WO2023161327A1 - Procédés de traitement de maladies inflammatoires et composé destiné à être utilisé dans ces procédés - Google Patents

Procédés de traitement de maladies inflammatoires et composé destiné à être utilisé dans ces procédés Download PDF

Info

Publication number
WO2023161327A1
WO2023161327A1 PCT/EP2023/054526 EP2023054526W WO2023161327A1 WO 2023161327 A1 WO2023161327 A1 WO 2023161327A1 EP 2023054526 W EP2023054526 W EP 2023054526W WO 2023161327 A1 WO2023161327 A1 WO 2023161327A1
Authority
WO
WIPO (PCT)
Prior art keywords
inducers
compound
cyp
cyp3a4
compounds
Prior art date
Application number
PCT/EP2023/054526
Other languages
English (en)
Inventor
Edouard HELLOT
Justine DAO
Natalia RUEDA RINCON
Original Assignee
Galapagos Nv
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 Galapagos Nv filed Critical Galapagos Nv
Publication of WO2023161327A1 publication Critical patent/WO2023161327A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • the present invention relates to improved methods for the use of the compound of the invention in the treatment of inflammatory diseases, and/or diseases associated with hypersecretion of IFNa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL- 12 and/or IL-23, by avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are cytochrome P450 (CYP) inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • CYP cytochrome P450
  • Janus kinases are cytoplasmic tyrosine kinases that transduce cytokine signaling from membrane receptors to STAT transcription factors.
  • JAK family members Four JAK family members are described, JAK1, JAK2, JAK3 and TYK2.
  • JAK family members Upon binding of the cytokine to its receptor, JAK family members auto- and/or transphosphorylate each other, followed by phosphorylation of STATs that then migrate to the nucleus to modulate transcription.
  • JAK-STAT intracellular signal transduction serves the interferons, most interleukins, as well as a variety of cytokines and endocrine factors such as EPO, TPO, GH, OSM, LIF, CNTF, GM-CSF and PRL (Vainchenker, Dusa, and Constantine scu 2008).
  • JAKinibs JAK inhibitors
  • JAKinibs JAK inhibitors with various degrees of selectivity profdes versus the JAK family members.
  • JAK inhibitors with various degrees of selectivity profdes versus the JAK family members.
  • targeting multiple JAK may not be detrimental (Brockman, Giovannetti, and Peters 2011)
  • developing selective JAK inhibitors would be very desirable to develop treatment course tailored to the needs of the patient despite the challenge it represents (Fabian et al. 2005).
  • JAK2 inhibition has proven useful in the treatment of polycythemia and myelofibrosis
  • undesirable effect associated with JAK2 inhibition were observed (O’Shea and Plenge 2012) thus rendering compounds with JAK2 inhibition components unsuitable for the treatment of non-JAK2 mediated diseases.
  • IL-6, IL-10, IL-11, IL-12, IL-13, IL-19, IL- 20, IL-22, IL-23, IL-27, IL-28, IL-29, IL-31, IL-35 and/or type 1 interferons signaling are dependent on TYK2 (Schwartz et al. 2016).
  • TYK2 is involved in type I interferons (including IFNa, INF[3), IL- 23 and IL- 12 signaling (Gillooly, Zhang, and Yang 2016). Since the activity of IL- 12 and IL-23 is particularly increased in patients with auto-immune diseases such as psoriasis and/or inflammatory bowel disorders, selective TYK2 inhibition may be particularly advantageous in the treatment of these diseases while avoiding JAK2 dependent erythropoietin (EPO) and thrombopoietin (TPO) signaling (Neubauer et al. 1998). [0005] Furthermore, TYK2 has been reported as a target for multiple autoimmune disorders, providing protection against inflammatory diseases as well as type 2 diabetes with a limited impact on the immune system (Dendrou et al. 2016).
  • Compound 1 is a small molecule inhibitor of JAK, a family of tyrosine kinases, more particularly TYK2, and is currently under investigation as a drug for the treatment of inflammatory diseases and/or diseases associated with hypersecretion of IFNa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL- 12 and/or IL-23.
  • interferonopathies especially type I interferonopathies
  • IL- 12 and/or IL-23 The identification and synthesis of Compound 1 have previously been described in WO2019/076716.
  • interferonopathies especially type I interferonopathies
  • IL-12 and/or IL-23 in particular diseases such as systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, polymyositis, Sjogren’s syndrome, psoriasis, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, trisomy 21, ulcerative colitis and/or Crohn’s disease.
  • Cytochrome P450 (CYP) enzymes are essential for the metabolism of many medicines and endogenous compounds (Danielson 2002).
  • the cytochrome P450 3 A family is the most abundant subfamily of the CYP isoforms in the liver. There are at least three isoforms: 3A4, 3A5 and 3A7 in adults, of which 3A4 is considered the most important of all CYP enzymes in the liver (Ince et al. 2013).
  • CYP enzymes can be inhibited or induced by drugs, which may result in clinically significant drugdrug interactions that may cause unanticipated adverse reactions or therapeutic failures (Lynch and Price 2007). It is therefore crucial to understand which combinations of drugs should be contraindicated or their co-administration avoided.
  • cytochrome P450 3A4 (CYP3A4) inhibitors are known (https://www.fda.gov/drugs/drug- interactions-labeling/drug-development-and-drug-interactions-table-substrates-inhibitors-and-inducers) and Itraconazole, given its strong CYP3A inhibition, has emerged as probe to evaluate clinical drug-drug interaction (DDI) studies (Liu et al. 2016).
  • Exposure of a drug may be influenced by the co-administration of a CYP3A4 inhibitor (Teo, Ho, and Chan 2015), leading to under or overdosing of said drug; therefore it is essential to ensure stable dose and exposure to avoid undesirable side effects or toxicity.
  • a CYP3A4 inhibitor Teo, Ho, and Chan 2015
  • P-glycoprotein also referred to as “Multidrug Resistance Protein (MDR1)” and by its gene name “ABCB1”
  • MDR1 Multidrug Resistance Protein
  • ABSCB1 ATP Binding Cassette transporters
  • P-glycoprotein plays an important role transporting drug substances outside the cell (efflux) influencing their elimination from the body.
  • P-glycoprotein is an important mediator of drug -drug interactions.
  • the pharmacokinetics of a drug may be altered when co-administered with compounds which inhibit or induce P-glycoprotein (Konig, Muller, and Fromm 2013). It is therefore crucial to understand which combinations of drugs should be contraindicated or their co-administration avoided.
  • a substance acting as P-gp inhibitor which can also act as CYP inhibitor
  • a substance acting as P-gp inhibitor may result in a reduced therapeutic effect due to a doble effect.
  • a P-gp inhibition-mediated enhanced the intracellular accumulation of the parent drug
  • a CYP inhibition-mediated may cause excessive drug accumulation of the parent drug and increased its toxicity, resulting in the need to reduce the dose of the therapeutic agent. Therefore, it is crucial to understand which combinations of drugs should be contraindicated or their co-administration avoided (Wandel et al. 1999).
  • the invention described herein is based upon the observation that the compound of the invention is metabolized by CYP3A4 and/or transported by P-gp, which could result in undesired drug-drug interactions when administered in combination with one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the present invention provides the compound of the invention for use in treating a patient in need of the compound of the invention therapy, characterized in that the treating comprises avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the present invention also provides the use of the compound of the invention in the manufacture of a medicament for treating a patient in need of therapy using the compound of the invention, characterized in that the treating comprises avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the present invention provides a method of administering treatment using the compound of the invention to a patient in need of therapy using the compound of the invention comprising administering the patient a therapeutically effective amount of the compound of the invention, and, avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the patient in need of therapy is a patient suffering from inflammatory diseases, and/or diseases associated with hypersecretion of IFNa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL- 12 and/or IL-23.
  • interferonopathies especially type I interferonopathies
  • diseases such as systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, polymyositis, Sjogren’s syndrome, psoriasis, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, trisomy 21, ulcerative colitis and/or Crohn’s disease.
  • the one or more compounds that may produce potentially serious side effects or toxicity or exhibit adverse drug interactions when co-administered with the compound of the invention are CYP inducers and/or P-gp inducers, more particularly CYP3A4 inducers and/or P-gp inducers, even more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the compound of the invention is according to Formula I below:
