WO2020123481A1 - Formulations combinées de taxanes et d'inhibiteurs de mtor - Google Patents

Formulations combinées de taxanes et d'inhibiteurs de mtor Download PDF

Info

Publication number
WO2020123481A1
WO2020123481A1 PCT/US2019/065440 US2019065440W WO2020123481A1 WO 2020123481 A1 WO2020123481 A1 WO 2020123481A1 US 2019065440 W US2019065440 W US 2019065440W WO 2020123481 A1 WO2020123481 A1 WO 2020123481A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
drug
docetaxel
nanoparticles
everolimus
Prior art date
Application number
PCT/US2019/065440
Other languages
English (en)
Inventor
Robin Humphreys
Paul Tardi
Leon WAN
Barry LIBOIRON
Original Assignee
Celator Pharmaceuticals Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Celator Pharmaceuticals Inc. filed Critical Celator Pharmaceuticals Inc.
Publication of WO2020123481A1 publication Critical patent/WO2020123481A1/fr

Links

Classifications

    • 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
    • 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
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • 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
    • A61K47/554Medicinal 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 the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles

Definitions

  • the invention relates to compositions and methods for improved delivery of combinations of therapeutic agents. More particularly, the invention concerns delivery systems which provide combinations of taxanes and mTOR inhibitors.
  • Taxanes are known antineoplastic agents that are typically used in combination with other agents to treat a variety of cancers.
  • taxanes such as paclitaxel
  • paclitaxel refer to a family of products extracted from the leaves and bark of the European yew tree ( Taxus Baccata and other species of the Taxus family), as well as semi-synthetic products, such as docetaxel, obtained from baccatin III or from 10-deacetylbaccatin III which are also extracted from yew.
  • Docetaxel acts by disrupting microtubule disassembly, resulting in inhibition of mitosis, and ultimately leading to apoptosis.
  • Examples of cancer which can be treated with docetaxel are: locally advanced or metastatic breast cancer, non-small cell lung cancer, hormone-refractory prostate cancer and ovarian cancer, and gastric cancer.
  • mTOR inhibitors are a class of agents which act to inhibit the mammalian target of rapamycin (mTOR) which is a serine/threonine-specific protein kinase that belongs to the family of phosphatidylinositol-3 kinase related kinases.
  • mTOR mammalian target of rapamycin
  • Everolimus is the 40-0(2-hydroxyethyl) derivative of rapamycin (sirolimus) and works similarly to sirolimus as an mTOR inhibitor.
  • Cellular effects of everolimus include enhancement of apoptosis in some tumour cell lines as well as inhibition of cell proliferation, migration, and angiogenesis in some human cancers.
  • Oral administration of everolimus has been approved or shown effective in the treatment of a number of cancers including advanced kidney cancer (US FDA approved in March 2009), subependymal giant cell astrocytoma (SEGA) associated with tuberous sclerosis (TS) in patients who are not suitable for surgical intervention (US FDA October 2010), progressive or metastatic pancreatic neuroendocrine tumors not surgically removable (May 2011), breast cancer in post-menopausal women with advanced hormone-receptor positive, HER2-negative type cancer, in conjunction with exemestane (US FDA July 2012) and progressive, well-differentiated non-functional, neuroendocrine tumors (NET) of gastrointestinal (GI) or lung origin with unresectable, locally advanced or metastatic disease (US FDA February 2016); as well as non cancer indications such as prevention of organ rejection after liver transplant (Feb 2013), prevention of organ rejection after renal transplant (US FDA April 2010), and, tuberous sclerosis complex-associated partial-onset seizures for adult and pediatric patients aged 2 years and older (US FDA April 2018).
  • Docetaxel is a front-line therapy for metastatic castration-resistant prostate cancer (mCRPC); however, resistance to docetaxel is a key problem in current prostate cancer management seeing as half of patients do not respond to therapy, and those who initially respond ultimately relapse.
  • the mTOR pathway has been implicated in prostate cancer chemoresistance and several studies indicate that mTOR inhibition is a valid strategy for docetaxel sensitization in prostate cancer cells.
  • Formulating these drugs in nanoparticulate carriers provides a novel approach to co-deliver such anticancer drug combinations with widely differing physicochemical properties, mechanisms of action and/or routes of administration while maintaining optimal drug:drug ratios in vivo thus resulting in enhanced efficacy. Disclosure of the Invention
  • the invention relates to compositions and methods for administering effective amounts of mTOR inhibitor and taxane drug combinations using nanoparticles that are stably associated therewith at least one taxane agent and one mTOR inhibitor.
  • These compositions allow the two or more agents to be delivered to the disease site in a coordinated fashion, thereby assuring that the agents will be present at the disease site at a desired ratio.
  • stably associated is meant that the pharmacokinetics of the delivery of each drug is controlled by its association with the nanoparticle so that the administered ratio is maintained after administration for sufficient time to permit a non-antagonistic ratio administered to exert its effect.
  • the administered ratio will be maintained in the blood for 1-6 hours or at least 1-4 hours.
  • PK pharmacokinetics
  • the invention provides a nanoparticle composition for parenteral administration comprising at least one taxane and one mTOR inhibitor associated with the nanoparticles at therapeutically effective ratios especially those that are non-antagonistic.
  • the therapeutically effective non-antagonistic ratio of the agents can be determined by assessing the biological activity or effects of the agents on relevant cell culture or cell-free systems, in addition to tumor homogenates from individual patient biopsies, over a range of concentrations, as well as in vivo. Frequent combinations are docetaxel with everolimus, among other taxane derivatives together with everolimus.
  • a combination comprising a taxane derivative and an mTOR inhibitor such as everolimus or sirolimus, among other known mTOR inhibitors. Any method which results in determination of a ratio of agents which maintains a desired therapeutic effect may be used, including in vivo analysis.
  • the composition comprises at least one taxane and one mTOR inhibitor in a ratio of the taxane to mTOR inhibitor which exhibits a desired biologic effect to relevant cells in culture, cell-free systems or tumor homogenates.
  • the ratio is a mole ratio at which the agents are non-antagonistic.
  • By“relevant” cells applicants refer to at least one cell culture or cell line which is appropriate for testing the desired biological effect.
  • these agents are used as antineoplastic agents
  • “relevant” cells are those of cell lines identified by the Developmental Therapeutics Program (DTP) of the National Cancer Institute (NCI)/National Institutes of Health (NIH) as useful in their anticancer drug discovery program.
  • DTP screen utilizes 60 different human tumor cell lines.
  • tumor homogenate refers to cells generated from the homogenization of patient biopsies or tumors. Extraction of whole tumors or tumor biopsies can be achieved through standard medical techniques by a qualified physician and homogenization of the tissue into single cells can be carried out in the laboratory using a number of methods well-known in the art.
  • the invention is directed to a method to deliver a therapeutically effective amount of an mTOR inhibitontaxane derivative combination (e.g., everolimus:docetaxel) to a desired target by administering the compositions of the invention.
  • an mTOR inhibitontaxane derivative combination e.g., everolimus:docetaxel
  • the invention is also directed to a method to deliver a therapeutically effective amount of an mTOR inhibitontaxane derivative combination by administering a taxane stably associated with a first delivery vehicle and an mTOR inhibitor stably associated with a second delivery vehicle.
  • the first and second delivery vehicles may be contained in separate vials, the contents of the vials being administered to a patient simultaneously or sequentially.
  • the ratio of the mTOR inhibitor and taxane derivative is non-antagonistic.
  • the taxane and/or mTOR inhibitor are provided as a drug conjugate.
