NZ613118B2 - Novel composition for the treatment of cystic fibrosis - Google Patents

Novel composition for the treatment of cystic fibrosis Download PDF

Info

Publication number
NZ613118B2
NZ613118B2 NZ613118A NZ61311812A NZ613118B2 NZ 613118 B2 NZ613118 B2 NZ 613118B2 NZ 613118 A NZ613118 A NZ 613118A NZ 61311812 A NZ61311812 A NZ 61311812A NZ 613118 B2 NZ613118 B2 NZ 613118B2
Authority
NZ
New Zealand
Prior art keywords
prostacyclin
inhibitor
inhibitors
group
treprostinil
Prior art date
Application number
NZ613118A
Other versions
NZ613118A (en
Inventor
Michael Freissmuth
Christina Gloeckel
Simon Keuerleber
Xaver Koenig
Original Assignee
Scipharm Sàrl
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 Scipharm Sàrl filed Critical Scipharm Sàrl
Priority claimed from PCT/EP2012/051880 external-priority patent/WO2012107363A1/en
Publication of NZ613118A publication Critical patent/NZ613118A/en
Publication of NZ613118B2 publication Critical patent/NZ613118B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41661,3-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. phenytoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • A61K31/5575Eicosanoids, e.g. leukotrienes or prostaglandins having a cyclopentane, e.g. prostaglandin E2, prostaglandin F2-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • A61K31/5578Eicosanoids, e.g. leukotrienes or prostaglandins having a pentalene ring system, e.g. carbacyclin, iloprost
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • A61K31/558Eicosanoids, e.g. leukotrienes or prostaglandins having heterocyclic rings containing oxygen as the only ring hetero atom, e.g. thromboxanes
    • A61K31/5585Eicosanoids, e.g. leukotrienes or prostaglandins having heterocyclic rings containing oxygen as the only ring hetero atom, e.g. thromboxanes having five-membered rings containing oxygen as the only ring hetero atom, e.g. prostacyclin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

Disclosed is a composition comprising at least one prostacyclin or prostacyclin analogue or a pharmaceutically acceptable salt thereof and at least one phosphodiesterase (PDE) 4 inhibitor for use in preventing or treating cystic fibrosis, wherein the prostacyclin analogue is selected from the group of Treprostinil, Iloprost, Cicaprost or Beraprost or pharmaceutically acceptable salts thereof. of Treprostinil, Iloprost, Cicaprost or Beraprost or pharmaceutically acceptable salts thereof.

Description

Novel composition for the treatment of Cystic Fibrosis The t invention provides compositions comprising a prostacyclin or prostacyclin analogue in combination with a phosphodiesterase tor 4 for use in preventing or ng cystic fibrosis as well as specific compositions.
Prostaglandin l2 (prostacyclin; epoprostenol, PGI2) is an oxygenated metabolite of arachidonic acid formed enzymatically by the sequential activities of cyclooxygenase and PGI synthase enzymes. it is produced constitutively by vascular endothelial and smooth muscle cells and is induced under inflammatory conditions in vascular cells and macrophages, PGl2 is a potent vasodilator and antithrombotic agent whose effects result from binding to a unique heptahelical G protein-coupled receptor termed the l prostanoid (IP)4 receptor. This receptor is coupled to G5— and tes adenylate cyclase, resulting in an acute burst of intracellular cAMP. Since expression of CFTR and mutated CFTR is CAMP-dependent, substances which enhance intracellular levels of CAMP are of interest for development of drugs for ent of CF. Most of these nces, such as forskolin, however, induce a rather unspecific elevation of CAMP, which may have also very harmful effects such as inflammation. Thus there is an unmet need of specific ers of CAMP in lung epithelial cells.
Treprostinil is a potent lP receptor agonist, gh its specificity for this receptor is unknown. Sprague RS. et al., Microcirculation 2008 Jul;15(5):461-71, showed that Prostacyclin analogues (UT—15, Remodulin) stimulate or—mediated CAMP synthesis and ATP release from rabbit and human erythrocytes.
Nucleic phosphodiesterase (PDE) is an enzyme that catalyzes the hydrolysis of CAMP and cyclic 3',5- ine monophosphate (cGMP) to inactive 5'—nucleotide products. CAMP and cGMP exhibit many intracellular s, mediated largely through their stimulatory effect on multisubstrate protein kinases. By inhibiting PDE, the level of cAMP and cGMP is increased, ing in relaxation of airway smooth muscle and inhibition of inflammatory cell activation. PDE4, PDE7 and PDE8 are specific for cAMP.
Phosphodiesterase inhibitors block one or more of the subtypes of the enzyme phosphodiesterase (PDE), therefore preventing the inactivation of the ellular second messengers cyclic adenosine monophosphate (CAMP) and cyclic ine monophosphate (cGMP) by the respective PDE subtype(s). lsozymes of cyclic-3', W0 2012/107363 ’-nucleotide PDE are a critically important component of the CAMP protein kinase A (PKA) signaling pathway. Eleven PDE families have been identified. The superfamily of PDE isozymes consists of at least nine gene families (types), PDE‘l to PDEi 1.
Some PDE families are very e and consist of several es and us isoform-splice variants.
Examples for unspecific PDE inhibitors are theophylline and related xanthine compounds, caffeine, aminophylline etc. Vinpocetine is a PDEi ive inhibitor.
Known PDE2 selective tors are EH NA or Anagrelide, PDE3 selective inhibitors are Enoximone or Milrinone.
PDE4 is the major etabolizing enzyme found in inflammatory and immune cells. PDE4 inhibitors have proven potential as anti—inflammatory drugs, especially in inflammatory pulmonary diseases such as asthma, COPD, and rhinitis.
They suppress the e of cytokines and other inflammatory s, and inhibit the production of reactive oxygen s. Known PDE4 inhibitors are for example Mesembrine, Rolipram, lbudilast etc.
PDE5 inhibitors are primarily metabolized by the cytochrome P450 enzyme CYP3A4. The potential exists for adverse drug interactions with other drugs which t or induce CYP3A4, including HIV protease inhibitors, ketoconazole, itraconazole, and other anti~hypertensive drugs such as Nitro-spray. Examples of PDE5 inhibitors are Sildenafil, Tadalafil, Verdenafil or Udenafil.
Cystic fibrosis (CF) is a genetic disease resulting from mutations in a 230 kb gene on chromosome 7 encoding a 1480 amino acid polypeptide known as the cystic fibrosis transmembrane conductance regulator (CFTR) which serves as a chloride channel in epithelial membranes. Over 1000 mutant alleles have been identified to date. The most common on, AF508, is the deletion of a phenylalanine residue at codon 508 in the cystic fibrosis transmembrane conductance regulator (CFTR) protein. This mutation results in a severe reduction in CFTR function, and leads to the classic cystic fibrosis phenotype characterized with abnormality in exocrine gland functions like raised sweat chloride, recurrent respiratory infection with iectasis, and early—onset of pancreatic insufficiency.
Clinically, CF is usually suspected when one or more l CF phenotypic features are present in a subject. This could be a chronic pulmonary disease alone or very often associated with gastrointestinal and ional abnormalities (e.g. pancreatic insufficiency and ent pancreatitis), salt loss syndromes and male W0 2012!107363 urogenital abnormalities (ie. obstructive ermia). In the human lung, thick, tenacious secretions obstruct the distal s and submucosal glands, which express CFTR. Ductular dilatation of these glands (associated with blockage by mucus) and the plastering of airway surfaces by thick, viscous, neutrophil dominated mucopurulent debris are among the pathological hallmarks of the disease. Pulmonary inflammation is another major cause of the decline in respiratory function in ts with cystic fibrosis and may precede the onset of chronic ion, Mucinous impaction and thick concretions within pancreatic ducts lead to chronic fibrosis, fatty replacement of the gland, or both in a large subgroup of subjects with a previous diagnosis of idiopathic or alcoholic pancreatitis.
Cystic fibrosis is the most common fatal inherited disease in the Caucasian population, affecting about 4 in 10,000 en. In the United States, the median age at death has increased from 8.4 years of age in 1969 to 14.3 years of age in 1998.
