CA2328871C - Methods for accelerating bone and cartilage growth and repair - Google Patents

Methods for accelerating bone and cartilage growth and repair Download PDF

Info

Publication number
CA2328871C
CA2328871C CA002328871A CA2328871A CA2328871C CA 2328871 C CA2328871 C CA 2328871C CA 002328871 A CA002328871 A CA 002328871A CA 2328871 A CA2328871 A CA 2328871A CA 2328871 C CA2328871 C CA 2328871C
Authority
CA
Canada
Prior art keywords
seq
group
bone
active agent
ala
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002328871A
Other languages
French (fr)
Other versions
CA2328871A1 (en
Inventor
Kathleen Rodgers
Gere Dizerega
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Southern California USC
Original Assignee
University of Southern California USC
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 University of Southern California USC filed Critical University of Southern California USC
Publication of CA2328871A1 publication Critical patent/CA2328871A1/en
Application granted granted Critical
Publication of CA2328871C publication Critical patent/CA2328871C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/30Insulin-like growth factors (Somatomedins), e.g. IGF-1, IGF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/29Parathyroid hormone (parathormone); Parathyroid hormone-related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/14Angiotensins: Related peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0655Chondrocytes; Cartilage
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/34Sugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/32Angiotensins [AT], angiotensinogen

Abstract

The present invention provides improved methods, kits, and compositions for enhancing bone, cartilage and cartilage repair, bone and prosthesis implantation, and attachment and fixation of cartilage and cartilage to bone or other tissues, and chondrocyte proliferation comprising the administratio n of an effective amount of angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof or AII AT2 type 2 receptor agonists.

Description

METHODS FOR ACCELERATING BONE AND CARTILAGE GROVVTI~ AND
REPAIR
Field of the Invention The present invention relates to methods, compositions, and kits for the repair, regeneration, and implantation of bone and cartilage:
Background of the Invention ~5 Natural mechanisms of repair, healing and augmentation are similar for bone and cartilage. (U.S. Patent No. 5;68,116) . Although repair, healing and augmentation require a complex series of events that are not well defined, it is known that specific, naturally occurring factors are required to achieve these objectives. Such factors are released'or migrate into the injured area, and stimulate osteoblasts and chondrocytes 2o and odontoblasts in bone and cartilage to stimulate matrix formation and remodeling of the wounded area. (ten Dijke et al., BioITechnology, 7:793-798 (1989)) Living bone tissue is continuously being replenished by the processes of resorption and deposition of bone matrix and minerals. This temporally and spatially coupled process, termed bone remodeling, is accomplished largely by two cell 25 populations, the osteocIasts and osteoblasts. (U.S. Patent No. 5,656,59g) The remodeling process is initiated when osteoclasts are recruited from the bone marrow or the circulation to the bone surface and remove a disk-shaped packet of bone. The bone matrix and mineral is subsequently replaced by a team of osteoblasts recruited to the resorbed bone surface from the bone marrow.
Osteoblasts are derived from local mesenchymal (stromal) precursors which differentiate into osteoblasts.
New bone can be formed by three basic mechanisms: osteogenesis, osteoconduction .and osteoinduction. (U.5. Patent No. 5,464,439 ) In osteogenic transplantation,. viable osteoblasts and 1o peri-osteoblasts are moved from one body location to another where they establish centers of bone formation. Cancellous bone and marrow grafts provide such viable cells. TGF beta has been shown to stimulate proliferation and matrix synthesis of osteoblastic cells (Centrella, et al. (1987) J. Biol. Chem. 262:2869-2874) and to inhibit the formation and activity of osteoclastic cells (Chenu, et al. (1988) Proc.
Natl. Acad.
Sci. U.S.A. 85:683-5687; Kiebzak et al. (1988) J. Bone Min. Res. 3:439-446), and to stimulate local bane formation in vivo. (Joyce, et al. (1990) J. Cell. Biol.
110:2195-2207; Noda and Camilliere (1989) Endocrinology 124:2991-2294). Other factors reported to stimulate bone growth include bone .morphogenetic proteins (WO
88/00205), insulin-like growth factor (IGF) (Endocrinol. Metab. 13:E367-72,1986), and parathyroid hormone (J. Bone & Min. Res. 1:377-381, 1986).
Members of the bone morphogenetic protein family have been shown to be useful for induction of cartilage and bone formation. For example, BMP-2 has been shown to be able to induce the formation of new cartilage andlor bone tissue in vivo in a rat ectopic implant model, see U.S. Pat. No. 5,013,649; in mandibular defects in dogs,
2 see Toriumi et ai.; Arch. Otolaryngol Head Neck Surg., 117:1101-1112 (1991);
and in femoral segmental defects in sheep, see Gerhart et al., Trans Orthop Res Soc, 16:172 (1991). Other members of the BMP family have also been shown to have osteogenic activity, including BMP-4, -6 and -7 (see Wozney, Bone Morphogenetic Proteins and Their Gene Expression, in Cellular and Molecular Biology of Bone, pp. 131-167 w (Academic Press, Inc. 1993)). BMP proteins have also been shown to demonstrate inductive and/or differentiation potentiating activity on a variety of other tissues, including cartilage. (U.S. Patent No. 5,700,774) l0 In the transplantation of large segments of cortical bone or allogenic .
banked bone, direct osteogenesis does not occur. Rather, osteoconduction occurs wherein the dead bone acts as a scaffold for the ingrowth of blood vessels, followed by the resorption of the implant and deposition of new bone. This process is very slow however, often requiring years to reunite a large segmental defect.
Osteoinduction is the phenotypic conversion of connective tissue into bone by an appropriate stimulus. As this concept implies, formation of bone can be induced at even non-skeletal sites. Osteoinductioiz is preferred over osteoconduction, as grafts of this type are typically incorporated into the host bone within a two-week period. In contrast, osteoconductive grafts have been found to be non-incorporated as long as one year after implantation. In order to provide an environment suitable for osteoinduction, a material should be selected which is not only capable of inducing osteogenesis throughout its volume, but is also biocompatible, non-inflammatory, and possesses the ability to be ultimately resorbed by the body and replaced with new, natural bone.
3 Among the pathological conditions associated with abnormal bone cell function are osteoporosis, osteoarthritis, Paget's disease, osteohalisteresis, osteomalacia, periodontal disease, bone loss resulting from multiple myeloma and other forms of cancer, bone loss resulting from side effects of other medical treatment (such as steroids), and age-related loss of bone mass. Inadequate organic matrix mass places an individual at risk of skeletal failure such that bone fractures can result from the minimal trauma of everyday life. Such fractures cause significant illness, or morbidity, inasmuch as there is insufficient repair or healing of the fractures. In certain pathologic conditions, osteoclast-mediated resorption is not regulated by osteoblasts but is driven to by cancer cells, infecting organisms or the host's immune cells: In those disease conditions, resorption of bone far exceeds bone formation. Such accelerated osteoclastic activity leads to excessive release of calcium from the inorganic mineral in bone, with a concomitant net Ioss .of skeletal mass, often with an attendant disturbance in calcium homeostasis in the form of elevated blood levels of calcium..
(L7.S. Patent 1s No.5,686,116) Although methods for directing new bone formation are known; improved methods that provide for accelerated bone growth are needed. For example, currently approved therapeutic agents for osteoporosis are antiresorptives. As such, they are not as effective in patients with established osteoporosis of either. type (decreased bone 2o density with fractures of the vertebrae and/or hip), or in patients with Type II
osteoporosis. In addition, the most accepted preventive agent for osteoporosis currently in use is estrogen therapy, which is not an acceptable therapeutic agent for women with a history of breast cancer or endometrial cancer or for men with osteoporosis.
4 Similarly, successful implantation and function of bone implants depends on bonding of the adjacent bone to the implant. (U.S. Patent No. 5,686,116) Such bonding requires bone repair by the formation of new matrix components at the interface between the implant and the bone proximate to the implant. An estimated ten percent of bone and joint prosthetic devices that are placed in people fail to function due to non-bonding of the bone to an implant. The resulting disability, often requires reoperation and reimplantation of the device. Furthermore, five to ten percent of all _ bone fiactures are never repaired. Although many methods have been proposed to cure these non-healing bone fractures, none has yet proven to be satisfactory.
Based on all of 1o the above, there clearly exists a need in the art for improved methods that provide for accelerated bone growth.
Cartilage is a specialized type of dense connective tissue consisting of cells embedded in a matrix. There are several kinds of cartilage. (LJ.S. Patent No.
5,736,372) Translucent cartilage having a homogeneous matrix containing collagenous fibers is found in articular cartilage, in costal cartilages, in the septum of the nose, in larynx and trachea. Articular cartilage is hyaline cartilage covering the articular surfaces of bones. Costal cartilage connects the true ribs and the sternum. Fibrous cartilage contains collagen fibers. Yellow cartilage is a network of elastic fibers holding cartilage cells which is primarily found in the epiglottis, the external ear, and the auditory tube. Cartilage is tissue made up of extracellular matrix primarily comprised of the organic compounds collagen, hyaluronic acid (a proteoglycan), and chondrocyte cells, which are responsible for cartilage production. Collagen, hyaluronic acid and water entrapped within these organic matrix elements yield the unique elastic properties and strength of cartilage.

