US20200360477A1 - Inhalable formulations for kinase inhibition - Google Patents

Inhalable formulations for kinase inhibition Download PDF

Info

Publication number
US20200360477A1
US20200360477A1 US16/874,190 US202016874190A US2020360477A1 US 20200360477 A1 US20200360477 A1 US 20200360477A1 US 202016874190 A US202016874190 A US 202016874190A US 2020360477 A1 US2020360477 A1 US 2020360477A1
Authority
US
United States
Prior art keywords
kinase
tyrosine kinase
kinases
receptor
oncogene
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.)
Pending
Application number
US16/874,190
Inventor
Ben Dake
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.)
Aerovate Therapeutics Inc
Original Assignee
Aerovate Therapeutics 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 Aerovate Therapeutics Inc filed Critical Aerovate Therapeutics Inc
Priority to US16/874,190 priority Critical patent/US20200360477A1/en
Assigned to AEROVATE THERAPEUTICS INC. reassignment AEROVATE THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DAKE, Ben
Publication of US20200360477A1 publication Critical patent/US20200360477A1/en
Assigned to AEROVATE THERAPEUTICS INC. reassignment AEROVATE THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DAKE, Ben
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0075Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a dry powder inhaler [DPI], e.g. comprising micronized drug mixed with lactose carrier particles
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • 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
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • 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
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the invention generally relates to inhalable formulations of kinase inhibitors to treat disease.
  • Pulmonary arterial hypertension is a condition involving elevated blood pressure in the arteries of the lungs with unknown causes and is differentiated from systemic hypertension. PAH is a progressive disease where resistance to blood flow increases in the lungs causing damage to the lungs, the pulmonary vasculature and the heart that can eventually lead to death. While symptoms are treatable with vasodilators and other medications, there is no known disease modifying therapy or cure and advanced cases can eventually require lung transplants.
  • PDGFR plays a significant role in the pathobiology of PAH and that the PDGFR-inhibiting effect of imatinib was therefore thought to contribute to its efficacy in treating PAH.
  • other tyrosine kinase inhibitors active against PDGFR have not shown the same efficacy.
  • PDGFR inhibitors such as dasatinib and nintedanib were found to induce or worsen PAH.
  • the invention is based on an in-depth molecular characterization of numerous different compounds. Results of those characterizations have led to discovery of a molecular profile associated with pulmonary arterial hypertension (PAH), which profile may be associated with other condition of the pulmonary cardiovascular system. Particularly, the invention recognizes that treating conditions such as PAH requires inhibiting activity of more than just PDGFR, and in fact requires inhibiting activity of a particular set of kinases.
  • PAH pulmonary arterial hypertension
  • compositions and methods of the invention target specific kinase combinations for inhibition to treat pulmonary and cardiovascular diseases such as PAH.
  • pulmonary and cardiovascular diseases such as PAH.
  • PDGFRs platelet-derived growth factor receptors
  • DDR1 discoidin domain receptor tyrosine kinase 1
  • Methods of the invention may include treating PAH or other pulmonary or cardiovascular diseases through the inhibition of one or more of PDGFR- ⁇ , PDGFR- ⁇ , DDR1, colony stimulating factor 1 receptor (CSF1R), tyrosine-protein kinase KIT (KIT), discoidin domain receptor tyrosine kinase 2 (DDR2), lymphocyte-specific protein tyrosine kinase (LCK), Abelson murine leukemia viral oncogene homolog 1 (ABL1), Abelson murine leukemia viral oncogene homolog 2 (ABL2), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C).
  • PDGFR- ⁇ PDGFR- ⁇
  • DDR1R colony stimulating factor 1 receptor
  • KIT tyrosine-protein kinase KIT
  • DDR2 discoidin domain receptor tyrosine kinase 2
  • LCK lymph
  • pulmonary or cardiovascular diseases such as PAH may be treated through inhibition of one or more of vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), PDGFR- ⁇ , PDGFR- ⁇ , DDR1, KIT, CSF1R, fyn related Src family tyrosine kinase (FRK), DDR2, LCK, LYN proto-oncogene, Src family tyrosine kinase (LYN), FYN proto-oncogene, Src family tyrosine kinase (FYN), and FGR proto-oncogene, Src family ty
  • methods of the invention provide effective treatments for various pulmonary and cardiovascular diseases such as PAH, pulmonary veno-occlusive disease (PVOD), idiopathic pulmonary fibrosis (IPF), and pulmonary capillary hemangiomatosis (PCH) as well as lung transplant rejection and pulmonary hypertension secondary to other diseases like heart failure with preserved ejection fraction (HFpEF) or schistosomiasis.
  • pulmonary and cardiovascular diseases such as PAH, pulmonary veno-occlusive disease (PVOD), idiopathic pulmonary fibrosis (IPF), and pulmonary capillary hemangiomatosis (PCH)
  • PAH pulmonary veno-occlusive disease
  • IPF idiopathic pulmonary fibrosis
  • PCH pulmonary capillary hemangiomatosis
  • lung transplant rejection and pulmonary hypertension secondary to other diseases like heart failure with preserved ejection fraction (HFpEF) or schistosomiasis.
  • HFpEF preserved
  • any route of administration may be applicable and certain formulations and routes of administration are exemplified here.
  • compounds are administered in inhalable form as dry powder or through a nebulizer.
  • Inhalable formulations can offer greater lung exposure than equivalent doses administered through conventional oral routes or by IV. Accordingly, where a relatively high oral dose would be required to achieve a target lung exposure, the same exposure can be achieved with much lower concentrations of drug delivered by inhalation.
  • methods of the invention circumvent some of the adverse effects associated with those high systemic concentrations. For example, as discussed above, systemically administered imatinib, while proving promising in the treatment of PAH, suffered from unacceptable rates of adverse effects including subdural hematoma.
  • certain methods of the invention provide inhalable compounds.
  • inhalable formulations of compounds A and/or B may be administered to treat cardiovascular and pulmonary diseases according to the invention.
  • formulations of the invention may be provided with a higher ratio of API (active pharmaceutical ingredient) than found in conventional formulations.
  • formulations comprising 50% or more kinase-inhibiting API are provided. Large volumes may be difficult or dangerous for patients to inhale. Therefore, minimizing the amount of non-API components in the formulation can improve patient comfort, safety, and compliance by reducing the overall amount of compound that is inhaled while still providing a therapeutically effective API concentration in target tissue.
  • compositions and methods of the invention can provide the load-reducing benefits discussed above while still delivering therapeutic results and avoiding the severe adverse events associated with other drug delivery routes.
  • the kinase inhibitor(s) or salts thereof used in the high-API compositions and methods of the invention can consist of entirely or almost entirely a single crystal form (e.g., greater than 80%, 85%, 90%, 95%, 99% or 100% of a single crystal form), thereby allowing for controlled and predictable dosing and patient response.
  • greater than 95% of the kinase inhibiting compound (e.g., compound A or B) or a salt thereof in the inhalable formulation may be present in a single crystal form.
  • inhalable kinase-inhibiting compounds may be micronized through wet or dry milling (e.g., jet milling) to achieve the desired particle size for dry powder formulations for inhalation.
  • Compounds or appropriate salts thereof may be micronized to particle sizes of about 0.5 ⁇ m to about 5 ⁇ m mass median aerodynamic diameter (MMAD) for desired deep lung penetration.
  • MMAD mass median aerodynamic diameter
  • Inhaled products may be limited in terms of the mass of powder that can be administered and certain salts will contribute significantly to the molecular weight of inhaled formulations. Accordingly, in certain embodiments, the free base of the kinase-inhibiting compound may be preferred over any salts thereof for efficient delivery of the active moiety to lung tissue. If required, various excipients or carriers can be added to the kinase inhibitor(s) or salts thereof before or after micronization depending on application.
  • carriers, excipients, conditioners, and force control agents such as lactose (which when used as a carrier may be conditioned with various solvents to increase separation of imatinib during inhalation), magnesium stearate, leucine, isoleucine, dileucine, trileucine, lecithin, distearylphosphatidylcholine (DSPC) or other lipid-based carriers, or various hydrophilic polymers where they exhibit appropriate physico-chemical properties may be included.
  • excipients or carriers are optional and that many embodiments of the invention do not require excipients or carriers.
  • API:carrier ratios may be greater than 50:50, 75:25, or 90:10. Additional ratios are contemplated as discussed below.
  • methods of the invention may include administering kinase-inhibiting inhalable formulations that exclude all or most amorphous forms of the compounds. Because crystal form can be important to drug pharmacokinetics and dosing, as well as physicochemical stability and avoiding amorphous content can therefore be important to providing predictable and efficient therapy.
  • Treatment methods of the invention may include the administration of kinase-inhibiting compounds to treat a variety of pulmonary and cardiovascular diseases. Doses may vary depending on the characteristics of the compound used (e.g., its kinase-binding profile) and the disease being treated. In various embodiments, dose ranges can include between about 10 mg to about 100 mg per dose for inhalation on a twice to four times per day schedule. About 0.1 mg to about 80 mg of the active imatinib compound may then be deposited within the lungs after inhalation.
  • the use of relatively high concentrations of API e.g., 50% or greater allows for the above doses to be achieved with less overall volume of inhalable compared to conventional formulations having 1%-3% API.
  • Methods of the invention may include administration of spray-dried kinase-inhibiting compounds or salts thereof for inhalation. While carriers such as lactose may be used after micronization to aid in delivery via inhalation, those carriers may generally comprise larger diameter particles and complication in the separation of the active imatinib compound may result in lower amounts of the inhaled compound reaching the lungs. Furthermore, the amount of active compound reaching the lungs may be less predictable using such carriers and methods, making dosing more complicated.
  • spray-dried methods may be used wherein the active kinase-inhibiting compound(s) or salts thereof along with various excipients or other additives may be micronized to a desired particle size and suspended or solubilized for spray-drying and inhalation.
  • the micronized kinase-inhibiting compound(s) may be suspended in a feedstock for the purposes of spray-drying to avoid the creation of amorphous or polymorphic imatinib content that may occur if dissolved in a solution (e.g. in an appropriate organic solvent or within an acidified aqueous solution) upon spray-drying.
  • a solution e.g. in an appropriate organic solvent or within an acidified aqueous solution
  • the inhalable formulation can retain the desired crystal structure, particle size, and low levels of amorphous content obtained before the micronization process.
  • Stable suspensions for spray-drying may be obtained through manipulation of factors affecting the solubility of the active compound such as pH, ionic strength, and dispersing agents or surfactants.
  • Excipients that may be used before micronization in the spray-drying methods described above include, for example, leucine, dileucine, trileucine, bulking agents such as trehalose or mannitol, lecithin, DSPC or other lipid-based carriers, citrate, or acetate.
  • aspects of the invention include methods of treating pulmonary arterial hypertension (PAH) that may comprise providing to a subject a therapeutically effective amount of an inhalable formulation of a compound in order to inhibit activity of a plurality of kinases comprising one or more platelet-derived growth factor receptors (PDGFRs) and discoidin domain receptor tyrosine kinase 1 (DDR1).
  • PDGFRs platelet-derived growth factor receptors
  • DDR1 discoidin domain receptor tyrosine kinase 1
  • the one or more PDGFRs can include PDGFR- ⁇ .
  • the one or more PDGFRs may include PDGFR- ⁇ .
  • the plurality of kinases can include colony stimulating factor 1 receptor (CSF1R).
  • CSF1R colony stimulating factor 1 receptor
  • the plurality of kinases may include tyrosine-protein kinase KIT (KIT).
  • KIT tyrosine-protein kinase KIT
  • DDR2 discoidin domain receptor tyrosine kinase 2
  • Methods of the invention may include inhibiting lymphocyte-specific protein tyrosine kinase (LCK).
  • the plurality of inhibited kinases can comprise at least Abelson murine leukemia viral oncogene homolog 1 (ABL1), Abelson murine leukemia viral oncogene homolog 2 (ABL2), colony stimulating factor 1 receptor (CSF1R), discoidin domain receptor tyrosine kinase 2 (DDR2), tyrosine-protein kinase KIT (KIT), lymphocyte-specific protein tyrosine kinase (LCK), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C).
  • ABL1 Abelson murine leukemia viral oncogene homolog 1
  • ABL2 Abelson murine leukemia viral oncogene homolog 2
  • CSF1R colony stimulating factor 1 receptor
  • DDR2 discoidin domain receptor tyrosine kinase 2
  • KIT tyrosine-protein kinase KIT
  • Certain aspects of the invention include methods of treating pulmonary arterial hypertension (PAH) that can include providing to a subject a therapeutically effective amount of an inhalable formulation of a compound in order to inhibit activity of two or more of vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), platelet-derived growth factor receptor ⁇ (PDGFR- ⁇ ), platelet-derived growth factor receptor ⁇ (PDGFR- ⁇ ), discoidin domain receptor tyrosine kinase 1 (DDR1), tyrosine-protein kinase KIT (KIT), colony stimulating factor 1 receptor (CSF1R), fyn related Src family ty
  • the invention relates to the treatment of various pulmonary and cardiovascular diseases through the targeted inhibition of specific combinations of kinases.
  • Diseases including pulmonary arterial hypertension (PAH), pulmonary veno-occlusive disease (PVOD), idiopathic pulmonary fibrosis (IPF), and pulmonary capillary hemangiomatosis (PCH) may be treated as well as lung transplant rejection and pulmonary hypertension secondary to other diseases like heart failure with preserved ejection fraction (HFpEF) or schistosomiasis using inhalable formulations of imatinib and salts thereof.
  • PAH pulmonary arterial hypertension
  • PVOD pulmonary veno-occlusive disease
  • IPF idiopathic pulmonary fibrosis
  • PCH pulmonary capillary hemangiomatosis
  • HFpEF preserved ejection fraction
  • schistosomiasis using inhalable formulations of imatinib and salts thereof.
  • methods of the invention can include targeted inhibition of combinations of Abelson murine leukemia viral oncogene homolog 1 (ABL1), colony stimulating factor 1 receptor (CSF1R), discoidin domain receptor tyrosine kinase 1 (DDR1), discoidin domain receptor tyrosine kinase 2 (DDR2), tyrosine-protein kinase KIT (KIT), lymphocyte-specific protein tyrosine kinase (LCK), platelet-derived growth factor receptor- ⁇ (PDGFR- ⁇ ), and platelet-derived growth factor receptor- ⁇ (PDGFR- ⁇ ).
  • ABL1 Abelson murine leukemia viral oncogene homolog 1
  • CSF1R colony stimulating factor 1 receptor
  • DDR1 discoidin domain receptor tyrosine kinase 1
  • DDR2 discoidin domain receptor tyrosine kinase 2
  • KIT tyrosine-protein kinase KIT
  • methods of the invention can include targeted inhibition of combinations of PDGFR- ⁇ , PDGFR- ⁇ , DDR1, CSF1R, KIT, DDR2, LCK, ABL1, Abelson murine leukemia viral oncogene homolog 2 (ABL2), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C).
  • PDGFR- ⁇ PDGFR- ⁇
  • DDR1R DDR1, CSF1R, KIT, DDR2, LCK
  • ABL1 Abelson murine leukemia viral oncogene homolog 2
  • ABL2 Abelson murine leukemia viral oncogene homolog 2
  • PI42C phosphatidylinositol 5-phosphate 4-kinase type-2 gamma
  • methods of the invention can include targeted inhibition of combinations of vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), PDGFR- ⁇ , PDGFR- ⁇ , DDR1, KIT, CSF1R, fyn related Src family tyrosine kinase (FRK), DDR2, LCK, LYN proto-oncogene, Src family tyrosine kinase (LYN), FYN proto-oncogene, Src family tyrosine kinase (FYN), and FGR proto-oncogene, Src family tyrosine kinase (VEGFR-2
  • methods of the invention may include targeted inhibition of combinations of PDGFR- ⁇ , PDGFR- ⁇ , DDR1, CSF1R, KIT, DDR2, LCK, ABL1, ABL2, and PI42C while not significantly inhibiting (e.g. less than 500 nM K d ) one or more of VEGFR-2, HCK, FLT4, RET, SRC, FRK, LYN, FYN, and FGR.
  • methods of the invention may include targeted inhibition of combinations of VEGFR-2, HCK, FLT4, RET, SRC, PDGFR- ⁇ , PDGFR- ⁇ , DDR1, KIT, CSF1R, FRK, DDR2, LCK, LYN, FYN, and FGR while not significantly inhibiting (e.g. less than 500 nM K d ) one or more of ABL2, and PI42C.
  • methods of the invention may include targeted inhibition of any combination of the above kinases via administration of a compound having an equilibrium dissociation constant (K d ) of less than about 1000 nM, 500 nM, less than about 400 nM, less than about 300 nM, less than about 200 nM, less than about 100 nM, less than about 75 nM, less than about 50 nM, less than about 40 nM, less than about 30 nM, less than about 20 nM, less than about 15 nM, or less than about 5 nM with respect to each of the targeted kinases.
  • K d equilibrium dissociation constant
  • Compound A as characterized below, is free base imatinib having the following structure:
  • the kinase-binding profile including the equilibrium dissociation constant (K d ) was determined for compound A as detailed in Example 1 below. The results of that characterization are shown in Table 1.
  • Compound B as characterized below, has the following structure:
  • the kinase-binding profile including the equilibrium dissociation constant (K d ) was determined for compound B as detailed in Example 3 below. The results of that characterization are shown in Table 2.
  • both compounds significantly bind and therefore inhibit a number of the same kinases while each further inhibits additional kinases not significantly affected by the other compound.
  • methods of the invention are able to identify target kinases for inhibition that are common to both compounds and that, therefore may be more likely to provide a therapeutic effect in treating cardiovascular or pulmonary diseases.
  • differences in kinase-inhibition between the two compounds may account for differences in compound efficacy in treating certain pulmonary and cardiovascular diseases and account for various adverse effects. Accordingly, in certain embodiments, methods of the invention may target only kinases inhibited by one or the other of the compounds to improve treatment outcomes and reduce the risk of adverse effects.
  • methods and compositions described herein may provide greater concentrations of a kinase-inhibiting compound in target lung tissue than obtained with equivalent doses administered orally or through IV.
  • those doses comprising a high percentage of the overall formulation, can be delivered in lower volume formulations than conventional formulations of between 1% and 3% API. Reducing the volume a patient must inhale can increase patient comfort and compliance, thereby improving results. Additionally, a higher percentage of API content can improve the API distribution and blend uniformity.
  • compositions of the invention allow for treatment of conditions of the pulmonary cardiovascular system (e.g., PAH) with lower doses and less inhalable volume than would be required in systemic administration, thereby lowering the risk of adverse events including subdural hematoma (See, Frost et al.).
  • PAH pulmonary cardiovascular system
  • the invention provides viable treatment methods for life threatening diseases that were heretofore too risky for practical application.
  • compounds of the invention may include formulations of a kinase inhibitor such as imatinib or salts thereof targeting combinations of kinases as discussed above.
  • the free base of the kinase-inhibiting compound may be used in a formulation (either in dry powder or suspension) for inhalation to treat a condition of the pulmonary cardiovascular system such as PAH.
  • Certain salt forms are also contemplated.
  • kinase inhibitor salts that were found to exhibit suitable thermal stability and few or single polymorphic forms include glycollate, isethionate, malonate, tartrate, and malate.
  • salt forms contemplated herein are xinafoate, furoate, trifenatate, HCl, sulfate, phosphate, lactate, maleate, fumarate, succinate, adipate, mesylate, and citrate.
  • formulations can comprise at least 50% of a kinase-inhibiting compound or a salt thereof.
  • formulations of the invention may include one or more excipients.
  • Excipients may include, for example, lactose in various forms (e.g., roller dried or spray dried). Larger lactose particles can be used as a carrier for inhalation of micronized formulations. The carrier particles, with their larger size, can be used to increase aerodynamic forces on the combined kinase inhibitor/carrier in order to aid in delivery through inhalation.
