US20190314324A1 - Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy - Google Patents

Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy Download PDF

Info

Publication number
US20190314324A1
US20190314324A1 US16/381,774 US201916381774A US2019314324A1 US 20190314324 A1 US20190314324 A1 US 20190314324A1 US 201916381774 A US201916381774 A US 201916381774A US 2019314324 A1 US2019314324 A1 US 2019314324A1
Authority
US
United States
Prior art keywords
cancer
mcl
agent
inhibitor
nanoparticle
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
US16/381,774
Inventor
Wenbin Lin
Christina Chan
Wenbo Han
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.)
University of Chicago
Original Assignee
University of Chicago
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 University of Chicago filed Critical University of Chicago
Priority to US16/381,774 priority Critical patent/US20190314324A1/en
Assigned to THE UNIVERSITY OF CHICAGO reassignment THE UNIVERSITY OF CHICAGO ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHAN, CHRISTINA, HAN, Wenbo, LIN, WENBIN
Publication of US20190314324A1 publication Critical patent/US20190314324A1/en
Priority to US16/859,594 priority patent/US11389422B2/en
Priority to US17/855,327 priority patent/US20230036839A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0038Radiosensitizing, i.e. administration of pharmaceutical agents that enhance the effect of radiotherapy
    • 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
    • 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
    • A61K47/54Medicinal 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 organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • 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
    • A61K47/69Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • A61K9/0009Galenical forms characterised by the drug release technique; Application systems commanded by energy involving or responsive to electricity, magnetism or acoustic waves; Galenical aspects of sonophoresis, iontophoresis, electroporation or electroosmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy

Definitions

  • NPs nanoparticles
  • MCL micheliolide
  • NPs NPs
  • X-ray irradiation and/or other therapeutic agents e.g., immune checkpoint inhibitors
  • the combinations can provide synergistic anticancer therapeutic efficacy.
  • Chemotherapy and radiotherapy are among the powerful anticancer therapies used across a multitude of cancer types.
  • the effective use of these therapies can be limited due to undesirable side effects to healthy cells at high dosages.
  • these therapies are often compromised by the development of drug and radio-resistance by tumor cells.
  • the presently disclosed subject matter provides a composition comprising a nanoparticle and micheliolide (MCL) or a derivative thereof.
  • the composition comprises a derivative of MCL, wherein said derivative is dimethylamino micheliolide (DMAMCL) or a lipid conjugate of MCL.
  • DMAMCL dimethylamino micheliolide
  • the MCL or the derivative thereof is attached to or sequestered within the nanoparticle.
  • the nanoparticle is selected from the group comprising a polymeric micelle, a liposome, a dendrimer, an organic polymer-based nanoparticle, a silica-based nanoparticle, a nanoscale coordination polymer, a nanoscale metal-organic framework, and an inorganic nanoparticle.
  • the nanoparticle comprises a core and one or more coating layers or agents covering at least a portion of an outer surface of the core.
  • the core comprises an organic polymer, a silica-based material, a nanoscale coordination polymer, a metal-organic framework, or an inorganic material.
  • the one or more coating agents or layers are selected from a metal oxide, a polymer, a single lipid layer, a lipid bilayer, and combinations thereof.
  • the one or more coating agents or layers further comprise a passivating agent, a targeting agent, and/or an imaging agent.
  • the MCL or derivative thereof is attached to or sequestered in the nanoparticle core. In some embodiments, the MCL or the derivative thereof is attached to or sequestered in a nanoparticle coating layer.
  • the composition further comprises a chemotherapeutic agent and/or an immunotherapy agent.
  • the chemotherapeutic agent is attached or sequestered in a nanoparticle core or a nanoparticle coating layer.
  • the chemotherapeutic agent is selected from the group comprising cisplatin, oxaliplatin, or prodrugs thereof; doxorubicin, daunorubicin, docetaxel, mitoxanthrone, paclitaxel, digitoxin, gemcitabine, methotrexate, leucovorin, pemetresed disodium, vinblastine, vincristine, vindesine, cytarabine, azathioprine, melphalan, imitinib, anastrozole, letrozole, carboplatin, etoposide, vinorelbine, and septacidin.
  • the immunotherapy agent is selected from the group comprising an anti-CD52 antibody, an anti-CD20 antibody, anti-CD47 antibody, an anti-GD2 antibody, polysaccharide K, a cytokine, Alemtuzumab, Ofatumumab, Rituximab, Zevalin, Adcetris, Kadcyla, Ontak, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • the immunotherapy agent is attached to or sequestered in a nanoparticle coating layer.
  • the presently disclosed subject matter provides a pharmaceutical composition
  • a pharmaceutical composition comprising a composition comprising a nanoparticle and micheliolide (MCL) or a derivative thereof and a pharmaceutically acceptable carrier.
  • MCL nanoparticle and micheliolide
  • the presently disclosed subject matter provides a method for treating a disease in a subject in need thereof, the method comprising: administering to the subject micheliolide (MCL) or a derivative thereof; and exposing at least a portion of the subject to ionizing irradiation energy.
  • administering the MCL or derivative thereof comprises administering a nanoparticle and/or a nanoscale coordination polymer (NCP) comprising the MCL or derivative thereof.
  • NCP nanoscale coordination polymer
  • the ionizing irradiation energy is X-rays or protons.
  • the disease is cancer.
  • the disease is selected from the group comprising a skin cancer, a connective tissue cancer, an adipose cancer, a breast cancer, a head and neck cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system (CNS) cancer, a retinal cancer, a blood cancer, a neuroblastoma, multiple myeloma, and a lymphoid cancer.
  • CNS central nervous system
  • the method further comprises administering to the subject an additional therapeutic agent or treatment selected from an immunotherapy agent and/or a cancer treatment, wherein the cancer treatment is selected from the group comprising surgery, chemotherapy, toxin therapy, cryotherapy and gene therapy.
  • the additional therapeutic agent or treatment comprises an immunotherapy agent.
  • the immunotherapy agent is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is selected from the group comprising a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • the presently disclosed subject matter provides a method for treating a disease in a subject in need thereof, the method comprising: administering to the subject micheliolide (MCL) or a derivative thereof; and administering to the subject one or more additional therapeutic agent or treatment.
  • administering to the subject MCL or a derivative thereof comprises administering to the subject a nanoparticle comprising MCL or a derivative thereof.
  • the one or more additional therapeutic agent or treatment agent is selected from a chemotherapeutic agent, an immune therapy agent, radiotherapy, surgery, chemotherapy, toxin therapy, cryotherapy and gene therapy.
  • the disease is cancer.
  • the disease is selected from the group comprising a skin cancer, a connective tissue cancer, an adipose cancer, a breast cancer, a head and neck cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system (CNS) cancer, a retinal cancer, a blood cancer, a neuroblastoma, multiple myeloma, and a lymphoid cancer.
  • CNS central nervous system
  • the one or more additional therapeutic agent or treatment comprises an immunotherapeutic agent.
  • the immunotherapeutic agent is an immune checkpoint inhibitor.
  • the one or more additional therapeutic agent or treatment further comprises radiotherapy. In some embodiments, the one or more additional therapeutic agent or treatment agent further comprises one or more of surgery, toxin therapy, cryotherapy and gene therapy. In some embodiments, the one or more additional treatment agent comprises chemotherapy.
  • compositions comprising MCL or a derivative thereof in a nanoparticle and/or in combination with another therapeutic agent, e.g., a chemotherapeutic agent or an immunotherapeutic agent, as well as methods of treating disease using the compositions, alone or in combination with X-ray irradiation and/or another therapy, e.g., immunotherapy.
  • another therapeutic agent e.g., a chemotherapeutic agent or an immunotherapeutic agent
  • FIG. 1 is a graph showing the in vitro cytotoxicity of combinations of dimethylamino micheliolide (DMAMCL) and irradiation in mouse glioma 261 (GL261) cells.
  • the cytotoxic effect of the combination is represented by a decrease in cell viability (in percentage (5%)).
  • the cytotoxicity was measured at two different concentrations of DMAMCL: 1 micromolar (1 ⁇ M, open triangles) and 10 ⁇ M (open circles).
  • the irradiation dose varied from 0 to 8 gray (Gy) as indicated on the x-axis.
  • cell viability data for GL261 cells treated with irradiation and phosphate buffered saline (PBS, filled squares) in place of the DMAMCL is also shown.
  • FIG. 2 is a graph showing the in vivo antitumor efficacy of combinations of dimethylamino micheliolide (DMAMCL) and irradiation in a mouse glioma 261 (GL261) tumor model in immunocompromised mice.
  • Mice were orally dosed with 50 milligrams per kilogram (mg/kg) DMAMCL or with phosphate buffered saline and irradiated or not irradiated with a 4 gray (Gy) irradiation dose about 4 hours later every day for up to 7 days.
  • FIG. 3 is a graph showing the in vivo antitumor efficacy of combinations of micheliolide (MCL), irradiation, and an immunotherapy agent in a mouse glioma 261 (GL261) tumor model in immunocompetent mice.
  • Mice were orally dosed with 50 milligrams per kilogram (mg/kg) MCL or phosphate buffered saline daily for up to 12 days. Some of the mice were irradiated with a 2 gray (Gy) irradiation dose about 4 hours after dosing with MCL. Some of the mice also received two 75 microgram ( ⁇ g) doses of a programmed death ligand 1 (PD-L1) antibody on days 0 and 4.
  • PD-L1 programmed death ligand 1
  • mice treated with PBS and not irradiated is shown in filled squares, data for mice treated with MCL and not irradiated is shown in open circles, data for mice treated with MCL and radiation (MCL (+)) is shown in open triangles, and data for mice treated with MCL, irradiation and antibody (MCL+ ⁇ -PD-L1 (+)) is shown in open stars. Tumor size was measured in cubic centimeters (cm 3 ).
  • FIG. 4 is a graph showing the intensity plot (intensity (in percentage (%)) versus size (diameter in nanometers (d.nm))) of oxaliplatin-containing nano-coordination polymers (OxNCP) also comprising micheliolide (MCL) as measured using dynamic light scattering (DLS).
  • OxNCP oxaliplatin-containing nano-coordination polymers
  • MCL micheliolide
  • DLS dynamic light scattering
  • chemotherapeutic agent includes a plurality of such chemotherapeutic agents, and so forth.
  • the term “about”, when referring to a value or to an amount of size (i.e., diameter), dose, weight, concentration, or percentage is meant to encompass variations of in one example ⁇ 20% or ⁇ 10%, in another example ⁇ 5%, in another example ⁇ 1%, and in still another example ⁇ 0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods.
  • Numerical ranges recited herein by endpoints include all numbers and fractions subsumed within that range (e.g. 1 to 5 includes, but is not limited to, 1, 1.5, 2, 2.75, 3, 3.90, 4, and 5).
  • the phrase “A, B, C, and/or D” includes A, B, C, and D individually, but also includes any and all combinations and sub-combinations of A, B, C, and D.
  • the phrase “consisting of” excludes any element, step, or ingredient not specified in the claim.
  • the phrase “consists of” appears in a clause of the body of a claim, rather than immediately following the preamble, it limits only the element set forth in that clause; other elements are not excluded from the claim as a whole.
  • alkyl can refer to C 1 -20 inclusive, linear (i.e., “straight-chain”), branched, or cyclic, saturated or at least partially and in some cases fully unsaturated (i.e., alkenyl and alkynyl) hydrocarbon chains, including for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl, octyl, ethenyl, propenyl, butenyl, pentenyl, hexenyl, octenyl, butadienyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, and allenyl groups.
  • Branched refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain.
  • Lower alkyl refers to an alkyl group having 1 to about 8 carbon atoms (i.e., a C 1-8 alkyl), e.g., 1, 2, 3, 4, 5, 6, 7, or 8 carbon atoms.
  • Higher alkyl refers to an alkyl group having about 10 to about 20 carbon atoms, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • alkyl refers, in particular, to C 1-8 straight-chain alkyls. In other embodiments, “alkyl” refers, in particular, to C 1-8 branched-chain alkyls.
  • Alkyl groups can optionally be substituted (a “substituted alkyl”) with one or more alkyl group substituents, which can be the same or different.
  • alkyl group substituent includes but is not limited to alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl.
  • alkyl chain there can be optionally inserted along the alkyl chain one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is hydrogen, lower alkyl (also referred to herein as “alkylaminoalkyl”), or aryl.
  • substituted alkyl includes alkyl groups, as defined herein, in which one or more atoms or functional groups of the alkyl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • Alkylene refers to a straight or branched bivalent aliphatic hydrocarbon group having from 1 to about 20 carbon atoms, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • the alkylene group can be straight, branched or cyclic.
  • the alkylene group also can be optionally unsaturated and/or substituted with one or more “alkyl group substituents.” There can be optionally inserted along the alkylene group one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms (also referred to herein as “alkylaminoalkyl”), wherein the nitrogen substituent is alkyl as previously described.
  • alkylene groups include methylene (—CH 2 —); ethylene (—CH 2 —CH 2 —); propylene (—(CH 2 ) 3 —); cyclohexylene (—C 6 H 10 —); —CH ⁇ CH—CH ⁇ CH—; —CH ⁇ CH—CH 2 —; —(CH 2 ) q —N(R)—(CH 2 ) r —, wherein each of q and r is independently an integer from 0 to about 20, e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and R is hydrogen or lower alkyl; methylenedioxyl (—O—CH 2 —O—); and ethylenedioxyl (—O—(CH 2 ) 2 —O—).
  • An alkylene group can have about 2 to about 3 carbon atoms and can further have 6-20 carbons.
  • aryl is used herein to refer to an aromatic substituent that can be a single aromatic ring, or multiple aromatic rings that are fused together, linked covalently, or linked to a common group, such as, but not limited to, a methylene or ethylene moiety.
  • the common linking group also can be a carbonyl, as in benzophenone, or oxygen, as in diphenylether, or nitrogen, as in diphenylamine.
  • aryl specifically encompasses heterocyclic aromatic compounds.
  • the aromatic ring(s) can comprise phenyl, naphthyl, biphenyl, diphenylether, diphenylamine and benzophenone, among others.
  • aryl means a cyclic aromatic comprising about 5 to about 10 carbon atoms, e.g., 5, 6, 7, 8, 9, or 10 carbon atoms, and including 5- and 6-membered hydrocarbon and heterocyclic aromatic rings.
  • the aryl group can be optionally substituted (a “substituted aryl”) with one or more aryl group substituents, which can be the same or different, wherein “aryl group substituent” includes alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, hydroxyl, alkoxyl, aryloxyl, aralkyloxyl, carboxyl, acyl, halo, nitro, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, acyloxyl, acylamino, aroylamino, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, arylthio, alkylthio, alkylene, and —NR′R′′, wherein R′ and R′′ can each be independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, and aralkyl.
  • substituted aryl includes aryl groups, as defined herein, in which one or more atoms or functional groups of the aryl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • aryl groups include, but are not limited to, cyclopentadienyl, phenyl, furan, thiophene, pyrrole, pyran, pyridine, imidazole, benzimidazole, isothiazole, isoxazole, pyrazole, pyrazine, triazine, pyrimidine, quinoline, isoquinoline, indole, carbazole, and the like.
  • arylene refers to a bivalent aromatic group, e.g., a bivalent phenyl or naphthyl group.
  • the arylene group can optionally be substituted with one or more aryl group substituents and/or include one or more heteroatoms.
  • arylene includes bivalent groups comprising more than one aromatic ring fused to or linked to one another (e.g., via a covalent bond or a linking group), such as bivalent biphenyl or bivalent terphenyl.
  • aralkyl refers to a -alkyl-aryl group, which can be substituted or unsubstituted.
  • An exemplary aralkyl group is benzyl.
  • acyl refers to the group —C( ⁇ O)R, wherein R is a substituted or unsubstituted alkyl, aralkyl, or aryl group.
  • amino refers to the group —N(R) 2 wherein each R is independently H, alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, or substituted aralkyl.
  • aminoalkyl and alkylamino can refer to the group —N(R) 2 wherein each R is H, alkyl or substituted alkyl, and wherein at least one R is alkyl or substituted alkyl.
  • Arylamine and “aminoaryl” refer to the group —N(R) 2 wherein each R is H, aryl, or substituted aryl, and wherein at least one R is aryl or substituted aryl, e.g., aniline (i.e., —NHC 6 H 5 ).
  • lipid can refer to a hydrophobic or amphiphilic small molecule, such as, but not limited to a fatty acid, a sterol, a fat-soluble vitamin, a phospholipid, a glycerolipid, a glycerophospholipid, a sphingolipid, a saccharolipid, or a polyketide.
  • the term “metal-organic framework” or “MOF” refers to a solid two- or three-dimensional network comprising both metal and organic components, wherein the organic components include at least one, and typically more than one carbon atom.
  • the material is crystalline.
  • the material is amorphous.
  • the material is porous.
  • the metal-organic matrix material is a coordination polymer, which comprises repeating units of coordination complexes comprising a metal-based secondary building unit (SBU), such as a metal ion or metal complex, and a bridging polydentate (e.g., bidentate or tridentate) organic ligand.
  • SBU metal-based secondary building unit
  • a bridging polydentate e.g., bidentate or tridentate
  • the material contains more than one type of SBU or metal ion.
  • the material can contain more than one type of organic bridging ligand.
  • nanoscale metal-organic framework can refer to a nanoscale particle comprising an MOF.
  • nanoscale particle refers to a structure having at least one region with a dimension (e.g., length, width, diameter, etc.) of less than about 1,000 nm.
  • the dimension is smaller (e.g., less than about 500 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 125 nm, less than about 100 nm, less than about 80 nm, less than about 70 nm, less than about 60 nm, less than about 50 nm, less than about 40 nm, less than about 30 nm or even less than about 20 nm).
  • the dimension is between about 20 nm and about 250 nm (e.g., about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 nm).
  • the nanoparticle is approximately spherical.
  • the characteristic dimension can correspond to the diameter of the sphere.
  • the nanomaterial can be disc-shaped, plate-shaped (e.g., hexagonally plate-like), oblong, polyhedral, rod-shaped, cubic, or irregularly-shaped.
  • the nanoparticle can comprise a core region (i.e., the space between the outer dimensions of the particle) and an outer surface (i.e., the surface that defines the outer dimensions of the particle).
  • the nanoparticle can have one or more coating layers surrounding or partially surrounding the nanoparticle core.
  • a spherical nanoparticle can have one or more concentric coating layers, each successive layer being dispersed over the outer surface of a layer closer to the center of the particle.
  • the presently disclosed nanoparticle can comprise a solid metal-organic framework matrix, which can comprise one or more pores or hollow interior regions.
  • the matrix can be amorphous or crystalline.
  • the nanoparticle core can further comprise one or more of: MCL or a derivative thereof; an optical imaging agent; and/or another therapeutic agent (e.g., an anticancer agent or an immunotherapeutic agent), which can be physically trapped within the matrix, coordinated to a metal ion of the matrix, or chemically bonded (e.g., to an organic bridging ligand in the matrix) via a covalent or ionic bond.
  • a chemotherapeutic agent or prodrug thereof can be an organic bridging ligand within a metal-organic matrix material that forms the core of the nanoparticle.
  • the matrix material is a metal bisphosphonate coordination polymer
  • the bisphosphonate can be a chemotherapeutic agent or prodrug thereof.
  • the core comprises a non-matrix therapeutic and/or imaging agent
  • said agents can be said to be “embedded” in the nanoparticle.
  • “Embedded” can refer to a therapeutic agent or an imaging agent that is bound, for example covalently bound or bound via a coordinative bond, inside the core of the particle (e.g., to a bisphosphonate, dicarboxylate, or metal ion of the matrix material).
  • the complex or agent can be “sequestered” (i.e., non-covalently/physically encapsulated) inside pores in the core or interact with a core material via hydrogen bonding, London dispersion forces, or any other non-covalent interaction.
  • polymer and “polymeric” refer to chemical structures that have repeating units (i.e., multiple copies of a given chemical substructure).
  • Polymers can be formed from polymerizable monomers.
  • a polymerizable monomer is a molecule that comprises one or more moieties that can react to form bonds (e.g., covalent or coordination bonds) with moieties on other molecules of polymerizable monomer.
  • bonds e.g., covalent or coordination bonds
  • each polymerizable monomer molecule can bond to two or more other molecules.
  • a polymerizable monomer will bond to only one other molecule, forming a terminus of the polymeric material.
  • Polymers can be organic, or inorganic, or a combination thereof.
  • inorganic refers to a compound or composition that contains at least some atoms other than carbon, hydrogen, nitrogen, oxygen, sulfur, phosphorous, or one of the halides.
  • an inorganic compound or composition can contain one or more silicon atoms and/or one or more metal atoms.
  • organic polymers are those that do not include silica or metal atoms in their repeating units.
  • exemplary organic polymers include polyvinylpyrrolidone (PVO), polyesters, polyamides, polyethers, polydienes, and the like.
  • PVO polyvinylpyrrolidone
  • Some organic polymers contain biodegradable linkages, such as esters or amides, such that they can degrade over time under biological conditions.
  • hydrophilic polymer generally refers to hydrophilic organic polymers, such as but not limited to, polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethyacrylamide, polydimethylacrylamide, polyhydroxylpropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethylenimine (PEI), polyethyleneglycol (i.e., PEG) or another hydrophilic poly(alkyleneoxide), polyglycerine, and polyaspartamide.
  • hydrophilic refers to the ability of a molecule or chemical species to interact with water. Thus, hydrophilic polymers are typically polar or have groups that can hydrogen bond to water.
  • bonding or “bonded” and variations thereof can refer to either covalent or non-covalent bonding. In some cases, the term “bonding” refers to bonding via a coordinate bond. The term “conjugation” can refer to a bonding process, as well, such as the formation of a covalent linkage or a coordinate bond.
