WO2012156310A1 - Novel peptides - Google Patents

Novel peptides Download PDF

Info

Publication number
WO2012156310A1
WO2012156310A1 PCT/EP2012/058770 EP2012058770W WO2012156310A1 WO 2012156310 A1 WO2012156310 A1 WO 2012156310A1 EP 2012058770 W EP2012058770 W EP 2012058770W WO 2012156310 A1 WO2012156310 A1 WO 2012156310A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
drug
bone
disorders
diseases
Prior art date
Application number
PCT/EP2012/058770
Other languages
French (fr)
Inventor
Lars Sävendahl
Farasat Zaman
Emma Eriksson
Original Assignee
Saevendahl Lars
Farasat Zaman
Emma Eriksson
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 Saevendahl Lars, Farasat Zaman, Emma Eriksson filed Critical Saevendahl Lars
Publication of WO2012156310A1 publication Critical patent/WO2012156310A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein

Definitions

  • the present invention relates to the field of medical therapeutics, and more particularly to novel therapeutic peptides, designated mutant humanin-like peptides (MHLPs) and fragments, analogs, derivatives, and variants thereof, which have beneficial activities on cartilage tissues and/or bone tissues, including prevention of negative effects of disorders/drugs on cartilage tissues and/or bone tissues by treatment with for example, but not limited to, glucocorticoids.
  • MHLPs mutant humanin-like peptides
  • the MHLPs are also useful in the prevention and/or treatment of cancers, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, heart and vascular diseases and metabolic diseases.
  • the present invention relates to a number of peptides named "mutant humanin-like peptides" with capacity to regulate biological pathways.
  • endochondral bone formation a process where resting zone chondrocytes are recruited to start active proliferation and then undergo differentiation, followed by apoptosis and later mineralization.
  • the balance between proliferation and differentiation is a crucial regulatory step controlled by various growth factors/hormones acting in both endocrine and
  • Glucocorticoids are widely used as a chemotherapy in cancers, antiinflammatory and immunosuppressive drugs both in children and adults with chronic diseases.
  • GCs Glucocorticoids
  • recent studies have revealed that long-term high- dose GC treatment often leads to growth failure in treated children, an effect which has been shown to be mediated through a negative effect on growth plate chondrocytes (Baron, Huang et al. 1992) (Loeb 1976).
  • Dexamethasone (Dexa) is a widely used glucocortcoid used in children and adults to treat different diseases such as, asthma, inflammation and cancer. Beside the positive effects of Dexa on inflammation and cancer, its long-term use has been linked to negative side effects on bone. Previous studies (pre-clinical and clinical) show that long-term use of Dexa can cause bone growth retardation in children and osteoporosis both in children and adults (Demoly 2008). It is well known that Dexa can alter the processes of proliferation and differentiation in chondrocytes and other cells.
  • CT chemotherapy
  • hepatoblastoma hepatoblastoma, head and neck cancers, nasopharyngeal carcinoma, esophageal/gastric junction adenocarcinoma, glaucoma and skin cancer.
  • 5-FU has been reported to suppress bone growth, an effect associated with rapid and significant suppression of cell proliferation in the growth plate and metaphysis and induction of apoptosis in the metaphysis (Xian, Howarth et al. 2004).
  • Other chemotherapeutic drugs such as etoposide and cyclophosphamide, administered alone or in combination, have been reported to damage the growth plate by causing structural and cellular changes in the growth plate cartilage (Xian, Cool et al. 2007).
  • proteasome inhibitors including Bortezomib (Velcade, PS-341 ), belong to another novel and promising group of CT that has past phase I clinical trials (Blaney, Bernstein et al. 2004) and are now under phase II clinical trials in pediatric cancers.
  • phase I clinical trials Blaney, Bernstein et al. 2004
  • secondary effects on normal bystander tissues of primary life saving modalities are so far unknown.
  • Bortezomib target essential cell populations within the growth plate causing permanent growth retardation and chondrocyte cell death, both in vitro and in vivo (Zaman, Menendez-Benito et al. 2007).
  • HN humanin
  • AD Alzheimer's disease
  • Mamiya and Ukai 2001 chemical-induced damage
  • HN is both an intracellular and secreted protein.
  • HN cerebrospinal fluid
  • HN has also been reported as an antagonist of Bax and Bid that induces survival in cancer cells (Guo, Zhai et al. 2003), cell survival by binding to putative cell-surface receptors (Ying, Iribarren et al. 2004) and as an IGFBP-3 partner that antagonizes the apoptotic actions of IGFBP-3 on cancer cells (Ikonen, Liu et al. 2003).
  • IGFBP-3 is one of a number of peptides including insulin, leptin, adiponectin, and resistin that have been shown to act in the central nervous system to regulate glucose metabolism (Muse, Lam et al. 2007) (Obici, Zhang et al. 2002).
  • IGFBP-3 is an HN partner that has pro- diabetogenic hypothalamic actions that are modulated by IGF-I (Muzumdar and Rao 2006).
  • HN has been reported as a wide spectrum survival factor (Nishimoto, Matsuoka et al. 2004), but its exact mechanism of action remains unclear.
  • HNG humanin-Gly14
  • HNG-F6A non-IGFBP-3 binding
  • colivelin hybrid peptide containing partial sequences of HN and ADNF9
  • HN and its analogs and derivatives have shown therapeutic potential for an array of diseases including Alzheimer's disease (AD), diabetes and kidney failure (Matsuoka and Hashimoto 2010) (Matsuoka 2009) (Xu, Chua et al. 2006) (Hoang, Park et al. 2010) (Singh and Mascarenhas 2008).
  • Alzheimer's disease-related neurotoxicity functional potentiation by isomerization and dimerization.” J Neurochem 85(6): 1521 -1538.
  • the present inventors have identified a number of mutant humanin-like peptides (MHLPs) having the capacity to regulate biological pathways, such as inflammation and exert an inhibitory effect on the death of chondrocyte cells.
  • the peptides are named "mutant humanin-like peptides" (MHLPs) and the present invention relates to the peptides themselves and their use in the treatment and/or prevention of bone- or cartilage disorders/diseases, bone fractures, skeletal chondrodysplasias, cancers, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, infertility, muscular disoders, aging, skin diseases, heart and vascular diseases and metabolic diseases.
  • a number of peptides have been designed, synthesized and tested for their capacity to modulate biological pathways in a cell model system. It was identified that chondrocyte cell death (which is the main cause of bone growth disorders under various conditions) is completely blocked by MHLPs, suggesting that the MHLPs according to the invention affect biological pathways. For instance, chondrocytes were treated with high dose glucocorticoids (GCs) and subsequent MHLP treatment completely rescued chondrocytes from undergoing apoptosis.
  • GCs glucocorticoids
  • the present findings show the use of MHLPs alone or in combination with chemotherapy completely prevents the negative effects of chemotherapy, wihout interfering with the primary effects of the drugs is of considerable clinical importance for their broader application.
  • a peptide comprising an amino acid sequence X1APX2X3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 ) wherein
  • Xi is selected from M, L, A, S, G, and P; X2 is selected from R, K, A, G, and E; X3 is selected from G, V, Y, and A; and fragments, analogs, derivatives, and variants thereof, provided that peptides wherein Xi is M; X 2 is R and X3 is G simultaneously, are excluded.
  • a peptide comprising an amino acid sequence
  • Xi is selected from M, L, A, S, G, and P; X2 is selected from R, K, A, G, and E; X3 is selected from G, V, Y, and A; provided that peptides wherein Xi is M; X 2 is R and X3 is G simultaneously, are excluded.
  • a peptide comprising an amino acid sequence
  • Xi is selected from L, A, S, G, and P.
  • a peptide comprising an amino acid sequence
  • X 2 is selected from K, A, G, and E.
  • a peptide comprising an amino acid sequence
  • X 3 is selected from V, Y, and A.
  • a peptide comprising:5 to SEQ ID NO:17; LAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :5);
  • AAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :6);
  • SAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO: ⁇ n
  • GAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :8);
  • PAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :9)
  • MAPAGFSCLLLLTGEIDLPVKRRA SEQ ID NO: :1 1 );
  • MAPEGFSCLLLLTGEIDLPVKRRA SEQ ID NO: 12
  • MAPRGVSCLLLLTGEIDLPVKRRA SEQ ID NO: 14
  • MAPRGYSCLLLLTGEIDLPVKRRA SEQ ID NO: 15
  • MAPRGASCLLLLTGEIDLPVKRRA SEQ ID NO: :16
  • MAPRGGSCLLLLTGEIDLPVKRRA SEQ ID NO:17.
  • a peptide said peptide being LAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:5).
  • peptide in another embodiment, there is provided a peptide, said peptide being AAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:6).
  • peptide in another embodiment, there is provided a peptide, said peptide being SAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:7).
  • a peptide said peptide being GAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:8). In another embodiment of this aspect, there is provided a peptide, said peptide being PAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:9).
  • peptide in another embodiment, there is provided a peptide, said peptide being MAPKGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:10).
  • a peptide said peptide being MAPAGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 1 ). In another embodiment of this aspect, there is provided a peptide, said peptide being MAPEGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:12).
  • peptide in another embodiment, there is provided a peptide, said peptide being MAPGGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:13).
  • peptide in another embodiment, there is provided a peptide, said peptide being MAPRGVSCLLLLTGEIDLPVKRRA (SEQ ID NO:14).
  • peptide in another embodiment, there is provided a peptide, said peptide being MAPRGYSCLLLLTGEIDLPVKRRA (SEQ ID NO:15).
  • a peptide said peptide being MAPRGASCLLLLTGEIDLPVKRRA (SEQ ID NO:16). In another embodiment of this aspect, there is provided a peptide, said peptide being MAPRGGSCLLLLTGEIDLPVKRRA (SEQ ID NO:17).
  • a peptide of the invention for use in therapy.
  • a pharmaceutical composition comprising a peptide of the invention, together with
  • a peptide of the invention for use in in the prevention and/or treatment of bone- or cartilages disorders/diseases, cancer, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes,
  • neurodegenerative diseases bone fractures, skeletal chondrodysplasias, infectious diseases, lung diseases, infertility, muscular disoders, aging, skin diseases, heart and vascular diseases and metabolic diseases.
  • a peptide of the invention for use in the prevention and/or treatment of bone- or cartilage disorders.
  • Said peptide may improve bone growth.
  • the peptide may improve bone growth by itself targeting the Wnt-signalling or beta-catenin pathway or together with growth hormone (GH) or insulin-like growth factor I (IGF-I). Further, said peptide may improve bone healing. Further, said peptide may prevent development of osteoporosis.
  • Said bone- or cartilage disorder may be a either a primary or secondary bone- or cartilage disorder. Further, said peptide may improve skin healing.
  • a peptide of the invention for use in the prevention and/or treatment of bone- or cartilage disorders wherein said disorder is drug-induced.
  • Said drug-induced disorder may have arisen from drugs used in the treatment of rheumatological disorders, respiratory disorders, gastrointestinal disorders, cardiovascular disorders, endocrinological disorders, cancer, neurodegenerative disorders, kidney disorders, liver disorders, dermatologic disorders, allergic disorders, inflammatory disorders, metabolic disorders, undesired immune response, undesired inflammatory response, obesity or diabetes.
  • a peptide of the invention for use in the prevention and/or treatment of bone- or cartilage disorders wherein said disorder is drug-induced bone growth impairment, short stature or osteoporosis.
  • Said drug may be an anti-inflammatory drug, such as a glucocorticoid drug.
  • Said glucocorticoid drug may be selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate,
  • budesonide ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide,
  • fluorometholone clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone, methylprednisolone
  • aceponate prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate,
  • glucocorticoid drug is dexamethasone.
  • a peptide of the invention for use in the prevention and/or treatment of bone- or cartilage disorders wherein said disorder is drug-induced by an anti-cancer drug.
  • Said anti-cancer drug may be a proteasome inhibitor, such as bortezomib,
  • a peptide of the invention for use in the prevention and/or treatment of bone- or cartilage disorders, wherein said peptide is administered in combination with another drug known to induce a bone- or cartilage disorder.
  • said another drug is selected from an antiinflammatory drug, a selective estrogen receptor modulator drug, an anti- androgen drug, an aromatase inhibitor drug and an anti-cancer drug.
  • said another drug is an anti-inflammatory drug, it may be a
  • glucocorticoid drug such as hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone,
  • methylprednisolone aceponate prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone
  • alclometasone fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal or mometasone furoate.
  • said glucocorticoid drug is dexamethasone.
  • said another drug is an anti-cancer drug
  • it may be a proteasome inhibitor, such as bortezomib, MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS, ZLVS.
  • said proteasome inhibitor is bortezomib.
  • said another drug may be selected from leuprolide, buserelin, nafarelin, histrelin, goserelin, deslorelin, cetrorelix, ganirelix, abarelix, degarelix, letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide, testolactone, tamoxifen, afimoxifene, 4- hydroxytamoxifen, arzoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, tamoxifentoremifene, spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride, dutasteride, cyclophosp
  • dexamethasone acetate dexamethasone sodium phosphate, dexasone, dexrazoxane, dhad, die, diodex, docetaxel, doxil, doxorubicin, doxorubicin liposomal, droxia, duralone, efudex, eligard, ellence, eloxatin, elspar, emcyt, epirubicin, epoetin alfa, erbitux, erlotinib, erwinia L-asparaginase,
  • a method of preventing and/or treating bone- or cartilage disorders comprising administering a therapeutically effective amount of a peptide of the present invention.
  • Said peptide may improve bone growth. Further, said peptide may improve bone healing. Further, said peptide may prevent development of osteoporosis.
  • Said bone- or cartilage disorder may be a either a primary or secondary bone- or cartilage disorder. Further, said peptide may improve skin healing.
  • a therapeutically effective amount of a peptide of the present invention wherein said disorder is drug-induced.
  • Said drug-induced disorder may have arisen from drugs used in the treatment of rheumatological disorders, respiratory disorders, gastrointestinal disorders, cardiovascular disorders, endocrinological disorders, cancer, neurodegenerative disorders, kidney disorders, liver disorders, dermatologic disorders, allergic disorders, inflammatory disorders, metabolic disorders, undesired immune response, undesired inflammatory response, obesity or diabetes.
  • Said drug may be an anti-inflammatory drug, such as a glucocorticoid drug.
  • Said glucocorticoid drug may be selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate,
  • budesonide ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide,
  • fluorometholone clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone, methylprednisolone
  • aceponate prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate,
  • glucocorticoid drug is dexamethasone.
  • Said anti-cancer drug may be a proteasome inhibitor, such as bortezomib, MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS.
  • said proteasome inhibitor is bortezomib.
  • a method of preventing and/or treating bone- or cartilage disorders comprising administering a therapeutically effective amount of a peptide of the present invention, wherein said peptide is administered in combination with another drug known to induce a bone- or cartilage disorder.
  • said another drug is selected from an anti- inflammatory drug, a selective estrogen receptor modulator drug, an anti- androgen drug, an aromatase inhibitor drug and an anti-cancer drug.
  • said another drug is an anti-inflammatory drug, it may be a
  • glucocorticoid drug such as hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone,
  • methylprednisolone aceponate prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate, beclomethasone monopropionate, paramethasone,
  • alclometasone fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal or mometasone furoate.
  • said glucocorticoid drug is dexamethasone.
  • said another drug is an anti-cancer drug
  • it may be a proteasome inhibitor, such as bortezomib, MLN9708, MLN273, MLN519, NPI-0052,
  • said proteasome inhibitor is bortezomib.
  • said another drug may be selected from leuprolide, buserelin, nafarelin, histrelin, goserelin, deslorelin, cetrorelix, ganirelix, abarelix, degarelix, letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide, testolactone, tamoxifen, afimoxifene, 4- hydroxytamoxifen, arzoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, tamoxifentoremifene, spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride, dutasteride, cyclophosp
  • dexamethasone acetate dexamethasone sodium phosphate, dexasone, dexrazoxane, dhad, die, diodex, docetaxel, doxil, doxorubicin, doxorubicin liposomal, droxia, duralone, efudex, eligard, ellence, eloxatin, elspar, emcyt, epirubicin, epoetin alfa, erbitux, erlotinib, erwinia L-asparaginase,
  • osteoarthritis or osteoarthrosis Another such a condition is delayed fracture healing which if untreated can lead to severe consequences including pseudoarthrosis.
  • a primary bone or cartilage disorder growth impairment, short stature, arthritis, osteoarthritis, rheumatoid arthritis, psoriasis arthritis, ankylosing spondylitis, osteoarthrosis, osteomyelitis fibrous dysplasia, fibrodysplasia ossificans, craniosynostosis, metabolic bone disease, osteitis deformans, osteogenesis imperfecta, scoliosis, leg length difference, bone dysplasia, osteopetrosis, osteomalacia, osteopenia, osteoporosis, osteitis fibrosa cystic, osteochondritis, osteonecrosis, osteosarcoma, bone tumor, osteochondroma, osteochondropathy, chondropathy, chondrodysplasia, achondrodysplasia, hypochondrodysplasia, chondrodystrophy,
  • a secondary bone or cartilage disorder is meant a disorder which is a consequence of another disorder not primarily involving bone or cartilage tissues.
  • Another disorder includes Cushing ' s syndrome which is caused by increased Cortisol production.
  • undesired immune response or undesired inflammatory response is meant responses which have undesired effects on bone or cartilage tissues.
  • Rheumatological disorders include, but are not limited to, reumatoid arhtritis, systemic lupus erythematosus, vasculitis, periarhteritis nodosa and
  • Respiratory disorders include, but are not limited to, asthma bronchiale, chronic obstructive lung disease and bronchiolitis obliterans.
  • Gastrointestinal disorders include, but are not limited to, colitis ulcerosa, Crohn ' s disease and pancreatitis.
  • Cardiovascular disorders include, but are not limited to, myocarditis, ischemia, hemolytic anemia and granulocytopenia.
  • Endocrinological disorders include, but are not limited to, adrenal disorder, glucocorticoid deficiency, congenital adrenal hyperplasia,
  • Cancer includes, but is not limited to, acute myeloid leukemia, multiple myeloma and lymphoma.
  • Neurodegenerative disorders include, but are not limited to, multiple sclerosis and encephalitis.
  • Kidney disorders include, but are not limited to,
  • Liver disorders include, but are not limited to, hepatitis and cholangitis.
  • Dermatologic disorders include, but are not limited to, psoriasis, exzema, myositis, dermatomyositis and dermatitis.
  • the disorder is drug-induced from a GnRH agonist drug, such as leuprolide, buserelin, nafarelin, histrelin, goserelin and deslorelin.
  • the disorder is drug-induced from a GnRH antagonist drug, such as cetrorelix, ganirelix, abarelix and degarelix.
  • the disorder is drug-induced from a selective estrogen receptor modulator drug, such as tamoxifen, afimoxifene, 4-hydroxytamoxifen, arzoxifene, avalycation, gammoxifene, femarelle, lasofoxifene, ormeloxifene, raloxifene and tamoxifentoremifene.
  • a selective estrogen receptor modulator drug such as tamoxifen, afimoxifene, 4-hydroxytamoxifen, arzoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene and tamoxifentoremifene.
  • the disorder is drug-induced from an anti- androgen drug, such as spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride and dutasteride.
  • an anti- androgen drug such as spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride and dutasteride.
