WO2011141891A1 - Use of cycloundecadepsipeptide compounds - Google Patents

Use of cycloundecadepsipeptide compounds Download PDF

Info

Publication number
WO2011141891A1
WO2011141891A1 PCT/IB2011/052096 IB2011052096W WO2011141891A1 WO 2011141891 A1 WO2011141891 A1 WO 2011141891A1 IB 2011052096 W IB2011052096 W IB 2011052096W WO 2011141891 A1 WO2011141891 A1 WO 2011141891A1
Authority
WO
WIPO (PCT)
Prior art keywords
axx
meleu
leu
meala
mebmt
Prior art date
Application number
PCT/IB2011/052096
Other languages
French (fr)
Inventor
Grégoire VUAGNIAUX
Bruno Gavillet
Rafael CRABBÉ
Original Assignee
Debio Recherche Pharmaceutique S.A.
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 Debio Recherche Pharmaceutique S.A. filed Critical Debio Recherche Pharmaceutique S.A.
Publication of WO2011141891A1 publication Critical patent/WO2011141891A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to cycloundecadepsipeptide compounds and analogues presenting mitochondrial permeability transition pore (MPTP) inhibitory activity and reduced cytotoxicity.
  • the present invention further relates to the pharmaceutical compositions containing cycloundecadepsipeptide compounds and analogues for use in the treatment and/or prevention of cell death associated disorders and diseases, related to the MPTP opening.
  • MPTP mitochondrial permeability transition pore
  • Oxidative stress accompanied by calcium overload, ATP depletion, and elevated phosphate levels induces mitochondrial permeability transition (MPT) with formation of nonspecific MPT pores (MPTP) in the inner mitochondrial membrane.
  • MPT mitochondrial permeability transition
  • MPTP nonspecific MPT pores
  • MPT results from opening of mitochondrial permeability transition pores (MPT pores or MPTP) (Halestrap 2010).
  • MPT induction is a key event in cell death and is implicated in a variety of disorders and diseases. For example:
  • MPT also appears to play a key role in damage caused by ischemia, as occurs in a heart attack and stroke (Baines, Kaiser et al. 2005; Nakagawa, Shimizu et al. 2005; Schinzel, Takeuchi et al. 2005; Halestrap 2010). • Research has shown that the MPT pore remains closed during ischemia, but opens once the tissues are reperfused with blood after the ischemic period, playing a role in reperfusion injury (Lim, Hausenloy et al. 2010).
  • MPT is also thought to underlie the cell death induced by Reye's syndrome, since chemicals that can cause the syndrome, like salicylate and valproate, cause
  • MPT may also play a role in mitochondrial autophagy (Lemasters, Nieminen et al. 1998).
  • Neurodegeneration a process that results in damage and death of neurons, is also a disorder where MPTP is found to be involved (Barrientos, Martinez et al. ; Du and Yan ; Du, Guo et al. 2008; Ibarra and Martinon 2009; Martin, Gertz et al. 2009).
  • MPTP plays an important role in muscular dystrophies (Palma, E., T. Tiepolo, et al. 2009; Millay, D. P., M. A. Sargent, et al. 2008
  • MPTP is involved in cell death induced by metabolic stress and/or oxidative stress and/or loss of cellular calcium homeostasis and/or aging cells and/or toxic agents (pathogens) and/or infectious agents (pathogens) (Burke et al. 2007; King et al. 2008; Sokol et al. 2005; Plotnikov et al. 2009).
  • MPTP is accepted as a therapeutic target for both pharmacological and conditional strategies to block pore formation by direct interaction with MPTP components or indirectly by decreasing MPTP inducers (Zorov, Juhaszova et al. 2009; Giorgio, Soriano et al. 2010).
  • sanglifehrin A as well as attenuation of reactive oxygen species accumulation through mitochondria-targeted antioxidants, is the most promising.
  • Inhibiting the opening of the MPTP can treat and/or prevent cell death associated disorders and diseases.
  • the MPTP opening has an important role in triggering organ dysfunction and pathology.
  • Cell death associated disorders and diseases, wherein the MPTP opening is involved include but are not limited to, ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
  • Ischaemia and ischaemia-reperfusion injury the opening of the MPTP may be induced by ischaemia-reperfusion and can play an important role in the following diseases or conditions:
  • cell death associated diseases or disorders relate to the MPTP opening, involve organ dysfunction or pathologies in which the opening of the MPTP may be also an important trigger for cell death and is induced by metabolic stress and/or oxidative stress and/or loss of cellular calcium homeostasis and/or aging cells and/or toxic agents (pathogens) and/or infectious agents (pathogens) including but not limited to:
  • muscular dysthrophies including but not limited to Ulrich congenital
  • ⁇ neurodegenerative diseases including but not limited to: amyothrophic lateral sclerosis (ALS) (Martin, Gertz et al. 2009; Martin 2010), Alzheimer disease (Du and Yan 2010), Parkinson disease , multiple sclerosis (Forte, Gold et al. 2007), Huntington disease (Brustovetsky, Houseovetsky et al. 2003)
  • Agents that block MPT include the immune suppressant cyclosporin A (CsA) N-methyl-Val-4-cyclosporin A (MeVal-4-CsA), a non-immunosuppressant derivative of CsA; another non-immunosuppressive agent, NIM81 1 , 2- aminoethoxydiphenyl borate (2-APB), and bongkrekic acid.
  • CsA immune suppressant cyclosporin A
  • MeVal-4-CsA N-methyl-Val-4-cyclosporin A
  • NIM81 1 another non-immunosuppressive agent
  • 2-APB 2- aminoethoxydiphenyl borate
  • bongkrekic acid bongkrekic acid.
  • DEBIOPHARM SA relates to the use of a cyclic undecapeptide for the preparation of a medicament for administration during a myocardial ischaemic event.
  • WO 2009/098533 (DEBIOPHARM SA) relates to the use of a non-immunosuppressive cyclosporin A derivative for preventing or reducing muscular degeneration in a subject suffering from Limb- Girdle muscular Dystrophy.
  • Naturally occurring cyclosporins and derivatives present MPTP inhibitory activity but simultaneously display cytotoxicity which limits their anti-cell death effect.
  • the aim of the present invention is to provide new series of compounds with reduced cytotoxicity resulting in enhanced anti-cell death activity.
  • the present invention provides a cydoundecadepsipeptide compound of Formula (I) Cyclo-(AXXi-AXX 2 -AXX3-AXX 4 -AXX5-AXX6-AXX 7 -D-Hiv-MeLeu-Leu-lvleVal) (I)
  • AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
  • AXX 2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH 2 CH 2 CH 2 OH), Nva, 5-hydroxy-Nva (Hnv);
  • AXX 3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
  • AXX 4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe, EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone;
  • AXX 5 is Leu, Val, lie, Gly, Abu;
  • AXX 6 is MeAla, Sar, MeLeu;
  • AXX 7 is Gly, Ala; for use in a method for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening,
  • the present invention further provides the use of a pharmaceutical composition for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening, comprising a compound of the invention, together with one or more pharmaceutically acceptable diluents or carriers.
  • the term “comprise” is generally used in the sense of include, that is to say permitting the presence of one or more features or components.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder or the disease as well as those in which the disorder is to be prevented. Hence, the mammal to be treated herein may have been diagnosed as having the disorder or the disease may be predisposed or susceptible to the disorder or the disease.
  • “Mammal” for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals or pet animals, such as dogs, horses, cats, cows, monkeys etc. Preferably, the mammal is human.
  • the term "therapeutically effective amount” refers to an amount of a drug effective to treat a disease or disorder in a mammal.
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • cell death associated disorders and diseases comprises disorders and diseases caused by, associated with, linked with, mediated by or accompanied by the MPTP opening or wherein the MPTP opening is involved.
  • Cell death encompasses the normal degeneration and death of living cells, also defined as the terminal failure of a cell to maintain essential life functions, or the point in the process of dying at which vital functions have ceased at the cellular level.
  • Cell death includes for example apoptosis as well as necrosis often defined as the morphological changes indicative of cell death caused by progressive enzymatic degradation; it may affect groups of cells or part of a structure or an organ.
  • a "disorder” refers to an underlying pathological disturbance in a symptomatic or asymptomatic organism relative to a normal organism, which may result, for example, from infection, stress or an acquired or congenital genetic imperfection.
  • the invention refers to a series of cycloundecadepsipeptide compounds, with or without immunosuppressant activity, presenting mitochondrial permeability transition pore (MPTP) inhibitory activity and reduced cytotoxicity.
  • MPTP mitochondrial permeability transition pore
  • AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
  • AXX 2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH 2 CH 2 CH 2 OH), Nva, 5-hydroxy-Nva;
  • AXX 3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
  • AXX 4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe,
  • EtTyr EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone;
  • AXX 5 is Leu, Val, lie, Gly, Abu;
  • AXX 6 is MeAla, Sar, MeLeu;
  • AXX 7 is Gly, Ala; for use in a method for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening, and wherein said
  • cycloundecadepsipeptide compound of Formula (I) comply with the two following criteria: (1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as:
  • said cycloundecadepsipeptide compound of Formula (I) is at least 5 fold less cytotoxic than CsA, and even more preferably at least ten-fold less cytotoxic than CsA, defined as:
  • CsA _ cell _ count _ IC 50 Calcium retention capacity is a sensitive measure of the propensity of mitochondria to open the Permeability Transition Pore (MPTP) after Ca 2+ uptake.
  • the IC2 (nM) is the concentration of compound necessary to double the CRC, relative to the CRC observed in the absence of any compound (CRCo).
  • cyclodepsipeptides are natural or synthetic compounds having sequences of amino and hydroxyl carboxylic acid residues (usually a-amino and a-hydroxy acids) and the residues are connected in a ring (see lUPAC Compendium of Chemical Terminology, 2 nd Edition (1997). Such cyclodepsipeptides are depicted as heterodetic peptides in which at least one amide bond has been replaced with an ester bond.
  • the compounds of the present invention comprise eleven residues, ten being a- amino acids and one being ⁇ -hydroxy acid. This ⁇ -hydroxy acid is
  • (2R)-2-hydroxy-3-methyl-butanoic acid also known as D-a-hydroxyisovaleric acid and abbreviated as H-D-Hiv-OH.
  • this hydroxyl acid is in position 8. It forms on the carboxylic acid end an amide bond with the amino group of the ⁇ -amino acid in position 9, namely N-methyl-leucine, and, on the hydroxyl end an ester bond with the carboxylic acid group of the a-amino acid in position 7, namely alanine or glycine.
  • the a-amino acids of Formula (I) are mentioned using the three letter code abbreviation usually used to name amino acids and their configuration is
  • An advantage of the compounds according to the present invention may lie in the impact of an ester bond within macro cyclic backbone in comparison to the regular cyclic amide backbone of the corresponding cycloundecapeptide compounds described in the prior art. Without being bound to theory, it is believed that the replacement of an amide by an ester bond between amino-acid AXX 7 in position 7 and D-HIV in position 8 results in a strong effect upon the conformational and physico-chemical properties such as increased conformational flexibility and lipophilicity as well as the absence of a hydrogen donor bond.
  • the new class of compounds also offers an improved preparation process, especially at an industrial scale.
  • the invention also encompasses chemical modifications of the compounds of formula I to prolong their circulating lifetimes.
  • suitable poly(ethylene glycol) derivatives that possess this property are described in e.g. US 2005171328 (NEKTAR THERAPEUTICS AL CORP) or US 6,713,454 (NOBEX CORP).
  • the compounds of the present invention are defined by
  • AXXi is MeBmt
  • AXX 2 is Abu. Val
  • AXX 3 is D-MeAla
  • AXX 4 is Melle, MeVal, EtVal;
  • AXX 5 is Leu, Val, lie, Gly, Abu;
  • AXX 6 is MeAla, Sar, MeLeu;
  • AXX 7 is Gly, Ala.
  • compounds of Formula (I) are selected among:
  • CsA cyclosporine A
  • the compounds of the present invention have reduced cytotoxicity, which allows (1 ) possible use of higher doses to achieve a maximal effect; (2) increased therapeutic window and (3) no lack of activity at high doses.
  • MPTP opening is a key event in the biochemical signaling that underlies mitochondria mediated cell death in various tissues, disorders and diseases.
  • the cell death associated disorders and diseases are selected from the group comprising ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
  • the cell death associated disorders and diseases, related to the MPTP opening are selected from the group comprising ischaemia and ischaemia- reperfusion injuries, muscular dysthrophies, neurodegeneration disorders and diseases, West Nile viral encephalitis, Japanese encephalitis, Chronic hepatitis and cirrhosis induced by Hepatitis C virus, Hepatitis B, delta agent, acute and chronic drug- or toxin-induced hepatotoxicity, alcoholic liver disease, cholestasis, Non Alcoholic Fatty Liver Disease, gastro-intestinal ulcerations caused by
  • NSAIDs transplant rejection, peripheral vascular insufficiency, renal insufficiency, obesity, diabetic micro-angiopathy, diabetic nephropathy, Maturity onset diabetes of the young type 4, acute and chronic pancreatitis, septicaemia, multi organ dysfunction syndrome, crush syndrome, congestive heart failure, atherosclerosis, hypertensive heart disease, cancer, bipolar disorder.
  • Ischaemia and ischaemia-reperfusion injuries are selected from the group comprising acute cerebral ischemia, acute stroke, acute myocardial infarction cardiac arrest, myocardial stunning, post-reperfusion arrhythmias, organ failure following therapeutic procedures such as thrombolytic therapy, coronary angioplasty, aortic cross-clamping or coronary bypass surgery, hypovolemic shock, storage/reperfusion injury of transplant organs, multi organ dysfunction syndrome.
  • Muscular dysthrophies are selected from the group comprising Ulrich congenital muscular dysthrophy, Bethlem myopathy, myosclerosis, limb girdle muscular dysthropy, Duchenne muscular dysthrophy, Becker muscular
  • Neurodegeneration disorders and diseases are selected from the group comprising traumatic brain injury, spinal cord injury, peripheral nerve injury, epilepsy-induced brain injury, amyothrophic lateral sclerosis (ALS), Alzheimer disease, Parkinson disease , multiple sclerosis, Huntington disease, axonal degeneration-induced neuropathic pain, diabetic neuropathy chemotherapy- induced neuropathic pain, or herpes-induced neuropathy.
  • the cell death associated disorders and diseases, related to the MPTP opening are selected among acute and chronic pancreatitis and muscular dystrophy Pancreatitis is an inflammation of the pancreas that can occur in two very different forms.
  • Acute pancreatitis is sudden while chronic pancreatitis is characterized by recurring or persistent abdominal pain with or without steatorrhea or diabetes mellitus.
  • Acute pancreatitis is a severe and frequently lethal disorder most commonly caused by gallstone disease and alcohol abuse, whereas chronic pancreatitis is a long-standing inflammation of the pancreas that alters its normal structure and functions.
  • Acute pancreatitis is characterized by acinar cell necrosis which contributes to pancreatic and extra-pancreatic autodigestion as well as systemic inflammatory response and multiple organ failure.
  • a key event in acinar cell necrosis is the formation of the MPTP within the inner mitochondrial membrane leading to permeabilization of the mitochondrial membrane.
  • the compounds have the ability to prevent acinar cell necrosis and attenuate the pancreatic injury.
  • Muscular dystrophies comprise a diverse group of genetic disorders that lead to muscle wasting and, in many instances, premature death. It was demonstrated (Millay et al., 2008) that mitochondrial-dependent necrosis represents a prominent disease mechanism in muscular dystrophy. Indeed one major mechanism leading to cellular necrosis is mitochondrial calcium overload, which secondarily enhances reactive oxygen species (ROS) generation and further promotes MPT
