of columns 1 and 12. The media was Dulbecco's Modified Eagles' Media (DMEM) (which included~10% Foetal Calf Serum (FCS) and -1 % 200mM L-Glutamine solution).
Plates were incubated overnight at 37°C before addition of drug if cells were adherent. 200 μΜ of the test compound solutions (in 100% DMSO) were serially diluted across a 96 well plate. Each resulting point was then further diluted 1/10 into sterile distilled water (SDW).
To the cell negative blanks and compound negative control wells, 10% DMSO was added at 5% v/v. Assay plates were incubated for the following durations at 37°C in 5% C02 in a humidified incubator for 72 hours. Following incubation, MTT solution to a final
concentration of 1.5μΜ was added to each well. The plates were then incubated for a further 4 hours at 37°C in 5% C02 in a humidified incubator. The media was then removed, and the dye was solubilised in 200μΙ DMSO (99.99%).
Plates were read at 540nm absorbance using an Envision plate reader. Data was analysed using Microsoft Excel and GraphPad Prism and IC50 values obtained.
Compound 11 has an IC50 of 1 1.7 pM in this assay.
Renal Cell and AML cell lines assays
The cytotoxicity of various free drug compounds was tested on a renal cell cancer cell line, 786-0, a Hodgkin lymphoma cell line, L428 and two AML cell lines, HL60 and HEL.
For a 96-hour assay, cells cultured in log-phase growth were seeded for 24 h in 96-well plates containing 150 μΙ_ RPMI 1640 supplemented with 20% FBS. Serial dilutions of test article (i.e., free drug) in cell culture media were prepared at 4x working concentration; 50 μΙ_ of each dilution was added to the 96-well plates. Following addition of test article, the cells were incubated with test articles for 4 days at 37 °C. Resazurin was then added to each well to achieve a 50 μΜ final concentration, and the plates were incubated for an additional 4 h at 37 °C. The plates were then read for the extent of dye reduction on a Fusion HT plate reader (Packard Instruments, Meridien, CT, USA) with excitation and emission wavelengths of 530 and 590 nm, respectively. The IC50 value, determined in triplicate, is defined here as the concentration that results in a 50% reduction in cell growth relative to untreated controls.
Referring to the following Table 1 , the para-aniline compound 11 showed markedly increased activity on these cell lines as compared to the mefa-aniline compound 19 in this assay.
Table 1 : IC50 Summary for Free Drugs [nM]
Figure imgf000097_0001
Referring to the following Table 2, the activity of compounds 28, 30 and 32 is shown on L428, 786-0, HEL, HL-60 and MCF-7 cells, as well as the activity for compound 19 on MCF-7 cells- Table 2: IC5o Summary for Free Drugs [nM]
Figure imgf000097_0002
Referring to the following Table 3, the activities of compounds 23, 25, are compared to that of compound 11 on 786-0, Caki-1 , MCF-7, HL-60, THP-1 , H EL, and TF1 cells. Cells were plated in 150 \iL growth media per well into black-sided clear-bottom 96-well plates (Costar, Corning) and allowed to settle for 1 hour in the biological cabinet before placing in the incubator at 37°C, 5% C02. The following day, 4X concentration of drug stocks were prepared, and then titrated as 10-fold serial dilutions producing 8-point dose curves and added at 50 μΙ per well in duplicate. Cells were then incubated for 48 hours at 37°C, 5% C02. Cytotoxicity was measure by incubating with 100 μΙ_ Cell Titer Glo (Promega) solution for 1 hour, and then luminescence was measured on a Fusion HT plate reader (Perkin Elmer). Data was processed with Excel (Microsoft) and Graph Pad (Prism) to produce dose response curves and IC50 values were generated and data collected.
Table 3: IC50 Summary for Free Drugs [nM]
Figure imgf000098_0001
In Examples 13 to 16, the following compounds are referred to by the compound numbers as show below:
Figure imgf000098_0002
Example 13: Synthesis of PBD Drug Linker Compounds
General Information. In the following examples, all commercially available anhydrous solvents were used without further purification. Analytical thin layer chromatography was performed on silica gel 60 F254 aluminum sheets (EMD Chemicals, Gibbstown, NJ). Radial chromatography was performed on Chromatotron apparatus (Harris Research, Palo Alto, CA). Analytical HPLC was performed on a Varian ProStar 210 solvent delivery system configured with a Varian ProStar 330 PDA detector. Samples were eluted over a C12 Phenomenex Synergi 2.0 x 150 mm, 4 μιη, 80 A reverse-phase column. The acidic mobile phase consisted of acetonitrile and water both containing either 0.05% trifluoroacetic acid or 0.1 % formic acid (denoted for each compound). Compounds were eluted with a linear gradient of acidic acetonitrile from 5% at 1 min post injection, to 95% at 1 1 min, followed by isocratic 95% acetonitrile to 15 min (flow rate = 1 .0 mL/min). LC-MS was performed on a ZMD Micromass mass spectrometer interfaced to an HP Agilent 1 100 HPLC instrument equipped with a C12 Phenomenex Synergi 2.0 x 150 mm, 4 μιη, 80 A reverse phase column. The acidic eluent consisted of a linear gradient of acetonitrile from 5% to 95% in 0.1 % aqueous formic acid over 10 min, followed by isocratic 95% acetonitrile for 5 min (flow rate = 0.4 mL/min). Preparative HPLC was carried out on a Varian ProStar 210 solvent delivery system configured with a Varian ProStar 330 PDA detector. Products were purified over a C12 Phenomenex Synergi 10.0 x 250 mm, 4 μιη, 80 A reverse phase column eluting with 0.1 % formic acid in water (solvent A) and 0.1 % formic acid in acetonitrile (solvent B). The purification method consisted of the following gradient of solvent A to solvent B: 90:10 from 0 to 5 min; 90:10 to 10:90 from 5 min to 80 min; followed by isocratic 10:90 for 5 min. The flow rate was 4.6 mL/min with monitoring at 254 nm. NMR spectral data were collected on a Varian Mercury 400 MHz spectrometer. Coupling constants (J) are reported in hertz.
Scheme 1
Figure imgf000099_0001
EEDQ, MeOH, CH2CI2
45%
Figure imgf000099_0002
(S)-2-((S)-2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanamido)-3- methylbutanamido)propanoic acid (36): To a solution of Val-Ala dipeptide 34 (200 mg, 1 .06 mmol) dissolved in 10.6 mL anhydrous DMF was added maleimidocaproyl NHS ester 35 (327 mg, 1 .06 mmol). Diisopropylethyamine (0.92 ml_, 5.3 mmol) was then added and the reaction was stirred under nitrogen at an ambient temperature for 18 h, at which time TLC and analytical HPLC revealed consumption of the starting material. The reaction was diluted with 0.1 M HCI (100 mL), and the aqueous layer was extracted with ethyl acetate (100 mL, 3x). The combined organic layer was washed with water and brine, then dried over sodium sulfate, filtered and concentrated. The crude product was dissolved in minimal methylene chloride and purified by radial chromatography on a 2 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (95:5 to 90:10 CH2CI2/MeOH) to provide 36 (158 mg, 39%) as an oily residue. TLC: Rf = 0.26, 10% MeOH in CH2CI2. 1H NMR (CDCI3) δ (ppm) 0.95 (d, J = 17 Hz, 3H), 0.98 (d, J = 17 Hz, 3H), 1 .30 (m, 2H), 1 .40 (d, J = 17 Hz,
3H), 1 .61 (m, 4H), 2.06 (m, 1 H), 2.25 (dt, J = 4, 19 Hz, 2H), 3.35 (s, 1 H), 3.49 (t, J = 17 Hz, 2H), 4.20 (d, J = 18 Hz, 1 H), 4.38 (m, 1 H), 6.80 (s, 2H). Analytical HPLC (0.1 % formic acid): tR 9.05 min. LC-MS: tR 1 1 .17 min, m/z (ES+) found 381.9 (M+H)+, m/z (ES") found 379.9 (M-H)\
6-(2,5-dioxo-2,5-dihydro-1H^yrrol-1 -yl)-W-((S)-1 -(((S)-1 -((4-((S)-7-methoxy-8-(3-(((S)-7 methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 - oxobutan-2-yl)hexanamide (38): A flame-dried 10 mL flask was charged with acid 36
(3.6 mg, 9.5 μιηοΙ), EEDQ (2.8 mg, 1 1 .4 μιηοΙ), and 0.33 mL anhydrous CH2CI2. Methanol (four drops, ~80 μί) was added to facilitate dissolution and the mixture was stirred under nitrogen for 1 h. PBD dimer 37 (5.7 mg, 7.9 μιηοΙ) was then added and the reaction was stirred at room temperature for 6 h, at which time LC-MS revealed conversion to product. The reaction was concentrated, dissolved in minimal CH2CI2, and purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 90:10 CH2CI2/MeOH) to provide the drug linker 38 (3.9 mg, 45%). TLC: Rf = 0.06, 5% MeOH in CH2CI2. Analytical HPLC (0.1 % formic acid): tR 1 1.51 min. LC-MS: tR 12.73 min, m/z (ES+) found 1089.6 (M+H)+, m/z (ES") found 1087.3 (M-H)\ Scheme 2
Figure imgf000101_0001
6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1 -yl)-W-(4-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4- methoxyphenyl)-5-oxo-5,11 a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8- yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2- yl)phenyl)hexanamide (40): To a flame-dried 10 mL flask was added PBD dimer 37 (25 mg, 34.4 μιηοΙ), which was dissolved in 1 .4 mL of a 10% MeOH in CHCI3 solvent mixture. Maleimidocaproic acid (39) was added (7.3 mg, 34.4 μιηοΙ), followed by EEDQ (10.2 mg, 41.3 μιηοΙ) and pyridine (6 μΙ_, 68.8 μιηοΙ). The reaction was stirred at room temperature under a nitrogen atmosphere for 14 h, at which time LC-MS revealed conversion to product. The reaction was concentrated, dissolved in minimal CH2CI2, and purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 90:10 CH2CI2/MeOH) to provide drug linker 40 (14.1 mg, 45%). LC-MS: tR 12.81 min, m/z (ES+) found 918.9 (M+H)+, m/z (ES") found 917.0 (M-H)\
Scheme 3
Figure imgf000101_0002
2^romo-W-(4-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,11a- dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11 a-dihydro- 1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)acetamide (41): To a flame-dried 10 mL flask was added PBD dimer 37 (16.5 mg, 22.7 μιηοΙ), which was dissolved in 0.9 mL of a 10% MeOH in CHCI3 solvent mixture. Bromoacetic acid was added (3.2 mg, 22.7 μιηοΙ), followed by EEDQ (6.8 mg, 27.2 μιηοΙ). The reaction was stirred at room temperature under a nitrogen atmosphere for 4 h, at which time LC-MS revealed conversion to product. The reaction was concentrated, dissolved in minimal CH2CI2, and purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOI-l mixtures (100:0 to 95:5 CH2CI2/MeOH) to provide drug linker 41 (9.9 mg, 52%). TLC: Rf = 0.09, 5% MeOH in CH2CI2. LC-MS: tR 12.44 min, m/z (ES+) found 848.1 (M+H)+, m/z (ES") found 845.7 (M- H)-.
Scheme 4
Figure imgf000102_0001
6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1 -yl)-W-((S)-1 -(((S)-1 -((3-((S)-7-methoxy-8-(3-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 - oxobutan-2-yl)hexanamide (43): A flame-dried 10 mL flask was charged with acid 36
(3.6 mg, 9.4 μιηοΙ), EEDQ (2.8 mg, 1 1.3 μιηοΙ), and 0.38 mL anhydrous CH2CI2 containing 1 % methanol. The reaction was stirred under nitrogen for 1 h; PBD dimer 42 (6.8 mg, 9.4 μιηοΙ) was then added and the reaction was stirred at room temperature for 2 h, at which time LC-MS revealed conversion to product. The reaction was concentrated, dissolved in minimal CH2CI2, and purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOI-l mixtures (100:0 to 90:10 CH2CI2/MeOI-l) to provide drug linker 43 (3.1 mg, 30%). TLC: Rf = 0.31 , 10% MeOH in CH2CI2. Analytical HPLC (0.1 % formic acid): tR 1 1.49 min. LC-MS: tR 12.28 min, m/z (ES+) found 1089.5 (M+H)+, m/z (ES") found 1087.3 (M-H)\
Scheme 5
Figure imgf000103_0001
EEDQ, pyridine
MeOH, CH2CI2
12%
Figure imgf000103_0002
6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1 -yl)-W-(3-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4- methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8- yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2- yl)phenyl)hexanamide (44): To a flame-dried 10 mL flask was added PBD dimer 42 (8.0 mg, 1 1 μιηοΙ), which was dissolved in 0.44 mL of a 10% MeOH in CH2CI2 solvent mixture. Maleimidocaproic acid (39) was added (2.3 mg, 1 1 μιηοΙ), followed by EEDQ (3.3 mg, 13.2 μιηοΙ) and pyridine (1 .8 μΙ_, 22 μιηοΙ). The reaction was stirred at room temperature under a nitrogen atmosphere for 3 h, at which time LC-MS revealed conversion to product. The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 90:10 CH2CI2/MeOH) to provide drug linker compound 44 (1 .2 mg, 12%). TLC: Rf = 0.45, 10% MeOH in CH2CI2. Analytical HPLC (0.05%
trifluoroacetic acid): tR 1 1.71 min. LC-MS: tR 12.63 min, m/z (ES+) found 919.1 (M+H)+, m/z (ES") found 917.1 (M-H)". Scheme 6
Figure imgf000104_0001
(2S,3y?,4S,5y?,6y?)-2-(2-(3-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)propanamido)- 4-((((3-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro- 1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)carbamoyl)oxy)methyl)phenoxy)-6- methyltetrahydro-2H-pyran-3,4,5-triyl triacetate (46): A flame-dried flask was charged with glucuronide linker intermediate 45 (reference: Jeffrey et al., Bioconjugate Chemistry, 2006, 17, 831-840) (15 mg, 20 μιτιοΙ), 1 .4 mL anhydrous CH2CI2, pyridine (20 μΙ_, 240 μιηοΙ), and then cooled to -78 °C under nitrogen. Diphosgene (3.0 μΙ_, 24 μιηοΙ) was then added and the reaction was stirred for 2 h at -78 °C, after which time a small aliquot was quenched with methanol and analyzed by LC-MS for formation of the methyl carbonate, which confirmed formation of the glucuronide chloroformate. PBD dimer 42 (15 mg, 20 μιηοΙ) was then dissolved in 0.7 mL anhydrous CH2CI2 and added dropwise to the reaction vessel. The reaction was warmed to 0 °C over 2 h and then diluted with 50 mL CH2CI2. The organic layer was washed with water (50 mL), brine (50 mL), dried over sodium sulfate, filtered and concentrated. The crude reaction product was purified by radial chromatography on a 1 mm chromatotron plate eluted 10% MeOH in CH2CI2 to provide 46 (5.7 mg, 19%). TLC: Rf = 0.47, 10% MeOH in CH2CI2. Analytical HPLC (0.1 % formic acid): tR 12.09 min. LC-MS: tR 14.05 min, m/z (ES+) found 1500.3 (M+H)+.
(2S,3S,4S,5R,6S)-6-(2-(3-aminopropanamido)-4-((((3-((S)-7-methoxy-8-(3-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)carbamoyl)oxy)methyl)phenoxy)-3,4,5- trihydroxytetrahydro-2H-pyran-2-carboxylic acid (47): A flask containing 46 (5.7 mg, 3.8 μιηοΙ) dissolved in a solvent mixture of 0.2 mL each of MeOH, tetrahydrofuran, and water was cooled to 0 °C. To the stirred solution was added lithium hydroxide
monohydrate (0.8 mg, 19 μιηοΙ) and the reaction was stirred at room temperature for 4 h, at which time LC-MS indicated conversion to product. Glacial acetic acid (1 .1 μί, 19 μιηοΙ) was added and the reaction was concentrated to provide 47, which was carried forward without further purification. LC-MS: tR 1 1 .59 min, m/z (ES+) found 1 138.4 (M+H)+.
(2S,3S,4S,5R,6S)-6-(2-(3-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 - yl)hexanamido)propanamido)-4-((((3-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4- methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8- yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 , 4]benzodiazepin-2- yl)phenyl)carbamoyl)oxy)methyl)phenoxy)-3,4,5-trihydroxytetrahydro-2H-pyran-2- carboxylic acid (48): To a solution of 47 (4.3 mg, 3.8 umol) dissolved in 0.38 mL anhydrous DMF was added maleimidocaproyl NHS ester 35 (1.2 mg, 3.8 umol), followed by diisopropylethylamine (4.0 uL, 22.8 umol). The reaction was stirred at room
temperature under nitrogen for 2 h, at which time LC-MS revealed conversion to product. The reaction was diluted with a mixture of acetonitrile (0.5 mL), DMSO (1 mL), water (0.5 mL), and then purified by preparative HPLC. The mobile phase consisted of A = water and B = acetonitrile, both containing 0.1 % formic acid. A linear elution gradient of 90:10 A:B to 10:90 A:B over 75 minutes was employed and fractions containing the desired product were lyophilized to provide drug linker compound 48 (1.2 mg, 24% over two steps).
Analytical HPLC (0.1 % formic acid): tR 10.85 min. LC-MS: tR 12.12 min, m/z (ES+) found 1331 .4 (M+H)+, m/z (ES") found 1329.5 (M-H)\ Scheme 7
Figure imgf000106_0001
6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1 -yl)-W-((S)-1 -(((S)-1 -((3-((S)-7-methoxy-8-((5-(((S)-
7- methoxy-2-(4-(4-methylpiperazin-1 -yl)phenyl)-5-oxo-5,11 a-dihydro-1H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)pentyl)oxy)-5-oxo-5,11a-dihydro-1H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 - oxobutan-2-yl)hexanamide (51): A flame-dried 10 mL flask was charged with acid 36 (2.7 mg, 7.1 μιηοΙ), EEDQ (2.1 mg, 8.5 μιηοΙ), and 0.28 mL anhydrous CH2CI2 containing 1 % methanol. The reaction was stirred under nitrogen for 1 h; PBD dimer 49 (5.8 mg, 7.1 μιηοΙ) was then added and the reaction was stirred at room temperature for 20 h, at which time LC-MS revealed conversion to product. The reaction was concentrated then purified by preparative HPLC and fractions containing the desired product were lyophilized to provide drug linker compound 51 (2.7 mg, 32%). Analytical HPLC (0.1 % formic acid): tR 9.17 min. LC-MS: tR 1 1.25 min, m/z (ES+) found 1 185.3 (M+H)+, m/z (ES") found 1 182.9 (M-H)\