  • Solvents may be used to condition the lactose surface such that the active component can be effectively separated from the lactose as it leaves the inhaler device and within the oral cavity when being used as a carrier.
  • Magnesium stearate can be used as a force-control agent or conditioning agent in various embodiments.
  • leucine can be used as a force-control agent including different forms of leucine (e.g. isoleucine) along with dileucine and even trileucine.
  • Lecithin phospholipids such as DSPC may be used as an excipient for dry powder inhalation.
  • excipients may include various hydrophilic polymers. See, for example, Karolewicz, B., 2016, A review of polymers as multifunctional excipients in drug dosage form technology, Saudi Pharm J., 24(5):525-536, incorporated herein by reference.
  • inhalable formulations may have API:carrier ratios of 50:50, 55:45, 60:40, 65:35, 70:30, 75:25, 80:20, 85:15, 90:10, or 95:5.
  • Certain inhalable formulations may be pure API with no additional components.
  • formulations may include a kinase inhibitor or salts thereof as the API in amounts greater than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, or 45%.
  • API ratios refer to % w/w.
  • micronized kinase inhibitor and salts thereof retain crystallinity, even after micronization and spray drying (as discussed in detail below).
  • kinase inhibiting formulations of the invention can include less than 50%, less than 25%, less than 20%, less than 10%, less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, or less than 1% amorphous API content by mass.
  • formulations of the invention include no observable amorphous content of the kinase-inhibiting compound.
  • the desired crystalline form and low amorphous content obtained during micronization is carried through to the spray-dried inhalable powder because the kinase inhibitor crystals may not be dissolved in the solution to a significant degree.
  • kinase inhibitor formulations of the invention may be significantly less hygroscopic than conventional salt compounds such as imatinib mesylate. Accordingly, the formulations of the invention are better suited for dry powder inhalation and can comprise less than 5% water content, less than 4%, less than 3%, less than 2%, or, in preferred embodiments, less than 1% water content.
  • inhalable formulations of the invention include kinase inhibitors or salts thereof present in a single crystal form.
  • the kinase inhibitor or a salt thereof may be present at greater than 75%, 80%, 85%, 90%, 95%, or, in preferred embodiments, greater than 99% in a single crystal form by mass.
  • the single crystal form may be, for example, imatinib in type A or type B in various embodiments. Crystalline purity can be estimated using any known method including, for example, x-ray powder diffraction (XRPD).
  • kinase inhibitors or salts thereof are provided in dry powder formulations for inhalation.
  • Dry powder can be administered via, for example, dry powder inhalers such as described in Berkenfeld, et al., 2015, Devices for Dry Powder Drug Delivery to the Lung, AAPS PharmaSciTech, 16(3):479-490, incorporated herein by reference.
  • Dry powder compounds may be divided into single doses for single, twice daily, three times daily, or four times daily inhalation to treat disorders such as PAH or other conditions of the pulmonary cardiovascular system.
  • the single doses may be divided into individual capsules or other formats compatible with the dry powder inhaler to be used.
  • suspensions having the characteristics described herein can be delivered via inhalation using, for example, a nebulizer.
  • Suspensions may offer advantages over solutions as discussed below.
  • micronization and particle diameter may be of particular importance for efficient delivery and the active kinase-inhibiting compound may be preferably micronized to a mass median diameter of 2 ⁇ m or less.
  • the suspension solution for nebulizer inhalation can be aqueous and doses may be divided into individual containers or compartments for sterile storage prior to use.
  • Micronized kinase inhibitor particle size can range from about 0.5 ⁇ m to about 5 ⁇ m depending on application (e.g., dry powder or suspension for inhalation). In preferred embodiments the size range is about 1 ⁇ m to about 3 ⁇ m in dry powder formulations to achieve deep lung penetration.
  • Dosages for treating PAH and other conditions of the pulmonary cardiovascular system may be in the range of between about 10 mg to about 100 mg per dose for inhalation on once, twice or three times per day schedule.
  • About 0.1 mg to about 80 mg of the kinase inhibitor(s) or salts thereof may then be deposited within the lung after inhalation.
  • about 10 mg to 30 mg of kinase-inhibiting compound may be given in a capsule for a single dry-powder inhalation dose with about 5 mg to about 10 mg of the compound to be expected to reach the lungs.
  • kinase inhibitor may be present at about 0.1 to about 1 mg/kg in a dose and may be administered one to four times a day to obtain the desired therapeutic results.
  • the kinase-inhibiting methods of the invention may be used to treat pulmonary hypertension as a result of schistosomiasis. See, for example, Li, et al., 2019, The ABL kinase inhibitor imatinib causes phenotypic changes and lethality in adult Schistosoma japonicum , Parasitol Res., 118(3):881-890; Graham, et al., 2010, Schistosomiasis-associated pulmonary hypertension: pulmonary vascular disease: the global perspective, Chest, 137(6 Suppl):20S-29S, the content of each of which is incorporated herein by reference.
  • Methods and compositions of the invention may be used to treat lung transplant recipients to prevent organ rejection. See, Keil, et al., 2019, Synergism of imatinib, vatalanib and everolimus in the prevention of chronic lung allograft rejection after lung transplantation (LTx) in rats, Histol Histopathol, 1:18088, incorporated herein by reference.
  • LTx chronic lung allograft rejection after lung transplantation
  • compositions described herein can be used to treat pulmonary veno-occlusive disease (PVOD).
  • PVOD pulmonary veno-occlusive disease
  • compounds and methods of the invention may be used to provide greater concentration at the target lung tissue through inhalation along with consistent, predictable pharmacokinetics afforded by low polymorphism and amorphous content.
  • the efficient localization of therapeutic compound at the target tissue allows for lower systemic exposure and avoidance of the adverse events associated with prolonged oral administration of certain kinase inhibitors such as imatinib mesylate.
  • Methods of the invention can include preparation of kinase-inhibiting formulations.
  • kinase inhibitors or salts thereof may be administered via inhalation in suspension or dry powder form.
  • Dry powder formulations may be obtained via any known method including, in preferred embodiments, jet milling. Jet milling can be used to grind active compounds and, potentially, various additives (e.g., excipients) using a jet (or jets) of compressed air or gas to force collisions between the particles as they transit at near sonic velocity around the perimeter of a toroidal chamber. The size reduction is the result of the high-velocity collisions between particles of the process material.
  • Outputs of the jet mill may allow particles to exit the apparatus once a desired size has been reached.
  • desired particle size for dry powder inhalation and other formulations may be in the range of about 0.5 ⁇ m to about 5 ⁇ m.
  • bulk compounds may be micronized to the desired size for inhalation via wet milling wherein the kinase inhibitor particles are suspended in a slurry and reduced through shearing or impact with a grinding media.
  • An unexpected finding of the invention is that, once micronized, free base kinase-inhibiting compounds retain crystallinity and are considerably less hygroscopic than certain salt forms (e.g., imatinib mesylate). Furthermore, micronized imatinib obtained using methods of the invention has been found to exhibit no apparent polymorphs other than the designated Type A and very low levels of amorphous content. Accordingly, this can result in improved stability of the drug substance and any drug product upon storage. Single crystal forms of imatinib or other kinase inhibitors such as described may allow for more predictable in vivo behavior and appropriate dosing can be determined.
  • kinase inhibitor formulations of the invention can be prepared for inhalation.
  • the dry powder kinase inhibitor can be combined with larger carrier particles such as lactose as discussed above.
  • a suspension can be formed of the kinase inhibitor compound(s).
  • the suspension may result from dry micronization followed by suspension of the resulting dry powder or can be obtained as the outcome of a wet milling procedure.
  • Suspensions of micronized crystal forms may be used in nebulized inhalation treatment or may be spray dried for dry powder treatments.
  • Spray drying methods may follow the following procedure. First, bulk kinase inhibitor compound may be micronized as described above to obtain particles in a desired size range. Then the micronized compound can be suspended in a solution such that it does not dissolve and instead retains the desired crystalline features (e.g., low polymorphism and amorphous content). The suspended particles can then be spray dried using any known method. Spray drying techniques are well characterized and described, for example, in Ziaee, et al., 2019, Spray drying of pharmaceuticals and biopharmaceuticals: Critical parameters and experimental process optimization approaches, Eur. J. Pharm. Sci., 127:300-318, and Weers et al., 2019, AAPS PharmSciTech. 2019 Feb. 7; 20(3):103.
  • Spray drying micronized kinase inhibitor compounds or salts thereof provides for uniform and predictable crystallinity and particle size and can avoid the need for large carrier molecules that may adversely affect the amount of inhaled drug that reaches the target lung tissue.
  • micronized drug particles may be suspended within a non-aqueous solvent or within an emulsion of a non-aqueous solvent which, in turn is emulsified or dispersed within an aqueous environment (e.g. oil in water) and spray-dried, resulting in crystalline drug particles.
  • the non-aqueous component may or may not be fugitive and thus could be removed completely during spray drying or, it could be retained, depending on the desired properties required.
  • each atomized droplet (mass median diameter ⁇ 10 ⁇ m) contains dispersed drug crystals. During the initial moments of the drying process, the more volatile aqueous phase begins to evaporate.
  • the rapidly receding atomized droplet interface drives enrichment of the slowly diffusing drug and emulsion particles at the interface. This leads to formation of a void space in the center of the drying droplet. As the drying process continues, the less volatile oil phase in the emulsion droplets evaporates, resulting in formation of hollow pores in their place. Overall, the resulting hollow spray-dried composite particles contain drug crystals.
  • formulation methods include manipulation of the suspension to prevent dissolution of the kinase inhibitor compound.
  • Aqueous solution factors such as pH, ionic strength and dispersing agents may be used to obtain a stable suspension for nebulized inhalation or spray drying.
  • the pH of the aqueous solution may be adjusted to prevent dissolution.
  • salt in the aqueous solution may be used to reduce solubility of the active compound crystals in certain embodiments.
  • a dispersing agent or surfactant e.g., Tween 20 or Tween 80
  • Tween 20 or Tween 80 may be added but should not cause dissolution of the kinase inhibitor in suspension.
  • excipients can be added to the suspension before spray drying.
  • the excipient may be a water-soluble excipient, such as leucine, dileucine, trileucine, trehalose, mannitol, citrate or acetate.
  • the excipient may be a water insoluble excipient, such as lecithin, distearylphosphatidylcholine (DSPC) or limonene.
  • DSPC distearylphosphatidylcholine
  • limonene such insoluble excipients may be dissolved in a non-aqueous medium that is miscible or immiscible with water, thereby creating an emulsion.
  • a liposomal dispersion could be created into which the suspended kinase inhibitor could be added and homogenized or where it could be spray dried in separate feedstocks.
  • each agent can readily be determined by the skilled person, having regard to typical factors such as the age, weight, sex and clinical history of the patient.
  • a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce the desired therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • compositions of the invention include a “therapeutically effective amount” or a “prophylactically effective amount” of one or more of the compounds of the present invention, or functional derivatives thereof.
  • An “effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, e.g., a diminishment or prevention of effects associated with PAH.
  • a therapeutically effective amount of a compound of the present invention or functional derivatives thereof may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the therapeutic compound to elicit a desired response in the subject.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the therapeutic agent are outweighed by the therapeutically beneficial effects.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to, or at an earlier stage of disease, the prophylactically effective amount may be less than the therapeutically effective amount. A prophylactically or therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the beneficial effects.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g. a therapeutic or prophylactic response). For example, a single inhalable bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigency of the therapeutic situation.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, without being toxic to the patient.
  • dosage unit refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the compound, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • therapeutically effective amount can be estimated initially either in cell culture assays or in animal models, usually rats, non-human primates, mice, rabbits, dogs, or pigs.
  • the animal model is also used to achieve a desirable concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in other subjects.
  • the therapeutically effective amount is sufficient to reduce PAH symptoms in a subject. In some embodiments, the therapeutically effective amount is sufficient to eliminate PAH symptoms in a subject.
  • Dosages for a particular patient can be determined by one of ordinary skill in the art using conventional considerations, (e.g. by means of an appropriate, conventional pharmacological protocol).
  • a physician may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the dose administered to a patient is sufficient to effect a beneficial therapeutic response in the patient over time, or, e.g., to reduce symptoms, or other appropriate activity, depending on the application.
  • the dose is determined by the efficacy of the particular formulation, and the activity, stability, or half-life of the compounds of the invention or functional derivatives thereof, and the condition of the patient, as well as the body weight or surface area of the patient to be treated.
  • the size of the dose is also determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular vector, formulation, or the like in a particular subject.
  • Therapeutic compositions comprising one or more compounds of the invention or functional derivatives thereof are optionally tested in one or more appropriate in vitro and/or in vivo animal models of disease, such as models of PAH, to confirm efficacy, tissue metabolism, and to estimate dosages, according to methods well known in the art.
  • dosages can be initially determined by activity, stability or other suitable measures of treatment vs. non-treatment (e.g., comparison of treated vs. untreated cells or animal models), in a relevant assay.
  • Administration can be accomplished via single or divided doses.
  • the aqueous suspension may contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents dispersing or wetting agents such as a naturally occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such a polyoxyethylene with partial esters derived from fatty acids and hexitol anhydrides, for example polyoxyethylene sorbitan monooleate.
  • suspending agents dispersing or wetting agents
  • a naturally occurring phosphatide for example lecithin
  • condensation products of an alkylene oxide with fatty acids for example polyoxyethylene stearate
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, mannitol, or trehalose.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents such as sucrose, mannitol, or trehalose.
  • sweetening agents such as sucrose, mannitol, or trehalose.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • composition means a composition comprising a compound as described herein and at least one component comprising pharmaceutically acceptable carriers, diluents, adjuvants, excipients, or vehicles, such as preserving agents, taste-masking agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • pharmaceutically acceptable carriers such as preserving agents, taste-masking agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • pharmaceutically acceptable carrier is used to mean any carrier, diluent, adjuvant, excipient, or vehicle, as described herein.
  • suspending agents include ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances.
  • Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like.
  • suitable carriers, diluents, solvents, or vehicles include water, ethanol, polyols, suitable mixtures thereof, vegetable oils (such as olive oil), and organic esters such as ethyl oleate.
  • excipients include lactose, milk sugar, sodium citrate, calcium carbonate, and dicalcium phosphate.
  • disintegrating agents include starch, alginic acids, and certain complex silicates.
  • lubricants include magnesium stearate, sodium lauryl sulphate, talc, as well as high molecular weight polyethylene glycols.
  • pharmaceutically acceptable means it is, within the scope of sound medical judgment, suitable for use in contact with the cells of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
  • imatinib compound A was screened against 468 quantifiable kinases in the human kinome (the set of protein kinases encoded in the human genome) to characterize binding affinity for each kinase.
  • KINOMEscanTM is based on a competition binding assay that quantitatively measures the ability of a compound to compete with an immobilized, active-site directed ligand. The assay is performed by combining three components: DNA-tagged kinase; immobilized ligand; and a test compound. The ability of the test compound to compete with the immobilized ligand is measured via quantitative PCR of the DNA tag.
  • kinases-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32° C. until lysis. The lysates were centrifuged and filtered to remove cell debris. Some kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection.
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the ligand-bound beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding.
  • Binding reactions were assembled by combining kinases, ligand-bound affinity beads, and test compounds in 1 ⁇ binding buffer (20% SeaBlock, 0.17 ⁇ PBS, 0.05% Tween 20, 6 mM DTT).
  • Test compounds were prepared as 111 ⁇ stocks in 100% DMSO.
  • K d s were determined using an 11-point 3-fold compound dilution series with three DMSO control points. All compounds for K d measurements were distributed by acoustic transfer (non-contact dispensing) in 100% DMSO.
  • the compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.9%. All reactions were performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml.
  • the assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1 ⁇ PBS, 0.05% Tween 20).
  • the beads were then re-suspended in elution buffer (1 ⁇ PBS, 0.05% Tween 20, 0.5 ⁇ M non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes.
  • the kinase concentration in the eluates was measured by qPCR.
  • Compound B was screened against 468 quantifiable kinases in the human kinome (the set of protein kinases encoded in the human genome) to characterize binding affinity for each kinase.
  • KINOMEscanTM is based on a competition binding assay that quantitatively measures the ability of a compound to compete with an immobilized, active-site directed ligand. The assay is performed by combining three components: DNA-tagged kinase; immobilized ligand; and a test compound. The ability of the test compound to compete with the immobilized ligand is measured via quantitative PCR of the DNA tag.
  • kinases-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32° C. until lysis. The lysates were centrifuged and filtered to remove cell debris. Some kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection.
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the ligand-bound beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding.
  • Binding reactions were assembled by combining kinases, ligand-bound affinity beads, and test compounds in 1 ⁇ binding buffer (20% SeaBlock, 0.17 ⁇ PBS, 0.05% Tween 20, 6 mM DTT).
  • Test compounds were prepared as 111 ⁇ stocks in 100% DMSO.
  • K d s were determined using an 11-point 3-fold compound dilution series with three DMSO control points. All compounds for K d measurements were distributed by acoustic transfer (non-contact dispensing) in 100% DMSO.
  • the compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.9%. All reactions were performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml.
  • the assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1 ⁇ PBS, 0.05% Tween 20).
  • the beads were then re-suspended in elution buffer (1 ⁇ PBS, 0.05% Tween 20, 0.5 ⁇ M non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes.
  • the kinase concentration in the eluates was measured by qPCR.

