US20160354482A1 - Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses - Google Patents

Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses Download PDF

Info

Publication number
US20160354482A1
US20160354482A1 US14/913,965 US201414913965A US2016354482A1 US 20160354482 A1 US20160354482 A1 US 20160354482A1 US 201414913965 A US201414913965 A US 201414913965A US 2016354482 A1 US2016354482 A1 US 2016354482A1
Authority
US
United States
Prior art keywords
formula
composition
compound
diastereomer
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/913,965
Inventor
Thomas Nittoli
Nareshkumar F. Jain
Thomas Patrick MARKOTAN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US14/913,965 priority Critical patent/US20160354482A1/en
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JAIN, NARESHKUMAR F., MARKOTAN, Thomas Patrick, NITTOLI, THOMAS
Publication of US20160354482A1 publication Critical patent/US20160354482A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • A61K47/48384
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/537Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines spiro-condensed or forming part of bridged ring systems
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • A61K47/48569
    • 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
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • A61K47/6809Antibiotics, e.g. antitumor antibiotics anthracyclins, adriamycin, doxorubicin or daunomycin
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • Proliferative diseases are characterized by uncontrolled growth and spread of abnormal cells. If the spread of these cells is not controlled, it can result in death.
  • Proliferative diseases for example cancer, can be treated by surgery, radiation, chemotherapy, hormone-based therapy and/or immunotherapy. A number of these treatments, particularly the chemotherapy, are based on the use of anti-proliferative drugs which limit the spread of the abnormal cells.
  • Anti-proliferative drugs typically are indiscriminate in their ability to kill cells, affecting both normal and abnormal cells based simply on whether a cell is replicating. Regardless, most anti-proliferative drugs require a relatively high concentration at the locale of the abnormal cell proliferation to be effective. It is this combination of providing sufficient anti-proliferative drug at a site of abnormal cell growth without also causing significant death to normal cells systemically or in the vicinity of these cells that this disclosure addresses.
  • conjugates of tumor targeted probes such as antibodies or growth factors
  • toxins such as pseudomonas or diphtheria toxins.
  • Conjugates for use in the treatment of cancer thereby target the anti-proliferative drug to a population of abnormal cells.
  • conjugates that include the toxin maytansine have been employed for the treatment of cancer. Maytansine has shown great effectiveness as an anti-proliferative agent but the compound's toxicity has proven problematic toward normal cells.
  • anti-proliferative compounds exhibit asymmetric structures, such as the Maytansinoid family of macrolides, and may therefore exist in the form of a racemic mixture, in the form of separate enantiomers with configuration “R” and “S”, or (+) and ( ⁇ ), per stereogenic center, and various diastereomers.
  • the present disclosure shows that targeted delivery of a single maytansinoid diastereomer exhibits improved inhibitory cell proliferation profile as compared to delivery of its respective mixture of diastereomers. Therefore, diastereomers in accordance with embodiments described herein can be used to prepare medicinal products with improved therapeutic profile that can be useful for the treatment of specific diseases and conditions, particularly cancer.
  • compositions comprising a plurality of drug molecules for formula (I):
  • compositions comprising a plurality of drug molecules of formula I, wherein n is 1, and R 1 and R 2 are each independently hydrogen.
  • composition comprising a plurality of drug molecules of formula I are present in the composition in a diastereomeric excess of about 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more.
  • the drug molecules of formula I are present in the composition in a diastereomeric excess of from about 90% to about 100%, or from about 95% to about 100%, or from about 98% to about 100%.
  • composition comprising a plurality of drug molecules of formula I wherein one of the at least two diastereomers is characterized by a 1 H NMR spectra of FIG. 1 .
  • the term “about”, when used in reference to a particular recited numerical value, means that the value may vary from the recited value by no more than 1%.
  • the expression “about 100” includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
  • compositions comprising a plurality of drug molecules of formula I wherein the drug molecules are present in the composition in a diastereomeric excess of greater than 50% and show greater anti-proliferative activity than a corresponding composition comprising drug molecules of formula I that are not present in a diastereomeric excess of greater than 50%.
  • compositions comprising a plurality of drug molecules of formula I further comprising a therapeutically effective amount of a second or additional agent including, for example, a chemotherapeutic agent, an anti-inflammatory agent, an antibiotic, and the like.
  • compositions comprising a plurality of drug molecules of formula I represented by the following structure:
  • one of the at least two diastereomers is a compound of formula (i)
  • one of the at least two diastereomers is a compound of formula (ii)
  • the present disclosure also provides a compound of formula (i) or (ii),
  • compositions comprising a plurality of ligand-drug conjugates of formula (II):
  • compositions comprising a plurality of ligand-drug conjugates of formula II, wherein n is 1, and R 1 and R 2 are each independently hydrogen.
  • compositions comprising a plurality of ligand-drug conjugates of formula II are present in the composition in a diastereomeric excess of about 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more.
  • the drug molecules of formula II are present in the composition in a diastereomeric excess of from about 90% to about 100%, or from about 95% to about 100%, or from about 98% to about 100%.
  • compositions comprising a plurality of ligand-drug conjugates of formula II in the compositions in a diastereomeric excess of greater than 50% and show greater antiproliferative activity than a corresponding composition comprising ligand-drug conjugates of formula II that are not present in a diastereomeric excess of more than 50%.
  • compositions comprising a plurality of ligand-drug conjugates of formula II, wherein the ligand is an antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof can be developed to specifically bind a tumor-associated antigen.
  • the n can be 1, and R 1 and R 2 can each be independently a hydrogen.
  • the disclosure provides compositions comprising a plurality of ligand-drug conjugates of formula II, wherein the ligand is an antibody, or antigen-binding fragment thereof which specifically binds a tumor-associated antigen, and the ligand-drug conjugates are present in the composition in a diastereomeric excess of more than about 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more.
  • compositions comprising a plurality of ligand-drug conjugates of formula II, wherein the antibody, or antigen-binding fragment thereof specifically binds a tumor-associated antigen and further wherein the tumor-associated antigen is selected from the group consisting of AFP, ALK, BAGE proteins, ⁇ -catenin, brc-abl, BRCA1, BORIS, CA9, carbonic anhydrase IX, caspase-8, CD20, CD40, CDK4, CEA, CLEC12A, cMET, CTLA4, cyclin-B1, CYP1B1, EGFR, EGFRvIII, ErbB2/Her2, ErbB3, ErbB4, ETV6-AML, EphA2, Fra-1, FOLR1, GAGE proteins (e.g., GAGE-1, -2), GD2, GD3, GloboH, glypican-3, GM3, gp100, Her2, HLA/B-raf, HLA/k-ras
  • the antibody, or antigen-binding fragment have amino acid sequences that vary from the above-mentioned antibodies but that retain the ability to bind a given tumor-associated antigen.
  • Such variant antibodies and antibody fragments can comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence, but exhibit biological activity that is essentially equivalent to that of the described antibodies.
  • Two conjugates are considered bioequivalent if, for example, they are pharmaceutical equivalents or pharmaceutical alternatives whose rate and extent of absorption do not show a significant difference when administered at the same molar dose under similar experimental conditions, either single dose or multiple dose.
  • Some conjugates will be considered equivalents or pharmaceutical alternatives if they are equivalent in the extent of their absorption but not in their rate of absorption and yet may be considered bioequivalent because such differences in the rate of absorption are intentional and are reflected in the labeling, are not essential to the attainment of effective body drug concentrations on, e.g., chronic use, and are considered medically insignificant for the particular drug product studied.
  • two conjugates are bioequivalent if there are no clinically meaningful differences in their safety, purity, and potency.
  • two conjugates are bioequivalent if a patient can be switched one or more times between the reference product and the biological product without an expected increase in the risk of adverse effects, including a clinically significant change in immunogenicity, or diminished effectiveness, as compared to continued therapy without such switching.
  • two conjugates are bioequivalent if they both act by a common mechanism or mechanisms of action for the condition or conditions of use, to the extent that such mechanisms are known.
  • Bioequivalence may be demonstrated by in vivo and in vitro methods.
  • Bioequivalence measures include, e.g., (a) an in vivo test in humans or other mammals, in which the concentration of the conjugate or its metabolites is measured in blood, plasma, serum, or other biological fluid as a function of time; (b) an in vitro test that has been correlated with and is reasonably predictive of human in vivo bioavailability data; (c) an in vivo test in humans or other mammals in which the appropriate acute pharmacological effect of the conjugate (or its target) is measured as a function of time; and (d) in a well-controlled clinical trial that establishes safety, efficacy, or bioavailability or bioequivalence of a conjugate.
  • the present disclosure also provides a novel method for preparation of a composition comprising a plurality of drug molecules of formula (I):
  • the disclosure provides a method for preparing composition comprising a plurality of drug molecules of formula I, wherein the method further comprises step (d), further wherein step (d) comprises purifying the compound of formula I obtained in step (c) as explained above.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the solid substrate is selected from the group consisting of silica gel, celite, alumina, a zeolite, and crushed molecular sieves.
  • the solid substrate is selected from the group consisting of silica gel, celite, alumina, a zeolite, and crushed molecular sieves.
  • Other like solid substrates may also be utilized as long as the solid substrate allows for proper positioning of the macrolide III to allow stereoselective addition of maleimide IV.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein n is 1, and R 1 and R 2 are each independently hydrogen.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the organic solvent comprises a polar aprotic solvent such as DMF, DMA, HMPT, NMP, acetonitrile, dioxane, acetone, DMSO, THF, ethyl acetate, methyl acetate, methylene chloride, propylene carbonate or mixtures thereof.
  • a polar aprotic solvent such as DMF, DMA, HMPT, NMP, acetonitrile, dioxane, acetone, DMSO, THF, ethyl acetate, methyl acetate, methylene chloride, propylene carbonate or mixtures thereof.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the polar aprotic solvent comprises acetonitrile.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the organic solvent and the water are present in ratio of from about 1:1 to about 4:1 or from about 1:1 to about 10:1.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the molar ratio of the starting material having formula III and the compound of formula IV is from about 1:1 to about 1:3.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, further comprising combining the compound of formula I with an antibody or antigen-binding fragment thereof to make an antibody-drug conjugate.
  • the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the compound of formula I is attached to the antibody or antigen-binding fragment via an S, O, or NR 3 .
  • the disclosure provides a method for preparing a composition comprising a plurality of drug molecules of formula I, wherein the drug molecules of formula I are present in the composition in a diastereomeric excess of about 60% 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more.
  • compositions comprising a plurality of drug molecules of formula I represented by the following structure:
  • compositions of the present invention in a diastereoselective excess of greater than 50%.
  • chromatographic separation of a racemic mixture or mixture of diastereomers e.g., HPLC on normal, reverse, or chiral stationary phase using polar aprotic solvent mixtures, etc.
  • compositions wherein a plurality of the drug molecules of formula I, or the ligand-drug conjugates of formula II, are contained within the compositions in a therapeutically effective amount, and further comprising a pharmaceutically acceptable diluent, carrier or excipient.
  • compositions comprising a plurality of drug molecules of formula II further comprising a therapeutically effective amount of a second chemotherapeutic agent.
  • compositions comprise a compound of formula (I) and/or (II) of the present disclosure which may be administered in combination with one or more additional compounds or therapies.
  • Co-administration and combination therapy are not limited to simultaneous administration, separately or together, but also include sequential administrations.
  • Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a composition comprising one or more compounds of formula (I) and/or (II) and one or more other therapeutic agents; as well as administration of a composition comprising compound of formula (I) and/or (II) of the present disclosure and one or more additional agent(s) in its own separate pharmaceutical dosage formulation.
  • a composition comprising a compound of formula (I) and/or (II) and, a cytotoxic agent, a chemotherapeutic agent, or a growth inhibitory agent can be administered to the patient together in a single dosage composition such as a combined formulation, or each agent can be administered in a separate dosage formulation.
  • a composition comprising a compound of formula (I) and/or (II) and one or more additional agents can be administered concurrently, or separately at staggered times, i.e., sequentially.
  • Non-limiting examples of such additional therapeutic agents include cytokine inhibitors (e.g., an interleukin-1 (IL-1) inhibitor (such as rilonacept or anakinra, a small molecule IL-1 antagonist, or an anti-IL-1 antibody); IL-18 inhibitor (such as a small molecule IL-18 antagonist or an anti-IL-18 antibody); IL-4 inhibitor (such as a small molecule IL-4 antagonist, an anti-IL-4 antibody or an anti-IL-4 receptor antibody); IL-6 inhibitor (such as a small molecule IL-6 antagonist, an anti-IL-6 antibody or an anti-IL-6 receptor antibody); aspirin; NSAIDs; steroids (e.g., prednisone, methotrexate, etc.); low dose cyclosporine A; tumor necrosis factor (TNF) or TNF receptor inhibitors (e.g., a small molecule TNF or TNFR antagonist or an anti-TNF or TNFR antibody); uric acid synthesis inhibitors (e.g.,
  • the additional therapeutic agent(s) may be administered prior to, concurrent with, or after the administration of the one or more compounds of formula (I) and/or (II) (for purposes of the present disclosure, such administration regimens are considered the administration of one or more compounds of formula (I) and/or (II) “in combination with” a therapeutic agent).
  • cytotoxic agent refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • the term is intended to include radioactive isotopes (e.g., I 131 , I 125 , Y 90 and Re 186 ), chemotherapeutic agents, and toxins such as enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof.
  • chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofos
  • anti-hormonal agents that act to regulate or inhibit hormone action on tumors
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • a “growth inhibitory agent” when used herein refers to a compound or composition which inhibits growth of a cell, especially a cancer cell either in vitro or in vivo.
  • growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest.
  • Classical M-phase blockers include the vincas (vincristine and vinblastine), TAXOL®, and topo II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin.
  • DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C.
  • compositions comprise a therapeutically effective amount of an active agent and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the United States Federal or State government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection.
  • a solubilizing agent such as lidocaine to ease pain at the site of the injection.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the active agents of the disclosure can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the active agent of the disclosure which will be effective in the treatment of a medical condition, can be determined by standard clinical techniques based on the present description.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each subject's circumstances.
  • suitable dosage ranges for intravenous administration are generally about 0.5 to 20 milligrams of active compound per kilogram body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the disclosure provides a method of treatment of a medical disorder in an individual suffering from the medical disorder comprising administering to the individual an effective amount of a composition comprising a compound of formula (I) and/or (II), and further comprising administering sequentially or consecutively an additional therapy or administering at least one additional therapeutic agent.