  • the invention is directed to a method to prepare a therapeutic composition comprising nanoparticles containing a ratio of at least one taxane and one mTOR inhibitor which provides a desired therapeutic effect
  • a method to prepare a therapeutic composition comprising nanoparticles containing a ratio of at least one taxane and one mTOR inhibitor which provides a desired therapeutic effect
  • method comprises providing a panel of at least one taxane and one mTOR inhibitor wherein the panel comprises at least one, but preferably a multiplicity of ratios of said drugs, testing the ability of the members of the panel to exert a biological effect on a relevant cell culture, cell-free system or tumor homogenate over a range of concentrations, selecting a member of the panel wherein the ratio provides a desired therapeutic effect on said cell culture, cell-free system or tumor homogenate over a suitable range of concentrations; and stably associating the ratio of drugs represented by the successful member of the panel into nanoparticulate drug delivery vehicles.
  • the abovementioned desired therapeutic effect is non-antagonistic (i.e. synergistic or additive).
  • the non-antagonistic ratios are selected as those that have a combination index (Cl) of ⁇ 1.1.
  • suitable nanoparticle formulations are designed such that they stably incorporate an effective amount of a taxane:mTOR inhibitor combination (e.g., docetaxeheverolimus) and allow for the sustained release of both drugs in vivo.
  • Preferred formulations contain drug conjugates of at least one of the taxane or mTOR inhibitor.
  • More preferred formulations contain drug conjugates of both a taxane and an mTOR inhibitor, such as for example, a drug conjugate of docetaxel and a drug conjugate of everolimus. Most preferred formulations contain drug conjugates of both docetaxel and everolimus wherein each is separately linked to a hydrophobic moiety such as cholesterol.
  • Figure 1 is a diagram outlining the method of the invention for determining an appropriate ratio of therapeutic agents to include in formulations.
  • Figure 2 shows the concentration of cells affected in C-33 A cells of everolimus and docetaxel alone and at 1: 1, 2:1 and 1:2 ratios. Also shown is an isobologram generated at 30% effective dose (ED) values.
  • ED effective dose
  • Figure 3A is a graph showing the concentration of docetaxel-cholesterol conjugate (Procet) remaining in the plasma (ug/mL) at various times after intravenous administration when the drug conjugate was formulated in nanoparticles.
  • Figure 3B is a graph showing % injected dose over time of docetaxel-cholesterol conjugate remaining in the plasma at various times after intravenous administration when the drug conjugate was formulated in nanoparticles.
  • Figure 3C is a graph showing the concentration of everolimus-cholesterol conjugate (ProEver) remaining in the plasma (ug/mL) at various times after intravenous administration when the drug conjugate was formulated in nanoparticles.
  • ProEver everolimus-cholesterol conjugate
  • Figure 3D is graph showing % injected dose over time of everolimus-cholesterol conjugate remaining in the plasma at various times after intravenous administration when the drug conjugate was formulated in nanoparticles.
  • Figure 4A is a graph showing the % of injected dose over time of both docetaxel- cholesterol and everolimus-cholesterol conjugates remaining in the plasma at various times after intravenous administration when the drug conjugates were formulated in PLA-PEG containing nanoparticles at a 1: 1: 12 drug:drug:polymer ratio.
  • Figure 4B is a graph showing the % of injected dose over time of both docetaxel- cholesterol and everolimus-cholesterol conjugates remaining in the plasma at various times after intravenous administration when the drug conjugates were formulated in PLA-PEG containing nanoparticles at a 1: 1:9 drug:drug:polymer ratio.
  • Figure 4C is a graph showing the % of injected dose over time of both docetaxel- cholesterol and everolimus-cholesterol conjugates remaining in the plasma at various times after intravenous administration when the drug conjugates were formulated in PLA-PEG containing nanoparticles at a 1: 1:6 drug:drug:polymer ratio.
  • Figure 5A is a graph showing the efficacy of co-loaded nanoparticles with docetaxeheverolimus drug conjugates at various ratios compared to the individual free drugs and the free drug cocktail when administered via i.v. against the PC3 (PTEN mut ) human prostate cancer cell model in nude mice.
  • mice were organized into appropriate groups consisting of a control (“Untreated”) and treatment groups including empty nanoparticles (“Vehicle”), free docetaxel (lOmg/kg), free everolimus (lOmg/kg), free cocktail of docetaxeheverolimus (10: 10 mg/kg) and docetaxeheverolimus drug conjugates co-loaded in nanoparticles at either a 1: 1, 1:5, 1: 10 or 5:1 ratios at various doses as indicated.
  • Untreated empty nanoparticles
  • Vehicle empty nanoparticles
  • free docetaxel lOmg/kg
  • free everolimus lOmg/kg
  • free cocktail of docetaxeheverolimus 10 mg/kg
  • docetaxeheverolimus drug conjugates co-loaded in nanoparticles at either a 1: 1, 1:5, 1: 10 or 5:1 ratios at various doses as indicated.
  • Figure 5B is a graph showing the efficacy of empty nanoparticles (“Vehicle”), free docetaxel (5 mg/kg), free everolimus (20 mg/kg), free drug cocktail (docetaxeheverolimus, 5 mg/kg:20mg/kg), docetaxel/everolimus co-loaded into nanoparticles (“DENP”) resulting in a final docetaxeheverolimus ratio of 1: 1 (5:5 docetaxeheverolimus mg/kg), or 5: 1 (45:9 docetaxeheverolimus mg/kg), docetaxel and /everolimus separately loaded into nanoparticles and co-administered (“NPD+NPE”) resulting in a final docetaxeheverolimus ratio of 5: 1 (15:3 docetaxeheverolimus mg/kg), and docetaxel (15 mg/kg) and everolimus ( 3 mg/kg) each separately encapsulated in nanoparticles and administered alone (“NPD” and “NPE”, respectively).
  • NPD do
  • Figure 5C is a graph showing the efficacy of empty nanoparticles (“Vehicle”), and docetaxel/everolimus co-loaded into nanoparticles (“DENP”) resulting in a final docetaxeheverolimus ratio of 1: 1 (10: 10 docetaxeheverolimus mg/kg), high dose 5: 1 (60: 12 docetaxel:everolimus mg/kg), 2: 1 (12.5:6 docetaxel :everoli mus mg/kg), and 5: 1 (15:3 docetaxekeverolimus mg/kg).
  • Figure 6 are a group of graphs showing the percent change in body weight over time for the study groups shown in Figure 5A.
  • Figure 7 is a graph showing the percent change in mean body weight for study groups given either free or encapsulated docetaxel or everolimus.
  • Figure 8 is a graph showing the efficacy of co-loaded nanoparticles with docetaxekeverolimus drug conjugates at various ratios compared to the free drug cocktail when administered via i.v. against the MDA-MB-231 (PTEN wt ) human breast adenocarcinoma cell model in nude mice.
  • Mice were organized into appropriate treatment groups consisting of empty nanoparticles (“Vehicle”), the free drug cocktail of docetaxekeverolimus (5:5 mg/kg) and docetaxekeverolimus drug conjugates co-loaded in nanoparticles (“NP D:E”) at either a 1: 1, 1:5, 1: 10 or 5: 1 ratios at various doses as indicated.
  • Figure 9 show the individual mice tumor volumes over time for the formulations shown in Figure 8 as well as for free docetaxel as indicated.
  • Figure 10A is a graph showing the efficacy of co-loaded nanoparticles with docetaxekeverolimus drug conjugates at various ratios compared to the free drug cocktail when administered via i.v. against the MDA-MB-468 (PTEN del ) human breast adenocarcinoma cell model in SCID Beige mice.
  • mice were organized into appropriate treatment groups consisting of a control (“Untreated”) and treatment groups including empty nanoparticles (“Vehicle”), free docetaxel, free everolimus, the free drug cocktail of docetaxekeverolimus (5:5mg/kg) and docetaxekeverolimus drug conjugates co-loaded in nanoparticles (“NP D:E”) at either a 1: 1, 1:5, 1: 10 or 5: 1 ratios at various doses as indicated.
  • Untreated empty nanoparticles
  • NP D:E docetaxekeverolimus drug conjugates co-loaded in nanoparticles
  • Figure 10B is a graph showing the efficacy of co-loaded nanoparticles with docetaxekeverolimus drug conjugates at various ratios compared to the free drug cocktail when administered via i.v. against the MDA-MB-468 (PTEN del ) human breast adenocarcinoma cell model in SCID Beige mice on an accelerated dosing schedule as compared to that shown in Figure 8A.
  • MDA-MB-468 PTEN del
  • Figure 12 is a graph showing the mean percent body weight change for the study groups shown in Figure 10A.
  • Figure 13 A is graph showing the concentration over time of docetaxel-cholesterol drug conjugate (“ProCet”) formulated in nanoparticles and stored at 4°C (“4C”) or room temperature (“RT”) for up to 3 months.