The mean age of death has increased from 14 years in 1969 to 32.4 years of age in 2003 (Cystic Fibrosis Foundation). For children born in the 19903, the median sur— vival is predicted to be over 40 years. A major contributor to the significant increase in life expectancy is improved ent of chronic respiratory tract infections and elimination of mucus in CF subjects as well as improved nutrition and r diagnosis.
Loss of the cystic fibrosis transmembrane conductance regulator (CFTR) anion conductance from the apical membranes of airway epithelia disrupts regulation of the airway surface liquid layer. This leads to impaired mucociliary clearance, air- way ion, and mation characteristic of cystic fibrosis (CF). The common AF508 mutation of CFTR is present on at least one allele in >90% of CF patients, and >50% of patients are homozygous for AFSOB, the rest being compound heterozygous. A central issue in CF disease is the inability of this common CFTR variant to achieve the native, folded state that will exit from the asmic reticulum (ER) and traffic to the epithelial cell apical membrane. lf ition of the native conformation is retarded, CFTR is thought to maintain excessive or prolonged interactions with molecular chaperones, which then target the protein for degradation by mechanisms that police the ER for misfolded or incompletely complexed proteins. ociated degradation (ERAD) involves ubiquitination of nt proteins and their delivery to the proteasome for digestion.
If ERAD lags behind the rate of protein synthesis, or during treatment with W0 2012(107363 proteasome inhibitors, aggregates of the mutant n accumulate. CFTR was the first integral membrane mammalian protein to be identified as a substrate for ubiquitin-proteasome mediated degradation, and it has served as a model for the growing list of diseases of n conformation, which account for a diverse set of pathological etiologies.
Essentially all of the AF508 CFTR produced by the cell is destroyed by ERAD.
Also, due to its complex folding pattern, 60—70% of the wild-type (wt) protein may be similarly degraded, although this may vary among cell types. The proteolytic cleavage patterns of the immature forms of wt and AF508 CFTR are similar, whereas the digestion pattern of mature wt CFTR is different. This finding ts the concept that at least a portion of the ER—retained mutant CFTR is present in an inter- mediate conformation that is formed along the normal CFTR folding pathway, as d to the formation of a variant protein structure. For AF508 CFTR, this inter— e conformation cannot proceed beyond a critical step in the folding process, but this implies that AF508 CFTR could be rescued if it were possible to facilitate this step.
A y of experimental conditions, such as reduced temperature, incubation with chemical chaperones, or pharmacological correctors, can e the escape of AF508 CFTR from the ER, yielding a functional anion channel at the cell surface. In addition, investigators have reported restoration of AF508 CFTR function by coexpression of various partial CFTR constructs or subdomains from wt CFTR.
However, a consensus as to which CFTR subdomains are effective in mutant protein rescue is not apparent, and the mechanism of this effect remains obscure. In on, CFTR fragment—induced rescue has been observed ily in cells exogenously overexpressing both the CFTR fragment and full-length AF508 CFTR.
W0 08/098196 describes the ent of pulmonary fibrosis using Treprostinil. Pulmonary fibrosis, however, is an interstitial lung disease that is caused by the accumulation of collagen fibres in the lung; this restricts the capacity of the lung to inhale air: the lung loses its compliance and the airway resistance increases (compliance = stance). As the e progresses there is also an increase in vascular resistance. The site of action of Treprostinil in pulmonary fibrosis is the vasculature and the interstitial space in the a.
Tissieres et al. describe studies using lloprost for the treatment of a patient with cystic fibrosis and secondary ary hypertension. It is disclosed that WO 07363 aeroiised lloprost was effective in lowering pulmonary artery pressure (The annals of ic surgery, vol, 78, no.3, E48-E50).
U82001/006979 A1 describes the use of prostacyclin derivatives like lloprost or Cicaprost for the treatment of fibrotic diseases Cystic fibrosis is ted to ary fibrosis because it is a disease that originates in the bronchial epithelium. Because of the absence of CFTR, there is too little water in the mucus that covers the bronchial epitheiium; accordingly, the cilia cannot move the thick mucus and mucociliary clearance breaks down (mucociliary clearance works like a conveyor belt, where the cilia beat rhythmically in a concen- trated manner to move the mucus back to the trachea and pharynx, from where it may be cleared by swallowing or coughing etc). If mucociliary breaks down, the ia cannot be removed from the bronchi, the i are colonized by bacteria and there are ed bouts of lung infections that destroy the lung. The situation can be remedied by restoring Cl- fluxes to the bronchial epitheiium. Thus, in cystic fibrosis the site of action is the airway epithelium of the bronchi. The site of action is anatomically distinct (lung interstitium vs. bronchial airway), involves a different set of cells (fibroblasts, vascular smooth muscle cells, endothelium versus ing and secreting bronchial epithelial cells) and presumably also involves different receptors (prostacyclin receptor vs possibly EP2-receptor). 05016345A1 and U82005101608A1 describe the use of PDE5 inhibitors for the treatment of pulmonary hypertension.
U82009325976A1 discloses new prostacyclin derivatives which may be used also in combination with a PDE 5 inhibitor for use in the treatment of pulmonary arterial hypertension.
Clinically used PDE inhibitors were tested for activating the chloride secretion in the setting of low CAMP levels as described by Cobb BR. et al., (Am J Respir Cell Moi Biol. 2003 Sep;29(3 Pt 1):410-418).
PDE 5 inhibitors, Sildenafii and Vardenafil, and their role in chloride ort in cystic fibrosis are described by Lubamba B. Et at. (Am J Respir Crit Care Med. 2008 Mar 1;177(5):506—515).
PDE 5 inhibitors and their role in AF508 CFTR l function is bed by Clarke Lane L. (J Respir Crit Care Med. 2008 Mar ‘l;177(5)1469-70).
In W02010106494A1 the use of mesembrine HCi, a known weak PD E4 inhibitor for treating ers is disclosed.
U820070249668 describes a composition containing a PDE inhibitor and a prostacyclin analogue to increase the ATP content in red blood cells.
Presently, no treatments of cystic fibrosis are available that significantly improve quality of life of patients over a longer . ore it is an object of the invention to provide compositions for treatment that can enhance the expression of AF508 CFTR and/or chloride channel function in epithelial cells of the lung or to at least provide the public with a useful alternative.
Short description of the invention: The object of the invention is achieved by ing a composition comprising at least one prostacyclin or an ue, a derivative or a pharmaceutically acceptable salt thereof in combination with at least one PDE 4 inhibitor for use in preventing or ng cystic fibrosis. As an alternative embodiment, said composition can r comprise at least one PDE5 and/or PD7 and/or PDE8 inhibitor.
Specifically, the cyclin analogue is selected from the group of Treprostinil, lloprost, Cicaprost or Beraprost or derivatives or pharmaceutically acceptable salts thereof. In one embodiment of the present invention the Treprostinil derivative is an acid derivative, a prodrug, a polymorph or an isomer of Treprostinil.
According to the invention, the PDE4 tor can be specifically selected from the group of Ro 4, lbudilast, Roflumilast and its N-Oxide, Cilomilast, BAY 19—8004, 003, AWD 12-281, SCH 351591, Ciclamilast, Piclamilast, CGH2466, Mesembrine, Rolipram, Luteolin and Drotaverine.
According to the invention, the PDE5 inhibitor can be specifically selected from Avanafil, Lodenafil, Mirodenafil, Sildenafil citrate, Tadalafil, Vardenafil and Udenafil; the PDE7 and PDE8 tors may be selected from Dipyridamol, BRL— 50481 and PF—04957325 According to a ic embodiment of the invention, the composition specifically consists of one type of cyclin ue and one type of PDE4 inhibitor.
As a specific embodiment, the composition comprises Treprostinil and a PDE4 inhibitor selected from the group of RO 20—1724, Roflumilast and ast.
A further embodiment of the invention, the composition comprises additional PDE inhibitors selected from PDE5, PDE7 or PDE8 inhibitors. in another embodiment, the invention provides a composition free of interferon.
Specifically, the inventive composition is formulated as a pharmaceutical composition.