  • the disorder is drug-induced from an aromatase inhibitor drug, such as letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide and testolactone.
  • an aromatase inhibitor drug such as letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide and testolactone.
  • Figure 1 Provides micrographs and data of potential protective effects of HNG and MHLPs of the invention on skin atrophy in mice (see Example 5 for further explanation).
  • compositions of the invention may be administered by means which include but are not limited to intravenous, oral, subcutaneous, intra-arterial, intramuscular, intracardial, intraspinal, intrathoracic, intraperitoneal, intraventricular, sublingual, transdermal, intranasal, and inhalation.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs as amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as naturally occurring amino acids that are later modified.
  • Amino acid analogs are compounds that have the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that differs from the general chemical structure of an amino acid, but that functions similar to a naturally occurring amino acid.
  • Endogenous refers to a protein, nucleic acid, lipid or other biomolecule produced or originating within the body or within cells, organs, tissues of the body of a subject.
  • Exogenous refers to a protein, nucleic acid, lipid, or other biomolecule originating outside the body of a subject.
  • Cartilage tissues refers to all tissues in the body where cartilage cells (chondrocytes) are the major cellular components. This includes but is not limited to joint cartilage, growth plate cartilage and intervertebral disc cartilage. “Bone tissues” refers to all tissues in the body where bone cells (osteocytes, osteoblasts and osteoclasts) are important cellular components. This includes but is not limited to long bones and vertebrae
  • Bone growth refers to a series of co-ordinated actions which take place at the epiphyseal growth plate of long bones by balanced cycle of cartilage growth, formation of matrix, calcification of cartilage that acts as a scaffold for bone formation and modelling (where bone is being continuously resorbed and replaced by new bone). Bone modelling is highly active in
  • Bone growth disorders occur when there is disruption of the normal cellular activity of chondrocytes (growth plate/articular) and/or bone cells.
  • “Bone metabolism” refers to a process involving bone modeling and bone remodeling where cells produce the substances and energy needed for their survival.
  • bone remodeling bone resorption by osteoclasts is followed by bone formation by osteoblasts.
  • Bone remodeling does not result in changes of bone shape but helps in repairing of microdamage.
  • modeling is the formation of new bone by osteoblasts at locations different from the sites of bone resorption by osteoclasts and results in bone growth.
  • Longitudinal bone growth refers endochondral bone formation that occurs in the growth plates (a thin layer of cartilage located on each end of long boneswhere in a series of co-ordinated steps where resting zone/stem-like chondrocytes are recruited to start active proliferation and then undergo differentiation, followed by apoptosis and later mineralization resulting in increased bone length.
  • Drug-induced longitudinal bone growth impairement refers to a condition when drugs given to the patient/subject (mammal, children, adolescents, adults, animal) to treat different diseases but as a side effect they also alter/inhibit the normal process of bone elongation causing longitudinal bone growth impairment/retardation.
  • “Drug-induced” refers to an effect which is primarily due to drug(s) given to patient/subject under different disease conditions. “Fragment” refers to a small part synthesised/produced/broken off/or detached from its original place.
  • Derivative refers to a substance derived/produced/obtained either directly or by modification or partial substitution.
  • Catch-up growth refers to acceleration of the growth rate in infants or young children above the limits of normal for age after a transient period of growth inhibition/impairment; it can be complete or incomplete.
  • peptide refers to any of various natural or synthetic compounds containing at least two or more amino acids linked by the carboxyl group of one amino acid to the amino group of another.
  • Polypeptide refers to chains of amino acids, and “proteins” are made up of one or more polypeptide molecules.
  • Polynucleotide refers to a polymeric form of nucleotides af at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, including single and double stranded forms of DNA.
  • polypeptide refers to a polymer (large molecules composed of repeating structural units typically connected by covalent chemical bonds) formed from the linking, in a defined order, of a- amino acids.
  • the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • endogenous expression of humanin in cartilage tissues and/or bone tissues refers to the local production and/or secretion of humanin within cartilage tissues and/or bone tissues.
  • agent that affects the local production and/or expression of humanin or another peptide expressed by the humanin gene refers to an agent that for example, but not limited to, triggers endogenous production of humanin or humanin like peptides/derivatives/analogs in cartilage tissues and/or bone tissues by the use of siRNA, miRNA, shRNA (plasmid and lentiviral).
  • MicroRNA refers to a class of small RNA molecules, about 21 nucleotides in length that regulate gene expression in a variety of ways.
  • RNA interference refers to a biological mechanism by which double- stranded RNA (dsRNA) induces gene silencing by targeting complementary mRNA for degradation.
  • siRNA small interfering RNA
  • RNAi RNA interference
  • short hairpin RNA refers to a sequence of RNA that makes a tight hairpin turn that can be used to silence gene expression via RNA
  • “Disorder that affects the endogenous expression of humanin in cartilage tissues and/or bone tissues negatively influencing cartilage tissues and/or bone tissues” refers to the presence, absence, increased or decreased level of humanin in cartilage tissues and/or bone tissues with or without treatment causing cartilage tissue and/or bone tissue disorders. For example, a patient suffering from increased or decreased and/or insufficient levels of locally produced humanin in cartilage tissues and/or bone tissues resulted from with/without treatment of any disease.
  • mutant-humanin-like peptides and derivates, analogs and variants thereof is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 85%, 95% or 98% identical to amino acid sequence of SEQ ID NO: 5-17.
  • mutant-humanin-like peptides (MHLPs) and dehvates, analogs and variants thereof has an amino acid sequence which is
  • sequence of the corresponding human humanin in that the sequence is at least about 30%-98% or more identical to the sequence of a reference sequence, such as the corresponding
  • amino acid(s) substitution is one in which one or more amino acid reidue(s) is replaced with an amino acid residue.
  • Families of amino acid residues have been defined in the art and include amino acids with basic side chains (e.g lysine, arginine, histidine), acidic side chains (e.g. aspartic acid, glutamic acid), uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g.
  • HLPs humanin and humanin-like peptides
  • dehvates, analogs and variants thereof can typically be replaced with another amino acid residue, preferably from the same side chain family.
  • non-essential amino acid residue is a residue that can be altered from the original sequence (e.g. a sequence of SEQ ID NO: 5-17) without abolishing or substantially altering the therapeutic activity of the peptide, whereas an "essential" amino acid residue is a residue that cannot be altered without introducing such a change.
  • nucleic acids or polypeptide sequences refers to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same when compared and aligned for maximum correspondence over a comparison window or designated region. Methods of aligning sequences for comparison are well-known in the art. Mutant humanin-like peptides (MHLPs) and derivates, analogs and variants provided herein may be modified chemically and/or biologically.
  • modifications include, but are not limited to, functional group introduction such as alkylation, acylation, amidation, esterification, halogenation, amination, carboxylation, and pegylation, functional group conversion such as oxidation, reduction, addition, and elimination, glycosylation, lipid compound introduction, phosporylation, and/or biotinylation.
  • modification(s) may for example stabilize and/or enhance the biological activity of mutant-humanin- like peptides (MHLPs) and derivates, analogs and variants thereof.
  • identifying methods for activity of humanin comprising inhibition, and/or stimulation or expression of humanin, such as the activity or expression in the cell is modulated by using mutant-humanin-like peptides (MHLPs).
  • Another aspect is a method for identifying modulation of
  • treating used herein includes, but is not limited to prevention, amelioration, alleviation, and/or eliminations of disease, disorder, or condition being treated or one or more symptoms of the disease, disorder or condition being treated, but also of therapies/treatments/drugs alone or in combination affecting cartilage tissues and/or bone tissues as well as improvement in the overall well being of patient, as measured by objective and/or subjective criteria.
  • Disease refers to a problem affecting cartilage tissues and/or bone tissues in a negative manner, but also of therapies/treatment/drugs alone or in combination affecting cartilage tissues and or bone tissues in a negative manner.
  • disorder that negativly affects cartilage tissues and/or bone tissues refers to a disorder, disease or condition that interact, alter, influences and/or disturbs the normal physiology and/or morphology and/or function of cartilage tissues and/or bone tissues.
  • X-ray examination Dual energy X-ray absorptiometry, Ultrasound, Computer tomography, Peripheral quantitative computer tomography, Magnetic resonance imaging, Visual inspection, Histological examination, Clinical examination, or Analysis of biological markers in the blood.
  • short stature used herein includes, but is not limited to, Familial short stature, Constitutional delay of growth and puberty, Idiopathic short stature, Small for gestational age, Intrauterine growth retardation, Growth hormone deficiency, Insulin-like growth factor-l deficiency, or growth impairment caused by a chronic disease and/or genetically determined disorder and/or syndrome.
  • bone dysplasia used herein includes, but is not limited to,
  • Achondrodysplasia Hypochondrodysplasia or a disorder and/or syndrome affecting cartilage and/or bone development.
  • osteomalacia used herein is defined as a disorder which involves the softening of the bones due to defective bone mineralization which can be caused by, but not limited to, Rickets, Hypoparathyroidism,
  • osteoporosis used herein is defined as a disease of bone that leads to an increased risk of fracture. In osteoporosis the bone mineral density is reduced and the bone microarchitecture is disrupted, and the amount and variety of proteins in bone may be altered.
  • skin atrophy used herein is defined as a condition when the skin becomes thin which could be secondary to a medical treatment or part of a natural aging process.
  • a “subject” of a method provided herein refers to any mammalian patient to which peptides or compositions of the invention can be beneficially administered.
  • the term “mammal” refers to humans and non-human primates, as well as experimental or veterinary animals, such as rabbits, rats mice, and other animals.
  • an "effective amount" of MHLPs and derivates, analogs and variants thereof is an amount sufficient to provide a measurable reduction in symptoms or other beneficial effect(s) with respect to a disease, therapy, disorder, or condition targeted for treatment.
  • Cancer refers generally to a disease characterized by uncontrolled, abnormal cell growth and proliferation.
  • a “tumor” or “neoplasm” is an abnormal mass of tissue that results from excessive, uncontrolled, and progressive cell division.
  • the present invention also provides methods of treating a subject having a disorder affecting cartilage tissues and/or bone tissues characterized by aberrant activity and/or aberrant expression of humanin or variant thereof, by administering an agent which is a modulator of the activity of humanin or variant or modulator thereof.
  • an agent which is a modulator of the activity of humanin or variant or modulator thereof.
  • pharmaceutical compositions comprising one or more peptides of the invention together with at least one pharmaceutically acceptable carrier or excipient.
  • Peptides of the invention can be incorporated into pharmaceutical
  • compositions suitable for administration typically comprise the peptide and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), intranasal, transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF; Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze- drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
  • compositions can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Phmogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active for transdermal administration, the active
  • the active peptides are prepared with carriers that will protect the peptide against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen,
  • polyorthoesters and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as
  • the active ingredient of the pharmaceutical composition of the present invention may be DNA encoding the polypeptide of the present invention.
  • examples of administration methods thereof include a method which administers a vector incorporating the DNA therein.
  • the vector include plasmids, adenovirus vectors, adeno- associated virus vectors, herpes virus vectors, vaccinia virus vectors, and retrovirus vectors.
  • the therapeutic agent can be expressed in vivo with efficiency by infecting organisms with the viral vectors.
  • a method which introduces the vector or the DNA into liposomes e.g., positively charged liposomes and positively charged cholesterol
  • administers the liposome can be used as an effective therapy.
  • the pharmaceutical composition of the present invention When used as a preventive and/or therapeutic agent for the diseases described above, it can be administered to mammals such as humans, mice, rats, rabbits, dogs, and cats.
  • the dose and number of doses of the pharmaceutical drug of the present invention may be changed appropriately according to the age, sex, and conditions of a subject to be administered, or administration routes.
  • a therapeutically effective amount of protein or polypeptide provided herein can range from about 0.001 to 500 mg/kg body weight, preferably about 0.01 to 25 mg/kg body weight, more preferably about 0.1 to 20 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight.
  • treatment of a subject with a therapeutically effective amount of a protein or polypeptide can include a single treatment or, preferably, can include a series of treatments.
  • a subject is treated with protein or polypeptide in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.
  • the effective dosage of antibody, protein, or polypeptide used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of diagnostic assays as described herein.
  • doses of protein or polypeptide agents depend upon a number of factors within the ken of the ordinarily skilled physician, veterinarian, or researcher.
  • the dose(s) will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable.
  • Exemplary doses include milligram or microgram amounts of the protein or polypeptide per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram.
  • appropriate doses of a protein or polypeptide depend upon the potency of the protein or polypeptide with respect to the expression or activity to be modulated. Such appropriate doses may be determined using the assays described herein.
  • a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific protein or polypeptide employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
  • Human chondrocyte cell line HCS-2/8 (20000 cells/well, in 96 wells plate), was cultured in DMEM/Ham's F12 (Invitrogen) supplemented with 20% FBS Thereafter, cells were treated with dexamethasone (25 ⁇ ) and MHLPs (1 OnM and 1000 nM) in test medium (DMEM-F12 supplemented with 1 % FBS) . Incubation was at 37°C in a humidified atmosphere of 95% air/5%
  • FVB mice female, 4 weeks old were treated with daily injections of dexamethasone (Dexa, 2mg/kg), humanin peptide (HNG, 1 g/mouse) or vehicle (saline) for 4 weeks. Animals were then killed and a skin biopsy (8mm) taken, fixed in 4% formaldehyde, embedded in paraffin, and sectioned. Skin thickness (subcutis and dermis) was measured on digital images captured in the light microscope.
  • dexamethasone Dexa, 2mg/kg
  • HNG humanin peptide
  • vehicle saline
  • HNG S14G-HN
  • MHLPs mutant-humanin-like peptides
  • the MHLPs of the present invention may be compared with the humanin sequence S14G-HN (HNG):
  • HNG Human chondrocytes
  • MHLP1 to MHLP13 were tested in corresponding concentrations, i.e. 10 nM, 100 nM and 1000 nM, respectively.
  • MHLPs protect human chondrocytes from toxic/anti-proliferative effects of GCs
  • the cell proliferation reagent 4-[3- (4-iodophenyl)-2(4-nitrophenyl)-2H-5-tetrazolio]-1 ,3-benzene disulfonate was used, which was purchased from Roche Diagnostics GmbH (Mannheim, Germany).
  • WST-1 is a colorimetric assay for the quantification of cell proliferation and cell viability. This assay is based on a water-soluble tetrazolium salt, which is cleaved to formazan by mitochondrial enzymes. The amount of the formazan dye formed is directly correlated to the number of metabolically active cells.
  • the HCS-2/8 cells were plated in 96-well plates (2 10 4 cells/well), and cultured for 72 hrs in DMEM/F12 with 20% FBS added. The cells were then washed once with 1 ⁇ PBS before the medium was changed to test medium, which contained 1 % FBS and for each experiment specific concentrations of effectors which included Dexamethasone (25 ⁇ , diluted in 99% Ethanol), HNG (10 or 1000 nM diluted in dH 2 O) and MHLPs (10 or 1000 nM diluted in dH 2 O). Control cells were incubated in medium with no effectors added but with corresponding amounts of 99% Ethanol and dH 2 O. After treatment, WST-1 was added, 20 ⁇ /well (final dilution 1 :10), and the reaction mixture was incubated for 1 hr at 37 C before read
  • MHLPs prevent apoptosis caused by dexamethasone in chondrocytes
  • apoptosis was studied with the detection and quantification of cytoplasmic histone-associated DNA fragments (mono- and oligonucleasomes) by photometric enzyme immunoassay (Cell Death Detection ELISA PLUS , Roche Diagnostics).
  • the cells were first treated as described for the proliferation assay and then lysated (100 ⁇ lysis buffer) and centrifuged. The supernatant was removed for analysis of cytoplasmic histone-associated DNA fragments according to the manufacturer's instructions.
  • the cells were treated with dexamethasone (25 ⁇ , diluted in 99% Ethanol), HNG (100 nM diluted in dH 2 O) and MHLPs (100 nM diluted in dH 2 O). Control cells were incubated in medium with no effectors added but with corresponding amounts of 99% Ethanol and dH 2 O.
  • HCS-2/8 chondrocytes (1 x10 6 per Petri-dish) in test medium (DMEM-F12 with 1 % FBS) were treated with Dexamethasone (25 ⁇ , diluted in 99% Ethanol), HNG (100 nM diluted in dH 2 O) and MHLPs (100 nM diluted in dH2O), and lipopolysacharide (LPS, 100 ng/ml).
  • Control cells were incubated in medium with no effectors added but with corresponding amounts of 99% Ethanol and dH 2 O. After 48 hrs, test medium was collected and analysed for release of cytokine (TNF-alpha).
  • TNF-alpha The detection of TNF-alpha was performed by using an ELISA kit (R&D systems, Minneapolis) according to the instructions provided by the manufacturer.
  • the data presented in Table 4 show that selected MHLPs do not interfer with the desired anti-inflammatory effects of GCs (dexamethasone).
  • FVB mice female, 4 weeks old were given a daily injection of
  • dexamethasone (2mg/kg), humanin peptide HNG (1 g/mice) or vehicle (saline). After 4 weeks of treatment, animals were killed and an 8 mm skin biopsy was collected (one from the neck and one from the back near the tail). The skin biopsies were fixed in 4% formladehyde and embedded in paraffin blocks.
  • Figure 1 a the upper left slide represents vehichle; upper right represents HNG (1 g/mouse); lower left represents dexamethasone
  • GCs dihydroxy-3-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)

Abstract

The present invention relates to the field of medical therapeutics, and more particularly to therapeutic peptides active in the treatment of bone-or cartilage disorders and other diseases. Provided herein is a series of novel peptides and fragments, analogs, derivatives, and variants thereof, structurally related to humanin,which have beneficial activities on cartilage tissues and/or bone tissues (including the prevention of negative effects of drugs on cartilage tissues and/or bone tissues), and in other diseases including, bone fractures, skeletal chondrodysplasias, cancers, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, infertility,muscular disorders, aging,skin diseases, heart and vascular diseases and metabolic diseases.