  • the disclosure relates to a method of treating a disease sensitive to treatment with said method, said method comprising parenterally administering to a patient in need thereof a therapeutically effective dose of the composition comprising a compound of formula (I) and/or (II).
  • compositions comprising compounds of formula (I), formula (II), formula (i), formula (ii), or a combination thereof and/or pharmaceutical formulations thereof in the manufacture of a medicament for the treatment, prevention and/or amelioration of a medical disorder.
  • compositions comprising compounds of formula (I), formula (II), formula (i), formula (ii), or a combination thereof and/or pharmaceutical formulations thereof in the manufacture of a medicament for the treatment, prevention and/or amelioration of a tumor.
  • compositions herein can also be useful in the treatment of a medical disorder selected from autoimmune diseases and other immunological diseases and dysfunctions, inflammatory diseases, infectious diseases, neurodegenerative diseases, bone disorders, and cardiovascular diseases. Further, any disorder that can benefit from the targeted delivery of a toxic substance to particular cells, cell types, tissues and/or organs is within the scope of the present invention.
  • compositions comprising mixtures of compounds as represented by formula (I) and formula (II).
  • conjugate refers to compound having a Ligand, linker and Biologically Active Molecule.
  • Illustrative examples include compounds of formula (II).
  • spacer refers to chemical building blocks of the linker used to spatially separate the Ligand from the Biologically Active Molecule and to allow for catabolism of the linker inside of cells.
  • macrolide refers to any Biologically Active Molecule having a macrolide ring.
  • alkyl refers to a hydrocarbon radical having a straight or branched chain or combinations thereof.
  • Alkyl radicals can be a univalent, a bivalent or a cyclic radical. Examples of univalent alkyl radicals are methyl, ethyl, 1-propyl, 2-propyl, n-butyl, isobutyl, sec-butyl, t-butyl, pentyl, neopentyl, hexyl, isohexyl, and the like.
  • bivalent alkyl radicals include
  • cyclic alkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • Typical alkyl radicals have from one to ten carbon atoms, one to nine carbon atoms, one to eight carbon atoms, one to seven carbon atoms, one to six carbon atoms, one to five carbon atoms, one to four carbon atoms, one to three carbon atoms, one to two carbon atoms or one carbon atom.
  • Typical cycloalkyl are 3 to 7 member monocyclic ring radicals.
  • pharmaceutically acceptable salt refers to both organic and inorganic salts of the conjugate compounds described herein, e.g., compounds of formula (I), and (II).
  • the salts are pharmaceutically acceptable and include: sulfate, citrate, nitrate, phosphate, ascorbate, bromide, gluconate, benzoate, oxalate, pantothenate, and the like.
  • pharmaceutically acceptable salts herein may include more than one charged atom in its structure as well as one or more counter ion. Preparation of conjugate compounds herein as pharmaceutically acceptable salts is well known to one of skill in the art.
  • ligand as used herein means any molecule or compound that specifically or selectively interacts with and/or binds to a second molecule or compound.
  • a ligand is an antibody or antigen-binding fragment thereof.
  • Other examples of ligands suitable for use in the context of the present invention include, e.g., aptamers, peptides that specifically interact with a particular antigen (e.g., peptibodies), receptor molecules, and antigen-binding scaffolds (e.g., DARPins, HEAT repeat proteins, ARM repeat proteins, tetratricopeptide repeat proteins, and other scaffolds based on naturally occurring repeat proteins, etc., [see, e.g., Boersma and Pluckthun, 2011 , Curr. Opin. Biotechnol. 22:849-857, and references cited therein]).
  • Antibodies exist as intact immunoglobulins, or as a number of well-characterized fragments produced by digestion with various peptidases. For example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′ 2 , a dimer of Fab which itself is a light chain joined to V H -C H by a disulfide bond.
  • the F(ab)′ 2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)′ 2 dimer into an Fab′ monomer.
  • the Fab′ monomer is essentially Fab with part of the hinge region.
  • antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology.
  • antibody also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv (scFv) single variable domains (Dabs)) or those identified using display libraries such as phage, E. coli or yeast display libraries (see, for example, McCafferty et al. (1990) Nature 348:552-554).
  • scFv single chain Fv
  • Dabs single variable domains
  • human antibody as used herein is intended to include antibodies having variable and constant regions derived from human immunoglobulin sequences.
  • the human mAbs of the invention may include amino acid residues not encoded by human immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the term “human antibody”, as used herein, is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species have been grafted onto human FR sequences.
  • terapéuticaally effective amount refers to an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • racemate as used herein means an equimolar mixture of a pair of enantiomers.
  • racemate also refers to a racemic mixture.
  • enantiomer refers to compounds which non-superimposable with the mirror images of each other. Enantiomers may exist in either the (R) or (S) configuration.
  • stereoselective synthesis refers to a chemical reaction that leads to formation of a single stereoisomer or an enantiomer-enriched mixture of isomers from among two or more possible stereoisomers.
  • diastereomeric excess refers to the difference between the mole fraction of the desired single diastereomer as compared to the remaining diastereomers in a composition. Diastereomeric excess is calculated as follows:
  • stereomerically pure refers to a compound wherein the indicated stereoisomer is present to a greater extent than other stereoisomers of that compound, e.g., the compound is present in diastereomeric excess.
  • the stereomerically pure compounds described herein comprise 80% or greater, 85% or greater, 90% or greater, 95% or greater, or 97% or greater by weight of one stereoisomer of the compound.
  • Conjugates in accordance with various embodiments described herein can be prepared by any known method in the art.
  • An illustrative protocol for producing conjugates is provided in the Examples below.
  • the conjugates can be prepared by i) reacting a Ligand with drug molecules of formula (I) to form a conjugate of formula (II), and ii) purifying the conjugate.
  • the conjugates are prepared by reacting a Ligand, linker and biologically active macrolide in a single reaction. Once the conjugates in accordance with the invention are prepared they can be purified.
  • compositions comprising drug molecules of formula (I) and/or compositions comprising conjugate compounds of formula (II) described herein can be evaluated for their ability to suppress proliferation of various cancer cell lines in vitro.
  • compositions comprising drug molecules or formula (I) and/or compositions comprising conjugate compounds of formula (II) can be applied to in vitro plated cancer cells for a predetermined number of days and surviving cells measured in assays by known methods.
  • FIG. 1 illustrates an 1 H-NMR spectrum of Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxysuccinate (5).
  • the 1 H-NMR spectrum is consistent with a single diastereomer present in at least 95% diastereomeric excess since the spectrum is not complicated by resonances attributable to the other diastereomer.
  • Example 2 sets forth the 1 H-NMR spectrum of the mixture of diastereomers).
  • FIG. 2 illustrates an 1 H-NMR spectrum of mixture of diastereomers of Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxysuccinate (6).
  • FIG. 3 illustrates that in SKBR3 cells the single diastereomer compound conjugate HER2-5 (in vitro and in vivo lots) possessed an IC50 value of 0.3 nM versus 0.9 nM for the mixture of diastereomer conjugate HER2-6.
  • FIG. 4 illustrates that in BT474 cells the single diastereomer compound conjugate HER2-5 (in vitro) in possessed an IC 50 value of 4.6 nM while the HER2-5 (in vivo) lot had an IC 50 value of 4.0 nM versus 11.6 nM for the mixture of diastereomer conjugate HER2-6.
  • FIG. 5 illustrates that in NCI-N87 cells the single diastereomer compound conjugate HER2-5 (in vitro) possessed an IC 50 value of 0.6 nM while the HER2-5 (in vivo) lot had an IC 50 value of 0.4 nM versus 1.0 nM for the mixture of diastereomer conjugate HER2-6.
  • FIG. 6 illustrates that in HEK293/hEGFRvIII cells the single diastereomer compound conjugate EGFRvIII-5 possessed an IC 50 value of 0.4 nM while the mixture of diastereomer conjugate EGFRvIII-6 had an IC 50 value of 0.5 nM.
  • FIG. 7 illustrates that in MMT/hEGFRvIII cells the single diastereomer compound conjugate EGFRvIII-5 possessed an IC 50 value of 0.5 nM while the mixture of diastereomer conjugate EGFRvIII-6 had an IC 50 value almost 20 fold higher at 9.8 nM.
  • FIG. 8 illustrates that in U251/hEGFRvIII cells the single diastereomer compound conjugate EGFRvIII-5 possessed an IC 50 value of 2.4 nM while the mixture of diastereomer conjugate EGFRvIII-6 had an IC 50 value of 3.3 nM.
  • FIG. 9 illustrates that in Ovcar-3 cells the single diastereomer compound conjugate MUC16-5 possessed an IC 50 value of 6.3 nM while the mixture of diastereomer conjugate MUC16-6 had an IC 50 value of 16.0 nM.
  • FIG. 10 illustrates that in PC3/MUC16long cells the single diastereomer compound conjugate MUC16-5 in possessed an IC 50 value of 0.34 nM while the mixture of diastereomer conjugate MUC16-6 had an IC 50 value at 0.80 nM.
  • FIG. 11 illustrates tumor growth curves in mice following dosing with HER2-5 and control reagents.
  • Mice received PBS vehicle ( ⁇ ), 300 ug/kg DM1-SMe ( ⁇ ) and Isotype Control mAb at 15 mg/kg ( ⁇ ).
  • Mice also received HER2 mAb ( ⁇ ), HER2-5 ( ⁇ ) and Isotype Control-5 ( ⁇ ) at doses of 1 mg/kg (A), 5 mg/kg (B) and 15 mg/kg (C).
  • Mice received 3 once weekly doses of conjugates and control agents as indicated by the black arrows (T. Groups are N 8, Mean ⁇ SE.).
  • FIG. 13 illustrates percentage change in animal weights following dosing with HER2-5 and control reagents.
  • Mice received PBS vehicle ( ⁇ ), 300 ug/kg DM1-SMe ( ⁇ ) and Isotype Control mAb at 15 mg/kg ( ⁇ ).
  • Mice also received HER2 mAb ( ⁇ ), HER2-5 ( ⁇ ) and Isotype Control-5 ( ⁇ ) at doses of 1 mg/kg (A), 5 mg/kg (B) and 15 mg/kg (C).
  • Mice received 3 once weekly doses of conjugates and control agents as indicated by the black arrows (T. Groups are N 8, Mean ⁇ SE.).
  • Proton NMR spectra (for compounds that could not be detected by UV) were acquired on a Varian Inova 300 MHz instrument, while mass spectra were collected on an Agilent 1100 series LC/MSD with electrospray ionization source and quadrupole ion trap analyzer. All conjugates were analyzed using a Bruker ultraFleXtreme MALDI-TOF-TOF mass spectrometer. All starting materials and solvents were purchased commercially and used without purification, unless otherwise noted.
  • the maleamic acid from Step A (36.8 g, 144 mmol) and sodium acetate (13.6 g, 165 mmol) were dissolved in acetic anhydride (368 mL), sealed in a glass reaction vessel, and heated to 120° C. for 3 hours.
  • the cooled reaction mixture (a black syrup) was poured onto water (3 L), stirred, and extracted with dichloromethane.
  • the organic layer was dried over Na 2 SO 4 , filtered over a sintered glass funnel, and the clear filtrate evaporated and dried under high vacuum giving the title compound as a yellow solid (7.00 g, 20%).
  • step B The product of the preceding step B (10.0 g, 42.1 mmol) was dissolved in dichloromethane (50 mL) under Ar, treated with N-hydroxysuccinimide (7.27 g, 63.2 mmol) and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDAC, 12.4 g, 64.5 mmol), and the reaction was stirred at ambient temperature overnight. The resulting brown solution was diluted with dichloromethane, washed with water and brine, dried over Na 2 SO 4 , filtered over a sintered glass funnel, and the filtrate concentrated and dried in vacuo.
  • EDAC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • DM1 The title compound, known in the art as DM1, was prepared using a modified version of the method described by Whitesides et al. ( J. Org. Chem., 1991, 56, 2648-2650). Maytansin-3-N-methyl-L-Ala-(3-methyldisulfanyl) propanamide (DM1-SMe, 2.42 g, 3.09 mmol, prepared in a manner similar to Ho and Carrozzella, U.S. Pat. Appl.
  • 2007/0037972 A1 was dissolved in acetonitrile (30 mL), treated with a solution of tris(2-carboxyethyl)phosphine hydrochloride (8.23 g, 28.7 mmol) in water (30 mL), the pH was raised to ca. 3 with the addition of saturated aqueous NaHCO 3 (5 mL), the flask was purged with Ar, and the reaction was stirred at ambient temperature under a rubber septum (vented due to effervescence). After 2 h, the reaction was treated with brine (ca. 100 mL), bubbled with Ar for 5 minutes (to remove the free methylmercaptan), and the phases separated.
  • Compound 5 comprises predominantly a single diastereomer of the linker-DM1 cytotoxic compound, whereas Compound 6 comprises a mixture of various linker-DM1 diastereomers.
  • the antibodies used in this Example were an anti-HER2 antibody having variable regions derived from humAb4D5-8 from Carter et al, PNAS 1992 89 4285, an anti-EGFRvIII antibody having variable regions derived from clone 131 from WO2013075048 A1, and an anti-MUC16 antibody having variable regions derived from clone 3A5 from WO2007001851.
  • the antibodies were expressed in CHO cells and purified by Protein A.
  • a non-binding isotype control antibody derived from an infectious disease antigen having no relation to oncology was also used in this Example.
  • the antibodies (10 mg/ml) in 50 mM HEPES, 150 mM NaCl, pH 8.0, and 10% (v/v) DMA were conjugated with a 6 fold excess of compound 5 or 6 for 1 hour at ambient temperature.
  • the conjugates were purified by size exclusion chromatography and sterile filtered. Protein and linker payload concentrations were determined by UV spectral analysis and MALDI-TOF mass spectrometry. Size-exclusion HPLC established that all conjugates used were >95% monomeric, and RP-HPLC established that there was ⁇ 0.5% unconjugated linker payload. Yields are reported in Table 1 based on protein. All conjugated antibodies were analyzed by UV for linker payload loading values according to Hamblett et al, Cancer Res 2004 10 7063 and by mass difference, native versus conjugated. The results are summarized in Table 1.
  • Cells were seeded in PDL-coated 96 well plates at 10,000 (SK-BR-3 and NCI-N87), 15,000 (BT-474), 3000 (Ovcar-3 and PC3/Muc16), 2000 (HEK293/hEGFRvIII), 1500 (U251/hEGFRvIII), or 400 (MMT/hEGFRvIII) cells per well in complete growth media and grown overnight.
  • serially diluted antibody-drug conjugates or free payload were added to the cells at final concentrations ranging from 1 ⁇ M to 1 pM and incubated for 3 days. Each concentration was run in duplicate and reported with the respective standard deviation.
  • the single diastereomer compound conjugate HER2-5 in vitro
  • the HER2-5 (in vivo) lot had an IC 50 value of 4.0 nM versus 11.6 nM for the mixture of diastereomer compound conjugate HER2-6 (Table 2, FIG. 4 ).
  • the naked HER2 antibody had little anti-proliferation activity.
  • the single diastereomer compound conjugate HER2-5 in vitro
  • the HER2-5 (in vivo) lot had an IC 50 value of 0.4 nM versus 1.0 nM for the mixture of diasteromer compound conjugate HER2-6 (Table 2, FIG. 5 ).
  • the naked HER2 antibody had little anti-proliferation activity.
  • the single diastereomer compound conjugate EGFRvIII-5 possessed an IC 50 value of 0.5 nM while the mixture of diastereomer compound conjugate EGFRvIII-6 had an IC 50 value of 9.8 nM (Table 2, FIG. 7 ).
  • the naked EGFRvIII antibody had little anti-proliferation activity.
  • the single diastereomer compound conjugate MUC16-5 possessed an IC 50 value of 6.3 nM while the mixture of diasteroemer compound conjugate MUC16-6 had an IC 50 value of 16.0 nM (Table 4, FIG. 8 ).
  • the naked Muc16 antibody had little anti-proliferation activity.
  • DM1-SMe maytansin-3-N-methyl-L-Ala-(3-methyldisulfanyl) propanamide
  • mice bearing HER2+ gastric cancer xenografts were assessed for efficacy.
  • 5 ⁇ 10 6 NCI-N87 cells (ATCC CRL-5822) were implanted subcutaneously into the lower right flank of CB-17 SCID mice (Taconic). Once tumors had reached an average volume of 150 mm 3 , mice were randomized in to groups of eight and dosed with HER2-5 or control reagents.
  • Control reagents included PBS vehicle, free DM1-SMe, isotype control, isotype control-5, or HER2. Mice received once weekly doses for three weeks and tumor volumes and body weights were monitored twice weekly throughout the study. Conjugates were dosed at 1, 5 and 15 mg/kg, as these doses had been shown to be effective in previous in vivo studies by Lewis-Phillips et al (Lewis-Phillips G et al., Can Res 2008).
  • HER2-5 demonstrated clear anti-tumor efficacy, with doses of 5 and 15 mg/kg leading to significant decreases in initial tumor volume and eradication of some tumors at the higher dose ( FIGS. 11 and 12 ). A significant delay in tumor growth relative to control agents was also observed in the 1 mg/kg dose level. No adverse events were observed following dosing, with mice receiving HER2-5 demonstrating robust weight gain throughout the study ( FIG. 13 ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The disclosure relates to compositions comprising diastereomer of a macrolide exhibiting improved therapeutic profile in the context of inhibiting cell proliferation compared to the corresponding compositions comprising mixture of diastereomers. The disclosure further provides drug-ligand conjugates formed using diastereomer of the macrolide. The disclosure also provides novel method of preparation of diastereomer of the macrolide and their therapeutic uses.