Any known administration forms can be used for administering the inventive combination, for example it can be intravenous or subcutaneous administration or inhalation administration, or in an orally available form selected from the group of sustained release forms, tablets and capsules.
According to a specific embodiment, the effective amount of Treprostinil or a pharmaceutically acceptable salt thereof is preferably of about 1.0 ng/kg of body weight, lbudilast is preferably up to 5x30 mg, preferably up to 2x30 mg the effective amount of the PDE4 inhibitor is approx. 0.5 mg. Additionally, one or more inhibitors of the group of PDE5 and PDE7 inhibitors may be contained in an effective amount of about 0.5mg of each of the inhibitors.
The present invention also provides an in vitro method for increasing the CAMP level in a cell wherein said cell is contacted with at least one prostacyclin or prostacyclin analogue and at least one PDE4 inhibitor or a pharmaceutically acceptable salt thereof. Additionally, a PDE5, PDE7 and/or PDE8 inhibitor may further be used ing to said method.
Specifically, the cell is an epithelial cell, more ically it may be a bronchoepithelial cell.
A therapeutic combination is also provided, comprising at least one cyclin or prostacyclin analogue and at least one PDE4 inhibitor or a pharmaceutically able salt thereof, wherein the prostacyclin analogue and the PDE4 tor are provided in amounts which together are sufficient to treat and/or t at least one symptom associated with cystic fibrosis. More specifically, the prostacyclin analogue and PDE4 inhibitor are formulated for administration by inhalation.
Said therapeutic ition may, according to an alternative embodiment, contain at least one further inhibitor selected from the group of PDE5, PDE7 and PDE8 inhibitors.
Figures: Fig. 1: lation of CAMP in IBS—‘l cells after incubation with stinil alone or in combination with the PDE 4 inhibitors lbudilast ) and azol ('lOOtJM).
Fig. 2: Accumulation of CAMP in lB3—1 cells after incubation with Treprostinil in combination with the PDE 4 inhibitors RO1724 (iOOpM) and Roflumilast (iOpM).
Fig. 3: Activation of a Cl-current by Treprostinil in the human bronchial epithelial IBB—1 cell line transiently expressing CFTR—wt.
Fig. 4: Accumulation of cAMP in lB3-1 cells stimulated by 10 uNl Treprostinil in the absence and presence of the indicated concentrations of Dipyridamole, lbudilast, R020-1724 or Roflumilast. Cells were metabolically prelabelled with [3H]adenine for 4 h and subsequently incubated with the indicated compounds for 30 min. The formation of [3H]cAMP was ined as outlined under Materials and s.
Data are means i s.e.m. (n=3).
Fig. 5: Concentration-response curve for Trepostinil—induced CAMP accumulation in [BB-1 cells. Cells were ted with increasing concentrations of Treprostinil in the e and presence of the indicated concentrations of lbudilast, R020-1724 or Roflumilast. Cells were metabolically prelabelled with [3H]adenine for 4 h and subsequently ted with the indicated compounds for 30 min. The formation of [3H]cAMP was determined as outlined under als and s.
Data are means -i_- s.e.m. (n=3).
Fig. 6: Effect of selected phosphodiesterase inhibitors on basal CAMP accumulation in lBB-i cells. Cells were metabolically prelabelled with [3H]adenine for 4 h and subsequently incubated in the absence (basal) and presence of the indicated concentrations of damole, Ro—ZO—i 724 or Roflumilast for 30 min. The levels of [3H]cAMP were determined as outlined under Materials and Methods. Data are means i s.e.m. (n=3).
Detailed description of the invention it has been surprisingly found by the inventors that cyclin or analogues or a pharmaceutically acceptable salt thereof in combination with a PDE4 inhibitor can be used for treating cystic fibrosis. It was shown that a combination of prostacyclin or prostacyclin analogues and PDE4 tors have istic effect in cAMP increase, specifically in the affected human airway epithelial cells, compared to the use of single substances. Said effect may further be enhanced by the presence of further PDE inhibitors ed from PDE5, PDE? and PDE8 tors.
Synthetic prostacyclin analogues can be for example, but are not limited to, Treprostinil, lloprost, Cicaprost or Beraprost.
W0 2012/107363 Treprostinil is marketed as RemodulinTM. Treprostinil is a (tR,2R,3aS,QaS)- [[2,3,3a,4,9,9a-hexahydro—2-hydroxy[(BS)-3—hydroxyoctyl]-1H-benz[t]tndenyl] oxy]acetic acid monosodium salt. lloprost is marketed as "llomedine" and is a 5-{(E)—(1S,58,6R,7R)~7—hydroxy— (3$, 4RS)—3—hydroxy—4—methy|octen-6—inyl]—bi-cyclo{3.3.0]octan-3— ylidene}pentanoic acid.
Beraprost is a 2,3,3a,8b-tetrahydro-2—lriydroxy—1-(3—hydroxymethylocten— 6-ynyl)-1H—cyclopenta(b)benzofuranbutanoic acid.
Cicaprost is a )[(3aS,4S,5R,6aS)—5-hydroxy—4-[(3$,4S)-3—hydroxy~4— methylnona—1,6—diynyl]—3,3a,4,5,6,6a—hexahydro-1H—pentalen—2—ylidene]ethoxy]acetic acid.
In reference to prostacyclin, PDE4 and PDES, PDE7 or PDE8 inhibitors, according to the present invention, the term ”prostacyclin analogues”, “inhibitor analogs" or “PDE4, PDE5, PDE 7 or PDE8 inhibitor analogs” means derivatives or analogues of said substances. The terms "analogue" or "derivative" relate to a chemical molecule that is similar to another chemical substance in structure and function, often ing structurally by a single element or group, which may differ by modification of more than one group (eg., 2, 3, or 4 groups) if it s the same function as the parental chemical substance. Such modifications are routine to skilled persons, and include, for example, additional or tuted chemical es, such as esters or amides of an acid, ting groups such as a benzyl group for an alcohol or thiol, and tert—butoxylcarbonyl groups for an amine. Also included are cations to alkyl side chains, such as alkyl substitutions (e.g., , dimethyl, ethyl, etc), modifications to the level of saturation or unsaturation of side chains, and the addition of modified groups such as substituted phenyl and phenoxy. Derivatives can also include conjugates, such as biotin or avidin moieties, enzymes such as horseradish peroxidase and the like, and radio—labeled, bioluminescent, chemoluminescent, or fluorescent moieties. r, moieties can be added to the agents described herein to alter their pharmacokinetic properties, such as to increase half—life in vivo or ex vivo, or to increase their cell penetration properties, among other desirable ties. Also included are prodrugs, which are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.) The term ”derivative" also includes within its scope alterations that have been made to a parent sequence ing additions, deletions, andlor substitutions that provide for functionally equivalent or functionally improved molecules.
Suitable prostacyclin or prostacyclin analogue derivatives include but are not limited to acid derivatives, pro-drugs, sustained release forms, inhaled forms and oral forms of Treprostinil, lloprost, Cicaprost or Beraprost.
According to a specific embodiment of the invention, the Treprostinil derivative is selected from the group of acid derivatives, gs, polymorphs or isomers of stinil.
Similarly, lloprost, Cioaprost or Beraprost derivatives can be acid derivatives, prodrugs, polymorphs or isomers therefrom. The term cyclin derivative also covers pharmaceutically acceptable salts thereof. A pharm aceutically able salt of a prostacyclin or a prostacyclin analogue of this invention can be formed between an acidic and a basic group of the nd, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
Specifically, physiologically acceptable salts of prostacyclin analogues include salts derived from bases. Base salts include ammonium salts (such as quaternary ammonium salts), alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium, salts with organic bases such as dicyclohexylamine and N-methyl—D-glucamine, and salts with amino acids such as arginine and lysine. ically, the use of Treprostinil is advantageous according to the invention.
Treprostinil can sfully enhance the expression of AF508 CFTR and/or the chloride channel function in epithelial cells of the lung of cystic is ts.
It has been surprisingly shown that a prostacyclin analogue in combination with a PDE4 tor and optionally in combination with a PDES and/or PDE7 and/or PDE8 tor leads to synergistic stimulation of CAMP production and/or increase of CAMP content in bronchoepithelial cells.