WO 00/02905 PCT/US99/15735 _ Chondrocytes produce both Type I and Type II collagens. Type II collagen is not found in bone, whereas Type I collagen is found in bone. (U.S. Patent No.
5,686,116) It has previously been shown that the endogenous growth factors TGF beta and BMP
induce both new cartilage and bone formation. Wozney et al. Science, 242:1528-(1988) and Sporn et al. J. Cell Biol. 105:1039-1045 (1987).
In cartilage, collagen synthesis is required for repair, healing and augmentation, as well as for the successful bonding of grafts and prosthetic devices. (U.S.
Patent No.
5,686,116) Collagen is the major structural protein responsible for the architectural integrity of cartilage. Thus, an adequate supply of chondrocytes is essential in order to t o produce sufficient amounts of collagen for repair, healing, and augmentation of cartilage. Other, noncollagen proteins, such as osteonectin, fibronectin and proteoglycans are also important for cartilage repair.
Cells such as synoviocytes that are found in joint spaces adjacent to cartilage have an important role in cartilage metabolism. Synoviocytes produce metallo-~5 proteinases, such as collagenases that are capable of breaking-down cartilage. TGF beta is known to inhibit cell-release (and probably synthesis) of metallo-proteinases and to induce chondrocytes (cartilage forming cells) to produce new matrix components and inhibit production of cartilage destructive enzymes so as to effect cartilage repair, healing and augmentation. Spom et al. ( 1987). It has also been shown that mice 2o deficient in parathyroid hormone-related peptide (PTIirP) exhibit abnormal cartilage maturation, indicating that PTHrP is an essential factor for chondrocyte development and maturation. (U.S. Patent No. 5,700,774) Cartilage implants are often used in reconstructive or plastic surgery such as rhinoplasty. There is a need in the art for methods that increase chondrocyte
6 WO 00/02905 PCT/US99/15735 _ proliferation and collagen synthesis, and thus inhibit cartilage destruction and enhance cartilage repair. Such methods would increase the clinical utility of cartilage repair including but not limited to cartilage grafts and healing of cartilage grafts.
Although some of the above methods have met with limited success, there remains a need in the art for improved methods for enhancing bone and cartilage repair, healing and augmentation, and for enhancing the attachment and fixation of bone and cartilage implants.
Summary of the Invention l0 The present invention provides methods, kits, and compositions for 1 ) enhancing bone and cartilage repair; 2) bone and prosthesis implantation; 3) attachment and fixation of cartilage to bone or other tissues; and methods, cell culture medium and kits for the proliferation of chondrocytes; all of which comprise the administration of angiotensinogen, angiotensin I {AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), All analogues, All fragments or analogues thereof or All ATZ type 2 receptor agonists.
These aspects and other aspects of the invention become apparent in light of the following detailed description.
Brief Description of the Figures Figure 1 is a bar graph showing the effect of AII, AIII, and GSD37B (10 p,g/ml) on chondrocyte proliferation.
Figure 2 is a bar graph showing the effect of AII, GSD36, GSD37B, GSD38B, and GSD28 (10 p.g/ml) of the invention on chondrocyte proliferation.
7 Figure 3 is a bar graph showing the effect of AII, 1GD, 2GD, and 3GD (10~g/ml) on chondrocyte proliferation.
Figure 4 is a bar graph showing the effect of AII, AII(1-7), GSD22A, GSD24B, and GSD28 (10 p,glml) of the invention on chondrocyte proliferation.
w Detailed Description of the Preferred Embodiments Within this application, unless otherwise stated, the techniques utilized may be to found in any of several well-known references such as: Molecular Cloning: A
Laboratory Manual (Sambrook, et al., 1989, Cold Spring Harbor Laboratory Press), Gene Expression Technology (Methods in Enzymology, Vol. 185, edited by D.
Goeddel, 1991. Academic Press, San Diego, CA), "Guide to Protein Purification"
in Methods in Enrymology (M.P. Deutshcer, ed., (1990) Academic Press, Inc.); PCR
IS Protocols: A Guide to Methods and Applications (Innis, et al. 1990.
Academic Press, San Diego, CA), Culture of Animal Cells: A Manual of Basic Technique, 2"d Eki.
(R.I.
Freshney. 1987. Liss, Inc. New York, NY), Gene Transfer and Expression Protocols, pp. 109-128, ed. E.J. Murray, The Humana Press Inc., Clifton, N.J.), and the Ambiori 1998 Catalog (Ambion, Austin, TX).
2o As defined herein the phrase "enhancing bone repair" refers to increasing the rate of new bone formation via bone remodeling, osteogenesis, osteoconduction and/or osteoinduction. The methods for enhancing bone repair in a mammal of the invention include those that stimulate bone formation and those that reverse bone loss.
The methods can thus be used for (1) providing a subject with an amount of a substance
8 WO 00/02905 PCT/US99/15735 _ sufficient to act prophylactically to prevent the development of a weakened and/or unhealthy state; or (2) providing a subject with a sufficient amount of a substance so as to alleviate or eliminate a disease state and/or the symptoms of a disease state, and a weakened and/or unhealthy state.
As used herein the term "enhancing cartilage repair" comprises healing and regeneration of cartilage injuries, tears, deformities or defects, and prophylactic use in preventing damage to cartilaginous tissue.
The present invention fulfills the need for methods to enhance bone repair in a mammal suffering from bone fractures, defects, and disorders which result in weakened bones such as osteoporosis, osteoarthritis, Paget's disease, osteohalisteresis, osteomalacia, periodontal disease, bone loss resulting from multiple myeloma and other forms of cancer, bone loss resulting from side effects of other medical treatment (such as steroids), and age-related loss of bone mass. In addition, bony ingrowth into various prosthetic devices can be greatly enhanced so that such artificial parts are firmly and t5 permanently anchored into the surrounding skeletal tissue through a natural osseous bridge.
The present invention further fulfills the need for methods to enhance the repair of cartilage in a mammal, by accelerating the proliferation of chondrocytes and thereby increasing the synthesis of collagen for use in cartilage repair. Such methods have 2o application in the healing of cartilage, for example articular cartilage tears, deformities and other cartilage defects in humans and other animals. The methods have prophylactic use in preventing damage to cartilaginous tissue, as well as use in the improved fixation of cartilage to bone or other tissues, and in repairing defects to cartilage tissue. De novo cartilaginous tissue formation induced by the compounds of
9 the present invention contributes to the repair of congenital, trauma induced, or other cartilage defects of other origin, and is also useful in surgery for attachment or repair of cartilage. The methods and compositions of the invention may also be useful in the treatment of arthritis and other cartilage defects. The methods of the present invention can also be used in. other indications wherein it is desirable to heal or regenerate cartilage tissue. Such indications include; without limitation, regeneration or repair of injuries to the articular cartilage. The methods of the present invention provide. an environment to attract cartilage-forming cells, stimulate growth of cartilage-forming cells or induce differentiation of progenitors of cartilage-forming cells and to chondrocytes.
The methods and kits of the present invention also pmvide improved chemically defined medium for accelerating the proliferation of chondrocytes (cartilage-forming cells). In another embodiment, the compositions and methods of the present invention can be used to treat chondrocytic cell lines, such as articular chondrocytes, in order to maintain chondrocytic phenotype and survival of the cells. The treated cell populations are therefore also useful for gene therapy applications.
U.S. Patent No. 5,015,629 to DiZerega describes a method for increasing the rate of healing of wound tissue, comprising the application to such tissue of angiotensin II
(AII) in an 2d amount which is sufficient for said increase. The application of All to wound tissue significantly increases the rate of wound healing, leading to a more rapid re-epithelialization and tissue repair. The term All refers to an octapeptide present in humans and other species having the sequence Asp-Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:I]. The biological formation of angiotensin is initiated by the action of renin on the plasma substrate angiotensinogen (Clouston et al., Genomics 2:240-( 1988); Kageyama et al, Biochemistry 23:3603-3609; Ohkubo et al., Proc. Natl.
Acad Sci. 80:2196-2200 (1983) The substance so formed is a decapeptide called angiotensin I (AI) which is converted to All by the angiotensin converting enzyme (ACE) which removes the C-terminal His-Leu residues from AI ESEQ ID NO: 37]. All is a known pressor agent and is commercially available.
Studies have shown that All increases mitogenesis and chemotaxis in cultured cells that are involved in wound repair, and also increases their release of growth factors and extracellular matrices (diZerega, U.S. Patent No. 5,015,629; Dzau et. al., J.
Mol. Cell. Cardiol. 21:57 (Supp III) 1989; Berk et. al., Hypertension 13:305-14 (/989);
Kawahara, et al., BBRC 150:52-9 (1988); Naftilan, et al., J. Clin. Invest.
83:1419-23 (1989); Taubman. et al., J. Biol. Chem. 264:526-530 (1989); Nakahara, et al., BBRC
184:811-8 (1992); Stouffer and Owens, Circ. Res. 70:820 (1992); Woliy et al., Arn. ,J.
Pathol. 140:95-107 (1992); Bell and Madri, Am. J. Pathol. 137:7-12 (1990). In addition, All was shown to be angiogenic in rabbit corneal eye and chick chorioallantoic membrane models (Fernandez, et al., J. Lab. Clin. Med. 105:141 (1985); LeNoble, et al., Eur. J. Pharmacol. 195:305-6 (1991). Additionally, All and angiotensin III analogs and fragments thereof have been shown to be effective in wound healing. (U.5. Patent No. 5,629,292; International Application No. WO
95/08565; International Application WO 95/08337; International Application No.
WO
96/39164; ) Previous studies have suggested that angiotensin I (AI) and All both stimulate bone resorption in vitro by osteoclasts incubated on bone slices, but only in the WO 00/02905 PCT/US99/15735 _ presence of osteoblastic cells, suggesting that the effect of angiotensin II
was not direct, but rather is mediated by a primary hormonal interaction on cells of the osteoblastic lineage. (Hatton et al., J. Endocrinol. 152:5-10 (1997)). AI stimulation of bone resorption was inhibited by ACE inhibitors, suggesting that the formation of All from AI was responsible for the stimulation of bone resorption. Neither AI nor All were shown to have any effect on osteoclast formation. Thus, this study suggests that local bone destruction may be mediated by AII's stimulation of bone resorption.
Other studies have demonstrated All stimulation of DNA and collagen synthesis in vitro on primary cultures of isolated, phenotypically immature osteoblasts derived from the periosteum of fetal rat calvaraiae and human adult trabecular bone.
(Lamparter et al., J. Cell. Physiol. 175:89-98 (1998)) No direct All effect was detected on primary cell populations with a mature osteoblast phenotype, and an indirect effect through AII-responsive osteoblastic precursor cells was proposed. Similar in vitro studies on osteoblast-rich populations of cells demonstrated a similar effect, while not ruling out stimulation of mature osteoblast proliferation. (Hiruma et al., Biochem and Biophys. Res. Commun. 230:176-178 (1997)) Another study suggests that All may decelerate the differentiation and bone formation of rat calvarial osteoblasts.
(Hagiwara et al., 3. of Endocrinology 156:543-550 (1998)) Based on all of the above studies, there is no expectation that the use of 2o angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, AII, All analogues, All fragments or analogues thereof or All ATZ type 2 receptor agonists would be effective in enhancing bone and cartilage repair, or effective in accelerating chondrocyte proliferation and collagen synthesis.

Previous studies in our laboratory have demonstrated that a class of All and All analogues and fragments stimulate the proliferation of mesenchymal stem cells, which give rise to the cells that make up bone and cartilage.
A peptide agonist selective for the AT2 receptor (AII has 100 times higher affinity for AT2 than AT1) has been identified. This peptide is p-aminophenylalanine 6-All ["(p-NH2-Phe)6-AII)"], Asp-Arg-Val-Tyr-Ile-xaa-Pro-Phe [SEQ ID N0.36]
wherein Xaa is p-NHZ-Phe.(Speth and Kim, BBRC 169:997-1006 (1990). This peptide gave binding characteristics comparable to AT2 antagonists in the experimental models to tested (Catalioto, et al., Eur. J. Pharmacol. 256:93-97 (1994); Bryson, et al., Eur. J.
Pharmacol. 225:119-127 (1992).
The effects of All receptor and All receptor antagonists have been examined in two experimental models of vascular injury and repair which suggest that both All receptor subtypes (AT1 and AT2) play a role in wound healing (Janiak et al., Hypertension 20:737-45 (1992); Prescott, et al., Am. J. Pathol. 139:1291-1296 (I991);
Kauffrnan, et al., Life Sci. 49:223-228 (1991); Viswanathan, et al:, Peptides 13:783-786 .
(1992); Kimura, et al., BBRC 187:1083-1090 (1992).
Many studies have focused upon AII(1-7) (AII residues 1-7) or other fragments of All to evaluate their activity. AII(1-7) elicits some, but not the full range of effects 2o elicited by AII. Pfeilschifter, et al., Eur. J. Pharmacol. 225:57-62 (1992); Jaiswal, et al., Hypertension 19(Supp. II):II-49-II-55 (1992); Edwards and Stack, J.
Pharmacol.
Exper. Ther. 266:506-510 (1993); Jaiswal, et al., J. Pharmacol. Exper. Ther.
265:664-673 (1991); Jaiswal, et al., Hypertension 17:1115-1120 (1991); Portsi, et a., Br. .I.
Pharmacol. 111:65?-654 {1994).

WO 00/02905 PGT/US99/15735 _ As hereinafter defined, a preferred class of AT2 agonists for use in accordance with the present invention comprises angiotensinogen, angiotensin I (AI), AI
analogues, AI fragments and analogues thereof, AII, All analogues, All fragments or analogues thereof or All ATZ type 2 receptor agonists having p-NH-Phe in a position s corresponding to a position 6 of AII. In addition to peptide agents, various nonpeptidic agents (e.g., peptidornirnetics) having the requisite AT2 agonist activity are further contemplated for use in accordance with the present invention.
The active All analogues, fragments of All and analogues thereof of particular interest in accordance with the present invention comprise a sequence consisting of at 1o least three contiguous amino acids of groups R'-R8 in the sequence of general formula I
R~ _Rz_R3_Ra_Rs_R6_R7~Rs in which Rt and RZ together form a group of formula X-R''-RB-, 15 wherein X is H or a one to three peptide group, or is absent, RA is suitably selected from H, Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2GIy, Pro, Bet, GIu(NH2), Gly, Asp(NH2) and Suc, RB is suitably selected from Arg, Lys, Ala, Orn, Citron, Ser(Ac), Sar, D-2o Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(P03)Z, Thr, Ala, Ser, homoSer and azaTyr;

WO 00/02905 PCT/US99/15735 _ RS is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NHZ-Phe;
R7 is Pro or Ala; and R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group, or is absent.
Compounds falling within the category of AT2 agonists useful in the practice of the invention include the All analogues set forth above subject to the restriction that R6 is p-NHz-Phe.
1 o Particularly preferred combinations for R" and RB are Asp-Arg, Asp-Lys, Glu-Arg and Glu-Lys. Particularly preferred embodiments of this class include the following: AII, AIII or AII(2-8), Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID N0:2];
AII(3-8), also known as desl-AIII or AIV, Val-Tyr-Ile-His-Pro-Phe [SEQ ID N0:3];
AII(1-7), Asp-Arg-Val-Tyr-Ile-His-Pro {SEQ ID N0:4]; AII(2-7). Arg-Val-Tyr-Ile-His-Pro is [SEQ ID NO:SJ; AII(3-7), Val-Tyr-Ile-His-Pro [SEQ ID N0:6]; AII(S-8), Ile-His-Pro-Phe [SEQ ID N0:7]; AII(1-6), Asp-Arg-Val-Tyr-Ile-His [SEQ ID N0:8]; AII(1-5), Asp-Arg-Val-Tyr-Ile [SEQ ID-N0:9]; AII(1-4), Asp-Arg-Val-Tyr [SEQ ID NO:10];
and AII(1-3), Asp-Arg-Val [SEQ ID NO:11]. Other preferred embodiments include:
Arg-norLeu-Tyr-Ile-His-Pro-Phe [SEQ ID N0:12] and Arg-Val-Tyr-norLeu-His-Pro-2o Phe [SEQ ID N0:13]. Still another preferred embodiment encompassed within the scope of the invention is a peptide having the sequence Asp-Arg-Pro-Tyr-Ile-His-Pro-Phe [SEQ ID N0:31 ]. AII(6-8), His-Pro-Phe [SEQ ID N0:14] and AII(4-8), Tyr-Ile-His-Pro-Phe [SEQ ID NO:15] were also tested and found not to be effective.