  • the chemical name of the compound of the invention is 4-methyl-5-[3-methyl-7-[(6-morpholin-4- ylpyridazin-3 -yl)amino]imidazo [4,5 -b]pyridin-5 -yl]oxypyridine-2 -carbonitrile (Compound 1 ) .
  • the compound of the invention useful in the pharmaceutical compositions and treatment methods disclosed herein is pharmaceutically acceptable as prepared and used.
  • the compound of the invention may be metabolized to yield biologically active metabolites.
  • analogue means one analogue or more than one analogue.
  • the Compound of the Invention means the compound of Formula I or Compound 1, which expression includes the pharmaceutically acceptable salts/cocrystals, and the solvates, e.g. hydrates, and the solvates of the pharmaceutically acceptable salts/cocrystals, where the context so permits.
  • reference to intermediates, whether or not they themselves are claimed, is meant to embrace their salts/cocrystals, and solvates, where the context so permits.
  • ‘Pharmaceutically acceptable salt/cocrystal’ refers to a salt and/or cocrystal of Compound 1 that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • such salts or cocrystals are non-toxic may be inorganic or organic acid addition salts and base addition salts.
  • Cocrystal refers to a crystalline material composed of Compound 1 and a cocrystal former ('coformer') in the same crystal lattice.
  • cocrystal and “co-crystal” are used interchangeably herein.
  • Reference to a certain ‘dose’ or ‘dosage’ of the Compound of the Invention refers to the equivalent weight of free base compound being administered, i.e. not including the weight of any salt, solvate or cocrystal counterpart or component.
  • ‘Pharmaceutically acceptable vehicle’ refers to a diluent, adjuvant, excipient or carrier with which the compound of the invention is administered.
  • Solvate refers to forms of the compound that are associated with a solvent, usually by a solvolysis reaction. This physical association includes hydrogen bonding.
  • Conventional solvents include water, EtOH, acetic acid and the like.
  • the compound of the invention may be prepared e.g. in crystalline form and may be solvated or hydrated.
  • Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances, the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • ‘Solvate’ encompasses both solution-phase and isolable solvates. Representative solvates include hydrates, ethanolates and methanolates.
  • Subject includes humans.
  • the terms ‘human’, ‘patient’ and ‘subject’ are used interchangeably herein.
  • Effective amount means the amount of the compound of the invention that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease.
  • the “effective amount” can vary depending on the disease and its severity, and the age, weight, etc, of the subject to be treated.
  • Preventing refers to a reduction in risk of acquiring or developing a disease or disorder (i.e. causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to a disease-causing agent, or predisposed to the disease in advance of disease onset.
  • prophylaxis is related to ‘prevention’ and refers to a measure or procedure the purpose of which is to prevent, rather than to treat or cure a disease.
  • prophylactic measures may include the administration of vaccines; the administration of low molecular weight heparin to hospital patients at risk for thrombosis due, for example, to immobilization; and the administration of an anti- malarial agent such as chloroquine, in advance of a visit to a geographical region where malaria is endemic or the risk of contracting malaria is high.
  • ‘Treating’ or ‘treatment’ of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e. arresting the disease or reducing the manifestation, extent or severity of at least one of the clinical symptoms thereof).
  • ‘treating’ or ‘treatment’ refers to ameliorating at least one physical parameter, which may not be discernible by the subject.
  • ‘treating’ or ‘treatment’ refers to modulating the disease or disorder, either physically, (e.g. stabilization of a discernible symptom), physiologically, (e.g. stabilization of a physical parameter), or both.
  • “treating” or “treatment” relates to slowing the progression of the disease.
  • inflammatory disease(s) refers to the group of conditions including, rheumatoid arthritis, osteoarthritis, juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, allergic airway disease (e.g. asthma, rhinitis), chronic obstructive pulmonary disease (COPD), inflammatory bowel diseases (e.g. Crohn’s disease, ulcerative colitis), endotoxin-driven disease states (e.g. complications after bypass surgery, acute kidney injury (AKI), Alport syndrome or chronic endotoxin states contributing to e.g. chronic cardiac failure), and related diseases involving cartilage, such as that of the joints.
  • allergic airway disease e.g. asthma, rhinitis
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • endotoxin-driven disease states e.g. complications after bypass surgery, acute kidney injury (AKI), Alport syndrome or chronic
  • the term refers to rheumatoid arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis and inflammatory bowel diseases. More particularly the term refers to psoriasis, rheumatoid arthritis and inflammatory bowel diseases, such as ulcerative colitis and Crohn’s disease.
  • IL-12 and/or IL-23 includes conditions such as systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, polymyositis, Sjogren’s syndrome, psoriasis, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, trisomy 21, ulcerative colitis and/or Crohn’s disease.
  • Adverse Event refers to any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment.
  • An Adverse Event can therefore be any unfavorable and/or unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product.
  • AEs may also include pre- or post-treatment complications that occur as a result of protocol mandated procedures, lack of efficacy, overdose or drug abuse/misuse reports. Pre-existing events that increase in severity or change in nature during or as a consequence of participation in the clinical study will also be considered AEs.
  • Treatment Emergent Adverse Event refers to any Adverse Event (AE) (or worsening of any Adverse Event (AE)) with an onset date on or after the start date of the respective treatment and no later than 30 days after the last dose of the respective treatment.
  • SESs Serious Adverse Event
  • AE Adverse Event
  • death a life-threatening event (an event in which the subject was at risk of death at the time of the event; it does not refer to an event that hypothetically might have caused death if it were more severe.)
  • in-subject hospitalization or prolongation of existing hospitalization persistent or significant disability/incapacity
  • a congenital anomaly/birth defect or a medically significant event (medical and scientific judgment should be exercised in deciding whether other situations should be considered serious such as important medical events that might not be immediately life-threatening or result in death or hospitalization but might jeopardize the subject or might require intervention to prevent one of the other outcomes listed in the definition above).
  • the term “respective medicament” “said medicament” or “contraindicated medicament” refers to the medicament or the one or more compounds that may produce potentially serious side effects or toxicity or exhibit adverse drug interactions when co-administered with the compound of the invention.
  • the term “avoiding the concomitant use or co-administration of’ comprises or relates to avoidance of the use of the contraindicated medicament by looking into alternatives to the respective medicament in a patient in need of therapy with the respective medicament.
  • discontinue and forms thereof, are contemplated to have as alternatives the terms cease, stop, suspend, and quit, and forms thereof.
  • the term “contraindicating” and forms thereof such as “contraindication” are contemplated to contain the instruction to not enter in the contraindicated activity.
  • CYP inducers refers to one or more compounds that decrease the AUC of substrates of a given CYP.
  • CYP inducers may be either weak, moderate or strong inducers.
  • the term refers to CYP3A4 inducers.
  • Examples of CYP3A4 inducers include Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Dexamethasone, Pioglitazone, Rifabutin, Rifampicin, St. John’s wort, Phenobarbital and Troglitazone.
  • the term refers to Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Pioglitazone, Rifabutin, Rifampicin, and St. John’s wort.
  • strong CYP3A4 inducers refers to one or more compounds that decrease the AUC of oral Midazolam or other specific 3A4 substrate by >5-fold or results in a >80% increase in its clearance.
  • strong CYP3A4 inducers include Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Pioglitazone, Rifabutin, Rifampicin, and St. John’s wort (https://www.fda.gov/drugs/drug-interactions-labeling/drug-development-and-drug-interactions-table- substrates-inhibitors-and-inducers).