Interestingly PDE8 inhibitors like Anagrelide and Cilostazol did not induce any accumulation of CAMP in experiments.
Given this ability to stimulate CAMP production through the IP receptor, and the limited presence of IP ors to a small number of cell—types (such as epithelial lung cells), a cyclin or analogue thereof, for example Treprostinil might induce expression and gating of CFTR and mutCFTR in a specific manner which can be used for treatment of CF, in particular, when combined with said PDE4 inhibitors to induce a long lasting increase in CAMP levels within the airway lium.
According to a further embodiment, said CAMP increase may further be induced by combination of a PDE4 inhibitor with further selected PDE inhibitors from the group of PDE5, PDE? and/or PDE8 inhibitors.
PDE4 inhibitors are approved for the treatment of COPD and asthma; the main target in COPD and asthma is to reduce the hyperreactivity of the smooth muscle cells lining the airways. Raising cAMP levels in smooth muscle cells has long been known to cause relaxation of the smooth , via action on myosin light chain kinase. In on, PDE4 inhibitors are thought to reduce the immune response that drives ic asthma by targeting monocytes, eosinophil and basophil granulocytes, B and T cells, eg. the inflammatory cells. Neither of these two mechanisms is relevant as a mode of action in cystic fibrosis. In cystic fibrosis, cAMP levels must be raised in a very different cellular compartment, is. the airway epithelium. In fact, to the best of our knowledge, there are no scientific reports that show that PDE4 is the relevant isoform that es receptor-mediated cAMP lation within the airway epithelium.
According to the present ion, any PDE4 inhibitor or its analogue can be used having inhibitory activity towards the PDE4 enzyme. Thus, it is not excluded that the PDE4 inhibitor can further inhibit other PDE s as well.
Specifically, the PDE4 inhibitor can be a specific PD E4 inhibitor, The PDE4 inhibitor of the invention can be, but is not d to Ro 20-1724, lbudilast, Roflumilast (3—(cyclopropylmethoxy)—N-(3,5—dichloropyridinyl)—4- (difluoromethoxy)benzamide) and its N-Oxide, Cilomilast, BAY 19—8004, CC3, AWD 12—281 (N—(3,5-dichloropyridinyl)-2—[1—(4—fluorobenzyl)hydroxy—1 H-indol-3—yl]-2— oxoacetamide), SCH 351591 (N—(S,5-dichloro—i -oxido—4—pyridinyl )-8—methoxy—2— (trifluoromethyl)~5-quinoline carboxamide), Ciclamilast, Piclamilast, CGH2466, Mesembrine, am, Luteolin and Drotaverine or functional analogs thereof.
More specifically, the ition for use of preventing or treating CF, specifically by raising the cAMP levels in the bronchoepithelial cells of individuals suffering from CF can specifically comprise Treprostinil and Roflumilast or Treprostinil and lbudilast or Treprostinil and Ro-20—i724. PDE5 inhibitors have been shown to increase cyclic nucleotide second messenger levels in the smooth muscle cells.
According to the present invention, any PDE5 inhibitor or its analogue can be used having inhibitory activity towards the PDE5 enzyme. Thus, it is not excluded that the PDE5 inhibitor can further inhibit other PDE enzymes as well.
According to the invention, the PDE5 inhibitor can be specifically selected from Avanafil (4-[(3-chIoro—4—methoxybenzyl)amino]—2— [2-(hydroxymethyl)-1—pyrrolidinyl]- N— imidinylmethyI)—5—pyrimidinecarboxamide), Lodenafil (bis~(2-{4-[4—ethoxy~3— hyl-7—oxo-3~propyI-6,7-dihydro-1 H-pyrazoloi4,3-d]pyrimidin—5—yl)~ benzenesulfonyllpiperazinyI}-ethyl)carbonate), Mirodenafil (5-ethyl-3,5-dihydro [5~([4~(2—hydroxyethyl)~1 —piperazinyl]sulfonyl)—2—propoxyphenyI]—7-propyI—4H— pyrrolo[3,2-d]pyrimidin—4—one), Sildenafil e (1 -[4—ethoxy(6,7-dihydro-1—methy|— 7-oxopropyl-1H-pyrazolo[4,3-d}pyrimidin—5-yl) phenylsulfonyl]—4-m ethylpiperazine), Tadalafil (BR-trans)—6—(i ,3-benzodioon—5-yl)— 2,3,6,7,12,12a-hexahydro~2~methyl- pyrazino [1', 221,6] [3,4~b]indole-‘I ,4—dione), Vardenafil (4—[2—ethoxy-5—(4— ethylpiperazin~1-y|)su|fonyI-pheny|]— 9-methyIpropyl- 3,5,6,8—tetrazabicyclo[4.3.0] nona—3,7,9—trien—2—one) or Udenafil (3—(1~methyl—7—oxo-3—propyl-4,7-dihydro—1H— pyrazolo[4,3—d]pyrim idin~5—yI)-N-[2-(1 -methylpyrroIidin-2—yl)ethyI] propoxybenzenesulfonamide) or any functional analogs thereof.
In a specific embodiment of the invention, the composition may comprise stinil, Roflumilast and optionally a PDE5, PDE7 or PDE8 inhibitor or Treprostinil, lbudilast and optionally PDE5, PDE7 or PDE8 inhibitor or Treprostinil, Ro—20-1724 and ally PDE5, PDE7 or PDE8 inhibitor.
In a further embodiment of the invention, the composition may se Beraprost. Roflumilast and optionally a PDE5, PDE7 or PDE8 inhibitor or Beraprost, lbudilast and optionally a PDE5, PDE7 or PDE8 inhibitor or Beraprost, Ro—20—1724 and optionally a PDE5, PDE7 or PDE8 inhibitor.
In a further embodiment of the invention, the composition may comprise lloprost, Roflumilast and optionally a PDE5, PDE7 or PDE8 inhibitor or Iloprost, lbudilast and optionally a PDE5, PDE7 or PDE8 inhibitor or Iloprost, Ro-20~1724 and optionally a PDE5, PDE7 or PDE8 inhibitor.
In a r ment of the invention, the com position may comprise ost, Roflumilast and optionally a PDE5, PDE7 or PDE8 tor or Cicaprost, ast and optionally a PDE5, PDE7 or PDE8 inhibitor or Cicaprost, Ro1724 and optionally a PDE5, PDE7 or PDE8 inhibitor.
According to the present invention, any PDE7 or PDE8 inhibitor or its analogue may be used having inhibitory activity towards the PDE7 or PDE8 enzyme.
Thus, it is not excluded that the PDE7 or PDE8 inhibitor can further inhibit other PDE enzymes as well.
According to the invention, the PDE7 inhibitor can be specifically selected from Dipyridamol and BRL 50481.
According to the invention, the PDE8 inhibitor can be specifically selected from 1,5-substituted nipecotic amides and 7325.
In an alternative embodiment of the invention, the composition may ically comprise stinil, Roflumilast and Dipyridamol or Treprostinil, lbudilast and Dipyridamol or Treprostinil, Ro‘l7’24 and Dipyridamol.
In a further alternative embodiment of the invention, the composition may specifically comprise Beraprost, Rotiumilast and Dipyridamol or Beraprost, lbudilast and Dipyridamol or Beraprost, 1724 and Dipyridamol. in yet a further alternative embodiment of the invention. the composition may specifically comprise lloprost, Roflumilast and Dipyridamol or lloprost, lbudilast and Dipyridamol 0r lloprost, Ro1724 and Dipyridamol. in an further embodiment of the invention, the composition may specifically comprise Cicaprost, Roflumilast and Dipyridamol or Cicaprost, lbudilast and Dipyridamol or Cicaprost, Ro—20—1724 and Dipyridamol.