WO 00/02905 PCTNS99/15735 _ In a particularly preferred embodiment of the methods for chondrocyte proliferation, collagen synthesis, cartilage repair, and attachment and fixation of cartilage to bone or other tissues, the active compounds of the present invention are selected from those comprising the following general formula:
R1-R2-R3-R4-RS-His-Pro-R6, wherein Rl is selected from the group consisting of Hydrogen, Gly, and Asp;
R2 is selected from the group consisting of Arg, Citron, or Ornithine;
R3 is selected from the group consisting of Val, Ile, Ala, Leu, and norLeu, or Pro;
to R4 is selected from Tyr, Tyr(P03)2, and Ala;
RS is selected from the group consisting of Ile, Ala, Val, Leu, and norLeu;
and R6 is Phe, Ile, or is absent.
Most particularly preferred embodiments of this class of compounds are selected from the group consisting of SEQ ID NO:1, SEQ ID N0:2, SEQ ID N0:4, SEQ ID NO: 13, SEQ ID NO: 18, SEQ ID N0:19, SEQ ID N0:12, SEQ ID N0:24, SEQ ID N0:26, SEQ ID NO: 32, SEQ ID N0:33, SEQ ID N0:38, SEQ ID N0:39, SEQ ID N0:40, SEQ ID N0:41, SEQ ID N0:42, SEQ ID N0:43, SEQ ID N0:44, and SEQ ID N0:45.
In a particularly preferred embodiment of the methods for bone repair and bone 2o and prosthesis implantation, the active compounds of the present invention are selected from those comprising the following general formula:
Asp-Arg-R1-R2-Ile-His-Pro-R2, wherein R1 is selected from the group consisting of Ile, Pro, Ala, Val, Leu, and norLeu;
R2 is selected from Tyr and Tyr(P03)2; and WO 00/02905 PCT/US99/15735 _ R3 is Phe, or is absent.
Most particularly preferred embodiments of this class of compounds are selected from the group consisting of SEQ ID NO:1, SEQ ID N0:4, SEQ ID N0:24, SEQ ID N0:31, SEQ ID N0:32, SEQ ID NO: 33, SEQ ID N0:41, and SEQ ID NO:
45.
Another class of compounds of particular interest in accordance with the present invention are those of the general formula II
RZ_Rs_Ra_Rs_R6_R~_Rs in which R2 is selected from the group consisting of H, Arg, Lys, Ala, l0 Orn, Citron, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(P03)2, Thr, Ser, homoSer, Ala, and azaTyr;
RS is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NHZ-Phe;
R' is Pro or Ala; and Rs is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
' A particularly preferred subclass of the compounds of general formula II has the formula RZ-R3-Tyr-RS-His-Pro-Phe [SEQ ID N0:16]

WO 00/02905 PCT/US99/15735 _ wherein R2, R3 and RS are as previously defined. Particularly preferred is angiotensin III of the formula Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID N0:2).
Other preferred compounds include peptides having the structures Arg-Val-Tyr-Gly-His-Pro-Phe [SEQ ID N0:17] and Arg-Val-Tyr-Ala-His-Pro-Phe [SEQ ID N0:18]. The s fragment AII(4-8) was ineffective in repeated tests; this is believed to be due to the exposed tyrosine on the N-terminus.
In the above formulas, the standard three-letter abbreviations for amino acid residues are employed. In the absence of an indication to the contrary, the L-form of the amino acid is intended. Other residues are abbreviated as follows:
t o TABLE 1 Abbreviation for Amino Acids Me2Gl N,N-dimeth 1 1 c 1 Bet 1-carboxy-N,N,N-trimethylmethanaminium hydroxide inner salt betaine Suc Succin 1 Phe Br -bromo-L- hen lalan 1 azaTyr aza-a'-homo-L-t os 1 Ac c 1-aminoc clo entane carbox lic acid Aib 2-aminoisobut 'c acid Sar N-meth 1 1 c 1 sarcosine Orn Ornithine It has been suggested that All and its analogues adopt either a gamma or a beta turn (Regoli, et al., Pharmacological Reviews 26:69 (1974). In general, it is believed that neutral side chains in position R3, RS and R7 may be involved in maintaining the WO 00/02905 PCT/US99/15735 _ appropriate distance between active groups in positions R4, R6 and Rg primarily responsible for binding to receptors and/or intrinsic activity. Hydrophobic side chains in positions R3, RS and Rg may also play an important role in the whole conformation of the peptide and/or contribute to the formation of a hypothetical hydrophobic pocket.
Appropriate side chains on the amino acid in position R2 may contribute to affinity of the compounds for target receptors and/or play an important role in the conformation of the peptide. For this reason, Arg and Lys are particularly preferred as R2.
For purposes of the present invention, it is believed that R3 may be involved in 1 o the formation of linear or nonlinear hydrogen bonds with RS (in the gamma turn model) or R6 (in the beta turn model). R3 would also participate in the first turn in a beta antiparallel structure (which has also been proposed as a possible structure).
In contrast to other positions in general formula I, it appears that beta and gamma branching are equally effective in this position. Moreover, a single hydrogen bond may be sufficient to maintain a relatively stable conformation. Accordingly, R3 may suitably be selected from Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr.
With respect to R4, conformational analyses have suggested that the side chain in this position (as well as in R3 and RS) contribute to a hydrophobic cluster believed to be essential for occupation and stimulation of receptors. Thus, R4 is preferably selected 2o from Tyr, Thr, Tyr (P03)2, homoSer, Ser and azaTyr. In this position, Tyr is particularly preferred as it may form a hydrogen bond with the receptor site capable of accepting a hydrogen from the phenolic hydroxyl (Regoli, et al. (1974), supra). R3 may also be suitably Ala.

WO 00/02905 PCT/US99/15735 _ In position R5, an amino acid with a (3 aliphatic or alicyclic chain is particularly desirable. Therefore, while Gly is suitable in position R5, it is preferred that the amino acid in this position be selected from Ile, Ala, Leu, norLeu, Gly and Val.
In the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, All analogues, fragments and analogues of fragments of particular interest in accordance with the present invention, R6 is His, Arg or 6-NHz-Phe. The unique properties of the imidazole ring of histidine (e.g., ionization at physiological pH, ability to act as proton donor or acceptor, aromatic character) are believed to contribute to its particular utility as R6. For example, conformational models suggest that His may ~o participate in hydrogen bond formation (in the beta model) or in the second turn of the antiparallel structure by influencing the orientation of R'. Similarly, it is presently considered that R' should be Pro in order to provide the most desirable orientation of R8. In position Rg, both a hydrophobic ring and an anionic carboxyl terminal appear to be particularly useful in binding of the analogues of interest to receptors;
therefore, Tyr and especially Phe are preferred for purposes of the present invention.
Analogues of particular interest include the following:

Angiotensin II Analogues All Amino Acid Sequence Analogue Sequence Identifier ~~
Name Analoue As -Ar -Val-T-Val-His-Pro-Phe SE ID NO:

Analoue Asn-Ar -Val-T-Val-His-Pro-Phe SE ID NO:

Analoue Ala-Pro-Gl -Ar -Ile-T r-Val-His-Pro-PheSE ID NO:
3 -As 21 Analoue Glu-Ar -Val-Tr-Ile-His-Pro-Phe SE ID NO:

Analoue As -L s-Val-T-Ile-His-Pro-Phe SE ID NO:

Analoue As -Ar -Ala-Tr-Ile-His-Pro-Phe SE ID NO:

Analoue As -Ar -Val-Thr-Ile-His-Pro-Phe SE ID
NO:

-Analogue Asp-Arg-Val-Ty_ EQ ID NO:
8 r-Leu-His=Pro-Phe ~ 26 Analoue As -Ar -Val-T-Ile-ArQ-Pro-Phe SE ID NO: 27 Analoa As -Ar -VaI-T-Ile-His-Ala-Phe SE ID NO: 28 Analoue As -Ar -Val-T-Ile-His-Pro-T SE ID NO: 29 Analoue Pro-Ar -IIe-His-Pro-Phe SE LD NO: 30 12 -Val-T

Analoue As -Ar -Pro-T-Ile-His-Pro-Phe SE ID NO: 31 Analoue As -Ar -Val-TP03 -Ile-His-Pro-Phe SE ID NO: 32 Analoa A -Ar -norLeu-T SE ID NO: 33 -Ile-His-Pro-Phe Analoue As -Ar -Val-T-riorLeu-His-Pro-Phe SE ID NO: 34 Analoue A -Ar -Val-homoSer-T SE ID NO: 3 5 17 -Ile-His-Pro-Phe The polypeptides of the instant invention may be synthesized by any conventional method, including, but not limited to, those set forth in J. M.
Stewart and J. D. Young, Solid Phase Peptide Synthesis, 2nd ed., Pierce Chemical Co., Rockford, 5 Ill. (1984) and J. Meienhofer, Hormonal Proteins and Peptides, Vol. 2, Academic Press, New York, (1973) for solid phase synthesis and E. Schroder and K.
Lubke, The Peptides, Vol. 1, Academic Press, New York, (1965) for solution synthesis.
In general, these methods involve the sequenfial addition of protected amino to acids to a growing peptide chain (LJ.S. Patent No. 5,693,616, ) Normally, either the amino or carboxyl group of the first amino acid and any reactive side chain group are protected. This protected amino acid is then either attached to an inert solid support, or utilized in solution, arid the next amino acid. in the sequence, also suitably protected, is added under conditions amenable t5 to formation of the amide linkage. After all the desired amino acids have been linked in the proper sequence, protecting groups and any solid support are removed to afford the crude polypeptide. The polypeptide is desalted and purified, preferably chromatob aphically, to yield. the final product.
Preferably, peptides are synthesized according to standard solid-phase zo methodologies, such as may be performed on an Applied Biosystems Model 430A

peptide synthesizer (Applied Biosystems, Foster City, Calif.), according to manufacturer's instructions. Other methods of synthesizing peptides or pepddomimetics, either by solid phase methodologies or in liquid phase, are well known to those skilled in the art.
In one aspect, the present invention provides methods and kits for enhancing bone and cartilage repair, implantation, and augmentation in a mammal comprising the administration of angiotensinogen, angiotensin I (AI), AI analogues, AI
fragments and analogues thereof, angiotensin II (AIn, All analogues, All fragments or analogues thereof or All AT2 type 2 receptor agonists (hereinafter referred to as "active agents").
The compounds can be administered alone or ~ in combination with other compounds that enhance bone and/or cartilage repair, implantation, and augmentation, including but not limited to bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, , insulin-like growth factor, and parathyroid hormone.
The active agents may be administered by any suitable route, including orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles.
Such vehicles may include a tantalum or hydroxyapatite scaffold as a vehicle with the compounds of the invention embedded therein. Alternatively, polymeric substrates can be used for compound delivery to the bone or cartilage such as the polymeric substrates disclosed in U.S. Patent Nos. 5,443,515; 5,171,273; 5,607,474; 4,916,207; ~
and 5,324,775. The term parenteral as used herein includes topical (i.e.; placement in to the bone), subcutaneous, intravenous, WO 00/02905 PCT/US99/15735 _ intraarterial, intramuscular, intrasternal, intratendinous, intraspinal, intracranial, intrathoracic, infusion techniques or intraperitoneally.
The active agents of the present invention can also be incorporated into a coating on the surface of a prosthetic device. Such coatings may be composed of a polymer that allows slow diffusion of the active agents at a rate sufficient to enhance bone attachment for a suitable period of time. Suitable coatings include, but are not limited to, hydroxyapatite, methacrylate and tricalcium phosphate. Further, the polymeric coatings can be applied only to the sites on the prosthetic device where bony ingrowth is desired. Bone grafts can be coated with or soaked or immersed in a rinse or 1o gel prior to implantation so as to impregnate the graft with the active agents. Topical or local administration of the active agents to either the site of implantation or the implant itself, is preferred, as it diminishes drug exposure for tissues and organs not requiring treatment.
The active agents may be made up in a solid form (including granules, powders or suppositories) or in a liquid form (e.g., solutions, suspensions, or emulsions). The compounds of the invention may be applied in a variety of solutions. Suitable solutions for use in accordance with the invention are sterile, dissolve sufficient amounts of the peptide, and are not harmful for the proposed application. In this regard, the compounds of the present invention are very stable but are hydrolyzed by strong acids 2o and bases. The compounds of the present invention are soluble in organic solvents and in aqueous solutions at pH 5-8.
The active agents may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc.