  • ROS reactive oxygen species
  • the compounds of the present invention may be obtained by applying classical peptide (solution or solid-phase peptide synthesis; in Houben - Weyl, Methods of Organic Chemistry, Vol. E 22d, Ed. -in-Chief: M. Goodman, Thieme Verlag, Stuttgart, 2003) and organic chemistry or biotechnology, for instance by applying the chemical tools as described by Wenger in Helv. Chim. Acta, 67, 502-25, 1984 or in Helv. Chim. Acta, 66, 2672-702 (1983) and by employing HATU as coupling reagent described in Rich, D.H. et al.. Comparative studies of the coupling of N- methylated, sterically hindered amino acids during SPPS, Tetr. Letters. 35, 5981 - 5984 (1994).
  • Fragment (Ax) may be as follows:
  • Fragment A which contains the a-hydroxy acid residue D-Hiv, may be obtained by degradation of a natural cydoundecadepsipeptide (namely Cyclo-(MeBmt-Thr-Sar- MeLeu-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal), the preparation of said cydoundecadepsipeptide being described either in US 5,1 16,816, Example 2, or in WO 02/092033, Example 4, Step 4-1 .
  • a natural cydoundecadepsipeptide namely Cyclo-(MeBmt-Thr-Sar- MeLeu-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal
  • ALISPORIVIR [D-MeAla] 3 -[EtVal] 4 -CsA as described in WO 2006/038088, DEBIOPHARM SA).
  • the synthesis of the expensive dipeptide derivative Boc-D-MeAla-EtVal-OH (including 4 chemical steps) is not needed.
  • some of the compounds contain a C-terminal Glycine, which facilitates the last step of the synthesis (macrolactonisation, no epimerisation).
  • the costs for reagents and starting compounds are considerably lower in case of the compounds according to the invention.
  • the "reagent of Meerwein" used for the ring opening reaction of CsA is not needed.
  • Fragment B (hexapeptide AXX 2 -AXX 7 ) can be efficiently obtained by standard solid-phase peptide synthesis using commercially starting compounds.
  • the overall yield for obtaining the pharmacologically interesting compounds can amount up to 80%.
  • the present invention further provides a pharmaceutical composition for treating and/or preventing cell death associated disorders or diseases, related to the MPTP opening, comprising a compound of the invention, together with one or more pharmaceutically acceptable diluents or carriers.
  • the compounds of the present invention may be administrated for instance parenterally or orally to a patient in need, for instance incorporated in a pre- concentrated microemultion.
  • the compounds of the present invention, their pharmaceutically acceptable salts and pro-drugs thereof, where applicable, may be administered in the form of a pharmaceutical composition in which they are in association with a
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, as hereinbefore defined, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • a pharmaceutically acceptable adjuvant diluent or carrier.
  • the compounds of the present invention may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi permeable matrices of solid hydrophobic polymers containing the compounds of the present invention, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained- release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl- methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • compositions of the invention may be formulated as creams, gels, solutions, ointments, suspensions or plasters etc. when intended for topical administration; for administration by inhalation, e.g. as aerosols or dry powders; for oral administration, e.g. in the form of tablets, capsules, gels, syrups, suspensions, solutions, powders or granules; for rectal or vaginal administration e.g. as suppositories; or for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular, intrathecal or infusion) as a sterile solution, suspension or emulsion.
  • parenteral injection including intravenous, subcutaneous, intramuscular, intravascular, intrathecal or infusion
  • the active compound of the invention may be administered by any conventional route. It may be administered parentally, e.g., in the form of injectable solutions or suspensions, or in the form of injectable deposit formulations. Preferably, it will be administered orally in the form of solutions or suspensions for drinking, tablets or capsules.
  • the pharmaceutical compositions of the invention typically comprise a cycloundecadepsipeptide compound of the invention and one or more
  • compositions for parenteral administration typically also include one or more excipients.
  • excipients include an isotonic agent, a buffer or other pH- controlling agent, and a preservative. These excipients may be added for maintenance of the composition and for the
  • the indicated dosage of a cycloundecadepsipeptide compound of the invention for daily to trice weekly administration may be from about 1 mg/kg to about 100 mg/kg, preferably from about 1 mg/kg to about 20 mg/kg.
  • the indicated corresponding dosage may be from about 1 mg/kg to about 50 mg/kg, preferably from about 1 mg/kg to about 25 mg/kg.
  • An effective amount of a cydoundecadepsipeptide compound of the invention is understood to be an amount that when administered in the course of a therapeutic regimen to a patient in need of treatment of MPTP opening mediated disorder results in an objective clinical response.
  • cydoundecadepsipeptide compound of the invention Additional factors include the weight of the patient, the age of the patient, and the general condition of the patient. A course of treatment will require repeated administration of a
  • composition of the invention Typically, an adequate drug dose will be administered in one single administration (i.e. a unique dose) or repeatedly for example 3-7 times per week, and the duration of treatment may be for example from about 4 weeks to 6 months, preferably from about 4 weeks to about 12 months.
  • a pharmaceutical composition of the present invention may comprise one or more other active ingredients which present MPTP inhibitory activity in addition to the compounds of the present invention. Compounds of the invention and such other active ingredients can be administered together as part of the same
  • the appropriate dose regimen, the amount of each dose administered, and specific intervals between doses of each active agent will depend upon the specific combination of active agents employed, the condition of the patient being treated, and other factors discussed in the previous section. Such additional active ingredients will generally be administered in amounts less than or equal to those for which they are effective as single therapeutic agents.
  • the FDA approved dosages for such active agents that have received FDA approval for administration to humans are publicly available.
  • the compounds of the invention may be administered as the sole ingredient or together with other drugs.
  • AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
  • AXX 2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH 2 CH 2 CH 2 OH), Nva, 5-hydroxy-Nva;
  • AXX 3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
  • AXX 4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe, EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone;
  • AXX 5 is Leu, Val, lie, Gly, Abu;
  • AXX 6 is MeAla, Sar, MeLeu;
  • AXX 7 is Gly, Ala; and wherein said cycloundecadepsipeptide compound of Formula (I) comply with the two following criteria:
  • said cycloundecadepsipeptide compound of Formula (I) is at least 5 fold less cytotoxic than CsA, and even more preferably at least ten-fold less cytotoxic than CsA, defined as:
  • the cell death associated disorders and diseases, wherein the MPTP opening is involved are selected from the group comprising ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
  • the cell death associated disorders and diseases, related to the MPTP opening are selected from the group comprising ischaemia and ischaemia- reperfusion injuries, muscular dysthrophies, neurodegeneration disorders and diseases, West Nile viral encephalitis, Japanese encephalitis, Chronic hepatitis and cirrhosis induced by Hepatitis C virus, Hepatitis B, delta agent, acute and chronic drug- or toxin-induced hepatotoxicity, alcoholic liver disease, cholestasis, Non Alcoholic Fatty Liver Disease, gastro-intestinal ulcerations caused by
  • NSAIDs transplant rejection, peripheral vascular insufficiency, renal insufficiency, obesity, diabetic micro-angiopathy, diabetic nephropathy, Maturity onset diabetes of the young type 4, acute and chronic pancreatitis, septicaemia, multi organ dysfunction syndrome, crush syndrome, congestive heart failure, atherosclerosis, hypertensive heart disease, cancer, bipolar disorder.
  • Ischaemia and ischaemia-reperfusion injuries are selected from the group comprising acute cerebral ischemia, acute stroke, acute myocardial infarction cardiac arrest, myocardial stunning, post-reperfusion arrhythmias, organ failure following therapeutic procedures such as thrombolytic therapy, coronary angioplasty, aortic cross-clamping or coronary bypass surgery, hypovolemic shock, storage/reperfusion injury of transplant organs, multi organ dysfunction syndrome.
  • Muscular dysthrophies are selected from the group comprising Ulrich congenital muscular dysthrophy, Bethlem myopathy, myosclerosis, limb girdle muscular dysthropy, Duchenne muscular dysthrophy, Becker muscular
  • Neurodegeneration disorders and diseases are selected from the group comprising traumatic brain injury, spinal cord injury, peripheral nerve injury, epilepsy-induced brain injury, amyothrophic lateral sclerosis (ALS), Alzheimer disease, Parkinson disease , multiple sclerosis, Huntington disease, axonal degeneration-induced neuropathic pain, diabetic neuropathy chemotherapy- induced neuropathic pain, or herpes-induced neuropathy.
  • traumatic brain injury spinal cord injury, peripheral nerve injury
  • epilepsy-induced brain injury amyothrophic lateral sclerosis (ALS), Alzheimer disease, Parkinson disease , multiple sclerosis, Huntington disease, axonal degeneration-induced neuropathic pain, diabetic neuropathy chemotherapy- induced neuropathic pain, or herpes-induced neuropathy.
  • ALS amyothrophic lateral sclerosis
  • Alzheimer disease Parkinson disease
  • multiple sclerosis Huntington disease
  • axonal degeneration-induced neuropathic pain diabetic neuropathy chemotherapy- induced neuropathic pain
  • the cell death associated disorders and diseases, related to the MPTP opening are selected among acute and chronic pancreatitis and muscular dystrophy
  • Another object of the invention is a method, comprising co-administration concomitantly or in sequence of a therapeutically effective amount of the compound according to the invention or the pharmaceutical composition of the invention and a co-agent selected from an agent having blocking properties on MPTP opening.
  • a pharmaceutical combination of the invention results in a beneficial effect, e. g. a synergistic therapeutic effect, compared to a monotherapy applying only one of its pharmaceutical active ingredients.
  • a synergistic combination is for example a combination of the
  • reaction mixture was diluted with DCM (60 mL) and washed with H 2 O (1 x 1 0 mL) and brine (1 x 1 0 mL).
  • the organic phase was filtered, dried (Na 2 SO 4 ), evaporated and dried under high vacuum.
  • the crude amine salt of H-MeLeu-Ala-OMe was used for the next coupling step without further purification.
  • Boc-D-MeAla-EtVal-OH was obtained starting from Boc-D-MeAla-Val-OMe using BuLi and Triethyloxoniumfluoroborate, followed by hydrolysis of the methylester according to literature [Jean Frangois. Guichou, PhD thesis entitled “De technical analogues de Cyclosporine A comme agent anti-VIH-1 ", Faculte des Sciences, Universite de Lausanne, 2001 , p.1 21 -1 22] . 2.6. Preparation of Boc-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe
  • the crude mixture was separated by CC to give the pentapeptide Boc-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as an oil.
  • the sample was repurified by semi-preparative RP-HPLC and the product was lyophilized to give the
  • D-MeAla-EtVal-Leu-MeLeu-Ala-OMe was used to the next coupling step without further purification.
  • a sample of crude product (23 mg) was re-purified by semi- preparative RP-HPLC and the peptide was lyophilized to give Fragment (Ba) H-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as a white powder.
  • the MPTP activity was evaluated by measuring the Calcium Retention Capacity (CRC) of mouse liver mitochondrial preparations.
  • CRC Calcium Retention Capacity
  • MPC Permeability Transition Pore
  • CRC of mitochondria was assessed fluorimetrically using the Ca2+ indicator Calcium Green-5N. Mitochondria was incubated 1 min with ranging concentrations of the test compound and the concentration of compound necessary to double the CRC (IC 2 ) was determined by constructing dose-response curves.
  • Mitochondria were isolated from the liver of 9-10 week old male C57BI/6j mice. After isolation the liver was kept at 4°C in a Tris-buffered (pH 7.4) sucrose-based isotonic solution and quickly minced and homogenized with a Teflon/glass potter. The suspension was spun for 6 min at 700 x g, and the resulting supernatant was spun for 6 min at 7,000 x g. The mitochondrial pellet was carefully resuspended in excess volume of sucrose buffer and spun again as above. The final mitochondrial pellet was resuspended with a minimal volume of sucrose buffer. Protein concentration was determined with the biuret method. Mitochondrial suspensions were used immediately and not beyond 4 h from isolation.
  • the CRC of mitochondrial preparations was assessed fluorimetrically in the presence of the Ca 2+ indicator Calcium Green-5N at room temperature using a Fluoroskan Ascent FL fluorometer (Thermo Electron Corporation).
  • Every treatment was represented by the mean value ⁇ SD of (1 ) the CRC and (2) the ratio between CRC (as just defined) and CRCo (concentration of Ca 2+ which opens the PTP in the absence of the compound).
  • IC2 (nM) was calculated as the concentration of compound necessary to double the CRC, relative to the CRC observed in the absence of any compound (CRCo).
  • Cytotoxicity was evaluated by measuring proliferation of HepG2 cells. Cells were incubated with ranging concentrations of the test compound for 72hrs. Cell proliferation is measured by the signal intensity of the incorporated nuclear dye. The half maximal inhibitory concentration (IC 5 0) was determined by constructing dose-response curves.
  • Cells were grown in RPMI 1640, 10%FBS, 2 mM L-alanyl-L-Glutamine, 1 mM Na Pyruvate or a special medium in a humidified atmosphere of 5% CO2 at 37°C. Cells were seeded into 384-well plates and incubated in a humidified atmosphere of 5% CO2 at 37°C. Compounds were added 24 hours post cell seeding. At the same time, a time zero untreated cell plate was generated.
  • IC 5 o is the test compound concentration that produces 50% of the cell proliferation inhibitory response or 50% cytotoxicity level.
  • AIM MathlQ based software
  • Table 1 shows that the selected compounds according to the invention have a MPTP inhibitory activity at least equal to half of the CsA activity as well as at least equal to half of the activity of the compounds of the prior art.
  • the selected compounds of the invention are at least three-fold less cytotoxic than CsA as well as least three-fold less cytotoxic than the compounds of the prior art.
  • Example 3 Acute pancreatitis Models The compounds have been shown to significantly ameliorate disease outcome in the bile acid-induced murine acute pancreatitis model which closely reproduce clinical pathologies.
  • the bile acid-induced murine acute pancreatitis model is similar to gallstone induced clinical acute pancreatitis, which results from retrograde passage of bile into the pancreatic duct when gallstones impact at the sphincter of Oddi.
  • Gallstone pancreatitis is the commonest form of the disease within the majority of countries in the world, except those where alcohol consumption is relatively very high.
  • Biliary pancreatitis was induced by retrograde perfusion of the pancreatic duct with 2 mM TLCS; sacrifice and assessments were made 24 hours later (Laukkarinen, J.M. et al, 2007).
  • the bile acid-induced murine acute pancreatitis model is a close representative of clinical acute pancreatitis, underlining the possible clinical utility of an early administration of compound 057 as a treatment for this disease.
  • OATP1 B1 organic anion transporting polypeptide 1 B1
  • Hepatitis C virus protein expression causes calcium-mediated mitochondrial bioenergetic dysfunction and nitro-oxidative stress.”