  • P-gp inducers refers to one or more compounds that decrease the AUC of substrates of P-gp.
  • substrates of p-gp include Dabigatran etexilate, digoxin, fexofenadine, Midazolam and Prazosin.
  • P-pg inducers include Barbiturates, Glucocorticoids, Modafinil, Oxcarbazepine, Pioglitazone, Apalutamide, Carbamazepine, Fosphenytoin, Lorlatinib, Phenytoin, Dexamethasone, Rifabutin, Rifampicin and St.
  • strong P-gp inducer refers to one or more compounds that decrease the AUC of oral P- gp substrate by >5-fold, for example Midazolam and Prazosin.
  • strong P-gp inducers include Barbiturates, Glucocorticoids, Modafinil, Oxcarbazepine, Pioglitazone, Carbamazepine, Phenytoin, Rifabutin, Rifampicin and St. John's Wort (https://www.fda.gov/drugs/drug-interactions- labeling/drug-development-and-drug-interactions-table-substrates-inhibitors-and-inducers).
  • a combined CYP3A4/P-gp inducer is a substance that upon administration to a subject increases CYP3A4 and P-gp mediated activity.
  • Examples of combined CYP3A4/P-gp inducers include phenytoin, carbamazepine, rifampicin, dexamethasone and St. John’s wort.
  • the term “rendered therapeutically ineffective” refers to a loss of efficacy of a medicament in a given patient or patient population by virtue of reduced exposure.
  • the present disclosure includes all isotopic forms of the compound of the invention provided herein, whether in a form (i) wherein all atoms of a given atomic number have a mass number (or mixture of mass numbers) which predominates in nature (referred to herein as the “natural isotopic form”) or (ii) wherein one or more atoms are replaced by atoms having the same atomic number, but a mass number different from the mass number of atoms which predominates in nature (referred to herein as an “unnatural variant isotopic form”). It is understood that an atom may naturally exists as a mixture of mass numbers.
  • unnatural variant isotopic form also includes embodiments in which the proportion of an atom of given atomic number having a mass number found less commonly in nature (referred to herein as an “uncommon isotope”) has been increased relative to that which is naturally occurring e.g. to the level of >20%, >50%, >75%, >90%, >95% or> 99% by number of the atoms of that atomic number (the latter embodiment referred to as an "isotopically enriched variant form").
  • the term “unnatural variant isotopic form” also includes embodiments in which the proportion of an uncommon isotope has been reduced relative to that which is naturally occurring.
  • Isotopic forms may include radioactive forms (i.e. they incorporate radioisotopes) and non-radioactive forms. Radioactive forms will typically be isotopically enriched variant forms.
  • An unnatural variant isotopic form of a compound may thus contain one or more artificial or uncommon isotopes such as deuterium ( 2 H or D), carbon-11 ( n C), carbon-13 ( 13 C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), nitrogen-15 ( 15 N), oxygen-15 ( 15 O), oxygen-17 ( 17 O), oxygen-18 ( 18 O), phosphorus-32 ( 32 P), sulphur-35 ( 35 S), chlorine-36 ( 36 C1), chlorine-37 ( 37 C1), fluorine-18 ( 18 F) iodine-123 ( 123 I), iodine-125 ( 125 I) in one or more atoms or may contain an increased proportion of said isotopes as compared with the proportion that predominates in nature in one or more atoms.
  • an artificial or uncommon isotopes such as deuterium ( 2 H or D), carbon-11 ( n C), carbon-13 ( 13 C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), nitrogen-15 ( 15 N), oxygen-15 ( 15 O), oxygen
  • Unnatural variant isotopic forms comprising radioisotopes may, for example, be used for drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • Unnatural variant isotopic forms which incorporate deuterium i.e. 2 H or D may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • unnatural variant isotopic forms may be prepared which incorporate positron emitting isotopes, such as n C, 18 F, 15 0 and 13 N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • PET Positron Emission Topography
  • Tautomers refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of 7i electrons and an atom (usually H). For example, enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base. Another example of tautomerism is the aci- and nitro- forms of phenylnitromethane that are likewise formed by treatment with acid or base. Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.
  • the compound of the invention may be metabolized to yield biologically active metabolites.
  • the compounds of the invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e. reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • a compound of the invention may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis.
  • the reaction mixture was stirred during one hour after the last addition.
  • the suspension was filtered on Dicalite 4158 (Carlo Erba, ref P8880014), washed with THF (IL) and the combined organic layers were concentrated.
  • the residue was slowly poured into a cold mixture of 20% ammoniac solution (100 mb) and water (2 L). The resulting solid was filtered, washed with water (2 L) and dried to afford the desired product.
  • the present invention provides the compound of the invention, or a pharmaceutical composition comprising the compound of the invention, for use in the treatment of a patient in need of therapy using the compound of the invention, characterized in that the treating comprises avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/ or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the patient in need of therapy is suffering from one or more inflammatory diseases, and/or diseases associated with hypersecretion of INFa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL- 12 and/or IL-23.
  • interferonopathies especially type I interferonopathies
  • the disease is systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, polymyositis, Sjogren’s syndrome, psoriasis, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, trisomy 21, ulcerative colitis or Crohn’s disease.
  • the present invention provides the compound of the invention, or a pharmaceutical composition comprising the compound of the invention for use in the manufacture of a medicament for use in the treatment of a patient in need of therapy using the compound of the invention, characterized in that the treatment comprises avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P- gp inducers.
  • the patient in need of therapy is suffering from one or more inflammatory diseases, and/or diseases associated with hypersecretion of INFa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL-12 and/or IL-23.
  • interferonopathies especially type I interferonopathies
  • the disease is systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, polymyositis, Sjogren’s syndrome, psoriasis, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, trisomy 21, ulcerative colitis or Crohn’s disease.
  • this invention provides methods of treatment of a patient in need of therapy thereof, which methods comprise the administration of an effective amount of the compound of the invention or one or more of the pharmaceutical compositions herein described wherein the treatment additionally comprises avoiding, contraindicating or discontinuing concomitant use or co- administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P- gp inducers.
  • the patient in need of therapy is suffering from one or more inflammatory diseases, and/or diseases associated with hypersecretion of INFa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL-12 and/or IL-23.
  • interferonopathies especially type I interferonopathies
  • the disease is systemic lupus erythematosus, cutaneous lupus erythematosus, lupus nephritis, dermatomyositis, polymyositis, Sjogren’s syndrome, psoriasis, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, trisomy 21, ulcerative colitis or Crohn’s disease.
  • the invention provides a method of administering the compound of the invention to a patient in need of therapy thereof, comprising administering to the patient a therapeutically effective amount of the compound of the invention, and avoiding (concomitant) use or (co-)administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the invention provides a method of administering the compound of the invention to a patient in need of therapy thereof, comprising discontinuing administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, and then administering a therapeutically effective amount of the compound of the invention.
  • the medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, is discontinued concurrently with starting administration of the compound of the invention.
  • the medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers is discontinued at least 12 hours to 1 week prior to or after starting the compound of the invention therapy. This time period, for example, can permit adequate time for tapering and withdrawal without adverse effects.
  • the medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers is discontinued to avoid an adverse drug interaction or to avoid an adverse event; in particular, a treatment-emergent adverse event (TEAE), the contraindicated medicament is preferably discontinued within at least 3 days prior to starting the compound of the invention therapy.