  • a “coordination complex” is a compound in which there is a coordinate bond between a metal ion and an electron pair donor, ligand or chelating group.
  • ligands or chelating groups are generally electron pair donors, molecules or molecular ions having unshared electron pairs available for donation to a metal ion.
  • coordinate bond refers to an interaction between an electron pair donor and a coordination site on a metal ion resulting in an attractive force between the electron pair donor and the metal ion.
  • coordinate bond refers to an interaction between an electron pair donor and a coordination site on a metal ion resulting in an attractive force between the electron pair donor and the metal ion.
  • the use of this term is not intended to be limiting, in so much as certain coordinate bonds also can be classified as having more or less covalent character (if not entirely covalent character) depending on the characteristics of the metal ion and the electron pair donor.
  • ligand refers generally to a species, such as a molecule or ion, which interacts, e.g., binds, in some way with another species. More particularly, as used herein, a “ligand” can refer to a molecule or ion that binds a metal ion in solution to form a “coordination complex.” See Martell, A. E., and Hancock, R. D., Metal Complexes in Aqueous Solutions , Plenum: New York (1996), which is incorporated herein by reference in its entirety. The terms “ligand” and “chelating group” can be used interchangeably.
  • bridging ligand can refer to a group that bonds to more than one metal ion or complex, thus providing a “bridge” between the metal ions or complexes.
  • Organic bridging ligands can have two or more groups with unshared electron pairs separated by, for example, an alkylene or arylene group. Groups with unshared electron pairs, include, but are not limited to, —CO 2 H, —NO 2 , amino, hydroxyl, thio, thioalkyl, —B(OH) 2 , —SO 3 H, POSH, phosphonate, and heteroatoms (e.g., nitrogen, oxygen, or sulfur) in heterocycles.
  • heteroatoms e.g., nitrogen, oxygen, or sulfur
  • coordination site when used herein with regard to a ligand, e.g., a bridging ligand, refers to an unshared electron pair, a negative charge, or atoms or functional groups cable of forming an unshared electron pair or negative charge (e.g., via deprotonation under at a particular pH).
  • an imaging agent refers to a chemical moiety that aids in the visualization of a sample.
  • an imaging agent can be a “contrast agent” and can refer to a moiety (a specific part of or an entire molecule, macromolecule, coordination complex, or nanoparticle) that increases the contrast of a biological tissue or structure being examined.
  • the contrast agent can increase the contrast of a structure being examined using, for example, magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) imaging, single photon emission computed tomography (SPECT) imaging, or a combination thereof (i.e., the contrast agent can be multimodal).
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • MRI contrast agent refers to a moiety that effects a change in induced relaxation rates of water protons in a sample.
  • optical imaging agent or “optical contrast agent” refer to a group that can be detected based upon an ability to absorb, reflect or emit light (e.g., ultraviolet, visible, or infrared light). Optical imaging agents can be detected based on a change in amount of absorbance, reflectance, or fluorescence, or a change in the number of absorbance peaks or their wavelength maxima. Thus, optical imaging agents include those which can be detected based on fluorescence or luminescence, including organic and inorganic dyes.
  • fluorophore and “fluorescent moiety” refer to species that can be excited by visible light or non-visible light (e.g., UV light).
  • fluorophores include, but are not limited to: quantum dots and doped quantum dots (e.g., a semiconducting CdSe quantum dot or a Mn-doped CdSe quantum dot), fluorescein, fluorescein derivatives and analogues, indocyanine green, rhodamine, triphenylmethines, polymethines, cyanines, phalocyanines, naphthocyanines, merocyanines, lanthanide complexes or cryptates, fullerenes, oxatellurazoles, LaJolla blue, porphyrins and porphyrin analogues and natural chromophores/fluorophores such as chlorophyll, carotenoids, flavonoids, bilins, phytochrome, phycobilin
  • cancer refers to diseases caused by uncontrolled cell division and/or the ability of cells to metastasize, or to establish new growth in additional sites.
  • malignant refers to cancerous cells or groups of cancerous cells.
  • cancers include, but are not limited to, skin cancers (e.g., melanoma), connective tissue cancers (e.g., sarcomas), adipose cancers, breast cancers, head and neck cancers, lung cancers (e.g., mesothelioma), stomach cancers, pancreatic cancers, ovarian cancers, cervical cancers, uterine cancers, anogenital cancers (e.g., testicular cancer), kidney cancers, bladder cancers, colon cancers, prostate cancers, central nervous system (CNS) cancers, retinal cancer, blood, neuroblastomas, multiple myeloma, and lymphoid cancers (e.g., Hodgkin's and non-Hodgkin's lymphomas).
  • skin cancers e.g., melanoma
  • connective tissue cancers e.g., sarcomas
  • adipose cancers e.g., breast cancers,
  • metalstatic cancer refers to cancer that has spread from its initial site (i.e., the primary site) in a patient's body.
  • anticancer drug refers to drugs (i.e., chemical compounds) or prodrugs known to, or suspected of being able to treat a cancer (i.e., to kill cancer cells, prohibit proliferation of cancer cells, or treat a symptom related to cancer).
  • chemotherapeutic refers to a non-MCL-based molecule that is used to treat cancer and/or that has cytotoxic ability.
  • Some traditional or conventional chemotherapeutic agents can be described by mechanism of action or by chemical compound class, and can include, but are not limited to, alkylating agents (e.g., melphalan), anthracyclines (e.g., doxorubicin), cytoskeletal disruptors (e.g., paclitaxel), epothilones, histone deacetylase inhibitors (e.g., vorinostat), inhibitors of topoisomerase I or II (e.g., irinotecan or etoposide), kinase inhibitors (e.g., bortezomib), nucleotide analogs or precursors thereof (e.g., methotrexate), peptide antibiotics (e.g., bleomycin), platinum based agents (e.g., cisplatin or oxaliplatin), retinoids (e.g., tretinoin), and vinka alkaloids (e.g
  • MCL Water-soluble micheliolide
  • the presently disclosed subject matter is related to the finding that MCL and related compounds can synergize with other disease treatments, particularly cancer treatments, such as chemotherapy and radiotherapy. Without being bound by any one theory, it is believed that the synergistic effects with regard to treating cancer can be related to the activity of the MCL or a related compound in sensitizing cancer cells to therapy and/or targeting resistant CSCs for selective cell death.
  • the combined efficacy of MCL and related compounds with the other treatment e.g., with radiation
  • immune checkpoint inhibitors can be further enhanced by immune checkpoint inhibitors.
  • nanoparticles containing MCL or a derivative thereof, and optionally one or more additional therapeutic agents (e.g., chemotherapeutic agents and/or immunotherapy agents), can be used to enhance the bioavailability and relative tumor deposition of MCL, its derivative and/or other agents, allowing for lower total doses and less frequent dosing.
  • additional therapeutic agents e.g., chemotherapeutic agents and/or immunotherapy agents
  • the presently disclosed subject matter relates to the use of MCL NPs, as well as to combinations of MCL or a derivative thereof and/or NPs with X-ray irradiation and/or other therapeutic agents, e.g., immune checkpoint inhibitors, for treating disease (e.g., cancer).
  • the combinations can provide synergistic anticancer therapeutic efficacy.
  • the presently disclosed subject matter provides a composition comprising: (i) a nanoparticle and (ii) MCL or a derivative thereof.
  • MCL has the following structure:
  • MCL The cyclization of parthenolide, a sesquiterpene lactone found in feverfew ( Tanacetum parthenium ), to form MCL was described in 1993. See Casta ⁇ eda-Acosta et al., J. Nat. Prod. 1993, 56(1), 90-98.
  • Exemplary derivatives of MCL include, but are not limited to, the compounds described in U.S. Pat. No. 9,255,078, the disclosure of which is incorporated herein by reference in its entirety.
  • the derivative of MCL comprises a compound wherein a carbon-carbon double bond has been replaced by a carbon-carbon single bond or a three-membered ring (e.g., an epoxide) and/or wherein one of the carbon atoms of the carbon-carbon double bond has been substituted, e.g., by a substituted or unsubstituted alkyl group; wherein an oxygen atom has been replaced by —CH 2 — or a —NR′— group, wherein R′ is an alkyl or substituted alkyl group; or wherein the hydroxyl group has been substituted by an alkyl, substituted alkyl, or acyl group.
  • the MCL derivative can be provided as a pharmaceutically acceptable salt.
  • the MCL derivative has a structure of the formula:
  • R is selected from H, alkyl, substituted alkyl, and acyl; and R 1 is selected from H, alkyl, substituted alkyl, and amino; or a pharmaceutically acceptable salt thereof.
  • the amino group is —N(R 2 )(R 3 ), wherein R 2 and R 3 are independently selected from H, alkyl (e.g., C 1-12 or C 1-6 alkyl), cycloalkyl, and substituted alkyl, or wherein R 2 and R 3 together form an alkylene group, e.g., propylene, butylene, or pentylene.
  • R is substituted alkyl, e.g., —CH 2 —O—R 4 , wherein R 4 is alkyl or alkenyl, optionally wherein R 4 is a lipid.
  • R 1 is —N(R 2 )(R 3 ).
  • R 2 and R 3 are each methyl, ethyl or propyl.
  • the MCL derivative is a lipid conjugate of MCL or a derivative thereof, e.g., a compound wherein the hydroxyl group of MCL is covalently linked (e.g., via one or more ether or ester groups) to a lipid or lipid derivative (e.g., a sterol, a fatty acid or a fatty alcohol).
  • the MCL derivative is dimethylamino micheliolide (DMAMCL) or a lipid conjugate of MCL or DMAMCL.
  • any suitable nanoparticle can be used, including but not limited to a polymeric micelle, a liposome, a dendrimer, an organic polymer-based nanoparticle, a silica-based nanoparticle, a nanoscale coordination polymer (NCP), a nanoscale metal-organic framework (nMOF), or an inorganic polymer, such as a gold or iron oxide nanoparticle.
  • the nanoparticle is a NCP and/or nMOF. Exemplary methods of preparing and using NCPs and nMOFs are described, for example in PCT International Patent Application Publication Nos. WO 2015/069926 and WO 2013/009701, U.S. Pat. No. 9,693,957, and U.S. Patent Application Publication No. 2016/0346204, the disclosures of each of which are incorporated herein by reference in their entireties.
  • the nanoparticle comprises a nMOF that comprises metal-containing clusters or ions coordinated to organic molecules, wherein the material comprises repeating coordination units.
  • the organic molecule (which can also be referred to as a “bridging ligand”) can include, for example, carboxylate, phosphate, amino, mercapto, or hydroxyl groups to form coordinate bonds with a metal ion.
  • the metal-containing clusters can be referred to as secondary building units (SBUs).
  • the SBUs are metal oxo clusters.
  • suitable SBUs can include, but are not limited to, Zr-oxo clusters, Hf-oxo clusters, Zn-oxo clusters, Ti-oxo clusters, Cu-carboxylate paddlewheels, and others.
  • the oxo clusters comprise anions selected from oxide (O 2 ⁇ ), hydroxide (OH ⁇ ), S 2 ⁇ , SH ⁇ , and formate (HCO 2 ⁇ ).
  • Each SBU is bonded to at least one other SBU via coordinative bonding to the same bridging ligand. Stated another way, each SBU is bonded via a coordinative bond to at least one bridging ligand, which is also coordinatively bonded to at least one other SBU.
  • each bridging ligand is an organic compound comprising multiple coordination sites.
  • the coordination sites can each comprise a group capable of forming a coordinate bond with a metal cation or a group capable of forming such a group.
  • each coordination site can comprise an unshared electron pair, a negative charge, or an atom or functional group capable of forming an unshared electron pair or negative charge.
  • Typical coordination sites include, but are not limited to, functional groups such as carboxylate and derivatives there (e.g., esters, amides, anhydrides), nitrogen-containing groups (e.g., amines, nitrogen-containing aromatic and non-aromatic heterocycles), alcohols, phenols and other hydroxyl-substituted aromatic groups; ethers, acetylacetonate (i.e., C( ⁇ O)CH 2 C( ⁇ O)CH 3 ), phosphonates, phosphates, thiols, and the like.
  • each bridging ligand comprises between 2 and 10 coordination sites (i.e., 2, 3, 4, 5, 6, 7, 8, 9, or 10 coordination sites).
  • each bridging ligand is capable of binding to two or three SBUs.
  • each of the organic bridging ligands is a dicarboxylate or a tricarboxylate.
  • the coordination sites of the organic bridging ligand can be bonded to the same polyvalent (e.g., divalent or trivalent) linking group, such as an alkylene or arylene group.
  • the bridging ligand further includes a group, such as an amino, hydroxyl, or thiol, that can form a covalent bond with a therapeutic agent (e.g., a chemotherapeutic or immunotherapeutic agent or MCL or a derivative thereof).
  • the bridging ligand can be amino-triphenyldicarboxylic acid (amino-TPDC).
  • the NCP is a metal bisphosphonate coordination polymer comprising a multivalent metal ion and a bisphosphonate. Any suitable multivalent metal ion can be used. In some embodiments, the multivalent metal ion is divalent. In some embodiments, the metal ion is Ca 2+ , Mg 2+ , Mn 2+ , Zn 2+ , or a combination thereof. In some embodiments, the metal is Zn 2+ . In some embodiments, the bisphosphonate is a chemotherapeutic prodrug. Thus, in some embodiments, a chemotherapeutic is present as a bridging ligand between metal ions in the NCP.
  • the bisphosphonate is a cisplatin, oxaliplatin, or carboplatin prodrug.
  • the prodrug can be prepared by providing a metal complex based on cisplatin, oxaliplatin, carboplatin or another platinum coordination complex chemotherapeutic and having two hydroxyl ligands, and contacting the metal complex with diethoxyphosphinyl isocyanate, diethoxyphosphinyl isothiocyanate, diethoxyphosphinyl-containing carboxylic anhydride, or diethoxyphosphinyl-containing acyl chloride to form metal ligands that can provide phosphonate groups available for further coordinative bonding.
  • the bisphosphonate can be a bisphosphonate ester of cis, cis-trans-[Pt(NH 3 ) 2 Cl 2 (OH) 2 ], a dihydroxyl complex based on cisplatin.
  • the bisphosphonate comprises a metal coordination complex and, therefore, includes a second metal ion, such as a Pt ion, in addition to the multivalent metal ion of the NCP.
  • the bisphosphonate is an organic compound comprising at least two phosphonate groups and is free of an additional metal ion.
  • the bisphosphonate can have a structure of the formula:
  • n and m are each independently an integer between 0 and 2 (i.e., each independently 0, 1, or 2); each R is independently H, alkyl, substituted alkyl, aralkyl, aryl, or substituted aryl; R 1 is H, hydroxyl, halo, alkyl, substituted alkyl, amino, alkoxy, or alkylthio; and R 2 is halo, alkyl, substituted alkyl, aralkyl, substituted aralkyl, alkoxy, alkylthio, aryloxy, arylthio or arylamino; or an anion or salt thereof.
  • the nanoparticle can comprise a core comprising one of the above-mentioned materials, such as an organic polymer, a silica-based matrix, a NCP, a nMOF, or an inorganic material, and further comprises one or more coating layers that surrounds at least a portion of the surface of the nanoparticle core and/or one or more coating agents, e.g., an agent attached to a coating layer or to the surface of the nanoparticle core.
  • one of the above-mentioned materials such as an organic polymer, a silica-based matrix, a NCP, a nMOF, or an inorganic material
  • one or more coating layers that surrounds at least a portion of the surface of the nanoparticle core and/or one or more coating agents, e.g., an agent attached to a coating layer or to the surface of the nanoparticle core.
  • Suitable coating agents or layers include, but are not limited to, metal oxide, polymer (e.g., a silica-based polymer, such as silica, poly(siloxane), or poly(silsesquioxane), or an organic or hydrophilic organic polymer), a single lipid layer, a lipid bilayer, or a combination thereof.
  • one or more of the coating agents or layers can be derivatized with a targeting agent (such as a RGD peptide or an antibody or antibody fragment) and/or a passivating agent (such as a hydrophilic polymer e.g., poly(ethylene glycol) (PEG)) and or an imaging agent (such as a fluorescent moiety).
  • a targeting agent such as a RGD peptide or an antibody or antibody fragment
  • a passivating agent such as a hydrophilic polymer e.g., poly(ethylene glycol) (PEG)
  • an imaging agent such as a fluorescent moiety
  • the one or more coating agents or layers are selected from the group comprising, but not limited to, dioleoyl L- ⁇ -phosphatidylethanolamine (DOPE), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dioleoyl-sn-glycero-3-phosphate sodium salt (DOPA), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), pegylated DSPE (e.g., DSPE-PEG 2k ), 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), anisamide-derivatized DOPE, cholesterol, silica, cRGfK-derivatized silica, PEG-derivatized silica, anisamide-PEG-derivatized silica, and combinations thereof.
  • DOPE dioleoyl L- ⁇ -phosphatidylethanolamine
  • DOPC 1,2-d
  • the MCL or derivative thereof is part of the nanoparticle.
  • the MCL or its derivative can be attached to or sequestered within the nanoparticle core.
  • the MCL derivative can include a chemical functional group that can form a covalent or ionic bond with an organic functional group or metal ion present in a component of an NCP or nMOF nanoparticle core.
  • the MCL or derivative thereof can be physically sequestered (e.g., encapsulated) in pores or channels in an NCP or nMOF nanoparticle core.
  • the MCL or its derivative can be attached (e.g., covalently or electrostatically) to a coating agent or layer or sequestered within a coating layer.
  • the MCL derivative can comprise a fatty acid or other lipid moiety and the MCL derivative can be sequestered in a lipid or lipid bilayer coating layer.
  • the nanoparticle comprises a therapeutic agent other than the MCL or its derivative, e.g., a chemotherapeutic agent or an immunotherapy agent.
  • a chemotherapeutic agent or immunotherapy agent can be used.
  • the chemotherapeutic agent can be selected from the group including, but not limited to, a platinum complex, such as cisplatin, oxaliplatin, carboplatin, or a prodrug thereof; doxorubicin; daunorubicin; docetaxel; mitoxanthrone; paclitaxel; digitoxin; gemcitabine; methotrexate; leucovorin; pemetresed disodium; vinblastine; vincristine; vindesine; cytarabine; azathioprine; melphalan; imitnib; anastrozole; letrozole; etoposide; vinorelbine; digoxin, and septacidin.
  • a platinum complex such as cisplatin
  • the nanoparticle can comprise more than one chemotherapeutic agent.
  • one or more chemotherapeutic agent is attached to or sequestered in a nanoparticle core.
  • one or more chemotherapeutic agent is attached to or sequestered in a nanoparticle coating layer.
  • the immunotherapy agent is selected from the group including, but not limited to, an anti-CD52 antibody, an anti-CD20 antibody, an anti-CD20 antibody, anti-CD47 antibody an anti-GD2 antibody, a radiolabeled antibody, an antibody-drug conjugate, polysaccharide K, a neoantigen, an anti-LAG3 antibody, an anti-4-IBB antibody, an anti-TIM3 antibody and a cytokine.
  • the cytokine is an interferon, an interleukin, or tumor necrosis factor alpha (TNF- ⁇ ).
  • TNF- ⁇ tumor necrosis factor alpha
  • the cytokine is selected from the group comprising IFN- ⁇ , INF- ⁇ , IL-2, IL-12 and TNF- ⁇ .
  • the immunotherapy agent is selected from the group comprising Alemtuzumab, Ofatumumab, Rituximab, Zevalin, Adcetris, Kadcyla and Ontak.
  • the immunotherapy agent is selected from the group comprising a PD-1 inhibitor or a PD-L1 inhibitor, such as, but not limited to, BMS-936559 or BMS-936558 from Bristol-Myers Squibb, MPDL3280A from Genentech, MK-3475 from Merck, CT-011 from Curetech, and MEDI4736 from MedImmune; a CTLA-4 inhibitor (e.g., ipilimumab, tremelimumab); an indoleamine-2,3-dioxygenase (IDO) inhibitor; and a CCR7 inhibitor.
  • a CTLA-4 inhibitor e.g., ipilimumab, tremelimumab
  • IDO indoleamine-2,3-dioxy
  • the IDO inhibitor can be any suitable IDO inhibitor, such as, but not limited to oxadiazole and other heterocyclic IDO inhibitors, e.g., as reported in U.S. Patent Application Publication Nos. 2006/0258719 and 2007/0185165, which are incorporated herein by reference in their entireties.
  • IDO inhibitors also include those described in PCT Publication WO 99/29310, incorporated herein by reference in its entirety, which reports methods for altering T cell-mediated immunity comprising altering local extracellular concentrations of tryptophan and tryptophan metabolites, BMS-986205 from Bristol-Myers Squibb or F001287 from Flexus, epacadostat from Incyte Corp., indoximod, IO-102, EOS-200271, HTI-1090, IO-101, KHK-2455, 1-methyl-DL-tryptophan, p-(3-benzofuranyl)-DL-alanine, p-[3-benzo(b)thienyl]-DL-alanine, and 6-nitro-L-tryptophan), and IDO inhibitors described in WO 03/087347, incorporated herein by reference in its entirety, also published as European Patent 1501918, which describes methods of making antigen-presenting cells for enhancing or reducing T cell tolerance.
  • the immunotherapy agent is an immune checkpoint inhibitor (e.g., an antibody immune checkpoint inhibitor, such as, but not limited to, a PD-1/PD-L1 antibody, a CTLA-4 antibody, an OX40 antibody, a TIM3 antibody, a LAG3 antibody, an anti-CD47 antibody).
  • an antibody immune checkpoint inhibitor such as, but not limited to, a PD-1/PD-L1 antibody, a CTLA-4 antibody, an OX40 antibody, a TIM3 antibody, a LAG3 antibody, an anti-CD47 antibody.