W-((S)-1 -(((S)-1 -((3-((S)-8-((5-(((S)-2-(4-(3-(dimethylamino)propoxy)phenyl)-7-methoxy- 5-0X0-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 , 4]benzodiazepin-8-yl)oxy)pentyl)oxy)-7- methoxy-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2- yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 -oxobutan-2-yl)-6-(2,5-dioxo- 2,5-dihydro-1 H-pyrrol-1 -yl)hexanamide (52): A flame-dried 10 mL flask was charged with acid 36 (3.7 mg, 9.7 μιηοΙ), EEDQ (2.9 mg, 1 1.6 μιηοΙ), and 0.4 mL anhydrous CH2CI2 containing 1 % methanol. The reaction was stirred under nitrogen for 1 h; PBD dimer 50 (8.0 mg, 9.7 μιηοΙ) was then added and the reaction was stirred at room temperature for 6 h, at which time LC-MS revealed the presence of product. The reaction was concentrated then purified by preparative HPLC and fractions containing the desired product were lyophilized to provide drug linker compound 52 (3.1 mg, 25%). Analytical HPLC (0.1 % formic acid): tR 9.45 min. LC-MS: tR 1 1.75 min, m/z (ES+) found 1 188.4 (M+H)+, m/z (ES") found 1 186.0 (M-H)\
Scheme 8
Figure imgf000107_0001
4-(2,5-dioxo-2,5-dihydrc-1 H-pyrrol-1 -yl)-N-((S)-1 -(((S)-1 -((4-((S)-7-methoxy-8-(3-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 - oxobutan-2-yl)benzamide (54): To a flame-dried 10 mL flask was added linker fragment 53 (7.7 mg, 20 μιηοΙ), which was dissolved in 0.33 mL of a 5% MeOH in CH2CI2 solvent mixture. EEDQ (6.1 mg, 25 μιηοΙ) was added and the reaction was stirred at room temperature under nitrogen for 15 minutes, at which time PBD dimer 37 (12 mg, 16.5 μιηοΙ) was added. The reaction was stirred at room temperature under a nitrogen atmosphere for an additional 3 h, at which time LC-MS revealed conversion to product. The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2Cl2/MeOH mixtures (100:0 to 90:10 CH2CI2/MeOH) to provide 54 (2.4 mg, 13%). TLC: Rf = 0.44, 10% MeOH in CH2CI2. Analytical HPLC (0.05% trifluoroacetic acid): tR 1 1.53 min. LC-MS: tR 12.61 min, m/z (ES+) found 1095.4 (M+H)+, m/z (ES") found 1093.9 (M-H)\ (S)-2-(2-iodoacetamido)-N-((S)-1 -((4-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4- methoxyphenyl)-5-oxo-5,11a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8- yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2- yl)phenyl)amino)-1 -oxopropan-2-yl)-3-methylbutanamide (56): A flame-dried flask was charged with linker 55 (7.8 mg, 22 μιηοΙ), which was dissolved in 0.37 mL of a 5% MeOH in CH2CI2 solvent mixture. EEDQ (6.8 mg, 27.5 μιηοΙ) was added and the reaction was stirred at room temperature under nitrogen for 15 minutes, at which time PBD dimer 37 (13 mg, 18 μιηοΙ) was added. The reaction was stirred at room temperature under a nitrogen atmosphere for an additional 4 h, at which time LC-MS revealed conversion to product. The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 80:20 CH2CI2/MeOH) to provide 56 (3.5 mg, 18%). Analytical HPLC (0.1 % formic acid): tR 1 1.43 min. LC-MS: tR 12.49 min, m/z (ES+) found 1064.6 (M+H)+, m/z (ES") found 1098.9 (M+2H20-H)\
Scheme 9
Figure imgf000108_0001
6- (2,5-dioxo-2,5-dihydrc-1 H-pyrrol-1 -yl)-N-((S)-1 -(((S)-1 -((4-((S)-7-methoxy-8-((5-(((S)-
7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)pentyl)oxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 - oxobutan-2-yl)hexanamide (58): To a flame-dried 10 mL flask was added linker fragment 36 (19 mg, 50 μιηοΙ), which was dissolved in 0.33 mL of a 5% MeOH in CH2CI2 solvent mixture. EEDQ (12.4 mg, 50 μιηοΙ) was added and the reaction was stirred at room temperature under nitrogen for 15 minutes, at which time PBD dimer 57 (12.5 mg, 16.6 μιηοΙ) was added. The reaction was stirred at room temperature under a nitrogen atmosphere for an additional 5 h, at which time LC-MS revealed conversion to product.
The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 80:20 CH2CI2/MeOH) to provide 58 (2.1 mg, 1 1 %). Analytical HPLC (0.1 % formic acid): tR 12.19 min. LC-MS: tR 12.58 min, m/z (ES+) found 1 1 17.8 (M+H)+, m/z (ES") found 1 133.7 (M+H20-H)\
Scheme 10
Figure imgf000109_0001
EEDQ, MeOH, CH2CI2
16%
Figure imgf000109_0002
(R)-2-((R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3- methylbutanamido)propanoic acid (60): A flame dried flask was charged with Fmoc-D- Valine (200 mg, 0.59 mmol) and 5.9 mL anhydrous THF. N-hydroxysuccinimide (75 mg, 0.65 mmol) was added, followed by diisopropylcarbodiimide (0.1 mL, 0.65 mmol), and the reaction was stirred at an ambient temperature overnight, at which time LC-MS revealed conversion to product. The reaction mixture was diluted with CH2CI2 and washed with water (50 mL), brine (50 mL), dried over sodium sulfate and concentrated to dryness. The material was carried forward without further purification. LC-MS: tR 13.89 min, m/z (ES+) found 437.0 (M+H)+. Crude Fmoc-D-Val-OSu (0.59 mmol) was dissolved in
dimethoxyethane (1 .5 mL) and THF (0.8 mL). D-alanine (73 mg, 0.89 mmol) was dissolved in 2.3 mL water and added to the reaction mixture, followed by sodium bicarbonate (99 mg, 1 .2 mmol). The resulting slurry was stirred at room temperature overnight, at which time the reaction had clarified and LC-MS revealed completion. The reaction was poured into 50 mL CH2CI2 and the organic layer was washed with 50 mL 0.1 M HCI and then brine, dried over sodium sulfate, and then concentrated to dryness. The crude product was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2 to provide 60 (128 mg, 54%). TLC: Rf = 0.18, 10% MeOH in CH2CI2. Analytical HPLC (0.1 % formic acid): tR 9.47 min. LC-MS: tR 13.09 min, m/z (ES+) found 41 1 .1 (M+H)+, m/z (ES") found 409.2 (M-H)\ (R)-2-((R)-2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanamido)-3- methylbutanamido)propanoic acid (61 ): Protected dipeptide 60 (70 mg, 0.37 mmol) was suspended in 6 mL anhydrous CH2CI2, cooled on ice under nitrogen, and 2 mL of diethylamine was added dropwise. The reaction was warmed to room temperature and stirred under nitrogen for 2 h, at which time HPLC revealed consumption of starting material. The reaction was diluted with 6 mL of chloroform and concentrated. The crude reaction residue was re-dissolved in 6 mL chloroform and concentrated twice, followed by drying on a vacuum line for 2 h. The deprotected dipeptide was then dissolved in 3.7 mL anhydrous DMF. MC-OSu (138 mg, 0.44 mmol) was then added, followed by
diisopropylethylamine (0.32 mL, 1.9 mmol). The reaction was stirred under a nitrogen atmosphere at room temperature overnight. Workup was achieved by pouring the reaction in to 50 mL 0.1 M HCI and extracting with ethyl acetate (50 mL, 3x). The combined organic layer was washed with water (50 mL) and brine (50 mL), dried over sodium sulfate, and concentrated. The crude product was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (99:1 to 95:5 CH2CI2/MeOH) to provide 61 (14 mg, 22%). 1H NMR (CD3OD) δ (ppm) 0.94 (d, J = 14 Hz, 3H), 0.98 (d, J = 14 Hz, 3H), 1.29 (m, 2H), 1.39 (d, J = 7.4 Hz, 3H), 1.61 (m, 4H), 2.05 (m, 1 H), 2.25 (dt, J = 1 .2, 7.4 Hz, 2H), 3.48 (t, J = 7 Hz, 2H), 4.19 (m, 1 H), 4.37 (m, 1 H), 6.78 (s, 2H). Analytical HPLC (0.1 % formic acid): tR 10.04 min. LC-MS: tR 1 1.22 min, m/z (ES+) found 382.1 (M+H)+, m/z (ES") found 380.0 (M-H)\ 6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)-N-((R)-1 -(((R)-1 -((4-((S)-7-methoxy-8-(3-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3-methyl-1 - oxobutan-2-yl)hexanamide (62): To a flame-dried 10 mL flask was added linker 61 (9.5 mg, 25 μιηοΙ), which was dissolved in 0.33 mL of a 5% MeOH in CH2CI2 solvent mixture. EEDQ (7.3 mg, 30 μιηοΙ) was added and the reaction was stirred at room temperature under nitrogen for 15 minutes, at which time PBD dimer 37 (12 mg, 16.5 μιηοΙ) was added. The reaction was stirred at room temperature under a nitrogen atmosphere for an additional 3 h, at which time LC-MS revealed conversion to product. The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 80:20 CH2CI2/MeOH) to provide 62 (2.8 mg, 16%). TLC: Rf = 0.39, 10% MeOH in CH2CI2. Analytical HPLC (0.1 % formic acid): tR 1 1.50 min. LC-MS: tR 12.50 min, m/z (ES+) found 1089.7 (M+H)+, m/z (ES") found 1088.0 (M-H)\
Scheme 11
Figure imgf000111_0001
EEDQ, MeOH, CH2CI2
11%
Figure imgf000111_0002
(S)-2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanamido)propanoic acid (64): L- alanine (58 mg, 0.65 mmol) was suspended in 6.5 mL anhydrous DMF and MC-OSu 35 (100 mg, 0.324 mmol) was then added. Diisopropylethylamine (0.28 mL, 1 .6 mmol) was added and the reaction was stirred overnight at room temperature under nitrogen. The reaction was then diluted with 50 mL 0.1 M HCI and the aqueous layer was then extracted with ethyl acetate (50 mL, 3x). The combined organic layer was then washed with water (50 mL) and brine (50 mL), dried over sodium sulfate, and then concentrated to dryness. The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (97.5:2.5 to 90:10 CH2CI2/MeOH) to provide 64 (25 mg, 27%). TLC: Rf = 0.25, 10% MeOH in CH2CI2. 1 H NMR (CD3OD) δ (ppm) 1 .30 (m, 2H), 1 .37 (d, J = 7.4 Hz, 3H), 1.60 (m, 4H), 2.21 (t, J = 7.4 Hz, 2H), 3.48 (t, J = 7 Hz, 2H), 4.35 (q, J = 7.4 Hz, 1 H), 6.78 (s, 2H). Analytical HPLC (0.1 % formic acid): tR 9.06 min.
6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)-N-((S)-1 -((4-((S)-7-methoxy-8-(3-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)hexanamide (65): To a flame-dried 10 mL flask was added linker 64 (14 mg, 50 μιηοΙ), which was dissolved in 0.66 mL of a 5% MeOH in CH2CI2 solvent mixture. EEDQ (15 mg, 60 μιηοΙ) was added and the reaction was stirred at room temperature under nitrogen for 15 minutes, at which time PBD dimer 37 (24 mg, 33 μιηοΙ) was added. The reaction was stirred at room temperature under a nitrogen atmosphere for an additional 4 h. The reaction was purified by radial chromatography on a 1 mm chromatotron plate eluted with CH2CI2/MeOH mixtures (100:0 to 90:10 CH2CI2/MeOH) to provide 65 (3.5 mg, 1 1 %). Analytical HPLC (0.1 % formic acid): tR 1 1.40 min. LC-MS: tR 12.39 min, m/z (ES+) found 990.6 (M+H)+, m/z (ES") found 989.0 (M-H)\
Scheme 12
Figure imgf000112_0001
peptide coupling agent
1
Figure imgf000112_0002
PBD dimer 57 linked directly through maleimidocaproyl spacer (Scheme 14): PBD dimer 57 is coupled to maleimidocaproic acid 39 employing the chemistry described in Scheme 2. Scheme 13
Figure imgf000113_0001
6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)-N-(2-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4- methoxyphenyl)-5-oxo-5,11a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8- yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2- yl)phenyl)hexanamide (68): To a mixture of the 66 (10 mg, 0.013 mmol) in CH2CI2 (300 μί) ν3ε added DIPEA and MC-CI (67) (3 mg, 0.013 mmol). After 1 h, an additional 3 equiv. of DIPEA (7 μΙ_) and 2 equiv. of the acid chloride (6 mg, 0.026 mmol) were added. After 1 h, an additional quantity of DIPEA (7 μΙ_) and acid chloride (6 mg, 0.026 mmol) were added. After an additional 3h, the reaction mixture was aspirated directly onto a 1 mm radial chromatotron plate and eluted with dichloromethane followed by a gradient of methanol (1 % to 5%) in dichloromethane. Product containing fractions, as a mixture with the starting aniline, were concentrated to a residue and dissolved in a mixture of 0.5 mL DMSO, 0.5 mL acetonitrile and 0.5 mL deionized water and was further purified by preparative HPLC. The major peak was collected and the fractions were combined, frozen and lyophilized to give 2.1 mg (18%): MS (ES+) m/z 919.2 [M+H]+.
Note: Acid chloride 67 was prepared by dissolving 100 mg of 39 in oxalyl chloride (5 mL). A drop of DMF was added and the mixture was stirred at an ambient temperature for several hours before being concentrated under reduced pressure. Dichloromethane was added and the mixture was concentrated a second time to afford an off-white solid which was used directly: 1H-NMR (400MHz, CDCI3) δ 6.70 (s, 2H), 3.46 (t, J = 7 Hz, 2H), 2.82 (t, J = 7.2 Hz, 2H), 1 .72 (pent, J = 7.6 Hz, 2H), 1.61 (pent, J = 7.4 Hz, 2H), 1.35 (pent, J = 7.6 Hz, 2H). Scheme 14
Figure imgf000114_0001
tert-butyl 2-(2-aminoacetamido)acetate (69): To a mixture of the glycine ferf-butyl ester hydrogen chloride salt (70) (484 mg, 2.9 mmol) in dichloromethane (25 mL) was added Fmoc-Gly-OH (71 ) (0.861 mg, 2.99 mmol), DIPEA (756 mg, 4.35 mmol) and HATU (1 .3 g, 3.5 mmol). The reaction mixture was stirred at an ambient temperature for 16 h and then poured into ethyl acetate and was washed with water (3X) and brine (1X). The organic phase was dried over MgS04, filtered and concentrated under reduced pressure. The resulting residue was purified via radial chromatography on a 2 mm plate eluting with 5% methanol/dichloromethane. Product containing fractions were concentrated under reduced pressure and treated with 20% piperidine/dichloromethane (10 mL) for 1 h, before being concentrated under reduced pressure and then purified twice via radial chromatography on a 2 mm plate eluting with a gradient of 5 to 10% methanol/dichloromethane to provide (200 mg, 37%): 1 H-NMR (400MHz, CDCI3) δ 7.62 (s, 1 H), 4.00 (s, 2H), 3.39 (s, 2H), 1 .47 (s, 9H).
2-(2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanamido)acetamido)acetic acid (72): To a solution of the amine 69 (200 mg, 0.1 1 mmol) in DMF (1 mL) was added 35 ( 350 mg, 0.1 1 mmol) and the reaction mixture was allowed to stir at an ambient temperature for 2 h. The mixture was concentrated under reduced pressure and was purify by radial chromatography on a 1 mm plate eluting with dichloromethane and a gradient of methanol (1 to 5%) in dichloromethane. Product containing fractions were concentrated under reduced pressure, dissolved in dichloromethane (4 mL) and treated with trifluoroacetic acid (4 mL). After 40 min the mixture was concentrated under reduced pressure and the resulting residue was dissolved in dichloromethane and concentrated to give 22.5 mg (19%) of 72 as white solid: 1 H-NMR (400 MHz, CD3OD) δ 6.79 (s, 2H), 3.93 (s, 2H), 3.89 (s, 2H), 3.49 (t, J = 6.8 Hz, 2H), 2.26 (t, J = 6.8 Hz, 2H), 1.61 (m, 4H), 1 .34 (m, 2H); MS (ES+) m/z 326.21 [M+H]+.
6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)-N-(2-((2-((4-((S)-7-methoxy-8-(3-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-2-oxoethyl)amino)-2-oxoethyl)hexanamide (73): To a mixture of 72 (15 mg, 0.046 mmol) in 5% methanol/dichloromethane (0.5 mL) was added EEDQ (1 1 mg, 0.046 mmol) and the mixture was stirred for 30 min at an ambient temperature, at which time 37 (16 mg, 0.023 mmol) was added. The reaction mixture was stirred for 3 h and was purified directly on a 1 mm radial chromatotron plate eluting with a 1 % to 4% methanol/dichloromethane gradient to give 6.8 mg (29%) of 73 as a yellow solid: MS (ES+) m/z 1033.57 [M+H]+. Scheme 15
Figure imgf000115_0001
(S)-tert-butyl 1 -((S)-pyrrolidine-2-carbonyl)pyrrolidine-2-carboxylate (74): To a mixture of L-proline-fert-butyl ester hydrogen chloride salt 75 (0.5 g, 2.9 mmol) in dichloromethane (50 mL) was added 76 (0.98 g, 2.99 mmol), DIPEA (756 mg, 4.35 mmol) and HATU (1.3 g, 3.5 mmol). The reaction mixture was allowed to stir at an ambient temperature for 16 h. The mixture was poured into ethyl acetate (100 mL) and was washed with 0.2 N HCI (50 mL), water (50 mL), brine (50 mL) and dried over MgS04. Chromatography was conducted on a 2 mm radial chromatotron plate eluting with 10% ethyl acetate in hexanes. Product-containing fractions were concentrated under reduced pressure, dissolved in dichloromethane (8 mL) and treated with piperidine (2 mL). The mixture was stirred for 1 h, concentrated under reduced pressure and purified on a 2 mm radial chromatotron plate eluting with 5% methanol/dichloromethane. This gave 200 mg (26%) of the dipeptide 74: 1H-NMR (400 MHz, CDCI3) δ 4.41 (m, 1 H), 4.17 (m, 1 H), 3.82 (m, 1 H), 3.57 (m, 4H), 3.2 (m, 1 H), 2.82 (m, 1 H), 2.83-1.65 (m, 5H), 1.44 (m, 9H). (S)-tert-butyl 1 -((S)-1 -(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanoyl)pyrrolidine-2- carbonyl)pyrrolidine-2-carboxylate (77): To a mixture of the amine 74 (200 mg, 0.75 mmol), 39 (190 mg, 0.9 mmol) and DIPEA (0.32 mL, 1 .8 mmol) was added HATU (342 mg, 0.9 mmol) and the mixture was allowed to stir at an ambient temperature for 5h. The mixture was poured into ethyl acetate (100 mL) and washed with water (3X100 mL) and brine (1X100 mL). The organic phase was dried over magnesium sulfate, filtered and concentrated. The resulting residue was subjected to radial chromatography on a 2 mm radial chromatotron plate eluting with dichloromethane followed by an increasing gradient of 1 to 5% methanol in dichloromethane. Two additional purifications, both eluting with a gradient of 1 to 5% methanol in dichloromethane, first on a 2 mm plate and then on a 1 mm plate afforded 1 13 mg (33%) of 77 as an white solid: 1H-NMR (400 MHz, CDCI3) δ 4.63 (m, 1 H), 4.41 (m, 1 H), 3.82 (m, 1 H), 3.6 3 (m, 1 H), 3.55 (m, 1 H), 3.45 (m, 3H), 2.38-1.83 (m, 10H), 1.70-1 .50 (m, 5H), 1.45 (m, 9H), 1.35 (m, 2H); MS (ES+) m/z 462.33 [M+H]+.
(S)-1 -((S)-1 -(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanoyl)pyrrolidine-2- carbonyl)pyrrolidine-2-carboxylic acid (78): To a mixture of the ferf-butyl ester 77 in dichloromethane (4 mL) was added trifluoroacetic acid (4 mL). After 40 min the reaction was determined to be complete by HPLC analysis. The mixture was concentrated under reduced pressure and the resulting residue was dissolved in dichloromethane and concentrated a second time to give 37 mg (100%) of 78 as a white solid: 1 H-NMR (400 MHz, CDCIs) δ 6.68 (s, 2H), 4.62 (m, 2H), 3.81 (m, 1 H), 3.70 (m, 1 H), 3.57 (m, 2H), 3.45 (m, 2H), 2.40-1.91 (m, 10H), 1 .70-1.45 (m, 4H), 1.33 (m, 2H); MS (ES+) m/z 406.2 [M+H]+.