  • ProCet docetaxel-cholesterol drug conjugate
  • Figure 13B is graph showing the concentration over time of everolimus-cholesterol drug conjugate (“ProEver”) formulated in nanoparticles and stored at 4°C or room temperature for up to 3 months.
  • ProEver everolimus-cholesterol drug conjugate
  • compositions comprising nanoparticles stably associated therewith at least one taxane and one mTOR inhibitor, wherein the taxane and mTOR inhibitor are present at non-antagonistic taxane/mTOR inhibitor ratios.
  • nanoparticle compositions provided herein will include nanoparticles stably associated therewith at least one taxane and one mTOR inhibitor in a ratio of the taxane / mTOR inhibitor which exhibits a non-antagonistic effect to relevant cells or tumor cell homogenates.
  • nanoparticle compositions of the invention will include nanoparticles stably associated therewith docetaxel and everolimus. More preferably, docetaxel and everolimus will be present in compositions of the invention at a docetaxel: everolimus ratio of between 100:1 and 1:100 or of between 40:1 and 1:40, even more preferably the ratio of docetaxel to everolimus will be in the range of 40:1 and 1:1, 30:1 and 1:1, 20:1 and 1:1 or 10:1 and 1:1.
  • nanoparticle compositions of the invention will include nanoparticles stably associated therewith docetaxel and everolimus, wherein the ratio of docetaxel to everolimus will be in the range of 40:1, 39:1, 38:1, 37:1, 36:1, 35:1, 34:1, 33:1,
  • the ratio of docetaxel to everolimus will be in the range of 10:1 and 4:1. In some embodiments, the ratio is a mole or molar ratio. In some embodiments, the ratio is a weight ratio.
  • the above described nanoparticles are administered with a third or fourth agent. Any therapeutic, diagnostic or cosmetic agent may be included.
  • At least one of the taxane or mTOR inhibitor is provided as a drug conjugate.
  • the taxane and/or mTOR inhibitor are conjugated to a hydrophobic moiety.
  • the hydrophobic moiety is a hydrophobic polymer or a natural product such as, but not limited to, cholesterol or a C22 fatty acid.
  • the nanoparticles of the present invention may be used not only in parenteral administration but also in topical, nasal, subcutaneous, intraperitoneal, intramuscular, aerosol or oral delivery or by the application of the delivery vehicle onto or into a natural or synthetic implantable device at or near the target site for therapeutic purposes or medical imaging and the like.
  • the nanoparticles of the invention are used in parenteral administration, most preferably, intravenous administration.
  • “therapeutic agent” or“drug” as used herein refers to chemical moieties used in a variety of therapeutic, including pharmaceutical applications.
  • the solubility range of the drugs range from“insoluble” in water or buffer, to those that are“sparingly soluble” or “soluble.”
  • insoluble in aqueous medium means that the substance can be dissolved in an aqueous solution at physiological ionic strength only to the extent of 0.05 mg/ml or less. It is recognized that almost no substances are completely insoluble in aqueous medium, and that the salt concentration or osmolality of the medium may also influence solubility.
  • “sparingly soluble” and“soluble” may be described in terms of reference to either“aqueous medium” as defined above or in“pure water.” Substances that are“soluble” in aqueous medium dissolve at least to the extent of being equal to or greater than 1.0 mg/ml of the physiological solution; substances that are“sparingly soluble” in aqueous medium dissolve only to the extent of less than 1.0 mg/ml but more than 0.05 mg/ml of the physiological solution.
  • Hydrophobic moiety is defined as a moiety which is insoluble in aqueous solution as defined above.
  • the hydrophobic moiety may be a hydrophobic polymer such as
  • PCL polycaprolactone
  • PCL polycaprolactone
  • It may be monovalent - i.e., have a suitable functional group for coupling only to a single active through a linker - or may be multivalent - i.e., able to couple to multiple actives through a linker.
  • An“amphiphilic stabilizer” or“amphiphilic moiety” is a compound having a molecular weight greater than about 500 that has a hydrophilic region and a hydrophobic region. Preferably the molecular weight is greater than about 1,000, or greater than about 1,500, or greater than about 2,000. Higher molecular weight moieties, e.g., 25,000 g/mole or 50,000 g/mole, may be used. “Hydrophobic” is defined as above. “Hydrophilic” in the context of the present invention refers to moieties that have a solubility in aqueous solution ( i.e ., a physiological solution as defined above) of at least 1.0 mg/ml.
  • aqueous solution i.e ., a physiological solution as defined above
  • Typical amphiphilic stabilizers are copolymers of hydrophilic regions and hydrophobic regions.
  • the hydrophobic region if taken alone, would exhibit a solubility in aqueous medium of less than 0.05 mg/ml and the hydrophilic region, if taken alone, would exhibit a solubility in aqueous medium of more than 1 mg/ml.
  • Examples include copolymers of polyethylene glycol and polylactic acid (PLA), including poly-L-lactide (PLLA), poly-D, L-lactide (a PDLA or PLLDA); or copolymers of polyethylene glycol and poly(lactic-co-glycolic acid) (PLGA or PLG) or copolymer of polyethylene glycol and polycaprolactone (PCL).
  • PLA polyethylene glycol and polylactic acid
  • PLLA poly-L-lactide
  • a PDLA or PLLDA L-lactide
  • PCL polycaprolactone
  • the weight ratio of the hydrophobic portion to the hydrophilic portion is between 8.5 and 12.5 and/or the hydrophobic portion has a molecular weight of 8- 25 kD.
  • the ratio of the hydrophobic portion to the hydrophilic portion is 4:1, 3:1, 2:1 or 1:1 and / or the hydrophobic portion has a molecular weight of 10 or 11 or 20 or 25 kD.
  • A“linker” refers to any covalent bond, to a divalent residue of a molecule, or to a chelator (in the case where the active (drug) is a metal ion or organic metallic compound, e.g., cisplatin) that allows the hydrophobic moiety to be attached to the active agent.
  • the linker may be selectively cleavable upon exposure to a predefined stimulus, thus releasing the active agent from the hydrophobic moiety.
  • the site of cleavage in the case of the divalent residue of a molecule may be at a site within the residue, or may occur at either of the bonds that couple the divalent residue to the agent or to the hydrophobic moiety.
  • the predefined stimuli include, for example, pH changes, enzymatic degradation, chemical modification or light exposure.
  • Convenient conjugates are often based on hydrolyzable or enzymatically cleavable bonds such as esters, carbonates, carbamates, disulfides and hydrazones.
  • the conditions under which the drug performs its function are not such that the linker is cleaved, but the drug is able to perform this function while still attached to the particle.
  • the linker is described as“non-cleavable,” although virtually any linker could be cleaved under some conditions; therefore,“non-cleavable” refers to those linkers that do not necessarily need to release the drug from the particle as the active performs its function. Taxanes
  • Taxanes are a class of widely used anticancer drugs. They are diterpenes which are naturally produced by plants belonging to the Taxus genus ( e.g ., Yews). “Taxanes” as used herein includes paclitaxel (TaxolTM), docetaxel (TaxotereTM), cabazitaxel and other taxane analogs or derivatives thereof. Paclitaxel is a used for treating a range of carcinomas. Another taxane, cabazitaxel, has been approved by the FDA to treat hormone-refractory prostate cancer.
  • the anti-tubulin agent docetaxel is one of the most broadly used chemotherapies, with approvals in locally advanced or metastatic breast cancer, head and neck cancer, gastric cancer, hormone -refractory prostate cancer and non- small-cell lung cancer. It is approximately twice as potent as paclitaxel (essentially due to docetaxel’ s effect on the centrosome of the mitotic spindle), however it has similar efficacy as paclitaxel which may be due to the fact that docetaxel is prone to cellular drug resistance via a number of different mechanisms.
  • Taxane drug conjugate for use of the invention is a docetaxel-cholesterol conjugate (or“docetaxel-linker-hydrophobic moiety”) shown below:
  • mTOR inhibitor as used herein is meant to include any inhibitor with modulates activity at any point along the mTOR pathway, including the PI3K/AKT/mTOR or“PAM” pathway (note that PI3K is Phosphoinositide 3-Kinase and AKT is Protein Kinase B).
  • Such inhibitors include, for example, rapamycin (sirolimus), temsirolimus, everolimus or other rapamycin derivatives (“rapalogs”). Some act through ATP competitive inhibition while others are non- ATP inhibitors, all of which are included herein.