  • one or more immunotherapy agent is attached to or sequestered in a nanoparticle core.
  • one or more immunotherapy agent is attached to or sequestered in a nanoparticle coating layer.
  • the nanoparticle comprises both MCL or a derivative thereof and one or more additional therapeutic agents (e.g., chemotherapeutic and/or immunotherapy agents).
  • the other therapeutic agent can be attached to or sequestered within the nanoparticle core or can be attached to or sequestered in a coating layer or agent.
  • the nanoparticle is an NCP or nMOF comprising a chemotherapeutic agent, e.g., cisplatin, oxaliplatin, or a prodrug thereof, and the nanoparticle further comprises a lipid coating layer comprising the MCL or its derivative, e.g., a MCL lipid conjugate.
  • both the MCL or its derivative and the additional therapeutic agent are attached to or sequestered in the nanoparticle core. In some embodiments, both the MCL or its derivative and the additional therapeutic agent are attached to or sequestered in a coating layer, attached to the nanoparticle core surface, or attached to a coating agent.
  • the composition comprises a mixture of nanoparticles, wherein one type of nanoparticle comprises MCL or a derivative thereof and another type of nanoparticle comprises one or more additional therapeutic agents (e.g., a chemotherapeutic, an immunotherapeutic and/or a nucleic acid therapeutic agent).
  • additional therapeutic agents e.g., a chemotherapeutic, an immunotherapeutic and/or a nucleic acid therapeutic agent.
  • the presently disclosed subject matter provides a composition comprising MCL or a derivative and/or nanoparticle thereof as described herein and a pharmaceutically acceptable carrier, e.g., a pharmaceutically acceptable carrier that is pharmaceutically acceptable in humans.
  • a pharmaceutically acceptable carrier e.g., a pharmaceutically acceptable carrier that is pharmaceutically acceptable in humans.
  • the composition can also include other components, such as, but not limited to anti-oxidants, buffers, bacteriostatics, bactericidal antibiotics, suspending agents, thickening agents, and solutes that render the composition isotonic with the bodily fluids of a subject to whom the composition is to be administered.
  • suitable formulations can include aqueous and non-aqueous sterile injection solutions that can contain anti-oxidants, buffers, bacteriostatics, bactericidal antibiotics, and solutes that render the formulation isotonic with the bodily fluids of the subject; and aqueous and non-aqueous sterile suspensions that can include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a frozen or freeze-dried (lyophilized) condition requiring only the addition of sterile liquid carrier, for example water for injections, immediately prior to use.
  • Some exemplary ingredients are sodium dodecyl sulfate (SDS), in one example in the range of 0.1 to 10 mg/ml, in another example about 2.0 mg/ml; and/or mannitol or another sugar, for example in the range of 10 to 100 mg/m I, in another example about 30 mg/m I; and/or phosphate-buffered saline (PBS).
  • SDS sodium dodecyl sulfate
  • PBS phosphate-buffered saline
  • formulations of the presently disclosed subject matter can include other agents conventional in the art having regard to the type of formulation in question.
  • sterile pyrogen-free aqueous and non-aqueous solutions can be used.
  • compositions disclosed herein can be used on a sample either in vitro (for example, on isolated cells or tissues) or in vivo in a subject (i.e. living organism, such as a patient).
  • a subject i.e. living organism, such as a patient.
  • the subject is a human subject, although it is to be understood that the principles of the presently disclosed subject matter indicate that the presently disclosed subject matter is effective with respect to all vertebrate species, including mammals, which are intended to be included in the terms “subject” and “patient”.
  • a mammal is understood to include any mammalian species for which employing the compositions and methods disclosed herein is desirable, particularly agricultural and domestic mammalian species.
  • the methods of the presently disclosed subject matter are particularly useful in warm-blooded vertebrates.
  • the presently disclosed subject matter concerns mammals and birds. More particularly provided are methods and compositions for mammals such as humans, as well as those mammals of importance due to being endangered (such as Siberian tigers), of economic importance (animals raised on farms for consumption by humans), and/or of social importance (animals kept as pets or in zoos) to humans, for instance, carnivores other than humans (such as cats and dogs), swine (pigs, hogs, and wild boars), ruminants (such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels), rodents (e.g., mice, rats, hamsters, gerbils, etc.) and horses.
  • carnivores other than humans such as cats and dogs
  • swine pigs, hogs, and wild boars
  • ruminants such as cattle,
  • poultry such as turkeys, chickens, ducks, geese, guinea fowl, and the like
  • livestock including, but not limited to domesticated swine (pigs and hogs), ruminants, horses, poultry, and the like.
  • Suitable methods for administration of a composition of the presently disclosed subject matter include, but are not limited to intravenous and intratumoral injection, oral administration, subcutaneous administration, intraperitoneal injection, intracranial injection, and rectal administration.
  • a composition can be deposited at a site in need of treatment in any other manner, for example by spraying a composition within the pulmonary pathways.
  • the particular mode of administering a composition of the presently disclosed subject matter depends on various factors, including the distribution and abundance of cells to be treated and mechanisms for metabolism or removal of the composition from its site of administration. For example, relatively superficial tumors can be injected intratumorally. By contrast, internal tumors can be treated following intravenous injection.
  • the method of administration encompasses features for regionalized delivery or accumulation at the site to be treated.
  • a composition is delivered intratumorally.
  • selective delivery of a composition to a target is accomplished by intravenous injection of the composition followed by irradiation (e.g., X-ray irradiation) of the target.
  • compositions of the presently disclosed subject matter can be formulated as an aerosol or coarse spray.
  • Methods for preparation and administration of aerosol or spray formulations can be found, for example, in U.S. Pat. Nos. 5,858,784; 6,013,638; 6,022,737; and 6,136,295.
  • an effective dose of a composition of the presently disclosed subject matter is administered to a subject.
  • An “effective amount” is an amount of the composition sufficient to produce detectable treatment (e.g., tumor size reduction, pain relief, reduction of the concentration of a disease-related blood marker, etc.).
  • Actual dosage levels of constituents of the compositions of the presently disclosed subject matter can be varied so as to administer an amount of the composition that is effective to achieve the desired effect for a particular subject and/or target.
  • the selected dosage level can depend upon the activity (e.g., nanoparticle drug loading and/or the IC 50 of the therapeutic components in certain cell types (e.g., cancer cell lines) of the composition and the route of administration.
  • a dose may be administered on an as needed basis or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, or 24 hours (or any range derivable therein) or 1, 2, 3, 4, 5, 6, 7, 8, 9, or times per day (or any range derivable therein).
  • a dose can be first administered before or after symptoms are exhibited by the subject; after a clinician evaluates the subject for the disease; or before, at about the same time as, or after administration of a second treatment (chemotherapy, radiation therapy, immunotherapy).
  • the patient is administered a first dose of a compound, such as MCL, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 hours (or any range derivable therein) or 1, 2, 3, 4, or 5 days after the subject exhibits signs or symptoms of the disease (or any range derivable therein).
  • a compound such as MCL
  • 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 hours or any range derivable therein
  • the subject can be treated for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more days (or any range derivable therein) or until symptoms of an the disease, such as tumor size, have disappeared or been reduced or after 6, 12, 18, or 24 hours or 1, 2, 3, 4, or 5 days after symptoms of the disease have disappeared or been reduced.
  • the presently disclosed subject matter provides a method for treating a disease (e.g., cancer or another hyperproliferative disease) by using the MCL or derivative and/or nanoparticle thereof and/or pharmaceutical composition thereof in combination with another treatment modality, e.g., a chemotherapeutic, an immunotherapeutic, radiation, surgery, etc.
  • another treatment modality e.g., a chemotherapeutic, an immunotherapeutic, radiation, surgery, etc.
  • the other treatment modality is radiation.
  • the presently disclosed subject matter provides a method of treating a disease in a subject in need thereof, the method comprising: administering to the subject an MCL or derivative and/or nanoparticle thereof and/or pharmaceutical composition thereof as described herein; and irradiating at least a portion of the subject with ionizing radiation, such as with x-rays and/or protons.
  • the method comprises administering a nanoparticle and/or NCP comprising MCL or a derivative thereof.
  • the subject is a mammal.
  • the subject is a human.
  • the patients are irradiated with a linear accelerator (LINAC), using conventional techniques, Intensity-Modulated Radiation Therapy (IMRT), Image Guided Radiation Therapy (IGRT), or Stereotactic Body Radio Therapy (SBRT), a 60 Co radiation source, an implanted radioactive seed such as the ones used in brachytherapy, an orthovoltage or supervoltage X-ray irradiator, a high energy electron beam generated from LINAC, or a proton source.
  • LINAC linear accelerator
  • IMRT Intensity-Modulated Radiation Therapy
  • IGRT Image Guided Radiation Therapy
  • SBRT Stereotactic Body Radio Therapy
  • the ⁇ -rays generated by a LINAC pass through an energy modulator (filter) before irradiating the patient, optionally wherein the filter comprises one or more element(s) with atomic number(s) of at least 20, further optionally wherein the filter comprises copper.
  • the filter has a thickness that is less than 5 mm, less than 4 mm, less than 3 mm, less than 2 mm, less than 1 mm, less than 0.5 mm, less than 0.4 mm, less than 0.3 mm, less than 0.2 mm, less than 0.1 mm.
  • X-rays are generated by placing radioactive sources inside the patient on a temporary or permanent basis.
  • a nanoparticle composition is injected along with the implantation of a radioactive source.
  • the radiation dosage regimen is generally defined in terms of Gray or Sieverts time and fractionation and can be carefully defined by the radiation oncologist.
  • the amount of radiation a subject receives can depend on various considerations, such as the location of the tumor in relation to other critical structures or organs of the body, and the extent to which the tumor has spread.
  • One illustrative course of treatment for a subject undergoing radiation therapy is a treatment schedule with a total dose of 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 Gray (Gy) or any derivable range therein.
  • the radiation dose can be administered to the subject in a single daily fraction of 1.0-2.0 Gy, 1.0-5.0 Gy, or 5.0-10.0 Gy 1, 2, 3, 4, or 5 days a week.
  • a Gy is an abbreviation for Gray and refers to 100 rad of dose.
  • Illustrative dosages used for photon-based radiation are measured in Gy, and in an otherwise healthy subject (that is, little or no other disease states present such as high blood pressure, infection, diabetes, etc.) for a solid epithelial tumor ranges from about 60 to about 80 Gy, and for a lymphoma ranges from about 20 to about 40 Gy.
  • Illustrative preventative (adjuvant) doses are typically given at about 45 to about 60 Gy in about 1.8 to about 2 Gy fractions for breast, head, and neck cancers.
  • the disease is cancer.
  • the disease is selected from the group including, but not limited to, a skin cancer, such as a melanoma; a connective tissue cancer, such as a sarcoma; an adipose cancer; a breast cancer; a head and neck cancer, such as a glioma; a lung cancer, such as mesothelioma; a stomach cancer; a pancreatic cancer; an ovarian cancer; a cervical cancer; a uterine cancer; an anogenital cancer, such as a testicular cancer; a kidney cancer; a bladder cancer; a colon cancer; a prostate cancer; a central nervous system (CNS) cancer; a retinal cancer; a blood cancer, such as leukemia; a neuroblastoma; multiple myeloma, and a lymphoid cancer, such as Hodgkin's or non-Hodgkin's lymphoma.
  • a skin cancer such as a melanom
  • the method can comprise administering the MCL or a derivative and/or nanoparticle thereof to the subject at about the same time as, or later than the administration of the radiation.
  • the MCL or derivative and/or nanoparticle thereof can be administered to the subject prior to a dose or course of radiation.
  • the method can further comprise administering to the subject an additional therapeutic agent or treatment.
  • the additional therapeutic agent or treatment is an immunotherapy agent or a cancer treatment selected from surgery, chemotherapy, toxin therapy, cryotherapy, and gene therapy.
  • the chemotherapy can be co-administered with the MCL or derivative thereof, e.g., in a nanoparticle comprising both MCL or a derivative thereof and a chemotherapeutic agent.
  • the additional therapeutic agent is an immunotherapeutic agent.
  • the additional therapeutic agent is an immune checkpoint inhibitor, such as, but not limited to, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • an immune checkpoint inhibitor such as, but not limited to, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • the additional therapeutic agent or treatment can be administered prior to, at the same time as, or later than the administration of the MCL or derivative and/or nanoparticle thereof.
  • administration of MCL or a derivative thereof alone, or of MCL or a derivative thereof in combination with X-ray irradiation treatment can be performed prior to surgery to reduce the size of a tumor.
  • the presently disclosed subject matter provides a method comprising treating a disease in a subject in need thereof, the method comprising: (i) administering to the subject MCL or a derivative and/or nanoparticle thereof and (ii) administering to the subject a chemotherapeutic agent, an immunotherapy agent, or radiotherapy.
  • the method comprises administering the MCL or derivative thereof and an immunotherapy agent.
  • the method comprises administering to the subject a nanoparticle comprising MCL or a derivative thereof.
  • the subject is a mammal. In some embodiments, the subject is a human.
  • the disease is cancer.
  • the disease is selected from the group including, but not limited to, a skin cancer, such as a melanoma; a connective tissue cancer, such as a sarcoma; an adipose cancer; a breast cancer; a head and neck cancer, such as a glioma; a lung cancer, such as mesothelioma; a stomach cancer; a pancreatic cancer; an ovarian cancer; a cervical cancer; a uterine cancer; an anogenital cancer, such as a testicular cancer; a kidney cancer; a bladder cancer; a colon cancer; a prostate cancer; a central nervous system (CNS) cancer; a retinal cancer; a blood cancer, such as leukemia; a neuroblastoma; multiple myeloma, and a lymphoid cancer, such as Hodgkin's or non-Hodgkin's lymphoma.
  • a skin cancer such as a melanom
  • the immunotherapeutic agent is an immune checkpoint inhibitor, such as, but not limited to, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • the immunotherapeutic agent can be formulated in the same pharmaceutical composition as the MCL or derivative and/or nanoparticle thereof or be part of (e.g., embedded in or attached to) the same nanoparticle as the MCL or derivative thereof.
  • the method further comprises administering to the subject a chemotherapeutic agent.
  • the chemotherapeutic agent can be provided in a nanoparticle, such as a nanoparticle further comprising the MCL or derivative thereof.
  • the chemotherapeutic agent is selected from the group including, but not limited to, a platinum complex, such as cisplatin, oxaliplatin, carboplatin, or a prodrug thereof; doxorubicin; daunorubicin; docetaxel; mitoxanthrone; paclitaxel; digitoxin; gemcitabine; methotrexate; leucovorin; pemetresed disodium; vinblastine; vincristine; vindesine; cytarabine; azathioprine; melphalan; imitnib; anastrozole; letrozole; etoposide; vinorelbine; digoxin, and septacidin.
  • a platinum complex such as cisplatin, o
  • the presently disclosed subject matter provides a method of treating a disease (e.g., cancer) in a subject in need thereof, wherein the method comprises: (i) administering to the subject MCL or a derivative and/or nanoparticle thereof; (ii) administering to the subject an immunotherapeutic agent (e.g., an immune checkpoint inhibitor); and (iii) irradiating at least a portion of the subject with ionizing radiation, such as with x-rays and/or protons.
  • the method can further comprise an additional therapeutic agent or treatment selected from the group including, but not limited to, surgery, toxin therapy, cryotherapy and gene therapy.
  • GL261 cells were seeded onto 96-well plates at a density of 2000 cells/well and treated with various concentrations of the dimethylamino Michael adduct of MCL (dimethylamino micheliolide, DMAMCL) for 24 hours to allow for sufficient cellular uptake and conversion to MCL before irradiation.
  • MCL dimethylamino micheliolide
  • the cells were irradiated with varying amounts of X-ray irradiation and then incubated for an additional 24 hours.
  • the culture medium was exchanged with fresh medium and further incubated for an additional 48 hours.
  • the cell viability was then detected by (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay (Promega, Madison, Wis., United States of America).
  • MTS 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)
  • mice were orally dosed with 50 mg/kg DMAMCL or PBS daily and irradiated with 4 Gy irradiation approximately 4 hrs after dosing, as applicable, for up to 7 days, starting when the tumors reached about 2-300 mm 3 .
  • a Precision X-ray X-RAD 225 Image Guided Biologic Irradiator (Precision X-Ray Irradiation, North Branford, Conn., United States of America) was used for all animal studies.
  • the instrument was set at 225 peak kilovoltage (kVp) and 13 milliampere (mA), with a 0.3 mm flat-board copper (Cu) filter installed before a 15-mm collimator.
  • kVp peak kilovoltage
  • mA milliampere
  • Cu flat-board copper
  • Fractionated irradiation allows for higher total doses of X-ray but showed little or no appreciable effects on tumor growth.
  • DMAMCL alone initially suppressed tumor growth, the tumors were ultimately not significantly different in size compared to those treated with irradiation alone. These results are consistent with in vitro results which showed varied effects leading to high average cell viability with large deviation in cells and tumors treated with irradiation alone.
  • the combination of DMAMCL and irradiation successfully controlled tumor growth leading to relatively stable tumor sizes over the course of treatment.
  • the combination of DMAMCL, irradiation, and PD-L1 antibody was evaluated on the glioma GL261 tumor model on wildtype C57BL/6 mice. See FIG. 3 .
  • the treatment began 14 days after the inoculation of 2 ⁇ 10 6 GL261 cells.
  • the mice were orally dosed with 50 mg/kg DMAMCL or PBS daily and irradiated with 2 Gy irradiation approximately 4 hrs after dosing, as applicable, for up to 12 days, starting when the tumors reached ⁇ 150-200 mm 3 .
  • One group of mice received two doses of 75 ⁇ g PD-L1 antibody on days 0 and 4.
  • DMAMCL alone showed no effect on tumor growth, but combination with irradiation suppressed tumor growth and ultimately led to tumor regression.
  • the combination of DMAMCL, irradiation, and a PD-L1 antibody controlled and regressed tumor growth immediately after treatment. By the cessation of treatment, 3/5 tumors treated with this combination were barely palpable.
  • DMAMCL showed synergy with irradiation to control and regress GL261 tumors, which was further enhanced by combination with an immune checkpoint inhibitor.
  • Micheliolide and its derivatives can be formulated into nanoscale coordination polymers (NCPs) for use in treating multiple cancer types in combination with radiotherapy.
  • NCPs nanoscale coordination polymers
  • Exemplary methods of preparing and using NCPs are described, for example in PCT International Patent Application Publication Nos. WO 2015/069926 and WO 2013/009701, U.S. Pat. No. 9,693,957, and U.S. Patent Application Publication No. 2016/0346204, the disclosures of each of which are incorporated herein by reference in their entireties.
  • MCL and its derivatives were loaded directly or as lipid-conjugates onto NCPs.
  • Equation 1 (Eq 1) of Scheme 1, below micheliolide (500 mg, 2 mmol) was dissolved in 20 mL anhydrous dichloromethane (DCM). 2.5 mL dimethylamine 2M solution in tetrahydrofuran (THF) was added to the solution. The mixture was stirred at room temperature for 8 hours and then solvent was removed by rotary evaporation. The solid residue was purified by column chromatography using 5% methanol in DCM to obtain pure DMAMCL.
  • DCM anhydrous dichloromethane
  • THF tetrahydrofuran
  • OxNCP/MCL was obtained by adding a 804 THF solution of DOPC, cholesterol, MCL (molar ratio 3.5:2:3), DSPE-PEG2k (20 mol %) and OxNCP bare particles to 500 ⁇ L 30% (v/v) EtOH/H 2 O at 50° C. THF and EtOH were evaporated and the solution was allowed to cool to room temperature before use. The particle size and distribution were determined by dynamic light scattering (DLS). See FIG. 4 . OxNCP/MCL has a Z-average diameter of 76 nm.