1 -(1 -(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanoyl)pyrrolidine-2-carbonyl)-N-(4- ((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,11a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)pyrrolidine-2-carboxamide (79): To a mixture of the 78 (9.3 mg, 0.023 mmol) in 5% methanol/dichloromethane (0.4 mL) was added EEDQ (7 mg, 0.027 mmol). The mixture was stirred for 15 min at an ambient temperature and then 37 (15 mg, 0.021 mmol) was added. The mixture was stirred for 4 h, the reaction mixture was diluted with dichloromethane (2mL) and was aspirated directly onto a 1 mm radial chromatotron plate. The product was eluted with a gradient of 1 to 5% methanol in dichloromethane to provide 6.8 mg (29%) of 79 as a yellow solid: MS (ES+) m/z 1 1 13.51 [M+H]+. Scheme 16
Figure imgf000117_0001
(S)-5-(allyloxy)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)-5- oxopentanoic acid (80): To a mixture of the 2-chlorotrityl resin (1.0 g, 1 .01 mmol) suspended in dichloromethane (10 ml) was added Fmoc-Glu-(OAIIyl)-OH (81 ) (409 mg, 1 .0 mmol) and DIPEA (173 μΙ_, 1 .0 mmol). The reaction mixture was shaken for 5 min, and an additional portion of DIPEA (260 μΙ_, 1.5 mmol) was added and the mixture was shaken for 1 h. Methanol (0.8 mL) was added and the mixture was shaken for 5 min, before being filtered and washed with DMF (6X), dichloromethane (6X), diethyl ether (6X) and dried under reduced pressure. The resulting resin was subjected to 20% piperidine in dichloromethane (10 mL) for 1 h, before being filtered and washed with DMF (6X), dichloromethane (6X), diethyl ether (6X) and dried under reduced pressure.
To a mixture of the Fmoc-Val-OH (82) (1 .03 g, 3.30 mmol)) in DMF (7 mL) was added DIPEA (1.0 mL) and HATU (1.1 g, 3.03 mmol). After thorough mixing, the solution as aspirated into a 10 mL syringe containing the resin prepared above. The mixture was capped and shaken for 16 h. The resin was washed with DMF (6X), dichloromethane (6X) and ether (6X). A small portion (10 mg) was isolated and treated with 20% TFA Dichloromethane and the resulting solution analyzed by LC-MS which revealed one high purity peak which displayed the correct mass (MS (ES+) m/z 509.28 [M+H]+). The remaining resin was then treated with 20% piperidine/DMF (8 mL) for 2 h, before being washed with DMF (6X), dichloromethane (6X), diethyl ether (6X) and dried under reduced pressure.
A mixture of allyl chloroformate (529 μΙ_, 5.05 mmol), DIPEA (1.7 mL, 10 mmol) in dichloromethane (10 mL) was prepared and aspirated into a syringe containing the resin above. The mixture was capped and shaken. After approximately 2 h, the reaction mixture was drained, and washed with dichloromethane (6X). A small portion of the resin (~10 mg) was cleaved with 20% TFA dichloromethane and analyzed by LC-MS for masses of starting material and product. The main component was still the unreacted amine, so the resin was again subjected to the conditions described above. After 4 h, the resin was washed with dichloromethane (6X), and then treated repeatedly with 5% TFA in dichloromethane (4 X 7 mL). The resulting solution was concentrated under reduced pressure. The mixture was purified on a 2 mm radial chromatotron plate eluting with 5% methanol/dichloromethane to give 107 mg of 80: 1H-NMR (400 MHz, CDCI3) δ 7.05 (s, 1 H), 5.90 (m, 2H), 5.57 (d, 1 H), 5.29 (d, J = 14.7 Hz, 2H), 5.22 (t, J = 10.9 Hz, 2H), 4.59 (m, 5H), 4.02 (m, 1 H), 2.60-2.40 (m, 2H), 2.37-2.18 (m, 1 H), 2.17-2.02 (m, 2H), 0.96 (d, J=6.4 Hz, 3H), 0.93 (d, J = 6.6 Hz, 3H); MS (ES+) m/z 371.12 [M+H]+.
(S)-allyl 4-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)-5-((4-((S)-7- methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-5-oxopentanoate (83): To a mixture of the acid 80 (30, 0.04 mmol) in 5% methanol/dichloromethane (1 mL) was added EEDQ (20 mg, 0.082 mmol). The mixture was stirred for 30 min at an ambient temperature and then 37 (30 mg, 0.04 mmol) was added and the mixture was stirred for approximately 5 h. Partially purification by aspirating directly onto a 1 mm radial chromatotron plate and eluting with a gradient of 1 % to 5% methanol/dichloromethane afforded a mixture of desired product and 37 (26 mg; ~3:1 respectively) which was carried forward without further purification.
(S)-4-((S)-2-amino-3-methylbutanamido)-5-((4-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2- (4-methoxyphenyl)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8- yl)oxy)propoxy)-5-oxo-5,11 a-dihydro-1H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2- yl)phenyl)amino)-5-oxopentanoic acid (84): To the mixture of 83 and 37 (26 mg) in anhydrous dichloromethane (3 mL) was added Ph3P (0.3 mg, 0.0012 mmol), pyrrolidine (4 μΙ_, 0.048 mmol) and tetrakis palladium (0.7 mg, 0.6 μΐηοΙ). After 2 h, an additional quantity (0.7 mg, 0.6 μΐηοΙ) of tetrakis palladium was added and the reaction was allowed to stir for an additional 1 hr before being concentrated under reduced pressure. The residue was dissolved in DMSO (1 mL), acetonitrile with 0.05% formic acid (1 mL) and water with 0.05% formic acid (1 mL) and purified by preparative reverse phase HPLC. A single fraction of product was collected and lyophilized to give 6 mg (14% for two steps) of 84: MS (ES+) m/z 1078.6 [M+H]+.
(S)-4-((S)-2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanamido)-3- methylbutanamido)-5-((4-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5- oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo- 5,11a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-5- oxopentanoic acid (85): To a mixture of the 84 (6 mg, 6 μΐηοΙ), and 35 (2 mg, 6 μΐηοΙ) in DMF (200 μί) was added DIPEA (3 μ\-, 18 μΐηοΙ) and the reaction mixture was stirred at an ambient temperature. After 1 h, an additional equivalent of 35 (2 mg, 6 μΐηοΙ) was added and the reaction was allowed to continue to stir at an ambient temperature for 3 h. A third equivalent of 35 (2 mg, 6 μΐηοΙ) was added and the mixture was stirred for approximately 1 h, concentrated under reduced pressure, dissolved in dichloromethane and aspirated directly onto a 1 mm radial chromatotron plate and eluted with 5% methanol in
dichloromethane. This gave 2.5 mg (36%) of high purity 85: MS (ES+) m/z 1 147.49 [M+H]+.
(21S,24S)-1 -(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)-21 -isopropyl-24-((4-((S)-7-methoxy- 8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5, 11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5, 11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)phenyl)carbamoyl)-3, 19,22 -trioxo-7,10,13,16-tetraoxa- 4,20,23-triazaheptacosan-27-oic acid (86): To a mixture of the 84 (8mg, 8.4 μΐηοΙ) and Mal-PEG4-NHS (87) (6.5 mg, 12.6 μΐηοΙ) in DMF (200 μΙ_) was added DIPEA (4.3 μΙ_, 25 μΐηοΙ). The reaction mixture was stirred at an ambient temperature for 2 h, and was concentrated under reduced pressure. The resulting residue was dissolved in
dichloromethane and aspirated onto a 1 mm radial chromatotron plate. The material was polar and did not chromatograph on the silica gel-based chromatotron plate. The plate was eluted with methanol to recover the mixture which was isolated under reduced pressure. The residual material was purified via preparative reverse phase HPLC. A single main peak eluted and the fractions were combined, frozen and lyophilized to a residue of 0.9 mg (8 %) of 86: MS (ES+) m/z 1353.04 [M+H]+.
Scheme 17
Figure imgf000120_0001
(S)-6-(dimethylamino)-2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 - yl)hexanamido)hexanoic acid (88): To a mixture of the 2-chlorotrityl resin (1 g, 1 .01 mmol) in CH2CI2 (10 ml) was added Fmoc-Lys(Me)2-OH (89) (432 mg, 1.0 mmol) and
DIPEA (433 μΙ_, 2.5 mmol). The reaction mixture was shaken for 1 h. Methanol (0.8 mL) was added and the mixture was shaken for an additional 5 min, before being filtered and washed with DMF (6x), dichloromethane (6x), diethyl ether (6x) and dried under reduced pressure. The dried resin was subjected to 20% piperidine in DMF (10 mL) for 1 h, before being filtered and washed with DMF (6X), dichloromethane (6X), diethyl ether (6X).
To a mixture of the 39 (3.0 mmol, 633 mg) in DMF (7 mL) was added DIPEA (1 .0 mL) and HATU (1.1 g, 3.03 mmol). After thorough mixing, the solution as aspirated into a 10 mL syringe containing the resin above. The mixture was capped, shaken for 16 h, filtered and the resin washed with DMF (6X), dichloromethane (6X), and ethyl ether (6X). The resin was by repeatedly treating with 5% TFA/dichloromethane (6 mLX5), shaking for 1 min, and then filtering. The resulting solution was concentrated under reduced pressure and under high vacuum. The material was purified by preparatory reverse phase HPLC to give 208 mg of 88: 1H-NMR (400 MHz, CD3OH/CDCI3 1 :1 mixture ) δ 6.73 (s, 2H), 4.41 (m, 1 H), 3.48 (t, 2H), 3.31 (s, 1 H), 3.03 (m, 2H), 2.84 (s, 6H), 2.22 (m, 2H), 1 .87 (m, 2H), 1.78- 1 .52 (m, 6H), 1 .43 (m, 2H), 1 .31 (pent, 2H); MS (ES+) m/z 386.28 [M+H]+.
(S)-6-(dimethylamino)-2-(6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)hexanamido)-N-(4-
((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5,11a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)hexanamide (90): To a mixture of the 88 (9.3 mg, 0.023 mmol) in 5% methanol/dichloromethane (400 μΙ_) was added EEDQ (7 mg, 0.027 mmol). The mixture was stirred for 30 min at an ambient temperature and then 37 (15 mg, 0.021 mmol) was added. After 4 h, the mixture was concentrated under reduced pressure, dissolved in a mixture of DMSO (1 ml_), acetonitrile (2 mL containing 0.05 % formic acid) and water (1 mL containing 0.05% formic acid) and purified by reverse-phase HPLC (method A). Product containing fractions were contaminated with 37, so the fractions were lyophilized to a residue and repurified as described above to give 0.5 mg (2 %) of pure 90: MS (ES+) m/z 537.46 [M+H]/2+.
Scheme 18
Figure imgf000121_0001
AIM ((S)-1 -(((S)-1 -((4-((S)-7-methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo- 5,11 a-dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5,11 a- dihydro-1 H-pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2- yl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (91): To a mixture of the 92 (45 mg,
0.123 mmol) in 5% methanol/dichloromethane (1 mL) was added EEDQ (30.4 mg, 0.123 mmol). The mixture was stirred for 30 min at an ambient temperature and then 37 (30 mg, 0.041 mmol) was added. The reaction mixture was stirred for approximately 5 h and then purified on a 1 mm radial chromatotron plate eluting with 5% methanol/dichloromethane to give 22mg (55%) of 91 which was not characterized but carried on directly.
1 -(3-(2,5-dioxo-2,5-dihydrc-1 H-pyrrol-1 -yl)propanamido)-N-((S)-1 -(((S)-1 -((4-((S)-7- methoxy-8-(3-(((S)-7-methoxy-2-(4-methoxyphenyl)-5-oxo-5, 11 a-dihydro-1 H- pyrrolo[2,1 -c][1 , 4]benzodiazepin-8-yl)oxy)propoxy)-5-oxo-5, 11 a-dihydro-1 H- pyrrolo[2,1 -c][1 ,4]benzodiazepin-2-yl)phenyl)amino)-1 -oxopropan-2-yl)amino)-3- methyl-1 -oxobutan-2-yl)-3,6,9,12-tetraoxapentadecan-15-amide (93): To a solution of the 91 (22 mg, 0.022 mmol) in anhydrous dichloromethane (3 mL) was added Ph3P (0.3 mg, 0.0012 mmol), pyrrolidine (4 μΙ_, 0.048 mmol) and tetrakis palladium (0.7 mg, 6 μΐηοΙ). After approximately 2h, the reaction mixture was purified on a 1 mm radial chromatotron plate eluting with 5% to 10% methanol/dichloromethane. The major band was collected and concentrated to a residue which was dissolved in DMF (0.2 mL) and reacted with NHS ester 87 (10 mg, 0.19 mmol). The reaction was allowed to stir for 30 min, concentrated and purified by radial chromatography on a 1 mm plate eluting with 5%
methanol/dichloromethane to give 3.2 mg (1 1 %) of 93: MS (ES+) m/z 1294.7 [M+H]+. Scheme 19
Figure imgf000122_0001
(E)-6-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)-N'-(4-((S)-7-methoxy-8-((5-(((S)-7- methoxy-2-(4-methoxyphenyl)-5-oxo-5,11 a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-8-yl)oxy)pentyl)oxy)-5-oxo-5,11a-dihydro-1 H-pyrrolo[2,1 - c][1 ,4]benzodiazepin-2-yl)benzylidene)hexanehydrazide (94): To a mixture of the aldehyde 95 (5.4 mg, 7 μΐηοΙ) in 5% methanol/dichloromethane at 0 °C was added the hydrazide-TFA salt 96 (4.5 mg, 14 μΐηοΙ). The reaction mixture was allowed to warm to an ambient temperature and stir for 5 h before being concentrated under reduced pressure and purified on a silica gel column eluting with 3% methanol/dichloromethane to give 2.2 mg (32%) of 94: MS (ES+) m/z 974.49 [M+H]+. Scheme 20
Figure imgf000123_0001
(S)-tert-butyl 2-((S)-2-amino-3-methylbutanamido)propanoate (97): To a mixture of the alanine-O-fert-butyl ester hydrogen chloride salt (98) (500 mg, 2.76 mmol) in
dichloromethane (5 mL) was added Fmoc-val-OSu (99) (1.09 g, 2.51 mmol). DIPEA (0.96 ml, 5.5 mmol) was added and the reaction mixture was allowed to stir at an ambient temperature for 16 h. The mixture was poured into dichloromethane (100 mL) and washed with 1 N HCI (50 mL) and water (50 mL) before being dried over magnesium sulfate. The material was chromatographed on a 2 mm radial chromatotron plate eluting with 1 to 5% methanol/dichloromethane gradient and product containing fractions were combined and concentrated. The resulting residue was dissolved in dichloromethane (16 mL) and piperidine (4 mL) was added. The mixture was stirred for 10 min before being
concentrated under reduced pressure. The resulting residue was chromatographed on a 2 mm plate eluting first with ammonia-saturated dichloromethane followed by 5% methanol in ammonia-saturated dichloromethane to give 494 mg (2.02 mmol, 81 % for two steps) of 97: 1H-NMR (400 MHz, CDCI3) δ 7.78 (bs, 1 H), 4.47 (m, 1 H), 3.30 (d, 1 H), 2.30 (m, 1 H), 1 .38 (d, 3H), 1.47 (s, 9H), 1.00 (d, J = 7.0 Hz, 3H), 0.84 (d, J = 6.9 Hz, 3H).
(S)-tert-butyl 2-((S)-2-(4-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)benzamido)-3- methylbutanamido)propanoate (100): To a mixture of the 97 (100 mg, 0.41 mmol) and 4-maleimidobenzoic acid (101 ) (98 mg, 0.45 mmol) was added dichloromethane (5 mL), followed by TBTU (157 mg, 0.49 mmol) and DIPEA (212 uL, 1 .23 mmol). The mixture was stirred at an ambient temperature for 16 h and then purified on a 2 mm radial chromatotron plate eluting with 50% ethyl acetate in hexanes to give 95 mg (51 %) of 100: 1H-NMR (400 MHz, CDCIs) δ 7.85 (d, J = 6.6 Hz, 2H), 7.42 (d, J = 6.6 Hz, 2H), 6.81 (s, 2H), 6.38 (bs, 1 H), 4.43 (m, 2H), 2.14 (sept, J = 6.6 Hz, 1 H), 1.41 (s, 9H), 1.31 (d, J = 7.0 Hz, 3H), 0.98 (m, 6H); MS (ES") m/z 441 .90 [M-H]\
(R)-2-((S)-2-(4-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)benzamido)-3- methylbutanamido)propanoic acid (53): To a mixture of 100 (47 mg, 0.1 1 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (5 mL) and the reaction mixture was monitored by TLC (50% ethyl acetate in hexane, after pumping down the TLC plate under high vacuum for 5 min). After 75 min, no starting material could be detected by TLC. The reaction was performed a second time using the same conditions and material from both reactions were combined and purified on a 2 mm radial chromatotron plate eluting with a gradient from 5-10% methanol in dichloromethane. The yield was 42 mg (49%) of 53: 1H- NMR (400 MHz, CDCI3) δ 7.92 (d, J = 6.6 Hz, 2H), 7.51 (d, J = 6.6 Hz, 2H), 7.0 (m, 1 H), 6.89 (s, 2H), 6.70 (s, 1 H), 4.60 M, 1 H), 2.22 (m, 1 H), 1 .18 (d, J = 6.6 Hz, 3H), 1.04 (m, 6H); MS (ES+) m/z 388.02 [M+H]+.
(S)-2-((S)-2-(2-iodoacetamido)-3-methylbutanamido)propanoic acid (102): To a mixture of the 97 (100 mg, 0.41 mmol) in dichloromethane was added iodoacetamide-NHS ester (103) (1 15 mg, 0.41 mmol) and the mixture was stirred at an ambient temperature. After 30 min, the mixture was aspirated onto a 1 mm chromototron plate and eluted with ethyl acetate in hexanes (1 :1 ). A single band was collected and the structure was confirmed: 1H-NMR (400 MHz, CDCI3) δ 6.70 (d, J = 7.8 Hz, 1 H), 6.27 (d, J = 7.0 Hz, 1 H), 4.45 (m, 1 H), 4.26 (dd, J = 8.6, 6.3 Hz, 1 H), 3.72 (quart, J = 1 1.3 Hz, 2H), 2.13 (sept, J = 6.5 Hz, 1 H), 1 .47 (s, 9H), 1 .38 (d, J = 7.1 Hz, 3H), 0.99 (m, 6H); MS (ES+) m/z 412.87
[M+H]+.
(S)-2-((S)-2-(2-iodoacetamido)-3-methylbutanamido)propanoic acid (55): See procedure for the synthesis of (R)-2-((S)-2-(4-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 - yl)benzamido)-3-methylbutanamido)propanoic acid (53). This gave 22 mg (15% for two steps): 1 H -NMR (400 MHz, D6-DMSO) δ 8.27 (d, J = 9.4 Hz, 1 H), 4.24 (m, 2H), 3.97 (bs, 2H), 3.83 (d, J = 9.4 Hz, 1 H), 3.71 (d, J = 9.6 Hz, 1 H), 2.07 (m, 1 H), 1.33 (d, J = 7.3 Hz, 3H), 0.93 (d, J = 6.7 Hz, 3H), 0.89 (d, J = 6.9 Hz, 3H); MS (ES") m/z 354.84 [M-H]\ Scheme 21
Figure imgf000125_0001
PBD dimers linked through aliphatic amines (Scheme 21). PBD dimers containing aliphatic amines, such as a benzyl amine (Example 9), are synthesized with peptidic linkers, the glucuronide linker, and/or linkers dependent on mAb degradation for release (i.e., non-cleavable linkers). Drug linkers conjugated through a benzyl amine will include: (1 ) a cleavable peptide employing chemistry similar to Scheme 1 ; (2) direct attachment with a maleimidocaproyi group (a noncleavable linker) (Scheme 2); (3) a glucuronide linker, prepared as described in Scheme 6.