Abstract

The present invention relates to cycloundecadepsipeptide compound analogues presenting mitochondrial permeability transition pore (MPTP) inhibitory activity and reduced cytotoxicity. The present invention further relates to a pharmaceutical composition containing cycloundecadepsipeptide compound and analogues for use in the treatment or prevention of cell death associated disorders or diseases, related to the MPTP opening.

Description

Use of cycloundecadepsipeptide compounds
Field of the invention
The present invention relates to cycloundecadepsipeptide compounds and analogues presenting mitochondrial permeability transition pore (MPTP) inhibitory activity and reduced cytotoxicity. The present invention further relates to the pharmaceutical compositions containing cycloundecadepsipeptide compounds and analogues for use in the treatment and/or prevention of cell death associated disorders and diseases, related to the MPTP opening.
Background of the invention
In addition to their central role in ATP synthesis, mitochondria play a critical role in cell death. Oxidative stress accompanied by calcium overload, ATP depletion, and elevated phosphate levels induces mitochondrial permeability transition (MPT) with formation of nonspecific MPT pores (MPTP) in the inner mitochondrial membrane. Mitochondrial permeability transition (MPT) is an increase in the permeability of the mitochondrial membranes to molecules of less than 1500 Daltons in molecular weight. MPT results from opening of mitochondrial permeability transition pores (MPT pores or MPTP) (Halestrap 2010). MPT induction is a key event in cell death and is implicated in a variety of disorders and diseases. For example:
• It is involved in cell death in excitotoxicity (Abramov and Duchen 2008; Malouitre, Dube et al. 2009), in which overactivation of glutamate receptors causes excessive calcium entry into the cell.
· MPT also appears to play a key role in damage caused by ischemia, as occurs in a heart attack and stroke (Baines, Kaiser et al. 2005; Nakagawa, Shimizu et al. 2005; Schinzel, Takeuchi et al. 2005; Halestrap 2010). • Research has shown that the MPT pore remains closed during ischemia, but opens once the tissues are reperfused with blood after the ischemic period, playing a role in reperfusion injury (Lim, Hausenloy et al. 2010).
• MPT is also thought to underlie the cell death induced by Reye's syndrome, since chemicals that can cause the syndrome, like salicylate and valproate, cause
MPT (Trost and Lemasters 1996).
• MPT may also play a role in mitochondrial autophagy (Lemasters, Nieminen et al. 1998).
• Cells exposed to toxic amounts of Ca2+ ionophores also undergo MPT and death by necrosis (Tazawa, Fujita et al. 2009).
• Neurodegeneration, a process that results in damage and death of neurons, is also a disorder where MPTP is found to be involved (Barrientos, Martinez et al. ; Du and Yan ; Du, Guo et al. 2008; Ibarra and Martinon 2009; Martin, Gertz et al. 2009).
· It has been shown that MPTP plays an important role in muscular dystrophies (Palma, E., T. Tiepolo, et al. 2009; Millay, D. P., M. A. Sargent, et al. 2008
• MPTP is involved in cell death induced by metabolic stress and/or oxidative stress and/or loss of cellular calcium homeostasis and/or aging cells and/or toxic agents (pathogens) and/or infectious agents (pathogens) (Burke et al. 2007; King et al. 2008; Sokol et al. 2005; Plotnikov et al. 2009).
Accordingly, MPTP is accepted as a therapeutic target for both pharmacological and conditional strategies to block pore formation by direct interaction with MPTP components or indirectly by decreasing MPTP inducers (Zorov, Juhaszova et al. 2009; Giorgio, Soriano et al. 2010). Inhibition of MPTP opening by reduction of CyP-D activity by nonimmunosuppressive analogs of cyclosporine A or
sanglifehrin A, as well as attenuation of reactive oxygen species accumulation through mitochondria-targeted antioxidants, is the most promising.
Inhibiting the opening of the MPTP can treat and/or prevent cell death associated disorders and diseases. As documented above, it is known that the MPTP opening has an important role in triggering organ dysfunction and pathology. Cell death associated disorders and diseases, wherein the MPTP opening is involved, include but are not limited to, ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
Ischaemia and ischaemia-reperfusion injury: the opening of the MPTP may be induced by ischaemia-reperfusion and can play an important role in the following diseases or conditions:
• acute cerebral ischemia (Schinzel, Takeuchi et al. 2005), acute stroke
(Halestrap 2010),
• acute myocardial infarction cardiac arrest, myocardial stunning, post- reperfusion arrhythmias, organ failure following therapeutic procedures such as thrombolytic therapy, coronary angioplasty, aortic cross-clamping or coronary bypass surgery (Baines, Kaiser et al. 2005; Nakagawa, Shimizu et al . 2005; Halestrap 2010)
• hypovolemic shock, storage/reperfusion injury of transplant organs (Raisky, Gomez et al. 2004), multi organ dysfunction syndrome.
Other cell death associated diseases or disorders, related to the MPTP opening, involve organ dysfunction or pathologies in which the opening of the MPTP may be also an important trigger for cell death and is induced by metabolic stress and/or oxidative stress and/or loss of cellular calcium homeostasis and/or aging cells and/or toxic agents (pathogens) and/or infectious agents (pathogens) including but not limited to:
Muscular dvsthrophies
• muscular dysthrophies, including but not limited to Ulrich congenital
muscular dysthrophy, Bethlem myopathy, myosclerosis, limb girdle muscular dysthropy, Duchenne muscular dysthrophy, Becker muscular dysthrophy, Emery-Dreifuss syndrome (Angelin, Tiepolo et al. 2007; Millay,
Sargent et al. 2008; Palma, Tiepolo et al. 2009; Tiepolo, Angelin et al. 2009; Wissing, Millay et al. 2010), Neurodegeneration:
• traumatic brain injury (Mazzeo, Beat et al. 2009), spinal cord injury
(Ravikumar, McEwen et al. 2007), or peripheral nerve injury (Barrientos, Martinez et al.), epilepsy-induced brain injury
· neurodegenerative diseases including but not limited to: amyothrophic lateral sclerosis (ALS) (Martin, Gertz et al. 2009; Martin 2010), Alzheimer disease (Du and Yan 2010), Parkinson disease , multiple sclerosis (Forte, Gold et al. 2007), Huntington disease (Brustovetsky, Brustovetsky et al. 2003)
· axonal degeneration-induced neuropathic pain, diabetic neuropathy
chemotherapy-induced neuropathic pain, or herpes-induced neuropathy (Barrientos, Martinez et al.)
Cell death induced by metabolic stress and/or oxidative stress and/or loss of cellular calcium homeostasis and/or age related cellular degeneration and/or diseases involving toxic agents and/or infectious pathogens:
• West Nile viral encephalitis (Morrey, Siddharthan et al. 2010), Japanese encephalitis(Tsao et al. J Gen Virol. 2008 Aug;89(Pt 8):1930-41 )
· Chronic hepatitis and cirrhosis induced by Hepatitis C virus (Piccoli, Scrima et al. 2007), Hepatitis B (Xia, Shen et al. 2005), delta agent,
• acute and chronic, drug- or toxin-induced hepatotoxicity (Soriano, Nicolosi et al. 2004; Masubuchi, Kano et al. 2006; Burke, Redeker et al. 2007), alcoholic liver disease (King, Swain et al. 2010; Shulga and Pastorino 2010), cholestasis, Non Alcoholic Fatty Liver Disease (Teodoro, Rolo et al.
2008), gastro-intestinal ulcerations caused by NSAIDs (LoGuidice,
Ramirez-Alcantra et al. Toxicol Sci. 2010),
• transplant rejection (Raisky, Gomez et al. 2004; Gomez, Raisky et al.
2006),
· peripheral vascular insufficiency, renal insufficiency,
• obesity (Devalaraja-Narashimha, Diener et al. 201 1 ), diabetic microangiopathy, diabetic nephropathy (Sun, Xiao et al. 2010), Maturity onset diabetes of the young type 4 (Fujimoto, Ford et al. 2010) • acute and chronic pancreatitis (Mukherjee, Criddle et al. 2008),
• septicaemia (Larche, Lancel et al. 2006), multi organ dysfunction syndrome, crush syndrome,
• congestive heart failure (Halestrap 2010), atherosclerosis (Di Lisa and
Bernardi 2009), hypertensive heart disease (Shabaz et al J Hypertens. 201
Sep; 28 Suppl 1 S25-32),
• cancer (Eliseev, Malecki et al. 2009)
• bipolar disorder (Kubota, Kasahara et al . 2010),
Agents that block MPT include the immune suppressant cyclosporin A (CsA) N-methyl-Val-4-cyclosporin A (MeVal-4-CsA), a non-immunosuppressant derivative of CsA; another non-immunosuppressive agent, NIM81 1 , 2- aminoethoxydiphenyl borate (2-APB), and bongkrekic acid. For example WO 2006/072639 (DEBIOPHARM SA) relates to the use of a cyclic undecapeptide for the preparation of a medicament for administration during a myocardial ischaemic event. Besides, WO 2009/098533 (DEBIOPHARM SA) relates to the use of a non-immunosuppressive cyclosporin A derivative for preventing or reducing muscular degeneration in a subject suffering from Limb- Girdle muscular Dystrophy.
Naturally occurring cyclosporins and derivatives present MPTP inhibitory activity but simultaneously display cytotoxicity which limits their anti-cell death effect.
It is know that cyclosporins are able to inhibit MPTP activity via their binding to mitochondrial cyclophilin and that MPTP modulation is involved in many
pathological processes. It is also known that cyclosporins display cytotoxicity.
Despite beneficial MPTP inhibition activity, the cyclosporins and derivatives display cytotoxicity which prevent their usage at higher doses and thus limit their therapeutic index. Overall, the anti-cell death effect of available cyclosporins and derivatives is limited by cytotoxicity. Thus there is a real need in obtaining compounds acting on MPTP and having reduced cytotoxicity which would result in an enhanced anti-cell death activity. Summary of the invention
Therefore the aim of the present invention is to provide new series of compounds with reduced cytotoxicity resulting in enhanced anti-cell death activity.
The present invention provides a cydoundecadepsipeptide compound of Formula (I) Cyclo-(AXXi-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-D-Hiv-MeLeu-Leu-lvleVal) (I)
1 2 3 4 5 6 7 8 9 10 1 1 in which:
AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
O-acetyl-MeBmt;
AXX2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH2CH2CH2OH), Nva, 5-hydroxy-Nva (Hnv);
AXX3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
D-MeSer(OCH2CH2OH), D-MeSer(OCH2CH2NEt2), D-MeAsp(OMe);
AXX4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe, EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone; AXX5 is Leu, Val, lie, Gly, Abu;
AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala; for use in a method for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening,
and wherein said cydoundecadepsipeptide compound of Formula (I) comply with the two following criteria:
(1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as: Compound _ of _ formula(I) _ CRC _ IC2 <
CsA _ CRC _ IC2 "
(2) at least three-fold less cytotoxic than CsA, defined as:
Compound _ of _ formula(I) _cell _ count _ IC50
CsA _ cell _ count _ IC50
The present invention further provides the use of a pharmaceutical composition for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening, comprising a compound of the invention, together with one or more pharmaceutically acceptable diluents or carriers.
Brief description of figures
Figure 1 shows plasma amylase, interleukin-6, pancreatic myeloperoxidase and histopathology scores following treatment in the bile acid-induced murine acute pancreatitis model (Student's t test: * = p < 0.05 comparing compound 057 with vehicle).
Detailed description of the invention
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are
incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting.
As used herein, the following definitions are supplied in order to facilitate the understanding of the present invention. The term "comprise" is generally used in the sense of include, that is to say permitting the presence of one or more features or components. "Treatment" refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder or the disease as well as those in which the disorder is to be prevented. Hence, the mammal to be treated herein may have been diagnosed as having the disorder or the disease may be predisposed or susceptible to the disorder or the disease.
"Mammal" for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals or pet animals, such as dogs, horses, cats, cows, monkeys etc. Preferably, the mammal is human. The term "therapeutically effective amount" refers to an amount of a drug effective to treat a disease or disorder in a mammal.
The terms "co-administration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
As used herein, the term "cell death associated disorders and diseases" comprises disorders and diseases caused by, associated with, linked with, mediated by or accompanied by the MPTP opening or wherein the MPTP opening is involved.
"Cell death" encompasses the normal degeneration and death of living cells, also defined as the terminal failure of a cell to maintain essential life functions, or the point in the process of dying at which vital functions have ceased at the cellular level. Cell death includes for example apoptosis as well as necrosis often defined as the morphological changes indicative of cell death caused by progressive enzymatic degradation; it may affect groups of cells or part of a structure or an organ. As referred to herein a "disorder" refers to an underlying pathological disturbance in a symptomatic or asymptomatic organism relative to a normal organism, which may result, for example, from infection, stress or an acquired or congenital genetic imperfection.
The invention refers to a series of cycloundecadepsipeptide compounds, with or without immunosuppressant activity, presenting mitochondrial permeability transition pore (MPTP) inhibitory activity and reduced cytotoxicity.
It is an object of the present invention to provide for a cycloundecadepsipeptide compound of Formula (I)
Cyclo-(AXXi-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-D-Hiv-MeLeu-Leu-lvleVal) (I)
1 2 3 4 5 6 7 8 9 10 1 1 in which:
AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
O-acetyl-MeBmt;
AXX2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH2CH2CH2OH), Nva, 5-hydroxy-Nva;
AXX3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
D-MeSer(OCH2CH2OH), D-MeSer(OCH2CH2NEt2), D-MeAsp(OMe);
AXX4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe,
EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone;
AXX5 is Leu, Val, lie, Gly, Abu;
AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala; for use in a method for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening, and wherein said
cycloundecadepsipeptide compound of Formula (I) comply with the two following criteria: (1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as:
Compound _ of _ formula(I) _ CRC _ IC2 <
CsA _ CRC _ IC2 " (2) at least three-fold less cytotoxic than CsA, defined as:
Compound _ of _ formula(I) _ cell _ count _ IC50 >
CsA _ cell _ count _ IC50
Preferably, said cycloundecadepsipeptide compound of Formula (I) is at least 5 fold less cytotoxic than CsA, and even more preferably at least ten-fold less cytotoxic than CsA, defined as:
Compound _of _ formula(I) _ cell _ count _ IC50 >
CsA _ cell _ count _ IC50 Calcium retention capacity (CRC) is a sensitive measure of the propensity of mitochondria to open the Permeability Transition Pore (MPTP) after Ca2+ uptake. The IC2 (nM) is the concentration of compound necessary to double the CRC, relative to the CRC observed in the absence of any compound (CRCo). According to lUPAC admitted definition, cyclodepsipeptides are natural or synthetic compounds having sequences of amino and hydroxyl carboxylic acid residues (usually a-amino and a-hydroxy acids) and the residues are connected in a ring (see lUPAC Compendium of Chemical Terminology, 2nd Edition (1997). Such cyclodepsipeptides are depicted as heterodetic peptides in which at least one amide bond has been replaced with an ester bond.
The compounds of the present invention comprise eleven residues, ten being a- amino acids and one being α-hydroxy acid. This α-hydroxy acid is
(2R)-2-hydroxy-3-methyl-butanoic acid, also known as D-a-hydroxyisovaleric acid and abbreviated as H-D-Hiv-OH. In Formula (I), this hydroxyl acid is in position 8. It forms on the carboxylic acid end an amide bond with the amino group of the α-amino acid in position 9, namely N-methyl-leucine, and, on the hydroxyl end an ester bond with the carboxylic acid group of the a-amino acid in position 7, namely alanine or glycine. The a-amino acids of Formula (I) are mentioned using the three letter code abbreviation usually used to name amino acids and their configuration is
L-configuration, unless otherwise specified. The residue numbering starts from AXXi representing N-methyl-(4R)-4-[(E)-2-butenyl]-4-methyl-L-threonine or MeBmt and its structural derivatives as defined above. When an alkyl group such as a methyl group Me or an ethyl group Et appears before the abbreviation of an amino acid, this means that such an alkyl group is fixed on the amino group of said amino acid residue.
An advantage of the compounds according to the present invention may lie in the impact of an ester bond within macro cyclic backbone in comparison to the regular cyclic amide backbone of the corresponding cycloundecapeptide compounds described in the prior art. Without being bound to theory, it is believed that the replacement of an amide by an ester bond between amino-acid AXX7 in position 7 and D-HIV in position 8 results in a strong effect upon the conformational and physico-chemical properties such as increased conformational flexibility and lipophilicity as well as the absence of a hydrogen donor bond.
It is believed that these structural features transform to pronounced differences in the physico-chemical, pharmaco-kinetic and biological properties of the
compounds of invention compared to the class of CsA derived analogues.
A possible rational for the observed higher tolerance to amino acid substitutions in systematic SAR studies may reside in an increase of the conformational space with respect to the bioactive conformation. In contrast to cycloundecapeptide analogues of the prior art, single or multiple replacements at positions in fragment between amino-acid in position 2 and amino-acid in position 7 of the natural cycloundecadepsipeptide, as obtained either from US 5,1 16,816, Example 2, or from WO 02/092033, Example 4, Step 4- 1 , result in the retention of high binding capacities to Cyclophilin as evidenced by the list of compounds meeting the criteria of the present invention. Most notably, an increase in the binding affinities to Cyp A of factors up to 2-4 for the
compounds of the invention compared to compound according to WO
2006/038088 i.e. [D-MeAla]3-[EtVal]4-CsA (Alisporivir) has been observed.
Apart from the improved active profile, the new class of compounds also offers an improved preparation process, especially at an industrial scale.