Abstract

Nanoparticles comprising micheliolide (MCL) or derivatives thereof, and optionally additional therapeutic agents, such as chemotherapeutic agents, are described. Also described are methods of treating diseases, such as cancer, comprising the use of combinations of MCL or a derivative thereof with X-ray irradiation and/or other therapeutic agents, such as immune checkpoint inhibitors. The use of the combinations can provide synergistic anticancer therapeutic efficacy, for example, as the MCL or derivative thereof can both sensitize cancer cells to therapy and target resistant cancer stem cells (CSCs) for selective cell death.

Description

    RELATED APPLICATIONS
  • The presently disclosed subject matter claims the benefit of U.S. Provisional Patent Application Ser. No. 62/657,112, filed Apr. 13, 2018, the disclosure of which is incorporated herein by reference in its entirety.
  • STATEMENT OF GOVERNMENT SUPPORT
  • This invention was made with government support under Grant No. U01-CA198989 awarded by the National Institutes of Health. The government has certain rights in the invention.
  • TECHNICAL FIELD
  • The presently disclosed subject matter relates to nanoparticles (NPs) comprising micheliolide (MCL) or a derivative thereof, as well as to the use of combinations of MCL or a derivative and/or NPs thereof in combination with X-ray irradiation and/or other therapeutic agents, e.g., immune checkpoint inhibitors, for treating disease. In some embodiments, the combinations can provide synergistic anticancer therapeutic efficacy.
  • ABBREVIATIONS
      • ° C.=degrees Celsius
      • percentage
      • microgram
      • micromolar
      • cm=centimeter
      • CSCs=cancer stem cells
      • DLS=dynamic light scattering
      • DOPC=1,2-dioleoyl-sn-glycero-3-phosphocholine
      • DSPE-PEG2k=1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)2000]
      • DMAMCL=dimethylamino micheliolide
      • ESI-MS=electrospray ionization mass spectrometry
      • EtOH=ethanol
      • Gy=gray
      • Kg=kilogram
      • kVp=peak kilovoltage
      • mA=milliampere
      • MCL=micheliolide
      • mg=milligram
      • mL=milliliter
      • mm=millimeter
      • mmol=millimole
      • MOF=metal-organic framework
      • NCP=nanoscale coordination polymer
      • nm=nanometer
      • nMOF=nanoscale metal-organic framework
      • NMR=nuclear magnetic resonance
      • NPs=nanoparticles
      • Ox=oxaliplatin
      • PD-L1=programmed death ligand 1
      • PEG=poly(ethylene glycol)
      • PBS=phosphate buffered saline
      • SBU=secondary building unit
      • THF=tetrahydrofuran
    BACKGROUND
  • Chemotherapy and radiotherapy are among the powerful anticancer therapies used across a multitude of cancer types. However, the effective use of these therapies can be limited due to undesirable side effects to healthy cells at high dosages. In addition, these therapies are often compromised by the development of drug and radio-resistance by tumor cells.
  • Accordingly, there is an ongoing need for additional cancer treatment methods and compositions, such as those with enhanced anticancer efficacy. For instance, there is an ongoing need for additional cancer treatment methods and compositions that can be used to treat drug- and/or radio-resistant cancers and/or that can be used to treat cancer while avoiding the development of drug- and/or radio-resistant cancer cells.
  • SUMMARY
  • In some embodiments, the presently disclosed subject matter provides a composition comprising a nanoparticle and micheliolide (MCL) or a derivative thereof. In some embodiments, the composition comprises a derivative of MCL, wherein said derivative is dimethylamino micheliolide (DMAMCL) or a lipid conjugate of MCL.
  • In some embodiments, the MCL or the derivative thereof is attached to or sequestered within the nanoparticle. In some embodiments, the nanoparticle is selected from the group comprising a polymeric micelle, a liposome, a dendrimer, an organic polymer-based nanoparticle, a silica-based nanoparticle, a nanoscale coordination polymer, a nanoscale metal-organic framework, and an inorganic nanoparticle.
  • In some embodiments, the nanoparticle comprises a core and one or more coating layers or agents covering at least a portion of an outer surface of the core. In some embodiments, the core comprises an organic polymer, a silica-based material, a nanoscale coordination polymer, a metal-organic framework, or an inorganic material. In some embodiments, the one or more coating agents or layers are selected from a metal oxide, a polymer, a single lipid layer, a lipid bilayer, and combinations thereof. In some embodiments, the one or more coating agents or layers further comprise a passivating agent, a targeting agent, and/or an imaging agent.
  • In some embodiments, the MCL or derivative thereof is attached to or sequestered in the nanoparticle core. In some embodiments, the MCL or the derivative thereof is attached to or sequestered in a nanoparticle coating layer.
  • In some embodiments, the composition further comprises a chemotherapeutic agent and/or an immunotherapy agent. In some embodiments, the chemotherapeutic agent is attached or sequestered in a nanoparticle core or a nanoparticle coating layer. In some embodiments, the chemotherapeutic agent is selected from the group comprising cisplatin, oxaliplatin, or prodrugs thereof; doxorubicin, daunorubicin, docetaxel, mitoxanthrone, paclitaxel, digitoxin, gemcitabine, methotrexate, leucovorin, pemetresed disodium, vinblastine, vincristine, vindesine, cytarabine, azathioprine, melphalan, imitinib, anastrozole, letrozole, carboplatin, etoposide, vinorelbine, and septacidin. In some embodiments, the immunotherapy agent is selected from the group comprising an anti-CD52 antibody, an anti-CD20 antibody, anti-CD47 antibody, an anti-GD2 antibody, polysaccharide K, a cytokine, Alemtuzumab, Ofatumumab, Rituximab, Zevalin, Adcetris, Kadcyla, Ontak, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor. In some embodiments, the immunotherapy agent is attached to or sequestered in a nanoparticle coating layer.
  • In some embodiments, the presently disclosed subject matter provides a pharmaceutical composition comprising a composition comprising a nanoparticle and micheliolide (MCL) or a derivative thereof and a pharmaceutically acceptable carrier.
  • In some embodiments, the presently disclosed subject matter provides a method for treating a disease in a subject in need thereof, the method comprising: administering to the subject micheliolide (MCL) or a derivative thereof; and exposing at least a portion of the subject to ionizing irradiation energy. In some embodiments, administering the MCL or derivative thereof comprises administering a nanoparticle and/or a nanoscale coordination polymer (NCP) comprising the MCL or derivative thereof. In some embodiments, the ionizing irradiation energy is X-rays or protons.
  • In some embodiments, the disease is cancer. In some embodiments, the disease is selected from the group comprising a skin cancer, a connective tissue cancer, an adipose cancer, a breast cancer, a head and neck cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system (CNS) cancer, a retinal cancer, a blood cancer, a neuroblastoma, multiple myeloma, and a lymphoid cancer.
  • In some embodiments, the method further comprises administering to the subject an additional therapeutic agent or treatment selected from an immunotherapy agent and/or a cancer treatment, wherein the cancer treatment is selected from the group comprising surgery, chemotherapy, toxin therapy, cryotherapy and gene therapy. In some embodiments, the additional therapeutic agent or treatment comprises an immunotherapy agent. In some embodiments, the immunotherapy agent is an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibitor is selected from the group comprising a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • In some embodiments, the presently disclosed subject matter provides a method for treating a disease in a subject in need thereof, the method comprising: administering to the subject micheliolide (MCL) or a derivative thereof; and administering to the subject one or more additional therapeutic agent or treatment. In some embodiments, administering to the subject MCL or a derivative thereof comprises administering to the subject a nanoparticle comprising MCL or a derivative thereof. In some embodiments, the one or more additional therapeutic agent or treatment agent is selected from a chemotherapeutic agent, an immune therapy agent, radiotherapy, surgery, chemotherapy, toxin therapy, cryotherapy and gene therapy.
  • In some embodiments, the disease is cancer. In some embodiments, the disease is selected from the group comprising a skin cancer, a connective tissue cancer, an adipose cancer, a breast cancer, a head and neck cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system (CNS) cancer, a retinal cancer, a blood cancer, a neuroblastoma, multiple myeloma, and a lymphoid cancer.
  • In some embodiments, the one or more additional therapeutic agent or treatment comprises an immunotherapeutic agent. In some embodiments, the immunotherapeutic agent is an immune checkpoint inhibitor.
  • In some embodiments, the one or more additional therapeutic agent or treatment further comprises radiotherapy. In some embodiments, the one or more additional therapeutic agent or treatment agent further comprises one or more of surgery, toxin therapy, cryotherapy and gene therapy. In some embodiments, the one or more additional treatment agent comprises chemotherapy.
  • Accordingly, it is an object of the presently disclosed subject matter to provide compositions comprising MCL or a derivative thereof in a nanoparticle and/or in combination with another therapeutic agent, e.g., a chemotherapeutic agent or an immunotherapeutic agent, as well as methods of treating disease using the compositions, alone or in combination with X-ray irradiation and/or another therapy, e.g., immunotherapy.
  • These and other objects are achieved in whole or in part in the presently disclosed subject matter. An object of the presently disclosed subject matter having been stated above, other objects and advantages will become apparent upon a review of the following descriptions, examples, and figures.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing the in vitro cytotoxicity of combinations of dimethylamino micheliolide (DMAMCL) and irradiation in mouse glioma 261 (GL261) cells. The cytotoxic effect of the combination is represented by a decrease in cell viability (in percentage (5%)). The cytotoxicity was measured at two different concentrations of DMAMCL: 1 micromolar (1 μM, open triangles) and 10 μM (open circles). The irradiation dose varied from 0 to 8 gray (Gy) as indicated on the x-axis. For comparison, cell viability data for GL261 cells treated with irradiation and phosphate buffered saline (PBS, filled squares) in place of the DMAMCL is also shown.
  • FIG. 2 is a graph showing the in vivo antitumor efficacy of combinations of dimethylamino micheliolide (DMAMCL) and irradiation in a mouse glioma 261 (GL261) tumor model in immunocompromised mice. Mice were orally dosed with 50 milligrams per kilogram (mg/kg) DMAMCL or with phosphate buffered saline and irradiated or not irradiated with a 4 gray (Gy) irradiation dose about 4 hours later every day for up to 7 days. Data for mice treated with PBS and not irradiated (PBS) is shown in open circles, data for mice treated with PBS and radiation (PBS (+)) is shown in filled circles, data for mice treated with DMAMCL and not irradiated is shown in open squares, and data for mice treated with DMAMCL and radiation (DMAMCL (+)) is shown in filled squares. Tumor size was measured in cubic millimeters (mm3).
  • FIG. 3 is a graph showing the in vivo antitumor efficacy of combinations of micheliolide (MCL), irradiation, and an immunotherapy agent in a mouse glioma 261 (GL261) tumor model in immunocompetent mice. Mice were orally dosed with 50 milligrams per kilogram (mg/kg) MCL or phosphate buffered saline daily for up to 12 days. Some of the mice were irradiated with a 2 gray (Gy) irradiation dose about 4 hours after dosing with MCL. Some of the mice also received two 75 microgram (μg) doses of a programmed death ligand 1 (PD-L1) antibody on days 0 and 4. Data for mice treated with PBS and not irradiated (PBS) is shown in filled squares, data for mice treated with MCL and not irradiated is shown in open circles, data for mice treated with MCL and radiation (MCL (+)) is shown in open triangles, and data for mice treated with MCL, irradiation and antibody (MCL+α-PD-L1 (+)) is shown in open stars. Tumor size was measured in cubic centimeters (cm3).
  • FIG. 4 is a graph showing the intensity plot (intensity (in percentage (%)) versus size (diameter in nanometers (d.nm))) of oxaliplatin-containing nano-coordination polymers (OxNCP) also comprising micheliolide (MCL) as measured using dynamic light scattering (DLS).
  • DETAILED DESCRIPTION
  • The presently disclosed subject matter will now be described more fully hereinafter with reference to the accompanying Examples, in which representative embodiments are shown. The presently disclosed subject matter can, however, be embodied in different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the embodiments to those skilled in the art.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this presently described subject matter belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the presently disclosed subject matter, representative methods, devices, and materials are now described. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. Throughout the specification and claims, a given chemical formula or name shall encompass all optical and stereoisomers, as well as racemic mixtures where such isomers and mixtures exist.
  • I. Definitions
  • While the following terms are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter.
  • Following long-standing patent law convention, the terms “a”, “an”, and “the” refer to “one or more” when used in this application, including the claims. Thus, for example, reference to “a chemotherapeutic agent” includes a plurality of such chemotherapeutic agents, and so forth.
  • Unless otherwise indicated, all numbers expressing quantities of size, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by the presently disclosed subject matter.
  • As used herein, the term “about”, when referring to a value or to an amount of size (i.e., diameter), dose, weight, concentration, or percentage is meant to encompass variations of in one example ±20% or ±10%, in another example ±5%, in another example ±1%, and in still another example ±0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods.
  • Numerical ranges recited herein by endpoints include all numbers and fractions subsumed within that range (e.g. 1 to 5 includes, but is not limited to, 1, 1.5, 2, 2.75, 3, 3.90, 4, and 5).
  • As used herein, the term “and/or” when used in the context of a listing of entities, refers to the entities being present singly or in combination. Thus, for example, the phrase “A, B, C, and/or D” includes A, B, C, and D individually, but also includes any and all combinations and sub-combinations of A, B, C, and D.
  • The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and “and/or.” As used herein “another” can mean at least a second or more.
  • The term “comprising”, which is synonymous with “including,” “containing,” or “characterized by” is inclusive or open-ended and does not exclude additional, unrecited elements or method steps. “Comprising” is a term of art used in claim language which means that the named elements are present, but other elements can be added and still form a construct or method within the scope of the claim.
  • As used herein, the phrase “consisting of” excludes any element, step, or ingredient not specified in the claim. When the phrase “consists of” appears in a clause of the body of a claim, rather than immediately following the preamble, it limits only the element set forth in that clause; other elements are not excluded from the claim as a whole.
  • As used herein, the phrase “consisting essentially of” limits the scope of a claim to the specified materials or steps, plus those that do not materially affect the basic and novel characteristic(s) of the claimed subject matter.
  • With respect to the terms “comprising”, “consisting of”, and “consisting essentially of”, where one of these three terms is used herein, the presently disclosed and claimed subject matter can include the use of either of the other two terms.
  • As used herein the term “alkyl” can refer to C1-20 inclusive, linear (i.e., “straight-chain”), branched, or cyclic, saturated or at least partially and in some cases fully unsaturated (i.e., alkenyl and alkynyl) hydrocarbon chains, including for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl, octyl, ethenyl, propenyl, butenyl, pentenyl, hexenyl, octenyl, butadienyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, and allenyl groups. “Branched” refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain. “Lower alkyl” refers to an alkyl group having 1 to about 8 carbon atoms (i.e., a C1-8 alkyl), e.g., 1, 2, 3, 4, 5, 6, 7, or 8 carbon atoms. “Higher alkyl” refers to an alkyl group having about 10 to about 20 carbon atoms, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms. In certain embodiments, “alkyl” refers, in particular, to C1-8 straight-chain alkyls. In other embodiments, “alkyl” refers, in particular, to C1-8 branched-chain alkyls.
  • Alkyl groups can optionally be substituted (a “substituted alkyl”) with one or more alkyl group substituents, which can be the same or different. The term “alkyl group substituent” includes but is not limited to alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl. In some embodiments, there can be optionally inserted along the alkyl chain one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is hydrogen, lower alkyl (also referred to herein as “alkylaminoalkyl”), or aryl.
  • Thus, as used herein, the term “substituted alkyl” includes alkyl groups, as defined herein, in which one or more atoms or functional groups of the alkyl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • “Alkylene” refers to a straight or branched bivalent aliphatic hydrocarbon group having from 1 to about 20 carbon atoms, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms. The alkylene group can be straight, branched or cyclic. The alkylene group also can be optionally unsaturated and/or substituted with one or more “alkyl group substituents.” There can be optionally inserted along the alkylene group one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms (also referred to herein as “alkylaminoalkyl”), wherein the nitrogen substituent is alkyl as previously described. Exemplary alkylene groups include methylene (—CH2—); ethylene (—CH2—CH2—); propylene (—(CH2)3—); cyclohexylene (—C6H10—); —CH═CH—CH═CH—; —CH═CH—CH2—; —(CH2)q—N(R)—(CH2)r—, wherein each of q and r is independently an integer from 0 to about 20, e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and R is hydrogen or lower alkyl; methylenedioxyl (—O—CH2—O—); and ethylenedioxyl (—O—(CH2)2—O—). An alkylene group can have about 2 to about 3 carbon atoms and can further have 6-20 carbons.
  • The term “aryl” is used herein to refer to an aromatic substituent that can be a single aromatic ring, or multiple aromatic rings that are fused together, linked covalently, or linked to a common group, such as, but not limited to, a methylene or ethylene moiety. The common linking group also can be a carbonyl, as in benzophenone, or oxygen, as in diphenylether, or nitrogen, as in diphenylamine. The term “aryl” specifically encompasses heterocyclic aromatic compounds. The aromatic ring(s) can comprise phenyl, naphthyl, biphenyl, diphenylether, diphenylamine and benzophenone, among others. In particular embodiments, the term “aryl” means a cyclic aromatic comprising about 5 to about 10 carbon atoms, e.g., 5, 6, 7, 8, 9, or 10 carbon atoms, and including 5- and 6-membered hydrocarbon and heterocyclic aromatic rings.
  • The aryl group can be optionally substituted (a “substituted aryl”) with one or more aryl group substituents, which can be the same or different, wherein “aryl group substituent” includes alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, hydroxyl, alkoxyl, aryloxyl, aralkyloxyl, carboxyl, acyl, halo, nitro, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, acyloxyl, acylamino, aroylamino, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, arylthio, alkylthio, alkylene, and —NR′R″, wherein R′ and R″ can each be independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, and aralkyl.
  • Thus, as used herein, the term “substituted aryl” includes aryl groups, as defined herein, in which one or more atoms or functional groups of the aryl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • Specific examples of aryl groups include, but are not limited to, cyclopentadienyl, phenyl, furan, thiophene, pyrrole, pyran, pyridine, imidazole, benzimidazole, isothiazole, isoxazole, pyrazole, pyrazine, triazine, pyrimidine, quinoline, isoquinoline, indole, carbazole, and the like.
  • The term “arylene” refers to a bivalent aromatic group, e.g., a bivalent phenyl or naphthyl group. The arylene group can optionally be substituted with one or more aryl group substituents and/or include one or more heteroatoms. The term “arylene” includes bivalent groups comprising more than one aromatic ring fused to or linked to one another (e.g., via a covalent bond or a linking group), such as bivalent biphenyl or bivalent terphenyl.
  • The term “aralkyl” as used herein refers to a -alkyl-aryl group, which can be substituted or unsubstituted. An exemplary aralkyl group is benzyl.
  • The term “acyl” refers to the group —C(═O)R, wherein R is a substituted or unsubstituted alkyl, aralkyl, or aryl group.
  • The term “amino” refers to the group —N(R)2 wherein each R is independently H, alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, or substituted aralkyl. The terms “aminoalkyl” and “alkylamino” can refer to the group —N(R)2 wherein each R is H, alkyl or substituted alkyl, and wherein at least one R is alkyl or substituted alkyl. “Arylamine” and “aminoaryl” refer to the group —N(R)2 wherein each R is H, aryl, or substituted aryl, and wherein at least one R is aryl or substituted aryl, e.g., aniline (i.e., —NHC6H5).
  • The term “lipid” can refer to a hydrophobic or amphiphilic small molecule, such as, but not limited to a fatty acid, a sterol, a fat-soluble vitamin, a phospholipid, a glycerolipid, a glycerophospholipid, a sphingolipid, a saccharolipid, or a polyketide.
  • As used herein, the term “metal-organic framework” or “MOF” refers to a solid two- or three-dimensional network comprising both metal and organic components, wherein the organic components include at least one, and typically more than one carbon atom. In some embodiments, the material is crystalline. In some embodiments, the material is amorphous. In some embodiments, the material is porous. In some embodiments, the metal-organic matrix material is a coordination polymer, which comprises repeating units of coordination complexes comprising a metal-based secondary building unit (SBU), such as a metal ion or metal complex, and a bridging polydentate (e.g., bidentate or tridentate) organic ligand. In some embodiments, the material contains more than one type of SBU or metal ion. In some embodiments, the material can contain more than one type of organic bridging ligand.
  • The term “nanoscale metal-organic framework” can refer to a nanoscale particle comprising an MOF.
  • The terms “nanoscale particle,” nanomaterial,” and “nanoparticle” refer to a structure having at least one region with a dimension (e.g., length, width, diameter, etc.) of less than about 1,000 nm. In some embodiments, the dimension is smaller (e.g., less than about 500 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 125 nm, less than about 100 nm, less than about 80 nm, less than about 70 nm, less than about 60 nm, less than about 50 nm, less than about 40 nm, less than about 30 nm or even less than about 20 nm). In some embodiments, the dimension is between about 20 nm and about 250 nm (e.g., about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 nm).
  • In some embodiments, the nanoparticle is approximately spherical. When the nanoparticle is approximately spherical, the characteristic dimension can correspond to the diameter of the sphere. In addition to spherical shapes, the nanomaterial can be disc-shaped, plate-shaped (e.g., hexagonally plate-like), oblong, polyhedral, rod-shaped, cubic, or irregularly-shaped.
  • The nanoparticle can comprise a core region (i.e., the space between the outer dimensions of the particle) and an outer surface (i.e., the surface that defines the outer dimensions of the particle). In some embodiments, the nanoparticle can have one or more coating layers surrounding or partially surrounding the nanoparticle core. Thus, for example, a spherical nanoparticle can have one or more concentric coating layers, each successive layer being dispersed over the outer surface of a layer closer to the center of the particle.
  • In some embodiments, the presently disclosed nanoparticle can comprise a solid metal-organic framework matrix, which can comprise one or more pores or hollow interior regions. The matrix can be amorphous or crystalline. In some embodiments, the nanoparticle core can further comprise one or more of: MCL or a derivative thereof; an optical imaging agent; and/or another therapeutic agent (e.g., an anticancer agent or an immunotherapeutic agent), which can be physically trapped within the matrix, coordinated to a metal ion of the matrix, or chemically bonded (e.g., to an organic bridging ligand in the matrix) via a covalent or ionic bond. In some embodiments, a chemotherapeutic agent or prodrug thereof can be an organic bridging ligand within a metal-organic matrix material that forms the core of the nanoparticle. For example, when the matrix material is a metal bisphosphonate coordination polymer, the bisphosphonate can be a chemotherapeutic agent or prodrug thereof.