Abstract

The invention relates to inhalable formulations configured to inhibit target combinations of kinases for the treatment of cardiovascular and pulmonary diseases such as pulmonary arterial hypertension (PAH).

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of, and priority to, U.S. Provisional Application Nos. 62/849,054, filed May 16, 2019; 62/849,056, filed May 16, 2019; 62/849,058, filed May 16, 2019; 62/849,059, filed May 16, 2019; 62/877,575, filed Jul. 23, 2019; 62/942,408, filed Dec. 2, 2019; 62/984,037, filed Mar. 2, 2020; and 62/958,481, filed Jan. 8, 2020; the content of each of which is hereby incorporated by reference herein in its entirety.
  • FIELD OF THE INVENTION
  • The invention generally relates to inhalable formulations of kinase inhibitors to treat disease.
  • BACKGROUND
  • Pulmonary arterial hypertension (PAH) is a condition involving elevated blood pressure in the arteries of the lungs with unknown causes and is differentiated from systemic hypertension. PAH is a progressive disease where resistance to blood flow increases in the lungs causing damage to the lungs, the pulmonary vasculature and the heart that can eventually lead to death. While symptoms are treatable with vasodilators and other medications, there is no known disease modifying therapy or cure and advanced cases can eventually require lung transplants.
  • It has been hypothesized that PDGFR plays a significant role in the pathobiology of PAH and that the PDGFR-inhibiting effect of imatinib was therefore thought to contribute to its efficacy in treating PAH. However, other tyrosine kinase inhibitors active against PDGFR have not shown the same efficacy. In fact, in certain instances PDGFR inhibitors such as dasatinib and nintedanib were found to induce or worsen PAH.
  • Therefore, up to this point, the exact mechanism of action for treating PAH has remained unknown. Similarly, it is not known what combination of kinases might be linked to PAH proliferation and might prove useful treatment targets. Accordingly, an effective treatment for PAH remains elusive.
  • SUMMARY
  • The invention is based on an in-depth molecular characterization of numerous different compounds. Results of those characterizations have led to discovery of a molecular profile associated with pulmonary arterial hypertension (PAH), which profile may be associated with other condition of the pulmonary cardiovascular system. Particularly, the invention recognizes that treating conditions such as PAH requires inhibiting activity of more than just PDGFR, and in fact requires inhibiting activity of a particular set of kinases.
  • Accordingly, compositions and methods of the invention target specific kinase combinations for inhibition to treat pulmonary and cardiovascular diseases such as PAH. By characterizing the binding affinity of imatinib and other PAH-effective compounds against a variety of kinases, new treatment targets and methods have been identified. Methods of the invention use the targeted inhibition of kinases such as platelet-derived growth factor receptors (PDGFRs) and discoidin domain receptor tyrosine kinase 1 (DDR1), preferably with a single compound, to treat PAH and other pulmonary and cardiovascular diseases.
  • Methods of the invention may include treating PAH or other pulmonary or cardiovascular diseases through the inhibition of one or more of PDGFR-β, PDGFR-α, DDR1, colony stimulating factor 1 receptor (CSF1R), tyrosine-protein kinase KIT (KIT), discoidin domain receptor tyrosine kinase 2 (DDR2), lymphocyte-specific protein tyrosine kinase (LCK), Abelson murine leukemia viral oncogene homolog 1 (ABL1), Abelson murine leukemia viral oncogene homolog 2 (ABL2), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C). In certain embodiments, pulmonary or cardiovascular diseases such as PAH may be treated through inhibition of one or more of vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), PDGFR-α, PDGFR-β, DDR1, KIT, CSF1R, fyn related Src family tyrosine kinase (FRK), DDR2, LCK, LYN proto-oncogene, Src family tyrosine kinase (LYN), FYN proto-oncogene, Src family tyrosine kinase (FYN), and FGR proto-oncogene, Src family tyrosine kinase (FGR).
  • By inhibiting combinations of the above kinases, through the administration of one or more compounds, methods of the invention provide effective treatments for various pulmonary and cardiovascular diseases such as PAH, pulmonary veno-occlusive disease (PVOD), idiopathic pulmonary fibrosis (IPF), and pulmonary capillary hemangiomatosis (PCH) as well as lung transplant rejection and pulmonary hypertension secondary to other diseases like heart failure with preserved ejection fraction (HFpEF) or schistosomiasis.
  • Any route of administration may be applicable and certain formulations and routes of administration are exemplified here. For example, in various embodiments, compounds are administered in inhalable form as dry powder or through a nebulizer. Inhalable formulations can offer greater lung exposure than equivalent doses administered through conventional oral routes or by IV. Accordingly, where a relatively high oral dose would be required to achieve a target lung exposure, the same exposure can be achieved with much lower concentrations of drug delivered by inhalation. By avoiding high systemic loads associated with other conventional administration routes, methods of the invention circumvent some of the adverse effects associated with those high systemic concentrations. For example, as discussed above, systemically administered imatinib, while proving promising in the treatment of PAH, suffered from unacceptable rates of adverse effects including subdural hematoma. In order to minimize the potential for similar risks based on the similar kinase-inhibition profiles, certain methods of the invention provide inhalable compounds.
  • Methods of the invention recognize that the inhibition of various combinations of specific kinases as discussed above are useful in treating certain cardiovascular and pulmonary ailments. Kinase-binding characterization has been performed for compounds A and B, as detailed below, and those compounds have been found to inhibit those various kinase combinations. Accordingly, in certain embodiments, inhalable formulations of compounds A and/or B may be administered to treat cardiovascular and pulmonary diseases according to the invention.
  • In certain embodiments, formulations of the invention may be provided with a higher ratio of API (active pharmaceutical ingredient) than found in conventional formulations. In certain embodiments, formulations comprising 50% or more kinase-inhibiting API are provided. Large volumes may be difficult or dangerous for patients to inhale. Therefore, minimizing the amount of non-API components in the formulation can improve patient comfort, safety, and compliance by reducing the overall amount of compound that is inhaled while still providing a therapeutically effective API concentration in target tissue.
  • Furthermore, aerodynamic properties important to inhalable drug uptake can more easily be managed when less of the formulation is required for carriers or other additives. By providing functional inhalable formulations with high concentrations of kinase inhibitors or salts thereof, compositions and methods of the invention can provide the load-reducing benefits discussed above while still delivering therapeutic results and avoiding the severe adverse events associated with other drug delivery routes.
  • In various embodiments, the kinase inhibitor(s) or salts thereof used in the high-API compositions and methods of the invention can consist of entirely or almost entirely a single crystal form (e.g., greater than 80%, 85%, 90%, 95%, 99% or 100% of a single crystal form), thereby allowing for controlled and predictable dosing and patient response. In certain embodiments, greater than 95% of the kinase inhibiting compound (e.g., compound A or B) or a salt thereof in the inhalable formulation may be present in a single crystal form.
  • In certain embodiments inhalable kinase-inhibiting compounds may be micronized through wet or dry milling (e.g., jet milling) to achieve the desired particle size for dry powder formulations for inhalation. Compounds or appropriate salts thereof may be micronized to particle sizes of about 0.5 μm to about 5 μm mass median aerodynamic diameter (MMAD) for desired deep lung penetration.
  • Inhaled products may be limited in terms of the mass of powder that can be administered and certain salts will contribute significantly to the molecular weight of inhaled formulations. Accordingly, in certain embodiments, the free base of the kinase-inhibiting compound may be preferred over any salts thereof for efficient delivery of the active moiety to lung tissue. If required, various excipients or carriers can be added to the kinase inhibitor(s) or salts thereof before or after micronization depending on application. For example, carriers, excipients, conditioners, and force control agents such as lactose (which when used as a carrier may be conditioned with various solvents to increase separation of imatinib during inhalation), magnesium stearate, leucine, isoleucine, dileucine, trileucine, lecithin, distearylphosphatidylcholine (DSPC) or other lipid-based carriers, or various hydrophilic polymers where they exhibit appropriate physico-chemical properties may be included. The skilled artisan will appreciate that excipients or carriers are optional and that many embodiments of the invention do not require excipients or carriers. In compounds including carriers or excipients, API:carrier ratios may be greater than 50:50, 75:25, or 90:10. Additional ratios are contemplated as discussed below.
  • In certain embodiments, methods of the invention may include administering kinase-inhibiting inhalable formulations that exclude all or most amorphous forms of the compounds. Because crystal form can be important to drug pharmacokinetics and dosing, as well as physicochemical stability and avoiding amorphous content can therefore be important to providing predictable and efficient therapy.
  • Treatment methods of the invention may include the administration of kinase-inhibiting compounds to treat a variety of pulmonary and cardiovascular diseases. Doses may vary depending on the characteristics of the compound used (e.g., its kinase-binding profile) and the disease being treated. In various embodiments, dose ranges can include between about 10 mg to about 100 mg per dose for inhalation on a twice to four times per day schedule. About 0.1 mg to about 80 mg of the active imatinib compound may then be deposited within the lungs after inhalation. The use of relatively high concentrations of API (e.g., 50% or greater) allows for the above doses to be achieved with less overall volume of inhalable compared to conventional formulations having 1%-3% API.
  • Methods of the invention may include administration of spray-dried kinase-inhibiting compounds or salts thereof for inhalation. While carriers such as lactose may be used after micronization to aid in delivery via inhalation, those carriers may generally comprise larger diameter particles and complication in the separation of the active imatinib compound may result in lower amounts of the inhaled compound reaching the lungs. Furthermore, the amount of active compound reaching the lungs may be less predictable using such carriers and methods, making dosing more complicated. Accordingly, spray-dried methods may be used wherein the active kinase-inhibiting compound(s) or salts thereof along with various excipients or other additives may be micronized to a desired particle size and suspended or solubilized for spray-drying and inhalation.
  • In certain embodiments, the micronized kinase-inhibiting compound(s) may be suspended in a feedstock for the purposes of spray-drying to avoid the creation of amorphous or polymorphic imatinib content that may occur if dissolved in a solution (e.g. in an appropriate organic solvent or within an acidified aqueous solution) upon spray-drying. By creating a stable suspension of micronized compound for spray-drying, once dried, the inhalable formulation can retain the desired crystal structure, particle size, and low levels of amorphous content obtained before the micronization process.
  • Stable suspensions for spray-drying may be obtained through manipulation of factors affecting the solubility of the active compound such as pH, ionic strength, and dispersing agents or surfactants. Excipients that may be used before micronization in the spray-drying methods described above include, for example, leucine, dileucine, trileucine, bulking agents such as trehalose or mannitol, lecithin, DSPC or other lipid-based carriers, citrate, or acetate.
  • Aspects of the invention include methods of treating pulmonary arterial hypertension (PAH) that may comprise providing to a subject a therapeutically effective amount of an inhalable formulation of a compound in order to inhibit activity of a plurality of kinases comprising one or more platelet-derived growth factor receptors (PDGFRs) and discoidin domain receptor tyrosine kinase 1 (DDR1). The one or more PDGFRs can include PDGFR-β. In certain embodiments, the one or more PDGFRs may include PDGFR-α.
  • The plurality of kinases can include colony stimulating factor 1 receptor (CSF1R). In some embodiments, the plurality of kinases may include tyrosine-protein kinase KIT (KIT). The plurality of kinases can include discoidin domain receptor tyrosine kinase 2 (DDR2). Methods of the invention may include inhibiting lymphocyte-specific protein tyrosine kinase (LCK).
  • In certain embodiments, the plurality of inhibited kinases can comprise at least Abelson murine leukemia viral oncogene homolog 1 (ABL1), Abelson murine leukemia viral oncogene homolog 2 (ABL2), colony stimulating factor 1 receptor (CSF1R), discoidin domain receptor tyrosine kinase 2 (DDR2), tyrosine-protein kinase KIT (KIT), lymphocyte-specific protein tyrosine kinase (LCK), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C). Each of the plurality of kinases may be inhibited with a Kd of 500 nM or lower.
  • Certain aspects of the invention include methods of treating pulmonary arterial hypertension (PAH) that can include providing to a subject a therapeutically effective amount of an inhalable formulation of a compound in order to inhibit activity of two or more of vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), platelet-derived growth factor receptor α (PDGFR-α), platelet-derived growth factor receptor β (PDGFR-β), discoidin domain receptor tyrosine kinase 1 (DDR1), tyrosine-protein kinase KIT (KIT), colony stimulating factor 1 receptor (CSF1R), fyn related Src family tyrosine kinase (FRK), discoidin domain receptor tyrosine kinase 2 (DDR2), lymphocyte-specific protein tyrosine kinase (LCK), LYN proto-oncogene, Src family tyrosine kinase (LYN), FYN proto-oncogene, Src family tyrosine kinase (FYN), and FGR proto-oncogene, Src family tyrosine kinase (FGR).
  • DETAILED DESCRIPTION
  • The invention relates to the treatment of various pulmonary and cardiovascular diseases through the targeted inhibition of specific combinations of kinases. Diseases including pulmonary arterial hypertension (PAH), pulmonary veno-occlusive disease (PVOD), idiopathic pulmonary fibrosis (IPF), and pulmonary capillary hemangiomatosis (PCH) may be treated as well as lung transplant rejection and pulmonary hypertension secondary to other diseases like heart failure with preserved ejection fraction (HFpEF) or schistosomiasis using inhalable formulations of imatinib and salts thereof.
  • Through comprehensive profiling of the kinase-binding properties of various compounds thought to effectively treat cardiovascular and pulmonary diseases like PAH, specific combinations of kinases have been identified as targets for inhibition. By treating patients with inhalable compounds that inhibit those target kinases, methods of the invention can provide effective treatment of many such diseases.