Scheme 22
Figure imgf000126_0001
Generic peptide linked 2-, 3-, and 4-aniline PBD dimers (Scheme 22). PBD dimers with anilines at the 2-, 3-, and 4-positions will be conjugated to peptide-based linkers, employing the chemistry described in Scheme 1 , or attached directly with maleimidocaproic acid, as exemplified in Scheme 2.
Example 14: Preparation of PDB Dimer Conjugates
Antibody-drug conjugates were prepared as previously described (see Doronina et al., Nature Biotechnology, 21 , 778-784 (2003)) or as described below. Briefly, for maleimide drug-linker the mAbs (4-5 mg/mL) in PBS containing 50 mM sodium borate at pH 7.4 were reduced with tris(carboxyethyl)phosphine hydrochloride (TCEP) at 37 °C. The progress of the reaction, which reduces interchain disulfides, was monitored by reaction with 5,5'- dithiobis(2-nitrobenzoic acid) and allowed to proceed until the desired level of thiols/mAb was achieved. The reduced antibody was then cooled to 0 °C and alkylated with 1.5 equivalents of maleimide drug-linker per antibody thiol. After 1 h, the reaction was quenched by the addition of 5 equivalents of N-acetyl cysteine. Quenched drug-linker was removed by gel filtration over a PD-10 column. The ADC was then sterile-filtered through a 0.22 μιη syringe filter. Protein concentration was determined by spectral analysis at 280 nm and 329 nm, respectively, with correction for the contribution of drug absorbance at 280 nm. Size exclusion chromatography was used to determine the extent of antibody aggregation and RP-HPLC confirmed the absence of remaining NAC-quenched drug- linker.
For halo acetamide-based drug linkers, conjugation was performed generally as follows: To a 10 mg/mL solution of reduced and reoxidized antibody (having introduced cysteines by substitution of S239C in the heavy chains (see infra)) in 10 mM Tris (pH 7.4), 50 mM NaCI, and 2 mM DTPA was added 0.5 volumes of propylene glycol. A 10 mM solution of acetamide-based drug linker in dimethylacetamide was prepared immediately prior to conjugation. An equivalent amount of propylene glycol as added to the antibody solution was added to a 6-fold molar excess of the drug linker. The dilute drug-linker solution was added to the antibody solution and the pH was adjusted to 8.0-8.5 using 1 M Tris (pH 9). The conjugation reaction was allowed to proceed for 45 minutes at 37 °C. The conjugation was verified by reducing and denaturing reversed phase PLRP-S chromatography. Excess drug linker was removed with Quadrasil MP resin (Sigma Aldrich; Product # 679526) and the buffer was exchanged into 10 mM Tris (pH 7.4), 50 mM NaCI, and 5% propylene glycol using a PD-10 desalting column (GE Heathcare; Product * 17-0851-01 ).
Engineered hlgG1 antibodies with introduced cysteines: CD70 antibodies containing a cysteine residue at position 239 of the heavy chain (hi F6d) were fully reduced by adding 10 equivalents of TCEP and 1 mM EDTA and adjusting the pH to 7.4 with 1 M Tris buffer (pH 9.0). Following a 1 hour incubation at 37 °C, the reaction was cooled to 22 °C and 30 equivalents of dehydroascorbic acid were added to selectively reoxidize the native disulfides, while leaving cysteine 239 in the reduced state. The pH was adjusted to 6.5 with 1 M Tris buffer (pH 3.7) and the reaction was allowed to proceed for 1 hour at 22 °C. The pH of the solution was then raised again to 7.4 by addition of 1 M Tris buffer (pH 9.0). 3.5 equivalents of the PBD drug linker in DMSO were placed in a suitable container for dilution with propylene glycol prior to addition to the reaction. To maintain solubility of the PBD drug linker, the antibody itself was first diluted with propylene glycol to a final concentration of 33% (e.g., if the antibody solution was in a 60 mL reaction volume, 30 mL of propylene glycol was added). This same volume of propylene glycol (30 mL in this example) was then added to the PBD drug linker as a diluent. After mixing, the solution of PBD drug linker in propylene glycol was added to the antibody solution to effect the conjugation; the final concentration of propylene glycol is 50%. The reaction was allowed to proceed for 30 minutes and then quenched by addition of 5 equivalents of N-acetyl cysteine. The ADC was then purified by ultrafiltration through a 30 kD membrane. (Note that the concentration of propylene glycol used in the reaction can be reduced for any particular PBD, as its sole purpose is to maintain solubility of the drug linker in the aqueous media.) Example 15: Determination of In Vitro Activity of Selected Conjugates
The in vitro cytotoxic activity of the selected antibody drug conjugates was assessed using a resazurin (Sigma, St. Louis, MO, USA) reduction assay (reference: Doronina et al., Nature Biotechnology, 2003, 27, 778-784). The antibody drug conjugates were prepared as described above in Example 13.
For the 96-hour assay, cells cultured in log-phase growth were seeded for 24 h in 96-well plates containing 150 μΙ_ RPMI 1640 supplemented with 20% FBS. Serial dilutions of ADC in cell culture media were prepared at 4x working concentration; 50 μΙ_ of each dilution was added to the 96-well plates. Following addition of ADC, the cells were incubated with test articles for 4 days at 37 °C. Resazurin was then added to each well to achieve a 50 μΜ final concentration, and the plates were incubated for an additional 4 h at 37 °C. The plates were then read for the extent of dye reduction on a Fusion HT plate reader (Packard Instruments, Meridien, CT, USA) with excitation and emission wavelengths of 530 and 590 nm, respectively. The IC50 value, determined in triplicate, is defined here as the concentration that results in a 50% reduction in cell growth relative to untreated controls.
Referring to Table 4 (infra), the in vitro cytotoxicity of ADCs having para-aniline PBD dimers using the 96 hour assay is shown. The ADCs were tested against CD70+ CD30" cell lines and a control CD70" CD30" cell line. The antibodies used were a CD70 antibody, humanized 1 F6 (see Published U.S. Application No. 2009-148942), a CD30 antibody, chimeric AC10 (see Published U.S. Application No. 2008-0213289) and a CD70 antibody (humanized 1 F6) having introduced cysteine residues at amino acid heavy chain position 239 (according to the EU numbering system) (indicated as h1 F6d). Conjugates having a maleimidyl-peptide linker (drug linker compound 38) had a lower IC50 than conjugates with a maleimidyl or acetamide-based linker (compounds 40 and 41 , respectively).
In vitro cytotoxic activity of ADCs bearing drug linkers derived from para-aniline PBD dimer 37: Table 4. In vitro cytotoxic activity on CD70+ cell lines (ng/mL), all ADCs 2 drugs/mAb
renal cell carcinoma AML
CD70+/30- CD70-/30-
786-0 Caki-1 769-P ACHN HEL9217 h1 F6d38 30 5 1378
F6-38 4 1 18 26
CAC1038 1052 4005 508
F640 71 13 1764
CAC1040 2644 1264
F641 580 1243
CAC1041 1 153 1 121
Referring to Table 5, the in vitro cytotoxicity of ADCs conjugate to PBD dimers on CD30+ cell lines using the 96 hour assay is shown. The ADCs were tested against CD30+CD70+ cell lines and a CD70" CD30+ cell line. The antibodies used were a CD70 antibody, humanized 1 F6 (see Published U.S. Application No. 2009-148942) and a CD30 antibody, chimeric AC10 (see Published U.S. Application No. 2008-0213289). Conjugates having a maleimidyl-peptide linker (drug linker compound 38) generally had a lower IC50 than conjugates with a maleimidyl or acetamide-based linker (compounds 40 and 41 , respectively).
Table 5. In vitro cytotoxic activity on CD30+ cell lines (ng/mL), all ADCs 2 drugs/mAb
ALCL Hodgkin lymphoma
CD70-/30+ CD70+/30+
Karoas 299 L428 L540cv L1236 Hs445 h1 F638 1 165 59 4 >10,000 5
CAC1038 0.8 7 3 2012 0.2 h1 F640 2195 7867 2557
CAC1040 621 3172 134
h1 F641 1330 3549 755
CAC1041 340 957 13
In vitro cytotoxic activity of ADCs bearing drug linkers derived from mefa-aniline PBD dimer Referring to Table 6, the in vitro cytotoxicity of ADCs containing PBD dimers on CD30+ cell lines using the 96 hour assay is shown. The activity was tested against CD30+CD70+ cell lines and a CD70" CD30+ cell line. The antibodies used were a CD70 antibody, humanized 1 F6 (see Published U.S. Application No. 2009-148942) and a CD70 antibody (humanized 1 F6) having introduced cysteine residues at amino acid heavy chain position 239
(according to the EU numbering system) (indicated as h1 F6d). Conjugates having a maleimidyl-peptide linker (drug linker compound 43) and a glucuronide linker (48) generally had a lower IC50 than conjugates with a maleimidyl-based linker (compound 44).
Table 6. In vitro cytotoxic activity on CD70+ cell lines (ng/mL)
Hodgkin
renal cell carcinoma lymphoma
Caki-1 786-0 L428
h1 F6d43 (2 dr/mAb) 7 39 >10,000
laG43 (2 dr/mAb) >10.000 >10.000
h1 F644 (3.5 dr/mAb) 1 124 2142
laG44 (3.5 dr/mAb) 1491 1242
h1 F6d48 (2 dr/mAb) 89 4093
lgG48 (2 dr/mAb) 2939 6376
In vitro cytotoxic activity of ADCs bearing drug linkers derived from para- and mefa-aniline PBD dimers 38 and 42 (respectively): Referring to Table 7, the in vitro cytotoxicity of ADCs containing PBD dimers on CD70+ cell lines using the 96 hour assay is shown. The activity was tested against CD70+ cell lines L428 and 7860 and a CD70" AML cell line. The antibodies used were a CD70 antibody, humanized 1 F6 (see Published U.S. Application No. 2009-148942) and a CD70 antibody (humanized 1 F6) having introduced cysteine residues at amino acid heavy chain position 239 (according to the EU numbering system) (indicated as hi F6d). Conjugates having a maleimidyl-peptide linker with a mefa-aniline (drug linker compound 43) were somewhat less active than those having a maleimidyl-peptide linker with a para-aniline (drug linker compound 38). Reducing the drug loading of the mefa-aniline compound to 2 per antibody reduced the activity. Conjugates with a glucuronide linker of the para-aniline compound (48) generally had a lower IC50 than conjugates with a maleimidyl-based linker (compound 39). Further, an aryl maleimide of the para-aniline compound (54) has no activity on these cell lines. Further, a conjugate having a maleimidyl linker conjugated directly to compound 42 has reduced activity as compared with conjugate hi F6-43 (data not shown).
Table 7. In vitro cytotoxic activity on CD70+ cell lines (ng/mL)
Hodgkin Renal cell
IvmDhoma carcinina control
L428 7860
h1 F6- 43 (4 dr/mAb) 404 11 1205
h1 F6d- 43 (2 dr/mAb) Max inhib.= 40% 200 1625
h1 F6d48 (2 dr/mAb) 4093 89 1964
h1 F6 54 (4 dr/mAb) No effect No effect No effect
h1 F6- 38 (2 dr/mAb) 230 (n=2) 25 (n = 3) 503
In vitro cytotoxic activity of ADCs bearing drug linkers derived from aniline-linked PBD dimers
Referring to Table 8, the in vitro cytotoxicity of ADCs containing PBD dimers on CD70+ cell lines using the 96 hour assay is shown. The activity was tested against CD70+ cell lines Caki-1 and L428 and a CD70" cell line. The antibody used was a CD70 antibody
(humanized 1 F6) having introduced cysteine residues at amino acid heavy chain position 239 (according to the EU numbering system) (indicated as hi F6d). Linkage of a PBD through an amine at the ortho position via a non-cleavable linker (compound 68) markedly reduced activity, as compared with an ADC linked via a para-aniline-linked deavable linker (compound 54). Compounds 73 and 85, having a deavable linker, showed comparable activity to compound 54; both of these compounds are linked via a para-aniline.
Compounds with deavable linkers requiring more stringeng cleavage, compounds 79 and 90, showed somewhat reduced activity, as compared to compound 54. Table 8. In vitro cytotoxic activity on CD70+ cell lines (ng/mL)
Figure imgf000132_0001
In vitro cytotoxic activity of ADCs bearing drug linkers derived from aniline-linked PBD dimers Referring to Table 9, the in vitro cytotoxicity of ADCs containing PBD dimers on CD70+ cell lines using the96 hour assay is shown. The activity was tested against CD70+ cell lines Caki-1 and L428 and two CD70" leukemia cell lines. The antibodies used were a CD70 antibody, humanized 1 F6 (see Published U .S. Application No. 2009-148942) and a CD70 antibody (humanized 1 F6) having introduced cysteine residues at amino acid heavy chain position 239 (according to the EU numbering system) (indicated as h 1 F6d). Compound 56, having a cleavable linker linked to the antibody via an acetamide showed comparable activity to compound 38. A glucuronide-linked version of the mefa-aniline linked PBD dimer, compound 48, demonstrated little activity in this assay. Compound 58, having five methylene groups in the PBD bridge, demonstrated comparable activity to compound 38, having three methylene groups in the PBD bridge. Table 9: In vitro cytotoxic activity on CD70+ cell lines (ng/
Renal Cell Leukemia
ADCs Caki-1 786-0 CD70" Line 1 CD70" Line 2
(CD70 #135,000) (CD70 #190,000)
h1 F6d-56 (1.8dr/Ab) 3 6 1672 Max lnh=50% h1 F6d-48 (0.6dr/Ab) Max lnh=45% Max lnh=35% No Effect No Effect h1 F6d-58 (1.9dr/Ab) 0.5 2 1750 4847
5 15
h1 F6d-38 (2dr/Ab) (3-5, n=4) (5-30, n=4) 2082 7188
Example 16: Determination of In Vivo Cytotoxicity of Selected Conjugates
All studies were conducted in concordance with the Animal Care and Use Committee in a facility fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care. In vivo tolerability was first assessed to ensure that the conjugates were tolerated at clinically relevant doses. BALB/c mice were treated with escalating doses of ADC formulated in PBS with 0.01 % Tween 20. Mice were monitored for weight loss following drug treatment; those that experienced 20% weight loss or other signs of morbidity were euthanized. The antibodies used were a CD70 antibody, humanized 1 F6 (see Published U.S. Application No. 2009-148942) and a CD30 antibody, chimeric AC10 (see Published U.S. Application No. 2008-0213289). Referring to Figure 1 , the results of a weight loss study are shown using cAC10-val-ala- SG3132(2) (cAC10-compound 38). A single dose of the conjugate administered at 5 mg administered either IP or IV resulted in little weight loss. A higher dose of the conjugate (15 mg/kg) caused weight loss in the mice. Referring to Figure 2, the results of a weight loss study are shown using hi F6-val-ala- SG3132(2) (h1 F6-compound 38). A single dose of the conjugate administered at 5 mg administered IP resulted in some weight loss. A higher dose of the conjugate (10 mg/kg) caused significant weight loss in the mice. Treatment studies were conducted in two CD70+ renal cell carcinoma xenograft models. Tumor (786-0 and Caki-1 ) fragments were implanted into the right flank of Nude mice. Mice were randomized to study groups (n=5) on day eight (786-0) or nine (Caki-1 ) with each group averaging around 100 mm3. The ADC or controls were dosed ip according to a q4dx4 schedule. Tumor volume as a function of time was determined using the formula (L x W2)/2. Animals were euthanized when tumor volumes reached 1000 mm3. Mice showing durable regressions were terminated around day 100 post implant.
Referring to Figure 3, the results of a treatment study using an h1 F6-val-ala-SG3132(2) (h1 F6-compound 38) conjugate are shown. A control conjugate, cAC10-val-ala-SG3132(2) (cAC10-compound 38), was also used. Mice administered doses of the h1 F6 conjugate at 0.1 mg/kg exhibited some tumor reduction, while higher doses at 0.3 mg/kg and 1 mg/kg appeared to exhibit complete tumor reduction. The control conjugate (non-binding) was less active the h1 F6 conjugates.
Referring to Figure 4, the results of a treatment study using an h1 F6-mc-val-ala-SG3132(2) (h1 F6-compound 38) conjugate are shown. A control conjugate, cAC10-mc-val-ala- SG3132(2) (cAC10-compound 38), was also used. Mice administered doses of the h1 F6 conjugate at 1 mg/kg appeared to exhibit complete tumor reduction. Mice administered lower doses at 0.3 mg/kg and 0.1 mg/kg exhibited lesser tumor reduction, respectively.
The control conjugate (non-binding) was less active the h1 F6 conjugate administered at a similar dose, although it exhibited more activity than the hi F6 conjugate administered at lower doses. The hi F6 conjugate was also more active than an hi F6-vc-MMAE conjugate (Published U.S. Application No. 2009-0148942) administered at higher doses.
Referring to Figure 5, the results of a treatment study using a two loaded antibody hi Fed- linked to compound 38 (h1 F6d-38) compared to a two-loaded non-binding control, HOOd conjugated to the same compound (hOOd-38). The model was a Caki subcutaneous model in Nude mice. Doses were 0.1 , 0.3 and 1 mg/kg q7dX2. The highest two doses of the h1 F6 conjugate demonstrated complete regressions as 1 mg/kg and substantial tumor delay at 0.3 mg/kg. The non-binding control demonstated tumor delay at the 1 mg/kg dose.
Referring to Figure 6, the results of a treatment study using a two loaded antibody hi F6d- linked to compound 38 (h1 F6d-38) compared to a two-loaded non-binding control, HOOd conjugated to the same compound (hOOd-38). The model was a 786-0 subcutaneous model in Nude mice. Doses were 0.1 , 0.3 and 1 mg/kg q7dX2. All three doses of the hi F6 conjugate demonstrated complete regressions or tumor delay, while the non-binding control demonstated tumor delay.