  • When the core comprises a non-matrix therapeutic and/or imaging agent, said agents can be said to be “embedded” in the nanoparticle. “Embedded” can refer to a therapeutic agent or an imaging agent that is bound, for example covalently bound or bound via a coordinative bond, inside the core of the particle (e.g., to a bisphosphonate, dicarboxylate, or metal ion of the matrix material). Alternatively, the complex or agent can be “sequestered” (i.e., non-covalently/physically encapsulated) inside pores in the core or interact with a core material via hydrogen bonding, London dispersion forces, or any other non-covalent interaction.
  • The terms “polymer” and “polymeric” refer to chemical structures that have repeating units (i.e., multiple copies of a given chemical substructure). Polymers can be formed from polymerizable monomers. A polymerizable monomer is a molecule that comprises one or more moieties that can react to form bonds (e.g., covalent or coordination bonds) with moieties on other molecules of polymerizable monomer. Generally, each polymerizable monomer molecule can bond to two or more other molecules. In some cases, a polymerizable monomer will bond to only one other molecule, forming a terminus of the polymeric material.
  • Polymers can be organic, or inorganic, or a combination thereof. As used herein, the term “inorganic” refers to a compound or composition that contains at least some atoms other than carbon, hydrogen, nitrogen, oxygen, sulfur, phosphorous, or one of the halides. Thus, for example, an inorganic compound or composition can contain one or more silicon atoms and/or one or more metal atoms.
  • As used herein “organic polymers” are those that do not include silica or metal atoms in their repeating units. Exemplary organic polymers include polyvinylpyrrolidone (PVO), polyesters, polyamides, polyethers, polydienes, and the like. Some organic polymers contain biodegradable linkages, such as esters or amides, such that they can degrade over time under biological conditions.
  • The term “hydrophilic polymer” as used herein generally refers to hydrophilic organic polymers, such as but not limited to, polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethyacrylamide, polydimethylacrylamide, polyhydroxylpropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethylenimine (PEI), polyethyleneglycol (i.e., PEG) or another hydrophilic poly(alkyleneoxide), polyglycerine, and polyaspartamide. The term “hydrophilic” refers to the ability of a molecule or chemical species to interact with water. Thus, hydrophilic polymers are typically polar or have groups that can hydrogen bond to water.
  • The terms “bonding” or “bonded” and variations thereof can refer to either covalent or non-covalent bonding. In some cases, the term “bonding” refers to bonding via a coordinate bond. The term “conjugation” can refer to a bonding process, as well, such as the formation of a covalent linkage or a coordinate bond.
  • A “coordination complex” is a compound in which there is a coordinate bond between a metal ion and an electron pair donor, ligand or chelating group. Thus, ligands or chelating groups are generally electron pair donors, molecules or molecular ions having unshared electron pairs available for donation to a metal ion.
  • The term “coordinate bond” refers to an interaction between an electron pair donor and a coordination site on a metal ion resulting in an attractive force between the electron pair donor and the metal ion. The use of this term is not intended to be limiting, in so much as certain coordinate bonds also can be classified as having more or less covalent character (if not entirely covalent character) depending on the characteristics of the metal ion and the electron pair donor.
  • As used herein, the term “ligand” refers generally to a species, such as a molecule or ion, which interacts, e.g., binds, in some way with another species. More particularly, as used herein, a “ligand” can refer to a molecule or ion that binds a metal ion in solution to form a “coordination complex.” See Martell, A. E., and Hancock, R. D., Metal Complexes in Aqueous Solutions, Plenum: New York (1996), which is incorporated herein by reference in its entirety. The terms “ligand” and “chelating group” can be used interchangeably. The term “bridging ligand” can refer to a group that bonds to more than one metal ion or complex, thus providing a “bridge” between the metal ions or complexes. Organic bridging ligands can have two or more groups with unshared electron pairs separated by, for example, an alkylene or arylene group. Groups with unshared electron pairs, include, but are not limited to, —CO2H, —NO2, amino, hydroxyl, thio, thioalkyl, —B(OH)2, —SO3H, POSH, phosphonate, and heteroatoms (e.g., nitrogen, oxygen, or sulfur) in heterocycles.
  • The term “coordination site” when used herein with regard to a ligand, e.g., a bridging ligand, refers to an unshared electron pair, a negative charge, or atoms or functional groups cable of forming an unshared electron pair or negative charge (e.g., via deprotonation under at a particular pH).
  • The term “imaging agent” refers to a chemical moiety that aids in the visualization of a sample. For example, an imaging agent can be a “contrast agent” and can refer to a moiety (a specific part of or an entire molecule, macromolecule, coordination complex, or nanoparticle) that increases the contrast of a biological tissue or structure being examined. The contrast agent can increase the contrast of a structure being examined using, for example, magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) imaging, single photon emission computed tomography (SPECT) imaging, or a combination thereof (i.e., the contrast agent can be multimodal).
  • The term “MRI contrast agent” refers to a moiety that effects a change in induced relaxation rates of water protons in a sample.
  • The terms “optical imaging agent” or “optical contrast agent” refer to a group that can be detected based upon an ability to absorb, reflect or emit light (e.g., ultraviolet, visible, or infrared light). Optical imaging agents can be detected based on a change in amount of absorbance, reflectance, or fluorescence, or a change in the number of absorbance peaks or their wavelength maxima. Thus, optical imaging agents include those which can be detected based on fluorescence or luminescence, including organic and inorganic dyes.
  • The terms “fluorophore” and “fluorescent moiety” refer to species that can be excited by visible light or non-visible light (e.g., UV light). Examples of fluorophores include, but are not limited to: quantum dots and doped quantum dots (e.g., a semiconducting CdSe quantum dot or a Mn-doped CdSe quantum dot), fluorescein, fluorescein derivatives and analogues, indocyanine green, rhodamine, triphenylmethines, polymethines, cyanines, phalocyanines, naphthocyanines, merocyanines, lanthanide complexes or cryptates, fullerenes, oxatellurazoles, LaJolla blue, porphyrins and porphyrin analogues and natural chromophores/fluorophores such as chlorophyll, carotenoids, flavonoids, bilins, phytochrome, phycobilins, phycoerythrin, phycocyanines, retinoic acid and analogues such as retinoins and retinates.
  • The term “cancer” as used herein refers to diseases caused by uncontrolled cell division and/or the ability of cells to metastasize, or to establish new growth in additional sites. The terms “malignant”, “malignancy”, “neoplasm”, “tumor,” “cancer” and variations thereof refer to cancerous cells or groups of cancerous cells.
  • Particular types of cancer include, but are not limited to, skin cancers (e.g., melanoma), connective tissue cancers (e.g., sarcomas), adipose cancers, breast cancers, head and neck cancers, lung cancers (e.g., mesothelioma), stomach cancers, pancreatic cancers, ovarian cancers, cervical cancers, uterine cancers, anogenital cancers (e.g., testicular cancer), kidney cancers, bladder cancers, colon cancers, prostate cancers, central nervous system (CNS) cancers, retinal cancer, blood, neuroblastomas, multiple myeloma, and lymphoid cancers (e.g., Hodgkin's and non-Hodgkin's lymphomas).
  • The term “metastatic cancer” refers to cancer that has spread from its initial site (i.e., the primary site) in a patient's body.
  • The terms “anticancer drug”, “chemotherapeutic”, and “anticancer prodrug” refer to drugs (i.e., chemical compounds) or prodrugs known to, or suspected of being able to treat a cancer (i.e., to kill cancer cells, prohibit proliferation of cancer cells, or treat a symptom related to cancer). In some embodiments, the term “chemotherapeutic” as used herein refers to a non-MCL-based molecule that is used to treat cancer and/or that has cytotoxic ability. Some traditional or conventional chemotherapeutic agents can be described by mechanism of action or by chemical compound class, and can include, but are not limited to, alkylating agents (e.g., melphalan), anthracyclines (e.g., doxorubicin), cytoskeletal disruptors (e.g., paclitaxel), epothilones, histone deacetylase inhibitors (e.g., vorinostat), inhibitors of topoisomerase I or II (e.g., irinotecan or etoposide), kinase inhibitors (e.g., bortezomib), nucleotide analogs or precursors thereof (e.g., methotrexate), peptide antibiotics (e.g., bleomycin), platinum based agents (e.g., cisplatin or oxaliplatin), retinoids (e.g., tretinoin), and vinka alkaloids (e.g., vinblastine).
  • II. General Considerations
  • Water-soluble micheliolide (MCL) is a derivative of a natural sesquiterpene lactone, compounds commonly used to treat inflammation. The presently disclosed subject matter is related to the finding that MCL and related compounds can synergize with other disease treatments, particularly cancer treatments, such as chemotherapy and radiotherapy. Without being bound by any one theory, it is believed that the synergistic effects with regard to treating cancer can be related to the activity of the MCL or a related compound in sensitizing cancer cells to therapy and/or targeting resistant CSCs for selective cell death. In some embodiments, the combined efficacy of MCL and related compounds with the other treatment (e.g., with radiation) can be further enhanced by immune checkpoint inhibitors. In some embodiments, nanoparticles (NPs) containing MCL or a derivative thereof, and optionally one or more additional therapeutic agents (e.g., chemotherapeutic agents and/or immunotherapy agents), can be used to enhance the bioavailability and relative tumor deposition of MCL, its derivative and/or other agents, allowing for lower total doses and less frequent dosing.
  • Accordingly, in some embodiments, the presently disclosed subject matter relates to the use of MCL NPs, as well as to combinations of MCL or a derivative thereof and/or NPs with X-ray irradiation and/or other therapeutic agents, e.g., immune checkpoint inhibitors, for treating disease (e.g., cancer). In some embodiments, the combinations can provide synergistic anticancer therapeutic efficacy.
  • III. Compositions
  • In some embodiments, the presently disclosed subject matter provides a composition comprising: (i) a nanoparticle and (ii) MCL or a derivative thereof. MCL has the following structure:
  • Figure US20190314324A1-20191017-C00001
  • The cyclization of parthenolide, a sesquiterpene lactone found in feverfew (Tanacetum parthenium), to form MCL was described in 1993. See Castañeda-Acosta et al., J. Nat. Prod. 1993, 56(1), 90-98. Exemplary derivatives of MCL include, but are not limited to, the compounds described in U.S. Pat. No. 9,255,078, the disclosure of which is incorporated herein by reference in its entirety. For example, in some embodiments, the derivative of MCL comprises a compound wherein a carbon-carbon double bond has been replaced by a carbon-carbon single bond or a three-membered ring (e.g., an epoxide) and/or wherein one of the carbon atoms of the carbon-carbon double bond has been substituted, e.g., by a substituted or unsubstituted alkyl group; wherein an oxygen atom has been replaced by —CH2— or a —NR′— group, wherein R′ is an alkyl or substituted alkyl group; or wherein the hydroxyl group has been substituted by an alkyl, substituted alkyl, or acyl group. In some embodiments, the MCL derivative can be provided as a pharmaceutically acceptable salt. In some embodiments, the MCL derivative has a structure of the formula:
  • Figure US20190314324A1-20191017-C00002
  • wherein
    Figure US20190314324A1-20191017-P00001
    represents a single or double bond; R is selected from H, alkyl, substituted alkyl, and acyl; and R1 is selected from H, alkyl, substituted alkyl, and amino; or a pharmaceutically acceptable salt thereof. In some embodiments, the amino group is —N(R2)(R3), wherein R2 and R3 are independently selected from H, alkyl (e.g., C1-12 or C1-6 alkyl), cycloalkyl, and substituted alkyl, or wherein R2 and R3 together form an alkylene group, e.g., propylene, butylene, or pentylene. In some embodiments, R is substituted alkyl, e.g., —CH2—O—R4, wherein R4 is alkyl or alkenyl, optionally wherein R4 is a lipid. In some embodiments,
    Figure US20190314324A1-20191017-P00001
    represents a single bond and R1 is —N(R2)(R3). In some embodiments, R2 and R3 are each methyl, ethyl or propyl. In some embodiments, the MCL derivative is a lipid conjugate of MCL or a derivative thereof, e.g., a compound wherein the hydroxyl group of MCL is covalently linked (e.g., via one or more ether or ester groups) to a lipid or lipid derivative (e.g., a sterol, a fatty acid or a fatty alcohol). In some embodiments, the MCL derivative is dimethylamino micheliolide (DMAMCL) or a lipid conjugate of MCL or DMAMCL.
  • Any suitable nanoparticle can be used, including but not limited to a polymeric micelle, a liposome, a dendrimer, an organic polymer-based nanoparticle, a silica-based nanoparticle, a nanoscale coordination polymer (NCP), a nanoscale metal-organic framework (nMOF), or an inorganic polymer, such as a gold or iron oxide nanoparticle. In some embodiments, the nanoparticle is a NCP and/or nMOF. Exemplary methods of preparing and using NCPs and nMOFs are described, for example in PCT International Patent Application Publication Nos. WO 2015/069926 and WO 2013/009701, U.S. Pat. No. 9,693,957, and U.S. Patent Application Publication No. 2016/0346204, the disclosures of each of which are incorporated herein by reference in their entireties.
  • In some embodiments, the nanoparticle comprises a nMOF that comprises metal-containing clusters or ions coordinated to organic molecules, wherein the material comprises repeating coordination units. The organic molecule (which can also be referred to as a “bridging ligand”) can include, for example, carboxylate, phosphate, amino, mercapto, or hydroxyl groups to form coordinate bonds with a metal ion. The metal-containing clusters can be referred to as secondary building units (SBUs). In some embodiments, the SBUs are metal oxo clusters. For example, suitable SBUs can include, but are not limited to, Zr-oxo clusters, Hf-oxo clusters, Zn-oxo clusters, Ti-oxo clusters, Cu-carboxylate paddlewheels, and others. In some embodiments, the oxo clusters comprise anions selected from oxide (O2−), hydroxide (OH), S2−, SH, and formate (HCO2 ). Each SBU is bonded to at least one other SBU via coordinative bonding to the same bridging ligand. Stated another way, each SBU is bonded via a coordinative bond to at least one bridging ligand, which is also coordinatively bonded to at least one other SBU.
  • Any suitable bridging ligand or ligands can be used. In some embodiments, each bridging ligand is an organic compound comprising multiple coordination sites. The coordination sites can each comprise a group capable of forming a coordinate bond with a metal cation or a group capable of forming such a group. Thus, each coordination site can comprise an unshared electron pair, a negative charge, or an atom or functional group capable of forming an unshared electron pair or negative charge. Typical coordination sites include, but are not limited to, functional groups such as carboxylate and derivatives there (e.g., esters, amides, anhydrides), nitrogen-containing groups (e.g., amines, nitrogen-containing aromatic and non-aromatic heterocycles), alcohols, phenols and other hydroxyl-substituted aromatic groups; ethers, acetylacetonate (i.e., C(═O)CH2C(═O)CH3), phosphonates, phosphates, thiols, and the like. In some embodiments, each bridging ligand comprises between 2 and 10 coordination sites (i.e., 2, 3, 4, 5, 6, 7, 8, 9, or 10 coordination sites). In some embodiments, each bridging ligand is capable of binding to two or three SBUs. In some embodiments, each of the organic bridging ligands is a dicarboxylate or a tricarboxylate. The coordination sites of the organic bridging ligand can be bonded to the same polyvalent (e.g., divalent or trivalent) linking group, such as an alkylene or arylene group. In some embodiments, the bridging ligand further includes a group, such as an amino, hydroxyl, or thiol, that can form a covalent bond with a therapeutic agent (e.g., a chemotherapeutic or immunotherapeutic agent or MCL or a derivative thereof). For instance, the bridging ligand can be amino-triphenyldicarboxylic acid (amino-TPDC).
  • In some embodiments, the NCP is a metal bisphosphonate coordination polymer comprising a multivalent metal ion and a bisphosphonate. Any suitable multivalent metal ion can be used. In some embodiments, the multivalent metal ion is divalent. In some embodiments, the metal ion is Ca2+, Mg2+, Mn2+, Zn2+, or a combination thereof. In some embodiments, the metal is Zn2+. In some embodiments, the bisphosphonate is a chemotherapeutic prodrug. Thus, in some embodiments, a chemotherapeutic is present as a bridging ligand between metal ions in the NCP. In some embodiments, the bisphosphonate is a cisplatin, oxaliplatin, or carboplatin prodrug. In some embodiments, the prodrug can be prepared by providing a metal complex based on cisplatin, oxaliplatin, carboplatin or another platinum coordination complex chemotherapeutic and having two hydroxyl ligands, and contacting the metal complex with diethoxyphosphinyl isocyanate, diethoxyphosphinyl isothiocyanate, diethoxyphosphinyl-containing carboxylic anhydride, or diethoxyphosphinyl-containing acyl chloride to form metal ligands that can provide phosphonate groups available for further coordinative bonding. Thus, in some embodiments, the bisphosphonate can be a bisphosphonate ester of cis, cis-trans-[Pt(NH3)2Cl2(OH)2], a dihydroxyl complex based on cisplatin. Accordingly, in some embodiments, the bisphosphonate comprises a metal coordination complex and, therefore, includes a second metal ion, such as a Pt ion, in addition to the multivalent metal ion of the NCP.
  • Alternatively, in some embodiments, the bisphosphonate is an organic compound comprising at least two phosphonate groups and is free of an additional metal ion. For example, the bisphosphonate can have a structure of the formula:
  • Figure US20190314324A1-20191017-C00003
  • wherein n and m are each independently an integer between 0 and 2 (i.e., each independently 0, 1, or 2); each R is independently H, alkyl, substituted alkyl, aralkyl, aryl, or substituted aryl; R1 is H, hydroxyl, halo, alkyl, substituted alkyl, amino, alkoxy, or alkylthio; and R2 is halo, alkyl, substituted alkyl, aralkyl, substituted aralkyl, alkoxy, alkylthio, aryloxy, arylthio or arylamino; or an anion or salt thereof.
  • In some embodiments, the nanoparticle can comprise a core comprising one of the above-mentioned materials, such as an organic polymer, a silica-based matrix, a NCP, a nMOF, or an inorganic material, and further comprises one or more coating layers that surrounds at least a portion of the surface of the nanoparticle core and/or one or more coating agents, e.g., an agent attached to a coating layer or to the surface of the nanoparticle core. Suitable coating agents or layers include, but are not limited to, metal oxide, polymer (e.g., a silica-based polymer, such as silica, poly(siloxane), or poly(silsesquioxane), or an organic or hydrophilic organic polymer), a single lipid layer, a lipid bilayer, or a combination thereof. In some embodiments, one or more of the coating agents or layers can be derivatized with a targeting agent (such as a RGD peptide or an antibody or antibody fragment) and/or a passivating agent (such as a hydrophilic polymer e.g., poly(ethylene glycol) (PEG)) and or an imaging agent (such as a fluorescent moiety). In some embodiments, the one or more coating agents or layers are selected from the group comprising, but not limited to, dioleoyl L-α-phosphatidylethanolamine (DOPE), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dioleoyl-sn-glycero-3-phosphate sodium salt (DOPA), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), pegylated DSPE (e.g., DSPE-PEG2k), 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), anisamide-derivatized DOPE, cholesterol, silica, cRGfK-derivatized silica, PEG-derivatized silica, anisamide-PEG-derivatized silica, and combinations thereof.
  • In some embodiments, the MCL or derivative thereof is part of the nanoparticle. For example, in some embodiments, the MCL or its derivative can be attached to or sequestered within the nanoparticle core. For example, in some embodiments, the MCL derivative can include a chemical functional group that can form a covalent or ionic bond with an organic functional group or metal ion present in a component of an NCP or nMOF nanoparticle core. In some embodiments, the MCL or derivative thereof can be physically sequestered (e.g., encapsulated) in pores or channels in an NCP or nMOF nanoparticle core. In some embodiments, the MCL or its derivative can be attached (e.g., covalently or electrostatically) to a coating agent or layer or sequestered within a coating layer. In some embodiments, the MCL derivative can comprise a fatty acid or other lipid moiety and the MCL derivative can be sequestered in a lipid or lipid bilayer coating layer.
  • In some embodiments, the nanoparticle comprises a therapeutic agent other than the MCL or its derivative, e.g., a chemotherapeutic agent or an immunotherapy agent. Any suitable chemotherapeutic agent or immunotherapy agent can be used. In some embodiments, the chemotherapeutic agent can be selected from the group including, but not limited to, a platinum complex, such as cisplatin, oxaliplatin, carboplatin, or a prodrug thereof; doxorubicin; daunorubicin; docetaxel; mitoxanthrone; paclitaxel; digitoxin; gemcitabine; methotrexate; leucovorin; pemetresed disodium; vinblastine; vincristine; vindesine; cytarabine; azathioprine; melphalan; imitnib; anastrozole; letrozole; etoposide; vinorelbine; digoxin, and septacidin. In some embodiments, the nanoparticle can comprise more than one chemotherapeutic agent. In some embodiments, one or more chemotherapeutic agent is attached to or sequestered in a nanoparticle core. In some embodiments, one or more chemotherapeutic agent is attached to or sequestered in a nanoparticle coating layer.
  • In some embodiments, the immunotherapy agent is selected from the group including, but not limited to, an anti-CD52 antibody, an anti-CD20 antibody, an anti-CD20 antibody, anti-CD47 antibody an anti-GD2 antibody, a radiolabeled antibody, an antibody-drug conjugate, polysaccharide K, a neoantigen, an anti-LAG3 antibody, an anti-4-IBB antibody, an anti-TIM3 antibody and a cytokine. In some embodiments, the cytokine is an interferon, an interleukin, or tumor necrosis factor alpha (TNF-α). In some embodiments, the cytokine is selected from the group comprising IFN-α, INF-γ, IL-2, IL-12 and TNF-α. In some embodiments, the immunotherapy agent is selected from the group comprising Alemtuzumab, Ofatumumab, Rituximab, Zevalin, Adcetris, Kadcyla and Ontak. In some embodiments, the immunotherapy agent is selected from the group comprising a PD-1 inhibitor or a PD-L1 inhibitor, such as, but not limited to, BMS-936559 or BMS-936558 from Bristol-Myers Squibb, MPDL3280A from Genentech, MK-3475 from Merck, CT-011 from Curetech, and MEDI4736 from MedImmune; a CTLA-4 inhibitor (e.g., ipilimumab, tremelimumab); an indoleamine-2,3-dioxygenase (IDO) inhibitor; and a CCR7 inhibitor. The IDO inhibitor can be any suitable IDO inhibitor, such as, but not limited to oxadiazole and other heterocyclic IDO inhibitors, e.g., as reported in U.S. Patent Application Publication Nos. 2006/0258719 and 2007/0185165, which are incorporated herein by reference in their entireties. IDO inhibitors also include those described in PCT Publication WO 99/29310, incorporated herein by reference in its entirety, which reports methods for altering T cell-mediated immunity comprising altering local extracellular concentrations of tryptophan and tryptophan metabolites, BMS-986205 from Bristol-Myers Squibb or F001287 from Flexus, epacadostat from Incyte Corp., indoximod, IO-102, EOS-200271, HTI-1090, IO-101, KHK-2455, 1-methyl-DL-tryptophan, p-(3-benzofuranyl)-DL-alanine, p-[3-benzo(b)thienyl]-DL-alanine, and 6-nitro-L-tryptophan), and IDO inhibitors described in WO 03/087347, incorporated herein by reference in its entirety, also published as European Patent 1501918, which describes methods of making antigen-presenting cells for enhancing or reducing T cell tolerance. Compounds having IDO inhibitory activity are further reported in WO 2004/094409, and in U.S. Patent Application Publication No. 2004/0234623, incorporated herein by reference in its entirety, and which is directed to methods of treating a subject with a cancer or an infection by the administration of an inhibitor of indoleamine-2,3-dioxygenase in combination with other therapeutic modalities. In some embodiments, the small molecule inhibitors are those disclosed in U.S. Pat. No. 8,088,803, which is incorporated by reference in its entirety herein. In some embodiments, the immunotherapy agent is an immune checkpoint inhibitor (e.g., an antibody immune checkpoint inhibitor, such as, but not limited to, a PD-1/PD-L1 antibody, a CTLA-4 antibody, an OX40 antibody, a TIM3 antibody, a LAG3 antibody, an anti-CD47 antibody). In some embodiments, one or more immunotherapy agent is attached to or sequestered in a nanoparticle core. In some embodiments, one or more immunotherapy agent is attached to or sequestered in a nanoparticle coating layer.