The invention also encompasses chemical modifications of the compounds of formula I to prolong their circulating lifetimes. Examples of suitable poly(ethylene glycol) derivatives that possess this property are described in e.g. US 2005171328 (NEKTAR THERAPEUTICS AL CORP) or US 6,713,454 (NOBEX CORP).
More preferably, the compounds of the present invention are defined by
Formula (I) in which
AXXi is MeBmt;
AXX2 is Abu. Val;
AXX3 is D-MeAla;
AXX4 is Melle, MeVal, EtVal;
AXX5 is Leu, Val, lie, Gly, Abu;
AXX 6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala.
According to one of the best mode of the invention, compounds of Formula (I) present the following formulae:
Compound Cycloundecadepsipeptide Sequence
001 Cyclo-(MeBmt-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-D-Hiv-MeLeu- Leu-MeVal)
009 Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Ala-D-Hiv-MeLeu- Leu-MeVal)
018 Cyclo-(MeBmt-Val-D-MeAla-MeVal-Leu-MeLeu-Ala-D-Hiv-MeLeu- Leu-MeVal)
020 Cyclo-(MeBmt-Thr-D-MeAla-MeVal-Leu-MeLeu-Ala-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeLys(Boc)-Leu-MeLeu-Ala-D-Hiv- MeLeu-Leu-MeVal)
Cyclo-(MeBmt-Val-D-MeAla-MeVal-Val-MeAla-Ala-D-Hiv-MeLeu-Leu- MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Val-MeAla-Ala-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Val-D-MeAla-EtVal-Leu-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-EtVal-Leu-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Val-D-MeAla-EtVal-Leu-MeAla-Ala-D-Hiv-MeLeu-Leu- MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-EtVal-Leu-MeAla-Ala-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Val-D-MeAla-MeVal-lle-MeLeu-Gly-D-Hiv-MeLeu-Leu- MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeVal-lle-MeAla-Gly-D-Hiv-MeLeu-Leu- MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeThr(fBu)-Leu-MeLeu-Ala-D-Hiv- MeLeu-Leu-MeVal)
Cyclo-(MeBmt-Val-D-MeAla-MeVal-Leu-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeVal-lle-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Val-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
Cyclo-(MeBmt-Val-D-MeAla-MeVal-Gly-Sar-Gly-D-Hiv-MeLeu-Leu- MeVal)
Cyclo-(MeBmt-Val-D-MeAla-MeVal-Val-Sar-Gly-D-Hiv-MeLeu-Leu- MeVal) 071 Cyclo-(MeBmt-Val-D-MeAla-MeVal-Abu-Sar-Gly-D-Hiv-MeLeu-Leu- MeVal)
073 Cyclo-(MeBmt-Abu-D-MeAla-Melle-Leu-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal)
More preferably, compounds of Formula (I) are selected among:
Compound Cycloundecadepsipeptide Sequence
009 Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Ala-D-Hiv-MeLeu- Leu-MeVal)
057 Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Gly-D-Hiv-MeLeu- Leu-MeVal) The above listed compounds correspond to an overall optimization of the most important criteria of the present invention, namely the reduction of cytotoxicity and the enhancement of anti-cell death activity. Notably, these compounds
demonstrate a much lower cytotoxicity and enhanced anti-cell death activity, as compared to cyclosporine A (CsA).
The compounds of the present invention have reduced cytotoxicity, which allows (1 ) possible use of higher doses to achieve a maximal effect; (2) increased therapeutic window and (3) no lack of activity at high doses. MPTP opening is a key event in the biochemical signaling that underlies mitochondria mediated cell death in various tissues, disorders and diseases.
Preferably the cell death associated disorders and diseases, wherein the MPTP opening is involved, are selected from the group comprising ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
More preferably, the cell death associated disorders and diseases, related to the MPTP opening, are selected from the group comprising ischaemia and ischaemia- reperfusion injuries, muscular dysthrophies, neurodegeneration disorders and diseases, West Nile viral encephalitis, Japanese encephalitis, Chronic hepatitis and cirrhosis induced by Hepatitis C virus, Hepatitis B, delta agent, acute and chronic drug- or toxin-induced hepatotoxicity, alcoholic liver disease, cholestasis, Non Alcoholic Fatty Liver Disease, gastro-intestinal ulcerations caused by
NSAIDs, transplant rejection, peripheral vascular insufficiency, renal insufficiency, obesity, diabetic micro-angiopathy, diabetic nephropathy, Maturity onset diabetes of the young type 4, acute and chronic pancreatitis, septicaemia, multi organ dysfunction syndrome, crush syndrome, congestive heart failure, atherosclerosis, hypertensive heart disease, cancer, bipolar disorder.
Even more preferably,
Ischaemia and ischaemia-reperfusion injuries are selected from the group comprising acute cerebral ischemia, acute stroke, acute myocardial infarction cardiac arrest, myocardial stunning, post-reperfusion arrhythmias, organ failure following therapeutic procedures such as thrombolytic therapy, coronary angioplasty, aortic cross-clamping or coronary bypass surgery, hypovolemic shock, storage/reperfusion injury of transplant organs, multi organ dysfunction syndrome.
Muscular dysthrophies are selected from the group comprising Ulrich congenital muscular dysthrophy, Bethlem myopathy, myosclerosis, limb girdle muscular dysthropy, Duchenne muscular dysthrophy, Becker muscular
dysthrophy, Emery-Dreifuss syndrome,
Neurodegeneration disorders and diseases are selected from the group comprising traumatic brain injury, spinal cord injury, peripheral nerve injury, epilepsy-induced brain injury, amyothrophic lateral sclerosis (ALS), Alzheimer disease, Parkinson disease , multiple sclerosis, Huntington disease, axonal degeneration-induced neuropathic pain, diabetic neuropathy chemotherapy- induced neuropathic pain, or herpes-induced neuropathy. Most preferably, the cell death associated disorders and diseases, related to the MPTP opening, are selected among acute and chronic pancreatitis and muscular dystrophy Pancreatitis is an inflammation of the pancreas that can occur in two very different forms. Acute pancreatitis is sudden while chronic pancreatitis is characterized by recurring or persistent abdominal pain with or without steatorrhea or diabetes mellitus. Acute pancreatitis is a severe and frequently lethal disorder most commonly caused by gallstone disease and alcohol abuse, whereas chronic pancreatitis is a long-standing inflammation of the pancreas that alters its normal structure and functions. Acute pancreatitis is characterized by acinar cell necrosis which contributes to pancreatic and extra-pancreatic autodigestion as well as systemic inflammatory response and multiple organ failure. A key event in acinar cell necrosis is the formation of the MPTP within the inner mitochondrial membrane leading to permeabilization of the mitochondrial membrane. This causes rapid depolarization of the mitochondrial membrane potential and uncouples ATP synthesis from oxidative metabolism leading to ATP depletion and necrosis (Mukherjee, R., et ai, 2008). Thus by inhibiting the formation of the MPTP, the compounds have the ability to prevent acinar cell necrosis and attenuate the pancreatic injury.
Muscular dystrophies comprise a diverse group of genetic disorders that lead to muscle wasting and, in many instances, premature death. It was demonstrated (Millay et al., 2008) that mitochondrial-dependent necrosis represents a prominent disease mechanism in muscular dystrophy. Indeed one major mechanism leading to cellular necrosis is mitochondrial calcium overload, which secondarily enhances reactive oxygen species (ROS) generation and further promotes MPT
(Mitochondrial Permeability Transition). Therefore the inhibition of MPTP opening provides a new treatment strategy for muscular dystrophies.
The compounds of the present invention may be obtained by applying classical peptide (solution or solid-phase peptide synthesis; in Houben - Weyl, Methods of Organic Chemistry, Vol. E 22d, Ed. -in-Chief: M. Goodman, Thieme Verlag, Stuttgart, 2003) and organic chemistry or biotechnology, for instance by applying the chemical tools as described by Wenger in Helv. Chim. Acta, 67, 502-25, 1984 or in Helv. Chim. Acta, 66, 2672-702 (1983) and by employing HATU as coupling reagent described in Rich, D.H. et al.. Comparative studies of the coupling of N- methylated, sterically hindered amino acids during SPPS, Tetr. Letters. 35, 5981 - 5984 (1994).
For instance, one of possible general scheme consists to prepare two fragments, namely Fragment A and Fragment B, containing the appropriate residues, with, when necessary the appropriate protective and activating groups, and, in the last steps of the preparation, to link them together to obtain an undecapeptide which is then cyclized to the cydoundecadepsipeptide. For instance, Fragment (Ax) may be as follows:
H-D-Hiv-MeLeu-Leu-MeVal-AXX OH (Ax)
and Fragment (Bx) as follows :
H-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-OR, (R being an alkyl group) (Bx), then Ax and Bx are coupled to the undecadepsipeptide Ax-Bx, H-D-Hiv-MeLeu- Leu-MeVal-AXXi-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-OR (R= alkyl, H)
and the final stage is the macrolactonisation.
Fragment A, which contains the a-hydroxy acid residue D-Hiv, may be obtained by degradation of a natural cydoundecadepsipeptide (namely Cyclo-(MeBmt-Thr-Sar- MeLeu-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal), the preparation of said cydoundecadepsipeptide being described either in US 5,1 16,816, Example 2, or in WO 02/092033, Example 4, Step 4-1 .
An advantage of the compounds according to the invention versus the prior art lies not only in its active profile (Example - Experimental Results), but also in its improved preparation process especially at an industrial scale.
1 . Starting from the natural compounds (CsA and Cyclo-(MeBmt-Thr-Sar-Mel_eu- Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal), the overall yields for the synthesis of cycloundecadepsipeptide analogues are > 50% compared to 20% for
ALISPORIVIR ([D-MeAla]3-[EtVal]4-CsA as described in WO 2006/038088, DEBIOPHARM SA). 2. The synthesis of the expensive dipeptide derivative Boc-D-MeAla-EtVal-OH (including 4 chemical steps) is not needed. In addition, some of the compounds contain a C-terminal Glycine, which facilitates the last step of the synthesis (macrolactonisation, no epimerisation). 3. The costs for reagents and starting compounds are considerably lower in case of the compounds according to the invention. Notably, the "reagent of Meerwein" used for the ring opening reaction of CsA is not needed.
4. Fragment B (hexapeptide AXX2-AXX7) can be efficiently obtained by standard solid-phase peptide synthesis using commercially starting compounds.
5. In optimizing the preparation of Fragment A from the natural starting compound as well as the condensation (fragment A with fragment B), macrocyclization and final purification steps, the overall yield for obtaining the pharmacologically interesting compounds can amount up to 80%.
6. Overall, the indicated aspects result in a good efficacy and consequently in cost of goods, in particular with respect to the synthesis of the compounds of invention at industrial scale.
The present invention further provides a pharmaceutical composition for treating and/or preventing cell death associated disorders or diseases, related to the MPTP opening, comprising a compound of the invention, together with one or more pharmaceutically acceptable diluents or carriers.
The compounds of the present invention may be administrated for instance parenterally or orally to a patient in need, for instance incorporated in a pre- concentrated microemultion. The compounds of the present invention, their pharmaceutically acceptable salts and pro-drugs thereof, where applicable, may be administered in the form of a pharmaceutical composition in which they are in association with a
pharmaceutically acceptable adjuvant, diluent or carrier, in order to prevent or treat cell death associated disorders or diseases, wherein the MPTP opening is involved.
The present invention also provides a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, as hereinbefore defined, in association with a pharmaceutically acceptable adjuvant, diluent or carrier. As to the appropriate excipients, diluents and
adjuvants, reference may be made to the standard literature describing these, e.g. to chapter 25.2 of Vol. 5 of "Comprehensive Medicinal Chemistry", Pergamon Press 1990, and to "Lexikon der Hilfsstoffe fur Pharmazie, Kosmetik und
angrenzende Gebiete", by H.P. Fiedler, Editio Cantor, 2002.
The compounds of the present invention may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi permeable matrices of solid hydrophobic polymers containing the compounds of the present invention, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained- release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl- methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and [gamma] ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as those used to prepare the medicament LUPRON DEPOT(TM) (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. The daily dose of the present invention will necessarily be varied depending upon the host treated, the particular route of administration, and the severity and kind of the illness being treated. Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient.
The pharmaceutical compositions of the invention may be formulated as creams, gels, solutions, ointments, suspensions or plasters etc. when intended for topical administration; for administration by inhalation, e.g. as aerosols or dry powders; for oral administration, e.g. in the form of tablets, capsules, gels, syrups, suspensions, solutions, powders or granules; for rectal or vaginal administration e.g. as suppositories; or for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular, intrathecal or infusion) as a sterile solution, suspension or emulsion.
The active compound of the invention may be administered by any conventional route. It may be administered parentally, e.g., in the form of injectable solutions or suspensions, or in the form of injectable deposit formulations. Preferably, it will be administered orally in the form of solutions or suspensions for drinking, tablets or capsules. The pharmaceutical compositions of the invention typically comprise a cycloundecadepsipeptide compound of the invention and one or more
pharmaceutically acceptable carrier substances. Typically, these compositions are concentrated and need to be combined with an appropriate diluent, e.g., water, prior to administration. Pharmaceutical compositions for parenteral administration typically also include one or more excipients. Optional excipients include an isotonic agent, a buffer or other pH- controlling agent, and a preservative. These excipients may be added for maintenance of the composition and for the
attainment of preferred ranges of pH (about 6.5-7.5) and osmolarity (about 300 mosm/L). Additional examples of formulations for oral administration can be found in U.S. Pat. Nos. 5,525,590 and 5,639,724, and U.S. Pat. Appl. 2003/0104992. By the oral route, the indicated dosage of a cycloundecadepsipeptide compound of the invention for daily to trice weekly administration may be from about 1 mg/kg to about 100 mg/kg, preferably from about 1 mg/kg to about 20 mg/kg. By the intravenous route, the indicated corresponding dosage may be from about 1 mg/kg to about 50 mg/kg, preferably from about 1 mg/kg to about 25 mg/kg. An effective amount of a cydoundecadepsipeptide compound of the invention is understood to be an amount that when administered in the course of a therapeutic regimen to a patient in need of treatment of MPTP opening mediated disorder results in an objective clinical response.
Numerous factors will be taken into consideration by a clinician when determining trial doses for testing efficacy of a pharmaceutical composition comprising a compound of the present invention against MPTP opening mediated disorders. Primary among these are the toxicity and half-life of the chosen
cydoundecadepsipeptide compound of the invention. Additional factors include the weight of the patient, the age of the patient, and the general condition of the patient. A course of treatment will require repeated administration of a
pharmaceutical composition of the invention. Typically, an adequate drug dose will be administered in one single administration (i.e. a unique dose) or repeatedly for example 3-7 times per week, and the duration of treatment may be for example from about 4 weeks to 6 months, preferably from about 4 weeks to about 12 months. A pharmaceutical composition of the present invention may comprise one or more other active ingredients which present MPTP inhibitory activity in addition to the compounds of the present invention. Compounds of the invention and such other active ingredients can be administered together as part of the same
pharmaceutical composition or can be administered separately as part of an appropriate dose regimen designed to obtain the benefits of the combination therapy. The appropriate dose regimen, the amount of each dose administered, and specific intervals between doses of each active agent will depend upon the specific combination of active agents employed, the condition of the patient being treated, and other factors discussed in the previous section. Such additional active ingredients will generally be administered in amounts less than or equal to those for which they are effective as single therapeutic agents. The FDA approved dosages for such active agents that have received FDA approval for administration to humans are publicly available. The compounds of the invention may be administered as the sole ingredient or together with other drugs.
Also encompassed is a method for preventing or treating cell death associated disorders or diseases, related to the MPTP opening, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of the pharmaceutical composition according to claim 4 or the cycloundecadepsipeptide compound of Formula (I) Cyclo-(AXXi-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-D-Hiv-MeLeu-Leu-lvleVal) (I)
1 2 3 4 5 6 7 8 9 10 1 1 in which:
AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
O-acetyl-MeBmt;
AXX2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH2CH2CH2OH), Nva, 5-hydroxy-Nva;
AXX3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
D-MeSer(OCH2CH2OH), D-MeSer(OCH2CH2NEt2), D-MeAsp(OMe);
AXX4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe, EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone; AXX5 is Leu, Val, lie, Gly, Abu;
AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala; and wherein said cycloundecadepsipeptide compound of Formula (I) comply with the two following criteria:
(1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as:
Compound _ of _ formula(I) _ CRC _ IC2 <
CsA _ CRC _ IC2 "
(2) at least three-fold less cytotoxic than CsA, defined as: Compound _ of _ formula(I) _ cell _ count _ IC50 >
CsA _ cell _ count _ IC50