Alternatively the composition may se Treprostinil, Roflumilast and BRL 50481 or Treprostinil, lbudilast and BRL 50481 or Treprostinil, Ro-20—1724 and BRL 50481. atively, the ition may specifically comprise Treprostinil, Roflumilast and PF—4957325 or Treprostinil, lbudilast and PF-4957325 or Treprostinil, Ro-20—1724 and PF~4957325. ing to the invention the term “at least one” or “a" means that one type of prostacyclin or prostacyclin analogue and one type of PDE4 inhibitor and optionally one or more of PDE5, PDE7 or PDE8 tors is t for use in the treatment or tion of cystic fibrosis, specifically for the use to se the CAMP level in bronchoepithelial cells. However, alternatively, the ition may also comprise more than one type of cyclin or prostacyclin analogue and more than one type of PDE4 inhibitor and optionally one or more of PDE5, PDE7 or PDE8 inhibitors, specifically two, three, four or more than four types or any combinations of W0 07363 prostacyclins or prostacyclin analogues and PDE4 and optionally PDE5, PDE7 and/or PDE8 inhibitors.
The invention further provides a specific composition comprising Treprostinil and one or more PDE4 inhibitors selected from the group of RO 20-1724, Roflumilast and lbudilast.
The inventive composition can be formulated as a pharmaceutical composition.
The composition of the ion can be present in any form which can be used for administration.
The ic dose of a compound administered according to this invention to obtain therapeutic or prophylactic effects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the route of administration, the age, weight and response of the individual patient, the condition being treated and the severity of the patient‘s symptoms.
In general, the compounds of the invention are most desirably administered at a concentration that will lly afford effective results without causing any s side effects and can be administered either as a single unit dose, or if desired, the dosage may be divided into convenient ts administered at suitable times hout the day.
The composition can be provided in a y of systemic and topical formulations. The systemic or topical formulations of the invention are selected from the group of oral, intrabuccal, intrapulmonary, rectal, intrauterine, intradermal, topical, dermal, parenteral, intratumor, intracranial, ulmonary, buccal, sublingual, nasal, subcutaneous, intravascular, intrathecal, inhalable, respirable, intraarticular, intracavitary, table, ermal, iontophoretic, intraocular, ophthalmic, vaginal, optical, intravenous, intramuscular, intraglanduiar, intraorgan, intraiymphatic, slow release and enteric coating formulations. The actual ation and compounding of these different formulations is known in the art and need not be detailed here, The composition may be administered once or several times a clay.
Formulations suitable for atory, nasal, intrapulmonary, and inhalation administration are preferred, as are topical, oral and parenteral formulations. In general, the formulations are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, g the product into desired formulations. itions suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing the composition as a powder or granules; as a solution or a suspension in an s or non—aqueous liquid; or as an oil-in-water or water-in-oil emulsion.
Compositions suitable for parenteral administration se e aqueous and non—aqueous injection solutions of the active compound, which preparations are preferably isotonic with the blood of the recipient. These preparations may contain anti—oxidants, buffers, bacteriostatic agents and s which render the compositions isotonic with the blood of the recipient. Aqueous and non—aqueous sterile suspensions may include suspending agents and thickening agents. The compositions may be presented in unit—dose or multi—dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried or lyophilized condition requiring only the addition of the sterile liquid carrier, for example, saline or water-for~injection immediately prior to use.
Nasal and instillable formulations comprise purified aqueous solutions of the active compound with preservative agents and isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucous membranes.
The composition disclosed according to the invention may be administered into the respiratory system either by inhalation, respiration, nasal stration or ulmonary instillation (into the lungs) of a subject by any le means, and are ably administered by generating an aerosol or spray comprised of powdered or liquid nasal, intrapulmonary, respirable or inhalable particles. The respirable or inhalable particles sing the active compound are inhaled by the subject, eg, by inhalation or by nasal administration or by instillation into the respiratory tract or the lung itself. The formulation may comprise respirable or inhalable liquid or solid les of the active compound that, in accordance with the present invention, include respirable or inhalable particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and continue into the bronchi and alveoli ofthe lungs. In l, particles ranging from about 0.05, about 0.1, about 0.5, about 1, about 2 to about 4, about 6, about 8, about 10 microns in diameter. More particularly, about 0.5 to less than about 5 pm in er, are respirable or inhalable. les of non—respirable size which are included in an aerosol or spray tend to deposit in the throat and be swallowed. The quantity of non-respirable particles in the aerosol is, W0 2012‘107363 thus, preferably minimized. For nasal administration or intrapulmonary instillation, a particle size in the range of about 8, about 10, about 20, about 25 to about 35, about 50, about 100, about 150, about 250, about 500 pm in diameter is preferred to ensure retention in the nasal cavity or for instillation and direct deposition into the lung. Liquid formulations may be squirted into the respiratory tract or nose and the lung, particularly when administered to newborns and infants.
Aerosols of liquid particles comprising the active compound may be produced by any suitable means, such as with a nebulizer. Nebulizers are commercially available devices which transform solutions or suspensions of the active ient into a therapeutic aerosol mist either by means of acceleration of a compressed gas, lly air or oxygen. le compositions for use in nebulizer consist of the active ient in liquid carrier, the active ingredient comprising up to 40% w/w composition, but preferably less than 20% w/w carrier being typically water or a dilute aqueous alcoholic solution, preferably made isotonic with body fluids by the addition of, for example sodium chloride. Optional additives include preservatives if the composition is not prepared sterile, for e, methyl hydroxybenzoate, anti- oxidants, flavoring , volatile oils, buffering agents and surfactants. ls of solid particles sing the active compound may likewise be produced with sold particulate medicament aerosol generator. Aerosol generators for administering solid particulate medicament, product les which are able, as explained above, and generate a volume of aerosol containing a predetermined metered dose of a medicament at a rate suitable for human stration. Examples of such aerosol generators include d dose inhalers and insufflators.
In one embodiment, the delivery device comprises a dry powder inhalator (DPI) that delivers single or multiple doses of the composition. The single dose inhalator may be provided as a disposable kit which is sterilely preloaded with enough formulation for one application. The inhalator may be provided as a rized inhalator, and the ation in a piercable or openable capsule or cartridge. The kit may optionally also comprise in a separate container an agent such as other therapeutic compounds, excipients, surfactants (intended as therapeutic agents as well as formulation ingredients), antioxidants, flavoring and coloring agents, fillers, volatile oils, buffering agents, dispersants, surfactants, antioxidants, flavoring agents, bulking agents, lants and preservatives, among other suitable additives for the different formulations.
W0 2012/107363 2012/051880 Due to the high lic stability of some prostacyclin analogues like Treprostinil, or if provided as lipid based or pegylated forms of the cyclins or prostacyclin analogues, the substances can also be administered as depot medicaments.
PDE4, PDE5, PDE 7 and PDE8 inhibitors are also metabolically stable, therefore the combination of the prostacyclin or prostacyclin analogue and the PDE4 tor optionally together with one or more of PDES, PDE 7 or PDE8 inhibitors can also be formulated as depot medicaments.
Aerosolized delivery of the composition may result in a more homogeneous distribution of the agent in a lung, so that deep lung delivery is obtained. y the dosage of application may be reduced due to the sustained presence of the agent at the site of action in the lung.
The composition can for example be given by a nebulizer. The advantage of the nebulizer method of delivery is that less of the nce reaches the ic circulation. The composition can be given several times a day, for e five to 10 times a day, however due to the synergistic effect of the prostacyclin or prostacyclin analogue and the PDE4, optionally in combination with one or more of PDE5, PDE7 and/or PDE8 inhibitors, the dosing frequency may generally be reduced.
The composition can be administered with any pharmaceutically acceptable substances or carriers or ents as known in the art. These can be for example, but are not restricted to water, neutralizing agents like NaOH, KOH, izers, DMSO, saline, betaine, taurine etc.
The term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the US.
The term "carrier” refers to a diluent, adjuvant, ent, or vehicle with which the pharmaceutical composition is administered. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable excipients include starch, glucose, lactose, sucrose, gelatine, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Examples of suitable pharmaceutical rs are described in "Remington's Pharmaceutical Sciences" by E.W. . The formulation should be selected according to the mode of administration.
The amount of the inventive composition can be selected by any skilled person, specifically the amount of the prostacyclins or cyclin analogues or pharmaceutically acceptable salts thereof, specifically the amount of Treprostinil is at least 1.0 ng/kg of body weight. The amount of PDE4 or PDE5 inhibitor can be easily ed by skilled persons. too. Specifically, the amount of PDE4 or PDE5 or PDE 7 or PDE8 inhibitor is about 0.5 mg for Roflumilast or about 30 mg ast, at least once per day, specifically at least two times/day.