  • In certain embodiments, methods of the invention can include targeted inhibition of combinations of Abelson murine leukemia viral oncogene homolog 1 (ABL1), colony stimulating factor 1 receptor (CSF1R), discoidin domain receptor tyrosine kinase 1 (DDR1), discoidin domain receptor tyrosine kinase 2 (DDR2), tyrosine-protein kinase KIT (KIT), lymphocyte-specific protein tyrosine kinase (LCK), platelet-derived growth factor receptor-α (PDGFR-α), and platelet-derived growth factor receptor-β (PDGFR-β). The above kinases were found to be inhibited by both compound A and compound B.
  • In certain embodiments, methods of the invention can include targeted inhibition of combinations of PDGFR-β, PDGFR-α, DDR1, CSF1R, KIT, DDR2, LCK, ABL1, Abelson murine leukemia viral oncogene homolog 2 (ABL2), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C). The above kinases were found to be inhibited by compound A.
  • In certain embodiments, methods of the invention can include targeted inhibition of combinations of vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), PDGFR-α, PDGFR-β, DDR1, KIT, CSF1R, fyn related Src family tyrosine kinase (FRK), DDR2, LCK, LYN proto-oncogene, Src family tyrosine kinase (LYN), FYN proto-oncogene, Src family tyrosine kinase (FYN), and FGR proto-oncogene, Src family tyrosine kinase (FGR). The above kinases were found to be inhibited by compound B
  • In certain embodiments, methods of the invention may include targeted inhibition of combinations of PDGFR-β, PDGFR-α, DDR1, CSF1R, KIT, DDR2, LCK, ABL1, ABL2, and PI42C while not significantly inhibiting (e.g. less than 500 nM Kd) one or more of VEGFR-2, HCK, FLT4, RET, SRC, FRK, LYN, FYN, and FGR.
  • In certain embodiments, methods of the invention may include targeted inhibition of combinations of VEGFR-2, HCK, FLT4, RET, SRC, PDGFR-α, PDGFR-β, DDR1, KIT, CSF1R, FRK, DDR2, LCK, LYN, FYN, and FGR while not significantly inhibiting (e.g. less than 500 nM Kd) one or more of ABL2, and PI42C.
  • In various embodiments, methods of the invention may include targeted inhibition of any combination of the above kinases via administration of a compound having an equilibrium dissociation constant (Kd) of less than about 1000 nM, 500 nM, less than about 400 nM, less than about 300 nM, less than about 200 nM, less than about 100 nM, less than about 75 nM, less than about 50 nM, less than about 40 nM, less than about 30 nM, less than about 20 nM, less than about 15 nM, or less than about 5 nM with respect to each of the targeted kinases.
  • Compound A, as characterized below, is free base imatinib having the following structure:
  • Figure US20200360477A1-20201119-C00001
  • The kinase-binding profile including the equilibrium dissociation constant (Kd) was determined for compound A as detailed in Example 1 below. The results of that characterization are shown in Table 1.
  • TABLE 1
    Compound Activity
    Kinase Remaining at 1 μM (%) Kd (nM)
    KIT 0.5 13
    PDGFR-β 0.15 14
    DDR1 0.2 0.7
    VEGFR2 70 >10000
    PDGFR-α 0.95 31
    CSF1R 1.6 11
    LCK 1.2 40
    DDR2 0.8 15
    ABL-1-nonphosphorolated 0.05 1.1
    HCK 81 >10000
    PIK4CB 59 >10000
    RET 98 >10000
    FRK 56 1500
    SRC 94 >10000
    LYN 30 890
    FYN 79 3100
    FLT4 87 >10000
    FGR 70 2400
  • Compound B, as characterized below, has the following structure:
  • Figure US20200360477A1-20201119-C00002
  • Compound B and some of its potential applications in treating pulmonary and cardiovascular disorders are discussed in U.S. Pat. Nos. 9,815,815; 9,925,184; 10,231,966; and 10,246,438; the content of each of which is incorporated herein by reference.
  • The kinase-binding profile including the equilibrium dissociation constant (Kd) was determined for compound B as detailed in Example 3 below. The results of that characterization are shown in Table 2.
  • TABLE 2
    Compound Activity
    Kinase Remaining at 1 μM (%) Kd (nM)
    KIT 0 4.7
    PDGFR-β 0 0.82
    DDR1 0.2 3.5
    VEGFR2 0.25 15
    PDGFR-α 0.35 4
    CSF1R 0.7 6.1
    LCK 1.1 22
    DDR2 1.8 19
    ABL-1-nonphosphorolated 3.3
    HCK 3.3 36
    PIK4CB 3.8 150
    RET 3.9 19
    FRK 5.3 11
    SRC 6.4 130
    LYN 7.3 24
    FYN 9.7 55
    FLT4 12 18
    FGR 21 92
  • As shown in the tables above, both compounds significantly bind and therefore inhibit a number of the same kinases while each further inhibits additional kinases not significantly affected by the other compound. By analyzing those combined profiles, in certain embodiments, methods of the invention are able to identify target kinases for inhibition that are common to both compounds and that, therefore may be more likely to provide a therapeutic effect in treating cardiovascular or pulmonary diseases.
  • Furthermore, the differences in kinase-inhibition between the two compounds may account for differences in compound efficacy in treating certain pulmonary and cardiovascular diseases and account for various adverse effects. Accordingly, in certain embodiments, methods of the invention may target only kinases inhibited by one or the other of the compounds to improve treatment outcomes and reduce the risk of adverse effects.
  • In certain embodiments methods and compositions described herein may provide greater concentrations of a kinase-inhibiting compound in target lung tissue than obtained with equivalent doses administered orally or through IV. Furthermore, those doses, comprising a high percentage of the overall formulation, can be delivered in lower volume formulations than conventional formulations of between 1% and 3% API. Reducing the volume a patient must inhale can increase patient comfort and compliance, thereby improving results. Additionally, a higher percentage of API content can improve the API distribution and blend uniformity. Accordingly, methods and compositions of the invention allow for treatment of conditions of the pulmonary cardiovascular system (e.g., PAH) with lower doses and less inhalable volume than would be required in systemic administration, thereby lowering the risk of adverse events including subdural hematoma (See, Frost et al.). Thus, the invention provides viable treatment methods for life threatening diseases that were heretofore too risky for practical application.
  • In certain embodiments, compounds of the invention may include formulations of a kinase inhibitor such as imatinib or salts thereof targeting combinations of kinases as discussed above. In certain embodiments, the free base of the kinase-inhibiting compound may be used in a formulation (either in dry powder or suspension) for inhalation to treat a condition of the pulmonary cardiovascular system such as PAH. Certain salt forms are also contemplated. In various embodiments, kinase inhibitor salts that were found to exhibit suitable thermal stability and few or single polymorphic forms include glycollate, isethionate, malonate, tartrate, and malate. Other salt forms contemplated herein are xinafoate, furoate, trifenatate, HCl, sulfate, phosphate, lactate, maleate, fumarate, succinate, adipate, mesylate, and citrate.
  • When the compounds of the present invention are administered as pharmaceuticals, to humans and mammals, they can be given alone or as a pharmaceutical composition containing, for example, 0.1 to 99.5% of active ingredient (e.g., imatinib or a salt thereof) in combination with a pharmaceutically acceptable carrier. In preferred embodiments, to reduce inhaled volumes for patients and improve patient outcomes, formulations can comprise at least 50% of a kinase-inhibiting compound or a salt thereof.
  • In certain embodiments, formulations of the invention may include one or more excipients. Excipients may include, for example, lactose in various forms (e.g., roller dried or spray dried). Larger lactose particles can be used as a carrier for inhalation of micronized formulations. The carrier particles, with their larger size, can be used to increase aerodynamic forces on the combined kinase inhibitor/carrier in order to aid in delivery through inhalation. Solvents may be used to condition the lactose surface such that the active component can be effectively separated from the lactose as it leaves the inhaler device and within the oral cavity when being used as a carrier. Magnesium stearate can be used as a force-control agent or conditioning agent in various embodiments. In some embodiments, leucine can be used as a force-control agent including different forms of leucine (e.g. isoleucine) along with dileucine and even trileucine.
  • Lecithin phospholipids such as DSPC may be used as an excipient for dry powder inhalation. In certain embodiments, excipients may include various hydrophilic polymers. See, for example, Karolewicz, B., 2016, A review of polymers as multifunctional excipients in drug dosage form technology, Saudi Pharm J., 24(5):525-536, incorporated herein by reference.
  • In the high-API-ratio formulations contemplated herein, carriers or excipients may make up the remainder of the formulation in amounts of 50% or less of the overall composition. In certain embodiments, inhalable formulations may have API:carrier ratios of 50:50, 55:45, 60:40, 65:35, 70:30, 75:25, 80:20, 85:15, 90:10, or 95:5. Certain inhalable formulations may be pure API with no additional components. In various embodiments, formulations may include a kinase inhibitor or salts thereof as the API in amounts greater than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, or 45%. As used herein, API ratios refer to % w/w.
  • In various embodiments, micronized kinase inhibitor and salts thereof retain crystallinity, even after micronization and spray drying (as discussed in detail below). For example, kinase inhibiting formulations of the invention can include less than 50%, less than 25%, less than 20%, less than 10%, less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, or less than 1% amorphous API content by mass. In preferred embodiments, formulations of the invention include no observable amorphous content of the kinase-inhibiting compound. Of particular note is, by suspending micronized kinase inhibitor particles in a solution as opposed to solubilizing, the desired crystalline form and low amorphous content obtained during micronization is carried through to the spray-dried inhalable powder because the kinase inhibitor crystals may not be dissolved in the solution to a significant degree.
  • Another unexpected result obtained with methods and formulations of the invention is that kinase inhibitor formulations of the invention may be significantly less hygroscopic than conventional salt compounds such as imatinib mesylate. Accordingly, the formulations of the invention are better suited for dry powder inhalation and can comprise less than 5% water content, less than 4%, less than 3%, less than 2%, or, in preferred embodiments, less than 1% water content.
  • As discussed above, in order to accurately and consistently model pharmacokinetics of formulations for proper dosing, low polymorphism is desired. To that end, inhalable formulations of the invention include kinase inhibitors or salts thereof present in a single crystal form. In various embodiments, the kinase inhibitor or a salt thereof may be present at greater than 75%, 80%, 85%, 90%, 95%, or, in preferred embodiments, greater than 99% in a single crystal form by mass. The single crystal form may be, for example, imatinib in type A or type B in various embodiments. Crystalline purity can be estimated using any known method including, for example, x-ray powder diffraction (XRPD).
  • In various embodiments, kinase inhibitors or salts thereof are provided in dry powder formulations for inhalation. Dry powder can be administered via, for example, dry powder inhalers such as described in Berkenfeld, et al., 2015, Devices for Dry Powder Drug Delivery to the Lung, AAPS PharmaSciTech, 16(3):479-490, incorporated herein by reference. Dry powder compounds may be divided into single doses for single, twice daily, three times daily, or four times daily inhalation to treat disorders such as PAH or other conditions of the pulmonary cardiovascular system. The single doses may be divided into individual capsules or other formats compatible with the dry powder inhaler to be used.
  • In other embodiments, suspensions having the characteristics described herein (e.g., low polymorphism and amorphous content) can be delivered via inhalation using, for example, a nebulizer. Suspensions may offer advantages over solutions as discussed below. For nebulized suspensions, micronization and particle diameter may be of particular importance for efficient delivery and the active kinase-inhibiting compound may be preferably micronized to a mass median diameter of 2 μm or less. The suspension solution for nebulizer inhalation can be aqueous and doses may be divided into individual containers or compartments for sterile storage prior to use.
  • Micronized kinase inhibitor particle size can range from about 0.5 μm to about 5 μm depending on application (e.g., dry powder or suspension for inhalation). In preferred embodiments the size range is about 1 μm to about 3 μm in dry powder formulations to achieve deep lung penetration.
  • Dosages for treating PAH and other conditions of the pulmonary cardiovascular system may be in the range of between about 10 mg to about 100 mg per dose for inhalation on once, twice or three times per day schedule. About 0.1 mg to about 80 mg of the kinase inhibitor(s) or salts thereof may then be deposited within the lung after inhalation. In certain embodiments about 10 mg to 30 mg of kinase-inhibiting compound may be given in a capsule for a single dry-powder inhalation dose with about 5 mg to about 10 mg of the compound to be expected to reach the lungs. In inhalable suspension embodiments, kinase inhibitor may be present at about 0.1 to about 1 mg/kg in a dose and may be administered one to four times a day to obtain the desired therapeutic results.
  • In certain embodiments, the kinase-inhibiting methods of the invention may be used to treat pulmonary hypertension as a result of schistosomiasis. See, for example, Li, et al., 2019, The ABL kinase inhibitor imatinib causes phenotypic changes and lethality in adult Schistosoma japonicum, Parasitol Res., 118(3):881-890; Graham, et al., 2010, Schistosomiasis-associated pulmonary hypertension: pulmonary vascular disease: the global perspective, Chest, 137(6 Suppl):20S-29S, the content of each of which is incorporated herein by reference.
  • Methods and compositions of the invention may be used to treat lung transplant recipients to prevent organ rejection. See, Keil, et al., 2019, Synergism of imatinib, vatalanib and everolimus in the prevention of chronic lung allograft rejection after lung transplantation (LTx) in rats, Histol Histopathol, 1:18088, incorporated herein by reference.
  • In certain embodiments, pharmaceutical compositions described herein can be used to treat pulmonary veno-occlusive disease (PVOD). See Sato, et al., 2019, Beneficial Effects of Imatinib in a Patient with Suspected Pulmonary Veno-Occlusive Disease, Tohoku J Exp Med. 2019 February; 247(2):69-73, incorporated herein by reference.
  • For treatment of any conditions of the pulmonary cardiovascular system for which kinase-inhibiting methods of the invention may produce a therapeutic effect, compounds and methods of the invention may be used to provide greater concentration at the target lung tissue through inhalation along with consistent, predictable pharmacokinetics afforded by low polymorphism and amorphous content. The efficient localization of therapeutic compound at the target tissue allows for lower systemic exposure and avoidance of the adverse events associated with prolonged oral administration of certain kinase inhibitors such as imatinib mesylate.