  • mTOR inhibitors of the invention are preferably present as a drug conjugate. More preferably the drug conjugate is provided in a nanoparticle.
  • Exemplified embodiments of mTOR inhibitor drug conjugates for use of the invention are everolimus-cholesterol drug conjugates (or “everolimus -linker-hydrophobic moiety”) linked through either a glycolate or succinate linkage such as the example shown below:
  • the preferred drug delivery approach applied here was to combine two well-known concepts, namely the use of drug conjugates and the utilization of micellar or nanoparticle delivery vehicles.
  • the goal of most prodrug or drug conjugate technologies is typically to make hydrophobic drugs more hydrophilic for increased solubility in an aqueous environment.
  • drugs are made more hydrophobic by linking them to a hydrophobic anchor (or“hydrophobic moiety”).
  • Micelles or lipophilic nanoparticle carriers are used to maintain these drug conjugates in aqueous environment since the individual drugs themselves are otherwise insoluble.
  • nanoparticle delivery systems employed in this work require the synthesis of taxane drug conjugates, docetaxel for example, to increase the hydrophobicity of the drug. While docetaxel itself is nearly insoluble in water, it possesses sufficient aqueous solubility that it can rapidly partition out of some delivery systems, typically with half-lives on the order of a few minutes.
  • the objective of this work was to control the pharmacokinetics of the formulated drug in vivo while maximizing efficacy.
  • a variety of drug conjugates were synthesized, characterized and formulated into nanoparticles to characterize the effects of iterative changes to the dmg-linker-hydrophobic moiety structural motif on partition kinetics and efficacy. A spectrum of in vitro and in vivo anti-tumor activity was observed, often dictated by the physical and chemical properties of the drug conjugate rather than the delivery vehicle itself.
  • Parameters that are likely to affect the in vivo availability of a drug when optimizing the design of formulations of the invention include: (1) the plasma elimination of the delivery vehicle; (2) the partitioning rate of the drug out of the particle; and (3) the hydrolysis rate of the drug conjugate
  • the nanoparticles remain intact upon i.v. administration, they are cleared relatively slowly from the central blood compartment, and drug conjugate hydrolysis is relatively rapid (preferably through enzymatic means rather than pH to avoid stability issues in the formulation).
  • the rate limiting process affecting drug availability in the compositions of the present invention may be the partitioning rate of the drug conjugate from the particle to the plasma.
  • a series of drug conjugates based on docetaxel and everolimus were investigated in order to achieve control of the pharmacokinetic behavior of a taxane/mTOR inhibitor drug combination in vivo.
  • Preferred hydrophobic moieties include polymers or natural products.
  • suitable hydrophobic polymeric moieties include but are not limited to polymers of the following: acrylates including methyl acrylate, ethyl acrylate, propyl acrylate, n-butyl acrylate (BA), isobutyl acrylate, 2-ethyl acrylate, and t-butyl acrylate; methacrylates including ethyl methacrylate, n-butyl methacrylate, and isobutyl methacrylate; acrylonitriles; methacrylonitrile; vinyls including vinyl acetate, vinylversatate, vinylpropionate, vinylformamide, vinylacetamide, vinylpyridines, and vinylimidazole; aminoalkyls including aminoalkylacrylates,
  • polydioxanone poly(ethylene terephthalate), poly(malic acid), poly(tartronic acid),
  • hydrophobic peptide-based polymers and copolymers based on poly(L-amino acids) Lavasanifar, A., et al, Advanced Drug Delivery Reviews (2002) 54:169-190
  • EVA poly(ethylene- vinyl acetate)
  • silicone rubber polyethylene, polypropylene, polydienes (polybutadiene, polyisoprene and hydrogenated forms of these polymers), maleic anhydride copolymers of vinyl-methylether and other vinyl ethers, polyamides (nylon 6,6), polyurethane, poly(ester urethanes), poly(ether urethanes), poly(ester- urea).
  • Particularly preferred polymeric hydrophobes include poly(ethylenevinyl acetate), poly (D,L-lactic acid) oligomers and polymers, poly (L-lactic acid) oligomers and polymers, poly (glycolic acid), copolymers of lactic acid and glycolic acid, poly (caprolactone), poly
  • Natural products as“hydrophobic moieties” should have functional groups or groups that can be converted to functional groups for conjugation including: hydrophobic vitamins (for example vitamin E, vitamins K and A), fatty acids (preferably long-chain or C18-C24 fatty acids), carotenoids and retinols (for example beta carotene, astaxanthin, trans and cis retinal, retinoic acid, folic acid, dihydrofolate, retinyl acetate, retinyl palmitate), cholecalciferol, calcitriol, hydroxycholecalciferol, ergocalciferol, i -tocopherol, ( ⁇ -tocopherol acetate, ( ⁇ -tocopherol nicotinate, cholesterol and estradiol.
  • hydrophobic vitamins for example vitamin E, vitamins K and A
  • fatty acids preferably long-chain or C18-C24 fatty acids
  • carotenoids and retinols for example beta carotene
  • hydrophobic moieties of the invention are cholesterol or a C18-C24 fatty acid.
  • it may be able to accommodate more than one, including substantially more than one drug through a multiplicity of linking sites.
  • Polymeric moieties may have as many as 100 sites whereby drugs could be linked.
  • Simpler hydrophobic moieties, such as vitamin E, may provide only one such site.
  • the number of drugs coupled to a single hydrophobic moiety may be only 1, or may be 2, 5, 10, 25, 100 and more, and all integers in between.
  • the polymers set forth above can readily be provided with a multiplicity of functional groups for coupling to the drug.
  • Difunctional hydrophobic moieties would include the hydrophobic polymer chains listed above that have two terminal OH, COOH, or NH2 groups. Multifunctional hydrophobic moieties include all of those listed above that have multiple OH, COOH, or NH2 groups on some or all of the monomer units on the polymer backbone. These functional groups are merely illustrative; other moieties which could form functional groups for linking include phenyl substituents, halo groups, and the like. Typically, when the hydrophobic moiety is a hydrophobic polymer, it may have multiple sites for linkage. When the hydrophobic moiety is a relatively small molecule, it will accommodate only the number of linkers for which it has available functional groups. Linkers or cross-linkers for use in the invention include succinate and diglycolate as well as other linkers such as those described in
  • Amphiphilic stabilizers and hydrophobic moieties for use in the invention also include those described in PCT/US2005/025549 or US Patent No. 8486924.
  • taxane agents and mTOR inhibitors will be encapsulated into nanoparticles at synergistic or additive (i.e . non-antagonistic) ratios. Determination of ratios of agents that display synergistic or additive combination effects may be carried out using various algorithms, based on the types of experimental data described below. These methods include isobologram methods (Loewe, et al., Arzneim-Forsch (1953) 3:285-290; Steel, et al., Int. J. Radiol. Oncol. Biol. Phys. (1979) 5:27-55), the fractional product method (Webb, Enzyme and Metabolic Inhibitors (1963) Vol. 1, pp.
  • Non-antagonistic ratios of two or more agents can also be determined using in vivo screening.
  • Non-antagonistic ratios of two or more agents can be determined for disease indications other than cancer and this information can be used to prepare therapeutic formulations of two or more drugs for the treatment of these diseases.
  • many measurable endpoints can be selected from which to define drug synergy, provided those endpoints are therapeutically relevant for the specific disease.
  • the in vitro studies on cell cultures will be conducted with “relevant” cells.
  • the choice of cells will depend on the intended therapeutic use of the agent.
  • In vitro studies on individual patient biopsies or whole tumors can be conducted with“tumor homogenate,” generated from homogenization of the tumor sample(s) into single cells.
  • In vivo studies will be conducted using animal studies, preferably with mice of various strains that bear a cancer cell or xenograft model.