For administration, the active agents are ordinarily combined with one or more adjuvants appropriate for the indicated route of administration. The compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulphuric acids, acacia, gelatin, sodium alginate, polyvinylpyrrolidine, and/or polyvinyl alcohol, and tableted or encapsulated for conventional administration.
Alternatively, the compounds of this invention may be dissolved in saline, water, polyethylene glycol, propylene glycol, carboxymethyl cellulose colloidal solutions, ethanol, corn oil, peanut oil, cottonseed oil, sesame oil, tragacanth gum, and/or various 1o buffers. Other adjuvants and modes of administration are well known in the pharmaceutical art. The carrier or diluent may include time delay material, such as glyceryl monostearate or glyceryl distearate alone or with a wax, or other materials well known in the art.
Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose.
The dosage regimen for enhancing bone and cartilage repair with the active agents is based on a variety of factors, including the type of injury, the age, weight, sex, 2o medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely by a physician using standard methods.
Dosage levels of the order of between 0.1 ng/kg and 10 mg/kg body weight of the active agents are useful for all methods of use disclosed herein.
za WO 00/02905 PCTNS99/15735.
As an example, the dosage regimen for accelerating bone and cartilage repair with the active agents wherein the compounds are embedded in a scaffold used for bone and/or cartilage, such as tantalum or hydroxyappetite, would be based upon the volume of bone to be filled and not upon the weight of the subject being treated.
Following s determination of the specific dose volume to be administered to the subject, calculated based on the defect, individual doses are prepared. The final delivered drug product will be mixed under aseptic conditions at a ratio of: 1 part (20%) of the active agents (in a concentration of between about .001 wg/ml to about 5 mg/ml) to 4 parts (80%) diluent.
1o The treatment regime will also vary depending on the disease being treated, based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. For example, the active agents are administered to an osteoporosis patient for up to 30 days. The therapy is administered I S for 1 to 6 times per day at dosages as described above.
In a preferred embodiment, the active agent is administered subcutaneously. A
suitable subcutaneous dose of active ingredient of active agent is preferably between about 0:1 ng/kg and about 10 mg/kg administered twice daily for a time sufficient to enhance bone or cartilage repair. In a more preferred embodiment, the concentration of 2o active agent is between about 100 ng/kg body weight and about 10.0 mg/kg body weight. In a most preferred embodiment, the concentration of active agent is between about 10 pg/kg body weight and about 10.0 mg/kg body weight. This dosage regimen maximizes the therapeutic benefits of the subject invention while minimizing the WO 00/02905 PCTIUS99/15735.
amount of agonist needed. Such an application minimizes costs as well as possible deleterious side effects.
For subcutaneous administration, the active ingredient may comprise from 0.0001% to 10% w/w, e.g., from 1% to 2% by weight of the formulation, although it may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1 % to 1 % of the formulation.
In another preferred embodiment of the present invention, the active agent is administered topically at the site of bone or cartilage loss or repair.
Suitable topical doses and active ingredient concentration in the formulation are as described for subcutaneous administration.
In another preferred embodiment, the active agent is administered at the desired site of bone or cartilage repair, such as via capsule delivery in gingival tissue at sites of bone resorption or in a scaffold surgically implanted at the site of a non-union bone fracture. Suitable doses and active ingredient concentration in the formulation are as described for subcutaneous administration.
In one embodiment of the invention, ex vivo methods are presented for enhancing bone repair via isolation of osteoblastic cell populations from a subject, contacting the isolated cell population with angiotensinogen, AI, AI
analogues, AI
fragments and analogues thereof, AII, All analogues, All fragments and analogues thereof and/or All ATZ type 2 receptor agonist, and subsequent reinfusion of the osteoblastic cell population into the subject. Methods for the isolation of osteoblastic cell population have been described. (Hiruma et al., 1997; Lamparter et al., 1998) In a preferred embodiment, human bone cells are cultured from outgrowths of trabecular bone fragments of the femoral head from patients undergoing hip replacement due to fracture, and treated by several consecutive collagenase digestion periods, using 1 mg/ml type II collagenase solution (Worthington Diagnostic Systems) for 20 minutes per digestion period.
In another embodiment of the invention, ex vivo or in vitro methods are presented for enhancing cartilage repair via isolation of chondrocyte cell populations from a subject, contacting the isolated cell population with the active agent, and subsequent reinfusion of the chondrocyte cell population into the subject.
Methods for the isolation of chondrocyte cell populations have been described. (Kato et al., J. Cell Biol., vol. 100, pages 477-485 (1985); U.S. Patent Nos. 4,642,120; 5,0-53,050;
and to 5,736,372, ) In a preferred embodiment, autologous or homologous bone marrow is obtained by aspiration with a bone biopsy needle from the iliac crest or femoral canal.
, (IJ.S.
Patent No. 5,053,050) The aspirated cells are injected into a phosphate buffered saline (PBS) containing 0.25% trypsin and injected sequentially through 17, 18 and 20 gauge needles to achieve a single cell suspension. The cells are plated in a density of 50-100 x 106 cells on 100 mm tissue culture dishes fed with BGJ b medium (GTBCO) with 15% F.C.S. (Fetal Calf Serum). The medium is changed daily or as required by the proliferation rate of the cells. The medium is supplemented with between about 0.1 nglml and 10 mg/ml active agent. The cells are subcultured weekly and after 5-2o subculturings an almost pure fibroblastic stromal cell population is achieved. This cell population is then trypsinized and put in a suspension culture at a density of 3-8 x 106 cellslml of medium and cultured above soft agar in a F-12 medium with 10%
F.C.S.
and SO ~g/ml sodium ascorbate added daily to the medium. The fibroblastic stromal cells start to aggregate immediately and after three-seven days most of the cells are in aggregates of 30-60 cells. All the aggregates express a chondrogenic phenotype, as determined by employing histochemical and immunohistochemical probes for analysis.
Although bone marrow derived chondrocytes are preferred, chondrocytes of autologous or homologous origin, or homologous committed chondrocytes, or any other progenital cells of mesenchymal origin can be used. It can be seen that this initial formulation comprises purification, proliferation and manipulation of a population expressing a chondrogenic phenotype. More specifically, the proliferating cells are from the class comprising bone marrow stroma cells, embryonal committed chondrocytes and any undifferentiated mesenchymal cells.
to In a preferred embodiment, the proliferadve effect of the active agents of the invention on cartilage cells is assessed by reactivity to an antibody directed against a protein lazown to be present in higher concentrations in proliferating cells than in non-proliferating cells, including but not limited to proliferating cell nuclear antigen (PCNA, or cyclin; Zymed Laboratories, South San Francisco, California). Viable cells 1s may also be identified using a technique such as the trypan blue exclusion assay. Cells to be reinfused into the subject are rinsed to remove all traces of culture fluid, resuspended in an appropriate, medium and then pelleted and rinsed several times.
After the final rinse, the cells are resuspended at between 0.7 x 106 and 50 x 106 cells per ml in an appropriate medium and administered as described below.
20 Alternatively, the effects of the active agents on matrix component synthesis in bone and cartilage cells are determined in organ culture and in isolated cells by measuring the levels of two matrix proteins, as described in U.S. Patent No.
5,686,116.
Type I collagen is the major matrix protein of bone and cartilage. Collagen is almost entirely composed'of proline, OH-WO 00/02905 PCT/US99/15735 _ proline, alanine and glycine. Thus, new-bone collagen synthesis can be determined by measuring the uptake of 3H-Pro or by following the appearance of 3H-OH-Pro, which its formed by conversion of proline subsequent to its incorporation into collagen.
Osteocalcin is a bone-specific matrix protein which is thought to be a cell signal for attracting osteoclasts to bone to initiate bone breakdown, and is also thought to slow or impede formation of newly mineralizing bone. Price et al., Proc. Natl. Acad.
Sci. USA, 79:7734-7738 (1982). Therefore, decreased osteocalcin synthesis is associated with bone repair.
Any synoviocyte (cartilage-forming) cells can be used. In a preferred t o embodiment, the cell line HIG-82, a permanent cartilage cell line which retains many features present in normal synoviocytes, is used. Georgescu et al., In Vitro Cell. Dev.
Biol., 24:1015-1022 (1988).
The synoviocytes are exposed to active agent for 24-48 hours, after which total cellular RNA is isolated by standard means. {Molecular Cloning: A Laboratory t5 Manual (Sambrook, et al., 1989, Cold Spring Harbor Laboratory Press)) Detection of mRNA for type I collagen can be performed via standard techniques in the art including but not limited to reverse transcription-polymerase chain reaction (RT-PCR) using primers specific to type I collagen or osteocalcin, or Northern blotting of the RNA
followed by hybridization with probes for type I collagen or osteocalcin.
(Molecular 2o Cloning: A Laboratory Manual (Sambrook, et al., 1989, Cold Spring Harbor Laboratory Press).
Alternatively, the expression of type I collagen synthesis is determined in organ culture as described in U.S. Patent No. 5,686,116. In a preferred embodiment, calvarial (skull) bones from newborn rats are placed in sterile culture dishes with a nutritive WO 00/02905 PCT/US99/15735 _ media to maintain viability. In this state, structural tissues grow by forming new matrix components (notably bone-specific collagen), but this growth is very slow.
Kream et al., Endocrinol., 116:296-302 (1985). Hemicalvaria from 21 day old fetal rats are incubated for 48 hours in the presence and absence of angiotensinogen, AI, AI
analogues, AI fragments and analogues thereof, AII, All analogues, All fragments and analogues thereof and/or All ATz type 2 receptor agonist at various concentrations. 3H-Pro is added for the final 18 hours of the incubation. Results showing that All increases the levels of proline (Pro) and hydroxyproline {OH-Pro) in fetal rat hemicalvaria demonstrate that All can increase the level of type I collagen synthesis in 1 o a normal bone culture system.
In another embodiment, the present invention comprises a method to enhance bone or cartilage repair in vivo by administration of the active agents of the invention.
In one embodiment, the compounds of the invention are injected into the subcutaneous tissue over the right calvarium of mice. Control vehicle is PBS supplemented with 1%
BSA. Heparin is administered at a dose of 50 units/ml. The animals are sacrificed on day 14 and bone growth is measured by histomorphometry.
Bone samples for quantitation are cleaned from adjacent tissues and fixed in
10% buffered formalin for 24-48 hours, decalcified in 14% EDTA for 1-3 weeks, processed through graded alcohols and embedded in paraffin wax. Three micron 2o sections of the calvaria and femurs are prepared. Representative sections are selected for histomorphometric assessment of the effects of the active agent on bone formation and bone resorption. Sections are measured by using a camera attachment to directly trace the microscopic image onto a digitizing plate. Bone changes are measured on sections collected 200 rnu m apart, over 4 adjacent 1 x 1 mm fields on both the injected WO 00/02905 PCT/US99/15735 _ and noninjected sides of the calvaria. New bone is identified by its woven, rather than lamellar structure, and osteoclasts and osteoblasts are identified by their distinctive morphology. Histomorphometry software (Osteomeasure, Osteometrix, Inc., Atlanta) is used to process digitizer input to determine cell counts and feature areas or perimeters.
Alternatively, the active agents of the invention can be used to potentiate osteoblast function in intact animals. In a preferred embodiment, a model for abnormal osteoblast activity, weanling Sprague-Dawley rats, are placed on a phosphate and vitamin D-deficient diet as per the manufacturer's instructions (the diet, #80039, Teklad, Madison, Wis.) The animals on the diet are also kept in the dark to prevent de novo vitamin D synthesis. Animals placed under such conditions show abnormal bone formation and a marked deficiency in total bone mass.
One group of the weanling rats on the diet is treated with active agent at between about 0.1 ng/kg and 10 mg/kg, given as a subcutaneous injection, every other day for 21 days. One group on the diet remains untreated and served as the control.
Littermate controls not on the diet and not treated with active agent supply blood samples at the time of sacrifice of the animals on the diet for determination of alkaline phosphatase activity. Upon sacrifice, the long bones are removed from the animals on the diet for subsequent analyses.
Serum alkaline phosphatase activity is used as a reliable indicator of osteoblast 2o activity, such that increased levels of alkaline phosphatase activity are evidence of the abnormal bone turnover in the experimental animals. Serum alkaline phosphatase activity is determined by measuring the hydrolysis of p-nitrophenyl phosphate by serum samples according to the method of Lowry et al., J. Biol. Chem., 207:19-(1954). Markedly elevated serum alkaline phosphatase activity indicates abnormal osteoblast activity. By contrast, normalization of bone cell function is evidenced by a decreased level of senun alkaline phosphate activity and increased bone mass (tested via determination of the ash weight of bones) relative to control animals.
Alternatively, a full thickness articular cartilage defect model in the femoral-patellar joint of adult rabbits is used to evaluate the ability of the compounds of the present invention to affect cartilage and bone repair, as described in U.S.
Patent No.
5,700,774. Adult New Zealand White rabbits are anesthetized and prepared for sterile surgery. A 3 x 3 mm defect through articular cartilage and into underlying subchondral bone is drilled into the patellar to groove of the knee joint. The defect is either left empty, filled with collagen sponge (controls), or with collagen sponge soaked with between about 0.1 ng/ml and 10 mg/ml active agent. The incision is closed and animals are allowed free movement within their cages for 4 weeks. After 4 weeks the animals are humanely euthanatized and the articular cartilage/subchondral bone defect is evaluated histologically for tissue t 5 architecture, quantity and quality of repair tissue. Northern analysis is performed for additional phenotyping.
In a further embodiment, dental and orthopedic implants can be coated with the compounds of the invention. In general, implant devices are coated with the active agents of the invention dissolved at a concentration in the range of 0.1 ng/ml to 10 2o mg/ml in phosphate-buffered saline (PBS) containing 2 mg/ml serum albumin.
The porous end of an implant is dipped in the solution and is air dried (or lyophilized) or implanted immediately into the bony site. The viscosity of the coating solution is increased, if desired, by adding hyaluronate at a final concentration of 0.1 mg/ml to 100 mg/ml or by adding other pharmaceutically acceptable excipie~nts.
Alternatively, the WO 00/02905 PCT/US99/15735 _ solution containing the active agent is mixed with collagen gel or human collagen (e.g.
Zyderm Registered TM Collagen Implant, Collagen Corp., Palo Alto, Calif.) to a final collagen concentration of 2 mg/ml to 100 mg/ml to form a paste or gel, which is then used to coat the porous end of the implant device. The coated implant device is placed into the bony site immediately or is air dried and rehydrated with PBS prior to implanting, with the objective of maximizing new bone formation into the implant while minimizing the ingrowth of soft tissue into the implant site.
In a further aspect, the present invention provides kits for enhancing bone or cartilage repair, wherein the kits comprise an effective amount of active agent for bone 1o or cartilage repair, and instructions for using the amount effective of active agent as a therapeutic. In a preferred embodiment, the kit further comprises a pharmaceutically acceptable carrier, such as those adjuvants described above. In another preferred embodiment, the kit further comprises a means for delivery of the active agent to a patient. Such devices include, but are not limited to syringes, matrical or micellar solutions, bandages, wound dressings, polymeric scaffolds, collagen vehicles, aerosol sprays, lipid foams, transdermal patches, topical administrative agents, polyethylene glycol polymers, carboxymethyl cellulose preparations, crystalloid preparations (e.g., saline, Ringer's lactate solution, phosphate-buffered saline, etc.), viscoelastics, polyethylene glycols, and polypropylene glycols. The means for delivery may either 2o contain the effective amount of the active agent, or rnay be separate from the compounds, which are then applied to the means for delivery at the time of use.
The kits may further comprise an amount effective for bone and/or cartilage repair, implantation, and augmentation of additional compounds, including but not limited to bone morphogenic protein-2, bone morphogenic protein-4, bone WO 00/02905 PCT/US99/15735 _ morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone. The kit may optionally contain a pharmaceutically acceptable carrier.
In another aspect of the present invention, an improved cell culture medium is provided for the proliferation of chondrocytes, wherein the improvement comprises addition to the cell culture medium of an effective amount of active agent to stimulate chondrocyte proliferation. Any cell culture media that can support the growth of chondrocytes can be used with the present invention. Such cell culture media include, but are not limited to Basal Media Eagle, Dulbecco's Modified Eagle Medium, Iscove's 1o Modified Dulbecco's Medium, McCoy's Medium, Minimum Essential Medium, F-10 Nutrient Mixtures, Opti-MEM~ Reduced-Serum Medium, RPMI Medium, and Macrophage-SFM Medium or combinations thereof.
The improved cell culture medium can be supplied in either a concentrated (ie:
10X) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophilizate. The cell culture may be either chemically defined, or may contain a serum supplement. Culture media is commercially available from many sources, such as GIBCO BRL (Gaithersburg, MD) and Sigma (St. Louis, MO).
In another embodiment, the methods of the present invention can be used to treat chondrocytic cell lines, such as articular chondrocytes, in order to maintain 2o chondrocytic phenotype and survival of the cells. The treated cell populations are therefore also useful for gene therapy applications.
In another preferred embodiment, the kit further comprises a sterile container.
The sterile container can comprise either a sealed container, such as a cell culture flask, WO 00/02905 PCT/US99/15735 _ a roller bottle, or a centrifuge tube, or a non-sealed container, such as a cell culture plate or rnicrotiter plate (Nunc; Naperville, IL).
In a further preferred embodiment, the kit further comprises an antibiotic supplement for inclusion in the reconstituted cell growth medium. Examples of appropriate antibiotic supplements include, but are not limited to actimonycin D, Fungizone~, kanamycin, neomycin, nystatin, penicillin, streptomycin, or combinations thereof (GIBCO).
A further obj ect of the present invention is to provide pharmaceutical compositions comprising the active agents as an ingredient for use in the methods of to the invention. The compositions comprise the active agents together with a compound or compounds that also enhance bone or cartilage implantation, repair and regeneration, including, but not limited to bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone, together with a pharmaceutically acceptable carrier, this term including any carrier which does not interfere with the effectiveness of the biological activity of the active agents and other compounds, and which is not toxic to the host to which it is administered.
Dosage and administration of the pharmaceutical compositions will vary depending on the disease being treated, based on a variety of factors, including the type of injury, the age, 2o weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed, as above. Thus, the dosage regimen may vary widely, but can be determined routinely by a physician using standard methods.