  • TEAE treatment-emergent adverse event
  • the medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers is discontinued within at least 4 days, or at least 5 days, or at least 6 days, or at least 7 days (or one week), or at least 8 days, or at least 9 days, or at least 10 days, or at least 11 days, or at least 12 days, or at least 13 days, or at least 14 days (or two weeks), or at least 15 days, or at least 16 days, or at least 17 days, or at least 18 days, or at least 19 days, or at least 20 days, or at least 21 days (or three weeks), or at least 22 days, or at least 23 days, or at least 24 days, or at least 25 days, or at least 26 days, or at least 27 days, or at least 28 days (or four weeks), or at least 29 days
  • the medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers is discontinued within at least 2 half-lives, or at least 3 half-lives, or at least 4 half-lives, or at least 5 half-lives, or at least 6 half-lives, or at least 7 half-lives, or at least 8 half-lives, or at least 9 half-lives, or at least 10 half-lives, prior to starting the compound of the invention therapy.
  • the medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers is discontinued no earlier than one month, 3 weeks, 2 weeks or 1 week before starting the compound of the invention therapy. Preferably, sufficient time is allowed for tapering and/or withdrawal of the contraindicated medicament.
  • the contraindicated medicament is preferably discontinued within at least 3 days after starting Compound 1 therapy.
  • the patient preferably avoids the use of the medicament or the one or more of compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers to allow sufficient time to avoid adverse drug interactions or adverse events following starting the compound of the invention therapy.
  • the invention provides a method of administering the compound of the invention therapy to a patient in need of thereof and in need of therapy with another medicament that may produce serious side effects or toxicity or may exhibit adverse drug interactions when administered with or alongside CYP3A4 inducers and/or P-gp inducers, comprising administering a therapeutically effective amount of the compound of the invention to the patient, and administering an alternative therapy with a medicament of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the method comprises concomitant use or co-administration of a medicament from the same class or mechanism of action or known to be a suitable alternative medicament for the respective therapy and that are not CYP inducers and/or not P-gp inducers, particularly not CYP3 A4 inducers and/or not P-gp inducers, and more particularly not strong CYP3A4 inducers and/or not strong P- gp inducers.
  • the method comprises discontinuing treatment with the medicament that may produce serious side effects or toxicity or may exhibit adverse drug interactions when administered with or alongside CYP3A4 inducers and/or P-gp inducers and commencing treatment with a medicament from the same class or mechanism of action or known to be a suitable alternative medicament for the respective therapy and the medicament or the one or more compounds that are not CYP inducers and/or P-gp inducers, particularly not CYP3A4 inducers and/or not P-gp inducers, and more particularly not strong CYP3A4 inducers and/or not strong P-gp inducers.
  • the administration of a therapeutically effective amount of the compound of the invention to a patient in need of therapy thereof can be improved.
  • the patient is advised that coadministration of the compound of the invention with a medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, can alter the therapeutic effect or adverse reaction profile of the compound of the invention and/or the respective medicament.
  • the patient if receiving therapy with a medicament or the one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, is advised that coadministration of the compound of the invention with the respective medicament can alter the therapeutic effect or adverse reaction profile of the compound of the invention and/or the respective medicament and that therapy with the respective medicament should be discontinued prior to commencing the compound of the invention therapy.
  • Injection dose levels range from about 0.01 mg/kg/h to at least 10 mg/kg/h, all for from about 1 to about 120 h and especially 24 to 96 h.
  • a preloading bolus may also be administered to achieve adequate steady state levels.
  • the maximum total dose is not expected to exceed about 1 g/day for a 40 to 80 kg human patient.
  • the regimen for treatment usually stretches over many months or years so oral dosing is preferred for patient convenience and tolerance.
  • one to four (1-4) regular doses daily especially one to three (1-3) regular doses daily, typically one to two (1-2) regular doses daily, and most typically one (1) regular dose daily are representative regimens.
  • dosage regimen can be every 1-14 days, more particularly 1-10 days, even more particularly 1-7 days, and most particularly 1-3 days.
  • each dose provides from about 1 mg to about 1000 mg daily dose of the compound of the invention, with particular doses each providing from about 10 mg to about 600 mg daily dose.
  • the compound of the invention is administered from about 60 mg to 200 mg daily dose.
  • the compound of the invention is administered at about 80 mg, 90 mg, 100 mg, 125 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 175 mg, 180 mg, 190 mg, or 200 mg daily dose for the treatment and/or prevention of inflammatory diseases, diseases associated with hypersecretion of IFNa and/or interferons (“interferonopathies”, especially type I interferonopathies), IL- 12 and/or IL-23.
  • interferonopathies especially type I interferonopathies
  • Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
  • the compound of the invention can be administered as the sole active agent or it can be administered in combination with other therapeutic agents, provided that concomitant use or co-administration treatment with a medicament or one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, is avoided, contraindicated or discontinued, said other therapeutic agents may demonstrate the same or a similar therapeutic activity and that are determined to be safe and efficacious for such combined administration.
  • co-administration of two (or more) agents allows for significantly lower doses of each to be used, thereby reducing the side effects seen.
  • the compound of the invention or a pharmaceutical composition comprising the compound of the invention is administered as a medicament.
  • said pharmaceutical composition additionally comprises a further active ingredient, provided said further active ingredient is not selected from one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • the compound of the invention is not an isotopic variant.
  • the compound of the invention according to any one of the embodiments herein described is present as the free base.
  • the compound of the invention according to any one of the embodiments herein described is a pharmaceutically acceptable salt or cocrystal.
  • the compound of the invention according to any one of the embodiments herein described is a solvate of the compound.
  • the compound of the invention according to any one of the embodiments herein described is a solvate of a pharmaceutically acceptable salt or cocrystal of the compound of the invention.
  • the exclusion of one or more of the specified variables from a group or an embodiment, or combinations thereof is also contemplated by the present invention.
  • the compound of the invention When employed as a pharmaceutical, the compound of the invention is typically administered in the form of a pharmaceutical composition. Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound of the invention according to Formula I. Generally, the compound of the invention is administered in a pharmaceutically effective amount. The amount of compound of the invention actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound of the invention administered, the age, weight, and response of the individual patient, the severity of the patient’s symptoms, and the like.
  • compositions of this invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intra-articular, intravenous, intramuscular, and intranasal.
  • the compound of the invention is preferably formulated as either injectable or oral compositions or as salves, as lotions or as patches all for transdermal administration.
  • the compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient, vehicle or carrier.
  • Typical unit dosage forms include prefdled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • the compound of the invention according to Formula I is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
  • Liquid forms suitable for oral administration may include a suitable aqueous or non-aqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like.