  • Nanoparticle formulations of the invention are preferably less than 100 nm, less than 80 nm, or more preferably 20-80 nm, or even more preferably 40-80 nm in average diameter. In some embodiments, the average diameter of nanoparticle formulations of the invention are preferably less than 70 nm. In one embodiment, nanoparticles are formed by assembling into a particle at least one taxane or mTOR inhibitor drug conjugate wherein the taxane or mTOR inhibitor is coupled through a linker to a hydrophobic moiety; and at least one amphiphilic stabilizer comprising a hydrophobic portion and a hydrophilic portion.
  • the amphiphilic stabilizer is copolymer which includes polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the weight ratio of hydrophobic portion to the hydrophilic portion of the amphiphilic stabilizer is in the range of 8:5 to 12:5, preferably 10:5 (or 2: 1) and wherein the hydrophobic portion has a molecular weight of 8 kD to 25 kD, or 8 kD-20 kD or 8 kD- 15 kD. This permits ready partition from the particles of the drug conjugate(s) in intact form whereupon release of the therapeutic agent itself when the drug conjugate is liberated to the bloodstream is relatively rapid.
  • preferred examples employ drug conjugates of docetaxel and everolimus and nanoparticle delivery vehicles to facilitate pharmacokinetic control.
  • these drugs By making these drugs more hydrophobic and consequently more compatible with polymer based delivery systems, the pharmacokinetics of the drug combination compositions can be controlled. It is also possible to adjust the properties of formulations containing additional antineoplastic agents such that their effective release rates in vivo are matched to that of the two-drug combinations.
  • Nanoparticle carriers can be used to suspend these drug conjugates and other agents in an aqueous environment.
  • exemplary pharmaceutical compositions are those that comprise nanoparticles formed from a drug conjugate of a taxane and a drug conjugate of an mTOR inhibitor, which drug conjugates are conjugates of said agents each coupled to a hydrophobic moiety through a linker wherein said nanoparticles also comprise a lipid and/or an amphiphilic stabilizer.
  • no lipid is required.
  • a targeting agent is included in the nanoparticles.
  • a targeting agent is attached to the amphiphilic stabilizer.
  • the amphiphilic stabilizer is a copolymer comprising PEG.
  • the invention also includes methods to administer the above combinations using the compositions of the invention, to combine the compositions of the invention with formulations of additional antineoplastic agents and administer these and to methods of preparing these compositions and formulations.
  • the nanoparticle delivery vehicles of the present invention may be used not only in parenteral administration but also in topical, nasal, subcutaneous, intraperitoneal, intramuscular, aerosol or oral delivery or by the application of the delivery vehicle onto or into a natural or synthetic implantable device at or near the target site for therapeutic purposes or medical imaging and the like.
  • the nanoparticle delivery vehicles of the invention are used in parenteral administration, most preferably, intravenous administration.
  • the nanoparticle delivery vehicles of the present invention may be used to treat cancer or other hematological disorders including, but not limited to, breast cancer (such as locally advanced or metastatic breast cancer), lung cancer (such as non-small cell lung cancer), prostate cancer (such as hormone-refractory prostate cancer or castration-resistant prostate cance), ovarian cancer, Head and Neck Squamous Cell Carcinoma (HNSCC) or gastric/GEJ cancers (e.g. tumors of the gastroesophageal junction).
  • breast cancer such as locally advanced or metastatic breast cancer
  • lung cancer such as non-small cell lung cancer
  • prostate cancer such as hormone-refractory prostate cancer or castration-resistant prostate cance
  • ovarian cancer such as Head and Neck Squamous Cell Carcinoma (HNSCC) or gastric/GEJ cancers (e.g. tumors of the gastroesophageal junction).
  • HNSCC Head and Neck Squamous Cell Carcinoma
  • gastric/GEJ cancers e.g. tumors of the
  • the cancer or hematological disorder is associated with a PTEN (Phosphatase and Tensin Homolog) mutation.
  • PTEN is a protein that, in humans, is encoded by the PTENgene. Mutations of this gene are a step in the development of many cancers. Genes corresponding to PTEN (orthologs) have been identified in most mammals for which complete genome data are available. PTEN acts as a tumor suppressor gene through the action of its phosphatase protein product. This phosphatase is involved in the regulation of the cell cycle, preventing cells from growing and dividing too rapidly. It is a target of many cancer drugs.
  • the protein encoded by this gene is a phosphatidylinositol-3,4,5-trisphosphate 3- phosphatase. It contains a tensin-like domain as well as a catalytic domain similar to that of the dual specificity protein tyrosine phosphatases. Unlike most of the protein tyrosine phosphatases, this protein preferentially dephosphorylates phosphoinositide substrates. It negatively regulates intracellular levels of phosphatidylinositol-3,4,5-trisphosphate in cells and functions as a tumor suppressor by negatively regulating Akt/PKB signaling or the PAM pathway.
  • cell used in the present invention include both PTEN wild-type (PTEN wt ) and thus no PAM pathway activation, as well as PTEN deleted (PTEN del ) which have PAM activated.
  • mTOR inhibitors as provided in formulations of the invention may be more active in these PTEN deleted cell lines or cancers.
  • the delivery vehicle compositions of the present invention may be administered to warm-blooded animals, including humans as well as to domestic avian species.
  • a qualified physician will determine how the compositions of the present invention should be utilized with respect to dose, schedule and route of administration using established protocols.
  • Such applications may also utilize dose escalation should agents encapsulated in delivery vehicle compositions of the present invention exhibit reduced toxicity to healthy tissues of the subject.
  • the pharmaceutical compositions of the present invention are administered parenterally, i.e., intraarterially, intravenously, intraperitoneally, subcutaneously, or intramuscularly. More preferably, the pharmaceutical compositions are administered intravenously or intraperitoneally by a bolus or infusional injection.
  • compositions comprising delivery vehicles of the invention are prepared according to standard techniques and may comprise water, buffered water, 0.9% saline, 0.3% glycine, 5% dextrose, iso-osmotic sucrose solutions and the like, including glycoproteins for enhanced stability, such as albumin, lipoprotein, globulin, and the like. These compositions may be sterilized by conventional, well-known sterilization techniques. The resulting aqueous solutions may be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, and the like.
  • the delivery vehicle suspension may include polymer or lipid-protective agents which protect the nanopoarticles against free-radical and peroxidative damages on storage. Free -radical quenchers, such as alpha-tocopherol and water-soluble iron-specific chelators, such as ferrioxamine, may be used.
  • At least 85%, 90%, or 95% of each drug is retained in the nanoparticles after storage for at least 3 months, 4 months, 5 months, 6 months, or greater than 6 months when stored at room temperature or when stored at 4 deg C.
  • the concentration of delivery vehicles in the pharmaceutical formulations can vary widely, such as from less than about 0.05%, usually at or at least about 2-5% to as much as 10 to 30% by weight and will be selected primarily by fluid volumes, viscosities, and the like, in accordance with the particular mode of administration selected. For example, the concentration may be increased to lower the fluid load associated with treatment
  • the pharmaceutical compositions of the present invention are administered intravenously. Dosage for the delivery vehicle formulations will depend on the ratio of drug to polymer and the administrating physician’s opinion based on age, weight, and condition of the patient.
  • suitable formulations for veterinary use may be prepared and administered in a manner suitable to the subject.
  • Preferred veterinary subjects include mammalian species, for example, non-human primates, dogs, cats, cattle, horses, sheep, and domesticated fowl.
  • Subjects may also include laboratory animals, for example, in particular, rats, rabbits, mice, and guinea pigs.
  • compositions may be formulated separately in individual compositions wherein each of the taxane and mTOR inhibitor are stably associated with appropriate nanoparticles.
  • These compositions can be administered separately to subjects as long as the pharmacokinetics of the delivery vehicles are coordinated so that the ratio of taxane:mTOR inhibitor administered is maintained at the target for treatment.