WO 00/02905 PCT/US99/15735 _ The present invention fulfills the need for methods to enhance bone and cartilage repair in a mammal suffering from a wide variety of disorders and injuries including, but not limited to bone fractures, defects, and disorders which result in weakened bones such as osteoporosis, osteoarthritis, Paget's disease, osteohalisteresis, osteomalacia, periodontal disease; bone loss resulting from multiple myeloma and other forms of cancer; bone loss resulting from side effects of other medical treatment (such as steroids); age-related loss of bone mass; articular cartilage tears, deformities and other cartilage defects such as arthritis and cartilaginous tissue damage.
1 o Example 1. Metlzod for Culture of Rabbit Chondrocytes Chondrocytes were isolated from the cartilage of the knee joints of adult rabbits and cultured according to the method of Okazaki et al. (Ann. Rheum. Dis.
55:181-186, 1996). Briefly, cartilage explants were minced into small pieces (approximately 1.5 mm by 1.5 mm). The tissue pieces were digested at 37°C sequentially in 0.05%
hyaluronidase (415 U/mg protein) for 10 minutes, 0.2% Type III trypsin (10,000 U/mg protein, Sigma) and 0.53 mM EDTA for 15 minutes, and 0.2% Type I collagenase {136 U/mg protein, Sigma) for 30 minutes. The samples were then washed and incubated overnight at 37°C in 5% COZ in air in Dulbecco's modified Eagle's medium enriched with 10% fetal calf serum, 3.5 mg/ml glucose, 0.2% Type I collagenase and antibiotics (100 U/ml penicillin and 100 p.g/ml streptomycin).
After this incubation, the cells were harvested and washed once with phosphate buffered saline (pH 7.2). The cells were then counted with a hemacytometer and resuspended at 1 x 105 celis/ml in Ham's FI2 supplemented with 10% fetal calf serum and antibiotics. A 10 ml aliquot of cells was transferred to collagen coated 25 cm2 WO 00/02905 PCT/US99/I5735 _ flasks (coated with collagen isolated from rat tail tendons) and incubated at 37°C in 5%
COZ in air. Five to seven days after the initiation of culture, the cells were detached with 0.05% trypsin-EDTA (Gibco-BRL) at 37°C in 5% COz in air. After detachment, the cells were washed once with phosphate buffered saline, centrifuged and resuspended at 200 cells/ml in Ham's F 12 supplemented with 10% fetal calf serum and antibiotics. Two hundred microliters of cells were then aliquoted into the collagen coated wells of a 96 well microtiter plate and allowed to adhere, after which, 10 pg/ml of the All and All analogue and fragment peptides listed in Table 3 were added to the wells to assess their effects on chondrocyte proliferation on days 1, 2, and 3 after 1 o culture initiation. Cell numbers were quantitated by staining the cells with Giemsa stain and counting the number of nuclei detected via microscopy. The data (FIG. 1-4) demonstrate that All and the All analogues and fragments all accelerated the chondrocyte proliferation.
Table 3. Designation for Analogues/Fragments Used in Example 1 Name Abbreviation Sequence SEQ ID NO:

GSD 37B Orn2-All D(Orn)VY>HPF SEQ ID N0:38 GSD 28 Ilea-All DRVYIHPI SEQ ID N0:39 GSD 24B Pro3-All DRPYIHPF SEQ ID N0:31 GSD 22A Ala4-AIII RVYAHPF SEQ ID N0:18 GSD36 Gly~-All GRVYIHPF SEQ ID N0:42 GSD38B Citron2-All D(Citron)VYIHPFSEQ ID N0:43 1GD Ala4-AII(1-7) DRVAIHP SEQ ID NO:40 2GD Pro3-AII(1-7) DRPYIHP SEQ ID N0:41 3GD Pro3Ala4-AII(1-7)DRPAIHP SEQ ID N0:44 AIII AII(2-8) RVYIHPF SEQ ID N0:2 AII(1-7) DRVYIHP SEQ ID N0:4 All DRVYIHPF SEQ ID NO.

Example 2. Bone.Healing Female, Sprague Dawley rats underwent intramuscular anesthesia with ketamine/rompum and were prepared for sterile surgery by shaving the surgical site and scrubbing with Betadine scrub followed by 70% ethanol. The rat was then placed on a sterile field in a lateral decubitis position facing the surgeon. The shaved legs were then covered with Betadine solution and draped aseptically. A skin incision was performed parallel to the long.axis of the right medial.diaphysis. The muscle was separated along fascial planes to expose the tibia. A defect of 1.4 mm in diameter was 1 o then drilled from the lateral side of the midshaft cortex so that the defect extended from one cortical side to the other, through the bone marrow. Sterile saline (0.9%
NaCI) for injection was then used to clean the surgical area of tissue debris and bone fragments.
Either hydron polymer solution (vehicle: 10% Hydron';~'60% ethanol, 1%
polyethylene glycol polymer) or peptide (AII, AII(1-7), or 9GD at 1 mg/ml} in hydron polymer solution was placed in the bone defect to fill the defect with polymer (approximately 0.1 ml of of er . The incision was closed with 3-0 Vicryl uture using continuous p Ym }
mattress suture. The animals were allowed to recover from anesthesia, given BuproneX
for analgesia and allowed free movement, until euthanasia 7 days later.
By gross observation, the defects ,that received the peptides of the invention 2o were more completely filled with new tissue that had begun to calcify. The majority of control defects were less than 50% filled with new tissue and no hardening of the tissue was observed. These data demonstrate that the peptides of the invention accelerate the formation of new bone tissue.

After gross evaluation, the muscle tissue was removed from the bone and the bones were placed in formalin for fixation. After 2 days in 10% buffered formaldehyde, the tissues were placed in a decalcifying solution (Rapid Bone Decal; M
American MasterTech Scientific, Inc. Lodi, CA) diluted 3:1 for 6 hours. After decalcification, the bone was cut in half along the long axis, embedded in paraffin, sectioned and stained with hematoxylin and eosin.
Microscopic evaluation of the tissue sections confirmed the gross observations.
TM
On day 7, the bones in which the defect had been filled with Hydron (the placebo) had a loose fibrin filler with the majority of cells observed being inflammatory in nature.
to Occasionally, a cell that appeared to be of mesenchymal origin or a blood vessels was seen at the site of injury. In all of the peptide treated animals, extensive stromal cell ingrowth with numerous blood vessels was observed. In approximately 50% of the peptide treated animals, tissue with the appearance of new bone was observed.
These data clearly support the ability of these angiotensin peptides to accelerate new bone 1 s formation.
Table 4. Designation for Analogues/Fragments used in Example 2 AII(1-7) DRVYIF-IP SEQ ID N0:4 2o All DRVYIFIPF SEQ ID NO. 1 9GD: NorLeu3-AII(1-7) DR(nor)YIH)? SEQ ID N0:45 It is to be understood that the invention is not to be limited to the exact details of operation, or to the exact compounds, compositions, methods, procedures or z5 embodiments shown and described, as obvious modifications and equivalents will be WO 00/02905 PCTNS99/15735 _ apparent to one skilled in the art, and the invention is therefore to be limited only by the full scope of the appended claims.