Description

  • Proliferative diseases are characterized by uncontrolled growth and spread of abnormal cells. If the spread of these cells is not controlled, it can result in death. Proliferative diseases, for example cancer, can be treated by surgery, radiation, chemotherapy, hormone-based therapy and/or immunotherapy. A number of these treatments, particularly the chemotherapy, are based on the use of anti-proliferative drugs which limit the spread of the abnormal cells. Anti-proliferative drugs, however, typically are indiscriminate in their ability to kill cells, affecting both normal and abnormal cells based simply on whether a cell is replicating. Regardless, most anti-proliferative drugs require a relatively high concentration at the locale of the abnormal cell proliferation to be effective. It is this combination of providing sufficient anti-proliferative drug at a site of abnormal cell growth without also causing significant death to normal cells systemically or in the vicinity of these cells that this disclosure addresses.
  • Various approaches to targeted drug delivery have been tried, including the use of conjugates of tumor targeted probes (such as antibodies or growth factors) with toxins such as pseudomonas or diphtheria toxins. Conjugates for use in the treatment of cancer thereby target the anti-proliferative drug to a population of abnormal cells. Recently, conjugates that include the toxin maytansine have been employed for the treatment of cancer. Maytansine has shown great effectiveness as an anti-proliferative agent but the compound's toxicity has proven problematic toward normal cells. A need exists for developing maytansine based conjugates that have sufficient activity for use as cancer therapeutics. The more active or effective the maytansine based conjugate is at inhibiting or killing a population of abnormal cells the lower the concentration of the conjugate is required, the benefit being a reduced overall risk of damaging normal cells.
  • Many anti-proliferative compounds exhibit asymmetric structures, such as the Maytansinoid family of macrolides, and may therefore exist in the form of a racemic mixture, in the form of separate enantiomers with configuration “R” and “S”, or (+) and (−), per stereogenic center, and various diastereomers. The present disclosure shows that targeted delivery of a single maytansinoid diastereomer exhibits improved inhibitory cell proliferation profile as compared to delivery of its respective mixture of diastereomers. Therefore, diastereomers in accordance with embodiments described herein can be used to prepare medicinal products with improved therapeutic profile that can be useful for the treatment of specific diseases and conditions, particularly cancer.
  • The present disclosure relates to compositions comprising a plurality of drug molecules for formula (I):
  • Figure US20160354482A1-20161208-C00001
  • wherein:
      • X is
  • Figure US20160354482A1-20161208-C00002
    Figure US20160354482A1-20161208-C00003
    Figure US20160354482A1-20161208-C00004
    Figure US20160354482A1-20161208-C00005
      • Y is Y1 or Y2 further wherein Y1 is
  • Figure US20160354482A1-20161208-C00006
      • or H;
  • Figure US20160354482A1-20161208-C00007
      • Y2 is —Cl, —Br, —I, or
      • Z is H or SO3H;
      • R1 and R2 are independently selected from H or alkyl;
      • each n is independently 0 or an integer from 1 to 50;
      • and wherein the drug molecules present in the composition comprises a mixture of at least two diastereomers, a first diastereomer and a second diastereomer, further wherein said first diastereomer and second diastereomer are otherwise identical, except that said first and second diastereomers have different stereochemical configuration at a chiral carbon represented by (*) in formula X, wherein said chiral carbon atom is a carbon atom that is bound to a sulfur atom, and said first or second diastereomer is present in a diastereomeric excess of greater than 50%.
  • In one embodiment, the disclosure provides compositions comprising a plurality of drug molecules of formula I, wherein n is 1, and R1 and R2 are each independently hydrogen.
  • In another embodiment, the composition comprising a plurality of drug molecules of formula I are present in the composition in a diastereomeric excess of about 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more.
  • In certain embodiments, the drug molecules of formula I are present in the composition in a diastereomeric excess of from about 90% to about 100%, or from about 95% to about 100%, or from about 98% to about 100%.
  • In another embodiment, the composition comprising a plurality of drug molecules of formula I wherein one of the at least two diastereomers is characterized by a 1H NMR spectra of FIG. 1.
  • As used herein, the term “about”, when used in reference to a particular recited numerical value, means that the value may vary from the recited value by no more than 1%. For example, as used herein, the expression “about 100” includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
  • In another embodiment, the disclosure provides compositions comprising a plurality of drug molecules of formula I wherein the drug molecules are present in the composition in a diastereomeric excess of greater than 50% and show greater anti-proliferative activity than a corresponding composition comprising drug molecules of formula I that are not present in a diastereomeric excess of greater than 50%.
  • Further, the disclosure herein provides compositions comprising a plurality of drug molecules of formula I further comprising a therapeutically effective amount of a second or additional agent including, for example, a chemotherapeutic agent, an anti-inflammatory agent, an antibiotic, and the like.
  • In an embodiment, the disclosure provides compositions comprising a plurality of drug molecules of formula I represented by the following structure:
  • Figure US20160354482A1-20161208-C00008
  • or mixtures thereof
  • in a diastereomeric excess of greater than 50%.
  • In one embodiment, one of the at least two diastereomers is a compound of formula (i)
  • Figure US20160354482A1-20161208-C00009
  • In another embodiment, one of the at least two diastereomers is a compound of formula (ii)
  • Figure US20160354482A1-20161208-C00010
  • The present disclosure also provides a compound of formula (i) or (ii),
  • Figure US20160354482A1-20161208-C00011
  • wherein the compound is steromerically pure.
  • The present disclosure also provides compositions comprising a plurality of ligand-drug conjugates of formula (II):
  • Figure US20160354482A1-20161208-C00012
  • wherein:
      • A is
  • Figure US20160354482A1-20161208-C00013
      • W is selected from S, O, or NR3;
      • L is a ligand;
      • further wherein:
      • L is capable of binding to a cell or cell population;
      • R1, R2 and R3 are each independently selected from H or alkyl;
      • n is 0 or an integer from 1 to 10;
      • p is an integer from 1 to 10;
      • and wherein the ligand-drug conjugates are present in the composition in a diastereomeric excess of greater than 50%.
  • In one embodiment, the disclosure provides compositions comprising a plurality of ligand-drug conjugates of formula II, wherein n is 1, and R1 and R2 are each independently hydrogen.
  • In an embodiment, the compositions comprising a plurality of ligand-drug conjugates of formula II are present in the composition in a diastereomeric excess of about 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more. In certain embodiments, the drug molecules of formula II are present in the composition in a diastereomeric excess of from about 90% to about 100%, or from about 95% to about 100%, or from about 98% to about 100%.
  • In another embodiment, the disclosure provides compositions comprising a plurality of ligand-drug conjugates of formula II in the compositions in a diastereomeric excess of greater than 50% and show greater antiproliferative activity than a corresponding composition comprising ligand-drug conjugates of formula II that are not present in a diastereomeric excess of more than 50%.
  • In an embodiment, the disclosure provides compositions comprising a plurality of ligand-drug conjugates of formula II, wherein the ligand is an antibody or antigen-binding fragment thereof. The antibody or antigen-binding fragment thereof can be developed to specifically bind a tumor-associated antigen. With respect to formula II, the n can be 1, and R1 and R2 can each be independently a hydrogen.
  • In certain embodiments, the disclosure provides compositions comprising a plurality of ligand-drug conjugates of formula II, wherein the ligand is an antibody, or antigen-binding fragment thereof which specifically binds a tumor-associated antigen, and the ligand-drug conjugates are present in the composition in a diastereomeric excess of more than about 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more.
  • In certain embodiments, the disclosure provides compositions comprising a plurality of ligand-drug conjugates of formula II, wherein the antibody, or antigen-binding fragment thereof specifically binds a tumor-associated antigen and further wherein the tumor-associated antigen is selected from the group consisting of AFP, ALK, BAGE proteins, β-catenin, brc-abl, BRCA1, BORIS, CA9, carbonic anhydrase IX, caspase-8, CD20, CD40, CDK4, CEA, CLEC12A, cMET, CTLA4, cyclin-B1, CYP1B1, EGFR, EGFRvIII, ErbB2/Her2, ErbB3, ErbB4, ETV6-AML, EphA2, Fra-1, FOLR1, GAGE proteins (e.g., GAGE-1, -2), GD2, GD3, GloboH, glypican-3, GM3, gp100, Her2, HLA/B-raf, HLA/k-ras, HLA/MAGE-A3, hTERT, IGF1R, LGR5, LMP2, MAGE proteins (e.g., MAGE-1, -2, -3, -4, -6, and -12), MART-1, mesothelin, ML-IAP, Muc1, Muc16 (CA-125), MUM1, NA17, NY-BR1, NY-BR62, NY-BR85, NY-ESO1, OX40, p15, p53, PAP, PAX3, PAX5, PCTA-1, PDGFR-α, PDGFR-β, PDGF-A, PDGF-B, PDGF-C, PDGF-D, PLAC1, PRLR, PRAME, PSMA (FOLH1), RAGE proteins, Ras, RGS5, Rho, SART-1, SART-3, Steap-1, Steap-2, survivin, TAG-72, TGF-β, TMPRSS2, Tn, TNFRSF17, TRP-1, TRP-2, tyrosinase, and uroplakin-3.
  • In some embodiments, the antibody, or antigen-binding fragment have amino acid sequences that vary from the above-mentioned antibodies but that retain the ability to bind a given tumor-associated antigen. Such variant antibodies and antibody fragments can comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence, but exhibit biological activity that is essentially equivalent to that of the described antibodies.
  • Two conjugates are considered bioequivalent if, for example, they are pharmaceutical equivalents or pharmaceutical alternatives whose rate and extent of absorption do not show a significant difference when administered at the same molar dose under similar experimental conditions, either single dose or multiple dose. Some conjugates will be considered equivalents or pharmaceutical alternatives if they are equivalent in the extent of their absorption but not in their rate of absorption and yet may be considered bioequivalent because such differences in the rate of absorption are intentional and are reflected in the labeling, are not essential to the attainment of effective body drug concentrations on, e.g., chronic use, and are considered medically insignificant for the particular drug product studied.
  • In one embodiment, two conjugates are bioequivalent if there are no clinically meaningful differences in their safety, purity, and potency.
  • In one embodiment, two conjugates are bioequivalent if a patient can be switched one or more times between the reference product and the biological product without an expected increase in the risk of adverse effects, including a clinically significant change in immunogenicity, or diminished effectiveness, as compared to continued therapy without such switching.
  • In one embodiment, two conjugates are bioequivalent if they both act by a common mechanism or mechanisms of action for the condition or conditions of use, to the extent that such mechanisms are known.
  • Bioequivalence may be demonstrated by in vivo and in vitro methods. Bioequivalence measures include, e.g., (a) an in vivo test in humans or other mammals, in which the concentration of the conjugate or its metabolites is measured in blood, plasma, serum, or other biological fluid as a function of time; (b) an in vitro test that has been correlated with and is reasonably predictive of human in vivo bioavailability data; (c) an in vivo test in humans or other mammals in which the appropriate acute pharmacological effect of the conjugate (or its target) is measured as a function of time; and (d) in a well-controlled clinical trial that establishes safety, efficacy, or bioavailability or bioequivalence of a conjugate.
  • The present disclosure also provides a novel method for preparation of a composition comprising a plurality of drug molecules of formula (I):
  • Figure US20160354482A1-20161208-C00014
  • wherein:
      • X is
  • Figure US20160354482A1-20161208-C00015
    Figure US20160354482A1-20161208-C00016
    Figure US20160354482A1-20161208-C00017
    Figure US20160354482A1-20161208-C00018
      • Y is Y1 or Y2 further wherein
      • Y1 is
  • Figure US20160354482A1-20161208-C00019
  • or H;
  • Figure US20160354482A1-20161208-C00020
      • Y2 is —Cl, —Br, —I, or
      • R1 and R2 are independently selected from H or alkyl;
      • each n is independently 0 or an integer from 1 to 50;
      • and wherein the drug molecules present in the composition comprises a mixture of at least two diastereomers, a first diastereomer and a second diastereomer, further wherein said first diastereomer and second diastereomer are otherwise identical, except that said first and second diastereomers have different stereochemical configuration at a chiral carbon represented by (*) in formula X, wherein said chiral carbon atom is a carbon atom that is bound to a sulfur atom, and said first or second diastereomer is present in a diastereomeric excess of greater than 50%, the method comprising:
      • (a) providing a mixture comprising
        • (i) a starting material which has a formula III:
  • Figure US20160354482A1-20161208-C00021
        • (ii) a compound of formula IV:
  • Figure US20160354482A1-20161208-C00022
      • Y1 is
  • Figure US20160354482A1-20161208-C00023
  • or H;
      • Y2 is —Cl, —Br, —I, or
  • Figure US20160354482A1-20161208-C00024
      • Z is H or SO3H;
      • R1 and R2 are independently selected from H or alkyl; and
      • each n is independently 0 or an integer from 1 to 50;
        • (iii) an organic solvent,
        • (iv) water, and
        • (v) a solid substrate;
      • (b) allowing the mixture of step (a) to react until some of the starting material is converted to the compound of formula I; and
      • (c) removing crude compound of formula I from the mixture of step (b).