Description

Novel peptides
FIELD OF THE INVENTION
The present invention relates to the field of medical therapeutics, and more particularly to novel therapeutic peptides, designated mutant humanin-like peptides (MHLPs) and fragments, analogs, derivatives, and variants thereof, which have beneficial activities on cartilage tissues and/or bone tissues, including prevention of negative effects of disorders/drugs on cartilage tissues and/or bone tissues by treatment with for example, but not limited to, glucocorticoids. The MHLPs are also useful in the prevention and/or treatment of cancers, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, heart and vascular diseases and metabolic diseases.
BACKGROUND OF THE INVENTION
The present invention relates to a number of peptides named "mutant humanin-like peptides" with capacity to regulate biological pathways.
Longitudinal bone growth occurs in the growth plate through a process called endochondral bone formation, a process where resting zone chondrocytes are recruited to start active proliferation and then undergo differentiation, followed by apoptosis and later mineralization. The balance between proliferation and differentiation is a crucial regulatory step controlled by various growth factors/hormones acting in both endocrine and
paracrine/autocrine ways. Any disturbance/alteration in these hormones systemically and/or locally within growth plate cartilage may lead to abnormal chondrogensis causing growth failure of long bones (Savendahl 2005). Glucocorticoids (GCs) are widely used as a chemotherapy in cancers, antiinflammatory and immunosuppressive drugs both in children and adults with chronic diseases. However, recent studies have revealed that long-term high- dose GC treatment often leads to growth failure in treated children, an effect which has been shown to be mediated through a negative effect on growth plate chondrocytes (Baron, Huang et al. 1992) (Loeb 1976). Furthermore, long-term use of GCs in adults (men and women) has also been reported to cause osteoporosis/bone fractures as a negative side effect. Dexamethasone (Dexa) is a widely used glucocortcoid used in children and adults to treat different diseases such as, asthma, inflammation and cancer. Beside the positive effects of Dexa on inflammation and cancer, its long-term use has been linked to negative side effects on bone. Previous studies (pre-clinical and clinical) show that long-term use of Dexa can cause bone growth retardation in children and osteoporosis both in children and adults (Demoly 2008). It is well known that Dexa can alter the processes of proliferation and differentiation in chondrocytes and other cells.
Further, use of Dexa in neonatals has also been reported to cause brain injury. High doses of GCs used as immunospressive agent may also trigger infections. Therefore, identification of new molecules/analogs/peptides with the potential to be used in combination with existing widely used drugs (such as glucocorticoids and other chemotherapeutic drugs) without altering actual effects of these drugs, but blocking the negative side effects is important for the development of new treatment strategies in different disorders.
During the past decades, the incidence of childhood cancers has increased (Mangano 1999). Moreover, the results of the cancer therapy in children and adolescents have improved and the number of childhood cancer survivors is increasing (Linet, Ries et al. 1999). However, there has been a significant increase in both acute and chronic toxicity associated with the more successful, but now highly intensive chemotherapy (CT) regimens (Linet, Ries et al. 1999). In view of the fact that the incidence of childhood cancers coincides with periods of rapid skeletal development, it has become particular important to understand the adverse effects of this modality of treatment (Pinkerton 1992). Disruption of the physiological cellular homeostasis of growth plate chondrocytes and/or bone cells results in skeletal growth disturbances. Consequently, numerous skeletal complications including short stature and osteoporosis are important long-term problems in adult cancer survivors. The most commonly used chemotherapy (CT) drugs to treat childhood cancers include alkalyting agents, antimetabolites, antibiotics, plant alkaloids (Balis 1997) alone or in combination with other drugs. Studies from in vitro experiments demonstrated direct effects on chondrocyte proliferation after treatment with various DNA-damaging CT drugs (cisplatin, etoposide, carboplatin and actinomycin) causing irreversible cell loss (Robson, Anderson et al. 1998). In vivo studies in male Wistar rats reported reduction in tibia length of 18% and 5% when animals received doxorubicin and methotrexate respectively (van Leeuwen, Kamps et al. 2000). In addition, several studies showed that CT alone is responsible for the decline in height seen during treatment for acute lymhoblastic leukemia (Halton, Atkinson et al. 1998) (Shalet and Price 1981 ) (Clayton, Shalet et al. 1988). Similarly, the antimetabolite 5-fluorouracil ( 5-FU) is used in the treatment of solid tumors not only in adult cancer patients but also in childhood solid tumors,
hepatoblastoma, head and neck cancers, nasopharyngeal carcinoma, esophageal/gastric junction adenocarcinoma, glaucoma and skin cancer. In pre-clinical studies 5-FU has been reported to suppress bone growth, an effect associated with rapid and significant suppression of cell proliferation in the growth plate and metaphysis and induction of apoptosis in the metaphysis (Xian, Howarth et al. 2004). Other chemotherapeutic drugs such as etoposide and cyclophosphamide, administered alone or in combination, have been reported to damage the growth plate by causing structural and cellular changes in the growth plate cartilage (Xian, Cool et al. 2007). Several clinical studies have shown a reduced bone mineral density after treatment with different CTs for different types of cancers (Arikoski, Komulainen et al. 1999) (Arikoski, Komulainen et al. 1999) (Arikoski, Kroger et al. 1999). The reduced bone mineral density after CT treatment may be permanent and significantly influences fracture risk later in life.
The proteasome inhibitors (Pis), including Bortezomib (Velcade, PS-341 ), belong to another novel and promising group of CT that has past phase I clinical trials (Blaney, Bernstein et al. 2004) and are now under phase II clinical trials in pediatric cancers. However, secondary effects on normal bystander tissues of primary life saving modalities are so far unknown.
Preliminary results from us indicates that Pis, such as MG262 and
Bortezomib target essential cell populations within the growth plate causing permanent growth retardation and chondrocyte cell death, both in vitro and in vivo (Zaman, Menendez-Benito et al. 2007).
Hashimoto and colleagues in 2001 identified humanin (HN), a novel 24 amino acid peptide (Hashimoto, Ito et al. 2001 ). In fact HN, encoded from the 16S ribosomal RNA of the mitochondrial DNA, was first identified in surviving neurons from an Alzheimer's disease (AD) patient, and described as a potent neurosurvival factor (Hashimoto, Niikura et al. 2001 ). Since then, its protective role has been described not only from various AD related insults, but also against prion-induced (Sponne, Fifre et al. 2004) and chemical- induced damage (Mamiya and Ukai 2001 ), thus broadening its role as a neuroprotective factor. Subsequently, it has been shown to be protective against many other cytotoxic agents (Kariya, Takahashi et al. 2003) and also protect non-neuronal cells such as smooth muscle cells (Jung and Van Nostrand 2003), rat pheochromocytoma cells (Kariya, Takahashi et al. 2002) and lymphocytes (Kariya, Takahashi et al. 2003). The HN cDNA shares complete identity with the mitochondrial 16SrRNA gene but spans only about half the length of the ribosomal RNA. HN is both an intracellular and secreted protein. Its transcripts of mitochondrial origin has been detected in normal mouse testis and colon (by immunoblot and immunohistochemical analyses) using specific antibodies against HN peptide (Kato, Iwamoto et al. 2003) and is present in cerebrospinal fluid (CSF), seminal fluid and serum. HN levels in CSF are few orders of magnitude higher than that in circulation, also present in kidney, brain, and the gastrointestinal tract. Interestingly, HN is highly conserved along evolution among species (between 90-100% homology), e.g. plants, nematodes, rats, mice, human and many other species (Guo, Zhai et al. 2003). So far, little has been discovered about the regulation of its production. HN has also been reported as an antagonist of Bax and Bid that induces survival in cancer cells (Guo, Zhai et al. 2003), cell survival by binding to putative cell-surface receptors (Ying, Iribarren et al. 2004) and as an IGFBP-3 partner that antagonizes the apoptotic actions of IGFBP-3 on cancer cells (Ikonen, Liu et al. 2003). Furthermore, IGFBP-3 is one of a number of peptides including insulin, leptin, adiponectin, and resistin that have been shown to act in the central nervous system to regulate glucose metabolism (Muse, Lam et al. 2007) (Obici, Zhang et al. 2002). Unlike these before mentioned peptides, IGFBP-3 is an HN partner that has pro- diabetogenic hypothalamic actions that are modulated by IGF-I (Muzumdar and Rao 2006). Finally, HN has been reported as a wide spectrum survival factor (Nishimoto, Matsuoka et al. 2004), but its exact mechanism of action remains unclear.
There are several analogs/derivatives/variants of HN with altered properties described, including humanin-Gly14 (HNG) (Hashimoto, Niikura et al. 2001 ) (Terashita, Hashimoto et al. 2003), HNG-F6A (non-IGFBP-3 binding) (Ikonen, Liu et al. 2003) and colivelin (hybrid peptide containing partial sequences of HN and ADNF9) (Chiba, Yamada et al. 2005). Interestingly, all these different analogs exert similar neuroprotective effects. HN and its analogs and derivatives have shown therapeutic potential for an array of diseases including Alzheimer's disease (AD), diabetes and kidney failure (Matsuoka and Hashimoto 2010) (Matsuoka 2009) (Xu, Chua et al. 2006) (Hoang, Park et al. 2010) (Singh and Mascarenhas 2008).
REFERENCES
Arikoski, P., J. Komulainen, et al. (1999). "Reduced bone density at
completion of chemotherapy for a malignancy." Arch Dis Child 80(2): 143-148.
Arikoski, P., J. Komulainen, et al. (1999). "Impaired development of bone mineral density during chemotherapy: a prospective analysis of 46 children newly diagnosed with cancer." J Bone Miner Res 14(12): 2002-2009.
Arikoski, P., H. Kroger, et al. (1999). "Disturbance in bone turnover in children with a malignancy at completion of chemotherapy." Med Pediatr Oncol 33(5): 455-461 .
Balis, F. M. (1997). "The Challenge of Developing New Therapies for
Childhood Cancers." Oncologist 2(1 ): l-ll.
Baron, J., Z. Huang, et al. (1992). "Dexamethasone acts locally to inhibit longitudinal bone growth in rabbits." Am J Physiol 263(3 Pt 1 ): E489- 492.
Blaney, S. M., M. Bernstein, et al. (2004). "Phase I study of the proteasome inhibitor bortezomib in pediatric patients with refractory solid tumors: a Children's Oncology Group study (ADVL0015)." J Clin Oncol 22(23): 4804-4809.
Chiba, T., M. Yamada, et al. (2005). "Development of a femtomolar-acting humanin derivative named colivelin by attaching activity-dependent neurotrophic factor to its N terminus: characterization of colivelin- mediated neuroprotection against Alzheimer's disease-relevant insults in vitro and in vivo." J Neurosci 25(44): 10252-10261 .
Clayton, P. E., S. M. Shalet, et al. (1988). "Growth in children treated for acute lymphoblastic leukaemia." Lancet 1 (8583): 460-462. De Luca, F., K. M. Barnes, et al. (2001 ). "Regulation of growth plate chondrogenesis by bone morphogenetic protein-2." Endocrinology 142(1 ): 430-436.
Demoly, P. (2008). "Safety of intranasal corticosteroids in acute
rhinosinusitis." Am J Otolaryngol 29(6): 403-413.
Guo, B., D. Zhai, et al. (2003). "Humanin peptide suppresses apoptosis by interfering with Bax activation." Nature 423(6938): 456-461 .
Halton, J. M., S. A. Atkinson, et al. (1998). "Growth and body composition in response to chemotherapy in children with acute lymphoblastic leukemia." Int J Cancer Suppl 11 : 81 -84.
Hashimoto, Y., Y. Ito, et al. (2001 ). "Mechanisms of neuroprotection by a novel rescue factor humanin from Swedish mutant amyloid precursor protein." Biochem Biophvs Res Commun 283(2): 460-468.
Hashimoto, Y., T. Niikura, et al. (2001 ). "Detailed characterization of
neuroprotection by a rescue factor humanin against various
Alzheimer's disease-relevant insults." J Neurosci 21(23): 9235-9245.
Hashimoto, Y., T. Niikura, et al. (2001 ). "A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer's disease genes and Abeta." Proc Natl Acad Sci U S A 98(1 1 ): 6336-6341 .
Hoang, P. T., P. Park, et al. (2010). "The neurosurvival factor Humanin
inhibits beta-cell apoptosis via signal transducer and activator of transcription 3 activation and delays and ameliorates diabetes in nonobese diabetic mice." Metabolism 59(3): 343-349.
Ikonen, M., B. Liu, et al. (2003). "Interaction between the Alzheimer's survival peptide humanin and insulin-like growth factor-binding protein 3 regulates cell survival and apoptosis." Proc Natl Acad Sci U S A
100(22): 13042-13047.
Jung, S. S. and W. E. Van Nostrand (2003). "Humanin rescues human
cerebrovascular smooth muscle cells from Abeta-induced toxicity." J Neurochem 84(2): 266-272. Kariya, S., N. Takahashi, et al. (2003). "Humanin improves impaired metabolic activity and prolongs survival of serum-deprived human lymphocytes." Mol Cell Biochem 254(1 -2): 83-89.
Kariya, S., N. Takahashi, et al. (2002). "Humanin inhibits cell death of serum deprived PC12h cells." Neuroreport 13(6): 903-907.
Kato, T., K. Iwamoto, et al. (2003). "No association of mutations and mRNA expression of WFS1/wolframin with bipolar disorder in humans."
Neurosci Lett 338(1 ): 21 -24.
Linet, M. S., L. A. Ries, et al. (1999). "Cancer surveillance series: recent trends in childhood cancer incidence and mortality in the United
States." J Natl Cancer Inst 91 (12): 1051 -1058.
Loeb, J. N. (1976). "Corticosteroids and growth." N Engl J Med 295(10): 547
552.
Mamiya, T. and M. Ukai (2001 ). "[Gly(14)]-Humanin improved the learning and memory impairment induced by scopolamine in vivo." Br J
Pharmacol 134(8): 1597-1599.
Mangano, J. J. (1999). "A rise in the incidence of childhood cancer in the
United States." Int J Health Serv 29(2): 393-408.
Matsuoka, M. (2009). "Humanin; a defender against Alzheimer's disease?"
Recent Pat CNS Drug Discov 4(1 ): 37-42.
Matsuoka, M. and Y. Hashimoto (2010). "Humanin and the receptors for humanin." Mol Neurobiol 41 (1 ): 22-28.
Muse, E. D., T. K. Lam, et al. (2007). "Hypothalamic resistin induces hepatic insulin resistance." J Clin Invest 117(6): 1670-1678.
Muzumdar, H. and M. Rao (2006). "Pulmonary dysfunction and sleep apnea in morbid obesity." Pediatr Endocrinol Rev 3 Suppl 4: 579-583.
Nishimoto, I., M. Matsuoka, et al. (2004). "Unravelling the role of Humanin."
Trends Mol Med 10(3): 102-105.
Obici, S., B. B. Zhang, et al. (2002). "Hypothalamic insulin signaling is
required for inhibition of glucose production." Nat Med 8(12): 1376- Owen, H. C, J. N. Miner, et al. (2007). "The growth plate sparing effects of the selective glucocorticoid receptor modulator, AL-438." Mol Cell
Endocrinol 264(1 -2): 164-170.
Pinkerton, C. R. (1992). "Avoiding chemotherapy related late effects in
children with curable tumours." Arch Pis Child 67(9): 1 1 16-1 1 19.
Robson, H., E. Anderson, et al. (1998). "Chemotherapeutic agents used in the treatment of childhood malignancies have direct effects on growth plate chondrocyte proliferation." J Endocrinol 157(2): 225-235.
Savendahl, L. (2005). "Hormonal regulation of growth plate cartilage." Horm Res 64 Suppl 2: 94-97.
Shalet, S. M. and D. A. Price (1981 ). "Effect of treatment of malignant disease on growth in children." Arch Pis Child 56(3): 235-236.
Singh, B. K. and D. D. Mascarenhas (2008). "Bioactive peptides control
receptor for advanced glycated end product-induced elevation of kidney insulin receptor substrate 2 and reduce albuminuria in diabetic mice." Am J Nephrol 28(6): 890-899.
Sponne, I., A. Fifre, et al. (2004). "Humanin rescues cortical neurons from
prion-peptide-induced apoptosis." Mol Cell Neurosci 25(1 ): 95-102. Terashita, K., Y. Hashimoto, et al. (2003). "Two serine residues distinctly
regulate the rescue function of Humanin, an inhibiting factor of
Alzheimer's disease-related neurotoxicity: functional potentiation by isomerization and dimerization." J Neurochem 85(6): 1521 -1538.
van Leeuwen, B. L, W. A. Kamps, et al. (2000). "Effect of single
chemotherapeutic agents on the growing skeleton of the rat." Ann
Oncol 11 (9): 1 121 -1 126.
Wassberg, E., S. Pahlman, et al. (1997). "The angiogenesis inhibitor TNP-470 reduces the growth rate of human neuroblastoma in nude rats." Pediatr
Res 41 (3): 327-333.
Xian, C. J., J. C. Cool, et al. (2007). "Cellular mechanisms for methotrexate chemotherapy-induced bone growth defects." Bone 41 (5): 842-850. Xian, C. J., G. S. Howarth, et al. (2004). "Effects of acute 5-fluorouracil chemotherapy and insulin-like growth factor-l pretreatment on growth plate cartilage and metaphyseal bone in rats." Bone 35(3): 739-749.
Xu, X., C. C. Chua, et al. (2006). "Humanin is a novel neuroprotective agent against stroke." Stroke 37(10): 2613-2619.
Ying, G., P. Iribarren, et al. (2004). "Humanin, a newly identified
neuroprotective factor, uses the G protein-coupled formylpeptide receptor-like-1 as a functional receptor." J Immunol 172(1 1 ): 7078- 7085.
Zaman, F., V. Menendez-Benito, et al. (2007). "Proteasome inhibition up- regulates p53 and apoptosis-inducing factor in chondrocytes causing severe growth retardation in mice." Cancer Res 67(20): 10078-10086.
DISCLOSURE OF THE INVENTION