  • Solid forms may include, for example, any of the following ingredients, or compound of the inventions of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, citric acid, Primogel, or com starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, citric acid, Primogel, or com
  • Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art.
  • the active compound of the invention according to Formula I in such compositions is typically a minor component, often being from about 0.05 to 10% by weight with the remainder being the injectable carrier and the like.
  • Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the active ingredients When formulated as an ointment, the active ingredients will typically be combined with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in-water cream base.
  • Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.
  • transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
  • the compound of the invention can also be administered in sustained release forms or from sustained release drug delivery systems.
  • sustained release materials can be found in Remington’s Pharmaceutical Sciences.
  • the compound of the invention according to Formula I may be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnesium stearate may be added as a lubricant. The mixture may be formed into 210-270 mg tablets (70-90 mg of active compound of the invention according to Formula I per tablet) in a tablet press.
  • the compound of the invention according to Formula I may be admixed as a dry powder with a starch diluent in an approximate 1 : 1 weight ratio.
  • the mixture may be filled into 250 mg capsules (125 mg of active compound of the invention according to Formula I per capsule).
  • the compound of the invention according to Formula I may be admixed with sucrose (1.75 g) and xanthan gum (4 mg) and the resultant mixture may be blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose (11:89, 50 mg) in water.
  • Sodium benzoate (10 mg) flavor, and color may be diluted with water and added with stirring. Sufficient water may then be added with stirring. Further sufficient water may be then added to produce a total volume of 5 mL.
  • the compound of the invention according to Formula I may be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnesium stearate may be added as a lubricant. The mixture may be formed into 450-900 mg tablets (150-300 mg of active compound of the invention according to Formula I) in a tablet press.
  • the compound of the invention according to Formula I may be dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of approximately 5 mg/mL.
  • Stearyl alcohol (250 g) and a white petrolatum (250 g) may be melted at about 75°C and then a mixture of the compound of the invention according to Formula I (50 g) methylparaben (0.25 g), propylparaben (0.15 g), sodium lauryl sulfate (10 g), and propylene glycol (120 g) dissolved in water (about 370 g) may be added and the resulting mixture may be stirred until it congeals.
  • Compound 1 was incubated at 3 concentrations (0.1, 0.5 and 1 pM) at 37 °C with HLM (with NADPH) at 0.5 and 2 mg/mL for up to 45 minutes to determine whether sufficient metabolism was observed to warrant further investigation.
  • Compoundl (0.1 pM) was also incubated with human recombinant CYP enzymes (1A2, 2B6, 2C8, 2C9, 2C19, 2D6 and 3A4) and NADPH at 37 °C for up to 45 min to identify potential enzymes involved in the metabolism of Compound 1 and to determine conditions for further enzyme kinetics investigations.
  • Compound 1 was incubated with human CYP3A4 recombinant enzyme at 11 substrate concentrations (0.08 - 200 pM) in triplicate at 37 °C with NADPH in order to determine the depletion rate constant at each concentration and thus calculate the Km and Vmax values.
  • Compound 1 showed low metabolism in HLM resulting in an intrinsic clearance up to 8.72 pL/min/mg protein.
  • Compound 1 is an in vitro substrate of the human CYP3A4 isoform.
  • Compound 1 was incubated at 3 concentrations (0.625, 2.00 and 6.25 pM; triplicates) in bidirectional permeability assay (monolayer substrate assay) with Madin-Darby canine kidney multidrug resistance 1 (MDCKII-MDR1) transfected cells for 0, 60 and 120 minutes to determine whether there was active transport (direction apical to basolateral [A-B] and basolateral to apical [B-A]). As an active transport was identified, Compound 1 (2 pM; triplicates) was incubated for 120 minutes with MDCKII-MDR1 cells in the presence of a strong MDR1 inhibitor (10 pM valspodar) to confirm the specificity of the MDR1 transporter.
  • a strong MDR1 inhibitor (10 pM valspodar
  • Compound 1 was incubated with MDCKII-MDR1 cells at 8 concentrations (0.50 - 6.0 pM) in triplicate for 15, 30, 60 and 120 minutes in order to determine the Km and Vmax values to test the unidirectional permeability B-A.
  • Bidirectional transport of Compound 1 was determined by quantification of Compound 1 in the samples using a LC-MS/MS analytical method.
  • Compound 1 is an in vitro substrate of the human MDR1 ABC efflux transporters.
  • the study of the current example is a non-randomized, fixed-sequence, open-label, drug-drug interaction study to evaluate the effect of multiple doses of Itraconazole on the single dose pharmacokinetics of Compound 1 in adult, healthy subjects.
  • the primary objective of this study is to evaluate the effect of Itraconazole on the PK of Compound 1.
  • the secondary objective of this study is to evaluate the safety and tolerability of Compound 1 when administered alone or in combination with Itraconazole and to evaluate the PK of Itraconazole, to confirm relevant exposure for CYP3A4 and P-gp inhibition.
  • the primary outcome measure is C max and AUCo- «> of Compound 1.
  • the secondary outcome measures are:
  • Safety and tolerability of Compound 1 when administered alone or in combination with Itraconazole safety and tolerability, assessed by the incidence and severity of TEAEs, treatment- emergent SAEs, and TEAEs leading to treatment discontinuation;
  • the study is divided in three different periods: a. Screening period: Day-21 to Day -1 b. Open-label study period: Day 1 to Day 11, comprising the periods: i. Open-label study period 1: Day 1 to Day 4 ii. Open-label study period 2: Day 5 to Day 11 c.
  • Subjects receive oral q.d. doses of 200 mg Itraconazole as a solution (10 mg/mL).
  • the study drugs are administered under fasting conditions on Day 1 (Compound 1 alone) and Day 8 (Compound 1 + Itraconazole).
  • Compound 1 is provided as a 25 mg capsule for oral administration.
  • Itraconazole is administered as a 10 mg/mL oral solution (commercially available as Sponarox). Composition of the solution: each milliliter of Sponarox oral solution contains 10 mg of Itraconazole as well as hydroxypropyl-P-cyclodextrin, sorbitol, propylene glycol, hydrochloric acid, flavouring, sodium saccharin, sodium hydroxide and purified water.
  • Subject must be able and willing to comply with restrictions on prior medication: all medication (including OTC and/or prescription medication, dietary supplements, nutraceuticals, vitamins and/or herbal supplements [e.g. St. John’s wort], oral hormonal contraceptives for WOCBP, and hormonal replacement therapy for postmenopausal subjects) except occasional paracetamol (maximum dose 2 g/day and a maximum of 10 g/2 weeks) should be discontinued at least 2 weeks or 5 half-lives of the drug, whichever is longer, prior to the first dose of Compound 1 administration and throughout the study.
  • all medication including OTC and/or prescription medication, dietary supplements, nutraceuticals, vitamins and/or herbal supplements [e.g. St. John’s wort], oral hormonal contraceptives for WOCBP, and hormonal replacement therapy for postmenopausal subjects
  • occasional paracetamol maximum dose 2 g/day and a maximum of 10 g/2 weeks
  • Negative screen for drugs (amphetamines, barbiturates, benzodiazepines, cannabis, cocaine, opiates, methadone, tricyclic antidepressants) and alcohol.
  • Female subjects should be of non-childbearing potential, defined as permanently surgically sterile (bilateral oophorectomy, i.e. surgical removal of ovaries, bilateral salpingectomy or hysterectomy, i.e. surgical removal ofuterus), orwithno menses for 12 ormore months without an alternative medical cause AND a follicle-stimulating hormone (FSH) level in the postmenopausal range.
  • FSH follicle-stimulating hormone
  • Female Subjects o In line with the HMA’s Clinical Trial Facilitation Group recommendation, female subjects are considered of non-childbearing potential if they meet one of the following criteria: o No menses for 12 or more months without an alternative medical cause. A high FSH level in the postmenopausal range may be used to confirm a postmenopausal state in women not using hormonal contraception or hormonal replacement therapy. However in the absence of 12 months of amenorrhea, a single FSH measurement is insufficient. o Permanently surgically sterile (bilateral oophorectomy, i.e. surgical removal of ovaries, bilateral salpingectomy or hysterectomy, i.e. surgical removal of uterus). o All other female subjects are considered to be WOCBP and must use one of the following highly effective methods of birth control prior to the first dose of Compound 1, during the clinical study and for at least 35 days after the last dose of Compound 1:
  • Sexual abstinence defined as refraining from heterosexual intercourse during the entire period of risk associated with the study treatments. The reliability of sexual abstinence needs to be evaluated in relation to the duration of the clinical study and the preferred and usual lifestyle of the subject. o A female subject is not allowed to use any of the following contraception methods in this study:
  • Intrauterine hormone-re leasing system ⁇ Periodic abstinence (e.g. calendar, symptothermal, post-ovulation methods), declaration of abstinence for the duration of a clinical study, withdrawal, spermicides only, and lactational amenorrhea method. Hormonal contraceptives are not allowed due to the potential interference with the drug-drug interaction evaluation. In case a WOCBP has a vasectomized partner, provided that partner is the sole sexual partner of the WOCBP clinical study participant and that the vasectomized partner has received medical assessment of the surgical success, then she is not required to use an additional form of contraception. Within these limits, the specific forms of contraception employed are left to the discretion of the subject, the investigator, and/or the subject’s physician. The safety of Compound 1 during breastfeeding is unknown. Nursing women are not allowed to take part in this clinical study.
  • Non-vasectomized male subjects with female partners of childbearing potential must be willing to use a condom from the time of the first dose of IP, during the clinical study, and until FU, in addition to having their female partner use one of the following forms of contraception:
  • Progesterone-only hormonal contraception associated with inhibition of ovulation oral, injectable, implantable.
  • Sexual abstinence defined as refraining from heterosexual intercourse is considered a highly effective contraceptive measure only if it is the preferred and usual lifestyle of the subject.
  • the reliability of sexual abstinence needs to be evaluated in relation to the duration of the clinical study.
  • Periodic abstinence e.g. calendar, symptothermal, post-ovulation methods
  • declaration of abstinence for the duration of a clinical study withdrawal, spermicides only, and lactational amenorrhea method are not acceptable methods of contraception.
  • hepatitis B virus surface antigen HBsAg
  • HCV hepatitis C virus
  • Baseline is defined as the last available assessment prior to the first intake of Compound 1.
  • a package or kit comprising the compound of the invention, optionally in a container, and, a package insert, package label, instructions or other labelling including information, recommendation or instruction regarding avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, in general, as described in different aspects and embodiments herein.
  • a package insert, package label, instructions or other labelling may include any one or more of the following information, recommendation, or instruction:
  • Informing or advising the patient that concurrent use of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers, should be discontinued, e.g. for at least 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, at least two weeks, at least 3 weeks or at least 4 weeks prior to compound 1 therapy;
  • Informing or advising the patient that concurrent use of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3 A4 inducers and/or strong P-gp inducers, can alter the therapeutic effect of the respective medicament or of the compound of the invention, e.g. decreases the therapeutic effect of Compound 1 or the respective medicament, and/or leads to adverse drug interactions or adverse events, in which case the instruction may further state that therefore concurrent use is contraindicated;
  • CYP inducers and/or P-gp inducers particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers;
  • the CYP3A4 inducer is selected from: Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Dexamethasone, Pioglitazone, Rifabutin, Rifampicin, St. John’s wort, Phenobarbital and Troglitazone.
  • the term refers to Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Pioglitazone, Rifabutin, Rifampicin, and St. John’s wort.
  • the strong CYP3A4 inducer is selected from: Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Pioglitazone, Rifabutin, Rifampicin, and St. John’s wort.
  • the P-gp inducers is selected from: Barbiturates, Glucocorticoids, Modafinil, Oxcarbazepine, Pioglitazone, Apalutamide, Carbamazepine, Fosphenytoin, Lorlatinib, Phenytoin, Dexamethasone, Rifabutin, Rifampicin and St. John's wort.
  • the strong P-gp inducer is selected from: Barbiturates, Glucocorticoids, Modafinil, Oxcarbazepine, Pioglitazone, Carbamazepine, Phenytoin, Rifabutin, Rifampicin and St. John's wort.
  • the CYP3A4 inducer and/or P- gp inducer is a combined CYP3A4/P-gp inducer, which is a substance that upon administration to a subject increases CYP3A4 and P-gp mediated activity, and is in particular phenytoin, carbamazepine, rifampicin, dexamethasone and St. John’s wort, especially rifampicin.
  • a method of administering treatment using the compound according to Formula I or a pharmaceutically acceptable salt/cocrystal thereof, or a solvate or the solvate of a salt/cocrystal thereof, to a patient in need of therapy thereof comprising administering the patient a therapeutically effective amount of compound 1, and avoiding, contraindicating or discontinuing concomitant use or co-administration of one or more compounds that are CYP inducers and/or P-gp inducers, particularly CYP3A4 inducers and/or P-gp inducers, and more particularly strong CYP3A4 inducers and/or strong P-gp inducers.
  • interferonopathies especially type I interferonopathies
  • IL- 12 and/or IL-23 IL- 12 and/or IL-23.
  • the compound for use according to embodiment 1, 4, 5, 6, 7, 8 or 9, the use of the compound according to embodiment 2, 4, 5, 6, 7, 8 or 9, or the method according to embodiment 3, 4, 5, 6, 7, 8 or 9, wherein the patient in need of therapy thereof has polymyositis.
  • the compound for use according to embodiment 1, 4, 5, 6, 7, 8 or 9, the use of the compound according to embodiment 2, 4, 5, 6, 7, 8 or 9, or the method according to embodiment 3, 4, 5, 6, 7, 8 or 9, wherein the patient in need of therapy thereof has trisomy 21.
  • cytochrome P450 (CYP) inducer is a CYP3A4 inducer and is one or more medicaments selected from: Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Dexamethasone, Pioglitazone, Rifabutin, Rifampicin, St. John’s wort, Phenobarbital and Troglitazone.
  • the P-gp inducer is one or more medicaments selected from: Barbiturates, Glucocorticoids, Modafinil, Oxcarbazepine, Pioglitazone, Apalutamide, Carbamazepine, Fosphenytoin, Lorlatinib, Phenytoin, Dexamethasone, Rifabutin, Rifampicin and St. John's wort.
  • a package or kit comprising: i. the compound according to Formula I or a pharmaceutically acceptable salt/cocrystal thereof, or a solvate or the solvate of a salt/cocrystal thereof, and ii. a package insert, package label, instructions or other labelling comprising instructions to avoid or discontinue or contraindication of concomitant use or co-administration of one or more compounds that are CYP inducers.
  • a package or kit according to embodiment 27 further comprising one or more of the features according to embodiments 1 to 26.
  • CYP inducer is a strong CYP3A4 inducer.
  • cytochrome P450 (CYP) inducer is a CYP3A4 inducer and is one or more medicaments selected from Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Dexamethasone, Pioglitazone, Rifabutin, Rifampicin, St.
  • cytochrome P450 (CYP) inducer is a CYP3A4 inducer and is one or more medicaments selected from Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Pioglitazone, Rifabutin, Rifampicin, St. John’s wort, Phenobarbital and Troglitazone.
  • CYP cytochrome P450
  • cytochrome P450 (CYP) inducer is a strong CYP3A4 inducer and is one or more medicaments selected from Barbiturates, Carbamazepine, Glucocorticoids, Modafinil, Oxcarbazepine, Phenytoin, Pioglitazone, Rifabutin, Rifampicin, and St. John’s wort.
  • the P-gp inducer is one or more medicaments selected from Barbiturates,
  • John's wort John's wort.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des procédés améliorés pour l'utilisation du composé selon la Formule (I) dans le traitement de maladies inflammatoires, et/ou de maladies associées à l'hypersécrétion d'INFα et/ou d'interférons ("interféronopathies", en particulier les interféronopathies de type I), l'IL-12 et/ou l'IL-23, évitant, contre-indiquant ou interrompant l'utilisation concomitante ou la co-administration d'un ou plusieurs composés qui sont des inducteurs de cytochrome P450 (CYP) et/ou des inducteurs de P-gp, en particulier des inducteurs de CYP3A4 et/ou des inducteurs de P-gp, et plus particulièrement des inducteurs de CYP3A4 forts et/ou des inducteurs de P-gp forts.
PCT/EP2023/054526 2022-02-24 2023-02-23 Procédés de traitement de maladies inflammatoires et composé destiné à être utilisé dans ces procédés WO2023161327A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP22315040 2022-02-24
EP22315040.0 2022-02-24
EP22184234.7 2022-07-11
EP22184234 2022-07-11