  • kits which include, in separate containers, a first composition comprising nanoparticles stably associated with at least one taxane (or taxane derivative) and, in a second container, a second composition comprising an mTOR inhibitor (preferably nanoparticles stably associated with at least one mTOR inhibitor). The containers can then be packaged into the kit.
  • the kit will also include instructions as to the mode of administration of the compositions to a subject, at least including a description of the ratio of amounts of each composition to be administered.
  • the kit is constructed so that the amounts of compositions in each container is pre-measured so that the contents of one container in combination with the contents of the other represent the correct ratio.
  • the containers may be marked with a measuring scale permitting dispensation of appropriate amounts according to the scales visible.
  • [cpd] is the compound concentration tested hill is the Hill-coefficient. Bottom and top are the asymptotic minimum and maximum of the curve.
  • Combination Index (Cl) determination A combination index (Cl) is determined for each everolimus:docetaxel dose using Calcusyn which is based on Chou and Talalay's theory of dose-effect analysis, in which a "median-effect equation" has been used to calculate a number of biochemical equations that are extensively used in the art. Derivations of this equation have given rise to higher order equations such as those used to calculate Combination Index (Cl).
  • Cl can be used to determine if combinations of more than one drug and various ratios of each combination are antagonistic (CI>1.1), additive (0.9 ⁇ CI>1.1) or synergistic (CKO.9).
  • Cl plots are typically illustrated with Cl representing the y-axis versus the proportion of cells affected, or fraction affected (f a ), on the x-axis (see Figure 1).
  • concentrations of the two compounds cpdl and cpd2 needed to reach a certain percentage cell viability V in combination are then compared to the concentrations needed as single agents:
  • [cpdl] 50 signifies the concentration of cpdl in a mixture that gives 50% viability.
  • ICso. cpdi would signify the IC50 of cpdl alone.
  • the Cl is labelled by %-effect, to follow conventions, so CI75 signifies the Cl at 25 % viability.
  • the data from these studies show the Cl of each cell line as a function of drug:drug ratio and illustrate that particular combinations of docetaxel and everolimus are antagonistic while others are synergistic or additive.
  • Curve shift analysis This analysis provides a visual confirmation of synergy.
  • the concentrations of the mixtures of compounds 1 and 2 ( cpdl and cpd2), and the single agents, were expressed in terms of IC50 equivalents (in‘units’ of IC50):
  • the dose-response signal was fitted by a 4-parameter logistics curve using XL-fit 5 (IDBS software)
  • Isobolograms An isobologram is a dose-oriented plot which reveals whether drug combinations are synergistic. It is defined at a certain effect level, which is usually 75 %. If the single agent curves do not achieve this efficacy level, the isobologram level is set at 50 % or 30 %. If single agents do not reach the 30 % effect, no isobologram is drawn. On the axis, the calculated doses of the single compounds are plotted that give the pre-set growth effect. Both points are connected with a straight line (additivity line). For the drug combinations, it is calculated which dilutions give the pre-set growth effect and the concentrations of the individual components at this point are plotted in the isobologram. In case of an additive drug effect, the drug combination will lie close to the additivity line. In case of synergy or antagonism, the points will lie under or above the line, respectively.
  • Figure 2 shows the cell viability of C-33 A cells as a function of drug concentration for free docetaxel, free everolimus and 3 combinations of docetaxel/everolimus as both overlays and as an Isobologram with 30% effect level.
  • Docetaxel and everolimus drug conjugates were generated by separately linking each drug to cholesterol through a diglycolate linker.
  • Each drug conjugate is combined with one or more co-polymers (e.g. PLA-PEG or PLGA-PEG) to generate nanoparticles.
  • Preferred copolymers are PEG copolymers that were used at the following molecular weights (“K” refers to thousands): PLA(10K)-PEG(5K);
  • PLGA( 10K)-PEG(5K) PLGA(10K)-PEG(2K); and PLGA(15K)-PEG(5K).
  • Nanoparticles comprising PLA-PEG showed optimal size and stability at a 2: 1 PLA:PEG ratio, and PLA(10K)-PEG(5K) was used in the studies below. Different
  • Nanoparticles were generated by rapidly mixing water and a miscible solvent containing the formulation components in a confined space using carefully controlled flow rates. Under conditions where the rate of mixing exceeds the rate of precipitation, these devices have been shown to achieve homogenous particle formation kinetics, resulting in minimum sized particles for a particular formulation composition.
  • Female BDF-1 mice (7-10 days old; 18-26 grams) were administered the nanoparticle-containing drug conjugates at a drug dose of 10 mg/kg and at the indicated time points (3 mice per time point), blood was collected by cardiac puncture and placed into EDTA coated microtainers.
  • Plasma docetaxel-cholesterol and everolimus-cholesterol levels were determined by HPLC-UV methods.
  • the total drug (counts) per mouse in the blood compartment was calculated assuming a plasma volume of 0.04125 mL per gram of body weight.
  • Nanoparticle formulations containing the two drug conjugates at fixed ratios shown to be non-antagonistic in tissue culture were developed allowing extended and coordinate in vivo drug release (see Figures 3A-3D). The antitumor activity of these formulations was then evaluated in PC3 human prostate cancer mouse models ( Figure 5).
  • Taxane and everolimus drug conjugates were generated by separately linking each to cholesterol through a diglycolate linker as described in Example 2. Nanoparticles comprising PLA(10K)-PEG(5K) were formed with docetaxel-cholesterol and everolimus-cholesterol drug conjugates at either a 1: 1, 1:5, 1: 10 or 5: 1 ratio. [0117] In order to perform tumor studies in mice, animals are inoculated subq. with about lxlO 6 PC3 tumors cells which were then allowed to grow for 14 days prior to initiation of treatment.
  • mice are inoculated subcutaneously with ⁇ 1 x 10 6 tumor cells on day 0 (one inoculum/mouse) in a volume of 50 pL.
  • tumors reach a defined size of approximately 150-to-250 mm 3 , either one-day prior to treatment or on the day of treatment (-day 10-14), all tumors are measured.
  • mice are organized into appropriate groups and consist of control and treatment groups such as, saline control, vehicle control, positive control and various dilutions of test articles. Mice are injected intravenously or via oral gavage for the everolimus control agent with the required volume of sample to administer the prescribed dose to the animals based on individual mouse weights as outlined in Table 1 below:
  • Tumor growth measurements were monitored using vernier calipers beginning on the day of treatment. Tumor length measurements (mm) were made from the longest axis and width measurements (mm) will be perpendicular to this axis. From the length and width
  • tumor volumes (cm 3 ) were calculated according to the equation (L X W 2 /2). Animal weights were collected at the time of tumor measurement. [0120] Individual mouse body weights were recorded at various days (generally two days apart such as Monday, Wednesday and Friday) during the efficacy study and for a period of 14-days after the last dosing.
  • Mean body weights for each group as a function of time are derived from Mean body weights for each group as a function of time.
  • Raw data including figures and tables are generated and include tumor growth vs. time, tumor growth inhibition, and tumor growth delay.
  • mice were organized into control (“Untreated”) and treatment groups including empty nanoparticles (“Vehicle”), free docetaxel, free everolimus, free drug cocktail and docetaxel/everolimus co-loaded in PLA-PEG nanoparticles (“NPD:E”) resulting in a final docetaxel: everolimus ratio of 1: 1 (5:5 docetaxel: everolimus mg/kg), 1:5 (5:25 docetaxel: everolimus mg/kg), 1: 10 (5:50 docetaxel: everolimus mg/kg), or 5: 1 (25:5 docetaxel: everolimus mg/kg).
  • Untreated empty nanoparticles
  • NPD:E docetaxel/everolimus co-loaded in PLA-PEG nanoparticles
  • mice Female NCr nu/nu mice were dosed post tumor cell inoculations as per Table 1 for the nanoparticle formulations (free drugs were given using standard dosing schedules as listed in Table 1 and shown in Figure 65A). Animals were weighed and monitored for survival and in-life observations are collected at the time of weight measurement. Figure 5A illustrates the results of these experiments.