Claims

1 . A Conjugate having formula I:
Figure imgf000136_0001
or a pharmaceutically acceptable salt or solvate thereof;
wherein L is a Ligand unit,
LU is a Linker unit,
p is 1 to 20; and
D is a Drug unit comprising a PBD dimer having the following formula II
Figure imgf000136_0002
wherein:
R2 is of formula III:
' 2'X ]
where A is a C5-7 aryl group, X is selected from the group comprising: -0-, -S-, -C(0)0-, - C(O)-, -NH(C=0)-, -N(RN)-, wherein RN is selected from the group comprising H, C1-4 alkyl and (C2H40)mCH3, where m is 1 to 3, and either:
(i) Q1 is a single bond, and Q2 is selected from a single bond and -Z-(CH2)n-, where Z is selected from a single bond, O, S and NH and is from 1 to 3; or
(ii) Q1 is -CH=CH-, and Q2 is a single bond;
R12 is a C5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, Ci-7 alkyl, C3-7 heterocyclyl, dimethyl- aminopropyloxy, piperazinyl and bis-oxy-Ci-3 alkylene;
R6 and R9 are independently selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR', nitro, Me3Sn and halo;
where R and R' are independently selected from optionally substituted C1 -12 alkyl, C3-2o heterocyclyl and C5-20 aryl groups;
R7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NHRR', nitro, Me3Sn and halo; either:
(a) R10 is H, and R11 is OH or ORA, where RA is C1-4 alkyl; (b) R and R form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or
(c) R10 is H and R1 1 is SOzM, where z is 2 or 3 and M is a monovalent
pharmaceutically acceptable cation;
R" is a C3-12 alkylene group, which chain may be interrupted by one or more heteroatoms, selected from the group consisting of O, S, NH, and an aromatic ring;
Y is selected from O, S, or NH;
R6 , R7 , R9 are selected from the same groups as R6, R7 and R9 respectively and R10 and R11 are the same as R10 and R11 , wherein if R11 and R11 are SOzM, M may represent a divalent pharmaceutically acceptable cation.
2. The Conjugate according to claim 1 , wherein R7 is selected from H , OH and OR.
3. The Conjugate according to claim 2, wherein R7 is a Ci-4 alkyloxy group. 4. The Conjugate according to one of the preceding claims, wherein Y is O.
5. The Conjugate according to any one of the preceding claims, wherein R" is C3-7 alkylene. 6. The Conjugate according to any one of the preceding claims, wherein R9 is H.
7. The Conjugate according to any one of the preceding claims, wherein R6 is selected from H and halo. 8. The Conjugate according to any one of the preceding claims, wherein A is phenyl.
9. The Conjugate according to any one of the preceding claims, wherein X is selected from -0-, -S-, or -NH- 10. The Conjugate according to any one of the preceding claims, wherein Q1 is a single bond.
1 1 . The Conjugate according to claim 10, wherein Q2 is a single bond.
12. The Conjugate according to claim 10, wherein Q2 is -Z-(CH2)n-, Z is O or S and n is 1 or 2.
13. The Conjugate according to any one of claims 1 to 9, wherein Q1 is -CH=CH-.
14. The Conjugate according to any one of claims 1 to 13, wherein R12 is a C5-7 aryl group.
15. The Conjugate according to claim 14, wherein R12 is phenyl.
16. The Conjugate according to any one of claims 1 to 13, wherein R12 is a C8-io aryl group.
17. The Conjugate according to any one of claims 1 to 16, wherein R12 bears one to three substituent groups.
18. The Conjugate according to any one of claims 1 to 17, wherein R10 and R11 and form a nitrogen-carbon double bond.
19. The Conjugate according to any one of claims 1 to 18, wherein R6 , R7 , R9 , R10 , R11 and Y' are the same as R6, R7, R9, R10, R11 and Y respectively.
20. The Conjugate according to any one of claims 1 -19, wherein the Linker unit (LU) has the formula 1 a or 1 b:
- "A1 a -L <-1 s -L <-2 y -
(la)
wherein:
-A1- is a Stretcher unit,
a is 1 or 2,
L1 - is a Specificity unit,
s is an integer ranging from 0 to 12,
-L2- is a Spacer unit, and
y is 0, 1 or 2, and
p is from 1- 20; or
Figure imgf000138_0001
I L - (A1 a- L2 y -D)p (lb) wherein:
-A1- is a Stretcher unit linked to a Stretcher unit (L2),
a is 1 or 2,
L1 - is a Specificity unit linked to a Stretcher unit (L2),
s is an integer ranging from 1 to 12,
-L2- is a Spacer unit,
y is 1 or 2, and
p is from 1 to 20.
The Conjugate of claim 20, wherein the Linker unit (LU) has formula 1 a. The Conjugate of claim 21 , wherein A1 is selected from:
Figure imgf000139_0001
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6;
Figure imgf000139_0002
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6;
Figure imgf000139_0003
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30; or
Figure imgf000140_0001
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, n is 0 or 1 , and m is 0 to 30.
23. The Conjugate of claim 21 , wherein A1 is:
Figure imgf000140_0002
where the asterisk indicates the point of attachment to L1, the wavy line indicates the point of attachment to the Ligand unit, and n is 0 to 6.
24. The Conjugate of claim 23, wherein n is 5.
25. The Conjugate of any of claims 20-24, wherein L1 comprises an amino acid sequence.
26. The Conjugate of claim 25, wherein L1 is a dipeptide.
27. The Conjugate of claim 26, wherein L1 is selected from the group consisting of valine-alanine, valine-citrulline and phenylalanine-lysine.
28. The Conjugate of any of claims 20-27, wherein y is 0.
29. The Conjugate of any of claims 20-27, wherein y is 1 or 2.
30. The Conjugate of claim 29, wherein L2 is:
Figure imgf000140_0003
where the asterisk indicates the point of attachment to the Drug unit, the wavy line indicates the point of attachment to the L1, Y is -N(H)-, -0-, -C(=0)N(H)- or -C(=0)0-, and n is 0 to 3.
31. The Conjugate of claim 30, wherein L2 is:
Figure imgf000141_0001
32. The use of a Conjugate, or a pharmaceutically acceptable salt or solvate thereof, according to any one of the preceding claims, in the manufacture of a medicament for treating a proliferative disease or an autoimmune disease.
33. The use of a Conjugate, or a pharmaceutically acceptable salt or solvate thereof, according to any one of the preceding claims for treating a proliferative disease or an autoimmune disease.
34. A method of treating a mammal having a proliferative disease or an autoimmune disease, comprising administering an effective amount of the Conjugate of any one of claims 1-32, or a pharmaceutically acceptable salt or solvate thereof.
PCT/US2011/032664 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates WO2011130613A1 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
NZ602932A NZ602932A (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates
KR1020167029858A KR101772354B1 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
AU2011239522A AU2011239522B2 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates
EP21213472.0A EP4039280A1 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
US13/641,219 US9242013B2 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates
CA2795349A CA2795349C (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
CN2011800298674A CN103068405A (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates
EA201290838A EA024118B1 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
EP11716754.4A EP2558127B1 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
BR112012026801A BR112012026801B8 (en) 2010-04-15 2011-04-15 targeted pyrrolobenzodiazepine conjugates, pharmaceutical composition, use thereof for treatment of a proliferative or autoimmune disease and drug binding
MX2012011900A MX2012011900A (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates.
KR1020127028121A KR101671360B1 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
JP2013505171A JP5870400B2 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazepine conjugates
ZA2012/07317A ZA201207317B (en) 2010-04-15 2012-10-01 Targeted pyrrolobenzodiazapine conjugates
IL222269A IL222269A (en) 2010-04-15 2012-10-09 Targeted pyrrolobenzodiazepine conjugates
US14/995,944 US9592240B2 (en) 2010-04-15 2016-01-14 Targeted pyrrolobenzodiazapine conjugates
US15/422,000 US20170143846A1 (en) 2010-04-15 2017-02-01 Targeted pyrrolobenzodiazapine conjugates
IL252864A IL252864B (en) 2010-04-15 2017-06-13 Targeted pyrrolobenzodiazapine conjugates
US15/951,753 US20180228916A1 (en) 2010-04-15 2018-04-12 Targeted pyrrolobenzodiazapine conjugates
US16/381,448 US10561739B2 (en) 2010-04-15 2019-04-11 Targeted pyrrolobenzodiazapine conjugates

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32462310P 2010-04-15 2010-04-15
US61/324,623 2010-04-15

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/641,219 A-371-Of-International US9242013B2 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates
US14/995,944 Continuation US9592240B2 (en) 2010-04-15 2016-01-14 Targeted pyrrolobenzodiazapine conjugates

Publications (1)

Publication Number Publication Date
WO2011130613A1 true WO2011130613A1 (en) 2011-10-20

Family

ID=44150264

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/032664 WO2011130613A1 (en) 2010-04-15 2011-04-15 Targeted pyrrolobenzodiazapine conjugates

Country Status (14)

Country Link
US (5) US9242013B2 (en)
EP (2) EP4039280A1 (en)
JP (1) JP5870400B2 (en)
KR (2) KR101772354B1 (en)
CN (2) CN107019804A (en)
AU (1) AU2011239522B2 (en)
BR (1) BR112012026801B8 (en)
CA (1) CA2795349C (en)
EA (1) EA024118B1 (en)
IL (2) IL222269A (en)
MX (2) MX2012011900A (en)
NZ (1) NZ602932A (en)
WO (1) WO2011130613A1 (en)
ZA (1) ZA201207317B (en)

Cited By (114)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012112708A1 (en) * 2011-02-15 2012-08-23 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and methods of preparation
WO2013041606A1 (en) 2011-09-20 2013-03-28 Spirogen Sàrl Pyrrolobenzodiazepines as unsymmetrical dimeric pbd compounds for inclusion in targeted conjugates
WO2013053873A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines
WO2013055993A1 (en) 2011-10-14 2013-04-18 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
WO2013053871A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines
US8426402B2 (en) 2009-02-05 2013-04-23 Immunogen, Inc. Benzodiazepine derivatives
WO2013119960A2 (en) 2012-02-08 2013-08-15 Stem Centrx, Inc. Novel modulators and methods of use
WO2013177481A1 (en) 2012-05-25 2013-11-28 Immunogen, Inc. Benzodiazepines and conjugates thereof
US8609104B2 (en) 2003-02-20 2013-12-17 Seattle Genetics, Inc. Treatment of B-cell cancers with anti-CD70 antibody-drug conjugates
WO2014057120A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2014057072A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
WO2014057118A1 (en) * 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
WO2014057122A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
WO2014057117A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2014057073A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014057115A1 (en) * 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-her2 antibody conjugates
WO2014057119A1 (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
WO2014057114A1 (en) * 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-psma antibody conjugates
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014143622A1 (en) * 2013-03-13 2014-09-18 Seattle Genetics, Inc. ACTIVATED CARBON FILTRATION FOR PURIFICATION OF BENZODIAZEPINE ADCs
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
WO2014165119A1 (en) 2013-03-13 2014-10-09 Seattle Genetics, Inc. Cyclodextrin and antibody-drug conjugate formulations
WO2015009740A2 (en) 2013-07-15 2015-01-22 Cell Signaling Technology, Inc. Anti-mucin 1 binding agents and uses thereof
US8986972B2 (en) 2012-02-24 2015-03-24 Stem Centrx, Inc. Nucleic acid encoding DLL3 antibodies
WO2015052321A1 (en) * 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2015077605A1 (en) 2013-11-25 2015-05-28 Seattle Genetics, Inc. Preparing antibodies from cho cell cultures for conjugation
JP2015520758A (en) * 2012-05-18 2015-07-23 シアトル ジェネティクス,インコーポレーテッド CD33 antibody and its use to treat cancer
JP2015535004A (en) * 2012-11-05 2015-12-07 ファイザー・インク Spiraisostatin analog
CN105246894A (en) * 2012-12-21 2016-01-13 斯皮罗根有限公司 Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016166304A1 (en) 2015-04-15 2016-10-20 Van Berkel Patricius Hendrikus Cornelis Site-specific antibody-drug conjugates
US9526798B2 (en) 2011-10-14 2016-12-27 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
WO2017004330A1 (en) 2015-06-30 2017-01-05 Seattle Genetics, Inc. Anti-ntb-a antibodies and related compositions and methods
US9562049B2 (en) 2012-12-21 2017-02-07 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9624227B2 (en) 2008-10-17 2017-04-18 Medimmune Limited Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
US9676850B2 (en) 2012-02-24 2017-06-13 Abbvie Stemcentrx Llc Anti SEZ6 antibodies and methods of use
WO2017129652A1 (en) * 2016-01-26 2017-08-03 Medimmune Limited Pyrrolobenzodiazepines
US9732084B2 (en) 2010-04-15 2017-08-15 Medimmune Limited Pyrrolobenzodiazepines used to treat proliferative diseases
WO2017160954A1 (en) 2016-03-15 2017-09-21 Seattle Genetics, Inc. Combinations of pbd-based antibody drug conjugates with bcl-2 inhibitors
US9777071B2 (en) 2013-12-12 2017-10-03 Abbvie Stemcentrx Llc Anti-DPEP3 antibodies and methods of use
WO2017186894A1 (en) * 2016-04-29 2017-11-02 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
WO2017201132A2 (en) 2016-05-18 2017-11-23 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
JP2017535246A (en) * 2014-09-05 2017-11-30 アッヴィ・ステムセントルクス・エル・エル・シー Novel anti-MFI2 antibody and method of use
WO2017223275A1 (en) 2016-06-24 2017-12-28 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited Pyrrolobenzodiazepines and 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
US9968687B2 (en) 2013-02-22 2018-05-15 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
WO2018091646A1 (en) * 2016-11-17 2018-05-24 Medimmune Limited Pyrrolobenzodiazepine conjugates
US9993566B2 (en) 2013-08-28 2018-06-12 Abbvie Stemcentrx Llc 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
US10035853B2 (en) 2013-08-28 2018-07-31 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
US10053511B2 (en) 2013-11-06 2018-08-21 Abbvie Stemcentrx Llc Anti-claudin antibodies and methods of use
US10058613B2 (en) 2015-10-02 2018-08-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
WO2018159582A1 (en) 2017-02-28 2018-09-07 学校法人近畿大学 Method for treating egfr-tki-resistant non-small cell lung cancer by administration of anti-her3 antibody-drug conjugate
WO2018175994A1 (en) 2017-03-24 2018-09-27 Seattle Genetics, Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
US10179820B2 (en) 2014-09-12 2019-01-15 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
GB201820725D0 (en) 2018-12-19 2019-01-30 Adc Therapeutics Sarl Pyrrolobenzodiazepine resistance
US20190083645A1 (en) * 2015-12-04 2019-03-21 Abbvie Stemcentrx Llc Novel anti-claudin antibodies and methods of use
WO2019065964A1 (en) 2017-09-29 2019-04-04 第一三共株式会社 Antibody-pyrrolobenzodiazepine derivative conjugate
WO2019096788A1 (en) 2017-11-14 2019-05-23 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2019104289A1 (en) 2017-11-27 2019-05-31 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
US10308721B2 (en) 2014-02-21 2019-06-04 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and drug conjugates for use in melanoma
WO2019126691A1 (en) 2017-12-21 2019-06-27 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
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
US10561739B2 (en) 2010-04-15 2020-02-18 Seattle Genetics Inc. Targeted pyrrolobenzodiazapine conjugates
WO2020059772A1 (en) 2018-09-20 2020-03-26 第一三共株式会社 Treatment of her3 mutant cancer by administration of anti-her3 antibody-drug conjugate
US10640508B2 (en) 2017-10-13 2020-05-05 Massachusetts Institute Of Technology Diazene directed modular synthesis of compounds with quaternary carbon centers
WO2020100954A1 (en) 2018-11-14 2020-05-22 第一三共株式会社 (anti-cdh6 antibody)-(pyrrolobenzodiazepine derivative) conjugate
WO2020127573A1 (en) 2018-12-19 2020-06-25 Adc Therapeutics Sa Pyrrolobenzodiazepine resistance
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2020173902A1 (en) * 2019-02-25 2020-09-03 Medimmune Limited Methods of synthesis and intermediates
CN111670045A (en) * 2017-12-22 2020-09-15 阿尔麦克探索有限公司 ROR 1-specific antigen-binding molecules
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2020191306A1 (en) * 2019-03-21 2020-09-24 Immunogen, Inc. Methods of preparing cell-binding agent-drug conjugates
WO2020196712A1 (en) 2019-03-27 2020-10-01 第一三共株式会社 Combination of antibody-pyrrolobenzodiazepine derivative conjugate and parp inhibitor
WO2020196475A1 (en) 2019-03-25 2020-10-01 第一三共株式会社 Anti-her2 antibody-pyrrolobenzodiazepine derivative conjugate
WO2020196474A1 (en) 2019-03-25 2020-10-01 第一三共株式会社 Antibody-pyrrolobenzodiazepine derivative conjugate
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
EP3756691A1 (en) 2015-06-12 2020-12-30 Seagen Inc. Anti-cd123 antibodies and conjugates thereof
US10899775B2 (en) 2015-07-21 2021-01-26 Immunogen, Inc. Methods of preparing cytotoxic benzodiazepine derivatives
WO2021024133A2 (en) 2019-08-06 2021-02-11 Glaxosmithkline Intellectual Property Development Limited Biopharmacuetical compositions and related methods
US10918735B2 (en) 2012-12-04 2021-02-16 Massachusetts Institute Of Technology Substituted pyrazino[1′,2′:1,5]pyrrolo[2,3-b]indole-1,4-diones for cancer treatment
US10918627B2 (en) 2016-05-11 2021-02-16 Massachusetts Institute Of Technology Convergent and enantioselective total synthesis of Communesin analogs
US10934359B2 (en) 2016-04-21 2021-03-02 Abbvie Stemcentrx Llc Anti-BMPR1B antibodies and methods of use
WO2021080608A1 (en) 2019-10-25 2021-04-29 Medimmune, Llc Branched moiety for use in conjugates
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
US11103593B2 (en) 2013-10-15 2021-08-31 Seagen Inc. Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
US11110179B2 (en) 2016-06-03 2021-09-07 Seagen Inc. Combination of CD33 antibody drug conjugates with chemotherapeutic agents
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11191771B2 (en) 2016-06-09 2021-12-07 Seagen Inc. Combinations of PBD-based antibody drug conjugates with FLT3 inhibitors
EP3939616A1 (en) 2017-02-08 2022-01-19 ADC Therapeutics SA Pyrrolobenzodiazepine-antibody conjugates
WO2022014698A1 (en) 2020-07-17 2022-01-20 第一三共株式会社 Method for producing antibody-drug conjugate
WO2022050300A1 (en) 2020-09-02 2022-03-10 第一三共株式会社 NOVEL ENDO-β-N-ACETYLGLUCOSAMINIDASE
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
WO2022153194A1 (en) 2021-01-13 2022-07-21 Memorial Sloan Kettering Cancer Center Antibody-pyrrolobenzodiazepine derivative conjugate
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
US11535634B2 (en) 2019-06-05 2022-12-27 Massachusetts Institute Of Technology Compounds, conjugates, and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines and uses thereof
WO2023012669A2 (en) 2021-08-03 2023-02-09 Glaxosmithkline Intellectual Property Development Limited Biopharmaceutical compositions and stable isotope labeling peptide mapping method
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
US11691982B2 (en) 2017-09-20 2023-07-04 Ph Pharma Co., Ltd. Thailanstatin analogs
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US11844839B2 (en) 2016-03-25 2023-12-19 Seagen Inc. Process for the preparation of pegylated drug-linkers and intermediates thereof
US11932650B2 (en) 2017-05-11 2024-03-19 Massachusetts Institute Of Technology Potent agelastatin derivatives as modulators for cancer invasion and metastasis