The present ion onally provides a method for increasing the cAMP level in a cell wherein said cell is contacted with at least one prostacyclin or prostacyclin analogue and at least one PDE4 and ally at least one of PDE5, PD7 or PDE8 inhibitors. The increase of cAMP in said cells can be at least 10%, preferably at least 25%, preferably at last 50%, more preferred at least 100% ed to single treatment with a prostacyclin or a PDE4 and/or PDE5 or PDE7 or PDE8 inhibitor.
A eutic combination, comprising at least one prostacyclin analogue and at least one PDE4 and optionally at least one PDE5 and/or PDE7 inhibitor and/or PDE8 inhibitor, wherein the prostacyclin analogue and PDE4 and/or PDE5 inhibitor and/or PDE7 inhibitor and/or PDE8 tor are provided in amounts which together are sufficient to treat and/or prevent at least one symptom associated with cystic fibrosis is provided, too. Specifically, an increase of the CAMP level in the epithelial cells of the lung of CF patients can be reached by administering the inventive therapeutic combination preparation. Specifically, at least one of the prostacyclin analogue and PDE4 and optionally one or more of PDE5, PDE7 or PDE8 inhibitors are ated for administration by inhalation.
In a specific embodiment of the present invention, a combination therapy is disclosed for treating cystic fibrosis. According to a ic embodiment, the symptoms associated with reduced CAMP levels in bronchoepithelial cells of patients with CF can be treated or prevented by using the ive combination therapy.
Possibly, one or more additional agents can also be administered.
The prostacyclin or prostacyclin analogue and the PDE4 and optionally the PDE5, PDE7 or PDE8 inhibitor may be administered together, for example in a single tablet or capsule or inhalation formulation or the PDE4 and optionally other PDE inhibitors of the invention as well as al additional agents may be administered separately from the prostacyclin or prostacyclin analogue.
The invention r provides a kit and its use for treating or ting a condition associated with cystic fibrosis in a subject, comprising (i) an effective amount of a prostacyclin or prostacyclin analogue, (ii) a PDE4 inhibitor, specifically Roflumilast, Ro1724 or lbudilast, and optionally one or more compounds ed form the group of PDE5, PDE?’ and PDE8 inhibitors, (iii) one or more pharmaceutically acceptable rs and/or additives, and (iv) instructions for use in treating or preventing cystic fibrosis in a subject, preferably a human.
Said components (i) and (ii) and (iii) can be in a form suitable for intravenous administration, for inhalation or for oral administration.
The examples described herein are rative of the present invention and are not intended to be limitations thereon. Different ments of the present invention have been described according to the present invention. Many modifications and ions may be made to the techniques described and illustrated herein without departing from the spirit and scope of the invention. Accordingly, it should be under- stood that the examples are illustrative only and are not limiting upon the scope of the invention.
Examples Example 1: 183—1 cells were plated on 6 well— plates (O.2*106 cells/well in Fig. 1; 0.4 * 106 cells in Fig. 2) in complete growth medium (LHC—8 + 5% F08). The following day, the adenine nucleotide pool was metabolically labeled by incubation with [3H]adenine (1 uCi/well) in Dulbecco’s Modified Eagle Medium (DMEM) containing adenosine deaminase (1 unit/ml) for 4h. Thereafter the medium was replaced with fresh ; the cells were stimulated by sole addition of Treprostinil (in logarithmically spaced concentrations ranging from 0.1 to 30 uM) or of Treprostinil in ation with the ted concentrations of PDE—inhibitors. After an incubation of 30 min, the cells were lysed by the addition of perchloric acid.
The formation of MP was determined by sequential chromatography on Dowex 50WX—4 and neutral alumina columns followed by liquid scintillation counting of the eluate. The assay was performed in triplicate.
The results are shown in figures 1 and 2. The difference between the m response in fig. 1 and fig. 2 is mainly due to the fact that the number of cells/well in fig. 2 is about twice as high as that employed in fig. 1.
W0 2012/107363 Example 2: lB3-1 cells endogenously s only mutated CFTR-AFSOB, which is retained within the cells. Using appropriated manipulations (e.g., pharmaco— chaperones or low temperature incubations), it is le to ocate the mutant CFTR-AF508 from the endoplasmic reticulum to the ER; when inserted at the cell surface, a CI— tance can be stimulated by elevating CAMP. The resulting Cl- conductance, however, is small. In order to unequivocally prove that the cAMP accumulation induced by Treprostinil ated into an activation of CFTR, we transiently expressed a GFP-tagged version of wild type CFTR (the GFP tag allowed for the identification of cells that sed the protein at the cell surface). As can be seen from Fig. 3, Treprostinil caused a robust activation of the current induced by a depolarization from -40 mV g potential to + 60 mV. The maximum effect was delayed, i.e. it was only obsen/ed several seconds after wash—in of the compound.
Likewise, there was also a hysteresis in the turn-off reaction; the current decayed to basal only ~ 100 s after washout. These delayed responses reflect the (i) intervening signaling cascade (i.e., the receptor-dependent activation of G3, Gas-dependent activation of CAMP formation and the ensuing protein kinase A-dependent phosphorylation of CFTR) and (ii) the delayed deactivation of increased CAMP by phosphodiesterases. r delays were also seen, if cells were stimulated with lin, a direct activator a adenylyl cyclase, which was used as a positive control.
These observations prove that Treprostinil can activate CFTR in bronchial epithelial cells.
Methods: Electrophysio/ogy The whole cel/ patch clamp technique was used for t recordings med at 22 i 15°C using an Axoclamp 2008 patch clamp amplifier (Axon Instruments). Pipettes had resistances n 1 and 2 M9 when filled with the recording pipette solution (composition: 110 mM CsCl, 5 mM EGTA, 2 mM M902, 1 mM K2.ATP, 10 mM Hepes, pH adjusted to 7.2 with CsOH). Voltage-clamp protocols and data acquisition were performed with pclamp 6.0 software (Axon Instruments).
Data were low-pass filtered at 2 kHz (-3 dB) and digitized at 10-20 kHz. Cells were continuously superfused with external solution (composition: 145 mM NaCl, 4.5 mM KCl, 2 mM CECIz, 1 mM MgC|2= 5 mM glucose, 10 mM Hepes: pH adjusted to 7.4 W0 2012/107363 with NaOH). When indicated, the external solution contained Treprostinil (1O uM) or forskolin (5 pri), switching between solutions was achieved by electronically controlled pressure valves.
Based on the results of example 1, a sustained response is expected, if Treprostinil is combined with PDE 4 or 5 inhibitors, e.g., 1O uM Roflumilast or 100 uM lbudilast or 10-100 uM Tadalafil or afil or Vardenafil.
Cell culture: IBB—1 cells were grown on dishes (Nunc, 3.5 cm diameter) covered with fibronectin (1O ug/mL) rat collagen l (30 ug/mL) and BSA 10 ug/mL) in LHC—8 medium (Gibco) containing 5% fetal calf serum (FCS). Cells were transiently transfected with a d driving the expression of human GFP—tagged wild type CFTR by using Lipofectamine plus® (lnvitrogen) according to the ctions of the manufacturer.
Representative current amplitudes recorded in the whole cell patch clamp configuration at + 60 mV. A transiently transfected lB3—1 cell expressing gged wild type CFTR was selected under fluorescent light and clamped to a holding potential at -40 mV. Depolarization was induced by a voltage step to + 60 mV for 50 ms and the current amplitude was recorded. Wash-in of Treprostinil (10 um final concentration, TP) was initiated at the time point 50 s and terminated at 125 s. lin was washed in at 275 s and was removed at 375 8. Results are shown in figure 3.