  • In some embodiments, the nanoparticle comprises both MCL or a derivative thereof and one or more additional therapeutic agents (e.g., chemotherapeutic and/or immunotherapy agents). The other therapeutic agent can be attached to or sequestered within the nanoparticle core or can be attached to or sequestered in a coating layer or agent. In some embodiments, the nanoparticle is an NCP or nMOF comprising a chemotherapeutic agent, e.g., cisplatin, oxaliplatin, or a prodrug thereof, and the nanoparticle further comprises a lipid coating layer comprising the MCL or its derivative, e.g., a MCL lipid conjugate. In some embodiments, both the MCL or its derivative and the additional therapeutic agent are attached to or sequestered in the nanoparticle core. In some embodiments, both the MCL or its derivative and the additional therapeutic agent are attached to or sequestered in a coating layer, attached to the nanoparticle core surface, or attached to a coating agent.
  • In some embodiments, the composition comprises a mixture of nanoparticles, wherein one type of nanoparticle comprises MCL or a derivative thereof and another type of nanoparticle comprises one or more additional therapeutic agents (e.g., a chemotherapeutic, an immunotherapeutic and/or a nucleic acid therapeutic agent).
  • IV. Pharmaceutical Compositions
  • In some embodiments, the presently disclosed subject matter provides a composition comprising MCL or a derivative and/or nanoparticle thereof as described herein and a pharmaceutically acceptable carrier, e.g., a pharmaceutically acceptable carrier that is pharmaceutically acceptable in humans. In some embodiments, the composition can also include other components, such as, but not limited to anti-oxidants, buffers, bacteriostatics, bactericidal antibiotics, suspending agents, thickening agents, and solutes that render the composition isotonic with the bodily fluids of a subject to whom the composition is to be administered.
  • For example, suitable formulations can include aqueous and non-aqueous sterile injection solutions that can contain anti-oxidants, buffers, bacteriostatics, bactericidal antibiotics, and solutes that render the formulation isotonic with the bodily fluids of the subject; and aqueous and non-aqueous sterile suspensions that can include suspending agents and thickening agents. The formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a frozen or freeze-dried (lyophilized) condition requiring only the addition of sterile liquid carrier, for example water for injections, immediately prior to use. Some exemplary ingredients are sodium dodecyl sulfate (SDS), in one example in the range of 0.1 to 10 mg/ml, in another example about 2.0 mg/ml; and/or mannitol or another sugar, for example in the range of 10 to 100 mg/m I, in another example about 30 mg/m I; and/or phosphate-buffered saline (PBS).
  • It should be understood that in addition to the ingredients particularly mentioned above, the formulations of the presently disclosed subject matter can include other agents conventional in the art having regard to the type of formulation in question. For example, sterile pyrogen-free aqueous and non-aqueous solutions can be used.
  • The methods and compositions disclosed herein can be used on a sample either in vitro (for example, on isolated cells or tissues) or in vivo in a subject (i.e. living organism, such as a patient). In some embodiments, the subject is a human subject, although it is to be understood that the principles of the presently disclosed subject matter indicate that the presently disclosed subject matter is effective with respect to all vertebrate species, including mammals, which are intended to be included in the terms “subject” and “patient”. Moreover, a mammal is understood to include any mammalian species for which employing the compositions and methods disclosed herein is desirable, particularly agricultural and domestic mammalian species.
  • As such, the methods of the presently disclosed subject matter are particularly useful in warm-blooded vertebrates. Thus, the presently disclosed subject matter concerns mammals and birds. More particularly provided are methods and compositions for mammals such as humans, as well as those mammals of importance due to being endangered (such as Siberian tigers), of economic importance (animals raised on farms for consumption by humans), and/or of social importance (animals kept as pets or in zoos) to humans, for instance, carnivores other than humans (such as cats and dogs), swine (pigs, hogs, and wild boars), ruminants (such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels), rodents (e.g., mice, rats, hamsters, gerbils, etc.) and horses. Also provided is the treatment of birds, including the treatment of those kinds of birds that are endangered, kept in zoos or as pets (e.g., parrots), as well as fowl, and more particularly domesticated fowl, for example, poultry, such as turkeys, chickens, ducks, geese, guinea fowl, and the like, as they are also of economic importance to humans. Thus, also provided is the treatment of livestock including, but not limited to domesticated swine (pigs and hogs), ruminants, horses, poultry, and the like.
  • Suitable methods for administration of a composition of the presently disclosed subject matter include, but are not limited to intravenous and intratumoral injection, oral administration, subcutaneous administration, intraperitoneal injection, intracranial injection, and rectal administration. Alternatively, a composition can be deposited at a site in need of treatment in any other manner, for example by spraying a composition within the pulmonary pathways. The particular mode of administering a composition of the presently disclosed subject matter depends on various factors, including the distribution and abundance of cells to be treated and mechanisms for metabolism or removal of the composition from its site of administration. For example, relatively superficial tumors can be injected intratumorally. By contrast, internal tumors can be treated following intravenous injection.
  • In one embodiment, the method of administration encompasses features for regionalized delivery or accumulation at the site to be treated. In some embodiments, a composition is delivered intratumorally. In some embodiments, selective delivery of a composition to a target is accomplished by intravenous injection of the composition followed by irradiation (e.g., X-ray irradiation) of the target.
  • For delivery of compositions to pulmonary pathways, compositions of the presently disclosed subject matter can be formulated as an aerosol or coarse spray. Methods for preparation and administration of aerosol or spray formulations can be found, for example, in U.S. Pat. Nos. 5,858,784; 6,013,638; 6,022,737; and 6,136,295.
  • In some embodiments, an effective dose of a composition of the presently disclosed subject matter is administered to a subject. An “effective amount” is an amount of the composition sufficient to produce detectable treatment (e.g., tumor size reduction, pain relief, reduction of the concentration of a disease-related blood marker, etc.). Actual dosage levels of constituents of the compositions of the presently disclosed subject matter can be varied so as to administer an amount of the composition that is effective to achieve the desired effect for a particular subject and/or target. The selected dosage level can depend upon the activity (e.g., nanoparticle drug loading and/or the IC50 of the therapeutic components in certain cell types (e.g., cancer cell lines) of the composition and the route of administration.
  • After review of the disclosure herein of the presently disclosed subject matter, one of ordinary skill in the art can tailor the dosages to an individual subject, taking into account the particular formulation, method of administration to be used with the composition, and nature of the target to be treated. Such adjustments or variations, as well as evaluation of when and how to make such adjustments or variations, are well known to those of ordinary skill in the art.
  • A dose may be administered on an as needed basis or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, or 24 hours (or any range derivable therein) or 1, 2, 3, 4, 5, 6, 7, 8, 9, or times per day (or any range derivable therein). A dose can be first administered before or after symptoms are exhibited by the subject; after a clinician evaluates the subject for the disease; or before, at about the same time as, or after administration of a second treatment (chemotherapy, radiation therapy, immunotherapy). In some embodiments, the patient is administered a first dose of a compound, such as MCL, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 hours (or any range derivable therein) or 1, 2, 3, 4, or 5 days after the subject exhibits signs or symptoms of the disease (or any range derivable therein). The subject can be treated for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more days (or any range derivable therein) or until symptoms of an the disease, such as tumor size, have disappeared or been reduced or after 6, 12, 18, or 24 hours or 1, 2, 3, 4, or 5 days after symptoms of the disease have disappeared or been reduced.
  • V. Methods of Treating Disease Using Combinations Comprising MCL or a Derivative Thereof and Another Therapeutic Agent and/or Radiation
  • In some embodiments, the presently disclosed subject matter provides a method for treating a disease (e.g., cancer or another hyperproliferative disease) by using the MCL or derivative and/or nanoparticle thereof and/or pharmaceutical composition thereof in combination with another treatment modality, e.g., a chemotherapeutic, an immunotherapeutic, radiation, surgery, etc. In some embodiments, the other treatment modality is radiation. Thus, in some embodiments, the presently disclosed subject matter provides a method of treating a disease in a subject in need thereof, the method comprising: administering to the subject an MCL or derivative and/or nanoparticle thereof and/or pharmaceutical composition thereof as described herein; and irradiating at least a portion of the subject with ionizing radiation, such as with x-rays and/or protons. In some embodiments, the method comprises administering a nanoparticle and/or NCP comprising MCL or a derivative thereof. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human.
  • In some embodiments, the patients are irradiated with a linear accelerator (LINAC), using conventional techniques, Intensity-Modulated Radiation Therapy (IMRT), Image Guided Radiation Therapy (IGRT), or Stereotactic Body Radio Therapy (SBRT), a 60Co radiation source, an implanted radioactive seed such as the ones used in brachytherapy, an orthovoltage or supervoltage X-ray irradiator, a high energy electron beam generated from LINAC, or a proton source.
  • In some embodiments, the γ-rays generated by a LINAC pass through an energy modulator (filter) before irradiating the patient, optionally wherein the filter comprises one or more element(s) with atomic number(s) of at least 20, further optionally wherein the filter comprises copper. In some embodiments, the filter has a thickness that is less than 5 mm, less than 4 mm, less than 3 mm, less than 2 mm, less than 1 mm, less than 0.5 mm, less than 0.4 mm, less than 0.3 mm, less than 0.2 mm, less than 0.1 mm.
  • In some embodiments, X-rays are generated by placing radioactive sources inside the patient on a temporary or permanent basis. In some embodiments, a nanoparticle composition is injected along with the implantation of a radioactive source.
  • The radiation dosage regimen is generally defined in terms of Gray or Sieverts time and fractionation and can be carefully defined by the radiation oncologist. The amount of radiation a subject receives can depend on various considerations, such as the location of the tumor in relation to other critical structures or organs of the body, and the extent to which the tumor has spread. One illustrative course of treatment for a subject undergoing radiation therapy is a treatment schedule with a total dose of 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 Gray (Gy) or any derivable range therein. The radiation dose can be administered to the subject in a single daily fraction of 1.0-2.0 Gy, 1.0-5.0 Gy, or 5.0-10.0 Gy 1, 2, 3, 4, or 5 days a week. A Gy is an abbreviation for Gray and refers to 100 rad of dose. Illustrative dosages used for photon-based radiation are measured in Gy, and in an otherwise healthy subject (that is, little or no other disease states present such as high blood pressure, infection, diabetes, etc.) for a solid epithelial tumor ranges from about 60 to about 80 Gy, and for a lymphoma ranges from about 20 to about 40 Gy. Illustrative preventative (adjuvant) doses are typically given at about 45 to about 60 Gy in about 1.8 to about 2 Gy fractions for breast, head, and neck cancers.
  • In some embodiments, the disease is cancer. In some embodiments, the disease is selected from the group including, but not limited to, a skin cancer, such as a melanoma; a connective tissue cancer, such as a sarcoma; an adipose cancer; a breast cancer; a head and neck cancer, such as a glioma; a lung cancer, such as mesothelioma; a stomach cancer; a pancreatic cancer; an ovarian cancer; a cervical cancer; a uterine cancer; an anogenital cancer, such as a testicular cancer; a kidney cancer; a bladder cancer; a colon cancer; a prostate cancer; a central nervous system (CNS) cancer; a retinal cancer; a blood cancer, such as leukemia; a neuroblastoma; multiple myeloma, and a lymphoid cancer, such as Hodgkin's or non-Hodgkin's lymphoma.
  • The method can comprise administering the MCL or a derivative and/or nanoparticle thereof to the subject at about the same time as, or later than the administration of the radiation. In some embodiments, the MCL or derivative and/or nanoparticle thereof can be administered to the subject prior to a dose or course of radiation.
  • In some embodiments, the method can further comprise administering to the subject an additional therapeutic agent or treatment. In some embodiments, the additional therapeutic agent or treatment is an immunotherapy agent or a cancer treatment selected from surgery, chemotherapy, toxin therapy, cryotherapy, and gene therapy. In some embodiments, the chemotherapy can be co-administered with the MCL or derivative thereof, e.g., in a nanoparticle comprising both MCL or a derivative thereof and a chemotherapeutic agent. In some embodiments, the additional therapeutic agent is an immunotherapeutic agent. In some embodiments, the additional therapeutic agent is an immune checkpoint inhibitor, such as, but not limited to, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
  • The additional therapeutic agent or treatment can be administered prior to, at the same time as, or later than the administration of the MCL or derivative and/or nanoparticle thereof. For example, in some embodiments, administration of MCL or a derivative thereof alone, or of MCL or a derivative thereof in combination with X-ray irradiation treatment, can be performed prior to surgery to reduce the size of a tumor.
  • In some embodiments, the presently disclosed subject matter provides a method comprising treating a disease in a subject in need thereof, the method comprising: (i) administering to the subject MCL or a derivative and/or nanoparticle thereof and (ii) administering to the subject a chemotherapeutic agent, an immunotherapy agent, or radiotherapy. In some embodiments, the method comprises administering the MCL or derivative thereof and an immunotherapy agent. In some embodiments, the method comprises administering to the subject a nanoparticle comprising MCL or a derivative thereof. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human.
  • In some embodiments, the disease is cancer. In some embodiments, the disease is selected from the group including, but not limited to, a skin cancer, such as a melanoma; a connective tissue cancer, such as a sarcoma; an adipose cancer; a breast cancer; a head and neck cancer, such as a glioma; a lung cancer, such as mesothelioma; a stomach cancer; a pancreatic cancer; an ovarian cancer; a cervical cancer; a uterine cancer; an anogenital cancer, such as a testicular cancer; a kidney cancer; a bladder cancer; a colon cancer; a prostate cancer; a central nervous system (CNS) cancer; a retinal cancer; a blood cancer, such as leukemia; a neuroblastoma; multiple myeloma, and a lymphoid cancer, such as Hodgkin's or non-Hodgkin's lymphoma.
  • In some embodiments, the immunotherapeutic agent is an immune checkpoint inhibitor, such as, but not limited to, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor. In some embodiments, the immunotherapeutic agent can be formulated in the same pharmaceutical composition as the MCL or derivative and/or nanoparticle thereof or be part of (e.g., embedded in or attached to) the same nanoparticle as the MCL or derivative thereof.
  • In some embodiments, the method further comprises administering to the subject a chemotherapeutic agent. In some embodiments, the chemotherapeutic agent can be provided in a nanoparticle, such as a nanoparticle further comprising the MCL or derivative thereof. In some embodiments, the chemotherapeutic agent is selected from the group including, but not limited to, a platinum complex, such as cisplatin, oxaliplatin, carboplatin, or a prodrug thereof; doxorubicin; daunorubicin; docetaxel; mitoxanthrone; paclitaxel; digitoxin; gemcitabine; methotrexate; leucovorin; pemetresed disodium; vinblastine; vincristine; vindesine; cytarabine; azathioprine; melphalan; imitnib; anastrozole; letrozole; etoposide; vinorelbine; digoxin, and septacidin.
  • In some embodiments, the presently disclosed subject matter provides a method of treating a disease (e.g., cancer) in a subject in need thereof, wherein the method comprises: (i) administering to the subject MCL or a derivative and/or nanoparticle thereof; (ii) administering to the subject an immunotherapeutic agent (e.g., an immune checkpoint inhibitor); and (iii) irradiating at least a portion of the subject with ionizing radiation, such as with x-rays and/or protons. In some embodiments, the method can further comprise an additional therapeutic agent or treatment selected from the group including, but not limited to, surgery, toxin therapy, cryotherapy and gene therapy.
  • EXAMPLES
  • The following Examples have been included to provide guidance to one of ordinary skill in the art for practicing representative embodiments of the presently disclosed subject matter. In light of the present disclosure and the general level of skill in the art, those of skill can appreciate that the following Examples are intended to be exemplary only and that numerous changes, modifications, and alterations can be employed without departing from the scope of the presently disclosed subject matter.
  • Example 1 Combinations of Micheliolide and Radiotherapy 1.1. In Vitro Cytotoxicity Against Glioma Cells GL261:
  • GL261 cells were seeded onto 96-well plates at a density of 2000 cells/well and treated with various concentrations of the dimethylamino Michael adduct of MCL (dimethylamino micheliolide, DMAMCL) for 24 hours to allow for sufficient cellular uptake and conversion to MCL before irradiation. The cells were irradiated with varying amounts of X-ray irradiation and then incubated for an additional 24 hours. The culture medium was exchanged with fresh medium and further incubated for an additional 48 hours. The cell viability was then detected by (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay (Promega, Madison, Wis., United States of America).
  • At doses up to 10 μM DMAMCL or 8 Gy irradiation, monotherapy DMAMCL or irradiation showed little to no toxicity. However, cells treated with 1 or 10 μM DMAMCL and X-ray irradiation of 8 Gy irradiation showed significant cytotoxicity that far exceeded the additive effects of either therapy alone. See FIG. 1.
  • 1.2. In Vivo Antitumor Activity:
  • The combination of DMAMCL and irradiation was evaluated on the glioma GL261 tumor model on immunocompromised Rag2−/− C57BL/6 mice. See FIG. 2. The mice were orally dosed with 50 mg/kg DMAMCL or PBS daily and irradiated with 4 Gy irradiation approximately 4 hrs after dosing, as applicable, for up to 7 days, starting when the tumors reached about 2-300 mm3. A Precision X-ray X-RAD 225 Image Guided Biologic Irradiator (Precision X-Ray Irradiation, North Branford, Conn., United States of America) was used for all animal studies. The instrument was set at 225 peak kilovoltage (kVp) and 13 milliampere (mA), with a 0.3 mm flat-board copper (Cu) filter installed before a 15-mm collimator. The irradiation was given in two successive fractions with incident angles of 0° and 180° for each mouse.
  • Fractionated irradiation allows for higher total doses of X-ray but showed little or no appreciable effects on tumor growth. Though DMAMCL alone initially suppressed tumor growth, the tumors were ultimately not significantly different in size compared to those treated with irradiation alone. These results are consistent with in vitro results which showed varied effects leading to high average cell viability with large deviation in cells and tumors treated with irradiation alone. In contrast, the combination of DMAMCL and irradiation successfully controlled tumor growth leading to relatively stable tumor sizes over the course of treatment.
  • Example 2 Combinations of Micheliolide, Radiotherapy and Immune Checkpoint Blockade
  • The combination of DMAMCL, irradiation, and PD-L1 antibody was evaluated on the glioma GL261 tumor model on wildtype C57BL/6 mice. See FIG. 3. The treatment began 14 days after the inoculation of 2×106 GL261 cells. The mice were orally dosed with 50 mg/kg DMAMCL or PBS daily and irradiated with 2 Gy irradiation approximately 4 hrs after dosing, as applicable, for up to 12 days, starting when the tumors reached ˜150-200 mm3. One group of mice received two doses of 75 μg PD-L1 antibody on days 0 and 4. DMAMCL alone showed no effect on tumor growth, but combination with irradiation suppressed tumor growth and ultimately led to tumor regression. The combination of DMAMCL, irradiation, and a PD-L1 antibody controlled and regressed tumor growth immediately after treatment. By the cessation of treatment, 3/5 tumors treated with this combination were barely palpable. DMAMCL showed synergy with irradiation to control and regress GL261 tumors, which was further enhanced by combination with an immune checkpoint inhibitor.
  • Example 3 Nanoparticle-Based Micheliolide
  • Micheliolide and its derivatives can be formulated into nanoscale coordination polymers (NCPs) for use in treating multiple cancer types in combination with radiotherapy. Exemplary methods of preparing and using NCPs are described, for example in PCT International Patent Application Publication Nos. WO 2015/069926 and WO 2013/009701, U.S. Pat. No. 9,693,957, and U.S. Patent Application Publication No. 2016/0346204, the disclosures of each of which are incorporated herein by reference in their entireties. In some embodiments, according to the presently disclosed subject matter, MCL and its derivatives were loaded directly or as lipid-conjugates onto NCPs.
  • 3.1. Synthesis of MCL Derivatives:
  • As shown in Equation 1 (Eq 1) of Scheme 1, below, micheliolide (500 mg, 2 mmol) was dissolved in 20 mL anhydrous dichloromethane (DCM). 2.5 mL dimethylamine 2M solution in tetrahydrofuran (THF) was added to the solution. The mixture was stirred at room temperature for 8 hours and then solvent was removed by rotary evaporation. The solid residue was purified by column chromatography using 5% methanol in DCM to obtain pure DMAMCL. 1H-NMR (CDCl3, 500 MHz): 1.32 (s, 3H), 1.70 (s, 3H), 1.82 (m, 2H), 2.06 (q, 1H), 2.19 (m, 4H), 2.27 (s, 6H), 2.41 (m, 2H), 2.62 (m, 2H), 2.67 (d, 2H), 2.72 (dd, 1H), 3.84 (t, 1H). ESI-MS: m/z=294.2 ([M+H]+, expected 294.2).
  • Figure US20190314324A1-20191017-C00004
  • As shown in Equation 2 (Eq 2) of Scheme 1, above, stearyl alcohol (1.62 g, 6 mmol) and paraformaldehyde (180 mg, 6 mmol) was mixed in a dry round bottom flask. Bromotrimethylsilane (5 mL) was added drop-wise to the mixture and the solution was stirred at room temperature until all solid dissolved. Solvents was then removed by rotary evaporation to obtain crude 1-(bromomethoxy)octadecane.
  • As shown in Equation 3 (Eq 3) of Scheme 1, above, micheliolide (MCL, 500 mg, 2 mmol) was dissolved in anhydrous THF. Sodium hydride (NaH, 100 mg, 2.5 mmol) was added portion-wise to the mixture. 1-(Bromomethoxy)octadecane dissolved in anhydrous THF was added drop-wise to the mixture and the mixture was stirred for 24 hours at room temperature. Solvents were removed and the solid residue was purified by column chromatography using 10% ethyl acetate (EtOAc) in hexanes to obtain (octadecyloxy)methoxymicheliolide. 1H-NMR (CDCl3, 500 MHz): 0.91 (t, 3H), 1.30 (m, 26H), 1.38 (s, 3H), 1.57 (m, 4H), 1.74 (s, 3H), 1.87 (m, 2H), 2.09 (d, 1H), 2.25 (m, 3H), 2.43 (dd, 1H), 2.71 (t, 1H), 2.92 (d, 1H), 3.66 (m, 5H), 3.76 (t, 1H), 4.83 (d, 1H), 4.95 (d, 1H), 5.44 (d, 1H), 6.19 (d, 1H). ESI-MS: m/z=531.4 ([M+H]+, expected 531.4)
  • 3.2. Loading of MCL onto Oxaliplatin (Ox)-Containing NCPs to Provide OxNCP/MCL:
  • OxNCP/MCL was obtained by adding a 804 THF solution of DOPC, cholesterol, MCL (molar ratio 3.5:2:3), DSPE-PEG2k (20 mol %) and OxNCP bare particles to 500 μL 30% (v/v) EtOH/H2O at 50° C. THF and EtOH were evaporated and the solution was allowed to cool to room temperature before use. The particle size and distribution were determined by dynamic light scattering (DLS). See FIG. 4. OxNCP/MCL has a Z-average diameter of 76 nm.
  • It will be understood that various details of the presently disclosed subject matter may be changed without departing from the scope of the presently disclosed subject matter. Furthermore, the foregoing description is for the purpose of illustration only, and not for the purpose of limitation.