  • mice were dosed with increasing amounts of docetaxel and/or everolimus.
  • Treatment groups included empty nanoparticles (“Vehicle”), free docetaxel (5 mg/kg), free everolimus (20 mg/kg), free drug cocktail (docetaxekeverolimus, 5 mg/kg:20mg/kg), docetaxel/everolimus co-loaded into nanoparticles (“DENP”) resulting in a final docetaxekeverolimus ratio of 1: 1 (5:5 docetaxekeverolimus mg/kg), or 5: 1 (45:9 docetaxekeverolimus mg/kg), docetaxel and /everolimus separately loaded into nanoparticles and co-administered (“NPD+NPE”) resulting in a final docetaxekeverolimus ratio of 5: 1 (15:3 docetaxekeverolimus mg/kg), and docetaxel (15 mg/kg) and everolimus ( 3 mg/kg) each separately encapsulated in
  • mice were also organized into treatment groups for various DENP formulations at increasing drug doses.
  • treatment groups included empty nanoparticles (“Vehicle”), and docetaxel/everolimus co-loaded into nanoparticles (“DENP”) resulting in a final docetaxel: everolimus ratio of 1: 1 (10: 10 docetaxel: everolimus mg/kg), high dose 5: 1 (60: 12 docetaxel: everolimus mg/kg), 2: 1 (12.5:6 docetaxel: everolimus mg/kg), and 5: 1 (15:3 docetaxel: everolimus mg/kg).
  • the results show that increasing the dosage amount of docetaxel at the 5: 1 ratio results in enhanced efficacy compared to a 1: 1 ratio.
  • Figure 7 is a graph showing the percent change in mean body weight for treatment groups that were given either free or nanoparticle-encapsulated docetaxel or everolimus (see Table 2 below).
  • the groups were dosed using 30 mg/kg, 45 mg/kg, 60 mg/kg or 90 mg/kg of docetaxel in a nanoparticle (“DNP”) and shows that there was no impact on body weight at doses up to 60 mg/kg.
  • Other groups were dosed with 15 mg/kg, 30 mg/kg, 45 mg/kg or 60 mg/kg everolimus in a nanoparticle (“ENP”) and the graph in Figure 7 shows that there was a reduction in body weight at amounts above 15 mg/kg encapsulated everolimus.
  • the antitumor activity of nanoparticle formulations containing the two drugs at various fixed drug ratios as described above were evaluated in the human breast adenocarcinoma xenograft mouse model with the human breast cancer cell line MDA-MB-231.
  • the MDA-MB- 231 cell line is a human breast cancer cell line that was established from a pleural effusion of a 51 -year-old Caucasian female with a metastatic mammary adenocarcinoma and is one of the most commonly used breast cancer cell lines in medical research laboratories.
  • MDA-MB-231 is a "basal" type and triple negative (ER, PR and HER2 negative) breast cancer cell line and is wild type for PTEN (Phosphatase and Tensin Homolog). It is generally considered as a positive control for the MDA-MB-468 breast cancer cell line which has a PTEN deletion (discussed in Example 5).
  • mTOR rapamycin
  • PI3K/AKT a vital component of signaling pathways involving PI3K/AKT
  • Everolimus an allosteric mTOR inhibitor that inhibits the mTOR functional complex mTORCl, is approved for treatment of estrogen receptor positive (ER+) breast cancer.
  • BEZ235 and GSK2126458 are ATP competitive mTOR inhibitors targeting both PI3K and mTORCl/2; AZD8055, AZD2014 and KU-0063794 are ATP competitive mTOR inhibitors targeting both mTORCl and mTORC2; and GDC-0941 is a pan- PI3K inhibitor.
  • the level of potentiation of everolimus inhibitory activity (measured by IC 5 o values) has been found to be cell line-specific and it is known that these breast cancer cell lines are sensitive to everolimus.
  • mice were organized into treatment groups including empty nanoparticles (“Vehicle”), free drug cocktail (both drugs at 5 mg/kg or “5mpk”) and docetaxel/everolimus co-loaded in PLA-PEG nanoparticles (“NPD:E”) resulting in a final docetaxel: everolimus drug conjugate ratio of 1: 1 (5:5 docetaxel: everolimus mg/kg), 1:5 (5:25 docetaxel: everolimus mg/kg), 1: 10 (5:50 docetaxel: everolimus mg/kg), or 5: 1 (25:5 docetaxel: everolimus mg/kg).
  • Vehicle empty nanoparticles
  • free drug cocktail both drugs at 5 mg/kg or “5mpk”
  • NPD:E docetaxel/everolimus co-loaded in PLA-PEG nanoparticles
  • mice were organized into control (“Untreated”) and treatment groups including empty nanoparticles (“Vehicle”), free docetaxel (5 mg/kg), free everolimus (5 mg/kg), free drug cocktail (both 5 mg/kg) and docetaxel/everolimus drug conjugates co-loaded in PLA-PEG nanoparticles (“NPD:E” or“NP DOC:EV”) resulting in a final docetaxel: everolimus ratio of 1:1 (5:5 docetaxel: everolimus mg/kg), 1:5 (5:25 docetaxel: everolimus mg/kg), 1:10 (5:50 docetaxel: everolimus mg/kg), or 5:1 (25:5 docetaxekeverolimus mg/kg).
  • mice were dosed post tumor cell inoculations as outlined in Table 4 for the nanoparticle formulations (free drugs were given using standard dosing schedules as shown in Table 4 and Figure 10A).
  • two different dosing schedules were tested; one on days 1, 8 and 15 as indicated above and the other on a q4d dosing schedule (days 1, 5, and 9).
  • the nanoparticle formulations were dosed on days 1, 8, and 15 (free drugs were given using standard dosing schedules as shown in Figure 10B). Animals were weighed and monitored for survival and in-life observations at the time of weight measurement.
  • Figure 11 illustrates the results of these experiments for individual mice in each treatment group.
  • Figure 12 shows the mean percent body weight for each of the treatment groups. Tolerability, expressed as the percent change in body weight of the mice, from the studies presented in Examples 3-5 are summarized in Table 5 below:
  • the antitumor activity of nanoparticle formulations containing the two drugs at various fixed drug ratios as described above are evaluated in the human ovarian carcinoma xenograft mouse model with the human ovarian cancer cell line A2780.
  • the A2780 cell line is a human ovarian cancer cell line that was established from an ovarian endometroid adenocarcinoma tumor in an untreated patient so the cell line has not been exposed to any anticancer drugs or chemicals.
  • the cell line has an epithelial morphology and it is commonly used as a model to observe the effects of, and test the potency of various chemicals, methods of delivery and treatments for ovarian cancer [0141] Tumors are grown and measured as described above and female NCr nu/nu mice are similarly treated and monitored. Mice are dosed as outlined in Table 6:
  • Vehicle 1 0.5% Methylcellulose : 0.2% Tween 80 in DI Water; 0.5%
  • mice are organized into treatment groups including empty nanoparticles (“Vehicle 1” and“Vehicle 2”), free drug cocktail (everolimus at lmg/kg or 0.25 mg/kg and docetaxel at lOmg/kg) and docetaxel/everolimus co-loaded in PLA-PEG nanoparticles (“NP DOC:EV”) resulting in a final docetaxel: everolimus drug conjugate ratio of 10:1 (10:1 docetaxel: everolimus mg/kg), 20:1 (10:0.5 docetaxel: everolimus mg/kg), or 40:1 (10:0.25 docetaxel: everolimus mg/kg).
  • mice are dosed post tumor cell inoculations as per Table 6 for the nanoparticle formulations (free drugs are given using standard dosing schedules as shown in Table 6). Animals are weighed, monitored for survival, tumors are measured and in-life observations are collected at the time of measurements.
  • nanoparticle formulations comprising docetaxel and everolimus cholesterol conjugates (at a 1: 1 ratio) was monitored by HPLC after long-term storage at 4°C and room temperature (“RT”).
  • Nanoparticle formulations stored in 300 mM sucrose solution at 4°C or RT were found to be physically stable for three months by both visual inspection and DLS size measurements.
  • the concentration of docetaxel-cholesterol conjugate (“ProCet”) was maintained for up to 3 months.
  • Figure 13B shows the same results for the everolimus drug conjugate, ProEver.
  • the concentration of the drug conjugates is maintained in the dosing solution at about 95% for up to 3 months demonstrating significant retention of the drugs upon storage.
  • no particle size change was observed in either 4°C or room temperature storage conditions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Botany (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des compositions et des procédés pour l'administration améliorée de combinaisons d'agents thérapeutiques. Plus particulièrement, l'invention concerne des systèmes d'administration qui fournissent des combinaisons de taxanes et d'inhibiteurs de mTOR et de dérivés de ceux-ci, ainsi que des procédés de traitement du cancer à l'aide de ceux-ci.
PCT/US2019/065440 2018-12-10 2019-12-10 Formulations combinées de taxanes et d'inhibiteurs de mtor WO2020123481A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862777704P 2018-12-10 2018-12-10
US62/777,704 2018-12-10