Example 3 introduction Previous observations indicated that, in human airway epithelial cells, the Treprostinil—induced CAMP accumulation was specifically enhanced by inhibitors of phosphodiesterase~4 (PDE4) ms. als and Methods Cell lines and cell culture: The following human bronchial epithelial cell lines were obtained through ATCC: SEAS-2B (ATCC 09), NuLi—1 (ATCC CRL—4011), IB3-1 (ATCC CRL-2777), CuFi-1 (ATCC 13). Cells were propagated using the culture conditions outlined in the ATCC recommendations, e.g., ISB~1 cells were maintained on dishes W0 2012/107363 2012/051880 coated with fibronectin (10 pg/ml) rat collagen | (30 pg/ml) and BSA 10 pg/mL) in LHC-8 medium (Gibco) ning 5% fetal calf serum (FCS) at 37°C in a 5 % C02 humified here. BEAS-ZB cells were maintained at 37°C in a 5 % CO2 humified atmosphere on dishes precoated with collagen IV (60 pg/ml in 0.25 % acetic acid) in BEGM medium (Lonza); the GA-1000 (gentamycin-amphotericin B mix) provided with the BEGM kit was not added to the medium, The level of endogenous expression of CFTR was too low to obtain a reliable signal. Accordingly, BEAS—ZB cells were transiently transfected with a plasmid driving the expression of human GFP—tagged wild type CFTR by using Lipofectamine plus® (lnvitrogen) according to the instructions of the manufacturer. Cells expressing this gged CFTR were identified by fluorescence microscopy and subjected to patch clamp recordings as outlined below.
CAMP accumulation assay: lBS—1 cells were seeded onto PDL—coated wells of 6—well plates (2 to 25*105 cells/well) in complete growth medium (LHC-8 + 5 % FCS). On the following day, the cellular adenine nucleotide pool was metabolically ed by tion with [3H]adenine (1 pCi/well) in Dulbecco’s Modified Eagle Medium (DMEM) in the presence of adenosine deaminase (5 pg/ml) for 4h. Subsequently, the medium was ed with fresh DMEM and the formation of CAMP was stimulated by addition of 5 uM forskolin, a direct activator of the adenylyl cyclase, or 10 uM treprostinil in the absence and presence of ent concentrations of the following phosphodiesterase (PDE) inhibitors: ast (0.3 - 1000 pM), Ro—20—1 724 (0.03 - 300 pM), roflumilast (1 nM — 10 uM), damole (0.01 - 100 pM), amrinone (1, 10, 100 pM), anagrelide (1, , 100 pM), enoximone (1, 10, 100 pM), milrinone (1, 10, 100 pM) and cilostazol (0.1 to 100 pM) for 20 min at 37°C. In some instances, the effect of these inhibitors on basal CAMP accumulation was examined by incubating cells in the absence of any additional stimulus with increasing concentrations of PDE—inhibitors (i.e., dipyridamole, ibudilast and Ro—20-1724 at 1, 10 and 100 pM; roflumilast at and 0.1, 1 and 10 pM of ilast). Concentration—response curves for treprostinil were obtained by adding treprostinil (0.1 to 30 pM) alone or in combination with 100 pM Ro1724, 100 pM ibudilast or 5 pM roflumilast. The reaction performed in triplicate was stopped by adding 2.5 % perchloric acid together with 01 mM (unlabelled) CAMP. [3H]cAMP was isolated by sequential chromatography on Dowex 5OW—X4 and W0 2012/107363 neutral alumina columns. The formation of MP was quantified by liquid scintillation counting.
Electrophysioiogy — patch clamp recordings: The whole cell patch clamp que was used for current recordings performed at 22 i 15°C using an Axoclamp 2008 patch clamp amplifier (Axon ments).
Pipettes had resistances n 1 and 2 MO when filled with the recording pipette solution (composition: 110 lel CsCl, 5 leI EGTA, 2 lel MgCI2, 1 lel K2.ATP, 10 mM Hepes, pH adjusted to 7.2 with CsOH). Voltage-clamp protocols and data acquisition were med with pclamp 6.0 software (Axon Instruments). Data were ss ed at 2 kHz (~3 dB) and digitized at 10—20 kHz. Cells were continuously superfused with external solution (composition: 145 mM NaCI, 4.5 mM KCl, 2 mM CaClZ, 1 mM MgCl2, 5 mM glucose, 10 mM Hepes, pH adjusted to 7.4 with NaOH).
When indicated, the external on contained Treprostinil (10 pM) or forskolin (5 ulVl), switching between solutions was achieved by onically controlled pressure valves.
Accumufation of CAMP in the lB3—‘i cell line in the absence and presence of phosphodiesterase inhibitors: The survey of oforms predicts that PDE—inhibitors ought to have a pronounced effect on CAMP accumulation in human bronchial epithelial cells. In addition, this analysis provided evidence for the presence of additional isoiorms of phosphodiesterases. Accordingly, the PDE-cir selective inhibitors roflumilast and ibudilast were tested and their effect was compared to that of several additional PDE- inhibitors: RO20—1724, a non—selective PDE-inhibitor with PDE4-preference; dipyridamole, which blocks the equilibrative nucleoside transporter—1 and —2 (ENT1— and ENT2) and, in addition, inhibits PDE5, PDE7A, PDE8A, PDE10A and PDE11 (Soderling et al., 1998; Hetman et at, 2000a&b; Omori & Kotera, 2007). Amrinone, milrinone and cilostazole, which are ive inhibitors of PDE3-isoforms; anagrelide, which inhibits PDE2 and PDE3. lbudilast is less selective than roflumilast and also inhibits PDE10- and PDEtt-isoforms. The approach focused on the regulation of CAMP-levels; hence the cGMP-specific enzymes PDES, PDE6 and PDE9 were not further considered r & Beavo, 2006; Omori & Kotera, 2007).
W0 2012/107363 Cells typically express many isoforms of yl cyclase. In many instances, receptors that are coupled to G5 do not have access to the entire cellular pools of adenylyl cyclases. In contrast, forskolin stimulates all isoforms of adenylyl cyclase. in the absence of phosphodiesterase inhibition, CAMP is rapidly hydrolysed such that it accumulates only to low levels at steady state. tion of odiesterase results in accumulation of CAMP. Roflumilast, ibudilast, dipyridamole and R0204 724 substantially enhanced the cAMP accumulation triggered by 5 uM forskolint Roflumilast was the most potent inhibitor and dipyridamol was a less potent inhibitor.
Ibudilast and R020-1724 were more effective than roflumilast. Taken together these data suggested that PDE4-isoforms contributed to a large extent to the ysis of cAMP. If cAMP accumulation was triggered by treprostinil, the concentration- response curve for all inhibitors were shifted to the left. This leftward shift indicates that cAMP generated via receptor stimulation is more readily accessible to degradation by phosphodiesterases. One possible explanation is the anchoring of phosphodiesterases in the vicinity of the receptors (Francis et al., 2011). The higher efficacy of damole also suggests a possible contribution by PDE8 or PDE10.
The main action of the phosphodiesterase inhibitors is to enhance cAMP accumulation: while Emax (119., the maximum effect increases), the apparent ty of the t (i.e., its ECso) is not shifted.
In the absence of an exogenously added agonist (or of lin), the PDE—inhibitors do not per se cause any appreciable increase in cAMP accumulationThis is to be expected; the basal activity of adenylate cyclase is very low and it requires input via receptor-dependent activation of (3.5 to catalyse the formation of cAMP. However, under cell e conditions — i.e., in defined media — there aren’t any agonists present.
PDE—inhibition es treprostinil-induced Cf currents through CFTR; Because inhibition of PDE4-isoforms enhanced treprostinil-induced cAMP accumulation, this manipulation was predicted to enhance the effect of treprostinil on chloride currents through the cystic fibrosis transmembrane conductance regulator/CF channel (CFTR). This was the case: reprostinil caused as ned activation of CFTR; the resulting outward t can be detected by voltage jumps from -20 to —80 mV. The addition of roflumilast (and of other PDE4 inhibitors such as W0 2012/107363 ibudilast and RO20—1724) caused an additional increase of the current. The current is carried by CFTR, because it is reversibly blocked by the specific inhibitor.
Conclusions 1) Human airway epithelial cells express several receptors that can be targeted by stinil to raise CAMP and thereby activate CFTR in human airway epithelial cells. 2) PDE4-isoforms are present in human airway epithelial cells and nhibitors effectively augment the response to treprostinil.