  • In certain embodiments, the disclosure provides a method for preparing composition comprising a plurality of drug molecules of formula I, wherein the method further comprises step (d), further wherein step (d) comprises purifying the compound of formula I obtained in step (c) as explained above.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the solid substrate is selected from the group consisting of silica gel, celite, alumina, a zeolite, and crushed molecular sieves. Other like solid substrates may also be utilized as long as the solid substrate allows for proper positioning of the macrolide III to allow stereoselective addition of maleimide IV.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein n is 1, and R1 and R2 are each independently hydrogen.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the organic solvent comprises a polar aprotic solvent such as DMF, DMA, HMPT, NMP, acetonitrile, dioxane, acetone, DMSO, THF, ethyl acetate, methyl acetate, methylene chloride, propylene carbonate or mixtures thereof.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the polar aprotic solvent comprises acetonitrile.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the organic solvent and the water are present in ratio of from about 1:1 to about 4:1 or from about 1:1 to about 10:1.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the molar ratio of the starting material having formula III and the compound of formula IV is from about 1:1 to about 1:3.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, further comprising combining the compound of formula I with an antibody or antigen-binding fragment thereof to make an antibody-drug conjugate.
  • In certain embodiments, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I, wherein the compound of formula I is attached to the antibody or antigen-binding fragment via an S, O, or NR3.
  • In an embodiment, the disclosure provides a method for preparing a composition comprising a plurality of drug molecules of formula I, wherein the drug molecules of formula I are present in the composition in a diastereomeric excess of about 60% 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more.
  • In an embodiment, the disclosure provides a method for preparing compositions comprising a plurality of drug molecules of formula I represented by the following structure:
  • Figure US20160354482A1-20161208-C00025
  • in a diastereoselective excess of greater than 50%. Although certain exemplary methods for preparing the compositions of the present invention are set forth in the working examples herein, other methods are contemplated within the scope of the present invention such as, e.g., chromatographic separation of a racemic mixture or mixture of diastereomers (e.g., HPLC on normal, reverse, or chiral stationary phase using polar aprotic solvent mixtures, etc.).
  • The present disclosure also relates to compositions wherein a plurality of the drug molecules of formula I, or the ligand-drug conjugates of formula II, are contained within the compositions in a therapeutically effective amount, and further comprising a pharmaceutically acceptable diluent, carrier or excipient.
  • In an embodiment, the disclosure provides compositions comprising a plurality of drug molecules of formula II further comprising a therapeutically effective amount of a second chemotherapeutic agent.
  • In numerous embodiments, the compositions comprise a compound of formula (I) and/or (II) of the present disclosure which may be administered in combination with one or more additional compounds or therapies. Co-administration and combination therapy are not limited to simultaneous administration, separately or together, but also include sequential administrations.
  • Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a composition comprising one or more compounds of formula (I) and/or (II) and one or more other therapeutic agents; as well as administration of a composition comprising compound of formula (I) and/or (II) of the present disclosure and one or more additional agent(s) in its own separate pharmaceutical dosage formulation. For example, a composition comprising a compound of formula (I) and/or (II) and, a cytotoxic agent, a chemotherapeutic agent, or a growth inhibitory agent can be administered to the patient together in a single dosage composition such as a combined formulation, or each agent can be administered in a separate dosage formulation. Where separate dosage formulations are used, a composition comprising a compound of formula (I) and/or (II) and one or more additional agents can be administered concurrently, or separately at staggered times, i.e., sequentially.
  • Non-limiting examples of such additional therapeutic agents include cytokine inhibitors (e.g., an interleukin-1 (IL-1) inhibitor (such as rilonacept or anakinra, a small molecule IL-1 antagonist, or an anti-IL-1 antibody); IL-18 inhibitor (such as a small molecule IL-18 antagonist or an anti-IL-18 antibody); IL-4 inhibitor (such as a small molecule IL-4 antagonist, an anti-IL-4 antibody or an anti-IL-4 receptor antibody); IL-6 inhibitor (such as a small molecule IL-6 antagonist, an anti-IL-6 antibody or an anti-IL-6 receptor antibody); aspirin; NSAIDs; steroids (e.g., prednisone, methotrexate, etc.); low dose cyclosporine A; tumor necrosis factor (TNF) or TNF receptor inhibitors (e.g., a small molecule TNF or TNFR antagonist or an anti-TNF or TNFR antibody); uric acid synthesis inhibitors (e.g., allopurinol); uric acid excretion promoters (e.g., probenecid, sulfinpyrazone, benzbromarone, etc.); other inflammatory inhibitors (e g, inhibitors of caspase-1, p38, IKK1/2, CTLA-4Ig, etc.); and/or corticosteroids. The additional therapeutic agent(s) may be administered prior to, concurrent with, or after the administration of the one or more compounds of formula (I) and/or (II) (for purposes of the present disclosure, such administration regimens are considered the administration of one or more compounds of formula (I) and/or (II) “in combination with” a therapeutic agent).
  • The term “cytotoxic agent” as used herein refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells. The term is intended to include radioactive isotopes (e.g., I131, I125, Y90 and Re186), chemotherapeutic agents, and toxins such as enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof.
  • A “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxanes, e.g., paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel (TAXOTERE®; Aventis Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • A “growth inhibitory agent” when used herein refers to a compound or composition which inhibits growth of a cell, especially a cancer cell either in vitro or in vivo. Examples of growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest. Classical M-phase blockers include the vincas (vincristine and vinblastine), TAXOL®, and topo II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin. Those agents that arrest G1 also spill over into S-phase arrest, for example, DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C.
  • The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of an active agent and a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable” means approved by a regulatory agency of the United States Federal or State government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin.
  • In an embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • The active agents of the disclosure can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • The amount of the active agent of the disclosure, which will be effective in the treatment of a medical condition, can be determined by standard clinical techniques based on the present description. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each subject's circumstances. However, suitable dosage ranges for intravenous administration are generally about 0.5 to 20 milligrams of active compound per kilogram body weight. Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • In one embodiment, the disclosure provides a method of treatment of a medical disorder in an individual suffering from the medical disorder comprising administering to the individual an effective amount of a composition comprising a compound of formula (I) and/or (II), and further comprising administering sequentially or consecutively an additional therapy or administering at least one additional therapeutic agent.
  • In another embodiment, the disclosure relates to a method of treating a disease sensitive to treatment with said method, said method comprising parenterally administering to a patient in need thereof a therapeutically effective dose of the composition comprising a compound of formula (I) and/or (II).
  • The present disclosure further includes the use of any of the compositions comprising compounds of formula (I), formula (II), formula (i), formula (ii), or a combination thereof and/or pharmaceutical formulations thereof in the manufacture of a medicament for the treatment, prevention and/or amelioration of a medical disorder.
  • The present disclosure further includes the use of any of the compositions comprising compounds of formula (I), formula (II), formula (i), formula (ii), or a combination thereof and/or pharmaceutical formulations thereof in the manufacture of a medicament for the treatment, prevention and/or amelioration of a tumor.
  • While the embodiments herein have predominantly been described in relation to antiproliferative disorders such as cancer, it is envisioned that the compositions herein can also be useful in the treatment of a medical disorder selected from autoimmune diseases and other immunological diseases and dysfunctions, inflammatory diseases, infectious diseases, neurodegenerative diseases, bone disorders, and cardiovascular diseases. Further, any disorder that can benefit from the targeted delivery of a toxic substance to particular cells, cell types, tissues and/or organs is within the scope of the present invention.
  • Finally, embodiments herein may include compositions comprising mixtures of compounds as represented by formula (I) and formula (II).
  • The references to certain embodiments made in the following description are considered illustrative only of the principles of the disclosure. Further, since numerous modifications and changes will readily be apparent to those skilled in the art, it is not intended to limit the disclosure to the exact construction and process shown as described herein. Accordingly, all suitable modifications and equivalents may be resorted to as falling within the scope of the disclosure and as defined by the claims that follow.
  • The words “comprise”, “comprising”, “include” and “including” when used in this specification and in the following claims are intended to specify the presence of the stated features, integers, components, or steps, but they do not preclude the presence or addition of one or more additional features, integers, components, or steps thereof.
  • General terms used in any of the embodiments herein can be defined as follows; however, the meaning stated should not be interpreted as limiting the scope of the term per se.
  • The term “conjugate” as used herein refers to compound having a Ligand, linker and Biologically Active Molecule. Illustrative examples include compounds of formula (II).
  • The term “spacer” as used herein refers to chemical building blocks of the linker used to spatially separate the Ligand from the Biologically Active Molecule and to allow for catabolism of the linker inside of cells.
  • The term “macrolide” as used herein refers to any Biologically Active Molecule having a macrolide ring.
  • The symbol
    Figure US20160354482A1-20161208-P00001
    denotes the points of attachment.
  • The term “alkyl” as used herein refers to a hydrocarbon radical having a straight or branched chain or combinations thereof. Alkyl radicals can be a univalent, a bivalent or a cyclic radical. Examples of univalent alkyl radicals are methyl, ethyl, 1-propyl, 2-propyl, n-butyl, isobutyl, sec-butyl, t-butyl, pentyl, neopentyl, hexyl, isohexyl, and the like. As a way of illustration examples of bivalent alkyl radicals include
  • Figure US20160354482A1-20161208-C00026
  • Examples of cyclic alkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. Typical alkyl radicals have from one to ten carbon atoms, one to nine carbon atoms, one to eight carbon atoms, one to seven carbon atoms, one to six carbon atoms, one to five carbon atoms, one to four carbon atoms, one to three carbon atoms, one to two carbon atoms or one carbon atom. Typical cycloalkyl are 3 to 7 member monocyclic ring radicals.
  • The phrase “pharmaceutically acceptable salt” as used herein refers to both organic and inorganic salts of the conjugate compounds described herein, e.g., compounds of formula (I), and (II). The salts are pharmaceutically acceptable and include: sulfate, citrate, nitrate, phosphate, ascorbate, bromide, gluconate, benzoate, oxalate, pantothenate, and the like. Note that pharmaceutically acceptable salts herein may include more than one charged atom in its structure as well as one or more counter ion. Preparation of conjugate compounds herein as pharmaceutically acceptable salts is well known to one of skill in the art.
  • The term “ligand” as used herein means any molecule or compound that specifically or selectively interacts with and/or binds to a second molecule or compound. In certain embodiments, a ligand is an antibody or antigen-binding fragment thereof. Other examples of ligands suitable for use in the context of the present invention include, e.g., aptamers, peptides that specifically interact with a particular antigen (e.g., peptibodies), receptor molecules, and antigen-binding scaffolds (e.g., DARPins, HEAT repeat proteins, ARM repeat proteins, tetratricopeptide repeat proteins, and other scaffolds based on naturally occurring repeat proteins, etc., [see, e.g., Boersma and Pluckthun, 2011, Curr. Opin. Biotechnol. 22:849-857, and references cited therein]).
  • Antibodies exist as intact immunoglobulins, or as a number of well-characterized fragments produced by digestion with various peptidases. For example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′2, a dimer of Fab which itself is a light chain joined to VH-CH by a disulfide bond. The F(ab)′2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)′2 dimer into an Fab′ monomer. The Fab′ monomer is essentially Fab with part of the hinge region. While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv (scFv) single variable domains (Dabs)) or those identified using display libraries such as phage, E. coli or yeast display libraries (see, for example, McCafferty et al. (1990) Nature 348:552-554).
  • Methods for preparing antibodies are known to the art. See, for example, Kohler & Milstein (1975) Nature 256:495-497; Harlow & Lane (1988) Antibodies: a Laboratory Manual, Cold Spring Harbor Lab., Cold Spring Harbor, N.Y.). Antibodies that are isolated from organisms other than humans, such as mice, rats, rabbits, cows, can be made more human-like through chimerization or humanization.
  • The term “human antibody” as used herein is intended to include antibodies having variable and constant regions derived from human immunoglobulin sequences. The human mAbs of the invention may include amino acid residues not encoded by human immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term “human antibody”, as used herein, is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species have been grafted onto human FR sequences.
  • The term “therapeutically effective amount” as used herein refers to an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • The term “racemate” as used herein means an equimolar mixture of a pair of enantiomers. The term racemate also refers to a racemic mixture.
  • The term “enantiomer” as used herein refers to compounds which non-superimposable with the mirror images of each other. Enantiomers may exist in either the (R) or (S) configuration.
  • The term “stereoselective synthesis” refers to a chemical reaction that leads to formation of a single stereoisomer or an enantiomer-enriched mixture of isomers from among two or more possible stereoisomers.
  • The term “diastereomeric excess” refers to the difference between the mole fraction of the desired single diastereomer as compared to the remaining diastereomers in a composition. Diastereomeric excess is calculated as follows:

  • (amount of single diastereomer)−(amount of other diastereomers)/1
      • For example, a composition that contains 90% of 1 and 10% of 2, 3, 4, or a mixture thereof has a diastereomeric excess of 80% [(90−10)/1]. A composition that contains 95% of 1 and 5% of 2, 3, 4, or a mixture thereof has a diastereomeric excess of 90% [(95−5)/1]. A composition that contains 99% of 1 and 1% of 2, 3, 4, or a mixture thereof has a diastereomeric excess of 98% [(99−1)/1]. The diastereomeric excess can similarly be calculated for any one of 1, 2, 3, or 4.
  • The term “stereomerically pure” as used herein refers to a compound wherein the indicated stereoisomer is present to a greater extent than other stereoisomers of that compound, e.g., the compound is present in diastereomeric excess. In some embodiments, the stereomerically pure compounds described herein comprise 80% or greater, 85% or greater, 90% or greater, 95% or greater, or 97% or greater by weight of one stereoisomer of the compound.
  • Conjugates in accordance with various embodiments described herein can be prepared by any known method in the art. An illustrative protocol for producing conjugates is provided in the Examples below.
  • In one embodiment, the conjugates can be prepared by i) reacting a Ligand with drug molecules of formula (I) to form a conjugate of formula (II), and ii) purifying the conjugate.
  • In an alternative embodiment, the conjugates are prepared by reacting a Ligand, linker and biologically active macrolide in a single reaction. Once the conjugates in accordance with the invention are prepared they can be purified.
  • In one embodiment, the compositions comprising drug molecules of formula (I) and/or compositions comprising conjugate compounds of formula (II) described herein can be evaluated for their ability to suppress proliferation of various cancer cell lines in vitro. For example, compositions comprising drug molecules or formula (I) and/or compositions comprising conjugate compounds of formula (II) can be applied to in vitro plated cancer cells for a predetermined number of days and surviving cells measured in assays by known methods.
  • The above specification, examples and data provide a complete description of the manufacture and use of the composition of the invention. Since many embodiments of the invention can be made without departing from the spirit and scope of the invention, the invention resides in the claims hereinafter appended.
  • All references cited herein and in the Examples that follow, including patents, published patent applications, and literature references, are expressly incorporated by reference in their entireties.
  • The description and Examples presented infra are provided to illustrate the subject invention. One of skill in the art will recognize these Examples are provided by way of illustration only and are not included for the purpose of limiting the invention.
  • Embodiments disclosed herein are illustrated in greater detail by means of the non-limiting examples described below.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 illustrates an 1H-NMR spectrum of Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxysuccinate (5). The 1H-NMR spectrum is consistent with a single diastereomer present in at least 95% diastereomeric excess since the spectrum is not complicated by resonances attributable to the other diastereomer. (For comparative purposes Example 2 sets forth the 1H-NMR spectrum of the mixture of diastereomers).
  • FIG. 2 illustrates an 1H-NMR spectrum of mixture of diastereomers of Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxysuccinate (6).
  • FIG. 3 illustrates that in SKBR3 cells the single diastereomer compound conjugate HER2-5 (in vitro and in vivo lots) possessed an IC50 value of 0.3 nM versus 0.9 nM for the mixture of diastereomer conjugate HER2-6.
  • FIG. 4 illustrates that in BT474 cells the single diastereomer compound conjugate HER2-5 (in vitro) in possessed an IC50 value of 4.6 nM while the HER2-5 (in vivo) lot had an IC50 value of 4.0 nM versus 11.6 nM for the mixture of diastereomer conjugate HER2-6.
  • FIG. 5 illustrates that in NCI-N87 cells the single diastereomer compound conjugate HER2-5 (in vitro) possessed an IC50 value of 0.6 nM while the HER2-5 (in vivo) lot had an IC50 value of 0.4 nM versus 1.0 nM for the mixture of diastereomer conjugate HER2-6.
  • FIG. 6 illustrates that in HEK293/hEGFRvIII cells the single diastereomer compound conjugate EGFRvIII-5 possessed an IC50 value of 0.4 nM while the mixture of diastereomer conjugate EGFRvIII-6 had an IC50 value of 0.5 nM.
  • FIG. 7 illustrates that in MMT/hEGFRvIII cells the single diastereomer compound conjugate EGFRvIII-5 possessed an IC50 value of 0.5 nM while the mixture of diastereomer conjugate EGFRvIII-6 had an IC50 value almost 20 fold higher at 9.8 nM.
  • FIG. 8 illustrates that in U251/hEGFRvIII cells the single diastereomer compound conjugate EGFRvIII-5 possessed an IC50 value of 2.4 nM while the mixture of diastereomer conjugate EGFRvIII-6 had an IC50 value of 3.3 nM.
  • FIG. 9 illustrates that in Ovcar-3 cells the single diastereomer compound conjugate MUC16-5 possessed an IC50 value of 6.3 nM while the mixture of diastereomer conjugate MUC16-6 had an IC50 value of 16.0 nM.
  • FIG. 10 illustrates that in PC3/MUC16long cells the single diastereomer compound conjugate MUC16-5 in possessed an IC50 value of 0.34 nM while the mixture of diastereomer conjugate MUC16-6 had an IC50 value at 0.80 nM.
  • FIG. 11 illustrates tumor growth curves in mice following dosing with HER2-5 and control reagents. Mice received PBS vehicle (▪), 300 ug/kg DM1-SMe () and Isotype Control mAb at 15 mg/kg (▾). Mice also received HER2 mAb (▴), HER2-5 (Δ) and Isotype Control-5 (∇) at doses of 1 mg/kg (A), 5 mg/kg (B) and 15 mg/kg (C). Mice received 3 once weekly doses of conjugates and control agents as indicated by the black arrows (T. Groups are N=8, Mean±SE.).
  • FIG. 12 illustrates change in tumor volume following dosing with HER2-5 and control reagents at termination of the PBS vehicle control group on Day 79 post tumor implantation. Individual tumor sizes are shown for each dosing group. Mice received PBS vehicle (▪), 300 ug/kg DM1-SMe () and Isotype Control mAb at 15 mg/kg (▾). Mice also received HER2 mAb (▴), HER2-5 (Δ) and Isotype Control-5 (∇) at doses of 1 mg/kg (A), 5 mg/kg (B) and 15 mg/kg (C). Groups are N=8, Mean±SD.
  • FIG. 13 illustrates percentage change in animal weights following dosing with HER2-5 and control reagents. Mice received PBS vehicle (▪), 300 ug/kg DM1-SMe () and Isotype Control mAb at 15 mg/kg (▾). Mice also received HER2 mAb (▴), HER2-5 (Δ) and Isotype Control-5 (∇) at doses of 1 mg/kg (A), 5 mg/kg (B) and 15 mg/kg (C). Mice received 3 once weekly doses of conjugates and control agents as indicated by the black arrows (T. Groups are N=8, Mean±SE.).
  • EXAMPLES Experimental Details
  • Proton NMR spectra (for compounds that could not be detected by UV) were acquired on a Varian Inova 300 MHz instrument, while mass spectra were collected on an Agilent 1100 series LC/MSD with electrospray ionization source and quadrupole ion trap analyzer. All conjugates were analyzed using a Bruker ultraFleXtreme MALDI-TOF-TOF mass spectrometer. All starting materials and solvents were purchased commercially and used without purification, unless otherwise noted.
  • Example 1 Synthesis of Maleimidylmethyl-4-trans-cyclohexylcarboxy-succinate (3): Maleimidylmethyl-4-trans-cyclohexanecarboxylic acid (2)
  • The title compound was prepared in two steps (Step A and Step B) using modified versions of the methods described by Marnett et al. (J. Med. Chem., 1996, 39, 1692-1670).
  • Step A:
  • A solution of trans-4-aminomethylcyclohexane carboxylic acid (7.00 g, 44.5 mmol) in 1,4-dioxane (70 mL) was treated with maleic anhydride (4.89 g, 49.9 mmol) and stirred at ambient temperature for 48 h. The reaction was evaporated in vacuo to a white solid that can be stored or carried on to the next step without further purification. 1H NMR (300 MHz, DMSO-d6) δ 9.11 (m, 1H), 6.44 (d, 1H, J=13 Hz), 6.24 (d, 1H, J=13 Hz), 3.05 (t, 2H, J=6 Hz), 2.13 (tt, 1H, J=12 Hz, 4 Hz), 1.90 (m, 2H), 1.75 (m, 2H), 1.44 (m, 1H), 1.28 (m, 2H), 0.96 (m, 2H).
  • Step B:
  • The maleamic acid from Step A (36.8 g, 144 mmol) and sodium acetate (13.6 g, 165 mmol) were dissolved in acetic anhydride (368 mL), sealed in a glass reaction vessel, and heated to 120° C. for 3 hours. The cooled reaction mixture (a black syrup) was poured onto water (3 L), stirred, and extracted with dichloromethane. The organic layer was dried over Na2SO4, filtered over a sintered glass funnel, and the clear filtrate evaporated and dried under high vacuum giving the title compound as a yellow solid (7.00 g, 20%). 1H NMR (300 MHz, CDCl3) δ 6.73 (s, 2H), 3.40 (d, 2H, J=7 Hz), 2.28 (m, 1H), 2.06 (m, 2H), 1.75 (m, 3H), 1.42 (m, 2H), 1.03 (m, 2H).
  • Maleimidylmethyl-4-trans-cyclohexanecarboxysuccinate (3)
  • The product of the preceding step B (10.0 g, 42.1 mmol) was dissolved in dichloromethane (50 mL) under Ar, treated with N-hydroxysuccinimide (7.27 g, 63.2 mmol) and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDAC, 12.4 g, 64.5 mmol), and the reaction was stirred at ambient temperature overnight. The resulting brown solution was diluted with dichloromethane, washed with water and brine, dried over Na2SO4, filtered over a sintered glass funnel, and the filtrate concentrated and dried in vacuo. This product was then dissolved in hot ethyl acetate, treated with activated charcoal (1.5 g), filtered, and the filtrate cooled. Filtration of the crystalline product, washing with cold ethyl acetate, and suction drying then gave the title compound as a tan solid (5.52 g, 39%). 1H NMR (300 MHz, CDCl3) δ 6.72 (s, 2H), 3.42 (m, 2H), 2.85 (br s, 4H), 2.56 (tt, 1H, J=12 Hz, 4 Hz), 2.18 (m, 2H), 1.80 (m, 2H), 1.70 (m, 1H), 1.56 (m, 2H), 1.09 (m, 2H).
  • Figure US20160354482A1-20161208-C00027
  • Example 2 Maytansin-3-N-methyl-L-alanine-propanamide-3-thiol (4)
  • The title compound, known in the art as DM1, was prepared using a modified version of the method described by Whitesides et al. (J. Org. Chem., 1991, 56, 2648-2650). Maytansin-3-N-methyl-L-Ala-(3-methyldisulfanyl) propanamide (DM1-SMe, 2.42 g, 3.09 mmol, prepared in a manner similar to Ho and Carrozzella, U.S. Pat. Appl. 2007/0037972 A1) was dissolved in acetonitrile (30 mL), treated with a solution of tris(2-carboxyethyl)phosphine hydrochloride (8.23 g, 28.7 mmol) in water (30 mL), the pH was raised to ca. 3 with the addition of saturated aqueous NaHCO3 (5 mL), the flask was purged with Ar, and the reaction was stirred at ambient temperature under a rubber septum (vented due to effervescence). After 2 h, the reaction was treated with brine (ca. 100 mL), bubbled with Ar for 5 minutes (to remove the free methylmercaptan), and the phases separated. The aqueous phase was extracted twice with ethyl acetate (EtOAc), saturated with NaCl, and extracted twice more with EtOAc. The combined organic layers were then dried over Na2SO4, filtered, and the filtrate concentrated and dried in vacuo to give the title compound as a white solid (2.24 g, 98%). 1H NMR (300 MHz, CDCl3/CD3OD) δ 6.76 (d, 1H, J=1.5 Hz), 6.63 (d, 1H, J=11 Hz), 6.59 (d, 1H, J=1.5 Hz), 6.35 (m, 2H), 5.59 (dd, 1H, J=15 Hz, 9 Hz), 5.36 (q, 1H, J=6.5 Hz), 4.68 (dd, 1H, J=12 Hz, 3 Hz), 4.21 (t, 1H, J=10 Hz), 3.92 (s, 3H), 3.60 (d, 1H, J=13 Hz), 3.42 (d, 1H, J=9 Hz), 3.29 (s, 3H), 3.14 (s, 3H), 3.05 (d, 1H, J=13 Hz), 2.95 (d, 1H, J=10 Hz), 2.77 (s, 3H), 2.75-2.47 (m, 6H), 2.11 (dd, 1H, J=14 Hz, 3 Hz), 1.58 (s, 3H), 1.47 (d, 1H, J=14 Hz), 1.40 (m, 1H), 1.22 (m, 6H), 0.73 (s, 3H). MS (ESI, pos.): calc'd for C35H48ClN3O10S, 737.3. found 738.3 (M+H), 720.3 (M−H2O+H).
  • Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxysuccinate (5)
  • The following procedure describes a new method not known in the art. The product of the preceding step (4, 2.23 g, 3.02 mmol) and Maleimidylmethyl-4-cyclohexanecarboxysuccinate (3, 1.50 g, 4.49 mmol) were dissolved in 4:1 acetonitrile/water (75 mL), treated with fine silica gel scraped from a preparative TLC plate (11.2 g), the flask purged with Ar, and the mixture stirred at ambient temperature under rubber septum. After 18 hours, more 3 (0.77 g, 2.3 mmol) and acetonitrile (MeCN, 10 mL) were added, and the reaction stirred an additional 6 hours. The mixture was filtered over Celite, solids washed with MeCN and ethyl acetate (EtOAc), and the filtrate concentrated in vacuo to a gold solid, which was purified by flash column chromatography on an 120 g silica gel cartridge (50-100% 1:1 EtOAc/MeCN in dichloromethane over 33 min, 75 mL/min) Evaporation and drying of the pure column fractions in vacuo gave the title compound as a cream-colored solid (2.09 g). Concentration of the impure fractions and repurification on an 80 g silica gel cartridge as above gave an additional batch of cream-colored solid (0.22 g), and brought the total yield of title compound to 2.31 g (71%). 1H NMR (300 MHz, CDCl3) δ 6.85 (d, 1H, J=4 Hz), 6.72 (m, 1H), 6.65 (d, 1H, J=4 Hz), 6.44 (dd, 1H, J=15 Hz, 11 Hz), 6.25 (s, 1H), 5.67 (dd, 1H, J=16 Hz, 9 Hz), 5.41 (m, 1H), 4.79 (d, 1H, J=11 Hz), 4.30 (t, 1H, J=11 Hz), 3.72 (m, 2H), 3.51 (d, 1H, J=9 Hz), 3.37 (m, 4H), 3.27 (m, 1H), 3.23 (s, 3H), 3.16-2.99 (m, 4H), 2.85 (m, 7H), 2.62 (m, 3H), 2.39 (ddd, 1H, J=19 Hz, 12 Hz, 4 Hz), 2.18 (br m, 2H), 1.77 (br m, 3H), 1.66 (s, 3H), 1.60-1.47 (m, 4H), 1.31 (m, 6H), 1.05 (m, 2H), 0.82 (s, 3H). MS (ESI, pos.): calc'd for CmH66ClN5O16S, 1071.4. found 1072.4 (M+H), 1054.9 (M−H2O+H); [α]20 589mm=−52.4 (c=0.00301, MeOH). See FIG. 1 for 1H-NMR of single diastereomer.