Publications (1)

Publication Number Publication Date
WO2023161327A1 true WO2023161327A1 (fr) 2023-08-31

Family

ID=85328782

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/054526 WO2023161327A1 (fr) 2022-02-24 2023-02-23 Procédés de traitement de maladies inflammatoires et composé destiné à être utilisé dans ces procédés

Country Status (1)

Country Link
WO (1) WO2023161327A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150118229A1 (en) * 2013-10-24 2015-04-30 Abbvie Inc. Jak1 selective inhibitor and uses thereof
WO2019076716A1 (fr) 2017-10-20 2019-04-25 Galapagos Nv Nouveaux composés et compositions pharmaceutiques associées pour le traitement de troubles inflammatoires
WO2021032582A1 (fr) * 2019-08-19 2021-02-25 Galapagos Nv Dérivés de pyrazolo [4,3-c] pyridine et leurs compositions pharmaceutiques pour le traitement de troubles inflammatoires

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150118229A1 (en) * 2013-10-24 2015-04-30 Abbvie Inc. Jak1 selective inhibitor and uses thereof
WO2019076716A1 (fr) 2017-10-20 2019-04-25 Galapagos Nv Nouveaux composés et compositions pharmaceutiques associées pour le traitement de troubles inflammatoires
US20210169879A1 (en) * 2017-10-20 2021-06-10 Galapagos Nv Novel compounds and pharmaceutical compositions thereof for the treatment of inflammatory disorders
WO2021032582A1 (fr) * 2019-08-19 2021-02-25 Galapagos Nv Dérivés de pyrazolo [4,3-c] pyridine et leurs compositions pharmaceutiques pour le traitement de troubles inflammatoires