The present inventors have identified a number of mutant humanin-like peptides (MHLPs) having the capacity to regulate biological pathways, such as inflammation and exert an inhibitory effect on the death of chondrocyte cells. The peptides are named "mutant humanin-like peptides" (MHLPs) and the present invention relates to the peptides themselves and their use in the treatment and/or prevention of bone- or cartilage disorders/diseases, bone fractures, skeletal chondrodysplasias, cancers, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, infertility, muscular disoders, aging, skin diseases, heart and vascular diseases and metabolic diseases.
A number of peptides have been designed, synthesized and tested for their capacity to modulate biological pathways in a cell model system. It was identified that chondrocyte cell death (which is the main cause of bone growth disorders under various conditions) is completely blocked by MHLPs, suggesting that the MHLPs according to the invention affect biological pathways. For instance, chondrocytes were treated with high dose glucocorticoids (GCs) and subsequent MHLP treatment completely rescued chondrocytes from undergoing apoptosis. The present findings show the use of MHLPs alone or in combination with chemotherapy completely prevents the negative effects of chemotherapy, wihout interfering with the primary effects of the drugs is of considerable clinical importance for their broader application. In a first aspect of the present invention, there is provided a peptide, comprising an amino acid sequence X1APX2X3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 ) wherein
Xi is selected from M, L, A, S, G, and P; X2 is selected from R, K, A, G, and E; X3 is selected from G, V, Y, and A; and fragments, analogs, derivatives, and variants thereof, provided that peptides wherein Xi is M; X2 is R and X3 is G simultaneously, are excluded.
In one embodiment of this aspect, there is provided a peptide, comprising an amino acid sequence
X1APX2X3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 ) wherein
Xi is selected from M, L, A, S, G, and P; X2 is selected from R, K, A, G, and E; X3 is selected from G, V, Y, and A; provided that peptides wherein Xi is M; X2 is R and X3 is G simultaneously, are excluded.
In another embodiment of this aspect, there is provided a peptide, comprising an amino acid sequence
X1APRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:2)
wherein
Xi is selected from L, A, S, G, and P. In another embodiment of this aspect, there is provided a peptide, comprising an amino acid sequence
MAPX2GFSCLLLLTGEIDLPVKRRA (SEQ ID NO:3)
wherein
X2 is selected from K, A, G, and E.
In another embodiment of this aspect, there is provided a peptide, comprising an amino acid sequence
MAPRX3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:4)
wherein
X3 is selected from V, Y, and A.
In another embodiment of this aspect, there is provided a peptide, said peptide being selected from a peptide represented by SEQ ID NO:5 to SEQ ID NO:17; LAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :5);
AAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :6);
SAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: ■n
GAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :8);
PAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :9);
MAPKGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :10);
MAPAGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :1 1 );
MAPEGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :12);
MAPGGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :13);
MAPRGVSCLLLLTGEIDLPVKRRA (SEQ ID NO: :14);
MAPRGYSCLLLLTGEIDLPVKRRA (SEQ ID NO: :15);
MAPRGASCLLLLTGEIDLPVKRRA (SEQ ID NO: :16); and
MAPRGGSCLLLLTGEIDLPVKRRA (SEQ ID NO: :17). In another embodiment of this aspect, there is provided a peptide, said peptide being LAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:5).
In another embodiment of this aspect, there is provided a peptide, said peptide being AAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:6).
In another embodiment of this aspect, there is provided a peptide, said peptide being SAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:7).
In another embodiment of this aspect, there is provided a peptide, said peptide being GAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:8). In another embodiment of this aspect, there is provided a peptide, said peptide being PAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:9).
In another embodiment of this aspect, there is provided a peptide, said peptide being MAPKGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:10).
In another embodiment of this aspect, there is provided a peptide, said peptide being MAPAGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 1 ). In another embodiment of this aspect, there is provided a peptide, said peptide being MAPEGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:12).
In another embodiment of this aspect, there is provided a peptide, said peptide being MAPGGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:13).
In another embodiment of this aspect, there is provided a peptide, said peptide being MAPRGVSCLLLLTGEIDLPVKRRA (SEQ ID NO:14).
In another embodiment of this aspect, there is provided a peptide, said peptide being MAPRGYSCLLLLTGEIDLPVKRRA (SEQ ID NO:15).
In another embodiment of this aspect, there is provided a peptide, said peptide being MAPRGASCLLLLTGEIDLPVKRRA (SEQ ID NO:16). In another embodiment of this aspect, there is provided a peptide, said peptide being MAPRGGSCLLLLTGEIDLPVKRRA (SEQ ID NO:17).
In another aspect of the invention, there is provided a peptide of the invention, for use in therapy. In another aspect of the invention, there is provided a pharmaceutical composition comprising a peptide of the invention, together with
pharmaceutically acceptable diluents and/or carriers. In another aspect of the invention, there is provided a peptide of the invention, for use in in the prevention and/or treatment of bone- or cartilages disorders/diseases, cancer, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes,
neurodegenerative diseases, bone fractures, skeletal chondrodysplasias, infectious diseases, lung diseases, infertility, muscular disoders, aging, skin diseases, heart and vascular diseases and metabolic diseases.
In one embodiment of this aspect, there is provided a peptide of the invention, for use in the prevention and/or treatment of bone- or cartilage disorders. Said peptide may improve bone growth. The peptide may improve bone growth by itself targeting the Wnt-signalling or beta-catenin pathway or together with growth hormone (GH) or insulin-like growth factor I (IGF-I). Further, said peptide may improve bone healing. Further, said peptide may prevent development of osteoporosis. Said bone- or cartilage disorder may be a either a primary or secondary bone- or cartilage disorder. Further, said peptide may improve skin healing.
In another embodiment of this aspect, there is provided a peptide of the invention, for use in the prevention and/or treatment of bone- or cartilage disorders wherein said disorder is drug-induced. Said drug-induced disorder may have arisen from drugs used in the treatment of rheumatological disorders, respiratory disorders, gastrointestinal disorders, cardiovascular disorders, endocrinological disorders, cancer, neurodegenerative disorders, kidney disorders, liver disorders, dermatologic disorders, allergic disorders, inflammatory disorders, metabolic disorders, undesired immune response, undesired inflammatory response, obesity or diabetes. In another embodiment of this aspect, there is provided a peptide of the invention, for use in the prevention and/or treatment of bone- or cartilage disorders wherein said disorder is drug-induced bone growth impairment, short stature or osteoporosis. Said drug may be an anti-inflammatory drug, such as a glucocorticoid drug. Said glucocorticoid drug may be selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate,
budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide,
fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone, methylprednisolone
aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate,
beclomethasone monopropionate, paramethasone, alclometasone, fludorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal and mometasone furoate. Preferably, said glucocorticoid drug is dexamethasone.
In another embodiment of this aspect, there is provided a peptide of the invention, for use in the prevention and/or treatment of bone- or cartilage disorders wherein said disorder is drug-induced by an anti-cancer drug. Said anti-cancer drug may be a proteasome inhibitor, such as bortezomib,
MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS. Preferably, said proteasome inhibitor is bortezomib. In another aspect of the invention, there is provided a peptide of the invention, for use in the prevention and/or treatment of bone- or cartilage disorders, wherein said peptide is administered in combination with another drug known to induce a bone- or cartilage disorder.
In one embodiment of this aspect, said another drug is selected from an antiinflammatory drug, a selective estrogen receptor modulator drug, an anti- androgen drug, an aromatase inhibitor drug and an anti-cancer drug. When said another drug is an anti-inflammatory drug, it may be a
glucocorticoid drug, such as hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone,
methylprednisolone aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone
dipropionate, beclomethasone monopropionate, paramethasone,
alclometasone, fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal or mometasone furoate. Preferably, said glucocorticoid drug is dexamethasone.
When said another drug is an anti-cancer drug, it may be a proteasome inhibitor, such as bortezomib, MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS, ZLVS. Preferably said proteasome inhibitor is bortezomib. Alternatively, said another drug may be selected from leuprolide, buserelin, nafarelin, histrelin, goserelin, deslorelin, cetrorelix, ganirelix, abarelix, degarelix, letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide, testolactone, tamoxifen, afimoxifene, 4- hydroxytamoxifen, arzoxifene, bazedoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, tamoxifentoremifene, spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride, dutasteride, cyclophosphamide, mechlorethamine, uramustine, melphalan, chlorambucil, ifosfamide, carmustine, lomustine, streptozocin, busulfan, thiotepa, cisplatin, carboplatin, nedaplatin, oxaliplatin, satraplatin, triplatin tetranitrate, procarbazine, altretamine, dacarbazine, mitozolomide, temozolomide, imatinib mesylate, erlotinib, gefitinib, sunitinib, roscovitine, bevacizumab, rapamycin, cyclosporin A, tacrolimus, 5-fluorouracil, methotrexate, etoposide, doxorubicin, actinomycine, vitamin A acid, 13-cis- retinoic acid, 2-chlorodeoxyadenosine, 5-azacitidine, 5-fluorouracil, 6- mercaptopurine, 6-thioguanine, abraxane, accutane, actinomycin-d, adriamycin, adrucil, afinitor, agrylin, ala-cort, aldesleukin, alemtuzumab, alitretinoin, alkaban-aq, alkeran, all-transretinoic acid, alpha interferon, altretamine, amethopterin, amifostine, aminoglutethimide, anagrelide, anandron, anastrozole, arabinosylcytosine, ara-c, aranesp, aredia, arimidex, aromasin, arranon, arsenic trioxide, asparaginase, avastin, azacitidine, bendamustine, bevacizumab, bexarotene, bicalutamide, blenoxane, bleomycin, busulfan, busulfex, calcium leucovorin, campath, camptosar, camptothecin-1 1 , capecitabine, carac, carboplatin, carmustine, carmustine wafer, casodex, cerubidine, cetuximab, chlorambucil, cisplatin, citrovorum factor, cladribine, cortisone, cosmegen, cyclophosphamide, cytadren, cytarabine, cytarabine liposomal, cytosar-U, Cytoxan, dacarbazine, dacogen, dactinomycin, darbepoetin alfa, dasatinib, daunomycin, daunorubicin, daunorubicin hydrochloride, daunorubicin liposomal, daunoxome, decadron, decitabine, delta-cortef, deltasone, denileukin diftitox, depocyt,
dexamethasone acetate, dexamethasone sodium phosphate, dexasone, dexrazoxane, dhad, die, diodex, docetaxel, doxil, doxorubicin, doxorubicin liposomal, droxia, duralone, efudex, eligard, ellence, eloxatin, elspar, emcyt, epirubicin, epoetin alfa, erbitux, erlotinib, erwinia L-asparaginase,
estramustine, ethyol, etopophos, etoposide, etoposide phosphate, eulexin, everolimus, evista, exemestane, fareston, faslodex, femara, filgrastim, floxuridine, fludara, fludarabine, fluoroplex, fluorouracil, fluorouracil (cream), fluoxymesterone, flutamide, folinic acid, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, gemzar, gleevec, gliadel wafer, goserelin, halotestin, herceptin, hexadrol, hexalen, hexamethylmelamine, hycamtin, hydrea, hydrocort acetate, hydrocortisone, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, hydrocortone phosphate, hydroxyurea, ibritumomab, ibritumomab tiuxetan, idamycin, idarubicin, ifex, ifosfamide, imatinib mesylate, imidazole carboxamide, interferon alfa, interleukin-2, interleukin-1 1 , intron A (interferon alfa-2b), iressa, irinotecan, isotretinoin, ixabepilone, ixempra, kidrolase (t), lanacort, lapatinib, l-asparaginase, lenalidomide, letrozole, leucovorin, leukeran, leukine, leuprolide, leurocristine, leustatin, liposomal ara-C, liquid pred, lomustine, L-sarcolysin, lupron, lupron depot, matulane, maxidex, mechlorethamine, mechlorethamine hydrochloride, medralone, medrol, megace, megestrol, megestrol acetate, melphalan, mercaptopurine, mesna, mesnex, methotrexate, methotrexate sodium, methylprednisolone, meticorten, mitomycin, mitomycin-c, mitoxantrone, m- prednisol, mustargen, mustine, mutamycin, myleran, mylocel, mylotarg, navelbine, nelarabine, neosar, neulasta, neumega, neupogen, nexavar, nilandron, nilutamide, nipent, nitrogen mustard, novaldex, novantrone, nplate, octreotide, octreotide acetate, oncospar, Oncovin, ontak, onxal, oprelvekin, orapred, orasone, oxaliplatin, paclitaxel, paclitaxel protein-bound,
pamidronate, panitumumab, panretin, paraplatin, pediapred, PEG Interferon, pegaspargase, pegfilgrastim, peginterferon alfa-2b, PEG-L-asparaginase, pemetrexed, pentostatin, phenylalanine mustard, platinol, platinol-AQ, prednisolone, prednisone, prelone, procarbazine, proleukin, prolifeprospan 20 with carmustine implant, purinethol, raloxifene, revlimid, rheumatrex, rituxan, rituximab, romiplostim, rubex, rubidomycin hydrochloride, sandostatin, sandostatin LAR, sargramostim, solu-cortef, solu-medrol, sorafenib, streptozocin, sunitinib, sutent, tamoxifen, tarceva, targretin, taxol, taxotere, temodar, temozolomide, temsirolimus, teniposide, thalidomide, thalomid, theracys, thioguanine, thioguanine tabloid, thiophosphoamide, thioplex, thiotepa, toposar, topotecan, toremifene, torisel, tositumomab, trastuzumab, treanda, tretinoin, trexall, trisenox, vectibix, velban, vepesid, vesanoid, viadur, vidaza, vinblastine, vinblastine sulfate, vincasar pfs, vincristine, vinorelbine, vinorelbine tartrate, vorinostat, vumon, xeloda, zanosar, zevalin, zinecard, zoladex, zoledronic acid, zolinza and zometa.
In another aspect of the invention, there is provided a method of preventing and/or treating bone- or cartilage disorders, bone fractures, skeletal chondrodysplasias, cancer, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes,
neurodegenerative diseases, infectious diseases, lung diseases, muscular disorders, aging, infertility, skin atrophy, heart and vascular diseases and metabolic diseases, comprising administering a therapeutically effective amount of a peptide of the present invention. In one embodiment of this aspect, there is provided a method of preventing and/or treating bone- or cartilage disorders, comprising administering a therapeutically effective amount of a peptide of the present invention. Said peptide may improve bone growth. Further, said peptide may improve bone healing. Further, said peptide may prevent development of osteoporosis. Said bone- or cartilage disorder may be a either a primary or secondary bone- or cartilage disorder. Further, said peptide may improve skin healing.
In another embodiment of this aspect, there is provided a method of preventing and/or treating bone- or cartilage disorders comprising
administering a therapeutically effective amount of a peptide of the present invention, wherein said disorder is drug-induced. Said drug-induced disorder may have arisen from drugs used in the treatment of rheumatological disorders, respiratory disorders, gastrointestinal disorders, cardiovascular disorders, endocrinological disorders, cancer, neurodegenerative disorders, kidney disorders, liver disorders, dermatologic disorders, allergic disorders, inflammatory disorders, metabolic disorders, undesired immune response, undesired inflammatory response, obesity or diabetes.
In another embodiment of this aspect, there is provided a method of preventing and/or treating bone- or cartilage disorders comprising
administering a therapeutically effective amount of a peptide of the present invention, wherein said disorder is drug-induced bone growth impairment, short stature or osteoporosis. Said drug may be an anti-inflammatory drug, such as a glucocorticoid drug. Said glucocorticoid drug may be selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate,
budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide,
fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone, methylprednisolone
aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate,
beclomethasone monopropionate, paramethasone, alclometasone, fludorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal and mometasone furoate. Preferably, said glucocorticoid drug is dexamethasone.
In another embodiment of this aspect, there is provided a method of preventing and/or treating bone- or cartilage disorders comprising
administering a therapeutically effective amount of a peptide of the present invention, wherein said disorder is drug-induced by an anti-cancer drug. Said anti-cancer drug may be a proteasome inhibitor, such as bortezomib, MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS. Preferably, said proteasome inhibitor is bortezomib.
In another aspect of the invention, there is provided a method of preventing and/or treating bone- or cartilage disorders comprising administering a therapeutically effective amount of a peptide of the present invention, wherein said peptide is administered in combination with another drug known to induce a bone- or cartilage disorder.