  • Methods of the invention can include preparation of kinase-inhibiting formulations. As noted above, kinase inhibitors or salts thereof may be administered via inhalation in suspension or dry powder form. Dry powder formulations may be obtained via any known method including, in preferred embodiments, jet milling. Jet milling can be used to grind active compounds and, potentially, various additives (e.g., excipients) using a jet (or jets) of compressed air or gas to force collisions between the particles as they transit at near sonic velocity around the perimeter of a toroidal chamber. The size reduction is the result of the high-velocity collisions between particles of the process material. Outputs of the jet mill may allow particles to exit the apparatus once a desired size has been reached. As noted herein, desired particle size for dry powder inhalation and other formulations may be in the range of about 0.5 μm to about 5 μm.
  • In certain embodiments, bulk compounds may be micronized to the desired size for inhalation via wet milling wherein the kinase inhibitor particles are suspended in a slurry and reduced through shearing or impact with a grinding media.
  • An unexpected finding of the invention is that, once micronized, free base kinase-inhibiting compounds retain crystallinity and are considerably less hygroscopic than certain salt forms (e.g., imatinib mesylate). Furthermore, micronized imatinib obtained using methods of the invention has been found to exhibit no apparent polymorphs other than the designated Type A and very low levels of amorphous content. Accordingly, this can result in improved stability of the drug substance and any drug product upon storage. Single crystal forms of imatinib or other kinase inhibitors such as described may allow for more predictable in vivo behavior and appropriate dosing can be determined.
  • Once micronized, in dry powder form, kinase inhibitor formulations of the invention, with their low polymorphic and amorphous content, can be prepared for inhalation. In certain embodiments, the dry powder kinase inhibitor can be combined with larger carrier particles such as lactose as discussed above.
  • In some embodiments a suspension can be formed of the kinase inhibitor compound(s). The suspension may result from dry micronization followed by suspension of the resulting dry powder or can be obtained as the outcome of a wet milling procedure. Suspensions of micronized crystal forms may be used in nebulized inhalation treatment or may be spray dried for dry powder treatments.
  • Spray drying methods may follow the following procedure. First, bulk kinase inhibitor compound may be micronized as described above to obtain particles in a desired size range. Then the micronized compound can be suspended in a solution such that it does not dissolve and instead retains the desired crystalline features (e.g., low polymorphism and amorphous content). The suspended particles can then be spray dried using any known method. Spray drying techniques are well characterized and described, for example, in Ziaee, et al., 2019, Spray drying of pharmaceuticals and biopharmaceuticals: Critical parameters and experimental process optimization approaches, Eur. J. Pharm. Sci., 127:300-318, and Weers et al., 2019, AAPS PharmSciTech. 2019 Feb. 7; 20(3):103. doi: 10.1208/s12249-018-1280-0, and 2018/0303753, each of which is incorporated herein by reference. Spray drying micronized kinase inhibitor compounds or salts thereof provides for uniform and predictable crystallinity and particle size and can avoid the need for large carrier molecules that may adversely affect the amount of inhaled drug that reaches the target lung tissue.
  • In spray-dried embodiments, micronized drug particles may be suspended within a non-aqueous solvent or within an emulsion of a non-aqueous solvent which, in turn is emulsified or dispersed within an aqueous environment (e.g. oil in water) and spray-dried, resulting in crystalline drug particles. The non-aqueous component may or may not be fugitive and thus could be removed completely during spray drying or, it could be retained, depending on the desired properties required. In such embodiments, each atomized droplet (mass median diameter ˜10 μm) contains dispersed drug crystals. During the initial moments of the drying process, the more volatile aqueous phase begins to evaporate. The rapidly receding atomized droplet interface drives enrichment of the slowly diffusing drug and emulsion particles at the interface. This leads to formation of a void space in the center of the drying droplet. As the drying process continues, the less volatile oil phase in the emulsion droplets evaporates, resulting in formation of hollow pores in their place. Overall, the resulting hollow spray-dried composite particles contain drug crystals.
  • As maintaining a stable solution of crystalline kinase inhibitor compound is important to many features of the formulations and methods of the invention, formulation methods include manipulation of the suspension to prevent dissolution of the kinase inhibitor compound. Aqueous solution factors such as pH, ionic strength and dispersing agents may be used to obtain a stable suspension for nebulized inhalation or spray drying. For example, the pH of the aqueous solution may be adjusted to prevent dissolution.
  • Additionally, the presence of ions in aqueous solution may tend to ‘salt out’ the active compound. The solubility of the kinase inhibitors and any salts thereof may decrease with salinity. Accordingly, salt in the aqueous solution may be used to reduce solubility of the active compound crystals in certain embodiments.
  • To promote dispersion and thoroughly deagglomerate the API particles, a dispersing agent or surfactant (e.g., Tween 20 or Tween 80) may be added but should not cause dissolution of the kinase inhibitor in suspension.
  • In certain embodiments, excipients can be added to the suspension before spray drying. In various embodiments, the excipient may be a water-soluble excipient, such as leucine, dileucine, trileucine, trehalose, mannitol, citrate or acetate. In other embodiment, the excipient may be a water insoluble excipient, such as lecithin, distearylphosphatidylcholine (DSPC) or limonene. Such insoluble excipients may be dissolved in a non-aqueous medium that is miscible or immiscible with water, thereby creating an emulsion. Alternatively, a liposomal dispersion could be created into which the suspended kinase inhibitor could be added and homogenized or where it could be spray dried in separate feedstocks.
  • The effective dosage of each agent can readily be determined by the skilled person, having regard to typical factors such as the age, weight, sex and clinical history of the patient. In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce the desired therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • If desired, the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • The pharmaceutical compositions of the invention include a “therapeutically effective amount” or a “prophylactically effective amount” of one or more of the compounds of the present invention, or functional derivatives thereof. An “effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, e.g., a diminishment or prevention of effects associated with PAH. A therapeutically effective amount of a compound of the present invention or functional derivatives thereof may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the therapeutic compound to elicit a desired response in the subject. A therapeutically effective amount is also one in which any toxic or detrimental effects of the therapeutic agent are outweighed by the therapeutically beneficial effects.
  • A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to, or at an earlier stage of disease, the prophylactically effective amount may be less than the therapeutically effective amount. A prophylactically or therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the beneficial effects.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g. a therapeutic or prophylactic response). For example, a single inhalable bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigency of the therapeutic situation. Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, without being toxic to the patient.
  • The term “dosage unit” as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the compound, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • In some embodiments, therapeutically effective amount can be estimated initially either in cell culture assays or in animal models, usually rats, non-human primates, mice, rabbits, dogs, or pigs. The animal model is also used to achieve a desirable concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in other subjects. Generally, the therapeutically effective amount is sufficient to reduce PAH symptoms in a subject. In some embodiments, the therapeutically effective amount is sufficient to eliminate PAH symptoms in a subject.
  • Dosages for a particular patient can be determined by one of ordinary skill in the art using conventional considerations, (e.g. by means of an appropriate, conventional pharmacological protocol). A physician may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained. The dose administered to a patient is sufficient to effect a beneficial therapeutic response in the patient over time, or, e.g., to reduce symptoms, or other appropriate activity, depending on the application. The dose is determined by the efficacy of the particular formulation, and the activity, stability, or half-life of the compounds of the invention or functional derivatives thereof, and the condition of the patient, as well as the body weight or surface area of the patient to be treated. The size of the dose is also determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular vector, formulation, or the like in a particular subject. Therapeutic compositions comprising one or more compounds of the invention or functional derivatives thereof are optionally tested in one or more appropriate in vitro and/or in vivo animal models of disease, such as models of PAH, to confirm efficacy, tissue metabolism, and to estimate dosages, according to methods well known in the art. In particular, dosages can be initially determined by activity, stability or other suitable measures of treatment vs. non-treatment (e.g., comparison of treated vs. untreated cells or animal models), in a relevant assay. Administration can be accomplished via single or divided doses.
  • In certain embodiments, in which an aqueous suspension is part of the manufacturing process, the aqueous suspension may contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents dispersing or wetting agents such as a naturally occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such a polyoxyethylene with partial esters derived from fatty acids and hexitol anhydrides, for example polyoxyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, mannitol, or trehalose.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • The term “pharmaceutical composition” means a composition comprising a compound as described herein and at least one component comprising pharmaceutically acceptable carriers, diluents, adjuvants, excipients, or vehicles, such as preserving agents, taste-masking agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • The term “pharmaceutically acceptable carrier” is used to mean any carrier, diluent, adjuvant, excipient, or vehicle, as described herein. Examples of suspending agents include ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Examples of suitable carriers, diluents, solvents, or vehicles include water, ethanol, polyols, suitable mixtures thereof, vegetable oils (such as olive oil), and organic esters such as ethyl oleate. Examples of excipients include lactose, milk sugar, sodium citrate, calcium carbonate, and dicalcium phosphate. Examples of disintegrating agents include starch, alginic acids, and certain complex silicates. Examples of lubricants include magnesium stearate, sodium lauryl sulphate, talc, as well as high molecular weight polyethylene glycols.
  • The term “pharmaceutically acceptable” means it is, within the scope of sound medical judgment, suitable for use in contact with the cells of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
  • INCORPORATION BY REFERENCE
  • References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made throughout this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes.
  • Equivalents
  • Various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including references to the scientific and patent literature cited herein. The subject matter herein contains important information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and equivalents thereof.
  • EXAMPLES Example 1
  • An inhalable form of imatinib (compound A) was screened against 468 quantifiable kinases in the human kinome (the set of protein kinases encoded in the human genome) to characterize binding affinity for each kinase.
  • Kinase binding was profiled using KINOMEscan® (Eurofins DiscoverX Corporation). KINOMEscan™ is based on a competition binding assay that quantitatively measures the ability of a compound to compete with an immobilized, active-site directed ligand. The assay is performed by combining three components: DNA-tagged kinase; immobilized ligand; and a test compound. The ability of the test compound to compete with the immobilized ligand is measured via quantitative PCR of the DNA tag.
  • Kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32° C. until lysis. The lysates were centrifuged and filtered to remove cell debris. Some kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection.
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays. The ligand-bound beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding. Binding reactions were assembled by combining kinases, ligand-bound affinity beads, and test compounds in 1× binding buffer (20% SeaBlock, 0.17×PBS, 0.05% Tween 20, 6 mM DTT). Test compounds were prepared as 111× stocks in 100% DMSO. Kds were determined using an 11-point 3-fold compound dilution series with three DMSO control points. All compounds for Kd measurements were distributed by acoustic transfer (non-contact dispensing) in 100% DMSO.
  • The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.9%. All reactions were performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1×PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (1×PBS, 0.05% Tween 20, 0.5 μM non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.
  • Example 2
  • Compound B, as detailed above, was screened against 468 quantifiable kinases in the human kinome (the set of protein kinases encoded in the human genome) to characterize binding affinity for each kinase.
  • Kinase binding was profiled using KINOMEscan® (Eurofins DiscoverX Corporation). KINOMEscan™ is based on a competition binding assay that quantitatively measures the ability of a compound to compete with an immobilized, active-site directed ligand. The assay is performed by combining three components: DNA-tagged kinase; immobilized ligand; and a test compound. The ability of the test compound to compete with the immobilized ligand is measured via quantitative PCR of the DNA tag.
  • Kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32° C. until lysis. The lysates were centrifuged and filtered to remove cell debris. Some kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection.
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays. The ligand-bound beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding. Binding reactions were assembled by combining kinases, ligand-bound affinity beads, and test compounds in 1× binding buffer (20% SeaBlock, 0.17×PBS, 0.05% Tween 20, 6 mM DTT). Test compounds were prepared as 111× stocks in 100% DMSO. Kds were determined using an 11-point 3-fold compound dilution series with three DMSO control points. All compounds for Kd measurements were distributed by acoustic transfer (non-contact dispensing) in 100% DMSO.
  • The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.9%. All reactions were performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1×PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (1×PBS, 0.05% Tween 20, 0.5 μM non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.