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"Drug Development and Drug Interactions: Table of Substrates, Inhibitors and Inducers", FDA, 2021
"Greene's Protective Groups in Organic Synthesis", 2006, WILEY
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BABON, JEFFREY J.ISABELLE S. LUCETJAMES M. MURPHYNICOS A. NICOLALEILA N. VARGHESE: "The Molecular Regulation of Janus Kinase", THE BIOCHEMICAL JOURNAL, vol. 462, no. 1, 2014, pages 1 - 13, Retrieved from the Internet <URL:https://doi.org/10.1042/BJ20140712>
BROEKMAN, FLEURELISA GIOVANNETTIGODEFRIDUS J. PETERS: "Tyrosine Kinase Inhibitors: Multi-Targeted", WORLD JOURNAL OF CLINICAL ONCOLOGY, vol. 2, no. 2, 2011, pages 80 - 93, Retrieved from the Internet <URL:https://doi.org/10.5306/wjco.v2.i2.80>
DANIELSON, P. B.: "The Cytochrome P450 Superfamily: Biochemistry, Evolution and Drug Metabolism in Humans", CURRENT DRUG METABOLISM, vol. 3, no. 6, 2002, pages 561 - 97, Retrieved from the Internet <URL:https://doi.org/10.2174/1389200023337054>
DENDROU, CALLIOPE A.ADRIAN CORTESLYDIA SHIPMANHAYLEY G. EVANSKATHRINE E. ATTFIELDLUKE JOSTINSTHOMAS BARBER ET AL.: "Resolving TYK2 Locus Genotype-to-Phenotype Diffe", SCIENCE TRANSLATIONAL MEDICINE, vol. 8, no. 363, 2016, pages 363 - 149, XP055904051, Retrieved from the Internet <URL:https://doi.org/10.1126/scitranslmed.aag1974> DOI: 10.1126/scitranslmed.aag1974
FABIAN, MILES A.WILLIAM H. BIGGSDANIEL K. TREIBERCOREY E. ATTERIDGEMIHAI D. AZIMIOARAMICHAEL G. BENEDETTITODD A. CARTER ET AL.: "A Small Molecule-Kinase Interaction Map", NATURE BIOTECHNOLOGY, vol. 23, no. 3, 2005, pages 329 - 36, Retrieved from the Internet <URL:https://doi.org/10.1038/nbt1068>
GILLOOLY, KATHLEENYIFAN ZHANGXIAOXIA YANG: "BMS-986165 Is a Highly Potent and Selective Allost", ACR MEETING ABSTRACTS, 2016, Retrieved from the Internet <URL:https://acrabstracts.org/abstract/bms-986165-is-a-highly-potent-and-selective-allosteric-inhibitor-oftyk2-blocks-il-12-il-23-and-type-i-interferon-signaling-and-provides-for-robust-efficacy-in-preclinical-models-of-systemic-lupus-e>
HU, BINGJIEXIN ZHOUMICHAEL A. MOHUTSKYPRASHANT V. DESAI.: "Structure-Property Relationships and Machine Learning Mo", MOLECULAR PHARMACEUTICS, vol. 17, no. 9, 2020, pages 3600 - 3608, Retrieved from the Internet <URL:https://doi.org/10.1021/acs.molpharmaceut.0c00637>
INCE, IBRAHIMCATHERIJNE A. J. KNIBBEMEINDERT DANHOFSASKIA N. DE WILDT: "Developmental Changes in the Expression and Function of Cytoc", CLINICAL PHARMACOKINETICS, vol. 52, no. 5, 2013, pages 333 - 45, Retrieved from the Internet <URL:https://doi.org/10.1007/s40262-013-0041-1>
JANNE T. BACKMAN ET AL: "Role of Cytochrome P450 2C8 in Drug Metabolism and Interactions", PHARMACOLOGICAL REVIEWS, vol. 68, no. 1, 31 December 2015 (2015-12-31), US, pages 168 - 241, XP055655323, ISSN: 0031-6997, DOI: 10.1124/pr.115.011411 *
KONIG, JORG, FABIAN MULLER, AND MARTIN F. FROMM.: "Transporters and Drug-Drug Interactions: Important Determinants", PHARMACOLOGICAL REVIEWS, vol. 65, no. 3, 2013, pages 944 - 66, Retrieved from the Internet <URL:https://doi.org/10.1124/pr.113.007518>
LIU, LICHUANAKINTUNDE BELLOMARK J. DRESSERDONALD HEALDSTEVEN FERENC KOMJATHYEDWARD O'MARAMARK ROGGES. AUBREY STOCHSARAH M. ROBERTS: "Best Practices for the Use of Itraconazole as a Repla", JOURNAL OF CLINICAL PHARMACOLOGY, vol. 56, no. 2, 2016, pages 143 - 51, XP055971002, Retrieved from the Internet <URL:https://doi.org/10.1002/jcph.562> DOI: 10.1002/jcph.562
LYNCH, TOMAMY PRICE: "The Effect of Cytochrome P450 Metabolism on Drug Resp.", AMERICAN FAMILY PHYSICIAN, vol. 76, no. 3, 2007, pages 391 - 96
NEUBAUER, H.A. CUMANOM. MULLERH. WUU. HUFFSTADTK. PFEFFER: "Jak2 Deficiency Defines an Essential Developme", CELL, vol. 93, no. 3, 1998, pages 397 - 409, Retrieved from the Internet <URL:https://doi.org/10.1016/s0092-8674(00)81168-x>
OLAVI PELKONEN ET AL: "Inhibition and induction of human cytochrome P450 enzymes: current status", ARCHIVES OF TOXICOLOGY, SPRINGER-VERLAG, BERLIN, DE, vol. 82, no. 10, 11 July 2008 (2008-07-11), pages 667 - 715, XP019652578, ISSN: 1432-0738, DOI: 10.1007/S00204-008-0332-8 *
O'SHEA, JOHN J.J.J PLENGE: "JAK and STAT Signaling Molecules in Imm", IMMUNITY, vol. 36, no. 4, 2012, pages 542 - 50, Retrieved from the Internet <URL:https://doi.org/10.1016/j.immuni.2012.03.014>
SCHWARTZ, DANIELLA M.MICHAEL BONELLIMASSIMO GADINAJOHN J. O'SHEA: "Type I/II Cytokines, JAKs, and New Stra", NATURE REVIEWS. RHEUMATOLOGY, vol. 12, no. 1, 2016, pages 25 - 36, XP055272570, Retrieved from the Internet <URL:https://doi.org/10.1038/nrrheum.2015.167> DOI: 10.1038/nrrheum.2015.167
TEO, YI LINGHAN KIAT HOALEXANDRE CHAN: "Metabolism-Related Pharmacokinetic Drug-drug Inter", BRITISH JOURNAL OF CLINICAL PHARMACOLOGY, vol. 79, no. 2, 2015, pages 241 - 53, Retrieved from the Internet <URL:https://doi.org/10.1111/bcp>
VAINCHENKER, WILLIAMALEXANDRA DUSASTEFAN N. CONSTANTINESCU: "JAKs in Pathology: Role of Janus Kinases in Hematopoietic", SEMINARS IN CELL & DEVELOPMENTAL BIOLOGY, vol. 19, no. 4, 2008, pages 385 - 93, XP025885305, Retrieved from the Internet <URL:https://doi.org/10.1016/j.semcdb.2008.07.002> DOI: 10.1016/j.semcdb.2008.07.002
WANDEL, C.R. B. KIMS. KAJIJIP. GUENGERICHG. R. WILKINSONA. J. WOOD: "P-Glycoprotein and Cytochrome P-450 3A Inhibition: Dissociation Of.", CANCER RESEARCH, vol. 59, no. 16, 1999, pages 3944 - 48
ZHI-XU HE: "Impact of physiological, pathological and environmental factors on the expression and activity of human cytochrome P450 2D6 and implications in precision medicine", 16 November 2015 (2015-11-16), XP055548600, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pubmed/26574146> [retrieved on 20190129], DOI: 10.3109/03602532.2015.1101131 *

Similar Documents

Publication Publication Date Title
US11931363B2 (en) Triazolopyrimidine compounds and uses thereof
US10676479B2 (en) Imidazolepyridine compounds and uses thereof
EP3601281B1 (fr) Formes cristallines de l&#39;acide 4-(1-(1,1-di(pyridin-2-yl)éthyl)-6-(3,5-diméthylisoxazol-4-yl)-1h- pyrrolo[3,2-b]pyridin-3-yl)benzoïque en tant que inhibiteur de bromodomaine
KR101687841B1 (ko) 톨-유사 수용체의 조절제
JP2021513973A (ja) ピリジン誘導体およびhiv感染を処置するためのその使用
US10689378B2 (en) Triazolopyridine compounds and uses thereof
EA032430B1 (ru) Конденсированные пиримидины для лечения вич
CA3077499C (fr) Inhibiteurs de la kinase p38 reduisant l&#39;expression du gene dux4 et des genes aval pour le traitement de la fshd
US20210213037A1 (en) Methods for treating fibrosis
JP2018510138A (ja) Olig2活性の阻害
KR102468670B1 (ko) Olig2 활성의 억제
WO2023125707A1 (fr) Régulateur de la voie p38 mapk/mk2, composition de celui-ci, son procédé de préparation et son utilisation
US20230226040A1 (en) Combination therapy comprising an fgfr inhibitor and a kras inhibitor
WO2023161327A1 (fr) Procédés de traitement de maladies inflammatoires et composé destiné à être utilisé dans ces procédés
WO2023161326A1 (fr) Procédés de traitement de maladies inflammatoires et composé destiné à être utilisé dans ces procédés
EP4041725B1 (fr) Composés de quinoléine et compositions en tant qu&#39;inhibiteurs de ezh2 pour le traitement des maladies de cancer
TWI797366B (zh) 吡咯并[1,2-b]嗒衍生物
AU2022310901A1 (en) Treatment of inflammatory diseases
US20230165863A1 (en) Methods and formulations for administration of thiocarbamate deriviatives a2a inhibitors
CN117677387A (zh) 炎性疾病的治疗
JP2016521740A (ja) 癌を治療するためのイミダゾピリダジン誘導体と有糸分裂阻害剤の組合せ
KR20230074132A (ko) Olig2 억제제를 사용하는 조합 요법

Legal Events

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

Ref document number: 23706788

Country of ref document: EP

Kind code of ref document: A1