WO 00/02905 PCT/US99/15735 _ SEQUENCE LISTING
<110> Rodgers, Kathleen diZerega, Gere <120> Methods for Accelerating Hone and Connective Tissue Growth and Repair <130> 98365C
<140> To be assigned <141> T999-07-11 <160> 45 <170> PatentIn Ver. 2.0 <210> 1 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII
<400> 1 Asp Arg Val Tyr Ile His Pro Phe <210> 2 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (2-B) <400> 2 Arg Val Tyr Ile His Pro Phe <210> 3 <211> 6 <212> PRT
<213> Artificial Sequence SUBSTITUTE SHEET (RULE 26) <223> Description of Artificial Sequence:AiI (3-8) <400> 3 Val Tyr Ile His Pro Phe <220> 4 <211> 7 <212> PRT
<213> PiYtificial Sequ~.z;!:e <220>
c223> Description of Artificial Sequence:AII (1-7) <400> 4 Asp Arg Val Tyr Ile His Pro <210> 5 <211> 6 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (2-7) <400> 5 Arg Val Tyr Ile His Pro <210> 6 <211> S
<212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (3-7) <400> 6 Val Tyr Ile His Pro <210> 7 SUBSTITUTE SHEET (RULE 26) <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (5-8) <400> 7 Ile His Pro Phe <210> 8 <211> 6 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (1-5) <400> 8 Asp Arg Val Tyr Ile His <210> 9 <211> 5 ~.212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (.-5) <400> 9 Asp Arg Val Tyr Ile <210> 10 <211> 4 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII il-:~) <400> 10 Asp Arg gal Tyr SUBSTITUTE SHEET (RULE 26) WO 00/02905 PCTNS99/15735 _ <210> 11 <211> 3 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (1-3) 2400> 1Z
Asp Arg Val <210> 12 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <220>
<221> MOD_RES
<222> (2) <223> Nle <400> 12 Arg Xaa Tyr Ile His Pro Phe <210> 13 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <220>
<221> MOD_RES
<222> (4) <223> Nle <400> 13 SUBSTTTUTE SHEET (RULE 2b) Arg ~;a? Tyr Xaa a=s Pro Phe <210> 1.1 <211> 3 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (6-8) <400> 14 His Pro Phe <210~ 15 <211> 5 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (4-8) <400> 15 Tyr Ile His Pro Phe <210> 16 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue class <220>
<221> UNSURE
<222> (1) <223> Xaa at poistion 1 can be Arg, Lys, Ala, orn, Ser, MeGly, D-Arg, or D-Lys <220>
<221> UNSURE
<222> 12) S
SUBSTITUTE SHEET (RULE 26) <223> ~aa at position 2 can be Val, :,la, ~eu, Nie, _Tie, Gly, Pro, Hib, Acp, or Tyr <220>
<221> UNSURE
<222> 141 <223> Xaa at position 4 can be Ile, Ala, Leu, Nle, Val, or Gly <400> 16 Xaa Xaa Tyr Xaa His Pro Phe <210> 17 <211> 7 <212> PRT
<213> Artificial.Sequence <220>
<223> Description of Artificial Sequence:AII analogue <400> 17 Arg Val Tyr Gly His Pro Phe <210> 18 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <400> 18 Arg Val Tyr Ala His Pro Phe <210> 19 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 1 SUBSTITUTE SHEET (RULE 26) WO 00/02905 PCT/US99/15735 _ Asp Arg ':'al Tyr Val His Prc Phe <210> 2:
<211> 8 <212> PPT
<213> Artificial Sequence <220>
'<223>-Description of Artificial Sequence:AII analogue 2 <400> 20 Asn Arg val Tyr Val His Pro Phe <210> 2i <211> ii <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 3 <400> 2I
Ala Pro Gly Asp Arg Ile T.yr Val His Pro Phe <210> 2~
<211> 8 <212> PPT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 4 <400> 22 Glu Arg :'al Tyr Ile His Pro Phe <210> 23 <211> 9 <212> PR:
<213> Artificial Sequence SUBSTITUTE SHEET (RULE 26) <220>
<223> Description of Artificial Sequence:AII a..~.alogue 5 <400> 23 Asp Lys Val Tyr Ile His Pro Phe <210> 24 <211> 8 '<212> 'PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 6 <400> 24 Asp Arg Ala Tyr Ile His Pro Phe <210> 25 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 7 <400> 25 Asp Arg Val Thr Ile His Pro Phe <210> 26 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial 5equence:AII analogue 8 <400> 26 Asp Arg Val Tyr Leu His Pro Phe SUBSTITUTE SKEET (RULE 26) <211> a <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 5 <400> 27 Asp Arg val Tyr Ile Arg Pro Phe <210> 28 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 10 <400> 28 Asp Arg Val Tyr Ile His Ala Phe <210> 2~
<211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 11 <400> 29 Asp Arg Val Tyr Ile His Pro Tyr <210> 30 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 12 <400> 30 SUBSTITUTE SHEET (RULE 26) Pro Arg Val Tyr Ile His Pro Phe <210> 31 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Ar:ificial Sequence:AII analogue 13 c400> 31 Asp Arg Pro Tyr Ile His Pro Phe <210> 32 <211> 8 c2I2 > PRT
<213> Artificial Sequence c220>
<223> Description of Artificial Sequence:AII analogue 14 c220>
<221> MOD RES
<222> (4)~
c223> PHOSPHORYLATION
c400> 32 Asp Arg val Tyr Ile His Pro Phe <210> 33 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 15 <220>
<221> MOD_RES
<222> (3) <223> Nle SUBSTITUTE SHEET (RULE 26) WO 00/02905 PCT/US99/15735 _ Asp Arg Xaa Tyr Ile His Pry Phe <210> 34 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<'223> Description of Artificial Sequence:AII analogue 16 <220>
<221> MOD_RES
<222> (5) <223> Nle <400> 34 Asp Arg Val Tyr Xaa His Pro Phe <210> 35 <211> 9 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 17 <220>
<221> MOD_RES
<222> (4) <223> homo Ser <400> 35 Asp Arg Val Ser Tyr Ile His Pro Phe <210> 36 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial
11 SUBSTITUTE SHEET (RULE 26) Sequence:p-aminophenylalanine 6 All <220>
<221> MOD RES
<222> (6) <223> p-aminophenylalanine <400> 36 Asp Arg Val Tyr Ile Xaa Pro Phe <210> 37 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:angiotensin I
<400> 37 Asp Arg Val Tyr Ile His Pro Phe His Leu <210> 38 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<221> MOD_RES
<222> (2) <223> Orn <220>
<223> Description of Artificial Sequence:GSD37H:
Orn2-All <900> 38 Asp Xaa Val Tyr Ile His Pro Phe <210> 39 <211> 8 <212> PRT
<213> Artificial Sequence
12 SUBSTITUTE SliEET (RULE 26) WO 00/02905 PCT/US99/15735 _ <220>
<223> Description of Artificial Sequence:GSD28: Ilea-All <400> 39 Asp Arg Val Tyr Ile His Pro Ile <210> 40 <211> 7 2212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:lGD:
Ala4-AII(1-7) <400> 40 Asp Arg Val Ala Ile His Pro <210> 41 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:2GD:
Pro3-AII(1-7) <400> 41 Asp Arg Pro Tyr Ile His Pro <210> 42 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Glyl-All <400> 42 Gly Arg Val Tyr Ile His Pro Phe
13 SUBSTITUTE SHEET (RULE 26) WO 00/02905 PCT/US99/15735 _ <210> 43 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:GSD38B:Citron2-All <220>
<221> MOD_RES
<222> (2) <223> Citron <400> 43 Asp Xaa Val Tyr IIe His Pro Phe <210> 44 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Pro3Ala4-AII(1-7) <400> 44 Asp Arg Pro Ala Ile His Pro <210> 45 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:9GD:
norleu3-AII(1-7) <220~
<221> MOD RES
<222> (3) <223> Nle
14 SUBSTTTUTE SHEET (RULE 26) WO 00/02905 PCT/US99/15735 _ <400> 45 Asp Arg Xaa Ty r Ile His Pro SUBSTTTUTE SHEET {RULE Z6)

Claims (52)

THE EMBODIMENTS OR THE INVENTION IN WHICH AN EXCLUSIVE
PROPERTY OR PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. A pharmaceutical composition comprising:
i) at least one active agent which comprises a sequence of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I

wherein R1 is selected from the group of H, Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc;
R2 is selected from the group of Arg, Lys, Ala, Orn, Citron, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group of Tyr, Tyr(PO3)2, Thr, Ala, Ser, homoSer and azaTyr;
R5 is selected from the group of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is selected from the group of His, Arg and 6-NH2-Phe;
R7 is selected from the group of Pro and Ala; and R8 is selected from the group of H, Phe, Phe(Br), Ile and Tyr;
with the proviso that when present as the N-terminal amino acid, R4 is not Tyr; and wherein said active agent is not angiotensin II [SEQ ID NO: 1] or His-Pro-Phe [SEQ ID NO:14];
ii) an effective amount of at least one compound selected from the group of hone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone; and iii) a pharmaceutically acceptable carrier;

wherein said pharmaceutical composition is suitable for enhancing bone repair, enhancing bone and prosthesis implantation, enhancing cartilage repair, or enhancing attachment and fixation of cartilage implants to bone or other tissue in a mammal.
2. A pharmaceutical composition comprising:
i) at least one active agent comprising an amino acid sequence selected from the group of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID
NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID
NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID
NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34;
SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID
NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, and SEQ ID NO:45;
ii) an effective amount of at least one compound selected from the group of bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone; and iii) a pharmaceutically acceptable carrier;
wherein said pharmaceutical composition is suitable for enhancing bone repair, enhancing bone and prosthesis implantation, enhancing cartilage repair, or enhancing attachment and fixation of cartilage implants to bone or other tissue in a mammal.
3. Use of the pharmaceutical composition according to Claim 1 or 2 for enhancing bone repair in a mammal.
4. The use according to Claim 3, wherein said bone repair is required as a result of osteoporosis, osteoarthritis, Paget's disease, osteohalisteresis, osteomalacia, periodontal disease, bone loss resulting from cancer, bone loss resulting from steroid treatment, age-related loss of bone mass or bone fracture.
5. Use of the pharmaceutical composition according to Claim 1 or 2 for enhancing bone and prosthesis implantation in a mammal.
6. Use of the pharmaceutical composition according to Claim 1 or 2 for enhancing cartilage repair in a mammal.
7. The use according to Claim 6, wherein said cartilage repair is required as a result of torn cartilage or arthritis.
8. Use of the pharmaceutical composition according to Claim 1 or 2 for enhancing attachment and fixation of cartilage implants to bone or other tissue in a mammal.
9. The use according to any one of Claims 3 - 8, wherein the dosage of said active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
10. Use of a pharmaceutical composition comprising:
i) at least one active agent which comprises a sequence of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I

wherein R1 is selected from the group of H, Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc;

R2 is selected from the group of Arg, Lys, Ala, Orn, Citron, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group of Tyr, Tyr(PO3)2, Thr, Ala, Ser, homoSer and azaTyr;
R5 is selected from the group of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is selected from the group of His, Arg and 6-NH2-Phe;
R7 is selected from the group of Pro and Ala; and R8 is selected from the group of H, Phe, Phe(Br), Ile and Tyr;
with the proviso that, when present as the N-terminal amino acid, R4 is not Tyr; and wherein said active agent is not angiotensin II [SEQ ID NO: 1] or His-Pro-Phe [SEQ ID NO:14]; and ii) a pharmaceutically acceptable carrier;
for enhancing bone repair, enhancing bone and prosthesis implantation, enhancing cartilage repair, or enhancing attachment and fixation of cartilage implants to bone or other tissue, in a mammal in need of such therapy.
11. The use according to Claim 10, wherein said pharmaceutical composition further comprises an effective amount of at least one compound selected from the group of bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone.
12. The use of at least one active agent comprising a sequence of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I

wherein R1 is selected from the group of H, Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc, R2 is selected from the group of Arg, Lys, Ala, Orn, Citron, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group of Tyr, Tyr(PO3)2, Thr, Ala, Ser, homoSer and azaTyr;
R5 is selected from the group of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is selected from the group of His, Arg and 6-NH2-Phe;
R7 is selected from the group of Pro and Ala; and R8 is selected from the group of H, Phe, Phe(Br), Ile and Tyr;
with the proviso that when present as an N-terminal amino acid, R4 is not Tyr; and wherein the active agent is not angiotensin II [SEQ ID NO: 1] or His-Pro-Phe [SEQ ID NO:14], for the manufacture of a medicament for enhancing bone repair, enhancing bone and prosthesis implantation, enhancing cartilage repair, or enhancing attachment and fixation of cartilage implants to bone or other tissue, in a mammal.
13. The use according to any one of Claims 10 - 12, wherein said active agent comprises at least 4 contiguous amino acids of general formula I.
14. The use according to any one of Claims 10 - 12, wherein said active agent comprises at least 5 contiguous amino acids of general formula I.
15. The use according to any one of Claims 10 - 12, wherein said active agent comprises at least 6 contiguous amino acids of general formula I.
16. The use according to any one of Claims 10 - 12, wherein said active agent comprises at least 7 contiguous amino acids of general formula I.
17. The use according to any one of Claims 10 - 12, wherein said active agent consists of 3 contiguous amino acids of general formula I.
18. The use according to any one of Claims 10 - 12, wherein said active agent consists of 4 contiguous amino acids of general formula I.
19. The use according to any one of Claims 10 - 12, wherein said active agent consists of 5 contiguous amino acids of general formula I.
20. The use according to any one of Claims 10 - 12, wherein said active agent consists of 6 contiguous amino acids of general formula I.
21. The use according to any one of Claims 10 - 20, wherein said active agent comprises an amino acid sequence selected from the group of angiotensinogen, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ
ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID
NO:12, SEQ ID NO:13, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ
ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID
NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ
ID NO:33, SEQ ID NO:34; SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID
NO:42, SEQ ID NO:43, SEQ ID NO:44, and SEQ ID NO:45.
22. The use according to any one of Claims 10 - 20, wherein said active agent consists of an amino acid sequence selected from the group of angiotensinogen, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ
ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID
NO:12, SEQ ID NO:13, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ
ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID
NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ
ID NO:33, SEQ ID NO:34; SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID
NO:42, SEQ ID NO:43, SEQ ID NO:44, and SEQ ID NO:45.
23. The use of said pharmaceutical composition of Claim 10 or Claim 11 or said medicament according to any one of Claims 12 - 22, for enhancing bone repair in a mammal.
24. The use according to Claim 23, wherein said bone repair is required as a result of osteoporosis, osteoarthritis, Paget's disease, osteohalisteresis, osteomalacia, periodontal disease, bone loss resulting from cancer, bone loss resulting from steroid treatment, age-related loss of bone mass or bone fracture.
25. The use of said pharmaceutical composition of Claim 10 or Claim 11 or said medicament according to any one of Claims 12 - 22, for enhancing bone and prosthesis implantation in a mammal.
26. The use of said pharmaceutical composition of Claim 10 or Claim 11 or said medicament according to any one of Claims 12 - 22, for enhancing cartilage repair in a mammal.
27. The use according to Claim 26, wherein said cartilage repair is required as a result of torn cartilage or arthritis.
28. The use of said pharmaceutical composition of Claim 10 or Claim 11 or said medicament according to any one of Claims 12 - 22, for enhancing attachment and fixation of cartilage implants to bone or other tissue in a mammal.
29. Use of a pharmaceutical composition comprising:

i) at least one active agent of general formula II

Asp-Arg-R1-R2-Ile-His-Pro-R3 wherein R1 is selected from the group of Ile, Pro, Ala, Val, Leu, and norLeu;
R2 is selected from the group of Tyr and Tyr(PO3)2; and R3 is selected from the group of H and Phe;
and wherein said active agent is not angiotensin II [SEQ ID NO: 1]; and ii) a pharmaceutically acceptable carrier;
for enhancing bone repair, or bone and prosthesis implantation, in a mammal in need of such therapy.
30. The use according to Claim 29, wherein said pharmaceutical composition further comprises an effective amount of at least one compound selected from the group of bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone.
31. The use of at least one active agent of general formula II

Asp-Arg-R1-R2-Ile-His-Pro-R3 wherein R1 is selected from the group of Ile, Pro, Ala, Val, Leu, and norLeu;
R2 is selected from the group of Tyr and Tyr(PO3)2; and R3 is selected from the group of H and Phe;
and wherein the active agent is not angiotensin II [SEQ ID NO: 1];
for the manufacture of a medicament for enhancing bone repair, or bone and prosthesis implantation, in a mammal.
32. The use according to any one of Claims 29 - 31, wherein said active agent comprises an amino acid sequence selected from the group of SEQ ID NO:4, SEQ
ID NO:24, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:41, and SEQ ID NO:45.
33. The use according to any one of Claims 29 - 31, wherein said active agent consists of an amino acid sequence selected from the group of SEQ ID NO:4, SEQ ID NO:24, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO: 33, SEQ ID
NO:41, and SEQ ID NO:45.
34. Use of a pharmaceutical composition comprising:
i) at least one active agent of general formula III

R1-R2-R3-R4-R5-His-Pro-R6 wherein R1 is selected from the group of H, Gly, and Asp;
R2 is selected from the group of Arg, Citron, and Ornithine;
R3 is selected from the group of Val, Ile, Ala, Leu, norLeu, and Pro;
R4 is selected from the group of Tyr, Tyr(PO3)2, and Ala;
R5 is selected from the group of Ile, Ala, Val, Leu, and norLeu; and R6 is selected from the group of H, Phe and Ile;
and wherein the active agent is not angiotensin II [SEQ ID NO: 1]; and ii) a pharmaceutically acceptable carrier;
for enhancing cartilage repair, or attachment and fixation of cartilage implants to bone or other tissue, in a mammal in need of such therapy.
35. The use according to Claim 34, wherein said pharmaceutical composition further comprises an effective amount of at least one compound selected from the group of bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone.
36. The use of at least one active agent of general formula III

R1-R2-R3-R4-R5-His-Pro-R6 wherein R1 is selected from the group of H, Gly, and Asp;
R2 is selected from the group of Arg, Citron, and Ornithine;
R3 is selected from the group of Val, Ile, Ala, Leu, norLeu, and Pro;
R4 is selected from the group of Tyr, Tyr(PO3)2, and Ala;
R5 is selected from the group of Ile, Ala, Val, Leu, and norLeu; and R6 is selected from the group of H, Phe and Ile;
and wherein the active agent is not angiotensin II [SEQ ID NO: 1], for the manufacture of a medicament for enhancing cartilage repair, or attachment and fixation of cartilage implants to bone or other tissue, in a mammal.
37. The use according to any one of Claims 34 - 36, wherein said active agent comprises a sequence selected from the group of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:13, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:12, SEQ ID
NO:24, SEQ ID NO:26, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:38, SEQ
ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, and SEQ ID NO:45.
38. The use according to any one of Claims 34 - 36, wherein said active agent consists of an amino acid sequence selected from the group of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:13, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:12, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:32, SEQ ID NO:33, SEQ ID
NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ
ID NO:43, SEQ ID NO:44, and SEQ ID NO:45.
39. The use according to any one of Claims 12, 31 or 36, wherein said medicament further comprises an effective amount of at least one compound selected from the group of bone morphogenic protein-2, bone morphogenic protein-4, bone morphogenic protein-6, bone morphogenic protein-7, transforming growth factor-beta, insulin-like growth factor, and parathyroid hormone.
40. A method for enhanced culture of osteoblasts, chondrocytes, or human bone cells in vitro, comprising contacting said osteoblasts, chondrocytes, or human bone cells with an amount effective to accelerate proliferation of said osteoblasts, chondrocytes, or human bone cells of at least one active agent comprising a sequence of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I

wherein R1 is selected from the group of H, Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc, R2 is selected from the group of Arg, Lys, Ala, Orn, Citron, Ser(Ac), Sar, D-Arg and D-Lys:
R3 is selected from the group of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group of Tyr, Tyr(PO3)2, Thr, Ala, Ser, homoSer and azaTyr;
R5 is selected from the group of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is selected from the group of His, Arg and 6-NH2-Phe;
R7 is selected from the group of Pro and Ala; and R8 is selected from the group of H, Phe, Phe(Br), Ile and Tyr;

with the proviso that when present as an N-terminal amino acid, R4 is not Tyr;
and wherein the active agent is not angiotensin II [SEQ ID NO: 1] or His-Pro-Phe [SEQ ID NO:14].
41. The method according to Claim 40 wherein said active agent comprises at least 4 contiguous amino acids of general formula I.
42. The method according to Claim 40 wherein said active agent comprises at least 5 contiguous amino acids of general formula I.
43. The method according to Claim 40 wherein said active agent comprises at least 6 contiguous amino acids of general formula I.
44. The method according to Claim 40 wherein said active agent comprises at least 7 contiguous amino acids of general formula I.
45. The method according to Claim 40 wherein said active agent consists of 3 contiguous amino acids of general formula I.
46. The method according to Claim 40 wherein said active agent consists of 4 contiguous amino acids of general formula I.
47. The method according to Claim 40 wherein said active agent consists of 5 contiguous amino acids of general formula I.
48. The method according to Claim 40 wherein said active agent consists of 6 contiguous amino acids of general formula I.
49. The method according to Claim 40 wherein said active agent consists of 7 contiguous amino acids of general formula I.
50. A kit for enhancing bone repair, enhancing bone and prosthesis implantation, enhancing cartilage repair or enhancing attachment and fixation of cartilage implants to bone or other tissue, in a mammal comprising:
i) the pharmaceutical composition according to Claim 1 or 2; and ii) instructions for use.
51. The kit according to Claims 50 further comprising a means of delivery of said pharmaceutical composition.
52. A kit for enhanced culture of osteoblasts, chondrocytes, or human bone cells in vitro, comprising:
i) a cell culture medium comprising an amount effective to accelerate proliferation of said osteoblasts, chondrocytes, or human bone cells of at least one active agent comprising a sequence of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I

wherein R1 is selected from the group of H, Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc, R2 is selected from the group of Arg, Lys, Ala, Orn, Citron, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group of Tyr, Tyr(PO3)2, Thr, Ala, Ser, homoSer and azaTyr;
R5 is selected from the group of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is selected from the group of His, Arg and 6-NH2-Phe;
R7 is selected from the group of Pro and Ala; and R8 is selected from the group of H, Phe, Phe(Br), Ile and Tyr;
with the proviso that when present as an N-terminal amino acid, R4 is not Tyr; and wherein the active agent is not angiotensin II [SEQ ID NO:1] or His-Pro-Phe [SEQ ID NO:14]; and ii) instructions for use.
CA002328871A 1998-07-13 1999-07-12 Methods for accelerating bone and cartilage growth and repair Expired - Fee Related CA2328871C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US9265398P 1998-07-13 1998-07-13
US60/092,653 1998-07-13
US13085599P 1999-04-22 1999-04-22
US60/130,855 1999-04-22
PCT/US1999/015735 WO2000002905A2 (en) 1998-07-13 1999-07-12 Methods for accelerating bone and cartilage growth and repair

Publications (2)

Publication Number Publication Date
CA2328871A1 CA2328871A1 (en) 2000-01-20
CA2328871C true CA2328871C (en) 2002-10-01

Family

ID=26785912

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002328871A Expired - Fee Related CA2328871C (en) 1998-07-13 1999-07-12 Methods for accelerating bone and cartilage growth and repair

Country Status (9)

Country Link
US (1) US6258778B1 (en)
EP (1) EP1094829B8 (en)
JP (2) JP3686335B2 (en)
AT (1) ATE250423T1 (en)
AU (1) AU756785B2 (en)
CA (1) CA2328871C (en)
DE (1) DE69911609T2 (en)
ES (1) ES2207258T3 (en)
WO (1) WO2000002905A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106456698A (en) * 2014-05-19 2017-02-22 特维娜有限公司 Synthesis of beta-arrestin effectors