In one embodiment of this aspect, said another drug is selected from an anti- inflammatory drug, a selective estrogen receptor modulator drug, an anti- androgen drug, an aromatase inhibitor drug and an anti-cancer drug.
When said another drug is an anti-inflammatory drug, it may be a
glucocorticoid drug, such as hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, cloprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone,
methylprednisolone aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate, beclomethasone monopropionate, paramethasone,
alclometasone, fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal or mometasone furoate. Preferably, said glucocorticoid drug is dexamethasone.
When said another drug is an anti-cancer drug, it may be a proteasome inhibitor, such as bortezomib, MLN9708, MLN273, MLN519, NPI-0052,
Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS, ZLVS. Preferably said proteasome inhibitor is bortezomib.
Alternatively, said another drug may be selected from leuprolide, buserelin, nafarelin, histrelin, goserelin, deslorelin, cetrorelix, ganirelix, abarelix, degarelix, letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide, testolactone, tamoxifen, afimoxifene, 4- hydroxytamoxifen, arzoxifene, bazedoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, tamoxifentoremifene, spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride, dutasteride, cyclophosphamide, mechlorethamine, uramustine, melphalan, chlorambucil, ifosfamide, carmustine, lomustine, streptozocin, busulfan, thiotepa, cisplatin, carboplatin, nedaplatin, oxaliplatin, satraplatin, triplatin tetranitrate, procarbazine, altretamine, dacarbazine, mitozolomide, temozolomide, imatinib mesylate, erlotinib, gefitinib, sunitinib, roscovitine, bevacizumab, rapamycin, cyclosporin A, tacrolimus, 5-fluorouracil, methotrexate, etoposide, doxorubicin, actinomycine, vitamin A acid, 13-cis- retinoic acid, 2-chlorodeoxyadenosine, 5-azacitidine, 5-fluorouracil, 6- mercaptopurine, 6-thioguanine, abraxane, accutane, actinomycin-d, adriamycin, adrucil, afinitor, agrylin, ala-cort, aldesleukin, alemtuzumab, alitretinoin, alkaban-aq, alkeran, all-transretinoic acid, alpha interferon, altretamine, amethopterin, amifostine, aminoglutethimide, anagrelide, anandron, anastrozole, arabinosylcytosine, ara-c, aranesp, aredia, arimidex, aromasin, arranon, arsenic trioxide, asparaginase, avastin, azacitidine, bendamustine, bevacizumab, bexarotene, bicalutamide, blenoxane, bleomycin, busulfan, busulfex, calcium leucovorin, campath, camptosar, camptothecin-1 1 , capecitabine, carac, carboplatin, carmustine, carmustine wafer, casodex, cerubidine, cetuximab, chlorambucil, cisplatin, citrovorum factor, cladribine, cortisone, cosmegen, cyclophosphamide, cytadren, cytarabine, cytarabine liposomal, cytosar-U, Cytoxan, dacarbazine, dacogen, dactinomycin, darbepoetin alfa, dasatinib, daunomycin, daunorubicin, daunorubicin hydrochloride, daunorubicin liposomal, daunoxome, decadron, decitabine, delta-cortef, deltasone, denileukin diftitox, depocyt,
dexamethasone acetate, dexamethasone sodium phosphate, dexasone, dexrazoxane, dhad, die, diodex, docetaxel, doxil, doxorubicin, doxorubicin liposomal, droxia, duralone, efudex, eligard, ellence, eloxatin, elspar, emcyt, epirubicin, epoetin alfa, erbitux, erlotinib, erwinia L-asparaginase,
estramustine, ethyol, etopophos, etoposide, etoposide phosphate, eulexin, everolimus, evista, exemestane, fareston, faslodex, femara, filgrastim, floxuridine, fludara, fludarabine, fluoroplex, fluorouracil, fluorouracil (cream), fluoxymesterone, flutamide, folinic acid, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, gemzar, gleevec, gliadel wafer, goserelin, halotestin, herceptin, hexadrol, hexalen, hexamethylmelamine, hycamtin, hydrea, hydrocort acetate, hydrocortisone, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, hydrocortone phosphate, hydroxyurea, ibritumomab, ibritumomab tiuxetan, idamycin, idarubicin, ifex, ifosfamide, imatinib mesylate, imidazole carboxamide, interferon alfa, interleukin-2, interleukin-1 1 , intron A (interferon alfa-2b), iressa, irinotecan, isotretinoin, ixabepilone, ixempra, kidrolase (t), lanacort, lapatinib, l-asparaginase, lenalidomide, letrozole, leucovorin, leukeran, leukine, leuprolide, leurocristine, leustatin, liposomal ara-C, liquid pred, lomustine, L-sarcolysin, lupron, lupron depot, matulane, maxidex, mechlorethamine, mechlorethamine hydrochloride, medralone, medrol, megace, megestrol, megestrol acetate, melphalan, mercaptopurine, mesna, mesnex, methotrexate, methotrexate sodium, methylprednisolone, meticorten, mitomycin, mitomycin-c, mitoxantrone, m- prednisol, mustargen, mustine, mutamycin, myleran, mylocel, mylotarg, navelbine, nelarabine, neosar, neulasta, neumega, neupogen, nexavar, nilandron, nilutamide, nipent, nitrogen mustard, novaldex, novantrone, nplate, octreotide, octreotide acetate, oncospar, Oncovin, ontak, onxal, oprelvekin, orapred, orasone, oxaliplatin, paclitaxel, paclitaxel protein-bound,
pamidronate, panitumumab, panretin, paraplatin, pediapred, PEG Interferon, pegaspargase, pegfilgrastim, peginterferon alfa-2b, PEG-L-asparaginase, pemetrexed, pentostatin, phenylalanine mustard, platinol, platinol-AQ, prednisolone, prednisone, prelone, procarbazine, proleukin, prolifeprospan 20 with carmustine implant, purinethol, raloxifene, revlimid, rheumatrex, rituxan, rituximab, romiplostim, rubex, rubidomycin hydrochloride, sandostatin, sandostatin LAR, sargramostim, solu-cortef, solu-medrol, sorafenib, streptozocin, sunitinib, sutent, tamoxifen, tarceva, targretin, taxol, taxotere, temodar, temozolomide, temsirolimus, teniposide, thalidomide, thalomid, theracys, thioguanine, thioguanine tabloid, thiophosphoamide, thioplex, thiotepa, toposar, topotecan, toremifene, torisel, tositumomab, trastuzumab, treanda, tretinoin, trexall, trisenox, vectibix, velban, vepesid, vesanoid, viadur, vidaza, vinblastine, vinblastine sulfate, vincasar pfs, vincristine, vinorelbine, vinorelbine tartrate, vorinostat, vumon, xeloda, zanosar, zevalin, zinecard, zoladex, zoledronic acid, zolinza and zometa. By a disorder is meant a disease or condition leading to an abnormality of normal functions. Such a condition includes, in accordance with the present invention, articular cartilage damage which if untreated can lead to
osteoarthritis or osteoarthrosis. Another such a condition is delayed fracture healing which if untreated can lead to severe consequences including pseudoarthrosis.
By a primary bone or cartilage disorder is meant growth impairment, short stature, arthritis, osteoarthritis, rheumatoid arthritis, psoriasis arthritis, ankylosing spondylitis, osteoarthrosis, osteomyelitis fibrous dysplasia, fibrodysplasia ossificans, craniosynostosis, metabolic bone disease, osteitis deformans, osteogenesis imperfecta, scoliosis, leg length difference, bone dysplasia, osteopetrosis, osteomalacia, osteopenia, osteoporosis, osteitis fibrosa cystic, osteochondritis, osteonecrosis, osteosarcoma, bone tumor, osteochondroma, osteochondropathy, chondropathy, chondrodysplasia, achondrodysplasia, hypochondrodysplasia, chondrodystrophy,
chondromalacia patellae, articular cartilage damage and delayed fracture healing.
By a secondary bone or cartilage disorder is meant a disorder which is a consequence of another disorder not primarily involving bone or cartilage tissues. Such another disorder includes Cushing's syndrome which is caused by increased Cortisol production.
By undesired immune response or undesired inflammatory response is meant responses which have undesired effects on bone or cartilage tissues. Rheumatological disorders include, but are not limited to, reumatoid arhtritis, systemic lupus erythematosus, vasculitis, periarhteritis nodosa and
sarcoidosis. Respiratory disorders include, but are not limited to, asthma bronchiale, chronic obstructive lung disease and bronchiolitis obliterans.
Gastrointestinal disorders include, but are not limited to, colitis ulcerosa, Crohn's disease and pancreatitis. Cardiovascular disorders include, but are not limited to, myocarditis, ischemia, hemolytic anemia and granulocytopenia. Endocrinological disorders include, but are not limited to, adrenal disorder, glucocorticoid deficiency, congenital adrenal hyperplasia,
adrenoleukodystrophy and pituitary disorder. Cancer includes, but is not limited to, acute myeloid leukemia, multiple myeloma and lymphoma.
Neurodegenerative disorders include, but are not limited to, multiple sclerosis and encephalitis. Kidney disorders include, but are not limited to,
glomerulonephritis and nephrotic syndrome. Liver disorders include, but are not limited to, hepatitis and cholangitis. Dermatologic disorders include, but are not limited to, psoriasis, exzema, myositis, dermatomyositis and dermatitis. In another aspect of the invention, the disorder is drug-induced from a GnRH agonist drug, such as leuprolide, buserelin, nafarelin, histrelin, goserelin and deslorelin. In another aspect of the invention, the disorder is drug-induced from a GnRH antagonist drug, such as cetrorelix, ganirelix, abarelix and degarelix.
In another aspect of the invention, the disorder is drug-induced from a selective estrogen receptor modulator drug, such as tamoxifen, afimoxifene, 4-hydroxytamoxifen, arzoxifene, bazedoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene and tamoxifentoremifene.
In another aspect of the invention, the disorder is drug-induced from an anti- androgen drug, such as spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide, ketoconazole, finasteride and dutasteride.
In another aspect of the invention, the disorder is drug-induced from an aromatase inhibitor drug, such as letrozole, anastrozole, exemestane, vorozole, formestane, fadrozole, aminoglutethimide and testolactone.
Brief description of the drawing
Figure 1 . Provides micrographs and data of potential protective effects of HNG and MHLPs of the invention on skin atrophy in mice (see Example 5 for further explanation).
Compositions of the invention may be administered by means which include but are not limited to intravenous, oral, subcutaneous, intra-arterial, intramuscular, intracardial, intraspinal, intrathoracic, intraperitoneal, intraventricular, sublingual, transdermal, intranasal, and inhalation.
The term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs as amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as naturally occurring amino acids that are later modified. Amino acid analogs are compounds that have the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refers to chemical compounds that have a structure that differs from the general chemical structure of an amino acid, but that functions similar to a naturally occurring amino acid.
"Endogenous" refers to a protein, nucleic acid, lipid or other biomolecule produced or originating within the body or within cells, organs, tissues of the body of a subject.
"Exogenous" refers to a protein, nucleic acid, lipid, or other biomolecule originating outside the body of a subject.
"Cartilage tissues" refers to all tissues in the body where cartilage cells (chondrocytes) are the major cellular components. This includes but is not limited to joint cartilage, growth plate cartilage and intervertebral disc cartilage. "Bone tissues" refers to all tissues in the body where bone cells (osteocytes, osteoblasts and osteoclasts) are important cellular components. This includes but is not limited to long bones and vertebrae
"Bone growth" refers to a series of co-ordinated actions which take place at the epiphyseal growth plate of long bones by balanced cycle of cartilage growth, formation of matrix, calcification of cartilage that acts as a scaffold for bone formation and modelling (where bone is being continuously resorbed and replaced by new bone). Bone modelling is highly active in
childhood/adolescence, and enables long bones to increase in diameter, change shape and develop a marrow cavity. The bone modelling process continues throughout adult life (this process in the adults is known as remodelling) with bone resorption equally balanced by bone formation in a healthy skeleton. Bone growth disorders occur when there is disruption of the normal cellular activity of chondrocytes (growth plate/articular) and/or bone cells. "Bone metabolism" refers to a process involving bone modeling and bone remodeling where cells produce the substances and energy needed for their survival. During bone remodeling, bone resorption by osteoclasts is followed by bone formation by osteoblasts. Bone remodeling does not result in changes of bone shape but helps in repairing of microdamage. Wherase, modeling is the formation of new bone by osteoblasts at locations different from the sites of bone resorption by osteoclasts and results in bone growth.
"Longitudinal bone growth" refers endochondral bone formation that occurs in the growth plates (a thin layer of cartilage located on each end of long boneswhere in a series of co-ordinated steps where resting zone/stem-like chondrocytes are recruited to start active proliferation and then undergo differentiation, followed by apoptosis and later mineralization resulting in increased bone length.
"Drug-induced longitudinal bone growth impairement" refers to a condition when drugs given to the patient/subject (mammal, children, adolescents, adults, animal) to treat different diseases but as a side effect they also alter/inhibit the normal process of bone elongation causing longitudinal bone growth impairment/retardation.
"Drug-induced" refers to an effect which is primarily due to drug(s) given to patient/subject under different disease conditions. "Fragment" refers to a small part synthesised/produced/broken off/or detached from its original place.
"Derivative" refers to a substance derived/produced/obtained either directly or by modification or partial substitution. "Catch-up growth" refers to acceleration of the growth rate in infants or young children above the limits of normal for age after a transient period of growth inhibition/impairment; it can be complete or incomplete.
The term "peptide" refers to any of various natural or synthetic compounds containing at least two or more amino acids linked by the carboxyl group of one amino acid to the amino group of another.
"Polypeptide" refers to chains of amino acids, and "proteins" are made up of one or more polypeptide molecules.
"Polynucleotide" refers to a polymeric form of nucleotides af at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, including single and double stranded forms of DNA.
The terms "polypeptide", "peptide" and "protein" herein refers to a polymer (large molecules composed of repeating structural units typically connected by covalent chemical bonds) formed from the linking, in a defined order, of a- amino acids. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
The term "endogenous expression of humanin in cartilage tissues and/or bone tissues" refers to the local production and/or secretion of humanin within cartilage tissues and/or bone tissues.
The phrase "agent that affects the local production and/or expression of humanin or another peptide expressed by the humanin gene" refers to an agent that for example, but not limited to, triggers endogenous production of humanin or humanin like peptides/derivatives/analogs in cartilage tissues and/or bone tissues by the use of siRNA, miRNA, shRNA (plasmid and lentiviral).
"MicroRNA (miRNA)" refers to a class of small RNA molecules, about 21 nucleotides in length that regulate gene expression in a variety of ways.
"RNA interference (RNAi)" refers to a biological mechanism by which double- stranded RNA (dsRNA) induces gene silencing by targeting complementary mRNA for degradation.
"small interfering RNA (siRNA)" refers to a class of double-stranded RNA molecules, 20-25 nucleotides in length, that play a variety of roles in cell. For example, siRNA is involved in the RNA interference (RNAi) pathway, where it interferes with the expression of a specific gene.
"short hairpin RNA (shRNA)" refers to a sequence of RNA that makes a tight hairpin turn that can be used to silence gene expression via RNA
interference. "Disorder that affects the endogenous expression of humanin in cartilage tissues and/or bone tissues negatively influencing cartilage tissues and/or bone tissues" refers to the presence, absence, increased or decreased level of humanin in cartilage tissues and/or bone tissues with or without treatment causing cartilage tissue and/or bone tissue disorders. For example, a patient suffering from increased or decreased and/or insufficient levels of locally produced humanin in cartilage tissues and/or bone tissues resulted from with/without treatment of any disease.
In some aspects, mutant-humanin-like peptides and derivates, analogs and variants thereof is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 85%, 95% or 98% identical to amino acid sequence of SEQ ID NO: 5-17. In some aspects, mutant-humanin-like peptides (MHLPs) and dehvates, analogs and variants thereof has an amino acid sequence which is
"substantially identical" to the sequence of the corresponding human humanin, in that the sequence is at least about 30%-98% or more identical to the sequence of a reference sequence, such as the corresponding
endogenous human humanin.
A "amino acid(s) substitution" is one in which one or more amino acid reidue(s) is replaced with an amino acid residue. Families of amino acid residues have been defined in the art and include amino acids with basic side chains (e.g lysine, arginine, histidine), acidic side chains (e.g. aspartic acid, glutamic acid), uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g. alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptphan), beta-branched side chains (e.g. threonine, valine, isoleucine) and aromatic side chains (e.g. tyrosine, phenylalanine, tryptophan, histidine). A predicted non-essential amino acid residue in humanin and humanin-like peptides (HLPs) and dehvates, analogs and variants thereof can typically be replaced with another amino acid residue, preferably from the same side chain family.