Families Citing this family (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8455622B2 (en) 2006-12-01 2013-06-04 Seattle Genetics, Inc. Variant target binding agents and uses thereof
MX2016001862A (en) * 2013-08-12 2016-08-03 Genentech Inc 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment.
AU2014312210A1 (en) * 2013-08-28 2016-04-07 Abbvie Stemcentrx Llc Engineered anti-DLL3 conjugates and methods of use
JP6332773B2 (en) 2013-09-02 2018-05-30 ハンジョウ ディーエーシー バイオテック シーオー.,エルティディ.Hangzhou Dac Biotech Co.,Ltd. Novel cytotoxic molecules for conjugation of drugs and cell binding molecules
MX2016003256A (en) 2013-09-12 2016-06-07 Halozyme Inc Modified anti-epidermal growth factor receptor antibodies and methods of use thereof.
JP6980384B2 (en) 2013-12-16 2021-12-15 ジェネンテック, インコーポレイテッド 1- (Chloromethyl) -2,3-dihydro-1H-benzo [E] indole dimer antibody-drug conjugate compound, and methods of use and treatment
CN105813653B (en) 2013-12-19 2020-06-26 西雅图基因公司 Methylene carbamate linkers for use with target-drug conjugates
EP3647322B1 (en) 2014-03-20 2021-10-20 Bristol-Myers Squibb Company Stabilized fibronectin based scaffold molecules
TW202214691A (en) 2014-03-21 2022-04-16 美商艾伯維有限公司 Anti-egfr antibodies and antibody drug conjugates
EP3221346B1 (en) 2014-11-21 2020-09-02 Bristol-Myers Squibb Company Antibodies comprising modified heavy constant regions
RS60631B1 (en) 2014-11-21 2020-09-30 Bristol Myers Squibb Co Antibodies against cd73 and uses thereof
EP3224277B1 (en) 2014-11-25 2020-08-26 Bristol-Myers Squibb Company Novel pd-l1 binding polypeptides for imaging
WO2016115201A1 (en) 2015-01-14 2016-07-21 Bristol-Myers Squibb Company Heteroarylene-bridged benzodiazepine dimers, conjugates thereof, and methods of making and using
BR112017014599A2 (en) 2015-01-14 2018-01-16 Bristol-Myers Squibb Company benzodiazepine dimers, conjugates thereof, and methods of preparation and use
JP2018510864A (en) 2015-03-10 2018-04-19 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Antibody capable of binding by transglutaminase and conjugate produced thereby
CA2987410A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
KR20180019592A (en) 2015-06-23 2018-02-26 브리스톨-마이어스 스큅 컴퍼니 Macrocycline benzodiazepine dimer, conjugate thereof, preparation method and uses
EP3733698A1 (en) 2015-09-23 2020-11-04 Bristol-Myers Squibb Company Glypican-3 binding fibronectin based scafflold molecules
JP2019500327A (en) 2015-11-30 2019-01-10 アッヴィ・インコーポレイテッド Anti-huLRRC15 antibody drug conjugate and method of use thereof
WO2017095808A1 (en) 2015-11-30 2017-06-08 Abbvie Inc. ANTI-huLRRC15 ANTIBODY DRUG CONJUGATES AND METHODS FOR THEIR USE
AU2016363013B2 (en) 2015-12-04 2022-03-10 Seagen Inc. Conjugates of quaternized tubulysin compounds
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
GB201521709D0 (en) * 2015-12-09 2016-01-20 Kings College London And Sec Dep For Health The PBD Antibacterial agents
EP3394096A1 (en) 2015-12-21 2018-10-31 Bristol-Myers Squibb Company Variant antibodies for site-specific conjugation
EA201891983A8 (en) 2016-03-04 2020-05-28 Бристол-Майерс Сквибб Компани COMBINED THERAPY BY ANTIBODIES TO CD73
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
WO2017180768A1 (en) * 2016-04-15 2017-10-19 Seattle Genetics, Inc. Combinations of cd33 antibody drug conjugates with hypomethylating agents
EP3468599A2 (en) 2016-06-08 2019-04-17 AbbVie Inc. Anti-cd98 antibodies and antibody drug conjugates
CN109563167A (en) 2016-06-08 2019-04-02 艾伯维公司 Anti- B7-H3 antibody and antibody drug conjugates
WO2017214456A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-cd98 antibodies and antibody drug conjugates
CA3027103A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
WO2017214335A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
AU2017204139B2 (en) 2016-06-17 2018-08-09 Magenta Therapeutics, Inc. Compositions and methods for the depletion of cells
WO2018048975A1 (en) 2016-09-09 2018-03-15 Bristol-Myers Squibb Company Use of an anti-pd-1 antibody in combination with an anti-mesothelin antibody in cancer treatment
MX2019008199A (en) 2017-01-06 2019-11-25 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and methods of inducing exon skipping.
US10576161B2 (en) 2017-01-20 2020-03-03 Magenta Therapeutics, Inc. Compositions and methods for the depletion of CD137+ cells
CA3052837A1 (en) 2017-02-28 2018-09-07 Seattle Genetics, Inc. Cysteine mutated antibodies for conjugation
TW201836647A (en) 2017-04-06 2018-10-16 美商艾伯維有限公司 Anti-prlr antibody-drug conjugates (adc) and uses thereof
KR20220167342A (en) 2017-05-25 2022-12-20 브리스톨-마이어스 스큅 컴퍼니 Antibodies comprising modified heavy constant regions
CN111587124B (en) 2017-06-23 2024-01-12 维洛斯生物股份有限公司 ROR1 antibody immunoconjugates
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
CN111278462A (en) * 2017-09-02 2020-06-12 艾伯维公司 anti-EGFR Antibody Drug Conjugates (ADCs) and uses thereof
WO2019071028A1 (en) 2017-10-04 2019-04-11 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and uses thereof
MX2020005860A (en) 2017-12-06 2020-09-09 Avidity Biosciences Inc Compositions and methods of treating muscle atrophy and myotonic dystrophy.
EP3724225A1 (en) 2017-12-15 2020-10-21 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
EP3829648A1 (en) 2018-07-27 2021-06-09 Promega Corporation Quinone-containing conjugates
JP2022513653A (en) 2018-11-28 2022-02-09 ブリストル-マイヤーズ スクイブ カンパニー Antibodies containing modified heavy chain constant regions
SI3886914T1 (en) 2018-11-30 2023-06-30 Bristol-Myers Squibb Company Antibody comprising a glutamine-containing light chain c-terminal extension, conjugates thereof, and methods and uses
US20220031860A1 (en) 2018-12-12 2022-02-03 Bristol-Myers Squibb Company Antibodies modified for transglutaminase conjugation, conjugates thereof, and methods and uses
IL297818A (en) 2018-12-21 2023-01-01 Avidity Biosciences Inc Anti-transferrin receptor antibodies and uses thereof
US20220096641A1 (en) * 2019-01-03 2022-03-31 Legochem Biosciences, Inc. Pyrrolobenzodiazepine dimer compound with improved safety and use thereof
EP3918323A4 (en) 2019-01-30 2022-12-28 TrueBinding, Inc. Anti-gal3 antibodies and uses thereof
US11578090B2 (en) 2019-06-06 2023-02-14 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
JP7448638B2 (en) * 2019-08-19 2024-03-12 沈陽薬科大学 Antibody variants and their uses
WO2021055306A1 (en) 2019-09-16 2021-03-25 Bristol-Myers Squibb Company Dual capture method for analysis of antibody-drug conjugates
JP2023537798A (en) 2020-03-19 2023-09-06 アビディティー バイオサイエンシーズ,インク. Compositions and methods for treating facioscapulohumeral muscular dystrophy
WO2021195469A1 (en) 2020-03-27 2021-09-30 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
JP2023526282A (en) 2020-05-13 2023-06-21 ボナム セラピューティクス,インク. Compositions of protein complexes and methods of use thereof
UY39610A (en) 2021-01-20 2022-08-31 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
AU2022345098A1 (en) 2021-09-16 2024-04-04 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
WO2023122347A2 (en) 2021-12-23 2023-06-29 Mirecule, Inc. Compositions for delivery of polynucleotides
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2053894A (en) 1979-07-17 1981-02-11 Green Cross Corp Benzodiazepines processes for producing them and compositions containing them
JPS58180487A (en) 1982-04-16 1983-10-21 Kyowa Hakko Kogyo Co Ltd Antibiotic dc-81 and its preparation
WO2000012508A2 (en) 1998-08-27 2000-03-09 Spirogen Limited Pyrrolbenzodiazepines
WO2004043963A1 (en) 2002-11-14 2004-05-27 Spirogen Limited Pyrrolobenzodiazepines
WO2005023814A1 (en) 2003-09-11 2005-03-17 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
WO2005082023A2 (en) 2004-02-23 2005-09-09 Genentech, Inc. Heterocyclic self-immolative linkers and conjugates
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
WO2006111759A1 (en) 2005-04-21 2006-10-26 Spirogen Limited Pyrrolobenzodiazepines
US20070092940A1 (en) 2004-09-23 2007-04-26 Genentech, Inc. Cysteine engineered antibodies and conjugates
EP1813614A1 (en) * 2006-01-25 2007-08-01 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives
WO2008070593A2 (en) 2006-12-01 2008-06-12 Seattle Genetics, Inc. Variant target binding agents and uses thereof
US20080213289A1 (en) 2002-07-31 2008-09-04 Seattle Genetics, Inc. Recombinant anti-cd30 antibodies and uses thereof
US20090148942A1 (en) 2005-04-19 2009-06-11 Mcdonagh Charlotte Humanized anti-cd70 binding agents and uses thereof
WO2010043880A1 (en) * 2008-10-17 2010-04-22 Spirogen Limited Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases

Family Cites Families (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3361742A (en) 1964-12-07 1968-01-02 Hoffmann La Roche 5-oxo-1h-pyrrolo-[2, 1-c][1, 4]-benzodiazepin-2-crylamides
US3523941A (en) 1967-03-06 1970-08-11 Hoffmann La Roche Benzodiazepine compounds and process for their preparation
US3524849A (en) 1967-10-27 1970-08-18 Hoffmann La Roche Process for the preparation of pyrrolo-benzodiazepine acrylamides and intermediates useful therein
IL33558A (en) 1968-12-30 1973-10-25 Fujisawa Pharmaceutical Co Antibiotic pyrrolo-benzodiazepine compound,its derivatives and processes for their production
JPS4843755B1 (en) 1969-06-26 1973-12-20
DE1965304A1 (en) 1968-12-30 1970-07-23 Fujisawa Pharmaceutical Co Benzdiazepinone compounds and processes for their preparation
JPS5382792U (en) 1976-12-10 1978-07-08
JPS6053033B2 (en) 1976-12-28 1985-11-22 財団法人微生物化学研究会 New anticancer antibiotic mazethramycin and its production method
JPS585916B2 (en) 1977-12-27 1983-02-02 株式会社ミドリ十字 New benzodiazepine compounds
JPS57131791A (en) 1980-12-31 1982-08-14 Fujisawa Pharmaceut Co Ltd Benzodiazepine derivative and its preparation
JPH0353356Y2 (en) 1981-02-06 1991-11-21
CA1173441A (en) 1981-02-27 1984-08-28 Hoffmann-La Roche Limited Imidazodiazepines
CA1185602A (en) 1981-02-27 1985-04-16 Emilio Kyburz Imidazodiazepines
CA1184175A (en) 1981-02-27 1985-03-19 Walter Hunkeler Imidazodiazepines
JPS58180487U (en) 1982-05-28 1983-12-02 松下電工株式会社 Light beam alarm assembly
US4427588A (en) 1982-11-08 1984-01-24 Bristol-Myers Company Process for conversion of oxotomaymycin to tomaymycin
US4427587A (en) 1982-11-10 1984-01-24 Bristol-Myers Company Total synthesis of antitumor antibiotics BBM-2040A and BBM-2040B
JPS59152329A (en) 1983-02-17 1984-08-31 Green Cross Corp:The Local disorder inhibitor
FR2586683B1 (en) 1985-08-29 1988-07-01 Centre Nat Rech Scient NOVEL NEOTHRAMYCIN DERIVATIVES, THEIR PREPARATION PROCESS AND THEIR APPLICATION AS MEDICAMENTS
JP2660201B2 (en) 1988-08-05 1997-10-08 塩野義製薬株式会社 Novel pyrrolo [1,4] benzodiazepine derivatives and senile dementia drugs
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.
GB9205051D0 (en) 1992-03-09 1992-04-22 Cancer Res Campaign Tech Pyrrolobenzodiazepine derivatives,their preparation,and compositions containing them
FR2696176B1 (en) 1992-09-28 1994-11-10 Synthelabo Piperidine derivatives, their preparation and their therapeutic application.
GB9316162D0 (en) 1993-08-04 1993-09-22 Zeneca Ltd Fungicides
DE69934618T2 (en) 1998-07-08 2007-05-03 E-Ink Corp., Cambridge Improved colored microencapsulated electrophoretic display
GB9818730D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Collections of compounds
GB9818732D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Collection of compounds
GB9818731D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Compounds
US6909006B1 (en) 1999-08-27 2005-06-21 Spirogen Limited Cyclopropylindole derivatives
US6660742B2 (en) 2000-09-19 2003-12-09 Taiho Pharmaceutical Co. Ltd. Compositions and methods of the use thereof achiral analogues of CC-1065 and the duocarmycins
US6362331B1 (en) 2001-03-30 2002-03-26 Council Of Scientific And Industrial Research Process for the preparation of antitumor agents
US6660856B2 (en) 2002-03-08 2003-12-09 Kaohsiung Medical University Synthesis of pyrrolo[2,1-c][1,4]benzodiazepine analogues
US20040138269A1 (en) 2002-10-11 2004-07-15 Sugen, Inc. Substituted pyrroles as kinase inhibitors
WO2004087717A1 (en) 2003-03-31 2004-10-14 Council Of Scientific And Industrial Research Non-cross-linking pyrrolo[2,1-c][1,4]benzodiazepines as potential antitumour agents and process thereof
GB0416511D0 (en) 2003-10-22 2004-08-25 Spirogen Ltd Pyrrolobenzodiazepines
AU2004284075A1 (en) 2003-10-22 2005-05-06 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Pyrrolobenzodiazepine derivatives, compositions comprising the same and methods related thereto
GB0404578D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Pyrrolobenzodiazepines
GB0404577D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Pyrrolobenzodiazepines
GB0404574D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Amino acids
DE102004010943A1 (en) 2004-03-03 2005-09-29 Degussa Ag Process for the preparation of N-protected 4-ketproline derivatives
EP1723152B1 (en) 2004-03-09 2015-02-11 Spirogen Sàrl Pyrrolobenzodiazepines
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
EP1831418A2 (en) 2004-12-24 2007-09-12 Showa Denko Kabushiki Kaisha Production method of thermoelectric semiconductor alloy, thermoelectric conversion module and thermoelectric power generating device
GB0508084D0 (en) * 2005-04-21 2005-06-01 Spirogen Ltd Pyrrolobenzodiazepines
AU2006269940C1 (en) * 2005-07-18 2013-11-07 Seagen Inc. Beta-glucuronide-linker drug conjugates
US20070154906A1 (en) 2005-10-05 2007-07-05 Spirogen Ltd. Methods to identify therapeutic candidates
ATE427949T1 (en) 2005-10-05 2009-04-15 Spirogen Ltd 4-A4-(5-OXO-2,3,5,11A-TETRAHYDRO-5H-PYRROLO A2, 1-CUA1,4UBENZODIAZEPINE-8-YLOXY)-BUTYRYLAMINOU-1 - PYRROLE-2-CARBONATE ALKYL ESTER DERIVATIVES AND RELATED COMPOUND FOR THE TREATMENT OF A PROLIFERATIVE DISEASE
CN101622276B (en) 2006-07-18 2015-04-22 赛诺菲-安万特 Antagonist antibody against EphA2 for the treatment of cancer
EP1914242A1 (en) 2006-10-19 2008-04-23 Sanofi-Aventis Novel anti-CD38 antibodies for the treatment of cancer
WO2008050140A2 (en) 2006-10-27 2008-05-02 Spirogen Limited Compounds for treatment of parasitic infection
SI2019104T1 (en) 2007-07-19 2013-12-31 Sanofi Cytotoxic agents comprising new tomaymycin derivatives and their therapeutic use
GB0722088D0 (en) 2007-11-09 2007-12-19 Spirogen Ltd Pyrrolobenzodiazepines
GB0722087D0 (en) 2007-11-09 2007-12-19 Spirogen Ltd Polyamides
GB0813432D0 (en) 2008-07-22 2008-08-27 Spirogen Ltd Pyrrolobenzodiazepines
GB0819097D0 (en) 2008-10-17 2008-11-26 Spirogen Ltd Pyrrolobenzodiazepines
EP3360879A1 (en) 2009-02-05 2018-08-15 ImmunoGen, Inc. Benzodiazepine derivatives as cytotoxic agents
FR2949469A1 (en) 2009-08-25 2011-03-04 Sanofi Aventis ANTICANCER DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
EP2534158B1 (en) 2010-02-09 2014-01-08 Bristol-Myers Squibb Company Benzylpyrrolidinone derivatives as modulators of chemokine receptor activity
GB201006340D0 (en) 2010-04-15 2010-06-02 Spirogen Ltd Synthesis method and intermediates
AU2011239525B2 (en) 2010-04-15 2015-04-09 Medimmune Limited Pyrrolobenzodiazepines used to treat proliferative diseases
AU2011239522B2 (en) 2010-04-15 2014-10-23 Medimmune Limited Targeted pyrrolobenzodiazapine conjugates
KR101738203B1 (en) 2010-04-15 2017-05-19 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
MX346635B (en) 2011-02-15 2017-03-27 Immunogen Inc Cytotoxic benzodiazepine derivatives.
AU2012311505B2 (en) 2011-09-20 2016-09-29 Medimmune Limited Pyrrolobenzodiazepines as unsymmetrical dimeric PBD compounds for inclusion in targeted conjugates
EA036202B1 (en) 2011-10-14 2020-10-14 Сиэтл Дженетикс, Инк. Pyrrolobenzodiazepines and targeted conjugates
AU2012322933B2 (en) 2011-10-14 2017-02-02 Medimmune Limited Synthesis method and intermediates useful in the preparation of pyrrolobenzodiazepines
KR101891859B1 (en) 2011-10-14 2018-08-24 메디뮨 리미티드 Pyrrolobenzodiazepines
EP2750713B1 (en) 2011-10-14 2015-09-16 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
JP6117801B2 (en) 2011-10-14 2017-04-19 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepine
US9526798B2 (en) 2011-10-14 2016-12-27 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
AU2013255613B2 (en) 2012-04-30 2017-06-22 Medimmune Limited Pyrrolobenzodiazepines
US9321774B2 (en) 2012-04-30 2016-04-26 Medimmune Limited Pyrrolobenzodiazepines
TW201408698A (en) 2012-07-09 2014-03-01 Genentech Inc Anti-CD79b antibodies and immunoconjugates
SG11201500087VA (en) 2012-07-09 2015-02-27 Genentech Inc Immunoconjugates comprising anti-cd22 antibodies
WO2014022679A2 (en) 2012-08-02 2014-02-06 Genentech, Inc. Anti-etbr antibodies and immunoconjugates
KR101412875B1 (en) 2012-10-04 2014-07-02 삼성전기주식회사 Gate driving circuit and inverter having the same
SI2906252T1 (en) 2012-10-12 2017-10-30 Adc Therapeutics Sa Pyrrolobenzodiazepine-anti-her2 antibody conjugates
HUE042731T2 (en) 2012-10-12 2019-07-29 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
HUE045435T2 (en) 2012-10-12 2019-12-30 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
AU2013328625B2 (en) 2012-10-12 2016-12-15 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
SI2906253T1 (en) 2012-10-12 2018-11-30 Adc Therapeutics Sa Pyrrolobenzodiazepine - anti-psma antibody conjugates
WO2014057118A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
BR112015008238A2 (en) 2012-10-12 2017-11-28 Adc Therapeutics Sarl pyrrolbenzodiazepine-anti-cd22 antibody conjugates
EP2906248B1 (en) 2012-10-12 2018-12-05 MedImmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2014057120A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
ES2680153T3 (en) 2012-10-12 2018-09-04 Adc Therapeutics Sa Anti-PSMA-pyrrolobenzodiazepine antibody conjugates
AU2013328673B2 (en) 2012-10-12 2017-07-13 Medimmune Limited Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
JP6307519B2 (en) 2012-12-21 2018-04-04 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepine and its conjugates
EA032986B1 (en) 2012-12-21 2019-08-30 Медимьюн Лимитед Pyrrolobenzodiazepines
MX2015010682A (en) 2013-02-22 2016-05-31 Stemcentrx Inc Novel antibody conjugates and uses thereof.
JP6340019B2 (en) 2013-03-13 2018-06-06 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepine and its conjugates
CA2905181C (en) 2013-03-13 2020-06-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof for providing targeted therapy
EA027910B1 (en) 2013-03-13 2017-09-29 Медимьюн Лимитед Pyrrolobenzodiazepines and conjugates thereof
US20160106861A1 (en) 2013-04-26 2016-04-21 Spirogen Sarl Axl antibody-drug conjugate and its use for the treatment of cancer
US20160256561A1 (en) 2013-10-11 2016-09-08 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
GB201317982D0 (en) 2013-10-11 2013-11-27 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
EP3054983B1 (en) 2013-10-11 2019-03-20 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2015052534A1 (en) 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepine-antibody conjugates
GB201317981D0 (en) 2013-10-11 2013-11-27 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
CA2931798C (en) 2013-12-16 2023-06-27 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
GB201406767D0 (en) 2014-04-15 2014-05-28 Cancer Rec Tech Ltd Humanized anti-Tn-MUC1 antibodies anf their conjugates
US10188746B2 (en) 2014-09-10 2019-01-29 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
GB201416112D0 (en) 2014-09-12 2014-10-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2053894A (en) 1979-07-17 1981-02-11 Green Cross Corp Benzodiazepines processes for producing them and compositions containing them
JPS58180487A (en) 1982-04-16 1983-10-21 Kyowa Hakko Kogyo Co Ltd Antibiotic dc-81 and its preparation
WO2000012508A2 (en) 1998-08-27 2000-03-09 Spirogen Limited Pyrrolbenzodiazepines
US20080213289A1 (en) 2002-07-31 2008-09-04 Seattle Genetics, Inc. Recombinant anti-cd30 antibodies and uses thereof
WO2004043963A1 (en) 2002-11-14 2004-05-27 Spirogen Limited Pyrrolobenzodiazepines
WO2005023814A1 (en) 2003-09-11 2005-03-17 Spirogen Limited Synthesis of protected pyrrolobenzodiazepines
WO2005082023A2 (en) 2004-02-23 2005-09-09 Genentech, Inc. Heterocyclic self-immolative linkers and conjugates
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
US20070092940A1 (en) 2004-09-23 2007-04-26 Genentech, Inc. Cysteine engineered antibodies and conjugates
US20090148942A1 (en) 2005-04-19 2009-06-11 Mcdonagh Charlotte Humanized anti-cd70 binding agents and uses thereof
WO2006111759A1 (en) 2005-04-21 2006-10-26 Spirogen Limited Pyrrolobenzodiazepines
EP1813614A1 (en) * 2006-01-25 2007-08-01 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives
WO2007085930A1 (en) 2006-01-25 2007-08-02 Sanofi-Aventis Cytotoxic agents comprising new tomaymycin derivatives and their therapeutic use
WO2008070593A2 (en) 2006-12-01 2008-06-12 Seattle Genetics, Inc. Variant target binding agents and uses thereof
WO2010043880A1 (en) * 2008-10-17 2010-04-22 Spirogen Limited Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases

Non-Patent Citations (35)

* Cited by examiner, † Cited by third party
Title
ALLEY, M.C. ET AL., CANCER RESEARCH, vol. 64, 2004, pages 6700 - 6706
ARIMA ET AL., J. ANTIBIOTICS, vol. 25, 1972, pages 437 - 444
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BOSE ET AL., TETRAHEDRON, vol. 48, 1992, pages 751 - 758
BOSE,O D.S. ET AL., J. AM. CHEM. SOC., vol. 114, 1992, pages 4939 - 4941
BURKE P J ET AL: "Novel immunoconjugates comprised of streptonigrin and 17-amino-geldanamycin attached via a dipeptide-p-aminobenzyl-amine linker system", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB, vol. 19, no. 10, 15 May 2009 (2009-05-15), pages 2650 - 2653, XP026085936, ISSN: 0960-894X, [retrieved on 20090405], DOI: 10.1016/J.BMCL.2009.03.145 *
DORONINA ET AL., NATURE BIOTECHNOLOGY, vol. 21, 2003, pages 778 - 784
GREENE, T.W., WUTS, G.M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS, INC., pages: 23 - 200
GREENE, T.W., WUTS, G.M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS, INC., pages: 503 - 549
GREENE, T.W., WUTS, G.M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS, INC., pages: 633 - 647
GREGSON, S. ET AL., J. MED. CHEM., vol. 44, 2001, pages 737 - 748
HARA ET AL., J. ANTIBIOTICS, vol. 41, 1988, pages 702 - 704
HARTLEY, J.A. ET AL., CANCER RESEARCH, vol. 64, 2004, pages 6693 - 6699
HOCHLOWSKI ET AL., J. ANTIBIOTICS, vol. 40, 1987, pages 145 - 148
HURLEY, NEEDHAM-VANDEVANTER, ACC. CHEM. RES., vol. 19, 1986, pages 230 - 237
ITOH ET AL., J. ANTIBIOTICS, vol. 41, 1988, pages 1281 - 1284
JEFFREY ET AL., BIOCONJUGATE CHEMISTRY, vol. 17, 2006, pages 831 - 840
JEFFREY SC ET AL: "DESIGN, SYNTHESIS, AND IN VITRO EVALUATION OF DIPEPTIDE-BASED ANTIBODY MINOR GROOVE BINDER CONJUGATES", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 48, no. 5, 10 May 2005 (2005-05-10), pages 1344 - 1358, XP008079143, ISSN: 0022-2623, DOI: 10.1021/JM040137Q *
KOHN: "Antibiotics III", 1975, SPRINGER-VERLAG, pages: 3 - 11
KONISHI ET AL., J. ANTIBIOTICS, vol. 37, 1984, pages 200 - 206
KUMINOTO ET AL., J. ANTIBIOTICS, vol. 33, 1980, pages 665 - 667
LANGLEY, THURSTON, J. ORG. CHEM., vol. 52, 1987, pages 91 - 97
LEBER ET AL., J. AM. CHEM. SOC., vol. 110, 1988, pages 2992 - 2993
LEIMGRUBER ET AL., J. AM. CHEM. SOC., vol. 87, 1965, pages 5791 - 5793
LEIMGRUBER ET AL., J. AM. CHEM. SOC., vol. 87, 1965, pages 5793 - 5795
MARTIN, C. ET AL., BIOCHEMISTRY, vol. 44, pages 4135 - 4147
Q DONG ET AL., TETRAHEDRON LETTERS, vol. 36, no. 32, 1995, pages 5681 - 5682
SHIMIZU ET AL., J. ANTIBIOTICS, vol. 29, 1982, pages 2492 - 2503
SMELLIE, M. ET AL., BIOCHEMISTRY, vol. 42, 2003, pages 8232 - 8239
TAKEUCHI ET AL., J. ANTIBIOTICS, vol. 29, 1976, pages 93 - 96
THOMAS FEY ET AL., J. ORG. CHEM., vol. 66, 2001, pages 8154 - 8159
THURSTON ET AL., CHEM. BRIT., vol. 26, 1990, pages 767 - 772
THURSTON ET AL., CHEM. REV., 1994, pages 433 - 465
THURSTON, D.E. ET AL., J. ORG. CHEM., vol. 61, 1996, pages 8141 - 8147
TSUNAKAWA ET AL., J. ANTIBIOTICS, vol. 41, 1988, pages 1366 - 1373