  • Figure US20160354482A1-20161208-C00028
  • Example 3 Mixture of diastereomers of Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxysuccinate (6)
  • A sample of the mixed stereoisomers of 5 was synthesized according to US Patent Application 20100129314, Example XI. 1H NMR (300 MHz, CDCl3) δ 6.85-6.6 (m), 6.4 (m), 6.1 (m), 5.8-5.4 (m), 5.2 (m), 4.92-4.79 (m), 4.4-4.1 (m), 4.03 (s), 3.82 (m), 3.8-2.2 (m), 2.1 (m), 2.07 (s), 2.0-0.8 (m). MS (ESI, pos.): calc'd for C51H66ClN5O16S, 1071.4. found 1072.4 (M+H). See FIG. 2 for 1H-NMR of mixture of diastereomers.
  • Example 4 Racemic Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxylic acid (8)
  • A solution of trans-1,4-(maleimidomethyl) cyclohexane-1-carboxylic acid 7 (167 mg, 0.701 mmol) in 1, 2-dimethoxyethane (8 mL) was added to a solution of 4 (340 mg, 0.461 mmol) in 1, 2-dimethoxyethane (15 mL). The mixture was then treated with pH 7.5 buffer (20 mL) and a few drops of saturated aqueous NaHCO3 to maintain the pH. The reaction mixture was stirred overnight at room temperature under argon and then concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography using a C18 column, 20-40 micron column (100 g), eluting with a gradient (10 95% over 25 mins) of acetonitrile (0.1% AcOH) in water (0.1% AcOH), and lyophilized to give 8 (330 mg, 0.338 mmol, 73% yield) as a white solid. MS m/z: 977.2 [MH+], 957.2 [M−18], 999.2 [M+Na]; Purity: >98% (by LC/MS).
  • Figure US20160354482A1-20161208-C00029
  • Example 5 Chiral Separation of 8 to Single Diastereomers (R*)-Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxylic acid (9) and (S*)-Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxylic acid (10) (S* and R* Represent a Single Stereoisomer of Unknown Chirality)
  • The diastereomeric mixture of compounds 8 (20 mg) was dissolved in 0.5 ml of acetonitrile and separated using semi-prep Chiral column (Chirlacel OJ, Solvent system, 6:1:1 Hexanes:IPA:Ethanol) to afford 3.5 mg of 10 as the faster-running compound, MS m/z: 977.2 [MH+], 957.2 [M−18], 999.2 [M+Na]; Purity: >95% (by LC/MS), RT=32 min and 4.6 mg of 9 as the slower-running compound, MS m/z: 977.2 [MH+], 957.2 [M−18], 999.2 [M+Na]; Purity: >95% (by LC/MS), Rf=48 min
  • Figure US20160354482A1-20161208-C00030
  • Example 6 Synthesis of (S*)-Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxylic acid (11)
  • A solution of 10 (2.5 mg, 0.0026 mmol) was dissolved in dichloromethane (1 mL), then treated with N-hydroxysuccinimide (NHS, 6.0 mg, 0.052 mmol) and 1-[3-(dimethylamino) propyl]-3-ethylcarbodiimide hydrochloride (EDCI, 13 mg, 0.065 mmol). The reaction mixture was stirred overnight at room temperature under argon, washed with water followed by brine, dried over anhydrous sodium sulfate, and filtered. The solvent was evaporated under reduced pressure to give the crude residue, which was purified by reverse phase chromatography using a C18, 20-40 micron column (30 g), eluting with a gradient (10-95% over 18 min) of acetonitrile (0.1% AcOH) in water (0.1% AcOH), and lyophilized to afford 11. MS m/z: 1073.2 [MH+], 1054.4 [M−18]; Purity: 95% (by LC/MS).
  • Figure US20160354482A1-20161208-C00031
  • Example 7 (R*)-Maytansin-3-N-methyl-L-alanine-propanamidyl-3-thio-3-succinimidyl-N-methylcyclohexyl-4-trans-carboxylic acid (12)
  • A solution of 9 (3, mg, 0.003 mmol) was dissolved in dichloromethane (1 mL), then treated with N-hydroxysuccinimide (NHS, 3.0 mg, 0.026 mmol) and 1-[3-(dimethylamino) propyl]-3-ethylcarbodiimide hydrochloride (EDCI, 7 mg, 0.036 mmol). The reaction mixture was stirred overnight at room temperature under argon, washed with water followed by brine, dried over anhydrous sodium sulfate, and filtered. The solvent was evaporated under reduced pressure to give the crude residue, which was purified by reverse phase chromatography using C18 column, 20-40 micron (15 g), eluting with a gradient (10-95% over 18 min) of acetonitrile (0.1% AcOH) in water (0.1% AcOH), lyophilized to afford 12. MS m/z: 1073.2 [MH+], 1054.4 [M−18]; Purity: 95% (by LC/MS).
  • Figure US20160354482A1-20161208-C00032
  • Example 8 Conjugate Preparation and Characterization
  • Two different maytansine-linker compositions prepared according to the previous Examples (Compound 5 and Compound 6) were conjugated to various anti-tumor antigen monoclonal antibodies. Compound 5 comprises predominantly a single diastereomer of the linker-DM1 cytotoxic compound, whereas Compound 6 comprises a mixture of various linker-DM1 diastereomers. The antibodies used in this Example were an anti-HER2 antibody having variable regions derived from humAb4D5-8 from Carter et al, PNAS 1992 89 4285, an anti-EGFRvIII antibody having variable regions derived from clone 131 from WO2013075048 A1, and an anti-MUC16 antibody having variable regions derived from clone 3A5 from WO2007001851.
  • The antibodies were expressed in CHO cells and purified by Protein A. A non-binding isotype control antibody derived from an infectious disease antigen having no relation to oncology was also used in this Example.
  • The antibodies (10 mg/ml) in 50 mM HEPES, 150 mM NaCl, pH 8.0, and 10% (v/v) DMA were conjugated with a 6 fold excess of compound 5 or 6 for 1 hour at ambient temperature. The conjugates were purified by size exclusion chromatography and sterile filtered. Protein and linker payload concentrations were determined by UV spectral analysis and MALDI-TOF mass spectrometry. Size-exclusion HPLC established that all conjugates used were >95% monomeric, and RP-HPLC established that there was <0.5% unconjugated linker payload. Yields are reported in Table 1 based on protein. All conjugated antibodies were analyzed by UV for linker payload loading values according to Hamblett et al, Cancer Res 2004 10 7063 and by mass difference, native versus conjugated. The results are summarized in Table 1.
  • TABLE 1
    ε252 nm (cm−1 M−1) ε280 nm (cm−1 M−1)
    Compound
    5 26790 5700
    6* 26790 5700
    Antibody
    HER2 81847 215388
    EGFRvIII 79579 209420
    MUC16 88671 248380
    Isotype Control 81718 233480
    Antibody Payload:Antibody Payload:Antibody
    Conjugate (UV) (MS) Yield %
    HER2-5 (in vivo) 2.7 2.7 66
    HER2-5 (in vitro) 3.1 2.4 75
    HER2-6 (in vitro) 2.9 2.4 70
    EGFRvIII-5 2.8 2.3 56
    EGFRvIII-6 2.9 2.2 56
    MUC16-5 2.4 2.0 76
    MUC16-6 2.3 2.1 96
    Isotype Control-5 3.3 3.3 67
    *Extinction coefficients were used from compound 5
  • Example 9 In Vitro Cytotoxicity Assays
  • Cells were seeded in PDL-coated 96 well plates at 10,000 (SK-BR-3 and NCI-N87), 15,000 (BT-474), 3000 (Ovcar-3 and PC3/Muc16), 2000 (HEK293/hEGFRvIII), 1500 (U251/hEGFRvIII), or 400 (MMT/hEGFRvIII) cells per well in complete growth media and grown overnight. For cell viability curves, serially diluted antibody-drug conjugates or free payload were added to the cells at final concentrations ranging from 1 μM to 1 pM and incubated for 3 days. Each concentration was run in duplicate and reported with the respective standard deviation. Cells were incubated with CCK8 (Dojindo) for the final 1-3 h and the absorbance at 450 nm (0D450) was determined on a Flexstation3 (Molecular Devices). Background OD450 levels from digitonin (40 nM) treated cells were subtracted from all wells and viability is expressed as a percentage of the untreated controls. IC50 values were determined from a four-parameter logistic equation over a 10-point response curve (GraphPad Prism). All curves and IC50 values are corrected for payload equivalents based on the loading from the MALDI-TOF experiment.
  • In SKBR3 cells (breast cancer line), natively expressing HER2 at 607 fold above isotype control binding, the single diastereomer compound conjugate HER2-5 (in vitro and in vivo lots) possessed an IC50 value of 0.3 nM versus 0.9 nM for the mixture of diastereomer compound conjugate HER2-6 (Table 2, FIG. 3). A small in vitro lot was conjugated first and only used for cell proliferation assays, while a larger in vivo lot was then conjugated and used for both in vitro and in vivo experiments. The naked HER2 antibody had little anti-proliferation activity.
  • In BT474 cells (breast cancer line), natively expressing HER2 at 426 fold above isotype control binding, the single diastereomer compound conjugate HER2-5 (in vitro) possessed an IC50 value of 4.6 nM while the HER2-5 (in vivo) lot had an IC50 value of 4.0 nM versus 11.6 nM for the mixture of diastereomer compound conjugate HER2-6 (Table 2, FIG. 4). The naked HER2 antibody had little anti-proliferation activity.
  • In NCI-N87 cells (breast cancer line), natively expressing HER2 at 869 fold above isotype control binding, the single diastereomer compound conjugate HER2-5 (in vitro) possessed an IC50 value of 0.6 nM while the HER2-5 (in vivo) lot had an IC50 value of 0.4 nM versus 1.0 nM for the mixture of diasteromer compound conjugate HER2-6 (Table 2, FIG. 5). The naked HER2 antibody had little anti-proliferation activity.
  • In HEK293/hEGFRvIII cells, expressing hEGFRvIII at 360 fold above isotype control binding, both conjugates (single and mixture of diastereomer) exhibited IC50 values of 0.4 nM (Table 3, FIG. 6). The naked EGFRvIII antibody had little anti-proliferation activity.
  • In MMT/hEGFRvIII cells, expressing hEGFRvIII at 280 fold above isotype control binding, the single diastereomer compound conjugate EGFRvIII-5 possessed an IC50 value of 0.5 nM while the mixture of diastereomer compound conjugate EGFRvIII-6 had an IC50 value of 9.8 nM (Table 2, FIG. 7). The naked EGFRvIII antibody had little anti-proliferation activity.
  • In U251/hEGFRvIII cells, expressing hEGFRvIII at 165 fold above isotype control binding, the single diastereomer compound conjugate EGFRvIII-5 possessed an IC50 value of 2.4 nM while the mixture of diastereomer compound conjugate EGFRvIII-6 had an IC50 value of 3.3 nM (Table 3, FIG. 7). The naked EGFRvIII antibody had little anti-proliferation activity.
  • In Ovcar-3 cells (ovarian cancer line), natively expressing MUC16 at 373 fold above isotype control binding, the single diastereomer compound conjugate MUC16-5 possessed an IC50 value of 6.3 nM while the mixture of diasteroemer compound conjugate MUC16-6 had an IC50 value of 16.0 nM (Table 4, FIG. 8). The naked Muc16 antibody had little anti-proliferation activity.
  • In PC3/MUC16 cells, expressing MUC16 at 105 fold above isotype control binding, the single diastereomer compound conjugate MUC16-5 possessed an IC50 value of 0.34 nM while the mixture of diastereomer compound conjugate MUC16-6 had an IC50 value of 0.8 nM (Table 4, FIG. 9). The naked Muc16 antibody had little anti-proliferation activity.
  • In FIGS. 3, 4, and 5 maytansin-3-N-methyl-L-Ala-(3-methyldisulfanyl) propanamide (DM1-SMe, prepared according to Ho and Carrozzella, U.S. Pat. Appl. 2007/0037972 A1) was chosen to represent the payload in these assays. Compound 4 would be too reactive to use in vitro or in vivo, thereby giving unreliable results.
  • The in vitro results are summarized in Tables 2-4 on a target basis below. This Example demonstrates that anti-tumor antibody-drug conjugates comprising the single diastereomer drug of the present invention, in most cases, exhibited greater in vitro killing potency than the corresponding antibody-drug conjugates comprising mixture of diastereomers. For the targets analyzed, the single diastereomer antibody-drug conjugates were typically on the order of 2- to 3-fold more potent than the corresponding mixture conjugates, depending on the particular cell lines tested.
  • TABLE 2
    Antibody BT474 IC50
    Conjugate SKBR3 IC50 (nM) (nM) N87 IC50 (nM)
    HER2-5 (in vivo) 0.3 4.0 0.4
    HER2-5 (in vitro) 0.3 4.6 0.6
    HER2-6 (in vitro) 0.9 11.6 1.0
  • TABLE 3
    Antibody HEK293/hEGFRvIII MMT/hEGFRvIII U251/hEGFRvIII
    Conjugate IC50 (nM) IC50 (nM) IC50 (nM)
    EGFRvIII-5 0.4 0.5 2.4
    EGFRvIII-6 0.5 9.8 3.3
  • TABLE 4
    Antibody Ovcar-3 IC50 PC3/MUC16
    Conjugate (nM) IC50 (nM)
    MUC16-5 6.3 0.34
    MUC16-6 16.0 0.80
  • Example 10 In Vivo Studies
  • To determine the in vivo efficacy of the anti-HER2 single-diastereomer conjugate (“HER2-5”), studies were performed in mice bearing HER2+ gastric cancer xenografts, as efficacy had been previously reported in this model by Barok et al (Barok M et al, Can Letters 2011). Specifically, 5×106 NCI-N87 cells (ATCC CRL-5822) were implanted subcutaneously into the lower right flank of CB-17 SCID mice (Taconic). Once tumors had reached an average volume of 150 mm3, mice were randomized in to groups of eight and dosed with HER2-5 or control reagents. Control reagents included PBS vehicle, free DM1-SMe, isotype control, isotype control-5, or HER2. Mice received once weekly doses for three weeks and tumor volumes and body weights were monitored twice weekly throughout the study. Conjugates were dosed at 1, 5 and 15 mg/kg, as these doses had been shown to be effective in previous in vivo studies by Lewis-Phillips et al (Lewis-Phillips G et al., Can Res 2008).
  • In the current N87 tumor model, HER2-5 demonstrated clear anti-tumor efficacy, with doses of 5 and 15 mg/kg leading to significant decreases in initial tumor volume and eradication of some tumors at the higher dose (FIGS. 11 and 12). A significant delay in tumor growth relative to control agents was also observed in the 1 mg/kg dose level. No adverse events were observed following dosing, with mice receiving HER2-5 demonstrating robust weight gain throughout the study (FIG. 13).