Claims (35)

What is claimed is:
1. A composition comprising a nanoparticle and micheliolide (MCL) or a derivative thereof.
2. The composition of claim 1, comprising a derivative of MCL, wherein said derivative is dimethylamino micheliolide (DMAMCL) or a lipid conjugate of MCL.
3. The composition of claim 1, wherein the MCL or the derivative thereof is attached to or sequestered within the nanoparticle.
4. The composition of claim 1, wherein the nanoparticle is selected from the group consisting of a polymeric micelle, a liposome, a dendrimer, an organic polymer-based nanoparticle, a silica-based nanoparticle, a nanoscale coordination polymer, a nanoscale metal-organic framework, and an inorganic nanoparticle.
5. The composition of claim 1, wherein the nanoparticle comprises a core and one or more coating layers or agents covering at least a portion of an outer surface of the core.
6. The composition of claim 5, wherein the core comprises an organic polymer, a silica-based material, a nanoscale coordination polymer, a metal-organic framework, or an inorganic material.
7. The composition of claim 5, wherein the one or more coating agents or layers are selected from a metal oxide, a polymer, a single lipid layer, a lipid bilayer, and combinations thereof.
8. The composition of claim 5, wherein the one or more coating agents or layers further comprise a passivating agent, a targeting agent, and/or an imaging agent.
9. The composition of claim 5, wherein the MCL or derivative thereof is attached to or sequestered in the nanoparticle core.
10. The composition of claim 5, wherein the MCL or the derivative thereof is attached to or sequestered in a nanoparticle coating layer.
11. The composition of claim 1, further comprising a chemotherapeutic agent and/or an immunotherapy agent.
12. The composition of claim 11, wherein the chemotherapeutic agent is attached or sequestered in a nanoparticle core or a nanoparticle coating layer.
13. The composition of claim 11, wherein the chemotherapeutic agent is selected from the group consisting of cisplatin, oxaliplatin, or prodrugs thereof; doxorubicin, daunorubicin, docetaxel, mitoxanthrone, paclitaxel, digitoxin, gemcitabine, methotrexate, leucovorin, pemetresed disodium, vinblastine, vincristine, vindesine, cytarabine, azathioprine, melphalan, imitinib, anastrozole, letrozole, carboplatin, etoposide, vinorelbine, and septacidin.
14. The composition of claim 11, wherein the immunotherapy agent is selected from the group consisting of an anti-CD52 antibody, an anti-CD20 antibody, anti-CD47 antibody, an anti-GD2 antibody, polysaccharide K, a cytokine, Alemtuzumab, Ofatumumab, Rituximab, Zevalin, Adcetris, Kadcyla, Ontak, a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
15. The composition of claim 11, wherein the immunotherapy agent is attached to or sequestered in a nanoparticle coating layer.
16. A pharmaceutical composition comprising a composition of claim 1 and a pharmaceutically acceptable carrier.
17. A method for treating a disease in a subject in need thereof, the method comprising:
administering to the subject micheliolide (MCL) or a derivative thereof; and
exposing at least a portion of the subject to ionizing irradiation energy.
18. The method of claim 17, wherein administering the MCL or derivative thereof comprises administering a nanoparticle and/or a nanoscale coordination polymer (NCP) comprising the MCL or derivative thereof.
19. The method of claim 17, wherein the ionizing irradiation energy is X-rays or protons.
20. The method of claim 17, wherein the disease is cancer.
21. The method of claim 17, wherein the disease is selected from the group consisting of a skin cancer, a connective tissue cancer, an adipose cancer, a breast cancer, a head and neck cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system (CNS) cancer, a retinal cancer, a blood cancer, a neuroblastoma, multiple myeloma, and a lymphoid cancer.
22. The method of claim 17, wherein the method further comprises administering to the subject an additional therapeutic agent or treatment selected from an immunotherapy agent and/or a cancer treatment, wherein the cancer treatment is selected from the group consisting of surgery, chemotherapy, toxin therapy, cryotherapy and gene therapy.
23. The method of claim 22, wherein the additional therapeutic agent or treatment comprises an immunotherapy agent.
24. The method of claim 23, wherein the immunotherapy agent is an immune checkpoint inhibitor.
25. The method of claim 24, wherein the immune checkpoint inhibitor is selected from the group consisting of a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, an IDO inhibitor, a CCR7 inhibitor, an OX40 inhibitor, a TIM3 inhibitor, and a LAG3 inhibitor.
26. A method for treating a disease in a subject in need thereof, the method comprising:
administering to the subject micheliolide (MCL) or a derivative thereof; and
administering to the subject one or more additional therapeutic agent or treatment.
27. The method of claim 26, wherein administering to the subject MCL or a derivative thereof comprises administering to the subject a nanoparticle comprising MCL or a derivative thereof.
28. The method of claim 26, wherein the one or more additional therapeutic agent or treatment agent is selected from a chemotherapeutic agent, an immune therapy agent, radiotherapy, surgery, chemotherapy, toxin therapy, cryotherapy and gene therapy.
29. The method of claim 26, wherein the disease is cancer.
30. The method of claim 26, wherein the disease is selected from the group consisting of a skin cancer, a connective tissue cancer, an adipose cancer, a breast cancer, a head and neck cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a central nervous system (CNS) cancer, a retinal cancer, a blood cancer, a neuroblastoma, multiple myeloma, and a lymphoid cancer.
31. The method of claim 26, wherein the one or more additional therapeutic agent or treatment comprises an immunotherapeutic agent.
32. The method of claim 31, wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
33. The method of claim 31, wherein the one or more additional therapeutic agent or treatment further comprises radiotherapy.
34. The method of claim 33, wherein the one or more additional therapeutic agent or treatment agent further comprises one or more of surgery, toxin therapy, cryotherapy and gene therapy.
35. The method of claim 26, wherein the one or more additional treatment agent comprises chemotherapy.
US16/381,774 2018-04-13 2019-04-11 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy Abandoned US20190314324A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/381,774 US20190314324A1 (en) 2018-04-13 2019-04-11 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
US16/859,594 US11389422B2 (en) 2018-04-13 2020-04-27 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
US17/855,327 US20230036839A1 (en) 2018-04-13 2022-06-30 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862657112P 2018-04-13 2018-04-13
US16/381,774 US20190314324A1 (en) 2018-04-13 2019-04-11 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/859,594 Division US11389422B2 (en) 2018-04-13 2020-04-27 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy

Publications (1)

Publication Number Publication Date
US20190314324A1 true US20190314324A1 (en) 2019-10-17

Family

ID=68161046

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/381,774 Abandoned US20190314324A1 (en) 2018-04-13 2019-04-11 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
US16/859,594 Active US11389422B2 (en) 2018-04-13 2020-04-27 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
US17/855,327 Pending US20230036839A1 (en) 2018-04-13 2022-06-30 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/859,594 Active US11389422B2 (en) 2018-04-13 2020-04-27 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
US17/855,327 Pending US20230036839A1 (en) 2018-04-13 2022-06-30 Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy

Country Status (1)

Country Link
US (3) US20190314324A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112745512A (en) * 2021-02-07 2021-05-04 河南农业大学 Copper metal-organic framework material based on metal copper and preparation method and application thereof
CN114010631A (en) * 2021-11-18 2022-02-08 上海市第六人民医院 Application of michelia lactone and derivatives thereof in treatment of traumatic craniocerebral injury
WO2022041123A1 (en) * 2020-08-28 2022-03-03 洛阳尚德药缘科技有限公司 Application of sesquiterpene lactone in preparing drug for treating optic neuritis
WO2022083632A1 (en) * 2020-10-23 2022-04-28 天津尚德药缘科技股份有限公司 Application of sesquiterpene lactone compound in preparation of drugs for alleviating radiotherapy-induced injuries
CN114470195A (en) * 2022-01-19 2022-05-13 同济大学 Antifungal preparation, inhibitor for inhibiting expression of cell PD-L1 and application
CN114469931A (en) * 2020-10-23 2022-05-13 天津尚德药缘科技股份有限公司 Application of sesquiterpene lactone compound in preparation of medicine for relieving radiation therapy injury
US11389422B2 (en) 2018-04-13 2022-07-19 The University Of Chicago Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
CN115025248A (en) * 2021-02-22 2022-09-09 西安电子科技大学 Iron-based ICG metal organic nano composite (MONs) and preparation method and application thereof
US11826426B2 (en) 2017-08-02 2023-11-28 The University Of Chicago Nanoscale metal-organic layers and metal-organic nanoplates for x-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
US11872311B2 (en) 2011-07-08 2024-01-16 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140234210A1 (en) * 2011-07-08 2014-08-21 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
US9255078B2 (en) * 2010-04-23 2016-02-09 Accendatech Micheliolide derivatives, medicinal composition, producing method and usage thereof
US20160279187A1 (en) * 2013-10-31 2016-09-29 University Of Hawaii Calpain inhibitors for ibd and colorectal cancer treatment

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6013638A (en) 1991-10-02 2000-01-11 The United States Of America As Represented By The Department Of Health And Human Services Adenovirus comprising deletions on the E1A, E1B and E3 regions for transfer of genes to the lung
US5858784A (en) 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US6022737A (en) 1995-11-02 2000-02-08 Amgen Inc. Formulations for non-viral in vivo transfection in the lungs
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
AU1628699A (en) 1997-12-05 1999-06-28 Medical College Of Georgia Research Institute, Inc. High-affinity tryptophan transporter
AU2002307243B2 (en) 2002-04-12 2008-01-03 Medical College Of Georgia Research Institute, Inc. Antigen-presenting cell populations and their use as reagents for enhancing or reducing immune tolerance
US7714139B2 (en) 2003-03-27 2010-05-11 Lankenau Institute For Medcial Research IDO inhibitors and methods of use
US7598287B2 (en) 2003-04-01 2009-10-06 Medical College Of Georgia Research Institute, Inc. Use of inhibitors of indoleamine-2,3-dioxygenase in combination with other therapeutic modalities
BRPI0608604B8 (en) 2005-05-10 2021-05-25 Incyte Corp indoleamine 2,3-dioxygenase modulators and pharmaceutical composition
CA2634198C (en) 2005-12-20 2014-06-03 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2009139939A2 (en) 2008-02-22 2009-11-19 The University Of North Carolina At Chapel Hill Hybrid nanoparticles as anti-cancer therapeutic agents and dual therapeutic/imaging contrast agents
PE20110308A1 (en) 2008-07-08 2011-06-10 Incyte Corp 1,2,5-OXADIAZOLES AS INHIBITORS OF INDOLAMINE 2,3-DIOXYGENASE
WO2015006893A1 (en) * 2013-07-15 2015-01-22 天津尚德药缘科技有限公司 Uses of sesquiterpene lactone compound and derivative thereof in preparation of drugs
JP6590802B2 (en) 2013-11-06 2019-10-16 ザ ユニバーシティ オブ シカゴThe University Of Chicago Nanoscale transporter for delivery or co-delivery of chemotherapeutic drugs, nucleic acids and photosensitizers
CN104876899A (en) 2014-02-28 2015-09-02 天津尚德药缘科技股份有限公司 Dimethylamino sphaelactone fumarate and use thereof
MA42161A (en) 2014-10-14 2017-08-23 Univ Chicago NANOPARTICLES FOR PHOTODYNAMIC THERAPY, X-RAY INDUCED PHOTODYNAMIC THERAPY, RADIOTHERAPY, CHEMOTHERAPY, IMMUNOTHERAPY, AND ANY COMBINATION OF THEM
US10806694B2 (en) 2014-10-14 2020-10-20 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
JP6643458B2 (en) 2015-08-17 2020-02-12 華為技術有限公司Huawei Technologies Co.,Ltd. User plane gateway updating method and apparatus
WO2017083595A1 (en) * 2015-11-10 2017-05-18 Massachusetts Institute Of Technology Complexes comprising a platinum compound and an immune checkpoint inhibitor and related methods
JP7090034B2 (en) 2016-05-20 2022-06-23 ザ ユニバーシティ オブ シカゴ Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy and any combination thereof
JP7364552B2 (en) 2017-08-02 2023-10-18 ザ ユニバーシティ オブ シカゴ Nanoscale metal-organic layers and metal-organic nanoplates for X-ray-induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
US11634400B2 (en) * 2017-08-19 2023-04-25 University Of Rochester Micheliolide derivatives, methods for their preparation and their use as anticancer and antiinflammatory agents
US20190314324A1 (en) 2018-04-13 2019-10-17 The University Of Chicago Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9255078B2 (en) * 2010-04-23 2016-02-09 Accendatech Micheliolide derivatives, medicinal composition, producing method and usage thereof
US20140234210A1 (en) * 2011-07-08 2014-08-21 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
US20160279187A1 (en) * 2013-10-31 2016-09-29 University Of Hawaii Calpain inhibitors for ibd and colorectal cancer treatment

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11872311B2 (en) 2011-07-08 2024-01-16 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
US11826426B2 (en) 2017-08-02 2023-11-28 The University Of Chicago Nanoscale metal-organic layers and metal-organic nanoplates for x-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
US11389422B2 (en) 2018-04-13 2022-07-19 The University Of Chicago Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
WO2022041123A1 (en) * 2020-08-28 2022-03-03 洛阳尚德药缘科技有限公司 Application of sesquiterpene lactone in preparing drug for treating optic neuritis
WO2022083632A1 (en) * 2020-10-23 2022-04-28 天津尚德药缘科技股份有限公司 Application of sesquiterpene lactone compound in preparation of drugs for alleviating radiotherapy-induced injuries
CN114469931A (en) * 2020-10-23 2022-05-13 天津尚德药缘科技股份有限公司 Application of sesquiterpene lactone compound in preparation of medicine for relieving radiation therapy injury
CN112745512A (en) * 2021-02-07 2021-05-04 河南农业大学 Copper metal-organic framework material based on metal copper and preparation method and application thereof
CN115025248A (en) * 2021-02-22 2022-09-09 西安电子科技大学 Iron-based ICG metal organic nano composite (MONs) and preparation method and application thereof
CN114010631A (en) * 2021-11-18 2022-02-08 上海市第六人民医院 Application of michelia lactone and derivatives thereof in treatment of traumatic craniocerebral injury
CN114470195A (en) * 2022-01-19 2022-05-13 同济大学 Antifungal preparation, inhibitor for inhibiting expression of cell PD-L1 and application

Also Published As

Publication number Publication date
US20200261403A1 (en) 2020-08-20
US20230036839A1 (en) 2023-02-02
US11389422B2 (en) 2022-07-19

Similar Documents

Publication Publication Date Title
US11389422B2 (en) Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
US10780045B2 (en) Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, chemotherapy, immunotherapy, and any combination thereof
US10806694B2 (en) Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
US11826426B2 (en) Nanoscale metal-organic layers and metal-organic nanoplates for x-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
US11246877B2 (en) Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy, and any combination thereof
US20130137916A1 (en) Radiotherapy combined with hypoxic cell sensitizers
EP4138784A1 (en) Metal-organic frameworks deliver small molecules and biomacromolecules for cancer immunotherapy
JP5642892B2 (en) A combination containing macitentan for the treatment of glioblastoma multiforme
CN1897949A (en) Combination therapy comprising the use of et-743 and paclitaxel for treating cancer
WO2016102735A1 (en) Stem cells irradiated for cancer treatment
JP7311177B2 (en) Combined use of A-NOR-5α androstane drugs with anticancer drugs
ES2341575T3 (en) MALIGNAS NEOPLASIAS THERAPY.
EP3052091B1 (en) Sustained release formulations containing methylglyoxal and their therapeutic applications
EA042139B1 (en) PHARMACEUTICAL COMPOSITIONS AND METHODS FOR CANCER TREATMENT
MXPA06005359A (en) Combination therapy comprising the use of et-743 and paclitaxel for treating cancer
NZ615005B2 (en) Combinations comprising macitentan for the treatment of glioblastoma multiforme

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNIVERSITY OF CHICAGO, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIN, WENBIN;CHAN, CHRISTINA;HAN, WENBO;SIGNING DATES FROM 20190406 TO 20190408;REEL/FRAME:048864/0293

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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