References Aronoff DM, Peres CM, Serezani CH, ger MN, Carstens JK, Coleman N, Moore BB, Peebles RS, Faccioli LH, Peters-Golden M (2007) Synthetic cyclin analogs differentially regulate macrophage on via distinct analog-receptor binding icities. J Immunol 178216284634.
Bender AT, Beavo JA (2006) Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev 582488 520 Francis SH, Blount MA, Corbin JD (2011) Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions. Physiol Rev 91 :651—690.
Hetman JM, Soderling SH, Glavas NA, Beavo JA. (2000a) Cloning and characterization of PDE7B, a cAM P—specific phosphodiesterase. Proc Natl Acad Sci U S A 97: 472—476 Hetman JM, Robas N, Baxendale R, Fidock M, Phillips SC, Soderling SH, Beavo JA (2000b) Cloning and terization of two splice variants of human phosphodiesterase 11A. Proc Natl Acad Sci U S A 97: 12891—12895 Houslay MD, Schafer P, Zhang KY (2005) Keynote review: phosphodiesterase-4 as a therapeutic target. Drug Discov Today 10:1503-1519 Omori K, Kotera J (2007) Overview of PDEs and their regulation. Circ. Res. 100309- Nikam VS, Wecker G, uly R, Rapp U, Szeiepusa K, Seeger W, Voswinckel R (2011) Treprostinii inhibits adhesion and differentiation of fibrocytes via CAMP and Rap dependent ERK vation. Am J Respir Cell Mol Biol 45: 692-703 W0 2012!107363 Soderling SH, Bayuga SJ, Beavo JA (1998) Cloning and Characterization of a CAMP- specific: cyclic nucleotide odiesterase. Proc Natl Acad Sci U S A 9528991- 8996.
Wright JM, Zeitlin PL, Cebotaru L, o SE, Guggino WB (2004) Gene expression profile analysis of 4—phenylbutyrate treatment of lBS-1 bronchial epithelial cell line demonstrates a major influence on heat-shock proteins. Physiol Genomi05161204—21 1

Claims (20)

Claims
1. Composition sing at least one prostacyclin or prostacyclin ue or a pharmaceutically acceptable salt thereof and at least one phosphodiesterase (PDE) 4 inhibitor for use in preventing or treating cystic is, wherein said prostacyclin analogue is selected from the group of Treprostinil, lloprost, Cicaprost or Beraprost or pharmaceutically acceptable salts thereof.
2. ition according to claim 1, wherein said prostacyclin analogue is selected from the group of acid derivatives of Treprostinil, polymorphs of Treprostinil or isomers of Treprostinil.
3. Composition according to claims 1 or 2, wherein said PDE4 inhibitor is selected from the group of Ro 4, ast, Roflumilast and its e, Cilomilast, BAY 19-8004, CC3, AWD 12—281, SCH 351591, Ciclamilast, Piclamilast, CGH2466, rine, Rolipram, Luteolin and Drotaverine.
4. Composition comprising Treprostinil and a PDE4 inhibitor selected from the group of RO 20-1724, Roflumilast and lbudilast.
5. Composition according to any one of claims 1 to 4, wherein a further PDE inhibitor selected from the group of PDE5, PDE? or PDE8 inhibitors is contained.
6. Composition according to claim 5, wherein said PDE5 inhibitor is selected from Avanafil, Lodenafil, Mirodenafil, Sildenafil citrate, Tadalafil, Vardenafil or Udenafil.
7. Composition according to claim 5, wherein said PDE? and PDE8 inhibitors are selected from Dipyridamol, BRL50481 and 7325.
8. Composition according to any one of claims 1 to 7 which is a pharmaceutical ition.
9. Composition according to any one of claims 1 to 8 which is formulated for inhalation.
10. Composition according to any one of claims 1 to 8 which is formulated for intravenous or subcutaneous administration or formulated as an orally available form ed from the group of sustained release forms, tablets and capsules.
11. Composition according to any one of claims 1 to 10 comprising an effective amount of stinil or a ceutically acceptable salt thereof that is at least 1.0 ng/kg of body .
12. Method for in vitro increasing the CAMP level in a cell, wherein said cell is contacted with at least one prostacyclin or prostacyclin analogue and at least one PDE4 inhibitor and optionally at least one inhibitor selected from the group of PDE5 inhibitors, PDE7 inhibitors or PDE8 tors or a pharmaceutically acceptable salt thereof.
13. A therapeutic combination, comprising at least one prostacyclin or cyclin analogue and at least one PDE4 inhibitor and at least one inhibitor selected from the group of PDE7 inhibitors or PDE8 inhibitors, wherein the prostacyclin or prostacyclin ue and PDE4 inhibitor and at least one inhibitor selected from the group of PDE7 inhibitors and PDE8 tors, and are provided in amounts which together are sufficient to treat and/or prevent at least one symptom associated with cystic fibrosis.
14. Therapeutic combination according to claim 13, wherein the prostacyclin or prostacyclin analogue and PDE4 inhibitor and at least one inhibitor selected from the group of PDE7 inhibitors and PDE8 inhibitors are ated for administration by inhalation.
15. The use, in the manufacture of a medicament for preventing or treating cystic fibrosis, of a composition comprising at least one prostacyclin or prostacyclin ue selected from the group of Treprostinil, lloprost, Cicaprost or Beraprost or a pharmaceutically acceptable salt thereof and at least one phosphodiesterase (PDE) 4 inhibitor.
16. The use of a therapeutic combination sing at least one prostacyclin or prostacyclin analogue and at least one PDE4 inhibitor and at least one inhibitor selected from the group of PDE7 inhibitors or PDE8 inhibitors, in the manufacture of a medicament for treating and/or preventing at least one symptom associated with cystic fibrosis, wherein the cyclin or prostacyclin analogue and PDE4 inhibitor and at least one inhibitor selected from the group of PDE7 inhibitors and PDE8 inhibitors.
17. A composition according to claim 1 or claim 4, substantially as herein described with reference to any one of the examples and/or figures f.
18. A method according to claim 12, substantially as herein described with reference to any one of the examples and/or s thereof.
19. A therapeutic combination according to claim 13, ntially as herein described with reference to any one of the examples and/or figures thereof.
20. A use according to claim 15 or 16, substantially as herein described with reference to any one of the examples and/or figures thereof. W0 2012!]07363
NZ613118A 2011-02-07 2012-02-03 Novel composition for the treatment of cystic fibrosis NZ613118B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP11153541 2011-02-07
EP11153541.5 2011-02-07
PCT/EP2012/051880 WO2012107363A1 (en) 2011-02-07 2012-02-03 Novel composition for the treatment of cystic fibrosis

Publications (2)

Publication Number Publication Date
NZ613118A NZ613118A (en) 2015-06-26
NZ613118B2 true NZ613118B2 (en) 2015-09-29

Family

ID=

Similar Documents

Publication Publication Date Title
EP2672957B1 (en) Novel composition for the treatment of cystic fibrosis
US10086001B2 (en) Composition for the treatment of cystic fibrosis
WO2012107364A1 (en) Novel composition for the treatment of cystic fibrosis
US20120010178A1 (en) Methods and compounds for treatment of neurodegenerative disorders
JP2020518642A (en) Compositions and methods for the prevention and treatment of hearing loss
JP2014505076A (en) Inhibitors of mTOR kinase as antiviral agents
US20220160872A1 (en) Drug Adsorbed Highly Porous Activated Carbon for Enhanced Drug Delivery
US20170216332A1 (en) Small molecule anti-scarring agents
NZ613118B2 (en) Novel composition for the treatment of cystic fibrosis
KR20080004564A (en) Pyrimidylaminobenzamide derivatives for hypereosinophilic syndrome
US20210379061A1 (en) Balipodect for treating or preventing autism spectrum disorders
US20240033236A1 (en) Methods of treating herpes viral infection with 4-phenylbutyrate (pba) or a pharmaceutically acceptable salt thereof
CN117999073A (en) Use of nadolol to treat chronic obstructive pulmonary disease by blocking the inhibitor protein-2 pathway