Cited By (280)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8609104B2 (en) 2003-02-20 2013-12-17 Seattle Genetics, Inc. Treatment of B-cell cancers with anti-CD70 antibody-drug conjugates
US9345785B2 (en) 2003-02-20 2016-05-24 Seattle Genetics, Inc. Treatment of renal cell carcinoma with anti-CD70 antibody-drug conjugates
US9624227B2 (en) 2008-10-17 2017-04-18 Medimmune Limited Unsymmetrical pyrrolobenzodiazepine-dimers for treatment of proliferative diseases
US9550787B2 (en) 2009-02-05 2017-01-24 Immunogen, Inc. Benzodiazepine derivatives
US10947315B2 (en) 2009-02-05 2021-03-16 Immunogen, Inc. Benzodiazepine derivatives
US8426402B2 (en) 2009-02-05 2013-04-23 Immunogen, Inc. Benzodiazepine derivatives
US9265841B2 (en) 2009-02-05 2016-02-23 Immunogen, Inc. Benzodiazepine derivatives
US11505617B2 (en) 2009-02-05 2022-11-22 Immunogen, Inc. 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
US10208127B2 (en) 2009-02-05 2019-02-19 Immunogen, Inc. Benzodiazepine derivatives
US9732084B2 (en) 2010-04-15 2017-08-15 Medimmune Limited Pyrrolobenzodiazepines used to treat proliferative diseases
US10561739B2 (en) 2010-04-15 2020-02-18 Seattle Genetics Inc. Targeted pyrrolobenzodiazapine conjugates
US9534000B2 (en) 2011-02-15 2017-01-03 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and methods of preparation
CN113896731A (en) * 2011-02-15 2022-01-07 伊缪诺金公司 Cytotoxic benzodiazepine derivatives
US9840564B2 (en) 2011-02-15 2017-12-12 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
WO2012112708A1 (en) * 2011-02-15 2012-08-23 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and methods of preparation
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
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
US10570212B2 (en) 2011-02-15 2020-02-25 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9868791B2 (en) 2011-02-15 2018-01-16 Immunogen, Inc. Methods of preparation of conjugates
US9353127B2 (en) 2011-02-15 2016-05-31 Immunogen, Inc. Methods of preparation of conjugates
US9169272B2 (en) 2011-02-15 2015-10-27 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US9434748B2 (en) 2011-02-15 2016-09-06 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US10364294B2 (en) 2011-02-15 2019-07-30 Immunogen, Inc. Methods of preparation of conjugates
US9399641B2 (en) 2011-09-20 2016-07-26 Medimmune Limited Pyrrolobenzodiazepines as unsymmetrical dimeric PBD compounds for inclusion in targeted conjugates
WO2013041606A1 (en) 2011-09-20 2013-03-28 Spirogen Sàrl Pyrrolobenzodiazepines as unsymmetrical dimeric pbd compounds for inclusion in targeted conjugates
US10328084B2 (en) 2011-10-14 2019-06-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
WO2013055993A1 (en) 2011-10-14 2013-04-18 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US9713647B2 (en) 2011-10-14 2017-07-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
WO2013053873A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines
JP2014528466A (en) * 2011-10-14 2014-10-27 スパイロジェン・エス・アー・エール・エルSpirogen Sarl Pyrrolobenzodiazepine
WO2013053871A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines
US9526798B2 (en) 2011-10-14 2016-12-27 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US9707301B2 (en) 2011-10-14 2017-07-18 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US10329352B2 (en) 2011-10-14 2019-06-25 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
US9399073B2 (en) 2011-10-14 2016-07-26 Seattle Genetics, Inc. Pyrrolobenzodiazepines
US9388187B2 (en) 2011-10-14 2016-07-12 Medimmune Limited Pyrrolobenzodiazepines
US9387259B2 (en) 2011-10-14 2016-07-12 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
EP3388435A1 (en) 2011-10-14 2018-10-17 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
EP3309162A1 (en) 2011-10-14 2018-04-18 Seattle Genetics, Inc. Targeted conjugates of pyrrolobenzodiazepines
WO2013119960A2 (en) 2012-02-08 2013-08-15 Stem Centrx, Inc. Novel modulators and methods of use
US9937268B2 (en) 2012-02-24 2018-04-10 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates and methods of use
US9764042B1 (en) 2012-02-24 2017-09-19 Abbvie Stemcentrx Llc Methods of making DLL3 antibody drug conjugates
US9480757B2 (en) 2012-02-24 2016-11-01 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9173959B1 (en) 2012-02-24 2015-11-03 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates
US10533051B2 (en) 2012-02-24 2020-01-14 Abbvie Stemcentrx Llc Anti SEZ6 antibodies and methods of use
US9486537B2 (en) 2012-02-24 2016-11-08 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US10137204B2 (en) 2012-02-24 2018-11-27 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates for treating cancer
US9155803B1 (en) 2012-02-24 2015-10-13 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates and methods of use
EP3095797A1 (en) 2012-02-24 2016-11-23 Stemcentrx, Inc. Anti dll3 antibodies and methods of use thereof
US9931420B2 (en) 2012-02-24 2018-04-03 Abbvie Stemcentrx Llc Methods of making DLL3 antibody drug conjugates
US9931421B2 (en) 2012-02-24 2018-04-03 Abbvie Stemcentrx Llc Methods of delivering DLL3 antibody drug conjugates
US9878053B2 (en) 2012-02-24 2018-01-30 Abbvie Stemcentrx Llc Methods of delivering DLL3 antibody drug conjugates
US9867887B1 (en) 2012-02-24 2018-01-16 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9861708B2 (en) 2012-02-24 2018-01-09 Abbvie Stemcentrx Llc Kits containing DLL3 antibody drug conjugates
US9855343B2 (en) 2012-02-24 2018-01-02 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US11033634B2 (en) 2012-02-24 2021-06-15 Abbvie Stemcentrx Llc Light chain variable regions
US9775916B1 (en) 2012-02-24 2017-10-03 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates for treating cancer
US9770518B1 (en) 2012-02-24 2017-09-26 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9133271B1 (en) 2012-02-24 2015-09-15 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates and methods of use
US9481727B2 (en) 2012-02-24 2016-11-01 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US8986972B2 (en) 2012-02-24 2015-03-24 Stem Centrx, Inc. Nucleic acid encoding DLL3 antibodies
US9676850B2 (en) 2012-02-24 2017-06-13 Abbvie Stemcentrx Llc Anti SEZ6 antibodies and methods of use
US9334318B1 (en) 2012-02-24 2016-05-10 Stemcentrx, Inc. Multivalent DLL3 antibodies
US9345784B1 (en) 2012-02-24 2016-05-24 Stemcentrx, Inc. Methods of delivering DLL3 antibody drug conjugates
US9107961B2 (en) 2012-02-24 2015-08-18 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates for treating cancer
US9352051B1 (en) 2012-02-24 2016-05-31 Stemcentrx, Inc. Kits containing DLL3 antibody drug conjugates
US9353182B2 (en) 2012-02-24 2016-05-31 Stemcentrx, Inc. Anti-DLL3 antibodies
US9089617B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates
US9358304B1 (en) 2012-02-24 2016-06-07 Stemcentrx, Inc. Methods of making DLL3 antibody drug conjugates
US9090683B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Methods of detection, diagnosis, and monitoring using anti-DLL3 antibodies
US9089616B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates and methods of use
US9089615B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Anti-DLL3 antibodies
US9587019B2 (en) 2012-05-18 2017-03-07 Seattle Genetics, Inc. CD33 antibodies and use of same to treat cancer
US10787514B2 (en) 2012-05-18 2020-09-29 Seattle Genetics, Inc. CD33 antibodies and use of same to treat cancer
JP2015520758A (en) * 2012-05-18 2015-07-23 シアトル ジェネティクス,インコーポレーテッド CD33 antibody and its use to treat cancer
EP3421048A1 (en) 2012-05-18 2019-01-02 Seattle Genetics, Inc. Cd33 antibodies and use of same to treat cancer
WO2013177481A1 (en) 2012-05-25 2013-11-28 Immunogen, Inc. Benzodiazepines and conjugates thereof
US10780181B2 (en) 2012-10-12 2020-09-22 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
JP2018168182A (en) * 2012-10-12 2018-11-01 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepines and conjugates thereof
KR101995619B1 (en) 2012-10-12 2019-07-03 에이디씨 테라퓨틱스 에스에이 Pyrrolobenzodiazepine-antibody conjugates
KR101995621B1 (en) 2012-10-12 2019-07-03 에이디씨 테라퓨틱스 에스에이 Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
AU2013328619B2 (en) * 2012-10-12 2016-11-17 Adc Therapeutics Sa Pyrrolobenzodiazepine - anti-PSMA antibody conjugates
US9415117B2 (en) 2012-10-12 2016-08-16 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
AU2013328625B2 (en) * 2012-10-12 2016-12-15 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
AU2013328628B2 (en) * 2012-10-12 2016-12-15 Adc Therapeutics Sa Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
KR20150083858A (en) * 2012-10-12 2015-07-20 에이디씨 테라퓨틱스 에스에이알엘 Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
KR20150083857A (en) * 2012-10-12 2015-07-20 에이디씨 테라퓨틱스 에스에이알엘 Pyrrolobenzodiazepine-antibody conjugates
US11690918B2 (en) 2012-10-12 2023-07-04 Medimmune Limited Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US11779650B2 (en) 2012-10-12 2023-10-10 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
CN104955485A (en) * 2012-10-12 2015-09-30 Adc疗法责任有限公司 Pyrrolobenzodiazepine-anti-her2 antibody conjugates
KR20150085813A (en) * 2012-10-12 2015-07-24 에이디씨 테라퓨틱스 에스에이알엘 Pyrrolobenzodiazepine - anti-psma antibody conjugates
WO2014057120A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
AU2013328623A1 (en) * 2012-10-12 2015-04-23 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2014057072A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
KR101986404B1 (en) 2012-10-12 2019-06-07 에이디씨 테라퓨틱스 에스에이 Pyrrolobenzodiazepine - anti-psma antibody conjugates
AU2013328623B8 (en) * 2012-10-12 2017-06-15 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
AU2013328674B2 (en) * 2012-10-12 2017-06-22 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11771775B2 (en) 2012-10-12 2023-10-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
KR20190126197A (en) * 2012-10-12 2019-11-08 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
CN105050661A (en) * 2012-10-12 2015-11-11 Adc疗法责任有限公司 Pyrrolobenzodiazepine-antibody conjugates
WO2014057118A1 (en) * 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
US9745303B2 (en) 2012-10-12 2017-08-29 Medimmune Limited Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
WO2014057122A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
WO2014057117A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
JP2016502504A (en) * 2012-10-12 2016-01-28 エイディーシー・セラピューティクス・エス・アー・エール・エルAdc Therapeutics Sarl Pyrrolobenzodiazepine-antibody conjugate
WO2014057073A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
JP2015533141A (en) * 2012-10-12 2015-11-19 スパイロジェン・エス・アー・エール・エルSpirogen Sarl Pyrrolobenzodiazepines and their complexes
US11701430B2 (en) 2012-10-12 2023-07-18 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2014057115A1 (en) * 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-her2 antibody conjugates
US10646584B2 (en) 2012-10-12 2020-05-12 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
KR102138220B1 (en) 2012-10-12 2020-07-27 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
US10994023B2 (en) 2012-10-12 2021-05-04 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10722594B2 (en) 2012-10-12 2020-07-28 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
JP2015534579A (en) * 2012-10-12 2015-12-03 エイディーシー・セラピューティクス・エス・アー・エール・エルAdc Therapeutics Sarl Pyrrolobenzodiazepine-antibody conjugate
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
JP2015534580A (en) * 2012-10-12 2015-12-03 エイディーシー・セラピューティクス・エス・アー・エール・エルAdc Therapeutics Sarl Pyrrolobenzodiazepine-antibody conjugate
CN104955485B (en) * 2012-10-12 2018-01-30 Adc疗法责任有限公司 Pyrrolobenzodiazepines Zhuo Anti-HER 2 conjugate
JP2015534578A (en) * 2012-10-12 2015-12-03 エイディーシー・セラピューティクス・エス・アー・エール・エルAdc Therapeutics Sarl Pyrrolobenzodiazepine-antibody conjugate
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
CN105050661B (en) * 2012-10-12 2018-03-30 Adc疗法责任有限公司 Pyrrolobenzodiazepines Zhuo antibody conjugates
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
JP2015534577A (en) * 2012-10-12 2015-12-03 エイディーシー・セラピューティクス・エス・アー・エール・エルAdc Therapeutics Sarl Pyrrolobenzodiazepine-antibody conjugate
CN105102003A (en) * 2012-10-12 2015-11-25 Adc疗法责任有限公司 Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2014057119A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2014057114A1 (en) * 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-psma antibody conjugates
WO2014057113A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine - anti-psma antibody conjugates
CN105102004A (en) * 2012-10-12 2015-11-25 Adc疗法责任有限公司 Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
US10799596B2 (en) 2012-10-12 2020-10-13 Adc Therapeutics S.A. Pyrrolobenzodiazepine-antibody conjugates
JP2015535004A (en) * 2012-11-05 2015-12-07 ファイザー・インク Spiraisostatin analog
US10918735B2 (en) 2012-12-04 2021-02-16 Massachusetts Institute Of Technology Substituted pyrazino[1′,2′:1,5]pyrrolo[2,3-b]indole-1,4-diones for cancer treatment
CN105246894A (en) * 2012-12-21 2016-01-13 斯皮罗根有限公司 Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
US9562049B2 (en) 2012-12-21 2017-02-07 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9567340B2 (en) 2012-12-21 2017-02-14 Medimmune Limited Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
CN110627797A (en) * 2012-12-21 2019-12-31 麦迪穆有限责任公司 Asymmetric pyrrolobenzodiazepine dimers for the treatment of proliferative and autoimmune diseases
JP2016505586A (en) * 2012-12-21 2016-02-25 スパイロジェン・エス・アー・エール・エルSpirogen Sarl Asymmetric pyrrolobenzodiazepine dimers for use in the treatment of proliferative and autoimmune diseases
US10478509B2 (en) 2013-02-22 2019-11-19 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates for treating cancer
RU2684468C2 (en) * 2013-02-22 2019-04-09 ЭББВИ СТЕМСЕНТРКС ЭлЭлСи Novel antibody conjugates and uses thereof
US9968687B2 (en) 2013-02-22 2018-05-15 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
AU2014248640B2 (en) * 2013-03-13 2018-03-01 Seagen Inc. Cyclodextrin and antibody-drug conjugate formulations
KR102274714B1 (en) 2013-03-13 2021-07-09 씨젠 인크. Cyclodextrin and antibody-drug conjugate formulations
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
KR20210087560A (en) * 2013-03-13 2021-07-12 씨젠 인크. Cyclodextrin and antibody-drug conjugate formulations
KR20150129697A (en) * 2013-03-13 2015-11-20 시애틀 지네틱스, 인크. Cyclodextrin and antibody-drug conjugate formulations
CN105209077A (en) * 2013-03-13 2015-12-30 麦迪穆有限责任公司 Pyrrolobenzodiazepines and conjugates thereof
WO2014143622A1 (en) * 2013-03-13 2014-09-18 Seattle Genetics, Inc. ACTIVATED CARBON FILTRATION FOR PURIFICATION OF BENZODIAZEPINE ADCs
KR102401525B1 (en) * 2013-03-13 2022-05-24 씨젠 인크. Cyclodextrin and antibody-drug conjugate formulations
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
KR102066319B1 (en) 2013-03-13 2020-01-14 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
EP3441072A1 (en) 2013-03-13 2019-02-13 Seattle Genetics, Inc. Activated carbon filtration for purification of benzodiazepine adcs
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014165119A1 (en) 2013-03-13 2014-10-09 Seattle Genetics, Inc. Cyclodextrin and antibody-drug conjugate formulations
IL273359A (en) * 2013-03-13 2020-05-31 Seattle Genetics Inc Cyclodextrin and antibody-drug conjugate formulations
US9453046B2 (en) 2013-03-13 2016-09-27 Seattle Genetics, Inc. Activated carbon filtration for purification of benzodiazepine ADCs
JP2019069947A (en) * 2013-03-13 2019-05-09 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepines and conjugates thereof
KR20150131210A (en) * 2013-03-13 2015-11-24 메디뮨 리미티드 Pyrrolobenzodiazepines and conjugates thereof
EP2970444A4 (en) * 2013-03-13 2016-11-09 Seattle Genetics Inc Cyclodextrin and antibody-drug conjugate formulations
US10391181B2 (en) 2013-03-13 2019-08-27 Seattle Genetics, Inc. Cyclodextrin and antibody-drug conjugate formulations
JP2016512540A (en) * 2013-03-13 2016-04-28 シアトル ジェネティックス, インコーポレイテッド Cyclodextrins and antibody-drug conjugate formulations
CN105209077B (en) * 2013-03-13 2019-06-11 麦迪穆有限责任公司 Pyrrolobenzodiazepines Zhuo and its conjugate
US9649390B2 (en) 2013-03-13 2017-05-16 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9610361B2 (en) 2013-03-13 2017-04-04 Seattle Genetics, Inc. Cyclodextrin and antibody-drug conjugate formulations
AU2018201985B2 (en) * 2013-03-13 2019-07-18 Seagen Inc. Cyclodextrin and antibody-drug conjugate formulations
US9987374B2 (en) 2013-03-13 2018-06-05 Seattle Genetics, Inc. Cyclodextrin and antibody-drug conjugate formulations
WO2015009740A2 (en) 2013-07-15 2015-01-22 Cell Signaling Technology, Inc. Anti-mucin 1 binding agents and uses thereof
EP3699200A1 (en) 2013-07-15 2020-08-26 Cell Signaling Technology, Inc. Anti-mucin 1 binding agents and uses thereof
US9993566B2 (en) 2013-08-28 2018-06-12 Abbvie Stemcentrx Llc SEZ6 modulators and methods of use
US10035853B2 (en) 2013-08-28 2018-07-31 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
EP3338793A1 (en) 2013-08-28 2018-06-27 AbbVie Stemcentrx LLC Novel sez6 modulators and methods of use
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2015052321A1 (en) * 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US9956298B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11103593B2 (en) 2013-10-15 2021-08-31 Seagen Inc. Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
US10053511B2 (en) 2013-11-06 2018-08-21 Abbvie Stemcentrx Llc Anti-claudin antibodies and methods of use
US10457976B2 (en) 2013-11-25 2019-10-29 Seattle Genetics, Inc. Preparing antibodies from CHO cell cultures for conjugation
WO2015077605A1 (en) 2013-11-25 2015-05-28 Seattle Genetics, Inc. Preparing antibodies from cho cell cultures for conjugation
US9777071B2 (en) 2013-12-12 2017-10-03 Abbvie Stemcentrx Llc Anti-DPEP3 antibodies and methods of use
US10189910B2 (en) 2013-12-12 2019-01-29 Abbvie Stemcentrx Llc Anti-DPEP3 antibodies and methods of use
US10308721B2 (en) 2014-02-21 2019-06-04 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and drug conjugates for use in melanoma
US10017580B2 (en) 2014-04-15 2018-07-10 ADC Therpeutics S.A. Humanized anti-Tn-MUC1 antibodies and their conjugates
US10428156B2 (en) 2014-09-05 2019-10-01 Abbvie Stemcentrx Llc Anti-MFI2 antibodies and methods of use
JP2017535246A (en) * 2014-09-05 2017-11-30 アッヴィ・ステムセントルクス・エル・エル・シー Novel anti-MFI2 antibody and method of use
EP3189079A4 (en) * 2014-09-05 2018-08-08 Abbvie Stemcentrx LLC Novel anti-mfi2 antibodies and methods of use
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10179820B2 (en) 2014-09-12 2019-01-15 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
US10556966B2 (en) 2014-09-12 2020-02-11 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
WO2016166304A1 (en) 2015-04-15 2016-10-20 Van Berkel Patricius Hendrikus Cornelis Site-specific antibody-drug conjugates
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
EP3756691A1 (en) 2015-06-12 2020-12-30 Seagen Inc. Anti-cd123 antibodies and conjugates thereof
US10912842B2 (en) 2015-06-12 2021-02-09 Seattle Genetics, Inc. CD123 antibodies and conjugates thereof
EP4302784A2 (en) 2015-06-30 2024-01-10 Seagen Inc. Anti-ntb-a antibodies and related compositions and methods
WO2017004330A1 (en) 2015-06-30 2017-01-05 Seattle Genetics, Inc. Anti-ntb-a antibodies and related compositions and methods
US10899775B2 (en) 2015-07-21 2021-01-26 Immunogen, Inc. Methods of preparing cytotoxic benzodiazepine derivatives
US10639373B2 (en) 2015-10-02 2020-05-05 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
US10058613B2 (en) 2015-10-02 2018-08-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US20190083645A1 (en) * 2015-12-04 2019-03-21 Abbvie Stemcentrx Llc Novel anti-claudin antibodies and methods of use
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
WO2017129652A1 (en) * 2016-01-26 2017-08-03 Medimmune Limited Pyrrolobenzodiazepines
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11027021B2 (en) 2016-03-15 2021-06-08 Seagen Inc. Combinations of PBD-based antibody drug conjugates with Bcl-2 inhibitors
WO2017160954A1 (en) 2016-03-15 2017-09-21 Seattle Genetics, Inc. Combinations of pbd-based antibody drug conjugates with bcl-2 inhibitors
US11844839B2 (en) 2016-03-25 2023-12-19 Seagen Inc. Process for the preparation of pegylated drug-linkers and intermediates thereof
US10934359B2 (en) 2016-04-21 2021-03-02 Abbvie Stemcentrx Llc Anti-BMPR1B antibodies and methods of use
WO2017186894A1 (en) * 2016-04-29 2017-11-02 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
US10918627B2 (en) 2016-05-11 2021-02-16 Massachusetts Institute Of Technology Convergent and enantioselective total synthesis of Communesin analogs
WO2017201132A2 (en) 2016-05-18 2017-11-23 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
US11110179B2 (en) 2016-06-03 2021-09-07 Seagen Inc. Combination of CD33 antibody drug conjugates with chemotherapeutic agents
US11191771B2 (en) 2016-06-09 2021-12-07 Seagen Inc. Combinations of PBD-based antibody drug conjugates with FLT3 inhibitors
WO2017223275A1 (en) 2016-06-24 2017-12-28 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2018091646A1 (en) * 2016-11-17 2018-05-24 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11813335B2 (en) 2017-02-08 2023-11-14 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11384098B2 (en) 2017-02-08 2022-07-12 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
EP3939616A1 (en) 2017-02-08 2022-01-19 ADC Therapeutics SA Pyrrolobenzodiazepine-antibody conjugates
WO2018159582A1 (en) 2017-02-28 2018-09-07 学校法人近畿大学 Method for treating egfr-tki-resistant non-small cell lung cancer by administration of anti-her3 antibody-drug conjugate
WO2018175994A1 (en) 2017-03-24 2018-09-27 Seattle Genetics, Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
US11730822B2 (en) 2017-03-24 2023-08-22 Seagen Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
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
US11932650B2 (en) 2017-05-11 2024-03-19 Massachusetts Institute Of Technology Potent agelastatin derivatives as modulators for cancer invasion and metastasis
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
US11691982B2 (en) 2017-09-20 2023-07-04 Ph Pharma Co., Ltd. Thailanstatin analogs
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
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
WO2019065964A1 (en) 2017-09-29 2019-04-04 第一三共株式会社 Antibody-pyrrolobenzodiazepine derivative conjugate
KR20200061376A (en) 2017-09-29 2020-06-02 다이이찌 산쿄 가부시키가이샤 Antibody-pyrrolobenzodiazepine derivative conjugate
US10640508B2 (en) 2017-10-13 2020-05-05 Massachusetts Institute Of Technology Diazene directed modular synthesis of compounds with quaternary carbon centers
WO2019096788A1 (en) 2017-11-14 2019-05-23 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11638760B2 (en) 2017-11-27 2023-05-02 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
WO2019104289A1 (en) 2017-11-27 2019-05-31 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
WO2019126691A1 (en) 2017-12-21 2019-06-27 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
CN111670045A (en) * 2017-12-22 2020-09-15 阿尔麦克探索有限公司 ROR 1-specific antigen-binding molecules
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
WO2020059772A1 (en) 2018-09-20 2020-03-26 第一三共株式会社 Treatment of her3 mutant cancer by administration of anti-her3 antibody-drug conjugate
WO2020100954A1 (en) 2018-11-14 2020-05-22 第一三共株式会社 (anti-cdh6 antibody)-(pyrrolobenzodiazepine derivative) conjugate
KR20210091711A (en) 2018-11-14 2021-07-22 다이이찌 산쿄 가부시키가이샤 Anti-CDH6 antibody-pyrrolobenzodiazepine derivative conjugate
WO2020127573A1 (en) 2018-12-19 2020-06-25 Adc Therapeutics Sa Pyrrolobenzodiazepine resistance
GB201820725D0 (en) 2018-12-19 2019-01-30 Adc Therapeutics Sarl Pyrrolobenzodiazepine resistance
WO2020173902A1 (en) * 2019-02-25 2020-09-03 Medimmune Limited Methods of synthesis and intermediates
US11833214B2 (en) 2019-03-21 2023-12-05 Immunogen, Inc. Methods of preparing cell-binding agent-drug conjugates
WO2020191306A1 (en) * 2019-03-21 2020-09-24 Immunogen, Inc. Methods of preparing cell-binding agent-drug conjugates
KR20210141630A (en) 2019-03-25 2021-11-23 다이이찌 산쿄 가부시키가이샤 Anti-HER2 antibody-pyrrolobenzodiazepine derivative conjugate
WO2020196475A1 (en) 2019-03-25 2020-10-01 第一三共株式会社 Anti-her2 antibody-pyrrolobenzodiazepine derivative conjugate
KR20210143237A (en) 2019-03-25 2021-11-26 다이이찌 산쿄 가부시키가이샤 Antibody-pyrrolobenzodiazepine derivative conjugate
WO2020196474A1 (en) 2019-03-25 2020-10-01 第一三共株式会社 Antibody-pyrrolobenzodiazepine derivative conjugate
WO2020196712A1 (en) 2019-03-27 2020-10-01 第一三共株式会社 Combination of antibody-pyrrolobenzodiazepine derivative conjugate and parp inhibitor
KR20210143839A (en) 2019-03-27 2021-11-29 다이이찌 산쿄 가부시키가이샤 Combination of antibody-pyrrolobenzodiazepine derivative conjugate and PARP inhibitor
US11535634B2 (en) 2019-06-05 2022-12-27 Massachusetts Institute Of Technology Compounds, conjugates, and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines and uses thereof
WO2020245283A1 (en) 2019-06-07 2020-12-10 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11484606B2 (en) 2019-06-07 2022-11-01 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2021024133A2 (en) 2019-08-06 2021-02-11 Glaxosmithkline Intellectual Property Development Limited Biopharmacuetical compositions and related methods
WO2021080608A1 (en) 2019-10-25 2021-04-29 Medimmune, Llc Branched moiety for use in conjugates
WO2022014698A1 (en) 2020-07-17 2022-01-20 第一三共株式会社 Method for producing antibody-drug conjugate
WO2022050300A1 (en) 2020-09-02 2022-03-10 第一三共株式会社 NOVEL ENDO-β-N-ACETYLGLUCOSAMINIDASE
WO2022153194A1 (en) 2021-01-13 2022-07-21 Memorial Sloan Kettering Cancer Center Antibody-pyrrolobenzodiazepine derivative conjugate
WO2023012669A2 (en) 2021-08-03 2023-02-09 Glaxosmithkline Intellectual Property Development Limited Biopharmaceutical compositions and stable isotope labeling peptide mapping method

Also Published As

Publication number Publication date
KR20130038254A (en) 2013-04-17
EP2558127B1 (en) 2022-01-19
US20180228916A1 (en) 2018-08-16
US20130028919A1 (en) 2013-01-31
KR20160128443A (en) 2016-11-07
US10561739B2 (en) 2020-02-18
CA2795349C (en) 2016-11-29
CN103068405A (en) 2013-04-24
KR101671360B1 (en) 2016-11-01
EA024118B1 (en) 2016-08-31
BR112012026801B1 (en) 2021-05-04
MX2012011900A (en) 2013-03-21
US20190336614A1 (en) 2019-11-07
BR112012026801B8 (en) 2021-05-25
NZ602932A (en) 2014-08-29
CN107019804A (en) 2017-08-08
MX2018013332A (en) 2020-11-12
US20160129013A1 (en) 2016-05-12
ZA201207317B (en) 2015-04-29
JP2013523896A (en) 2013-06-17
KR101772354B1 (en) 2017-08-28
US9592240B2 (en) 2017-03-14
US9242013B2 (en) 2016-01-26
EA201290838A1 (en) 2013-05-30
CA2795349A1 (en) 2011-10-20
JP5870400B2 (en) 2016-03-01
BR112012026801A2 (en) 2016-09-20
EP2558127A1 (en) 2013-02-20
IL252864A0 (en) 2017-08-31
IL222269A (en) 2017-06-29
IL222269A0 (en) 2012-12-31
AU2011239522B2 (en) 2014-10-23
IL252864B (en) 2018-01-31
US20170143846A1 (en) 2017-05-25
AU2011239522A1 (en) 2012-11-01
EP4039280A1 (en) 2022-08-10

Similar Documents

Publication Publication Date Title
US10561739B2 (en) Targeted pyrrolobenzodiazapine conjugates
US10328084B2 (en) Pyrrolobenzodiazepines and targeted conjugates
CA2795353C (en) Pyrrolobenzodiazepines used to treat proliferative diseases
AU2012322934B2 (en) Pyrrolobenzodiazepines
EP3309162A1 (en) Targeted conjugates of pyrrolobenzodiazepines
WO2013053871A1 (en) Pyrrolobenzodiazepines
EP2751120A1 (en) Pyrrolobenzodiazepines as unsymmetrical dimeric pbd compounds for inclusion in targeted conjugates

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180029867.4

Country of ref document: CN

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

Ref document number: 11716754

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 201290838

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2795349

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2011716754

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013505171

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2012/011900

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 13641219

Country of ref document: US

ENP Entry into the national phase

Ref document number: 20127028121

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2011239522

Country of ref document: AU

Date of ref document: 20110415

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 9658/DELNP/2012

Country of ref document: IN

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012026801

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012026801

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20121015