Families Citing this family (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US20040033950A1 (en) * 2000-09-26 2004-02-19 Hock Janet M. Method of increasing bone toughness and stiffness and reducing fractures
ATE255920T1 (en) * 1999-02-01 2003-12-15 Genetics Inst Llc METHODS AND COMPOSITIONS FOR HEALING AND REPAIRING ARTicular cartilage
US6730775B1 (en) 1999-03-23 2004-05-04 University Of Southern California Methods for limiting scar and adhesion formation
CA2685349C (en) 1999-11-15 2013-09-17 Bio Syntech Canada Inc. Temperature-controlled and ph-dependant self-gelling biopolymeric aqueous solution
US6747008B1 (en) 2000-06-19 2004-06-08 University Of Southern California Methods for treating and preventing alopecia
IL153490A0 (en) 2000-06-29 2003-07-06 Biosyntech Canada Inc Composition and method for the repair and regeneration of cartilage and other tissues
US20060094112A1 (en) * 2001-03-07 2006-05-04 Omotunde Babalola Biological scaffold
EP1395566B1 (en) 2001-05-31 2007-09-12 Vicore Pharma AB Tricyclic compounds useful as angiotensin ii agonists
WO2003061653A1 (en) * 2002-01-22 2003-07-31 Pfizer Limited 3-(imidazolyl)-2-aminopropanoic acids for use as tafi-a inhibitors for the treatment of thrombotic diseases
WO2003072059A2 (en) * 2002-02-27 2003-09-04 Wake Forest University Angiotensin-(1-7) and angiotensin-(1-7) agonists for inhibition of cancer cell growth
US7957401B2 (en) * 2002-07-05 2011-06-07 Geos Communications, Inc. System and method for using multiple communication protocols in memory limited processors
EP1534191A4 (en) * 2002-08-20 2010-11-03 Exactech Inc Composition for the carrying and delivery of bone growth inducing material and methods for producing and applying the composition
EP1613267A2 (en) * 2003-03-27 2006-01-11 Children's Hospital Medical Center Use of fgf- 18 protein, target proteins and their respective encoding nucleotide sequences to induce cartilage formation
US7067123B2 (en) 2003-04-29 2006-06-27 Musculoskeletal Transplant Foundation Glue for cartilage repair
US7488348B2 (en) 2003-05-16 2009-02-10 Musculoskeletal Transplant Foundation Cartilage allograft plug
US7901457B2 (en) 2003-05-16 2011-03-08 Musculoskeletal Transplant Foundation Cartilage allograft plug
CN100551452C (en) * 2003-05-21 2009-10-21 株式会社Jms Serum production is with container and use the method for regenerative medicine of this container
US7606217B2 (en) * 2003-07-02 2009-10-20 I2 Telecom International, Inc. System and method for routing telephone calls over a voice and data network
CN101072572B (en) * 2003-07-09 2013-12-11 华沙整形外科股份有限公司 Isolation of bone marrow fraction rich in connective tissue growth components and the use thereof to promote connective tissue formation
AU2003272814A1 (en) 2003-10-02 2005-05-19 Ibrahim Al-Habdan Peptide for promoting healing of fractures
WO2006020981A2 (en) * 2004-08-12 2006-02-23 University Of Southern California Methods for accelerating bone growth and repair
US7309589B2 (en) 2004-08-20 2007-12-18 Vironix Llc Sensitive detection of bacteria by improved nested polymerase chain reaction targeting the 16S ribosomal RNA gene and identification of bacterial species by amplicon sequencing
US7837740B2 (en) 2007-01-24 2010-11-23 Musculoskeletal Transplant Foundation Two piece cancellous construct for cartilage repair
US7815926B2 (en) 2005-07-11 2010-10-19 Musculoskeletal Transplant Foundation Implant for articular cartilage repair
EP1926459B1 (en) 2005-09-19 2015-01-07 Histogenics Corporation Cell-support matrix having narrowly defined uniformly vertically and non-randomly organized porosity and pore density and a method for preparation thereof
DE602007012693D1 (en) 2006-05-26 2011-04-07 Baxter Healthcare Sa INJECTABLE FIBRIN COMPOSITION FOR BONE GAIN
EP2446862B1 (en) 2006-06-20 2017-08-09 Ben Gurion University Of The Negev Research And Development Authority Amphiphilic peptides and hydrogel matrices thereof for bone repair
JP5579445B2 (en) * 2007-01-22 2014-08-27 オルソトロフィックス インコーポレイテッド Peptide compositions and methods for promoting cartilage formation
US8435551B2 (en) 2007-03-06 2013-05-07 Musculoskeletal Transplant Foundation Cancellous construct with support ring for repair of osteochondral defects
CN103785058B (en) * 2007-04-23 2016-12-07 巴克斯特国际公司 Comprise the fibrin composition of strontium compound
EP2217260B1 (en) 2007-12-04 2016-11-09 Ben-Gurion University Of The Negev Research And Development Authority Amphiphilic peptide matrices for treatment of osteoporosis
WO2009111069A1 (en) 2008-03-05 2009-09-11 Musculoskeletal Transplant Foundation Cancellous constructs, cartilage particles and combinations of cancellous constructs and cartilage particles
US20090227507A1 (en) * 2008-03-10 2009-09-10 University Of Southern California Angiotensin (1-7) Dosage Forms and Uses Thereof
ES2712891T3 (en) * 2008-11-20 2019-05-16 Cartiheal 2009 Ltd Solid forms for tissue repair
WO2010111617A2 (en) 2009-03-27 2010-09-30 Van Andel Research Institute Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
AT508569A1 (en) * 2009-07-23 2011-02-15 Affiris Ag PHARMACEUTICAL COMPOUND
WO2012048275A2 (en) 2010-10-08 2012-04-12 Caridianbct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
EP2455388A1 (en) 2010-11-23 2012-05-23 LanthioPep B.V. Novel angiotensin type 2 (AT2) receptor agonists and uses thereof.
WO2012103347A1 (en) * 2011-01-26 2012-08-02 Whitson Stanley William Hydrogel bioscaffold and coating for bone and tooth repair
EA026440B1 (en) 2011-02-02 2017-04-28 Университи Оф Сатерн Калифорния Methods for treating diabetic foot ulcers
ES2387057B1 (en) 2011-02-16 2013-07-19 Valeria Lucila Sainz Prestel USE OF A VEGETABLE EXTRACT AS AN ACTIVE PRINCIPLE FOR THE DEVELOPMENT OF A PRODUCT WITH PHARMACOLOGICAL ACTIVITY FOR THE TREATMENT OF TISSULAR INJURIES AND PROCEDURE OF OBTAINING THE EXTRACT
WO2013172956A1 (en) 2012-05-14 2013-11-21 University Of Southern California Methods for limiting development of a skin wound
US8557958B1 (en) 2012-06-18 2013-10-15 Tarix Pharmaceuticals Ltd. Compositions and methods for treatment of diabetes
CA2918486C (en) * 2012-07-20 2020-12-08 The Regents Of The University Of California Methods for producing cartilage and bone
US8633158B1 (en) 2012-10-02 2014-01-21 Tarix Pharmaceuticals Ltd. Angiotensin in treating brain conditions
WO2014074806A1 (en) 2012-11-08 2014-05-15 Smith & Nephew, Inc-- Improved reattachment of detached cartilage to subchondral bone
AU2013342255B2 (en) 2012-11-08 2017-05-04 Smith & Nephew, Inc. Methods and compositions suitable for improved reattachment of detached cartilage to subchondral bone
US9623084B2 (en) 2013-03-15 2017-04-18 University Of Southern California Methods for treating multiple sclerosis
EP2967049B1 (en) 2013-03-15 2020-09-02 University Of Southern California Methods, compounds, and compositions for the treatment of angiotensin-related diseases
CA2916701A1 (en) 2013-07-03 2015-01-08 The Arizona Board Of Regents On Behalf Of The University Of Arizona Method for treating cognitive dysfunction
US10633625B2 (en) 2013-11-16 2020-04-28 Terumo Bct, Inc. Expanding cells in a bioreactor
US9333233B2 (en) 2014-02-25 2016-05-10 Tarix Pharmaceuticals Ltd. Methods and compositions for the delayed treatment of stroke
EP3122866B1 (en) 2014-03-25 2019-11-20 Terumo BCT, Inc. Passive replacement of media
CA2955656C (en) 2014-07-21 2023-03-14 The Arizona Board Of Regents On Behalf Of The University Of Arizona Ang-(1-7) derivative oligopeptides and methods for using and producing the same
US10183055B2 (en) 2014-07-21 2019-01-22 Arizona Board Of Regents On Behalf Of The University Of Arizona Ang-(1-7) derivative oligopeptides for the treatment of pain and other indications
JP6830059B2 (en) 2014-09-26 2021-02-17 テルモ ビーシーティー、インコーポレーテッド Scheduled cell feeding
US10077420B2 (en) 2014-12-02 2018-09-18 Histogenics Corporation Cell and tissue culture container
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
EP3464565A4 (en) 2016-05-25 2020-01-01 Terumo BCT, Inc. Cell expansion
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
WO2018184028A2 (en) 2017-03-31 2018-10-04 Terumo Bct, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
JP2022542437A (en) 2019-08-02 2022-10-03 ランティオペプ ベスローテン ヴェンノーツハップ Angiotensin type 2 (AT2) receptor agonists for treating cancer

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL68218A (en) 1983-03-23 1985-12-31 Univ Ramot Compositions for cartilage repair comprising embryonal chondrocytes
IL83003A (en) 1986-07-01 1995-07-31 Genetics Inst Osteoinductive factors
US5013649A (en) 1986-07-01 1991-05-07 Genetics Institute, Inc. DNA sequences encoding osteoinductive products
US5736372A (en) 1986-11-20 1998-04-07 Massachusetts Institute Of Technology Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
US5053050A (en) 1988-04-29 1991-10-01 Samuel Itay Compositions for repair of cartilage and bone
US5162430A (en) 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
US5171273A (en) 1989-01-13 1992-12-15 University Of Medicine And Dentistry Of New Jersey Synthetic collagen orthopaedic structures such as grafts, tendons and other structures
US5015629A (en) 1989-06-26 1991-05-14 University Of Southern California Tissue repair
US5686116A (en) 1990-01-12 1997-11-11 New York Society For The Relief Of The Ruptured And Crippled, Maintaining The Hospital For Special Surgery Methods of enhancing repair, healing and augmentation of bone implants
ES2076467T3 (en) 1990-10-31 1995-11-01 El Gendler FLEXIBLE MEMBRANES PRODUCED WITH ORGANIC BONE MATTER FOR THE REPAIR AND RECONSTRUCTION OF PARTS OF THE SKELETON.
JPH07503869A (en) 1992-02-14 1995-04-27 ボード・オヴ・リージェンツ,ザ・ユニヴァーシティ・オヴ・テキサス・システム Multiphasic bioerodible implant materials or carriers and methods of manufacture and use thereof
US5589452A (en) 1992-07-14 1996-12-31 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
EP0730465B1 (en) 1993-09-24 2001-12-12 The University Of Southern California Use of angiotensin iii and analogs thereof in tissue repair
AU697504B2 (en) 1993-09-24 1998-10-08 University Of Southern California Use of angiotensin II analogs in tissue repair
US5955430A (en) 1993-09-24 1999-09-21 University Of Southern California Use of angiotensin II fragments and analogs thereof in tissue repair
US5443515A (en) 1994-01-26 1995-08-22 Implex Corporation Vertebral body prosthetic implant with slidably positionable stabilizing member
US5656598A (en) 1994-03-08 1997-08-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Use of fibroblast growth factors to stimulate bone growth
US5614515A (en) 1994-11-17 1997-03-25 University Of Southern California Lazaroid-based compositions and method for preventing adhesion formation using the same
US5834432A (en) 1995-06-06 1998-11-10 The University Of Southern California Use of angiotensin II Type 2 receptor agonists in tissue repair
EP0831796A1 (en) 1995-06-07 1998-04-01 University Of Southern California Method for reducing or preventing post-surgical adhesion formation using 5-lipoxygenase inhibitors
JP3422609B2 (en) 1995-11-14 2003-06-30 花王株式会社 Disposable diapers
US5700774A (en) 1996-03-26 1997-12-23 Genetics Institute, Inc. Compositions comprising bone morphogenic proteins and truncated parathyroid hormone related peptide, and methods of inducing cartilage by administration of same
US6110895A (en) 1996-12-16 2000-08-29 University Of Southern California Method of promoting healing in skin grafts
US6335195B1 (en) 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
AU6650098A (en) 1997-02-04 1998-08-25 University Of Southern California Method for accelerating healing of thermal injuries
JP2002507383A (en) 1997-11-26 2002-03-12 ユニヴァースティ オブ サザーン カリフォルニア Methods for promoting hematopoietic and mesenchymal cell proliferation and differentiation
JP4347522B2 (en) 1997-12-12 2009-10-21 ユニバーシティ オブ サザン カリフォルニア Wound healing composition
WO1999046285A2 (en) * 1998-03-11 1999-09-16 University Of Southern California Method of promoting production of living tissue equivalents

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106456698A (en) * 2014-05-19 2017-02-22 特维娜有限公司 Synthesis of beta-arrestin effectors

Also Published As

Publication number Publication date
ES2207258T3 (en) 2004-05-16
EP1094829B8 (en) 2004-09-22
JP3686335B2 (en) 2005-08-24
AU4986999A (en) 2000-02-01
JP2005120094A (en) 2005-05-12
AU756785B2 (en) 2003-01-23
US6258778B1 (en) 2001-07-10
JP2002520334A (en) 2002-07-09
EP1094829A2 (en) 2001-05-02
DE69911609D1 (en) 2003-10-30
CA2328871A1 (en) 2000-01-20
ATE250423T1 (en) 2003-10-15
EP1094829B1 (en) 2003-09-24
DE69911609T2 (en) 2004-07-01
WO2000002905A3 (en) 2000-02-24
WO2000002905A2 (en) 2000-01-20

Similar Documents

Publication Publication Date Title
CA2328871C (en) Methods for accelerating bone and cartilage growth and repair
US20050187163A1 (en) Methods for accelerating bone, cartilage, and connective tissue growth
EP0805853B1 (en) Lineage-directed induction of human mesenchymal stem cell differentiation
Matsuda et al. Mitogenic, chemotactic, and synthetic responses of rat periodontal ligament fibroblastic cells to polypeptide growth factors in vitro
JP4171727B2 (en) Methods for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US20020146823A1 (en) Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
Vukičević et al. Discovery and clinical applications of bone morphogenetic proteins
WO1996023059A9 (en) Lineage-directed induction of human mesenchymal stem cell differentiation
CZ20011893A3 (en) Pharmaceutical preparation, methods for treating health state and use of a peptide
EP0691849B1 (en) Oesteoblast growth stimulating compostion containing pdgf and vitamin d
CA2310852A1 (en) Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
CA2320404A1 (en) Method of promoting embryonic stem cell proliferation
AU750029B2 (en) Method of promoting neuronal cell proliferation and diffrentiation
MXPA01000405A (en) Methods for accelerating bone and cartilage growth and repair
CA2251808A1 (en) Methods for inducing bone formation
GB2343182A (en) Use of GnRH-II and analogues thereof for the treatment of osteoporosis
MXPA00008070A (en) Method of promoting embryonic stem cell proliferation
MXPA00008071A (en) Method of promoting neuronal cell proliferation and diffrentiation
WO2006020981A2 (en) Methods for accelerating bone growth and repair
MXPA00007704A (en) Method of promoting keratinocyte proliferation

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20160712