Claims (10)

What is claimed is:
1. A method of treating pulmonary arterial hypertension (PAH), the method comprising providing to a subject a therapeutically effective amount of an inhalable formulation of a compound in order to inhibit activity of a plurality of kinases comprising one or more platelet-derived growth factor receptors (PDGFRs) and discoidin domain receptor tyrosine kinase 1 (DDR1).
2. The method of claim 1 wherein the one or more PDGFRs comprises PDGFR-β.
3. The method of claim 2 wherein the one or more PDGFRs comprises PDGFR-α.
4. The method of claim 3 wherein the plurality of kinases comprises colony stimulating factor 1 receptor (CSF1R).
5. The method of claim 4 wherein the plurality of kinases comprises tyrosine-protein kinase KIT (KIT).
6. The method of claim 5 wherein the plurality of kinases comprises discoidin domain receptor tyrosine kinase 2 (DDR2).
7. The method of claim 6 wherein the plurality of kinases comprises lymphocyte-specific protein tyrosine kinase (LCK).
8. The method of claim 1 wherein the plurality of kinases comprise Abelson murine leukemia viral oncogene homolog 1 (ABL1), Abelson murine leukemia viral oncogene homolog 2 (ABL2), colony stimulating factor 1 receptor (CSF1R), discoidin domain receptor tyrosine kinase 2 (DDR2), tyrosine-protein kinase KIT (KIT), lymphocyte-specific protein tyrosine kinase (LCK), and phosphatidylinositol 5-phosphate 4-kinase type-2 gamma (PI42C).
9. The method of claim 8 wherein each of the plurality of kinases are inhibited with a Kd of 500 nM or lower.
10. A method of treating pulmonary arterial hypertension (PAH), the method comprising providing to a subject a therapeutically effective amount of an inhalable formulation of a compound in order to inhibit activity of a plurality of kinases comprising vascular endothelial growth factor receptor 2 (VEGFR-2), HCK proto-oncogene, Src family tyrosine kinase (HCK), fms related receptor tyrosine kinase 4 (FLT4), ret proto-oncogene (RET), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), platelet-derived growth factor receptor α (PDGFR-α), platelet-derived growth factor receptor β (PDGFR-β), discoidin domain receptor tyrosine kinase 1 (DDR1), tyrosine-protein kinase KIT (KIT), colony stimulating factor 1 receptor (CSF1R), fyn related Src family tyrosine kinase (FRK), discoidin domain receptor tyrosine kinase 2 (DDR2), lymphocyte-specific protein tyrosine kinase (LCK), LYN proto-oncogene, Src family tyrosine kinase (LYN), FYN proto-oncogene, Src family tyrosine kinase (FYN), and FGR proto-oncogene, Src family tyrosine kinase (FGR).
US16/874,190 2019-05-16 2020-05-14 Inhalable formulations for kinase inhibition Pending US20200360477A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/874,190 US20200360477A1 (en) 2019-05-16 2020-05-14 Inhalable formulations for kinase inhibition