Preferably, said cycloundecadepsipeptide compound of Formula (I) is at least 5 fold less cytotoxic than CsA, and even more preferably at least ten-fold less cytotoxic than CsA, defined as:
Compound _of _ formula(I) _ cell _ count _ IC50 >
CsA _ cell _ count _ IC50 Preferably the cell death associated disorders and diseases, wherein the MPTP opening is involved, are selected from the group comprising ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
More preferably, the cell death associated disorders and diseases, related to the MPTP opening, are selected from the group comprising ischaemia and ischaemia- reperfusion injuries, muscular dysthrophies, neurodegeneration disorders and diseases, West Nile viral encephalitis, Japanese encephalitis, Chronic hepatitis and cirrhosis induced by Hepatitis C virus, Hepatitis B, delta agent, acute and chronic drug- or toxin-induced hepatotoxicity, alcoholic liver disease, cholestasis, Non Alcoholic Fatty Liver Disease, gastro-intestinal ulcerations caused by
NSAIDs, transplant rejection, peripheral vascular insufficiency, renal insufficiency, obesity, diabetic micro-angiopathy, diabetic nephropathy, Maturity onset diabetes of the young type 4, acute and chronic pancreatitis, septicaemia, multi organ dysfunction syndrome, crush syndrome, congestive heart failure, atherosclerosis, hypertensive heart disease, cancer, bipolar disorder.
Even more preferably,
Ischaemia and ischaemia-reperfusion injuries are selected from the group comprising acute cerebral ischemia, acute stroke, acute myocardial infarction cardiac arrest, myocardial stunning, post-reperfusion arrhythmias, organ failure following therapeutic procedures such as thrombolytic therapy, coronary angioplasty, aortic cross-clamping or coronary bypass surgery, hypovolemic shock, storage/reperfusion injury of transplant organs, multi organ dysfunction syndrome.
Muscular dysthrophies are selected from the group comprising Ulrich congenital muscular dysthrophy, Bethlem myopathy, myosclerosis, limb girdle muscular dysthropy, Duchenne muscular dysthrophy, Becker muscular
dysthrophy, Emery-Dreifuss syndrome,
Neurodegeneration disorders and diseases are selected from the group comprising traumatic brain injury, spinal cord injury, peripheral nerve injury, epilepsy-induced brain injury, amyothrophic lateral sclerosis (ALS), Alzheimer disease, Parkinson disease , multiple sclerosis, Huntington disease, axonal degeneration-induced neuropathic pain, diabetic neuropathy chemotherapy- induced neuropathic pain, or herpes-induced neuropathy.
Most preferably, the cell death associated disorders and diseases, related to the MPTP opening, are selected among acute and chronic pancreatitis and muscular dystrophy
Another object of the invention is a method, comprising co-administration concomitantly or in sequence of a therapeutically effective amount of the compound according to the invention or the pharmaceutical composition of the invention and a co-agent selected from an agent having blocking properties on MPTP opening.
The administration of a pharmaceutical combination of the invention results in a beneficial effect, e. g. a synergistic therapeutic effect, compared to a monotherapy applying only one of its pharmaceutical active ingredients. For instance, a synergistic combination is for example a combination of the
cydoundecadepsipeptide compound of the invention with an interferon, optionally conjugated to a polymer. All patents, patent applications and publications cited herein shall be considered to have been incorporated by reference in their entireties.
Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and
modifications without departing from the spirit or essential characteristics thereof. The invention also includes all of the steps, features, compositions and
compounds referred to or indicated in this specification, individually or collectively, and any and all combinations or any two or more of said steps or features. The present disclosure is therefore to be considered as in all aspects illustrated and not restrictive, the scope of the invention being indicated by the appended Claims, and all changes which come within the meaning and range of equivalency are intended to be embraced therein.
Various references are cited throughout this Specification, each of which is incorporated herein by reference in its entirety.
The foregoing description will be more fully understood with reference to the following Examples. Such Examples, are, however, exemplary of methods of practicing the present invention and are not intended to limit the scope of the invention.
Examples
Example 1 :
Preparation of cvcloundecadepsipeptide (compound 001 ):
cyclo-(MeBmt-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal)
(001 ) 1. Preparation of Fragment (Aa), starting from natural
cycloundecadepsipeptide Cyclo-(MeBmt-Thr-Sar-MeLeu-Leu-Mel_eu-Ala-D- Hiv-MeLeu-Leu-MeVal)
(see US 5,1 16,816, Example 2, or in WO 02/092033), Example 4, Step 4-1 ): H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-OH (Aa)
1.1. Preparation of Cyclo-(MeBmt-Thr(0-(N-lmidazolyl)carbonyl)-Sar-Mel_eu- Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal) (1A).
A solution of Cyclo-(MeBmt-Thr-Sar-MeLeu-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu- MeVal) (US 5,1 16,816, Example 2, or in WO 02/092033, Example 4, Step 4-1 ) (3.00 g, 2.40 mmol, 1 .0 equiv.) and 1 ,1 '-carbonyldiimidazole (1 .17 g, 7.21 mmol, 3.0 equiv.) in 15 mL of anhydrous CH2CI2 was stirred at room temperature for 2.5 h. Progress of the reaction was monitored by analytical UPLC. 81 .1 % conversion was obtained. Additional amount of 1 ,1 '-carbonyldiimidazole (0.39 g, 2.40 mmol, 1 .0 equiv.) was added to the reaction mixture. After additional stirring for 16 h, a 98.8% conversion was obtained. The solution was evaporated under reduced pressure. The residue was dissolved in AcOEt (45 mL) and washed successively with 10% citric acid (45 mL) and brine (45 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure to give a mixture of two compounds (1A) and (2A) as a white powder
UPLC-ESI-MS (m/z): (1A): 1342.75 [M + H]+
Figure imgf000027_0001
calc. 1342.73), (2A): 1274.77 [M + H]+ ([CesH aNioOis]*; calc. 1274.65)
1.2. Preparation of Cyclo-(MeBmt-Thr(0,N-carbonyl)-Sar-Mel_eu-l_eu- MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal) (2A). A solution of compound (1 A) as obtained in the preceding reaction (3.41 g, 2.40 mmol) in dry DMSO (15 mL) was stirred and heated at 100 °C for 2 h under argon atmosphere. Progress of the reaction was monitored by analytical UPLC. Full conversion was obtained. Subsequently, the solution was dissolved in AcOEt (45 mL) and washed successively with HCI 1 M (30 mL) and 23.2% aq. NaCI solution (15 mL) then 1 1 .6% aq. NaCI solution (45 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure to give the
cycloundecadepsipeptide Cyclo-(MeBmt-Thr(0,N-carbonyl)-Sar-MeLeu-Leu- MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal) (2A) as white powder UPLC-ESI-MS (m/z): 1274.65 [M + H]+ ([C65Hi i3NioOi5]+; calc. 1274.65.
1.3. Preparation of H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-OMe (fragment (Aa) methyl ester (3A).
To a solution of the cyclodepsipeptide as obtained in the preceding reaction (1 .00 g, 0.78 mmol) in 30 ml_ of MeOH (dry), cooled at 0 °C, was added KOMe (0.24 g, 2.35 mmol) in one portion, under argon. After 5 h at 0 °C then 1 h at room temperature, a control of the reaction advancement by UPLC indicates 98.6% conversion. The solution was cooled at 0 °C then neutralized with an aq. solution of 10% citric acid. Methanol was evaporated. The aq. mixture was poured into 100 ml_ of a mixture of AcOEt NaCI 23.2% (1 :1 v/v). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. Crude mixture was separated by column chromatography; to give the pentapeptide -D-Hiv-MeLeu- Leu-MeVal-MeBmt-OMe (3A
UPLC-ESI-MS (m/z): 669.16 [M + H]+ ([C35H65N4O8]+; calc. 669.48).
1.4. Preparation of Fragment (Aa) H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-OH (4).
A solution of the pentapeptide as obtained in the preceding reaction (3A) (1 .15 g, 1 .72 mmol, 1 .0 equiv.) in THF (8.6 mL) and water (1 .1 mL) was cooled to 0 °C in an ice-water bath, then 2 M LiOH (1 .72 mL, 3.44 mmol, 2.0 equiv.) was added within 20 seconds. The cooling bath was then removed and the mixture (pH = 12- 13) was stirred at room temperature for about 3 h. A control of the reaction advancement by UPLC indicates a full conversion. Subsequently, the solution was cooled to 0 °C and neutralized with 10% citric acid. Tetrahydrofurane was evaporated under reduced pressure. The residue was dissolved in AcOEt (50 mL) and washed with 10% citric acid (3 mL) and 23.2% aq. NaCI solution (50 mL) (pH 3). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure to give Fragment (Aa) H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-OH (4) as a white powder
UPLC-ESI-MS (m/z) 655.21 [M + H]+ ([C3 H63N4O8]+; calc. 655.46). 2. Preparation of Fragment (Bb) :
H-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe (Bb) 2.1. Preparation of Boc-MeLeu-Ala-OMe
To a solution of commercially available H-Ala-OMe HCI (2.00 g, 14.32 mmol, 1 .0 equiv.) in dry DCM (1 30 mL), cooled at 0 °C, was added under argon DIPEA (14.68 mL, 86.0 mmol, 6.0 equiv.). Subsequently, after 1 5 min, HATU (6.52 g, 1 7.1 8 mmol, 1 .2 equiv.) and Boc-MeLeu-OH (3.51 2 g, 14.32 mmol, 1 .0 equiv.) were added in one portion. After 15 min at 0 °C then 48 h at room temperature (RT = 2 days 1 5 min), a control of the reaction advancement by TLC indicates the completion. The reaction was quenched by addition of 1 0% NaHCO3 (20 mL) and stirred 15 min. Reaction mixture was diluted with DCM (200 mL) and washed with 1 0% citric acid (1 χ 40 mL), H2O (1 χ 40 mL) and brine (1 χ 40 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. Crude mixture was separated by CC to give the ester Boc-MeLeu-Ala-OMe as a pale yellowish oil. The sample (30 mg) was repurified by semi-preparative
RP-HPLC and the peptide was lyophilized to give the ester Boc-MeLeu-Ala-OMe as a white powder.
2.2. Preparation of H-MeLeu-Ala-OMe
A solution of dipeptide as obtained in the preceding reaction (1 .70 g, 5.14 mmol, 1 .0 equiv.) in TFA/DCM (1 0 mL, 2:3 v/v) was kept at 0 °C for 1 .5 h, and the solvents removed under reduced pressure. The crude product was dried under high vacuum (20 min). Subsequently, DCM (1 0 mL) was added and the mixture was triturated at 0 °C with DIPEA to pH 7-8 to neutralize an excess of TFA.
Subsequently, reaction mixture was diluted with DCM (60 mL) and washed with H2O (1 x 1 0 mL) and brine (1 x 1 0 mL). The organic phase was filtered, dried (Na2SO4), evaporated and dried under high vacuum. The crude amine salt of H-MeLeu-Ala-OMe was used for the next coupling step without further purification. 2.3. Preparation of Boc-Leu-MeLeu-Ala-OMe
To a solution of dipeptide as obtained in the preceding reaction (7) (2.17 g, 9.42 mmol, 1 .0 equiv.) in dry DCM (108 ml_), cooled at 0 °C, was added under argon DIPEA (6.45 ml_, 37.68 mmol, 4.0 equiv.). Subsequently, after 10 min, HATU (4.65 g, 12.25 mmol, 1 .3 equiv.) and Boc-Leu-OH (2.39 g, 10.36 mmol, 1 .1 equiv.) were added in one portion. After 15 min at 0 °C then 15 h at room temperature (15 h 15 min), a control of the reaction advancement by TLC indicates the completion. The reaction was quenched by addition of 10% NaHCO3 (10 ml_) and stirred 15 min. Reaction mixture was diluted with DCM (50 ml_) and washed with 10% citric acid (1 χ 20 ml_), H2O (1 χ 20 ml_) and brine (1 χ 20 ml_). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. Crude mixture was separated by CC to give the tripeptide Boc-Leu- MeLeu-Ala-OMe as a yellowish oil. The sample was re-purified by semi- preparative RP-HPLC and the peptide lyophilized to give the tripeptide Boc-Leu- MeLeu-Ala-OMe as a white powder.
2.4. Preparation of H-Leu-MeLeu-Ala-OMe A solution of the tripeptide as obtained in the preceding reaction (2.00 g, 4.50 mmol, 1 .0 equiv.) in TFA DCM (10 mL, 2:3 v/v) was kept at 0 °C for 2 h, and the solvents removed under reduced pressure. The product was dried under high vacuum (20 min) to give the crude material. Subsequently, DCM (20 mL) was added and the mixture was triturated at 0 °C with DIPEA to pH 7-8 (Lackmus- paper) to neutralize an excess of TFA. After evaporation and drying the crude amine H-Leu-MeLeu-Ala-OMe was used to the next coupling step without further purification. The sample (25 mg) was repurified by semi-preparative RP-HPLC and the product was lyophilized to give the tripeptide H-Leu-MeLeu-Ala-OMe as a solid 2.5. Preparation of Boc-D-MeAla-EtVal-OH
Boc-D-MeAla-EtVal-OH was obtained starting from Boc-D-MeAla-Val-OMe using BuLi and Triethyloxoniumfluoroborate, followed by hydrolysis of the methylester according to literature [Jean Frangois. Guichou, PhD thesis entitled "De nouveaux analogues de Cyclosporine A comme agent anti-VIH-1 ", Faculte des Sciences, Universite de Lausanne, 2001 , p.1 21 -1 22] . 2.6. Preparation of Boc-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe
To a solution of the tripeptide H-Leu-MeLeu-Ala-OMe (0.096 g, 0.291 mmol, 1 .0 equiv.) in dry DCM (8 mL), cooled at -8 °C, was added under argon DIPEA (0.1 5 mL, 0.87 mmol, 3.0 equiv.). Subsequently, after 1 0 min, HATU (0.1 3 g, 0.35 mmol, 1 .2 equiv.) and the dipeptide Boc-D-MeAla-EtVal-OH (0.1 g,
0.29 mmol, 1 .0 equiv.) were added in one portion. After 1 0 min at -8 °C then 40 min at room temperature, a control of the reaction advancement by HPLC indicates the completion. The reaction was quenched by addition of 1 0% NaHCO3 (1 mL) and stirred 1 5 min. Reaction mixture was diluted with DCM (20 mL) and washed with 1 0% citric acid (1 x 5 mL), H2O (1 x 5 mL) and brine (1 x 5 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude mixture was separated by CC to give the pentapeptide Boc-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as an oil. The sample was repurified by semi-preparative RP-HPLC and the product was lyophilized to give the
pentapeptide Boc-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as a white powder.
2.7. Preparation of H-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe
A solution of the pentapeptide as obtained in the preceding reaction (0.45 g, 0.69 mmol, 1 .0 equiv.) in TFA DCM (4.0 mL, 2 :3 v/v) was kept at 0 °C for 1 .5 h, and the solvents removed under reduced pressure. The residue was dried under high vacuum (0.5 h). Subsequently, DCM (1 0 mL) was added and the mixture was triturated at 0 °C with DIPEA to pH 7-8 (Lackmus-paper) to neutralize an excess of TFA. After evaporation and drying the crude amine salt of the pentapeptide H-D- MeAla-EtVal-Leu-MeLeu-Ala-OMe was used to the next coupling step without further purification. A sample (30 mg) was purified by semi-preparative RP-HPLC and the peptide was lyophilized to give the free amine of the pentapeptide H-D- MeAla-EtVal-Leu-MeLeu-Ala-OMe as a white powder. 2.8. Preparation of Boc-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe
To a solution of the crude pentapeptide as obtained in the preceding reaction (0.2 g, 0.36 mmol, 1 .0 equiv.) in dry DCM (10 mL), cooled at 0 °C, was added under argon DIPEA (0.18 mL, 1 .08 mmol, 3.0 equiv.). Subsequently, after 5 min, HATU (0.19 g, 0.5 mmol, 1 .4 equiv.) and Boc-Abu-OH (0.088 g, 0.42 mmol, 1 .1 equiv.) were added in one portion. After 15 min at 0 °C then 1 .15 h at room temperature (RT = 1 .5 h), a control of the reaction advancement by TLC indicates the completion. The reaction was quenched by addition of 10% NaHCO3 (2 mL) and stirred 15 min. Reaction mixture was diluted with DCM (50 mL) and washed with 10% citric acid (1 x 10 mL), H2O (1 χ 10 mL) and brine (1 x 10 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. Crude mixture was separated by CC to give the hexapeptide Boc-Abu- D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as a yellowish oil . A sample
(30 mg) was repurified by semi-preparative RP-HPLC and the product lyophilized to give the hexapeptide Boc-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as a white powder. 2.9. Preparation of H-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe Fragment (Bb)
A solution of the hexapeptide as obtained in the preceding reaction (0.187 g, 0.25 mmol, 1 .0 equiv.) in TFA DCM (3.0 mL, 2:3 v/v) was kept at 0 °C for 1 .5 h, and the solvents removed under reduced pressure. The crude product was dried under high vacuum (0.5 h). Subsequently, DCM (3 mL) was added and the mixture was triturated at 0 °C with DIPEA to pH 7-8 (Lackmus-paper) to remove TFA. After evaporation and drying the crude amine H-Abu-
D-MeAla-EtVal-Leu-MeLeu-Ala-OMe was used to the next coupling step without further purification. A sample of crude product (23 mg) was re-purified by semi- preparative RP-HPLC and the peptide was lyophilized to give Fragment (Ba) H-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as a white powder.
UPLC-ESI-MS (m/z) 641 .259 [M + H]+
Figure imgf000032_0001
calc.641 .4602). 3. Coupling of fragment (Aa) and fragment (Bb)
3.1. Preparation of H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-Abu-D-MeAla-EtVal- Leu-MeLeu-Ala-OMe
To a solution of the hexapeptide H-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-OMe as obtained in the preceding reaction (0.16 g, 0.25 mmol, 1 .0 equiv.) in dry DCM (10 mL), cooled at 0 °C, was added under argon DIPEA (0.214 mL, 1 .26 mmol, 5.0 equiv.). Subsequently, after 5 min, HATU (0.12 g, 0.31 mmol, 1 .25 equiv.) and the pentadepsipeptide H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-OH (4) as obtained above (0.16 g, 0.25 mmol, 1 .0 equiv.) were added in one portion. After 15 min at 0 °C then 1 .15 h at room temperature (RT = 1 .5 h), a control of the reaction advancement by TLC indicates the completion. The reaction was quenched by addition of 10% NaHCO3 (3 mL) and stirred 15 min. Reaction mixture was diluted with DCM (40 mL) and washed with 10% citric acid (1 x 8 mL), H2O (1 x 8 mL) and brine (1 x 8 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. Crude mixture was separated by CC to give the undecapeptide H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-Abu-D-MeAla-EtVal-Leu- MeLeu-Ala-OMe as a white foam. An analytical sample (18 mg) was repurified by semi-preparative RP-HPLC and the product was lyophilized to give the
undecapeptide H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-Abu-D-MeAla-EtVal-Leu- MeLeu-Ala-OMe as a white powder.
3.2. Preparation of H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-Abu-D-MeAla-EtVal- Leu-MeLeu-Ala-OH
A solution of the undecapeptide methylester as obtained in the preceding reaction (0.27 g, 0.21 mmol, 1 .0 equiv.) in THF (2.5 mL) was cooled to 0 °C in an ice-water bath, then 0.2 M LiOH (2.1 1 mL, 0.42 mmol, 2.0 equiv.) was added dropwise over 10 min. The cooling bath was then removed and the mixture (pH = 12-13;
Lackmus-paper) was stirred at room temperature for 1 h 20 min (TLC control). Subsequently, the solution was cooled to 0 °C and acidified with 1 .0 M HCI to pH 3-4. The solution was evaporated under reduced pressure. The residue was dissolved in AcOEt (40 mL) and washed with 10% citric acid
(1 x 8 mL), H2O (1 x 8 mL) and brine (1 x 8 mL). The organic phase was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude mixture was separated by CC to give the acid H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-Abu- D-MeAla-EtVal-Leu-MeLeu-Ala-OH as a white powder. The sample (40 mg) was repurified by semi-preparative RP-HPLC and the product was lyophilized to give the undecapeptide H-D-Hiv-MeLeu-Leu-MeVal-MeBmt-Abu-D-MeAla-EtVal-Leu- MeLeu-Ala-OH as a white powder. 4. Macrolactonisation : Preparation of cyclo-(MeBmt-Abu-D-MeAla-EtVal- Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal) (compound 001)
To a solution of DMAP (0.048 g, 0.39 mmol, 4.0 equiv.) and benzotriazol- 1 -yloxytripyrrolidinophosphonium hexafluorophosphate (PyBOP) (0.102 g, 0.196 mmol, 2.0 equiv.) in DCM (12 mL) was added dropwise over 1 .5 h a solution of the undecapeptide as obtained in the preceding reaction (0.124 g, 0.098 mmol, 1 .0 equiv.) in DCM (4 mL). The mixture was stirred after the addition of acid was complete (RT = 24 h), then transferred to a separating funnel. The solution was washed with 1 M HCI (2 mL) and the organic layer was separated and dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography, thereby giving the
cycloundecadepsipeptide cyclo-(MeBmt-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-D- Hiv-MeLeu-Leu-MeVal) (la) as a white solid. A sample (77 mg) was purified by semi-preparative RP-HPLC and the product was lyophilized to give the
cycloundecadepsipeptide cyclo-(MeBmt-Abu-D-MeAla-EtVal-Leu-MeLeu-Ala-D- Hiv-MeLeu-Leu-MeVal) (lb) as a white powder.
UPLC-ESI-MS (m/z) 1245.787 [M + H]+ ([CesH /NioOia]*; calc. 1245.8802), 623.465 [M/2 + H]+ (calc. 623.444). Other cycloundecadepsipeptide compounds of Formula (I) according to the invention can be prepared according to analogous reaction pathway as described above or any other techniques known to the skilled in the art. Example 2: BIOLOGICAL DATA
MPTP inhibitory activity The MPTP activity was evaluated by measuring the Calcium Retention Capacity (CRC) of mouse liver mitochondrial preparations. CRC is a sensitive measure of the propensity of mitochondria to open the Permeability Transition Pore (MPTP) after Ca2+ uptake. CRC of mitochondria was assessed fluorimetrically using the Ca2+ indicator Calcium Green-5N. Mitochondria was incubated 1 min with ranging concentrations of the test compound and the concentration of compound necessary to double the CRC (IC2) was determined by constructing dose-response curves.
Experimental procedure
Mitochondria were isolated from the liver of 9-10 week old male C57BI/6j mice. After isolation the liver was kept at 4°C in a Tris-buffered (pH 7.4) sucrose-based isotonic solution and quickly minced and homogenized with a Teflon/glass potter. The suspension was spun for 6 min at 700 x g, and the resulting supernatant was spun for 6 min at 7,000 x g. The mitochondrial pellet was carefully resuspended in excess volume of sucrose buffer and spun again as above. The final mitochondrial pellet was resuspended with a minimal volume of sucrose buffer. Protein concentration was determined with the biuret method. Mitochondrial suspensions were used immediately and not beyond 4 h from isolation.
The CRC of mitochondrial preparations was assessed fluorimetrically in the presence of the Ca2+ indicator Calcium Green-5N at room temperature using a Fluoroskan Ascent FL fluorometer (Thermo Electron Corporation). The
experiments were done in 96-well plates with 100μΙ of mitochondrial suspension, adding 20 μΜ Ca2+ (2μΙ of a 1 mM Ca2+ solution) every minute and monitoring Ca- green fluorescence changes at Aex 485 nm, Aem 538 nm.
All compounds were dissolved in DMSO to a final concentration of 10 mM, and stored at 4°C. Data analysis
Every treatment was represented by the mean value ± SD of (1 ) the CRC and (2) the ratio between CRC (as just defined) and CRCo (concentration of Ca2+ which opens the PTP in the absence of the compound).
The IC2 (nM) was calculated as the concentration of compound necessary to double the CRC, relative to the CRC observed in the absence of any compound (CRCo).
Cytotoxicity
Cytotoxicity was evaluated by measuring proliferation of HepG2 cells. Cells were incubated with ranging concentrations of the test compound for 72hrs. Cell proliferation is measured by the signal intensity of the incorporated nuclear dye. The half maximal inhibitory concentration (IC50) was determined by constructing dose-response curves.
Experimental Procedure
Cells were grown in RPMI 1640, 10%FBS, 2 mM L-alanyl-L-Glutamine, 1 mM Na Pyruvate or a special medium in a humidified atmosphere of 5% CO2 at 37°C. Cells were seeded into 384-well plates and incubated in a humidified atmosphere of 5% CO2 at 37°C. Compounds were added 24 hours post cell seeding. At the same time, a time zero untreated cell plate was generated.
After a 72 hour incubation period, cells were fixed and stained with nuclear dye. Automated fluorescence microscopy was carried out using a GE Healthcare IN Cell Analyzer 1000, and images were collected with a 4X objective.
Data Analysis
Twelve bit tiff images were acquired using the InCell Analyzer 1000 3.2 and analyzed with Developer Toolbox 1 .6 software. The cell proliferation assay output is referred to as the relative cell count. Relative cell count IC5o is the test compound concentration that produces 50% of the cell proliferation inhibitory response or 50% cytotoxicity level. IC5o values were calculated using nonlinear regression to fit data to a sigmoidal 4 point, 4 parameter One-Site dose response model, where: y (fit) = A + [(B - A)/(1 + ((C/x) Λ D))]. Curve-fitting and IC50 calculations are performed using a custom data reduction engine MathlQ based software (AIM).
Experimental Results
For selected compounds having MPTP inhibitory activity and cytotoxicity as defined in the present invention
(1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as:
Compound _ of _ formula(I) _ CRC _ IC2 <
CsA _ CRC _ IC2
(2) at least three-fold less cytotoxic than CsA, defined as:
Compound _ of _ formula(I) _ cell _ count _ IC50 >
CsA cell count IC,n
Compound CRC IC2 Compound cell count IC5o
Compound
/ CsA CRC \C2 / CsA cell count IC5o
CsA 1 .0 1 .0
001 1 .2 13.6
009 0.5 13.6
018 0.3 3.8
020 0.4
036 13.6
044 0.3 045 0.3 3.1
049 0.8 12.5
050 0.5 13.6
051 0.5 4.3
052 0.7 13.6
055 0.3 13.6
056 0.3 3.1
057 0.4 13.6
058 13.6
062 0.4 13.6
064 0.2 9.6
065 0.3 8.8
067 1 .7 5.9
070 0.1 9.1
071 5.2
073 0.5 13.6
Table 1
Conclusion:
Table 1 shows that the selected compounds according to the invention have a MPTP inhibitory activity at least equal to half of the CsA activity as well as at least equal to half of the activity of the compounds of the prior art. In addition the selected compounds of the invention are at least three-fold less cytotoxic than CsA as well as least three-fold less cytotoxic than the compounds of the prior art.
Example 3: Acute pancreatitis Models The compounds have been shown to significantly ameliorate disease outcome in the bile acid-induced murine acute pancreatitis model which closely reproduce clinical pathologies. The bile acid-induced murine acute pancreatitis model is similar to gallstone induced clinical acute pancreatitis, which results from retrograde passage of bile into the pancreatic duct when gallstones impact at the sphincter of Oddi. Gallstone pancreatitis is the commonest form of the disease within the majority of countries in the world, except those where alcohol consumption is relatively very high.
Biliary pancreatitis was induced by retrograde perfusion of the pancreatic duct with 2 mM TLCS; sacrifice and assessments were made 24 hours later (Laukkarinen, J.M. et al, 2007).
Results:
Single administration of 10 mg/kg compound 057 given by intraperitoneal injection one hour after pancreatic ductal infusion of bile acid abolished or markedly reduced histopathological changes, plasma amylase and interleukin-6, pancreatic myeloperoxidase and histopathology scores as represented in Figure 1 .
The bile acid-induced murine acute pancreatitis model is a close representative of clinical acute pancreatitis, underlining the possible clinical utility of an early administration of compound 057 as a treatment for this disease.
List of Abbreviations
Abu L-a-amino-n-butyric acid
Ac acetyl
AcOEt ethyl acetate
AcOH acetic acid
Ala L-alanine
Boc teff-butyloxycarbonyl
BSEP bile salt export pump
f-Bu fert-butyl
Ca(MeO)2 calcium methoxide
CC column chromatography
DCM dichloromethane
DIPEA Λ/,/V-diisopropylethylamine
DMAP 4-(/V,/V-dimethylamino)pyhdine
DMSO dimethyl sulfoxide
Eq equivalent
ESI-MS electrospray ionization mass spectrometry
EtVal /V-ethyl-L-valine
Fmoc 9-fluorenylmethoxycarbonyl
HATU A/-[(dimethylamino)-1 H-1 ,2,3-triazolo[4,5-b]pyridine-1 -yl- methylene]-/V-methylmethanaminium hexafluorophosphate N- oxide
D-Hiv D-hydroxyisovaleric acid
g gramme
HCOOH formic acid
HPLC high performance liquid chromatography
HR-MALDI-MS high resolution MALDI-TOF mass spectrometry
HR-Q-TOF-MS high resolution Q-TOF mass spectrometry
lm2CO Ν,Ν-carbonyl-dimidazole (CDI)
Kg kilogramme
KOMe potassium methoxide
L liter MALDI matrix-assisted laser desorption/ionization
MALDI-TOF time-of-flight mass spectrometry
Me methyl
D-MeAla /V-methyl-D-alanine
MeBmt A/-methyl-(4R)-4-[(E)-2-butenyl]-4,4-dimethyl-L-threonine
MeLeu /V-methyl-L-leucine
MeCN acetonitrile
MeOH methanol
MeVal /V-methyl-L-valine
mg milligramme
ml_ milliliter
MRP2 multidrug resistance associated protein 2
MS mass spectrometry
MtBE methyl-tertio-butyl-ether
NMR nuclear magnetic resonance
NTCP sodium-taurocholate cotransporting polypeptide
OATP1 B1 organic anion transporting polypeptide 1 B1
PyBOP (benzotriazol-1 -yloxy)-tris(pyrrolidino)phosphonium- hexafluorophosphate
RP reverse-phase
RP-HPLC reverse-phase HPLC
RT reaction time
Thr L-threonine
TFA trifluoroacetic acid
tR retention time
THF tetrahydrofuran
Tj temperature of the jacket
TLC thin layer chromatography
Tr temperature into the reactor
UPLC ultra performance liquid chromatography
Vol. volumes (1g of key raw material means 1 volume) References
Abramov, A. Y. and M. R. Duchen (2008). "Mechanisnns underlying the loss of mitochondrial membrane potential in glutamate excitotoxicity." Biochim
Biophvs Acta 1777(7-8): 953-964.
Angelin, A., T. Tiepolo, et al. (2007). "Mitochondrial dysfunction in the
pathogenesis of Ullrich congenital muscular dystrophy and prospective therapy with cyclosporins." Proc Natl Acad Sci U S A 104(3): 991 -996.
Baines, C. P., R. A. Kaiser, et al. (2005). "Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death." Nature
434(7033): 658-662.
Barrientos, S. A., N. W. Martinez, et al. "Axonal degeneration is mediated by the mitochondrial permeability transition pore." J Neurosci 31(3): 966-978.
Brustovetsky, N., T. Brustovetsky, et al. (2003). "Increased susceptibility of striatal mitochondria to calcium-induced permeability transition." J Neurosci 23(12): 4858-4867.
Burke, A. S., K. Redeker, et al. (2007). "Mechanisms of chloroform-induced
hepatotoxicity: oxidative stress and mitochondrial permeability transition in freshly isolated mouse hepatocytes." J Toxicol Environ Health A 70(22):
1936-1945.
Devalaraja-Narashimha, K., A. M. Diener, et al. (201 1 ). "Cyclophilin D deficiency prevents diet-induced obesity in mice." FEBS Lett 585(4): 677-682.
Di Lisa, F. and P. Bernardi (2009). "A CaPful of mechanisms regulating the
mitochondrial permeability transition." J Mol Cell Cardiol 46(6): 775-780.
Du, H., L. Guo, et al. (2008). "Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in
Alzheimer's disease." Nat Med 14(10): 1097-1 105.
Du, H. and S. S. Yan "Mitochondrial permeability transition pore in Alzheimer's disease: cyclophilin D and amyloid beta." Biochim Biophvs Acta 1802(1 ):
198-204.
Du, H. and S. S. Yan (2010). "Mitochondrial permeability transition pore in
Alzheimer's disease: cyclophilin D and amyloid beta." Biochim Biophvs Acta 1802(1 ): 198-204. Eliseev, R. A., J. Malecki, et al. (2009). "Cyclophilin D interacts with Bcl2 and exerts an anti-apoptotic effect." J Biol Chem 284(15): 9692-9699.
Forte, M., B. G. Gold, et al. (2007). "Cyclophilin D inactivation protects axons in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis." Proc Natl Acad Sci U S A 104(18): 7558-7563.
Fujimoto, K., E. L. Ford, et al. (2010). "Loss of Nix in Pdx1 -deficient mice prevents apoptotic and necrotic beta cell death and diabetes." J Clin Invest 120(1 1 ):
4031 -4039.
Giorgio, V., M. E. Soriano, et al. (2010). "Cyclophilin D in mitochondrial
pathophysiology." Biochim Biophvs Acta 1797(6-7): 1 1 13-1 1 18.
Gomez, L, O. Raisky, et al. (2006). "Link between immune cell infiltration and
mitochondria-induced cardiomyocyte death during acute cardiac graft rejection." Am J Transplant 6(3): 487-495.
Halestrap, A. P. (2010). "A pore way to die: the role of mitochondria in reperfusion injury and cardioprotection." Biochem Soc Trans 38(4): 841 -860.
Ibarra, A. and S. Martinon (2009). "Pharmacological approaches to induce
neuroregeneration in spinal cord injury: an overview." Curr Drug Discov Technol 6(2): 82-90.
King, A. L., T. M. Swain, et al. (2010). "Chronic ethanol consumption enhances sensitivity to Ca(2+)-mediated opening of the mitochondrial permeability transition pore and increases cyclophilin D in liver." Am J Physiol
Gastrointest Liver Physiol 299(4): G954-966.
Kubota, M., T. Kasahara, et al. (2010). "Therapeutic implications of down- regulation of cyclophilin D in bipolar disorder." Int J Neuropsvchopharmacol 13(10): 1355-1368.
Larche, J., S. Lancel, et al. (2006). "Inhibition of mitochondrial permeability
transition prevents sepsis-induced myocardial dysfunction and mortality." J Am Coll Cardiol 48(2): 377-385.
Laukkarinen, J.M., Van Acker, G.J., Weiss, E.R., Steer, M.L., Perides, G. A mouse model of acute biliary pancreatitis induced by retrograde pancreatic duct infusion of Na-taurocholate._Guf 56, 1590-1598 (2007). Lemasters, J. J., A. L. Nieminen, et al. (1998). "The mitochondrial permeability transition in cell death: a common mechanism in necrosis, apoptosis and autophaqy." Biochim Biophvs Acta 1366(1 -2): 177-196.
Lim, S. Y., D. J. Hausenloy, et al. (2010). "Mitochondrial cyclophilin-D as a
potential therapeutic target for post-myocardial infarction heart failure." J
Cell Mol Med.
Malouitre, S., H. Dube, et al. (2009). "Mitochondrial targeting of cyclosporin A
enables selective inhibition of cyclophilin-D and enhanced cytoprotection after glucose and oxygen deprivation." Biochem J 425(1 ): 137-148.
Martin, L. J. (2010). "The mitochondrial permeability transition pore: a molecular target for amyotrophic lateral sclerosis therapy." Biochim Biophvs Acta
1802(1 ): 186-197.
Martin, L. J., B. Gertz, et al. (2009). "The mitochondrial permeability transition pore in motor neurons: involvement in the pathobiology of ALS mice." Exp
Neurol 218(2): 333-346.
Masubuchi, Y., S. Kano, et al. (2006). "Mitochondrial permeability transition as a potential determinant of hepatotoxicity of antidiabetic thiazolidinediones." Toxicology 222(3): 233-239.
Mazzeo, A. T., A. Beat, et al. (2009). "The role of mitochondrial transition pore, and its modulation, in traumatic brain injury and delayed neurodegeneration after TBI." Exp Neurol 218(2): 363-370.
Millay, D. P., M. A. Sargent, et al. (2008). "Genetic and pharmacologic inhibition of mitochondrial-dependent necrosis attenuates muscular dystrophy." Nat Med 14(4): 442-447.
Morrey, J. D., V. Siddharthan, et al. (2010). Discovery and Treatment of
Respiratory Neurological Sequelae in West Nile Virus Infected Hamsters. The Twenty-Third International Conference on Antiviral Research, San- Francisco.
Mukherjee, R., D. N. Criddle, et al. (2008). "Mitochondrial injury in pancreatitis." Cell Calcium 44(1 ): 14-23.
Nakagawa, T., S. Shimizu, et al. (2005). "Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death."
Nature 434(7033): 652-658. Palma, E., T. Tiepolo, et al. (2009). "Genetic ablation of cyclophilin D rescues mitochondrial defects and prevents muscle apoptosis in collagen VI myopathic mice." Hum Mol Genet 18(1 1 ): 2024-2031 .
Piccoli, C, R. Scrima, et al. (2007). "Hepatitis C virus protein expression causes calcium-mediated mitochondrial bioenergetic dysfunction and nitro-oxidative stress." Hepatoloqy 46(1 ): 58-65.
Raisky, O., L. Gomez, et al. (2004). "Mitochondrial permeability transition in
cardiomyocyte apoptosis during acute graft rejection." Am J Transplant
4(7): 1071 -1078.
Ravikumar, R., M. L. McEwen, et al. (2007). "Post-treatment with the cyclosporin derivative, NIM81 1 , reduced indices of cell death and increased the volume of spared tissue in the acute period following spinal cord contusion." J
Neurotrauma 24(10): 1618-1630.
Schinzel, A. C, O. Takeuchi, et al. (2005). "Cyclophilin D is a component of
mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia." Proc Natl Acad Sci U S A 102(34): 12005-12010. Shulga, N. and J. G. Pastorino (2010). "Ethanol sensitizes mitochondria to the permeability transition by inhibiting deacetylation of cyclophilin-D mediated by sirtuin-3." J Cell Sci 123(Pt 23): 41 17-4127.
Soriano, M. E., L. Nicolosi, et al. (2004). "Desensitization of the permeability
transition pore by cyclosporin a prevents activation of the mitochondrial apoptotic pathway and liver damage by tumor necrosis factor-alpha." J Biol
Chem 279(35): 36803-36808.
Sun, L, L. Xiao, et al. (2010). "p66Shc mediates high-glucose and angiotensin II- induced oxidative stress renal tubular injury via mitochondrial-dependent apoptotic pathway." Am J Physiol Renal Physiol 299(5): F1014-1025.
Tazawa, H., C. Fujita, et al. (2009). "Involvement of cyclophilin D in mitochondrial permeability transition induction in intact cells." Arch Biochem Biophys
481 (1 ): 59-64.
Teodoro, J. S., A. P. Rolo, et al. (2008). "Differential alterations in mitochondrial function induced by a choline-deficient diet: understanding fatty liver disease progression." Mitochondrion 8(5-6): 367-376. Tiepolo, T., A. Angelin, et al. (2009). "The cyclophilin inhibitor Debio 025
nornnalizes mitochondrial function, muscle apoptosis and ultrastructural defects in Col6a1 -/- myopathic mice." Br J Pharmacol 157(6): 1045-1052. Trost, L. C. and J. J. Lemasters (1996). "The mitochondrial permeability transition: a new pathophysiological mechanism for Reye's syndrome and toxic liver injury." J Pharmacol Exp Ther 278(3): 1000-1005.
Wissing, E. R., D. P. Millay, et al. (2010). "Debio-025 is more effective than
prednisone in reducing muscular pathology in mdx mice." Neuromuscul
Disord 20(1 1 ): 753-760.
Xia, W. L, Y. Shen, et al. (2005). "Inhibitory effect of cyclosporine A on hepatitis B virus replication in vitro and its possible mechanisms." Hepatobiliary
Pancreat Pis Int 4(1 ): 18-22.
Zorov, D. B., M. Juhaszova, et al. (2009). "Regulation and pharmacology of the mitochondrial permeability transition pore." Cardiovasc Res 83(2): 213-225.

Claims

Claims
1. A cydoundecadepsipeptide compound of Formula (I):
Cyclo-(AXXi-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-D-Hiv-MeLeu-Leu-lvleVal) (I)
1 2 3 4 5 6 7 8 9 10 11 in which:
AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
O-acetyl-MeBmt;
AXX2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH2CH2CH2OH), Nva, 5-hydroxy-Nva;
AXX3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
D-MeSer(OCH2CH2OH), D-MeSer(OCH2CH2NEt2), D-MeAsp(OMe);
AXX4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe, EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone; AXX5 is Leu, Val, lie, Gly, Abu;
AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala; for use in a method for treating and/or preventing cell death associated disorders or diseases, related to the MPTP opening,
and wherein said cydoundecadepsipeptide compound of Formula (I) comply with the two following criteria:
(1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as:
Compound _ of _ formula(I) _ CRC _ IC2 <
CsA _ CRC _ IC2 "
(2) at least three-fold less cytotoxic than CsA, defined as: Compound _ of _ formula(I) _ cell _ count _ IC50 >
CsA _ cell _ count _ IC50
2. The cycloundecadepsipeptide compound of claim 1 , wherein said
cycloundecadepsipeptide compound of Formula (I) is at least ten-fold less cytotoxic than CsA, defined as:
Compound _of _ formula(I) _ cell _ count _ IC50 >
CsA _ cell _ count _ IC50
3. The cycloundecadepsipeptide compound according to any of claims 1 -2, characterised in that, in Formula (I),
AXXi is MeBmt
AXX2 is Abu. Val;
AXX3 is D-MeAla;
AXX4 is Melle, MeVal, EtVal ;
AXX5 is Leu, Val, lie, Gly, Abu;
AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala.
4. The cycloundecadepsipeptide compound according to any of claims 1 -3, wherein said compound is selected from the group comprising
Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal), and Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Gly-D-Hiv-MeLeu-Leu- MeVal).
5. The cycloundecadepsipeptide compound according to any of claims 1 -4, wherein the cell death associated disorders or diseases, related to the MPTP opening, are selected from the group comprising ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
6. The cycloundecadepsipeptide compound according to any of claims 1 -5, wherein the cell death associated disorders or diseases, related to the MPTP opening, are selected from the group comprising ischaemia and ischaemia- reperfusion injuries, muscular dysthrophies, neurodegeneration disorders and diseases, West Nile viral encephalitis, Japanese encephalitis, Chronic hepatitis and cirrhosis induced by Hepatitis C virus, Hepatitis B, delta agent, acute and chronic drug- or toxin-induced hepatotoxicity, alcoholic liver disease, cholestasis, Non Alcoholic Fatty Liver Disease, gastro-intestinal ulcerations caused by
NSAIDs, transplant rejection, peripheral vascular insufficiency, renal insufficiency, obesity, diabetic micro-angiopathy, diabetic nephropathy, Maturity onset diabetes of the young type 4, acute and chronic pancreatitis, septicaemia, multi organ dysfunction syndrome, crush syndrome, congestive heart failure, atherosclerosis, hypertensive heart disease, cancer, bipolar disorder.
7. The cycloundecadepsipeptide compound according to claim 6, wherein ischaemia and ischaemia-reperfusion injuries are selected from the group comprising acute cerebral ischemia, acute stroke, acute myocardial infarction cardiac arrest, myocardial stunning, post-reperfusion arrhythmias, organ failure following therapeutic procedures such as thrombolytic therapy, coronary angioplasty, aortic cross-clamping or coronary bypass surgery, hypovolemic shock, storage/reperfusion injury of transplant organs, multi organ dysfunction syndrome.
8. The cycloundecadepsipeptide compound according to claim 6, wherein muscular dysthrophies are selected from the group comprising Ulrich congenital muscular dysthrophy, Bethlem myopathy, myosclerosis, limb girdle muscular dysthropy, Duchenne muscular dysthrophy, Becker muscular dysthrophy, Emery- Dreifuss syndrome,
9. The cycloundecadepsipeptide compound according to claim 6, wherein neurodegeneration disorders and diseases are selected from the group comprising traumatic brain injury, spinal cord injury, peripheral nerve injury, epilepsy-induced brain injury, amyothrophic lateral sclerosis (ALS), Alzheimer disease, Parkinson disease, multiple sclerosis, Huntington disease, axonal degeneration-induced neuropathic pain, diabetic neuropathy chemotherapy-induced neuropathic pain, or herpes-induced neuropathy.
10. The cycloundecadepsipeptide compound according to any of claims 1 -6, wherein the cell death associated disorders or diseases, related to the MPTP opening, are selected from the group comprising acute and chronic pancreatitis and muscular dystrophy.
1 1 . A pharmaceutical composition for use in a method for treating and/or preventing cell death associated disorders and diseases, related to the MPTP opening, comprising a cycloundecadepsipeptide compound according to any of claims 1 -10, together with one or more pharmaceutically acceptable diluents or carriers. 12. A method for preventing or treating cell death associated disorders or diseases, related to the MPTP opening, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of the
pharmaceutical composition according to claim 1 1 or the cycloundecadepsipeptide compound of Formula (I):
Cyclo-(AXXi-AXX2-AXX3-AXX4-AXX5-AXX6-AXX7-D-Hiv-MeLeu-Leu-MeVal) (I)
1 2 3 4 5 6 7 8 9 10 1 1 in which:
AXXi is MeBmt, 4-fluoro-MeBmt, dihydro-MeBmt, 8-hydroxy-MeBmt;
O-acetyl-MeBmt;
AXX2 is Abu, Val, Thr, Thr(OMe), Thr(OAc), Thr(OCOCH2CH2CH2OH), Nva, 5-hydroxy-Nva;
AXX3 is D-MeAla, D-3-fluoro-MeAla, D-MeSer, D-MeSer(OAc),
D-MeSer(OCH2CH2OH), D-MeSer(OCH2CH2NEt2), D-MeAsp(OMe);
AXX4 is Melle, MeMet, MeVal, MeThr, MeThr(OAc), MeAla, EtVal, Etlle, EtPhe, EtTyr, EtThr(OAc), MeThr(OAc), MeTyr, MeTyr(OAc), MeTyr(OMe), MePhe, MeMet(Ox) wherein the sulphur atom of methionine is sulphoxyde or sulphone; AXX5 is Leu, Val, lie, Gly, Abu; AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala; and wherein said cydoundecadepsipeptide compound of Formula (I) comply with the two following criteria:
(1 ) MPTP inhibitory activity at least equal to half of the CsA activity, defined as:
Compound _ of _ formula(I) _ CRC _ IC2
< 2
CsA _ CRC _ IC2
(2) at least three-fold less cytotoxic than CsA, defined as:
Compound _ of _ formula(I) _ cell _ count _ IC
≥2
CsA cell count IC,n
13. The method of claim 12, wherein said cydoundecadepsipeptide compound of Formula (I) is at least ten-fold less cytotoxic than CsA, defined as:
Compound _of _ formula(I) _ cell _ count _ IC,
> 10
CsA cell count IC^
14. The method of any of claims 12-13, wherein in Formula (I),
AXXi is MeBmt
AXX2 is Abu. Val;
AXX3 is D-MeAla;
AXX4 is Melle, MeVal, EtVal ;
AXX5 is Leu, Val, lie, Gly, Abu;
AXX6 is MeAla, Sar, MeLeu; and
AXX7 is Gly, Ala.
15. The method of any of claims 12-14, wherein the cydoundecadepsipeptide compound of Formula (I) is selected from the group comprising Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Ala-D-Hiv-MeLeu-Leu-MeVal), and Cyclo-(MeBmt-Abu-D-MeAla-MeVal-Leu-MeLeu-Gly-D-Hiv-MeLeu-Leu- MeVal)
16. The method of any of claims 12-15, wherein the cell death associated disorders or diseases, related to the MPTP opening, are selected from the group comprising ischaemia-reperfusion injuries, metabolic and/or oxidative stress disorders, diseases involving loss of cellular calcium homeostasis, age related cellular degeneration, diseases involving toxic and/or infectious pathogens.
17. The method of any of claims 12-16, wherein the cell death associated disorders or diseases, related to the MPTP opening, are selected from the group comprising ischaemia and ischaemia-reperfusion injuries, muscular dysthrophies, neurodegeneration disorders and diseases, West Nile viral encephalitis, Japanese encephalitis, Chronic hepatitis and cirrhosis induced by Hepatitis C virus, Hepatitis B, delta agent, acute and chronic drug- or toxin-induced hepatotoxicity, alcoholic liver disease, cholestasis, Non Alcoholic Fatty Liver Disease, gastro-intestinal ulcerations caused by NSAIDs, transplant rejection, peripheral vascular insufficiency, renal insufficiency, obesity, diabetic micro-angiopathy, diabetic nephropathy, Maturity onset diabetes of the young type 4, acute and chronic pancreatitis, septicaemia, multi organ dysfunction syndrome, crush syndrome, congestive heart failure, atherosclerosis, hypertensive heart disease, cancer, bipolar disorder.
18. The method of any of claims 12-17, wherein the cell death associated disorders or diseases, related to the MPTP opening, are selected from the group comprising acute and chronic pancreatitis and muscular dystrophy.
PCT/IB2011/052096 2010-05-12 2011-05-12 Use of cycloundecadepsipeptide compounds WO2011141891A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IB2010052117 2010-05-12
IBPCT/IB2010/052117 2010-05-12

Publications (1)

Publication Number Publication Date
WO2011141891A1 true WO2011141891A1 (en) 2011-11-17

Family

ID=42330991

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2011/052096 WO2011141891A1 (en) 2010-05-12 2011-05-12 Use of cycloundecadepsipeptide compounds

Country Status (1)

Country Link
WO (1) WO2011141891A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104768968A (en) * 2012-10-02 2015-07-08 西普拉里斯有限公司 Cyclic depsipeptide compounds and their uses

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US5116816A (en) 1987-06-19 1992-05-26 Sandoz Ltd. Cyclosporin peptolides having an α-hydroxycarboxylic acid at position 8
US5525590A (en) 1987-06-17 1996-06-11 Sandoz Ltd. Cyclosporins and their use as pharmaceuticals
US5639724A (en) 1984-07-24 1997-06-17 Sandoz Ltd. Cyclosporin galenic forms
WO2002092033A1 (en) 2001-05-17 2002-11-21 Lg Household & Health Care Ltd. The use of nonimmunosuppressive [$g(g)-hydroxy-n-methyl-l-leucine4] cyclosporin derivatives for treating hair loss
US20030104992A1 (en) 2001-10-12 2003-06-05 Or Yat Sun Cyclosporin analogs for the treatment of autoimmune diseases
US6713454B1 (en) 1999-09-13 2004-03-30 Nobex Corporation Prodrugs of etoposide and etoposide analogs
US20050171328A1 (en) 1996-09-26 2005-08-04 Nektar Therapeutics Al, Corporation Soluble, degradable poly(ethylene glycol) derivatives for conrollable release of bound molecules into solution
WO2006038088A1 (en) 2004-10-01 2006-04-13 Debiopharm Sa Use of [d-meala]3-[etval]4-cyclosporin for the treatment of hepatitis c infection and pharmaceutical composition comprising said [d-meala]3-[etval]4-cyclosporin
WO2006072639A1 (en) 2005-01-10 2006-07-13 Debiopharm S.A. Use of a cyclic undecapeptide for the preparation of a medicament for administration during myocardial ischaemic events
WO2009098533A1 (en) 2008-02-08 2009-08-13 Debiopharm Sa Non -immunosuppressive cyclosporin for the treatment of muscular dystrophy

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US5639724A (en) 1984-07-24 1997-06-17 Sandoz Ltd. Cyclosporin galenic forms
US5525590A (en) 1987-06-17 1996-06-11 Sandoz Ltd. Cyclosporins and their use as pharmaceuticals
US5116816A (en) 1987-06-19 1992-05-26 Sandoz Ltd. Cyclosporin peptolides having an α-hydroxycarboxylic acid at position 8
US20050171328A1 (en) 1996-09-26 2005-08-04 Nektar Therapeutics Al, Corporation Soluble, degradable poly(ethylene glycol) derivatives for conrollable release of bound molecules into solution
US6713454B1 (en) 1999-09-13 2004-03-30 Nobex Corporation Prodrugs of etoposide and etoposide analogs
WO2002092033A1 (en) 2001-05-17 2002-11-21 Lg Household & Health Care Ltd. The use of nonimmunosuppressive [$g(g)-hydroxy-n-methyl-l-leucine4] cyclosporin derivatives for treating hair loss
US20030104992A1 (en) 2001-10-12 2003-06-05 Or Yat Sun Cyclosporin analogs for the treatment of autoimmune diseases
WO2006038088A1 (en) 2004-10-01 2006-04-13 Debiopharm Sa Use of [d-meala]3-[etval]4-cyclosporin for the treatment of hepatitis c infection and pharmaceutical composition comprising said [d-meala]3-[etval]4-cyclosporin
WO2006072639A1 (en) 2005-01-10 2006-07-13 Debiopharm S.A. Use of a cyclic undecapeptide for the preparation of a medicament for administration during myocardial ischaemic events
WO2009098533A1 (en) 2008-02-08 2009-08-13 Debiopharm Sa Non -immunosuppressive cyclosporin for the treatment of muscular dystrophy

Non-Patent Citations (60)

* Cited by examiner, † Cited by third party
Title
"Comprehensive Medicinal Chemistry", vol. 5, 1990, PERGAMON PRESS
"IUPAC Compendium of Chemical Terminology", 1997
"Remington's Pharmaceutical Sciences", 1980
ABRAMOV, A. Y., M. R. DUCHEN: "Mechanisms underlying the loss of mitochondrial membrane potential in glutamate excitotoxicity", BIOCHIM BIOPHVS ACTA, vol. 1777, no. 7-8, 2008, pages 953 - 964
ANGELIN, A., T. TIEPOLO ET AL.: "Mitochondrial dysfunction in the pathogenesis of Ullrich congenital muscular dystrophy and prospective therapy with cyclosporins", PROC NATL ACAD SCI U S A, vol. 104, no. 3, 2007, pages 991 - 996
BAINES, C. P., R. A. KAISER ET AL.: "Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death", NATURE, vol. 434, no. 7033, 2005, pages 658 - 662
BARRIENTOS, S. A., N. W. MARTINEZ ET AL.: "Axonal degeneration is mediated by the mitochondrial permeability transition pore", J NEUROSCI, vol. 31, no. 3, pages 966 - 978
BRUSTOVETSKY, N., T. BRUSTOVETSKY ET AL.: "Increased susceptibility of striatal mitochondria to calcium-induced permeability transition", J NEUROSCI, vol. 23, no. 12, 2003, pages 4858 - 4867
BURKE, A. S., K. REDEKER ET AL.: "Mechanisms of chloroform-induced hepatotoxicity: oxidative stress and mitochondrial permeability transition in freshly isolated mouse hepatocytes", J TOXICOL ENVIRON HEALTH A, vol. 70, no. 22, 2007, pages 1936 - 1945
DEVALARAJA-NARASHIMHA, K., A. M. DIENER ET AL.: "Cyclophilin D deficiency prevents diet-induced obesity in mice", FEBS LETT, vol. 585, no. 4, 2011, pages 677 - 682
DI LISA, F., P. BERNARDI: "A CaPful of mechanisms regulating the mitochondrial permeability transition", J MOL CELL CARDIOL, vol. 46, no. 6, 2009, pages 775 - 780
DU, H., L. GUO ET AL.: "Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer's disease", NAT MED, vol. 14, no. 10, 2008, pages 1097 - 1105
DU, H., S. S. YAN: "Mitochondrial permeability transition pore in Alzheimer's disease: cyclophilin D and amyloid beta", BIOCHIM BIOPHVS ACTA, vol. 1802, no. 1, pages 198 - 204
DU, H., S. S. YAN: "Mitochondrial permeability transition pore in Alzheimer's disease: cydophilin D and amyloid beta", BIOCHIM BIOPHVS ACTA, vol. 1802, no. 1, 2010, pages 198 - 204
ELISEEV, R. A., J. MALECKI ET AL.: "Cyclophilin D interacts with Bcl2 and exerts an anti-apoptotic effect", J BIOL CHEM, vol. 284, no. 15, 2009, pages 9692 - 9699
FORTE, M., B. G. GOLD ET AL.: "Cyclophilin D inactivation protects axons in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis", PROC NATL ACAD SCI U S A, vol. 104, no. 18, 2007, pages 7558 - 7563
FUJIMOTO, K., E. L. FORD ET AL.: "Loss of Nix in Pdx1-deficient mice prevents apoptotic and necrotic beta cell death and diabetes", J CLIN INVEST, vol. 120, no. 11, 2010, pages 4031 - 4039
GIORGIO, V., M. E. SORIANO ET AL.: "Cyclophilin D in mitochondrial pathophysiology", BIOCHIM BIOPHVS ACTA, vol. 1797, no. 6-7, 2010, pages 1113 - 1118
GOMEZ, L., O. RAISKY ET AL.: "Link between immune cell infiltration and mitochondria-induced cardiomyocyte death during acute cardiac graft rejection", AM J TRANSPLANT, vol. 6, no. 3, 2006, pages 487 - 495
H.P. FIEDLER: "Lexikon der Hilfsstoffe fur Pharmazie, Kosmetik und angrenzende Gebiete", 2002
HALESTRAP, A. P.: "A pore way to die: the role of mitochondria in reperfusion injury and cardioprotection", BIOCHEM SOC TRANS, vol. 38, no. 4, 2010, pages 841 - 860
HELV. CHIM. ACTA, vol. 66, 1983, pages 2672 - 702
HOUBEN - WEYL: "Methods of Organic Chemistry", vol. E 22D, 2003, THIEME VERLAG
IBARRA, A., S. MARTINON: "Pharmacological approaches to induce neuroregeneration in spinal cord injury: an overview", CURR DRUCI DISCOV TECHNOL, vol. 6, no. 2, 2009, pages 82 - 90
JEAN FRANGOIS. GUICHOU: "De nouveaux analogues de Cyclosporine A comme agent anti-VIH-1", PHD THESIS, 2001, pages 121 - 122
KING, A. L., T. M. SWAIN ET AL.: "Chronic ethanol consumption enhances sensitivity to Ca(2+)-mediated opening of the mitochondrial permeability transition pore and increases cyclophilin D in liver", AM J PHYSIOL GASTROINTEST LIVER PHVSIOL, vol. 299, no. 4, 2010, pages 954 - 966
KUBOTA, M., T. KASAHARA ET AL.: "Therapeutic implications of down- regulation of cyclophilin D in bipolar disorder", INT J NEUROPSVCHOPHARMACOL, vol. 13, no. 10, 2010, pages 1355 - 1368
LARCHE, J., S. LANCEL ET AL.: "Inhibition of mitochondrial permeability transition prevents sepsis-induced myocardial dysfunction and mortality", J AM COLL CARDIOL, vol. 48, no. 2, 2006, pages 377 - 385
LAUKKARINEN, J.M., VAN ACKER, G.J., WEISS, E.R., STEER, M.L., PERIDES, G.: "A mouse model of acute biliary pancreatitis induced by retrograde pancreatic duct infusion of Na-taurocholate", GUT, vol. 56, 2007, pages 1590 - 1598
LEMASTERS, J. J., A. L. NIEMINEN ET AL.: "The mitochondrial permeability transition in cell death: a common mechanism in necrosis, apoptosis and autophagy", BIOCHIM BIOPHVS ACTA, vol. 1366, no. 1-2, 1998, pages 177 - 196
LIM, S. Y., D. J. HAUSENLOY ET AL.: "Mitochondrial cyclophilin-D as a potential therapeutic target for post-myocardial infarction heart failure", J CELL MOL MED, 2010
LOGUIDICE, RAMIREZ-ALCANTRA ET AL., TOXICOL SCI., 2010
MALOUITRE, S., H. DUBE ET AL.: "Mitochondrial targeting of cyclosporin A enables selective inhibition of cyclophilin-D and enhanced cytoprotection after glucose and oxygen deprivation", BIOCHEM J, vol. 425, no. 1, 2009, pages 137 - 148
MARTIN, L. J., B. GERTZ ET AL.: "The mitochondrial permeability transition pore in motor neurons: involvement in the pathobiology of ALS mice", EXO NEUROL, vol. 218, no. 2, 2009, pages 333 - 346
MARTIN, L. J.: "The mitochondrial permeability transition pore: a molecular target for amyotrophic lateral sclerosis therapy", BIOCHIM BIOPHVS ACTA, vol. 1802, no. 1, 2010, pages 186 - 197
MASUBUCHI, Y., S. KANO ET AL.: "Mitochondrial permeability transition as a potential determinant of hepatotoxicity of antidiabetic thiazolidinediones", TOXICOLOGY, vol. 222, no. 3, 2006, pages 233 - 239
MAZZEO, A. T., A. BEAT ET AL.: "The role of mitochondrial transition pore, and its modulation, in traumatic brain injury and delayed neurodegeneration after TBI", EXP NEUROL, vol. 218, no. 2, 2009, pages 363 - 370
MILLAY, D. P., M. A. SARGENT ET AL.: "Genetic and pharmacologic inhibition of mitochondrial-dependent necrosis attenuates muscular dystrophy", NAT MED, vol. 14, no. 4, 2008, pages 442 - 447
MORREY, J. D., V. SIDDHARTHAN ET AL.: "Discoverv and Treatment of Respiratorv Neuroloqical Sequelae in West Nile Virus Infected Hamsters", THE TWENTY-THIRD INTERNATIONAL CONFERENCE ON ANTIVIRAL RESEARCH, 2010
MUKHERJEE, R., D. N. CRIDDLE ET AL.: "Mitochondrial injury in pancreatitis", CELL CALCIUM, vol. 44, no. 1, 2008, pages 14 - 23
NAKAGAWA, T., S. SHIMIZU ET AL.: "Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death", NATURE, vol. 434, no. 7033, 2005, pages 652 - 658
PALMA, E., T. TIEPOLO ET AL.: "Genetic ablation of cyclophilin D rescues mitochondrial defects and prevents muscle apoptosis in collagen VI myopathic mice", HUM MOL GENET, vol. 18, no. 11, 2009, pages 2024 - 2031
PICCOLI, C., R. SCRIMA ET AL.: "Hepatitis C virus protein expression causes calcium-mediated mitochondrial bioenergetic dysfunction and nitro-oxidative stress", HEPATOLOGY, vol. 46, no. 1, 2007, pages 58 - 65
RAISKY, 0., L. GOMEZ ET AL.: "Mitochondrial permeability transition in cardiomyocyte apoptosis during acute graft rejection", AM J TRANSPLANT, vol. 4, no. 7, 2004, pages 1071 - 1078
RAVIKUMAR, R., M. L. MCEWEN ET AL.: "Post-treatment with the cyclosporin derivative, NIM811, reduced indices of cell death and increased the volume of spared tissue in the acute period following spinal cord contusion", J NEUROTRAUMA, vol. 24, no. 10, 2007, pages 1618 - 1630
RICH, D.H. ET AL.: "Comparative studies of the coupling of N-methylated, sterically hindered amino acids during SPPS", TETR. LETTERS, vol. 35, 1994, pages 5981 - 5984
SCHINZEL, A. C., O. TAKEUCHI ET AL.: "Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia", PROC NATL ACAD SCI U S A, vol. 102, no. 34, 2005, pages 12005 - 12010
SHABAZ ET AL., J HYPERTENS., vol. 28, no. 1, pages 25 - 32
SHULGA, N., J. G. PASTORINO: "Ethanol sensitizes mitochondria to the permeability transition by inhibiting deacetylation of cyclophilin-D mediated by sirtuin-3", J CELL SCI, vol. 123, 2010, pages 4117 - 4127
SORIANO, M. E., L. NICOLOSI ET AL.: "Desensitization of the permeability transition pore by cyclosporin a prevents activation of the mitochondrial apoptotic pathway and liver damage by tumor necrosis factor-alpha", J BIOL CHEM, vol. 279, no. 35, 2004, pages 36803 - 36808
SUN, L., L. XIAO ET AL.: "p66Shc mediates high-glucose and angiotensin 11- induced oxidative stress renal tubular injury via mitochondrial-dependent apoptotic pathway", AM J PHVSIOL RENAL PHVSIOL, vol. 299, no. 5, 2010, pages 1014 - 1025
TAZAWA, H., C. FUJITA ET AL.: "Involvement of cyclophilin D in mitochondrial permeability transition induction in intact cells", ARCH BIOCHEM BIOPHVS, vol. 481, no. 1, 2009, pages 59 - 64
TEODORO, J. S., A. P. ROLO ET AL.: "Differential alterations in mitochondrial function induced by a choline-deficient diet: understanding fatty liver disease progression", MITOCHONDRION, vol. 8, no. 5-6, 2008, pages 367 - 376
TIEPOLO, T., A. ANGELIN ET AL.: "The cyclophilin inhibitor Debio 025 normalizes mitochondrial function, muscle apoptosis and ultrastructural defects in Col6a1-/- myopathic mice", BR J PHARMACOL, vol. 157, no. 6, 2009, pages 1045 - 1052
TROST, L. C., J. J. LEMASTERS: "The mitochondrial permeability transition: a new pathophysiological mechanism for Reye's syndrome and toxic liver injury", J PHARMACOL EXP THER, vol. 278, no. 3, 1996, pages 1000 - 1005
TSAO ET AL., J GEN VIROL., vol. 89, August 2008 (2008-08-01), pages 1930 - 41
WENGER, HELV. CHIM. ACTA, vol. 67, 1984, pages 502 - 25
WISSING, E. R., D. P. MILLAY ET AL.: "Debio-025 is more effective than prednisone in reducing muscular pathology in mdx mice", NEUROMUSCUL DISORD, vol. 20, no. 11, 2010, pages 753 - 760
XIA, W. L., Y. SHEN ET AL.: "Inhibitory effect of cyclosporine A on hepatitis B virus replication in vitro and its possible mechanisms", HEPATOBILIARV PANCREAT DIS INT, vol. 4, no. 1, 2005, pages 18 - 22
ZOROV, D. B., M. JUHASZOVA ET AL.: "Regulation and pharmacology of the mitochondrial permeability transition pore", CARDIOVASC RES, vol. 83, no. 2, 2009, pages 213 - 225

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104768968A (en) * 2012-10-02 2015-07-08 西普拉里斯有限公司 Cyclic depsipeptide compounds and their uses
US9382295B2 (en) 2012-10-02 2016-07-05 Cypralis Limited Cyclic depsipeptide compounds and their uses
US9895412B2 (en) 2012-10-02 2018-02-20 Cypralis Limited Cyclic depsipeptide compounds and their uses

Similar Documents

Publication Publication Date Title
US6927208B1 (en) Cyclosporin with improved activity profile
AU2019203671B2 (en) Cyclotides as immunosuppressive agents
AU2009312535B2 (en) Cycloundecadepsipeptide compounds and use of said compounds as a medicament
KR102020886B1 (en) Polymyxin derivative and uses thereof
CN111704653A (en) Inhibitor polypeptide compounds targeted to fibronectin derived peptides and uses thereof
KR20140041689A (en) Beta - hairpin peptidomimetics as cxc4 antagonists
CH637111A5 (en) POLYPEPTIDE COMPOUNDS WITH THERMAL OR ANTAGONIST ACTIVITY AND METHODS OF SYNTHESIS THEREOF.
SK285332B6 (en) Use of pentosan polysulphate for the manufacture of a pharmaceutical composition for preventing or reducing the development of nephrotoxicity or renal dysfunction and pharmaceutical composition
WO2011141891A1 (en) Use of cycloundecadepsipeptide compounds
CA2282720C (en) Dolastatin-15 derivatives in combination with taxanes
KR101633772B1 (en) Non-immunosuppressive Cyclosporin for the treatment of a muscular dystrophy
Jędrzejczak et al. Synthesis and biological activity of cyclolinopeptide A analogues modified with γ3-bis (homophenylalanine)

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: 11726954

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11726954

Country of ref document: EP

Kind code of ref document: A1