Claims (30)

What is claimed is:
1. A composition comprising a plurality of drug molecules of formula I:
Figure US20160354482A1-20161208-C00033
wherein:
X is
Figure US20160354482A1-20161208-C00034
Figure US20160354482A1-20161208-C00035
Figure US20160354482A1-20161208-C00036
Figure US20160354482A1-20161208-C00037
Y is Y1 or Y2 further wherein
Y1 is
Figure US20160354482A1-20161208-C00038
or H;
Y2 is —Cl, —Br, —I, or
Figure US20160354482A1-20161208-C00039
Z is H or SO3H;
R1 and R2 are independently selected from H or alkyl;
n is an integer from 0 to 50;
and wherein the drug molecules present in the composition comprises a mixture of at least two diastereomers, a first diastereomer and a second diastereomer, further wherein said first diastereomer and second diastereomer are otherwise identical, except that said first and second diastereomers have different stereochemical configuration at a chiral carbon represented by (*) in formula X, wherein said chiral carbon atom is a carbon atom that is bound to a sulfur atom, and said first or second diastereomer is present in a diastereomeric excess of greater than 50%.
2. The composition of claim 1, wherein n is 1, and R1 and R2 are each independently hydrogen.
3. The composition of claim 1, wherein the drug molecules are present in the composition in a diastereomeric excess of at least 95%.
4. The composition of claim 1, wherein formula I is represented by:
Figure US20160354482A1-20161208-C00040
or mixtures thereof in a diastereomeric excess of greater than 50%.
5. A composition comprising a plurality of ligand-drug conjugates of Formula II:
Figure US20160354482A1-20161208-C00041
wherein:
A is
Figure US20160354482A1-20161208-C00042
W is selected from S, O, or NR3;
L is a ligand;
further wherein:
L is capable of binding to a cell or cell population;
R1, R2 and R3 are each independently selected from H or alkyl;
n is an integer from 0 to 10;
p is an integer from 1 to 10;
and wherein the ligand-drug conjugates are present in the composition in a diastereomeric excess of greater than 50%.
6. The composition of claim 5, wherein the ligand is an antibody or an antigen-binding fragment thereof, W is NH, and R1, R2 are each independently selected from H.
7. The composition of claim 6, wherein the antibody or antigen-binding fragment thereof specifically binds a tumor-associated antigen.
8. The composition of claim 7, wherein the ligand-drug conjugates are present in the composition in a diastereomeric excess of more than 95%.
9. The composition of claim 7, wherein the tumor-associated antigen is selected from the group consisting of AFP, ALK, BAGE proteins, β-catenin, brc-abl, BRCA1, BORIS, CA9, carbonic anhydrase IX, caspase-8, CD40, CDK4, CEA, CTLA4, CLEC12A, cyclin-B1, CYP1B1, EGFR, EGFRvIII, ErbB2/Her2, ErbB3, ErbB4, ETV6-AML, EphA2, Fra-1, FOLR1, GAGE proteins (e.g., GAGE-1, -2), GD2, GD3, GloboH, glypican-3, GM3, gp100, Her2, HLA/B-raf, HLA/k-ras, HLA/MAGE-A3, hTERT, LMP2, MAGE proteins (e.g., MAGE-1, -2, -3, -4, -6, and -12), MART-1, mesothelin, ML-IAP, Muc1, Muc16 (CA-125), MUM1, NA17, NY-BR1, NY-BR62, NY-BR85, NY-ESO1, OX40, p15, p53, PAP, PAX3, PAX5, PCTA-1, PLAC1, PRLR, PRAME, PSMA (FOLH1), RAGE proteins, Ras, RGS5, Rho, SART-1, SART-3, Steap-1, Steap-2, survivin, TAG-72, TGF-β, TMPRSS2, Tn, TRP-1, TRP-2, tyrosinase, and uroplakin-3.
10. A method for preparing composition comprising a plurality of drug molecules of formula I:
Figure US20160354482A1-20161208-C00043
wherein:
X is
Figure US20160354482A1-20161208-C00044
Figure US20160354482A1-20161208-C00045
Figure US20160354482A1-20161208-C00046
Figure US20160354482A1-20161208-C00047
Y is Y1 or Y2 further wherein
Y1 is
Figure US20160354482A1-20161208-C00048
or H;
Y2 is —Cl, —Br, —I, or
Figure US20160354482A1-20161208-C00049
R1 and R2 are independently selected from H or alkyl;
n is an integer from 0 to 50;
and wherein the drug molecules present in the composition comprises a mixture of at least two diastereomers, a first diastereomer and a second diastereomer, further wherein said first diastereomer and second diastereomer are otherwise identical, except that said first and second diastereomers have different stereochemical configuration at a chiral carbon represented by (*) in formula X, wherein said chiral carbon atom is a carbon atom that is bound to a sulfur atom, and said first or second diastereomer is present in a diastereomeric excess of greater than 50%,
the method comprising:
(a) providing a mixture comprising
(i) a starting material which has a formula III:
Figure US20160354482A1-20161208-C00050
(ii) a compound of formula IV:
Figure US20160354482A1-20161208-C00051
Figure US20160354482A1-20161208-C00052
Y1 is
Figure US20160354482A1-20161208-C00053
or H;
Y2 is —Cl, —Br, —I, or
Figure US20160354482A1-20161208-C00054
Z is H or SO3H;
R1 and R2 are independently selected from H or alkyl; and
each n is an integer from 0 to 50;
(iii) an organic solvent,
(iv) water, and
(v) a solid substrate;
(b) allowing the mixture of step (a) to react until some of the starting material is converted to the compound of formula I; and
(c) removing crude compound of formula I from the mixture of step (b).
11. The method of claim 10, further comprising (d) purifying the compound of formula I obtained in step (c).
12. The method of claim 10, wherein the solid substrate is selected from the group consisting of silica gel, celite, alumina, a zeolite, and crushed molecular sieves.
13. The method of claim 10, wherein n is 1, and R1 and R2 are each independently hydrogen.
14. The method of claim 10, wherein the organic solvent comprises a polar aprotic solvent.
15. The method of claim 14, wherein the polar aprotic solvent comprises acetonitrile.
16. The method of claim 10, wherein the organic solvent and the water are present in ratio from about 1:1 to about 4:1 or from about 1:1 to about 10:1.
17. The method of claim 10, wherein the molar ratio of the starting material having formula III and the compound of formula IV is from about 1:1 to about 1:3.
18. The method of claim 10, further comprising combining the compound of formula I with an antibody or antigen-binding fragment thereof to make an antibody-drug conjugate.
19. The method of claim 18, wherein the compound of formula I is attached to the antibody or antigen-binding fragment via an S, O, or NR3.
20. The method of claim 10, wherein the formula I is represented by the following structure:
Figure US20160354482A1-20161208-C00055
in a diastereomeric excess of greater than 50%.
21. The composition of claim 1 or 5, wherein the drug molecules or the ligand-drug conjugates are contained within the composition in a therapeutically effective amount, and further comprising a pharmaceutically acceptable diluent, carrier or excipient.
22. The composition of claim 21, further comprising a therapeutically effective amount of a second chemotherapeutic agent.
23. A method of treating a disease sensitive to treatment with said method, said method comprising parenterally administering to a patient in need thereof a therapeutically effective dose of the composition of claim 1 or 5.
24. The method of claim 23, wherein said disease is selected from tumor, autoimmune disease, and inflammatory disease.
25. A composition comprising a plurality of drug molecules of formula I:
Figure US20160354482A1-20161208-C00056
wherein:
X is
Figure US20160354482A1-20161208-C00057
Y is
Figure US20160354482A1-20161208-C00058
or H;
R1 and R2 are each independently selected from H or alkyl;
n is an integer from 0 to 10; and
wherein the drug molecules are present in the composition in an diastereomeric excess of greater than 50% and show greater anti-proliferative activity than a corresponding composition comprising drug molecules of formula I that are not present in a diastereomeric excess of greater than 50%.
26. The composition of claim 1, wherein one of the at least two diastereomers is a compound of formula (i)
Figure US20160354482A1-20161208-C00059
27. The composition of claim 1, wherein one of the at least two diastereomers is a compound of formula (ii)
Figure US20160354482A1-20161208-C00060
28. The composition of claim 1, wherein one of the at least two diastereomers is characterized by a 1H NMR spectra of FIG. 1.
29. A compound of formula (i):
Figure US20160354482A1-20161208-C00061
wherein the compound is stereomerically pure.
30. A compound of formula (ii):
Figure US20160354482A1-20161208-C00062
wherein the compound is stereomerically pure.
US14/913,965 2013-08-26 2014-08-26 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses Abandoned US20160354482A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/913,965 US20160354482A1 (en) 2013-08-26 2014-08-26 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361869954P 2013-08-26 2013-08-26
US201461934313P 2014-01-31 2014-01-31
US14/913,965 US20160354482A1 (en) 2013-08-26 2014-08-26 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
PCT/US2014/052757 WO2015031396A1 (en) 2013-08-26 2014-08-26 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/052757 A-371-Of-International WO2015031396A1 (en) 2013-08-26 2014-08-26 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/189,925 Continuation US11596635B2 (en) 2013-08-26 2018-11-13 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses

Publications (1)

Publication Number Publication Date
US20160354482A1 true US20160354482A1 (en) 2016-12-08

Family

ID=51542449

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/913,965 Abandoned US20160354482A1 (en) 2013-08-26 2014-08-26 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US16/189,925 Active 2037-06-28 US11596635B2 (en) 2013-08-26 2018-11-13 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US18/145,811 Abandoned US20230321109A1 (en) 2013-08-26 2022-12-22 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/189,925 Active 2037-06-28 US11596635B2 (en) 2013-08-26 2018-11-13 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US18/145,811 Abandoned US20230321109A1 (en) 2013-08-26 2022-12-22 Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses

Country Status (17)

Country Link
US (3) US20160354482A1 (en)
EP (1) EP3038624A1 (en)
JP (1) JP6608823B2 (en)
KR (1) KR102252925B1 (en)
CN (1) CN105530942B (en)
AU (1) AU2014311361B2 (en)
BR (1) BR112016004023A2 (en)
CA (1) CA2921412A1 (en)
CL (1) CL2016000408A1 (en)
EA (1) EA038192B1 (en)
HK (1) HK1220115A1 (en)
IL (1) IL244203B (en)
MX (1) MX2016002149A (en)
PH (1) PH12016500380A1 (en)
SG (1) SG11201601230RA (en)
WO (1) WO2015031396A1 (en)
ZA (1) ZA201601534B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018213077A1 (en) 2017-05-18 2018-11-22 Regeneron Pharmaceuticals, Inc. Cyclodextrin protein drug conjugates
WO2019094395A2 (en) 2017-11-07 2019-05-16 Regeneron Pharmaceuticals, Inc. Hydrophilic linkers for antibody drug conjugates
US11142578B2 (en) 2016-11-16 2021-10-12 Regeneron Pharmaceuticals, Inc. Anti-MET antibodies, bispecific antigen binding molecules that bind MET, and methods of use thereof
US11345715B2 (en) 2013-03-15 2022-05-31 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US11446389B2 (en) 2016-01-25 2022-09-20 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US11596635B2 (en) 2013-08-26 2023-03-07 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US11814428B2 (en) 2019-09-19 2023-11-14 Regeneron Pharmaceuticals, Inc. Anti-PTCRA antibody-drug conjugates and uses thereof
US11866502B2 (en) 2020-10-22 2024-01-09 Regeneron Pharmaceuticals, Inc. Anti-FGFR2 antibodies and methods of use thereof
US11896682B2 (en) 2019-09-16 2024-02-13 Regeneron Pharmaceuticals, Inc. Radiolabeled MET binding proteins for immuno-PET imaging and methods of use thereof
US11958910B2 (en) 2020-02-28 2024-04-16 Regeneron Pharmaceuticals, Inc. Bispecific antigen binding molecules that bind HER2, and methods of use thereof