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US201962849058P 2019-05-16 2019-05-16
US201962849056P 2019-05-16 2019-05-16
US201962849059P 2019-05-16 2019-05-16
US201962849054P 2019-05-16 2019-05-16
US201962877575P 2019-07-23 2019-07-23
US201962942408P 2019-12-02 2019-12-02
US202062958481P 2020-01-08 2020-01-08
US202062984037P 2020-03-02 2020-03-02
US16/874,190 US20200360477A1 (en) 2019-05-16 2020-05-14 Inhalable formulations for kinase inhibition

Publications (1)

Publication Number Publication Date
US20200360477A1 true US20200360477A1 (en) 2020-11-19

Family

ID=73228465

Family Applications (13)

Application Number Title Priority Date Filing Date
US16/874,111 Active US11229650B2 (en) 2019-05-16 2020-05-14 Inhalable imatinib formulations, manufacture, and uses thereof
US16/874,168 Pending US20200360279A1 (en) 2019-05-16 2020-05-14 Nebulized imatinib formulations, manufacture, and uses thereof
US16/874,190 Pending US20200360477A1 (en) 2019-05-16 2020-05-14 Inhalable formulations for kinase inhibition
US16/874,143 Pending US20200360277A1 (en) 2019-05-16 2020-05-14 Inhalable formulations of imatinib, imatinib metabolites, imatinib salts, their manufacture, and uses thereof
US16/874,128 Pending US20200360276A1 (en) 2019-05-16 2020-05-14 Inhalable imatinib metabolite formulations, manufacture, and uses thereof
US16/874,118 Active US11298355B2 (en) 2019-05-16 2020-05-14 Inhalable imatinib formulations, manufacture, and uses thereof
US16/874,122 Active US11413289B2 (en) 2019-05-16 2020-05-14 Inhalable imatinib formulations, manufacture, and uses thereof
US17/582,599 Active 2040-05-21 US11806349B2 (en) 2019-05-16 2022-01-24 Inhalable imatinib formulations, manufacture, and uses thereof
US17/685,704 Pending US20220184080A1 (en) 2019-05-16 2022-03-03 Nebulized imatinib formulations, manufacture, and uses thereof
US17/887,866 Active US11813263B2 (en) 2019-05-16 2022-08-15 Inhalable imatinib formulations, manufacture, and uses thereof
US17/963,607 Pending US20230056721A1 (en) 2019-05-16 2022-10-11 Inhalable imatinib formulations, manufacture, and uses thereof
US18/377,561 Pending US20240108622A1 (en) 2019-05-16 2023-10-06 Inhalable imatinib formulations, manufacture, and uses thereof
US18/378,949 Pending US20240041879A1 (en) 2019-05-16 2023-10-11 Inhalable imatinib formulations, manufacture, and uses thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US16/874,111 Active US11229650B2 (en) 2019-05-16 2020-05-14 Inhalable imatinib formulations, manufacture, and uses thereof
US16/874,168 Pending US20200360279A1 (en) 2019-05-16 2020-05-14 Nebulized imatinib formulations, manufacture, and uses thereof

Family Applications After (10)

Application Number Title Priority Date Filing Date
US16/874,143 Pending US20200360277A1 (en) 2019-05-16 2020-05-14 Inhalable formulations of imatinib, imatinib metabolites, imatinib salts, their manufacture, and uses thereof
US16/874,128 Pending US20200360276A1 (en) 2019-05-16 2020-05-14 Inhalable imatinib metabolite formulations, manufacture, and uses thereof
US16/874,118 Active US11298355B2 (en) 2019-05-16 2020-05-14 Inhalable imatinib formulations, manufacture, and uses thereof
US16/874,122 Active US11413289B2 (en) 2019-05-16 2020-05-14 Inhalable imatinib formulations, manufacture, and uses thereof
US17/582,599 Active 2040-05-21 US11806349B2 (en) 2019-05-16 2022-01-24 Inhalable imatinib formulations, manufacture, and uses thereof
US17/685,704 Pending US20220184080A1 (en) 2019-05-16 2022-03-03 Nebulized imatinib formulations, manufacture, and uses thereof
US17/887,866 Active US11813263B2 (en) 2019-05-16 2022-08-15 Inhalable imatinib formulations, manufacture, and uses thereof
US17/963,607 Pending US20230056721A1 (en) 2019-05-16 2022-10-11 Inhalable imatinib formulations, manufacture, and uses thereof
US18/377,561 Pending US20240108622A1 (en) 2019-05-16 2023-10-06 Inhalable imatinib formulations, manufacture, and uses thereof
US18/378,949 Pending US20240041879A1 (en) 2019-05-16 2023-10-11 Inhalable imatinib formulations, manufacture, and uses thereof

Country Status (14)

Country Link
US (13) US11229650B2 (en)
EP (2) EP3969111A4 (en)
JP (1) JP2022532431A (en)
KR (1) KR20220050839A (en)
CN (1) CN114514016A (en)
AU (1) AU2020274521A1 (en)
BR (1) BR112021023014A2 (en)
CA (1) CA3140641A1 (en)
IL (1) IL288111A (en)
JO (1) JOP20210305A1 (en)
MX (1) MX2021014029A (en)
SG (1) SG11202112719XA (en)
WO (2) WO2020232236A1 (en)
ZA (1) ZA202109070B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11806349B2 (en) 2019-05-16 2023-11-07 Aerovate Therapeutics, Inc. Inhalable imatinib formulations, manufacture, and uses thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20230131179A (en) * 2020-11-17 2023-09-12 유나이티드 쎄러퓨틱스 코포레이션 Inhaled Imatinib for Pulmonary Hypertension
EP4247378A1 (en) * 2020-11-23 2023-09-27 Aerovate Therapeutics, Inc. Imatinib formulations, manufacture, and uses thereof
EP4274577A1 (en) * 2021-01-06 2023-11-15 InCarda Therapeutics, Inc. Inhalable imatinib formulation
KR102634754B1 (en) * 2021-07-07 2024-02-08 애니머스큐어 주식회사 Composition for prevention or treatment of muscle diseases containing Imatinib
WO2023147868A1 (en) * 2022-02-04 2023-08-10 Justus-Liebig-Universität Giessen Inhaled imatinib for treatment of pulmonary hypertension

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015188243A1 (en) * 2014-06-10 2015-12-17 Cristália Produtos Químicos Farmacêuticos Ltda PROCESS FOR PREPARING IMATINIB AND IMATINIB MESYLATE NON-NEEDLE SHAPED α2 FORM