A "non-essential" amino acid residue is a residue that can be altered from the original sequence (e.g. a sequence of SEQ ID NO: 5-17) without abolishing or substantially altering the therapeutic activity of the peptide, whereas an "essential" amino acid residue is a residue that cannot be altered without introducing such a change.
The terms "identical" or percent "identity", in the context of two or more nucleic acids or polypeptide sequences, refers to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same when compared and aligned for maximum correspondence over a comparison window or designated region. Methods of aligning sequences for comparison are well-known in the art. Mutant humanin-like peptides (MHLPs) and derivates, analogs and variants provided herein may be modified chemically and/or biologically. Examples of such modifications include, but are not limited to, functional group introduction such as alkylation, acylation, amidation, esterification, halogenation, amination, carboxylation, and pegylation, functional group conversion such as oxidation, reduction, addition, and elimination, glycosylation, lipid compound introduction, phosporylation, and/or biotinylation. Such modification(s) may for example stabilize and/or enhance the biological activity of mutant-humanin- like peptides (MHLPs) and derivates, analogs and variants thereof. In another aspect, identifying methods for activity of humanin comprising inhibition, and/or stimulation or expression of humanin, such as the activity or expression in the cell is modulated by using mutant-humanin-like peptides (MHLPs). Another aspect is a method for identifying modulation of
transcription, splicing, translation of humanin.
The term "treating" used herein includes, but is not limited to prevention, amelioration, alleviation, and/or eliminations of disease, disorder, or condition being treated or one or more symptoms of the disease, disorder or condition being treated, but also of therapies/treatments/drugs alone or in combination affecting cartilage tissues and/or bone tissues as well as improvement in the overall well being of patient, as measured by objective and/or subjective criteria.
"Disease", "disorder", or "condition" herein refers to a problem affecting cartilage tissues and/or bone tissues in a negative manner, but also of therapies/treatment/drugs alone or in combination affecting cartilage tissues and or bone tissues in a negative manner. The terms "disorder that negativly affects cartilage tissues and/or bone tissues" refers to a disorder, disease or condition that interact, alter, influences and/or disturbs the normal physiology and/or morphology and/or function of cartilage tissues and/or bone tissues. This is assessed by different means, such as, but not limited to, X-ray examination, Dual energy X-ray absorptiometry, Ultrasound, Computer tomography, Peripheral quantitative computer tomography, Magnetic resonance imaging, Visual inspection, Histological examination, Clinical examination, or Analysis of biological markers in the blood.
The term "short stature" used herein includes, but is not limited to, Familial short stature, Constitutional delay of growth and puberty, Idiopathic short stature, Small for gestational age, Intrauterine growth retardation, Growth hormone deficiency, Insulin-like growth factor-l deficiency, or growth impairment caused by a chronic disease and/or genetically determined disorder and/or syndrome.
The term "bone dysplasia" used herein includes, but is not limited to,
Achondrodysplasia, Hypochondrodysplasia or a disorder and/or syndrome affecting cartilage and/or bone development.
The term "osteomalacia" used herein is defined as a disorder which involves the softening of the bones due to defective bone mineralization which can be caused by, but not limited to, Rickets, Hypoparathyroidism,
Pseudohypoparathyroidism, Hypophospatemia, Renal tubular acidosis, Cancer, Malabsorption or Malnutrition.
The term "osteoporosis" used herein is defined as a disease of bone that leads to an increased risk of fracture. In osteoporosis the bone mineral density is reduced and the bone microarchitecture is disrupted, and the amount and variety of proteins in bone may be altered. The term "skin atrophy" used herein is defined as a condition when the skin becomes thin which could be secondary to a medical treatment or part of a natural aging process.
A "subject" of a method provided herein refers to any mammalian patient to which peptides or compositions of the invention can be beneficially administered. The term "mammal" refers to humans and non-human primates, as well as experimental or veterinary animals, such as rabbits, rats mice, and other animals.
In some aspects, an "effective amount" of MHLPs and derivates, analogs and variants thereof is an amount sufficient to provide a measurable reduction in symptoms or other beneficial effect(s) with respect to a disease, therapy, disorder, or condition targeted for treatment.
"Cancer" refers generally to a disease characterized by uncontrolled, abnormal cell growth and proliferation. A "tumor" or "neoplasm" is an abnormal mass of tissue that results from excessive, uncontrolled, and progressive cell division.
The present invention also provides methods of treating a subject having a disorder affecting cartilage tissues and/or bone tissues characterized by aberrant activity and/or aberrant expression of humanin or variant thereof, by administering an agent which is a modulator of the activity of humanin or variant or modulator thereof. Also provided herein are pharmaceutical compositions comprising one or more peptides of the invention together with at least one pharmaceutically acceptable carrier or excipient.
Peptides of the invention can be incorporated into pharmaceutical
compositions suitable for administration. Such compositions typically comprise the peptide and a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), intranasal, transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as
ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF; Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze- drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Phmogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. For administration by inhalation, the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means.
For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active
compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
In one aspect, the active peptides are prepared with carriers that will protect the peptide against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen,
polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as
pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,81 1 . The active ingredient of the pharmaceutical composition of the present invention may be DNA encoding the polypeptide of the present invention. When the DNA encoding the oligopeptide is used as a gene therapy agent for the disease described above, examples of administration methods thereof include a method which administers a vector incorporating the DNA therein. Examples of the vector include plasmids, adenovirus vectors, adeno- associated virus vectors, herpes virus vectors, vaccinia virus vectors, and retrovirus vectors. The therapeutic agent can be expressed in vivo with efficiency by infecting organisms with the viral vectors. Alternatively, a method which introduces the vector or the DNA into liposomes (e.g., positively charged liposomes and positively charged cholesterol) and administers the liposome can be used as an effective therapy.
When the pharmaceutical composition of the present invention is used as a preventive and/or therapeutic agent for the diseases described above, it can be administered to mammals such as humans, mice, rats, rabbits, dogs, and cats. The dose and number of doses of the pharmaceutical drug of the present invention may be changed appropriately according to the age, sex, and conditions of a subject to be administered, or administration routes.
A therapeutically effective amount of protein or polypeptide provided herein can range from about 0.001 to 500 mg/kg body weight, preferably about 0.01 to 25 mg/kg body weight, more preferably about 0.1 to 20 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight.
The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a protein or polypeptide can include a single treatment or, preferably, can include a series of treatments. In a preferred example, a subject is treated with protein or polypeptide in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. It will also be appreciated that the effective dosage of antibody, protein, or polypeptide used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of diagnostic assays as described herein.
It is understood that appropriate doses of protein or polypeptide agents depend upon a number of factors within the ken of the ordinarily skilled physician, veterinarian, or researcher. The dose(s) will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable. Exemplary doses include milligram or microgram amounts of the protein or polypeptide per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram. It is furthermore understood that appropriate doses of a protein or polypeptide depend upon the potency of the protein or polypeptide with respect to the expression or activity to be modulated. Such appropriate doses may be determined using the assays described herein. When one or more of these proteins or polypeptides is to be administered to an animal (e.g., a human) in order to modulate expression or activity, a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained. In addition, it is understood that the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific protein or polypeptide employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
Having now generally described various aspects and aspects of the invention, the same will be more readily understood through reference to the following examples which are provided by way of illustration, and are not intended to be limiting, unless specified.
Examples
Material and methods
Cell Culture
Human chondrocyte cell line HCS-2/8 (20000 cells/well, in 96 wells plate), was cultured in DMEM/Ham's F12 (Invitrogen) supplemented with 20% FBS Thereafter, cells were treated with dexamethasone (25 μΜ) and MHLPs (1 OnM and 1000 nM) in test medium (DMEM-F12 supplemented with 1 % FBS) . Incubation was at 37°C in a humidified atmosphere of 95% air/5%
CO2. After 72 hrs, WST-1 cell viability/proliferation reagent was added in each well and cells were further incubated at 37°C for 45 minutes. Cell viability and proliferation, DNA fragmentation (apoptosis), and TNF-alpha cytokine levels were analysed by ELISA reader. The cells were subcultivated every week and given fresh medium every 3-4 days. Experiments were run in duplicate/triplicates. Data are presented in table 2 and 3.
Animal Studies
FVB mice (female, 4 weeks old) were treated with daily injections of dexamethasone (Dexa, 2mg/kg), humanin peptide (HNG, 1 g/mouse) or vehicle (saline) for 4 weeks. Animals were then killed and a skin biopsy (8mm) taken, fixed in 4% formaldehyde, embedded in paraffin, and sectioned. Skin thickness (subcutis and dermis) was measured on digital images captured in the light microscope. Example 1.
Selecive mutations in the S14G-HN (HNG) peptide (A) was performed and total 13 different unique mutant-humanin-like peptides ( MHLPs) were developed. The peptides were synthesized using conventional techniques, available to the skilled person. The peptide sequences and name designations are set out in Table 1 below.
Table 1. Mutant humanin-like peptides (MHLPs) of the invention.
Name Amino acid sequence SEQ ID NO
MHLP1 LAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO:5
MHLP2 AAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO:6
MHLP3 SAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO:7
MHLP4 GAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO:8
MHLP5 PAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO:9
MHLP6 MAPKGFSCLLLLTGEIDLPVKRRA SEQ ID NO:10
MHLP7 MAPAGFSCLLLLTGEIDLPVKRRA SEQ ID NO:1 1
MHLP8 MAPEGFSCLLLLTGEIDLPVKRRA SEQ ID NO:12
MHLP9 MAPGGFSCLLLLTGEIDLPVKRRA SEQ ID NO:13
MHLP10 MAPRGVSCLLLLTGEIDLPVKRRA SEQ ID NO:14
MHLP1 1 MAPRGYSCLLLLTGEIDLPVKRRA SEQ ID NO:15
MHLP12 MAPRGASCLLLLTGEIDLPVKRRA SEQ ID NO:16
MHLP13 MAPRGGSCLLLLTGEIDLPVKRRA SEQ ID NO:17
The MHLPs of the present invention may be compared with the humanin sequence S14G-HN (HNG):
Name Amino acid sequence
S14G-HN MAPRGFSCLLLLTGEIDLPVKRRA SEQ ID NO:18 (HNG) Human chondrocytes (HCS-2/8) were treated with Dexamethasone, LPS and selected MHLPs, as follows (further described in consecutive examples):
SEQ ID NO:18 (comparative)
Dexamethasone (D): 25 uM
LPS: 100 ng/ml
HNG : 10 nM
HNG : 100 nM
HNG 1000 nM
Each of MHLP1 to MHLP13 (SEQ ID NO:5 to SEQ ID NO:17) were tested in corresponding concentrations, i.e. 10 nM, 100 nM and 1000 nM, respectively.
Example 2.
MHLPs protect human chondrocytes from toxic/anti-proliferative effects of GCs
To investigate effects on cell proliferation, the cell proliferation reagent 4-[3- (4-iodophenyl)-2(4-nitrophenyl)-2H-5-tetrazolio]-1 ,3-benzene disulfonate (WST-1 ) was used, which was purchased from Roche Diagnostics GmbH (Mannheim, Germany). WST-1 is a colorimetric assay for the quantification of cell proliferation and cell viability. This assay is based on a water-soluble tetrazolium salt, which is cleaved to formazan by mitochondrial enzymes. The amount of the formazan dye formed is directly correlated to the number of metabolically active cells. The HCS-2/8 cells were plated in 96-well plates (2 104 cells/well), and cultured for 72 hrs in DMEM/F12 with 20% FBS added. The cells were then washed once with 1 χ PBS before the medium was changed to test medium, which contained 1 % FBS and for each experiment specific concentrations of effectors which included Dexamethasone (25 μΜ, diluted in 99% Ethanol), HNG (10 or 1000 nM diluted in dH2O) and MHLPs (10 or 1000 nM diluted in dH2O). Control cells were incubated in medium with no effectors added but with corresponding amounts of 99% Ethanol and dH2O. After treatment, WST-1 was added, 20 μΙ/well (final dilution 1 :10), and the reaction mixture was incubated for 1 hr at 37 C before read
absorbance at 450 nm.
The data presented in Table 2 show that some of the MHLPs are more potent than the native HNG peptide to prevent the anti-proliferative effects of dexamethasone in chondrocytes.
Table 2. Effect of MHLPs on cell proliferation
Figure imgf000045_0001
Figure imgf000046_0001
Example 3.
MHLPs prevent apoptosis caused by dexamethasone in chondrocytes
In HCS-2/8 chondrocytic cells, apoptosis was studied with the detection and quantification of cytoplasmic histone-associated DNA fragments (mono- and oligonucleasomes) by photometric enzyme immunoassay (Cell Death Detection ELISAPLUS, Roche Diagnostics). The cells were first treated as described for the proliferation assay and then lysated (100 μΙ lysis buffer) and centrifuged. The supernatant was removed for analysis of cytoplasmic histone-associated DNA fragments according to the manufacturer's instructions. The cells were treated with dexamethasone (25 μΜ, diluted in 99% Ethanol), HNG (100 nM diluted in dH2O) and MHLPs (100 nM diluted in dH2O). Control cells were incubated in medium with no effectors added but with corresponding amounts of 99% Ethanol and dH2O.
The data presented in Table 3 clearly demonstrate that MHLPs are effective to prevent DNA damage caused by dexamethasone. Further, some of the MHLPs are highly potent and more effective than native HNG peptide. Interestingly, some of the MHLPs induce higher level of cell death/apoptosis than native HNG which can have relevance to cancer treatment. Table 3. Anti-apoptotic effects of MHLPs
Figure imgf000047_0001
Figure imgf000048_0001
Example 4.
MHLPs do not interfere with the anti-inflammatory effects of GCs
HCS-2/8 chondrocytes (1 x106 per Petri-dish) in test medium (DMEM-F12 with 1 % FBS) were treated with Dexamethasone (25 μΜ, diluted in 99% Ethanol), HNG (100 nM diluted in dH2O) and MHLPs (100 nM diluted in dH2O), and lipopolysacharide (LPS, 100 ng/ml). Control cells were incubated in medium with no effectors added but with corresponding amounts of 99% Ethanol and dH2O. After 48 hrs, test medium was collected and analysed for release of cytokine (TNF-alpha). The detection of TNF-alpha was performed by using an ELISA kit (R&D systems, Minneapolis) according to the instructions provided by the manufacturer. The data presented in Table 4 show that selected MHLPs do not interfer with the desired anti-inflammatory effects of GCs (dexamethasone).
Table 4. Combined treatment of MHLPs and GCs (Dexamethasone) in inflammation.
Group TNF-alpha levels
(OD, ELISA)
Control (dH2O+Ethanol) 0.097
Lipopolysacharide 0.139
Lipopolysacharide + Dexamethasone 0.109
Lipopolysacharide + Dexamethasone + HNG 0.100 Lipopolysacharide + Dexamethasone + MHLP1 0.106
Lipopolysacha de + Dexamethasone + MHLP2 0.093
Example 5.
Potential prevention of skin atrophy by humanin/MHLPs
FVB mice (female, 4 weeks old) were given a daily injection of
dexamethasone (2mg/kg), humanin peptide HNG (1 g/mice) or vehicle (saline). After 4 weeks of treatment, animals were killed and an 8 mm skin biopsy was collected (one from the neck and one from the back near the tail). The skin biopsies were fixed in 4% formladehyde and embedded in paraffin blocks. In Figure 1 a, the upper left slide represents vehichle; upper right represents HNG (1 g/mouse); lower left represents dexamethasone
(2mg/kg); and lower right represents dexamethasone (2mg/kg) and humanin peptide HNG (1 g/mouse). The biopsies were sectioned and skin thickness (subcutis and dermis) was measured by using the Image-Pro software. Each group contained 9 animals. Data are presented in Figure 1 b. The humanin peptide tended to prevent dexamethasone-induced skin atrophy. It should be pointed out that a relatively low dose of dexamethasone was used and therefore the experimental conditions were not optimal to study
glucocorticoid-induced skin atrophy and its potential prevention by humanin. In future experiments, a higher dose of GCs (dexamethasone, 5mg/kg) will be applied on the skin or given systemically with or without humanin petide HNG and different concentrations of MHLPs. Skin biopsies will be collected and analysed for skin thickness.