Publications (1)

Publication Number Publication Date
WO2020123481A1 true WO2020123481A1 (fr) 2020-06-18

Family

ID=71077491

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/065440 WO2020123481A1 (fr) 2018-12-10 2019-12-10 Formulations combinées de taxanes et d'inhibiteurs de mtor

Country Status (1)

Country Link
WO (1) WO2020123481A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8486924B2 (en) * 2007-11-28 2013-07-16 Celator Pharmaceuticals, Inc. Taxane delivery system
US20180153863A1 (en) * 2015-06-29 2018-06-07 Abraxis Bioscience, Llc Methods of treating solid tumors using nanoparticle mtor inhibitor combination therapy
US20180207104A1 (en) * 2015-07-15 2018-07-26 Celator Pharmaceuticals, Inc. Improved nanoparticle delivery systems

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8486924B2 (en) * 2007-11-28 2013-07-16 Celator Pharmaceuticals, Inc. Taxane delivery system
US20180153863A1 (en) * 2015-06-29 2018-06-07 Abraxis Bioscience, Llc Methods of treating solid tumors using nanoparticle mtor inhibitor combination therapy
US20180207104A1 (en) * 2015-07-15 2018-07-26 Celator Pharmaceuticals, Inc. Improved nanoparticle delivery systems

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DEEPA A. RAO, DUC X. NGUYEN, GYAN P. MISHRA, BHUVANA SHYAM DODDAPANENI, ADAM W. G. ALANI: "Preparation and Characterization of Individual and Multi-drug Loaded Physically Entrapped Polymeric Micelles", JOURNAL OF VISUALIZED EXPERIMENTS, no. 102, XP055720187, DOI: 10.3791/53047 *

Similar Documents

Publication Publication Date Title
Nobili et al. Overcoming tumor multidrug resistance using drugs able to evade P‐glycoprotein or to exploit its expression
US6667337B2 (en) Combination therapy for cancer
Abouzeid et al. Polyethylene glycol-phosphatidylethanolamine (PEG-PE)/vitamin E micelles for co-delivery of paclitaxel and curcumin to overcome multi-drug resistance in ovarian cancer
KR101943230B1 (ko) 항암제로서 라파마이신 및 알부민을 포함하는 나노입자
Danhier et al. Active and passive tumor targeting of a novel poorly soluble cyclin dependent kinase inhibitor, JNJ-7706621
Graham-Gurysh et al. Synergistic drug combinations for a precision medicine approach to interstitial glioblastoma therapy
US11413253B2 (en) Nanoparticle delivery systems
JP2019178175A (ja) Hki−272とビノレルビンとを含有する抗悪性腫瘍剤の組合せ
EP1009436B1 (fr) Derive de l'acridine servant a augmenter la biodisponibilite orale du taxol
US10980891B2 (en) Oligolactic acid conjugates and micelles with enhanced anticancer efficacy
Jeswani et al. Advances in the delivery of cancer therapeutics: a comprehensive review
JP2024053046A (ja) 抗がん剤の送達のためのステレオコンプレックス
KR20210005714A (ko) 비스플루오로알킬-1,4-벤조디아제피논 화합물을 포함하는 조합 조성물 및 이의 사용 방법
JP2019112461A (ja) Nedd8活性化酵素阻害剤及び化学療法剤の投与
Rogers et al. Sequence-dependent synergism between the new generation platinum agent ZD0473 and paclitaxel in cisplatin-sensitive and-resistant human ovarian carcinoma cell lines
WO2020123481A1 (fr) Formulations combinées de taxanes et d'inhibiteurs de mtor
Edelman Novel taxane formulations and microtubule-binding agents in non–small-cell lung cancer
CN109195590A (zh) 聚合纳米颗粒
US20060216341A1 (en) Compositions for treating drug resistance
KR20180006417A (ko) 카바지탁셀 및 암 치료에 대한 그의 용도
CN114272209A (zh) 一种化学抗肿瘤药物的纳米脂质微球、以及该纳米脂质微球与肿瘤免疫治疗药物的联合应用
McClay Taxanes and Epothilones in Cancer Treatment
Bae i, United States Patent (10) Patent No.: US 8,858,965 B2

Legal Events

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

Ref document number: 19894832

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19894832

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 19894832

Country of ref document: EP

Kind code of ref document: A1