Family Cites Families (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO1996025918A1 (en) * 1995-02-24 1996-08-29 Nanosystems L.L.C. Aerosols containing nanoparticle dispersions
GB0009468D0 (en) 2000-04-17 2000-06-07 Vectura Ltd Improvements in or relating to formulations for use in inhaler devices
AU2003248813A1 (en) * 2002-07-05 2004-01-23 Beth Israel Deaconess Medical Center Combination of mtor inhibitor and a tyrosine kinase inhibitor for the treatment of neoplasms
GB2398565A (en) 2003-02-18 2004-08-25 Cipla Ltd Imatinib preparation and salts
US20040204439A1 (en) 2003-04-14 2004-10-14 Staniforth John Nicholas Composition, device, and method for treating sexual dysfunction via inhalation
GB0327723D0 (en) 2003-09-15 2003-12-31 Vectura Ltd Pharmaceutical compositions
EP2233174B1 (en) 2004-01-21 2016-07-20 Emory University Compositions and use of tyrosine kinase inhibitors to treat pathogenic infection
US7507821B2 (en) 2004-12-30 2009-03-24 Chemagis Ltd. Process for preparing Imatinib
SI1833815T1 (en) 2004-12-30 2011-01-31 Inst Farmaceutyczny A process for preparation of imatinib base
EP1695728A1 (en) 2005-02-23 2006-08-30 Activaero GmbH Component for an inhalation device, an inhalation device with this component and a method of controlling for such a component
EP1700614B1 (en) 2005-03-08 2013-05-08 Activaero GmbH Inhalation device
BRPI0613540A2 (en) 2005-06-03 2011-01-18 Elan Pharma Int Ltd nanoparticulate imatinib mesylate formulations
ATE373498T1 (en) 2005-07-06 2007-10-15 Activaero Gmbh ADJUSTABLE VALVE AND INHALATION DEVICE
EP2111863B1 (en) 2005-10-26 2012-03-28 Asahi Kasei Pharma Corporation Fasudil in combination with bosentan for the treament of pulmonary arterial hypertension
BRPI0602338A (en) 2006-03-24 2007-12-11 Univ Kyushu Nat Univ Corp organic compounds
US20060223817A1 (en) 2006-05-15 2006-10-05 Chemagis Ltd. Crystalline imatinib base and production process therefor
US20080181958A1 (en) 2006-06-19 2008-07-31 Rothrock Ginger D Nanoparticle fabrication methods, systems, and materials
US20080103305A1 (en) 2006-10-26 2008-05-01 Macdonald Peter Process for the preparation of imatinib
EP1988089A1 (en) * 2006-10-26 2008-11-05 Sicor, Inc. Imatinib base, and imatinib mesylate and processes for preparation thereof
US20130177598A1 (en) * 2007-02-27 2013-07-11 The University Of North Carolina At Chapel Hill Discrete size and shape specific pharmaceutical organic nanoparticles
WO2008136010A1 (en) 2007-05-07 2008-11-13 Natco Pharma Limited A process for the preparation of highly pure imatinib base
RU2519647C2 (en) 2007-07-13 2014-06-20 Нестек С.А. Selection of medications for lung cancer therapy by means of antibody-based matrices
EP2022525B1 (en) 2007-08-02 2012-12-26 Activaero GmbH Device and system for targeting aerosolized particles to a specific area of the lungs
EP2033674A1 (en) 2007-09-06 2009-03-11 Activaero GmbH Inhalation device
CA2732789A1 (en) 2008-08-13 2010-02-18 Novartis Ag Treatment of pulmonary arterial hypertension
US8834848B2 (en) 2009-02-04 2014-09-16 Activaero Gmbh Research & Development Flow and volume regulated inhalation for treatment of severe oral corticosteroid-dependent asthma
US20100330130A1 (en) 2009-05-22 2010-12-30 Actavis Group Ptc Ehf Substantially pure imatinib or a pharmaceutically acceptable salt thereof
CZ2009570A3 (en) 2009-08-26 2011-03-09 Zentiva, K. S. Preparation, stabilization and use of imatinib mesylate polymorphs for development of medicinal forms
US20130034534A1 (en) 2009-09-29 2013-02-07 Philipp Kroneberg Method for treatment of patients with cystic fibrosis
WO2011039782A1 (en) 2009-09-29 2011-04-07 Ind-Swift Laboratories Limited Processes for preparing imatinib and pharmaceutically acceptable salts thereof
US20110306763A1 (en) 2009-12-10 2011-12-15 Shanghai Parling Pharmatech Co., Ltd. Process for the preparation of imatinib and salts thereof
WO2011095835A1 (en) 2010-02-02 2011-08-11 Actavis Group Ptc Ehf Highly pure imatinib or a pharmaceutically acceptable salt thereof
WO2011100282A2 (en) 2010-02-09 2011-08-18 Dr. Reddy's Laboratories Ltd. Imatinib mesylate polymorphs
WO2011114337A1 (en) 2010-03-15 2011-09-22 Natco Pharma Limited Process for the preparation of highly pure crystalline imatinib base
EA024088B1 (en) * 2010-06-18 2016-08-31 КРКА, д.д., НОВО МЕСТО Alpha-form of imatinib mesylate, processes for preparation thereof and pharmaceutical composition comprising the same
TR201007005A2 (en) 2010-08-23 2011-09-21 Mustafa Nevzat İlaç Sanayi̇i̇ A.Ş. Imatinib base production method
WO2012090221A1 (en) 2010-12-29 2012-07-05 Cadila Healthcare Limited Novel salts of imatinib
US10105356B2 (en) 2011-01-31 2018-10-23 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
ES2913095T3 (en) 2011-01-31 2022-05-31 Avalyn Pharma Inc Aerosolized pirfenidone and pyridone analog compounds and uses thereof
CN106420676B (en) 2011-04-26 2020-04-28 维克多瑞有限责任公司 Administration of iloprost as an aerosol bolus
PL2846859T3 (en) 2012-03-09 2017-07-31 Vectura Gmbh Mixing channel for an inhalation device and inhalation device
CN105560246B (en) * 2012-08-04 2019-07-26 正大天晴药业集团南京顺欣制药有限公司 The preparation and its pharmaceutical composition of imatinib mesylate α crystallization
NZ705985A (en) * 2012-09-17 2018-08-31 Pfizer Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using same
ES2635056T3 (en) 2012-10-26 2017-10-02 Vectura Gmbh Inhalation device for use in aerosol therapy
EP2914244B1 (en) 2012-10-31 2024-01-03 Vectura GmbH Administration of aerosolised iloprost
CN109568319A (en) 2013-01-10 2019-04-05 普尔莫凯恩股份有限公司 Non-selective kinase inhibitor
US20150352111A1 (en) * 2013-01-10 2015-12-10 Pulmokine, Inc. Therapeutic Indications of Kinase Inhibitors
CA2919498C (en) * 2013-07-31 2023-07-25 Windward Pharma, Inc. Aerosol nintedanib compounds and uses thereof
JP6401273B2 (en) 2013-08-16 2018-10-10 ベクチュラ・ゲーエムベーハー Inhaler administration system
US9925184B2 (en) 2013-10-11 2018-03-27 Pulmokine, Inc. Spray-dry formulations
WO2015106150A1 (en) 2014-01-10 2015-07-16 Genoa Pharmaceuticals Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
CN103910711A (en) * 2014-03-24 2014-07-09 福建天泉药业股份有限公司 Imatinib free alkali crystal form and preparation method thereof
CN107206069A (en) 2014-10-10 2017-09-26 埃博灵克斯股份有限公司 The treatment of rsv infection
US11229915B2 (en) 2015-09-09 2022-01-25 Vectura Limited Jet milling method
JP7077219B2 (en) 2015-09-09 2022-05-30 ノバルティス アーゲー Targeted delivery of spray-dried product to the lungs
AU2017351638A1 (en) 2016-10-26 2019-06-13 Genea Biocells USA (Holdings), Inc. Improved generation of muscle lineage cells and therapeutic uses thereof
AU2018336812B2 (en) 2017-09-19 2023-08-31 Evapco, Inc. Air-cooled heat transfer device with integrated and mechanized air pre-cool system
JP2021534079A (en) 2018-08-22 2021-12-09 アヴァリン ファーマ インク. Composition of specially formulated inhaled nintedanib and nintedanib salt
BR112021023014A2 (en) 2019-05-16 2022-02-08 Aerovate Therapeutics Inc Imatinib formulations, manufacture, and uses thereof
US11464776B2 (en) 2019-05-16 2022-10-11 Aerovate Therapeutics, Inc. Inhalable imatinib formulations, manufacture, and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015188243A1 (en) * 2014-06-10 2015-12-17 Cristália Produtos Químicos Farmacêuticos Ltda PROCESS FOR PREPARING IMATINIB AND IMATINIB MESYLATE NON-NEEDLE SHAPED α2 FORM

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Larson et al. ("Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study" Blood, vol. 111(8), April 2008 pp. 4022-1028). *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11806349B2 (en) 2019-05-16 2023-11-07 Aerovate Therapeutics, Inc. Inhalable imatinib formulations, manufacture, and uses thereof
US11813263B2 (en) 2019-05-16 2023-11-14 Aerovate Therapeutics, Inc. Inhalable imatinib formulations, manufacture, and uses thereof

Also Published As

Publication number Publication date
JP2022532431A (en) 2022-07-14
US20230086230A1 (en) 2023-03-23
US20200360377A1 (en) 2020-11-19
US20230056721A1 (en) 2023-02-23
US20240108622A1 (en) 2024-04-04
EP3969111A1 (en) 2022-03-23
WO2020232238A1 (en) 2020-11-19
AU2020274521A1 (en) 2021-12-16
EP3968963A4 (en) 2023-02-01
BR112021023014A2 (en) 2022-02-08
US11813263B2 (en) 2023-11-14
US11806349B2 (en) 2023-11-07
US11413289B2 (en) 2022-08-16
US20200360376A1 (en) 2020-11-19
CA3140641A1 (en) 2020-11-19
SG11202112719XA (en) 2021-12-30
US20220184080A1 (en) 2022-06-16
WO2020232236A1 (en) 2020-11-19
IL288111A (en) 2022-01-01
MX2021014029A (en) 2022-02-21
EP3969111A4 (en) 2023-08-16
EP3968963A1 (en) 2022-03-23
US20200360276A1 (en) 2020-11-19
US20200360275A1 (en) 2020-11-19
US20200360279A1 (en) 2020-11-19
US20220218706A1 (en) 2022-07-14
KR20220050839A (en) 2022-04-25
US20200360277A1 (en) 2020-11-19
CN114514016A (en) 2022-05-17
JOP20210305A1 (en) 2023-01-30
US11229650B2 (en) 2022-01-25
US11298355B2 (en) 2022-04-12
US20240041879A1 (en) 2024-02-08
ZA202109070B (en) 2023-01-25

Similar Documents

Publication Publication Date Title
US20200360477A1 (en) Inhalable formulations for kinase inhibition
JP7066321B2 (en) Aerosols of pirfenidone and pyridone analog compounds, and their use
RU2497524C2 (en) Intrapulmonary administration of fuoroquinolone
US11464776B2 (en) Inhalable imatinib formulations, manufacture, and uses thereof
JP2023171770A (en) Inhalable composition of clofazimine and methods of use thereof
Lee et al. A novel inhaled multi-pronged attack against respiratory bacteria
JP2017523189A (en) Dry powder formulation for inhalation
KR20220080127A (en) Inhalable dry powder composition for lung disease
US20240130966A1 (en) Inhalable imatinib formulations, manufacture, and uses thereof
JP2024506379A (en) Inhalable imatinib formulations, their manufacture and use
US11667612B2 (en) Compositions and methods for delivering pharmaceutical agents
AU2021382051A1 (en) Imatinib formulations, manufacture, and uses thereof
WO2023235267A2 (en) Nintedanib and nintedanib combination dry powder compositions and uses
WO2024013152A1 (en) Inhalable formulation for use in the treatment of bacterial lung infections

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

AS Assignment

Owner name: AEROVATE THERAPEUTICS INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DAKE, BEN;REEL/FRAME:053204/0247

Effective date: 20200427

AS Assignment

Owner name: AEROVATE THERAPEUTICS INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DAKE, BEN;REEL/FRAME:056022/0796

Effective date: 20210416

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

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: FINAL REJECTION MAILED

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: 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

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: FINAL REJECTION MAILED