Claims

1 . A peptide, comprising an amino acid sequence
X1APX2X3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 )
wherein
Xi is selected from M, L, A, S, G, and P;
X2 is selected from R, K, A, G, and E;
X3 is selected from G, V, Y, and A; and
fragments, analogs, derivatives, and variants thereof, provided that peptides wherein Xi is M; X2 is R and X3 is G simultaneously, are excluded.
2. A peptide according to claim 1 , comprising an amino acid sequence X1APX2X3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:1 )
wherein
Xi is selected from M, L, A, S, G, and P;
X2 is selected from R, K, A, G, and E;
X3 is selected from G, V, Y, and A;
provided that peptides wherein Xi is M; X2 is R and X3 is G
simultaneously, are excluded.
3. A peptide accoding to claim 1 or 2, comprising an amino acid sequence X1APRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO:2)
wherein Xi is selected from L, A, S, G, and P.
A peptide accoding to claim 1 or 2, comprising an amino acid sequence
MAPX2GFSCLLLLTGEIDLPVKRRA (SEQ ID NO:3)
wherein
X2 is selected from K, A, G, and E.
A peptide accoding to claim 1 or 2, comprising an amino acid sequence
MAPRX3FSCLLLLTGEIDLPVKRRA (SEQ ID NO:4)
wherein
X3 is selected from V, Y, and A.
6. A peptide according to any one of claims 1 to 5, said peptide being selected from a peptide of SEQ ID NO:5 to SEQ ID NO:17
LAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :5);
AAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :6);
SAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: ■n
GAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :8);
PAPRGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :9);
MAPKGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :10);
MAPAGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :1 1 );
MAPEGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :12);
MAPGGFSCLLLLTGEIDLPVKRRA (SEQ ID NO: :13);
MAPRGVSCLLLLTGEIDLPVKRRA (SEQ ID NO: :14); MAPRGYSCLLLLTGEIDLPVKRRA (SEQ ID NO:15); MAPRGASCLLLLTGEIDLPVKRRA (SEQ ID NO:16); and MAPRGGSCLLLLTGEIDLPVKRRA (SEQ ID NO:17). 7. A peptide according to any one of claims 1 to 6, for use in therapy.
8. A pharmaceutical composition comprising a peptide according to any one of claims 1 to 6 together with pharmaceutically acceptable diluents and/or carriers.
9. A peptide according to any one of claims 1 to 6, for use in the prevention and/or treatment of bone- or cartilages disorders, bone fractures, skeletal chondrodysplasias, cancer, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, infertility, aging, muscular disorders, skin diseases, heart and vascular diseases and metabolic diseases. 10. A peptide according to any one of claims 1 to 6, for use in the prevention and/or treatment of bone- or cartilage disorders.
1 1 . The peptide for use, according to claim 10, wherein said peptide
improves bone growth.
12. The peptide for use, according to claim 10, wherein said peptide
together with insulin-growth factor I improves bone growth.
13. The peptide for use, according to claim 10, wherein said peptide
together with growth hormone improves bone growth.
14. The peptide for use, according to claim 10, wherein said peptide
together with growth hormone improves bone healing.
15. The peptide for use, according to claim 10, wherein said peptide prevents development of osteoporosis. 16. The peptide for use according to claim 10, wherein said bone- or
cartilage disorder is either a primary or secondary bone- or cartilage disorder.
17. The peptide for use according to any one of claims 10 to 16, wherein said bone- or cartilage disorder is drug-induced.
18. The peptide for use according to claim 17, wherein said bone- or
cartilage disorder is drug-induced from drugs used in the treatment of rheumatological disorders, respiratory disorders, gastrointestinal disorders, cardiovascular disorders, endocrinological disorders, cancer, neurodegenerative disorders, kidney disorders, liver disorders, dermatologic disorders, allergic disorders, inflammatory disorders, metabolic disorders, undesired immune response, undesired
inflammatory response, obesity or diabetes.
19. The peptide for use according to claim 17, wherein said drug-induced bone- or cartilage disorder is selected from drug-induced bone growth impairment, short stature and osteoporosis. 20. The peptide for use according to any one of claims 17 to 19, wherein said drug is an anti-inflammatory drug, preferably a glucocorticoid drug.
21 . The peptide for use according to claim 20, wherein said glucocorticoid drug is selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, doprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone,
methylprednisolone, methyl prednisolone aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate, beclomethasone monopropionate, paramethasone, alclometasone, fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal and mometasone furoate, preferably dexamethasone.
The peptide for use according to any one of claims 17 to 19, wherein said drug is an anti-cancer drug, preferably a proteasome inhibitor.
The peptide for use according to claim 22, wherein said proteasome inhibitor is selected from bortezomib, MLN9708, MLN273, MLN519, NPI- 0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS- 519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS, preferably bortezomib.
24. The peptide for use according to any one of claims 9 to 23, wherein said peptide is administered in combination with another drug known to induce a bone- or cartilage disorder.
25. The peptide for use according to claim 24, wherein said another drug is selected from an anti-inflammatory drug, a selective estrogen receptor modulator drug, an anti-androgen drug, an aromatase inhibitor drug and an anti-cancer drug. The peptide for use according to claim 25, wherein said another drug is an anti-inflammatory drug, preferably a glucocorticoid drug.
The peptide for use according to claim 26, wherein said glucocorticoid drug is selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, doprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone,
methylprednisolone, methylprednisolone aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone, dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, bedomethasone dipropionate, bedomethasone monopropionate, paramethasone, alclometasone, fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal and mometasone furoate, preferably dexamethasone.
The peptide for use according to claim 25, wherein said another drug is an anti-cancer drug, preferably a proteasome inhibitor.
The peptide for use according to claim 28, wherein said proteasome inhibitor is selected from bortezomib, MLN9708, MLN273, MLN519, NPI- 0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS- 519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS, preferably bortezomib. The peptide for use according to claim 24, wherein said another drug is selected from leuprolide, buserelin, nafarelin, histrelin, goserelin, deslorelin, cetrorelix, ganirelix, abarelix, degarelix, letrozole,
anastrozole, exemestane, vorozole, formestane, fadrozole,
aminoglutethimide, testolactone, tamoxifen, afimoxifene, 4- hydroxytamoxifen, arzoxifene, bazedoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, tamoxifentoremifene,
spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide ketoconazole, finasteride, dutasteride, cyclophosphamide,
mechlorethamine, uramustine, melphalan, chlorambucil, ifosfamide, carmustine, lomustine, streptozocin, busulfan, thiotepa, cisplatin, carboplatin, nedaplatin, oxaliplatin, satraplatin, triplatin tetranitrate, procarbazine, altretamine, dacarbazine, mitozolomide, temozolomide, imatinib mesylate, eriotinib, gefitinib, sunitinib, roscovitine, bevacizumab rapamycin, cyclosporin A, tacrolimus, 5-fluorouracil, methotrexate, etoposide, doxorubicin, actinomycine, vitamin A acid, 13-cis-retinoic acid, 2-chlorodeoxyadenosine, 5-azacitidine, 5-fluorouracil, 6- mercaptopurine, 6-thioguanine, abraxane, accutane, actinomycin-d, adriamycin, adrucil, afinitor, agrylin, ala-cort, aldesleukin, alemtuzumab, alitretinoin, alkaban-aq, alkeran, all-transretinoic acid, alpha interferon, altretamine, amethopterin, amifostine, aminoglutethimide, anagrelide, anandron, anastrozole, arabinosylcytosine, ara-c, aranesp, aredia, arimidex, aromasin, arranon, arsenic trioxide, asparaginase, avastin, azacitidine, bendamustine, bevacizumab, bexarotene, bicalutamide, blenoxane, bleomycin, busulfan, busulfex, calcium leucovorin, campath, camptosar, camptothecin-1 1 , capecitabine, carac, carboplatin, carmustine, carmustine wafer, casodex, cerubidine, cetuximab, chlorambucil, cisplatin, citrovorum factor, cladribine, cortisone, cosmegen, cyclophosphamide, cytadren, cytarabine, cytarabine liposomal, cytosar-U, Cytoxan, dacarbazine, dacogen, dactinomycin, darbepoetin alfa, dasatinib, daunomycin, daunorubicin, daunorubicin hydrochloride, daunorubicin liposomal, daunoxome, decadron, decitabine, delta-cortef, deltasone, denileukin diftitox, depocyt, dexamethasone acetate, dexamethasone sodium phosphate, dexasone, dexrazoxane, dhad, die, diodex, docetaxel, doxil, doxorubicin,
doxorubicin liposomal, droxia, duralone, efudex, eligard, ellence, eloxatin, elspar, emcyt, epirubicin, epoetin alfa, erbitux, eriotinib, erwinia
L-asparaginase, estramustine, ethyol, etopophos, etoposide, etoposide phosphate, eulexin, everolimus, evista, exemestane, fareston, faslodex, femara, filgrastim, floxuridine, fludara, fludarabine, fluoroplex,
fluorouracil, fluorouracil (cream), fluoxymesterone, flutamide, folinic acid, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, gemzar, gleevec, gliadel wafer, goserelin, halotestin, herceptin, hexadrol, hexalen, hexamethylmelamine, hycamtin, hydrea, hydrocort acetate, hydrocortisone, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, hydrocortone phosphate, hydroxyurea, ibritumomab, ibritumomab tiuxetan, idamycin, idarubicin, ifex, ifosfamide, imatinib mesylate, imidazole carboxamide, interferon alfa, interleukin-2, interleukin-1 1 , intron A (interferon alfa-2b), iressa, irinotecan,
isotretinoin, ixabepilone, ixempra, kidrolase (t), lanacort, lapatinib, I- asparaginase, lenalidomide, letrozole, leucovorin, leukeran, leukine, leuprolide, leurocristine, leustatin, liposomal ara-C, liquid pred, lomustine, L-sarcolysin, lupron, lupron depot, matulane, maxidex, mechlorethamine, mechlorethamine hydrochloride, medralone, medrol, megace, megestrol, megestrol acetate, melphalan, mercaptopurine, mesna, mesnex, methotrexate, methotrexate sodium,
methylprednisolone, meticorten, mitomycin, mitomycin-c, mitoxantrone, m-prednisol, mustargen, mustine, mutamycin, myleran, mylocel, mylotarg, navelbine, nelarabine, neosar, neulasta, neumega, neupogen, nexavar, nilandron, nilutamide, nipent, nitrogen mustard, novaldex, novantrone, nplate, octreotide, octreotide acetate, oncospar, Oncovin, ontak, onxal, oprelvekin, orapred, orasone, oxaliplatin, paclitaxel, paclitaxel protein-bound, pamidronate, panitumumab, panretin, paraplatin, pediapred, PEG Interferon, pegaspargase, pegfilgrastim, peg interferon alfa-2b, PEG-L-asparaginase, pemetrexed, pentostatin, phenylalanine mustard, platinol, platinol-AQ, prednisolone, prednisone, prelone, procarbazine, proleukin, prolifeprospan 20 with carmustine implant, purinethol, raloxifene, revlimid, rheumatrex, rituxan, rituximab, romiplostim, rubex, rubidomycin hydrochloride, sandostatin, sandostatin
LAR, sargramostim, solu-cortef, solu-medrol, sorafenib, streptozocin, sunitinib, sutent, tamoxifen, tarceva, targretin, taxol, taxotere, temodar, temozolomide, temsirolimus, teniposide, thalidomide, thalomid, theracys, thioguanine, thioguanine tabloid, thiophosphoamide, thioplex, thiotepa, toposar, topotecan, toremifene, torisel, tositumomab, trastuzumab, treanda, tretinoin, trexall, trisenox, vectibix, velban, vepesid, vesanoid, viadur, vidaza, vinblastine, vinblastine sulfate, vincasar pfs, vincristine, vinorelbine, vinorelbine tartrate, vorinostat, vumon, xeloda, zanosar, zevalin, zinecard, zoladex, zoledronic acid, zolinza and zometa.
A peptide according to any one of claims 1 to 6, for use in skin healing
A method of preventing and/or treating bone- or cartilage disorders, bone fractures, skeletal chondrodysplasias, cancer, autoimmune diseases, fibrotic diseases, inflammatory diseases, obesity, type I and type II diabetes, neurodegenerative diseases, infectious diseases, lung diseases, muscular disorders, aging, infertility, skin atrophy, heart and vascular diseases and metabolic diseases, comprising administering a therapeutically effective amount of a peptide according to any one of claims 1 to 6, to a patient in need thereof.
A method of preventing and/or treating bone- or cartilage disorders, comprising administering a therapeutically effective amount of a peptide according to any one of claims 1 to 6, to a patient in need thereof.
The method according to claim 33, wherein said peptide improves bone growth.
35. The method according to claim 33, wherein said peptide together with insulin-growth factor I improves bone growth.
36. The method according to claim 33, wherein said peptide together with growth hormone improves bone growth.
37. The method according to claim 33, wherein said peptide together with growth hormone improves bone healing. 38. The method according to claim 33, wherein said peptide prevents
development of osteoporosis.
39. The method according to claim 33, wherein said bone- or cartilage
disorder is either a primary or secondary bone- or cartilage disorder.
40. The method according to any one of claims 33 to 39, wherein said bone- or cartilage disorder is drug-induced.
41 . The method according to claim 40, wherein said bone- or cartilage
disorder is drug-induced from drugs used in the treatment of
rheumatological disorders, respiratory disorders, gastrointestinal disorders, cardiovascular disorders, endocrinological disorders, cancer, neurodegenerative disorders, kidney disorders, liver disorders, dermatologic disorders, allergic disorders, inflammatory disorders, metabolic disorders, undesired immune response, undesired
inflammatory response, obesity or diabetes.
42. The method according to claim 40, wherein said drug-induced bone- or cartilage disorder is selected from drug-induced bone growth
impairment, short stature and osteoporosis.
43. The method according to any one of claims 40 to 42, wherein said drug is an anti-inflammatory drug, preferably a glucocorticoid drug. The method according to claim 43, wherein said glucocorticoid drug is selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, doprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone, methylprednisolone aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone,
dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate, beclomethasone
monopropionate, paramethasone, alclometasone, fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal and mometasone furoate, preferably dexamethasone.
The method according to any one of claims 40 to 42, wherein said drug is an anti-cancer drug, preferably a proteasome inhibitor.
The method according to claim 45, wherein said proteasome inhibitor is selected from bortezomib, MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS, preferably bortezomib. 47. The method according to any one of claims 32 to 46, wherein said
peptide is administered in combination with another drug known to induce a bone- or cartilage disorder.
48. The method according to claim 47, wherein said another drug is selected from an anti-inflammatory drug, a selective estrogen receptor modulator drug, an anti-androgen drug, an aromatase inhibitor drug and an anti-cancer drug.
49. The method according to claim 48, wherein said another drug is an antiinflammatory drug, preferably a glucocorticoid drug.
50. The method according to claim 49, wherein said glucocorticoid drug is selected from hydrocortisone, hydrocortisone buteprate, hydrocortisone butyrate, budesonide, ciclesonide, cortisone acetate, deflazacort, medrysone, tixocortol, doprednol, halcinonide, pregnadiene, rimexolone, flunisolide, triamcinolone, amcinonide, fluocinolone acetonide, fluocinonide, fluorometholone, clocortolone, diflucortolone, fluocortin, desoximetasone, prednisone, prednisolone, methylprednisolone, methylprednisolone aceponate, prednicarbate, prednylidene, desonide, fluprednisolone, difluprednate, fluperolone, meprednisone,
dexamethasone, betamethasone, triamcinolone, beclometasone, clobetasone, diflorasone, halometasone, ulobetasol, fludrocortisone acetate, beclomethasone dipropionate, beclomethasone
monopropionate, paramethasone, alclometasone, fluclorolone, flumetasone, fluprednidene, triamcinolone, flunisolide, cortivazol, fluticasone, fluticasone propionate, fluticasone furoate, loteprednol, fludroxycortide, formocortal and mometasone furoate, preferably dexamethasone.
51 . The method according to claim 48, wherein said another drug is an anticancer drug, preferably a proteasome inhibitor.
52. The method according to claim 51 , wherein said proteasome inhibitor is selected from bortezomib, MLN9708, MLN273, MLN519, NPI-0052, Disulfiram, Ritonavir, Lactacystin, CEP-18770, Carfilzomib, PS-519, MG132, MG 262, ALLN, Fellutamide B, Tyropeptin A, Omuralide, Salinosporamide A, Eponeomycine, Epoxomicin, TMC-95A, Syringolin A, Glidobactin A, TMC-95 analogs, NLVS and ZLVS, preferably bortezomib.
The method according to claim 47, wherein said another drug is selected from leuprolide, buserelin, nafarelin, histrelin, goserelin, deslorelin, cetrorelix, ganirelix, abarelix, degarelix, letrozole,
anastrozole, exemestane, vorozole, formestane, fadrozole,
aminoglutethimide, testolactone, tamoxifen, afimoxifene, 4- hydroxytamoxifen, arzoxifene, bazedoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, tamoxifentoremifene,
spironolactone, cyproterone acetate, flutamide, nilutamide, bicalutamide ketoconazole, finasteride, dutasteride, cyclophosphamide,
mechlorethamine, uramustine, melphalan, chlorambucil, ifosfamide, carmustine, lomustine, streptozocin, busulfan, thiotepa, cisplatin, carboplatin, nedaplatin, oxaliplatin, satraplatin, triplatin tetranitrate, procarbazine, altretamine, dacarbazine, mitozolomide, temozolomide, imatinib mesylate, eriotinib, gefitinib, sunitinib, roscovitine, bevacizumab rapamycin, cyclosporin A, tacrolimus, 5-fluorouracil, methotrexate, etoposide, doxorubicin, actinomycine, vitamin A acid, 13-cis-retinoic acid, 2-chlorodeoxyadenosine, 5-azacitidine, 5-fluorouracil, 6- mercaptopurine, 6-thioguanine, abraxane, accutane, actinomycin-d, adriamycin, adrucil, afinitor, agrylin, ala-cort, aldesleukin, alemtuzumab, alitretinoin, alkaban-aq, alkeran, all-transretinoic acid, alpha interferon, altretamine, amethopterin, amifostine, aminoglutethimide, anagrelide, anandron, anastrozole, arabinosylcytosine, ara-c, aranesp, aredia, arimidex, aromasin, arranon, arsenic trioxide, asparaginase, avastin, azacitidine, bendamustine, bevacizumab, bexarotene, bicalutamide, blenoxane, bleomycin, busulfan, busulfex, calcium leucovorin, campath, camptosar, camptothecin-1 1 , capecitabine, carac, carboplatin, carmustine, carmustine wafer, casodex, cerubidine, cetuximab, chlorambucil, cisplatin, citrovorum factor, cladribine, cortisone, cosmegen, cyclophosphamide, cytadren, cytarabine, cytarabine liposomal, cytosar-U, Cytoxan, dacarbazine, dacogen, dactinomycin, darbepoetin alfa, dasatinib, daunomycin, daunorubicin, daunorubicin hydrochloride, daunorubicin liposomal, daunoxome, decadron, decitabine, delta-cortef, deltasone, denileukin diftitox, depocyt, dexamethasone acetate, dexamethasone sodium phosphate, dexasone, dexrazoxane, dhad, die, diodex, docetaxel, doxil, doxorubicin,
doxorubicin liposomal, droxia, duralone, efudex, eligard, ellence, eloxatin, elspar, emcyt, epirubicin, epoetin alfa, erbitux, erlotinib, erwinia
L-asparaginase, estramustine, ethyol, etopophos, etoposide, etoposide phosphate, eulexin, everolimus, evista, exemestane, fareston, faslodex, femara, filgrastim, floxuridine, fludara, fludarabine, fluoroplex,
fluorouracil, fluorouracil (cream), fluoxymesterone, flutamide, folinic acid, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, gemzar, gleevec, gliadel wafer, goserelin, halotestin, herceptin, hexadrol, hexalen, hexamethylmelamine, hycamtin, hydrea, hydrocort acetate, hydrocortisone, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, hydrocortone phosphate, hydroxyurea, ibritumomab, ibritumomab tiuxetan, idamycin, idarubicin, ifex, ifosfamide, imatinib mesylate, imidazole carboxamide, interferon alfa, interleukin-2, interleukin-1 1 , intron A (interferon alfa-2b), iressa, irinotecan,
isotretinoin, ixabepilone, ixempra, kidrolase (t), lanacort, lapatinib, I- asparaginase, lenalidomide, letrozole, leucovorin, leukeran, leukine, leuprolide, leurocristine, leustatin, liposomal ara-C, liquid pred, lomustine, L-sarcolysin, lupron, lupron depot, matulane, maxidex, mechlorethamine, mechlorethamine hydrochloride, medralone, medrol, megace, megestrol, megestrol acetate, melphalan, mercaptopurine, mesna, mesnex, methotrexate, methotrexate sodium,
methylprednisolone, meticorten, mitomycin, mitomycin-c, mitoxantrone, m-prednisol, mustargen, mustine, mutamycin, myleran, mylocel, mylotarg, navelbine, nelarabine, neosar, neulasta, neumega, neupogen, nexavar, nilandron, nilutamide, nipent, nitrogen mustard, novaldex, novantrone, nplate, octreotide, octreotide acetate, oncospar, Oncovin, ontak, onxal, oprelvekin, orapred, orasone, oxaliplatin, paclitaxel, paclitaxel protein-bound, pamidronate, panitumumab, panretin, paraplatin, pediapred, PEG Interferon, pegaspargase, pegfilgrastim, peginterferon alfa-2b, PEG-L-asparaginase, pemetrexed, pentostatin, phenylalanine mustard, platinol, platinol-AQ, prednisolone, prednisone, prelone, procarbazine, proleukin, prolifeprospan 20 with carmustine implant, purinethol, raloxifene, revlimid, rheumatrex, rituxan, rituximab, romiplostim, rubex, rubidomycin hydrochloride, sandostatin, sandostatin LAR, sargramostim, solu-cortef, solu-medrol, sorafenib, streptozocin, sunitinib, sutent, tamoxifen, tarceva, targretin, taxol, taxotere, temodar, temozolomide, temsirolimus, teniposide, thalidomide, thalomid, theracys, thioguanine, thioguanine tabloid, thiophosphoamide, thioplex, thiotepa, toposar, topotecan, toremifene, torisel, tositumomab, trastuzumab, treanda, tretinoin, trexall, trisenox, vectibix, velban, vepesid, vesanoid, viadur, vidaza, vinblastine, vinblastine sulfate, vincasar pfs, vincristine, vinorelbine, vinorelbine tartrate, vorinostat, vumon, xeloda, zanosar, zevalin, zinecard, zoladex, zoledronic acid, zolinza and zometa.
A method of healing skin, comprising administering a therapeutically effective amount of a peptide according to any one of claims 1 to 6.
PCT/EP2012/058770 2011-05-13 2012-05-11 Novel peptides WO2012156310A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SE1150432 2011-05-13
SE1150432-1 2011-05-13

Publications (1)

Publication Number Publication Date
WO2012156310A1 true WO2012156310A1 (en) 2012-11-22

Family

ID=46148836

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/058770 WO2012156310A1 (en) 2011-05-13 2012-05-11 Novel peptides

Country Status (1)

Country Link
WO (1) WO2012156310A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170061084A (en) * 2015-11-25 2017-06-02 연세대학교 원주산학협력단 Pharmaceutical composition for wound healing containing Humanin or analogue thereof as an active ingredient

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
WO2008115525A2 (en) * 2007-03-19 2008-09-25 Ontherix, Inc. Metal-binding therapeutic peptides
WO2011076880A1 (en) * 2009-12-22 2011-06-30 Emma Eriksson Methods and use related to humanin and humanin-like peptides

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
WO2008115525A2 (en) * 2007-03-19 2008-09-25 Ontherix, Inc. Metal-binding therapeutic peptides
WO2011076880A1 (en) * 2009-12-22 2011-06-30 Emma Eriksson Methods and use related to humanin and humanin-like peptides

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
ARIKOSKI, P.; H. KROGER ET AL.: "Disturbance in bone turnover in children with a malignancy at completion of chemotherapy.", MED PEDIATR ONCOL, vol. 33, no. 5, 1999, pages 455 - 461
ARIKOSKI, P.; J. KOMULAINEN ET AL.: "Impaired development of bone mineral density during chemotherapy: a prospective analysis of 46 children newly diagnosed with cancer.", J BONE MINER RES, vol. 14, no. 12, 1999, pages 2002 - 2009
ARIKOSKI, P.; J. KOMULAINEN ET AL.: "Reduced bone density at completion of chemotherapy for a malignancy", ARCH DIS CHILD, vol. 80, no. 2, 1999, pages 143 - 148
BALIS, F. M.: "The Challenge of Developing New Therapies for Childhood Cancers.", ONCOLOGIST, vol. 2, no. 1, 1997, pages I - II
BARON, J.; Z. HUANG ET AL.: "Dexamethasone acts locally to inhibit longitudinal bone growth in rabbits.", AM J PHYSIOL, vol. 263, 1992, pages E489 - 492
BLANEY, S. M.; M. BERNSTEIN ET AL.: "Phase I study of the proteasome inhibitor bortezomib in pediatric patients with refractory solid tumors: a Children's Oncology Group study (ADVL0015", J CLIN ONCOL, vol. 22, no. 23, 2004, pages 4804 - 4809
CHIBA, T.; M. YAMADA ET AL.: "Development of a femtomolar-acting humanin derivative named colivelin by attaching activity-dependent neurotrophic factor to its N terminus: characterization of colivelin- mediated neuroprotection against Alzheimer's disease-relevant insults in vitro and in vivo.", J NEUROSCI, vol. 25, no. 44, 2005, pages 10252 - 10261, XP002532185, DOI: doi:10.1523/JNEUROSCI.3348.05.2005
CLAYTON, P. E.; S. M. SHALET ET AL.: "Growth in children treated for acute lymphoblastic leukaemia", LANCET, vol. 1, no. 8583, 1988, pages 460 - 462
DATABASE Geneseq [Online] 27 November 2008 (2008-11-27), "Metal-binding domain variant humanin peptide, SEQ ID NO:189.", XP002683075, retrieved from EBI accession no. GSP:ATR80906 Database accession no. ATR80906 *
DE LUCA, F.; K. M. BARNES ET AL.: "Regulation of growth plate chondrogenesis by bone morphogenetic protein-2.", ENDOCRINOLOQV, vol. 142, no. 1, 2001, pages 430 - 436
DEMOLY, P.: "Safety of intranasal corticosteroids in acute rhinosinusitis.", AM J OTOLARVNQOL, vol. 29, no. 6, 2008, pages 403 - 413, XP025583140, DOI: doi:10.1016/j.amjoto.2007.11.004
GUO, B.; D. ZHAI ET AL.: "Humanin peptide suppresses apoptosis by interfering with Bax activation.", NATURE, vol. 423, no. 6938, 2003, pages 456 - 461, XP002982057, DOI: doi:10.1038/nature01627
HALTON, J. M.; S. A. ATKINSON ET AL.: "Growth and body composition in response to chemotherapy in children with acute lymphoblastic leukemia", INT J CANCER SUPPL, vol. 11, 1998, pages 81 - 84
HASHIMOTO, Y.; T. NIIKURA ET AL.: "A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer's disease genes and Abeta.", PROC NATL ACAD SCI U S A, vol. 98, no. 11, 2001, pages 6336 - 6341, XP002982058, DOI: doi:10.1073/pnas.101133498
HASHIMOTO, Y.; T. NIIKURA ET AL.: "Detailed characterization of neuroprotection by a rescue factor humanin against various Alzheimer's disease-relevant insults.", J NEUROSCI, vol. 21, no. 23, 2001, pages 9235 - 9245, XP001154175
HASHIMOTO, Y.; Y. ITO ET AL.: "Mechanisms of neuroprotection by a novel rescue factor humanin from Swedish mutant amyloid precursor protein.", BIOCHEM BIOPHVS RES COMMUN, vol. 283, no. 2, 2001, pages 460 - 468, XP002280098, DOI: doi:10.1006/bbrc.2001.4765
HOANG, P. T.; P. PARK ET AL.: "The neurosurvival factor Humanin inhibits beta-cell apoptosis via signal transducer and activator of transcription 3 activation and delays and ameliorates diabetes in nonobese diabetic mice.", METABOLISM, vol. 59, no. 3, 2010, pages 343 - 349, XP026901224, DOI: doi:10.1016/j.metabol.2009.08.001
IKONEN, M.; B. LIU ET AL.: "Interaction between the Alzheimer's survival peptide humanin and insulin-like growth factor-binding protein 3 regulates cell survival and apoptosis.", PROC NATL ACAD SCI U S A, vol. 100, no. 22, 2003, pages 13042 - 13047, XP002338653, DOI: doi:10.1073/pnas.2135111100
JUNG, S. S.; W. E. VAN NOSTRAND: "Humanin rescues human cerebrovascular smooth muscle cells from Abeta-induced toxicity.", J NEUROCHEM, vol. 84, no. 2, 2003, pages 266 - 272
KARIYA, S.; N. TAKAHASHI ET AL.: "Humanin improves impaired metabolic activity and prolongs survival of serum-deprived human lymphocytes.", MOL CELL BIOCHEM, vol. 254, no. 1-2, 2003, pages 83 - 89
KARIYA, S.; N. TAKAHASHI ET AL.: "Humanin inhibits cell death of serum-deprived PC12h cells.", NEUROREPORT, vol. 13, no. 6, 2002, pages 903 - 907, XP009016247, DOI: doi:10.1097/00001756-200205070-00034
KATO, T.; K. IWAMOTO ET AL.: "No association of mutations and mRNA expression of WFS1/wolframin with bipolar disorder in humans.", NEUROSCI LETT, vol. 338, no. 1, 2003, pages 21 - 24, XP002380338, DOI: doi:10.1016/S0304-3940(02)01334-4
LINET, M. S.; L. A. RIES ET AL.: "Cancer surveillance series: recent trends in childhood cancer incidence and mortality in the United States", J NATL CANCER INST, vol. 91, no. 12, 1999, pages 1051 - 1058
LOEB, J. N.: "Corticosteroids and growth.", N ENQL J MED, vol. 295, no. 10, 1976, pages 547 - 552
MAMIYA, T.; M. UKAI: "Gly(14)]-Humanin improved the learning and memory impairment induced by scopolamine in vivo.", BR J PHARMACOL, vol. 134, no. 8, 2001, pages 1597 - 1599, XP001153677, DOI: doi:10.1038/sj.bjp.0704429
MANGANO, J. J.: "A rise in the incidence of childhood cancer in the United States.", INT J HEALTH SERV, vol. 29, no. 2, 1999, pages 393 - 408
MATSUOKA, M.: "Humanin; a defender against Alzheimer's disease?", RECENT PAT CNS DRUG DISCOV, vol. 4, no. 1, 2009, pages 37 - 42
MATSUOKA, M.; Y. HASHIMOTO: "Humanin and the receptors for humanin.", MOL NEUROBIOL, vol. 41, no. 1, 2010, pages 22 - 28, XP009172930, DOI: doi:10.1007/s12035-009-8090-z
MUSE, E. D.; T. K. LAM ET AL.: "Hypothalamic resistin induces hepatic insulin resistance.", J CLIN INVEST, vol. 117, no. 6, 2007, pages 1670 - 1678
MUZUMDAR, H.; M. RAO: "Pulmonary dysfunction and sleep apnea in morbid obesity.", PEDIATR ENDOCRINOL REV, vol. 3, 2006, pages 579 - 583
NISHIMOTO, I.; M. MATSUOKA ET AL.: "Unravelling the role of Humanin.", TRENDS MOL MED, vol. 10, no. 3, 2004, pages 102 - 105, XP009173036, DOI: doi:10.1016/j.molmed.2004.01.001
OBICI, S.; B. B. ZHANG ET AL.: "Hypothalamic insulin signaling is required for inhibition of glucose production.", NAT MED, vol. 8, no. 12, 2002, pages 1376 - 1382
OWEN, H. C.; J. N. MINER ET AL.: "The growth plate sparing effects of the selective glucocorticoid receptor modulator, AL-438.", MOL CELL ENDOCRINOL, no. 1-2, 2007, pages 264
PINKERTON, C. R.: "Avoiding chemotherapy related late effects in children with curable tumours.", ARCH DIS CHILD, vol. 67, no. 9, 1992, pages 1116 - 1119
ROBSON, H.; E. ANDERSON ET AL.: "Chemotherapeutic agents used in the treatment of childhood malignancies have direct effects on growth plate chondrocyte proliferation.", J ENDOCRINOL, vol. 157, no. 2, 1998, pages 225 - 235
SAVENDAHL, L.: "Hormonal regulation of growth plate cartilage.", HORM RES, vol. 64, no. 2, 2005, pages 94 - 97
SHALET, S. M.; D. A. PRICE: "Effect of treatment of malignant disease on growth in children.", ARCH DIS CHILD, vol. 56, no. 3, 1981, pages 235 - 236
SINGH, B. K.; D. D. MASCARENHAS: "Bioactive peptides control receptor for advanced glycated end product-induced elevation of kidney insulin receptor substrate 2 and reduce albuminuria in diabetic mice.", AM J NEPHROL, vol. 28, no. 6, 2008, pages 890 - 899
SPONNE, I.; A. FIFRE ET AL.: "Humanin rescues cortical neurons from prion-peptide-induced apoptosis.", MOL CELL NEUROSCI, vol. 25, no. 1, 2004, pages 95 - 102, XP002508984, DOI: doi:10.1016/j.mcn.2003.09.017
TERASHITA, K.; Y. HASHIMOTO ET AL.: "Two serine residues distinctly regulate the rescue function of Humanin, an inhibiting factor of Alzheimer's disease-related neurotoxicity: functional potentiation by isomerization and dimerization.", J NEUROCHEM, vol. 85, no. 6, 2003, pages 1521 - 1538, XP001154174, DOI: doi:10.1046/j.1471-4159.2003.01797.x
VAN LEEUWEN, B. L.; W. A. KAMPS ET AL.: "Effect of single chemotherapeutic agents on the growing skeleton of the rat.", ANN ONCOL, vol. 11, no. 9, 2000, pages 1121 - 1126
WASSBERG, E.; S. PAHLMAN ET AL.: "The angiogenesis inhibitor TNP-470 reduces the growth rate of human neuroblastoma in nude rats.", PEDIATR RES, vol. 41, no. 3, 1997, pages 327 - 333
XIAN, C. J.; G. S. HOWARTH ET AL.: "Effects of acute 5-fluorouracil chemotherapy and insulin-like growth factor-I pretreatment on growth plate cartilage and metaphyseal bone in rats.", BONE, vol. 35, no. 3, 2004, pages 739 - 749, XP004567964, DOI: doi:10.1016/j.bone.2004.04.027
XIAN, C. J.; J. C. COOL ET AL.: "Cellular mechanisms for methotrexate chemotherapy-induced bone growth defects.", BONE, vol. 41, no. 5, 2007, pages 842 - 850, XP022304112, DOI: doi:10.1016/j.bone.2007.07.021
XU, X.; C. C. CHUA ET AL.: "Humanin is a novel neuroprotective agent against stroke", STROKE, vol. 37, no. 10, 2006, pages 2613 - 2619, XP002713880, DOI: doi:10.1161/01.STR.0000242772.94277.1f
YING, G.; P. IRIBARREN ET AL.: "Humanin, a newly identified neuroprotective factor, uses the G protein-coupled formylpeptide receptor-like-1 as a functional receptor", J !MMUNO!, vol. 172, no. 11, 2004, pages 7078 - 7085
ZAMAN, F.; V. MENENDEZ-BENITO ET AL.: "Proteasome inhibition up- regulates p53 and apoptosis-inducing factor in chondrocytes causing severe growth retardation in mice.", CANCER RES, vol. 67, no. 20, 2007, pages 10078 - 10086
ZAPALA BARBARA ET AL: "Humanins, the neuroprotective and cytoprotective peptides with antiapoptotic and anti-inflammatory properties", PHARMACOLOGICAL REPORTS, vol. 62, no. 5, September 2010 (2010-09-01), pages 767 - 777, XP002683076, ISSN: 1734-1140 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170061084A (en) * 2015-11-25 2017-06-02 연세대학교 원주산학협력단 Pharmaceutical composition for wound healing containing Humanin or analogue thereof as an active ingredient
KR101885591B1 (en) * 2015-11-25 2018-08-06 주식회사 바이오에프디엔씨 Pharmaceutical composition for wound healing containing Humanin or analogue thereof as an active ingredient

Similar Documents

Publication Publication Date Title
US20130053323A1 (en) Methods and use related to humanin and humanin-like peptides
TWI683825B (en) Single-chain trail-receptor agonist proteins
JP2017043604A (en) Amphipathic peptide for blocking thorax air leakage
US20150202314A1 (en) Site-specific labeling and targeted delivery of proteins for the treatment of cancer
US20160129030A1 (en) Treatment of mtor hyperactive related diseases and disorders
US20200031873A1 (en) Repurposing cell penetrating peptides and their novel derivatives and iopromide and iodo-aryl carbonates for treatment of senescence-related diseases and disorders
WO2012156310A1 (en) Novel peptides
US9044448B2 (en) X-ray contrast media compositions and methods of using the same
WO2020201444A1 (en) Repurposing small molecules for senescence-related diseases and disorders
WO2017072080A1 (en) Single-chain tl1a receptor agonist proteins
CA3232856A1 (en) Kras antagonists
WO2012081038A2 (en) Anticancer compounds and targeting cancer with the same
US9044449B2 (en) X-ray contrast media compositions and methods of using the same to treat, reduce or delay the onset of CNS inflammation and inflammation associated conditions
US20160214941A1 (en) Compounds for lnhibition of Unregulated Cell Growth
US20180360784A1 (en) X-ray contrast media compositions and methods of using the same to treat, reduce or delay the onset of migraine
US20230348506A1 (en) Macrocyclic pantetheine derivatives and uses thereof
US11191806B2 (en) Polymyxin-based pharmaceutical composition for treating infectious diseases
US20240024436A1 (en) Method for treating cancer
US20230008508A1 (en) Combination therapies for the treatment of cancer
JP6791977B2 (en) Frostan-3ol derivative as a skeletal muscle hypertrophy agent
WO2012081039A1 (en) Molecules with anticancer activity and uses thereof

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: 12723135

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: 12723135

Country of ref document: EP

Kind code of ref document: A1