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016160615A1 (en) * 2015-03-27 2016-10-06 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
SG10202112460VA (en) 2015-07-06 2021-12-30 Regeneron Pharma Multispecific antigen-binding molecules and uses thereof
WO2017190079A1 (en) 2016-04-28 2017-11-02 Regeneron Pharmaceuticals, Inc. Methods of making multispecific antigen-binding molecules
DK3515487T3 (en) 2016-09-23 2023-08-28 Regeneron Pharma Bispecific anti-MUC16-CD3 antibodies and anti-MUC16 drug conjugates
IL295509A (en) 2016-09-23 2022-10-01 Regeneron Pharma Anti-steap2 antibodies, antibody-drug conjugates, and bispecific antigen-binding molecules that bind steap2 and cd3, and uses thereof
IL266917B2 (en) 2016-11-29 2023-10-01 Regeneron Pharma Anti-human prolactin receptor (prlr) antibody-drug conjugates and use thereof in combination therapy of prlr positive breast cancer
CA3097711A1 (en) 2018-04-30 2019-11-07 Regeneron Pharmaceuticals, Inc. Antibodies, and bispecific antigen-binding molecules that bind her2 and/or aplp2, conjugates, and uses thereof
MX2020012350A (en) 2018-05-17 2021-01-29 Regeneron Pharma Anti-cd63 antibodies, conjugates, and uses thereof.
EP3927435A1 (en) 2019-02-21 2021-12-29 Regeneron Pharmaceuticals, Inc. Methods of treating ocular cancer using anti-met antibodies and bispecific antigen binding molecules that bind met
JP2023502929A (en) 2019-11-15 2023-01-26 シージェン インコーポレイテッド Methods of treating HER2-positive breast cancer with tucatinib in combination with an anti-HER2 antibody-drug conjugate
MX2022012821A (en) 2020-04-16 2022-11-07 Regeneron Pharma Diels-alder conjugation methods.
AU2021308190A1 (en) 2020-07-13 2023-02-02 Regeneron Pharmaceuticals, Inc. Camptothecin analogs conjugated to a glutamine residue in a protein, and their use
WO2022103724A1 (en) 2020-11-10 2022-05-19 Regeneron Pharmaceuticals, Inc. Selenium antibody conjugates
CN115215780B (en) * 2022-04-22 2023-08-08 上海格苓凯生物科技有限公司 Method for preparing heterobifunctional crosslinking agent SMCC by using N, N-disuccinimidyl carbonate

Family Cites Families (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
JPS5566585A (en) 1978-11-14 1980-05-20 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
CA2026147C (en) 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US20070258987A1 (en) 2000-11-28 2007-11-08 Seattle Genetics, Inc. Recombinant Anti-Cd30 Antibodies and Uses Thereof
EP1258255A1 (en) 2001-05-18 2002-11-20 Boehringer Ingelheim International GmbH Conjugates of an antibody to CD44 and a maytansinoid
US20090068178A1 (en) * 2002-05-08 2009-03-12 Genentech, Inc. Compositions and Methods for the Treatment of Tumor of Hematopoietic Origin
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
EP1638606B1 (en) 2003-06-27 2016-01-06 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
GB0316294D0 (en) 2003-07-11 2003-08-13 Polytherics Ltd Conjugated biological molecules and their preparation
TW200539855A (en) 2004-03-15 2005-12-16 Wyeth Corp Calicheamicin conjugates
JP5234734B2 (en) 2004-06-01 2013-07-10 ジェネンテック, インコーポレイテッド Antibody-drug conjugates and methods
CA2587589A1 (en) 2004-11-29 2006-06-22 Seattle Genetics, Inc. Engineered antibodies and immunoconjugates
MX2007007011A (en) 2004-12-09 2007-09-21 Johnson & Johnson Anti-integrin immunoconjugates, methods and uses.
US7750116B1 (en) 2006-02-18 2010-07-06 Seattle Genetics, Inc. Antibody drug conjugate metabolites
RU2483080C2 (en) 2006-10-27 2013-05-27 Дженентек, Инк. Antibodies and immunoconjugates and their application
WO2008122039A2 (en) 2007-04-02 2008-10-09 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Selenocysteine mediated hybrid antibody molecules
US7723485B2 (en) 2007-05-08 2010-05-25 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
WO2008153744A2 (en) 2007-05-23 2008-12-18 Ventana Medical Systems, Inc. Polymeric carriers for immunohistochemistry and in situ hybridization
SI2254571T1 (en) * 2008-03-18 2015-10-30 Genentech, Inc. Combinations of an anti-her2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
UA108598C2 (en) 2008-04-30 2015-05-25 HIGH-EFFICIENT CONJUGATES AND HYDROPHILIC STAPLING AGENTS (LINERS)
EP2326349B1 (en) 2008-07-21 2015-02-25 Polytherics Limited Novel reagents and method for conjugating biological molecules
EP2403538B1 (en) 2009-03-04 2017-10-04 Polytherics Limited Conjugated proteins and peptides
WO2011018611A1 (en) 2009-08-10 2011-02-17 Ucl Business Plc Reversible covalent linkage of functional molecules
UY32914A (en) 2009-10-02 2011-04-29 Sanofi Aventis ANTIBODIES SPECIFICALLY USED TO THE EPHA2 RECEIVER
UY32913A (en) 2009-10-02 2011-04-29 Sanofi Aventis NEW MAITANSINOIDS AND THE USE OF SUCH MAITANSINOIDS TO PREPARE CONJUGATES WITH AN ANTIBODY
EP2486023A4 (en) * 2009-10-06 2014-05-07 Immunogen Inc Potent conjugates and hydrophilic linkers
WO2011081928A2 (en) 2009-12-14 2011-07-07 The Regents Of The University Of Michigan Compositions and methods for altering cocaine esterase activity
EA024730B1 (en) 2010-04-15 2016-10-31 Медимьюн Лимитед Pyrrolobenzodiazepine compounds, conjugates thereof, pharmaceutical compositions comprising said conjugates, and use of said conjugates
US20130244905A1 (en) 2010-07-06 2013-09-19 Ed Grabczyk Reporter for RNA Polymerase II Termination
EP3960865A1 (en) 2010-08-02 2022-03-02 Regeneron Pharmaceuticals, Inc. Mice that make binding proteins comprising vl domains
PL2611785T3 (en) 2010-09-01 2014-10-31 Bayer Ip Gmbh N-(tetrazol-5-yl)- and n-(triazol-5-yl)aryl carboxylic acid amides and use of same as herbicides
BR112013010569A2 (en) 2010-10-29 2017-07-04 Immunogen Inc non-antagonistic and immunoconjugated egfr binding molecules thereof
CN106349254A (en) 2010-11-03 2017-01-25 伊缪诺金公司 Cytotoxic agents comprising new ansamitocin derivatives
CA2836361C (en) 2011-05-16 2020-11-10 Koninklijke Philips N.V. Bio-orthogonal drug activation
EP3170821B1 (en) 2011-05-27 2021-09-15 Ambrx, Inc. Compositions containing, methods involving, and uses of non-natural amino acid linked dolastatin derivatives
US8815226B2 (en) 2011-06-10 2014-08-26 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
BR112013032928A2 (en) 2011-06-21 2017-01-24 Immunogen Inc "peptide-binding maytansinoid derivatives and conjugates thereof, pharmaceutical composition comprising and use thereof"
CN103987384A (en) 2011-10-14 2014-08-13 西雅图基因公司 Pyrrolobenzodiazepines and targeted conjugates
EA028457B1 (en) 2011-10-14 2017-11-30 Медимьюн Лимитед Pyrrolobenzodiazepines
WO2013053872A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Synthesis method and intermediates useful in the preparation of pyrrolobenzodiazepines
US9387259B2 (en) 2011-10-14 2016-07-12 Seattle Genetics, Inc. Pyrrolobenzodiazepines and targeted conjugates
WO2013068874A1 (en) 2011-11-11 2013-05-16 Pfizer Inc. Antibody-drug conjugates
WO2013085925A1 (en) 2011-12-05 2013-06-13 Igenica, Inc. Antibody-drug conjugates and related compounds, compositions, and methods
GB201210770D0 (en) 2012-06-18 2012-08-01 Polytherics Ltd Novel conjugation reagents
CA2876365A1 (en) 2012-06-19 2013-12-27 Polytherics Limited Novel process for preparation of antibody conjugates and novel antibody conjugates
HUE038285T2 (en) 2012-10-23 2018-10-29 Synaffix Bv Modified antibody, antibody-conjugate and process for the preparation thereof
NO2789793T3 (en) 2012-10-24 2018-01-27
WO2014080251A1 (en) 2012-11-24 2014-05-30 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and their uses for conjugation of drugs to cell binding molecules
TW201425336A (en) * 2012-12-07 2014-07-01 Amgen Inc BCMA antigen binding proteins
CN103333179B (en) 2012-12-21 2017-06-16 百奥泰生物科技(广州)有限公司 Class maytansine derivative and its production and use
CN103254213B (en) 2012-12-21 2015-02-25 百奥泰生物科技(广州)有限公司 Preparation method of maytenin-like ester and composition used in method
JP6847388B2 (en) 2013-03-15 2021-03-31 レゲネロン ファーマシューティカルス,インコーポレーテッド Bioactive molecules, their conjugates, and therapeutic uses
CN103254311B (en) 2013-05-09 2015-05-13 齐鲁制药有限公司 Method for preparing antibody-maytansine alkaloid medicine conjugate
WO2014194030A2 (en) 2013-05-31 2014-12-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2014197854A1 (en) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Novel linkers for antibody-drug conjugates and related compounds, compositions, and methods of use
NZ714765A (en) 2013-06-06 2021-12-24 Pf Medicament Anti-c10orf54 antibodies and uses thereof
WO2014197871A2 (en) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Antibody-drug conjugates, compositions and methods of use
WO2014197866A1 (en) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Modified antibodies and related compounds, compositions, and methods of use
US9545451B2 (en) 2013-08-21 2017-01-17 Regeneron Pharmaceuticals, Inc. Anti-PRLR antibodies and methods for killing PRLR-expressing cells
AU2014311361B2 (en) 2013-08-26 2018-11-29 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US9493413B2 (en) 2013-11-27 2016-11-15 Redwood Bioscience, Inc. Hydrazinyl-pyrrolo compounds and methods for producing a conjugate
CN105793268B (en) 2013-12-02 2019-03-15 香港浸会大学 There are two the cancer resistance Caulis Mayteni chlorins compounds for condensing big ring for tool
CN106573074B (en) 2014-06-02 2022-04-12 里珍纳龙药品有限公司 Bioactive molecule conjugates, reagents and methods of preparation and therapeutic uses thereof
WO2016160615A1 (en) 2015-03-27 2016-10-06 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
JP6934010B2 (en) 2016-01-25 2021-09-08 レゲネロン ファーマシューティカルス,インコーポレーテッド Maytansinoid derivatives, their conjugates, and how to use them

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11345715B2 (en) 2013-03-15 2022-05-31 Regeneron Pharmaceuticals, Inc. Biologically active molecules, conjugates thereof, and therapeutic uses
US11596635B2 (en) 2013-08-26 2023-03-07 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
US11446389B2 (en) 2016-01-25 2022-09-20 Regeneron Pharmaceuticals, Inc. Maytansinoid derivatives, conjugates thereof, and methods of use
US11142578B2 (en) 2016-11-16 2021-10-12 Regeneron Pharmaceuticals, Inc. Anti-MET antibodies, bispecific antigen binding molecules that bind MET, and methods of use thereof
WO2018213077A1 (en) 2017-05-18 2018-11-22 Regeneron Pharmaceuticals, Inc. Cyclodextrin protein drug conjugates
WO2019094395A2 (en) 2017-11-07 2019-05-16 Regeneron Pharmaceuticals, Inc. Hydrophilic linkers for antibody drug conjugates
US11896682B2 (en) 2019-09-16 2024-02-13 Regeneron Pharmaceuticals, Inc. Radiolabeled MET binding proteins for immuno-PET imaging and methods of use thereof
US11814428B2 (en) 2019-09-19 2023-11-14 Regeneron Pharmaceuticals, Inc. Anti-PTCRA antibody-drug conjugates and uses thereof
US11958910B2 (en) 2020-02-28 2024-04-16 Regeneron Pharmaceuticals, Inc. Bispecific antigen binding molecules that bind HER2, and methods of use thereof
US11866502B2 (en) 2020-10-22 2024-01-09 Regeneron Pharmaceuticals, Inc. Anti-FGFR2 antibodies and methods of use thereof

Also Published As

Publication number Publication date
US20190151323A1 (en) 2019-05-23
WO2015031396A1 (en) 2015-03-05
NZ716768A (en) 2021-10-29
CA2921412A1 (en) 2015-03-05
CN105530942A (en) 2016-04-27
KR20160045146A (en) 2016-04-26
NZ756518A (en) 2021-10-29
HK1220115A1 (en) 2017-04-28
US20230321109A1 (en) 2023-10-12
BR112016004023A2 (en) 2022-11-16
EA201600191A1 (en) 2016-07-29
ZA201601534B (en) 2023-11-29
IL244203B (en) 2018-12-31
US11596635B2 (en) 2023-03-07
AU2014311361A1 (en) 2016-03-03
JP2016528304A (en) 2016-09-15
KR102252925B1 (en) 2021-05-18
EA201600191A8 (en) 2018-04-30
EA038192B1 (en) 2021-07-21
JP6608823B2 (en) 2019-11-20
IL244203A0 (en) 2016-04-21
AU2014311361B2 (en) 2018-11-29
PH12016500380A1 (en) 2016-05-16
SG11201601230RA (en) 2016-03-30
EP3038624A1 (en) 2016-07-06
CL2016000408A1 (en) 2017-02-24
CN105530942B (en) 2019-10-11
MX2016002149A (en) 2016-10-28

Similar Documents

Publication Publication Date Title
US20230321109A1 (en) Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
KR102459468B1 (en) Conjugation linkers, cell binding molecule-drug conjugates containing the likers, methods of making and uses such conjugates with the linkers
EP3737419B1 (en) Phosphoalkyl polymers comprising biologically active compounds
KR20200083605A (en) Programmable polymeric drugs
KR20200067132A (en) Programmable polymeric drugs
EP3724200A1 (en) Ionic polymers comprising biologically active compounds
EP3737417A1 (en) Phosphoalkyl ribose polymers comprising biologically active compounds
WO2019140301A1 (en) Polymers with rigid spacing groups comprising biologically active compounds
EP3952918A1 (en) Programmable polymeric drugs
CN113698335A (en) Specific conjugation of cell binding molecules
CN115210227A (en) Heterobifunctional molecules as TEAD inhibitors
JP2023528412A (en) Anti-BCMA antibody-drug conjugates and methods of use
CA2921412C (en) Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
NZ756914A (en) Printing-fluid cartridge, set of printing-fluid cartridges, and system including the printing-fluid cartridge and printing-fluid consuming apparatus
NZ757070B2 (en) Printing-fluid cartridge, set of printing-fluid cartridges, and system including the printing-fluid cartridge and printing-fluid consuming apparatus
NZ756518B2 (en) Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
NZ756914B2 (en) Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
NZ716768B2 (en) Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
NZ757070A (en) Fire extinguisher with internal mixing and gas cartridge
CN116284054B (en) Ecteinascidins compound, antibody drug conjugate and application thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NITTOLI, THOMAS;JAIN, NARESHKUMAR F.;MARKOTAN, THOMAS PATRICK;SIGNING DATES FROM 20141106 TO 20141107;REEL/FRAME:039781/0340

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION