WO2012153333A1 - Régénération et réparation de tissu mésenchymateux par utilisation d'amélogénine - Google Patents

Régénération et réparation de tissu mésenchymateux par utilisation d'amélogénine Download PDF

Info

Publication number
WO2012153333A1
WO2012153333A1 PCT/IL2012/050163 IL2012050163W WO2012153333A1 WO 2012153333 A1 WO2012153333 A1 WO 2012153333A1 IL 2012050163 W IL2012050163 W IL 2012050163W WO 2012153333 A1 WO2012153333 A1 WO 2012153333A1
Authority
WO
WIPO (PCT)
Prior art keywords
amelogenin
ligament
tissue
treating
cells
Prior art date
Application number
PCT/IL2012/050163
Other languages
English (en)
Inventor
Dan DEUTSCH
Amir HAZE
Anat Blumenfeld
Original Assignee
Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd.
Hadasit Medical Research Services And Development Ltd.
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 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd., Hadasit Medical Research Services And Development Ltd. filed Critical Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd.
Priority to US14/116,796 priority Critical patent/US20140073765A1/en
Publication of WO2012153333A1 publication Critical patent/WO2012153333A1/fr
Priority to US14/471,262 priority patent/US9957314B2/en
Priority to US15/899,621 priority patent/US11034753B2/en

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
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders

Definitions

  • the present invention in some embodiments thereof, relates to the use of amelogenin for enhancing the regeneration of mesenchymal tissue, and more specifically ligaments and tendons.
  • Ligaments and tendons are elastic collagenous tissues with similar composition and hierarchical structure. Their strength is related to the number and size of collagen fibrils. Water is the predominant component of ligaments and tendons at 55-70% (higher in tendons). Additional components include collagen (type I predominant, some type III) accounting for 70-80% of the dry weight, elastin, proteoglycans, glycosaminoglycans and glycoproteins (e.g. fibronectin, thrombospondin). Fibrillar collagen type I provides ligaments and tendons their high tensile strength and is responsible for the hierarchical structure.
  • Ligaments play an essential role in mediating normal movement and stability of joints, connecting bones to each other in order to restrict their relative motion. They are also involved in proprioception since they comprise mechanoreceptors which serve to protect from extremes of motion, thereby, maintaining the stability of joints or providing feedback control that changes muscle activity when resistance to movement is encountered.
  • Tendon injuries can also be chronic, usually elicited by repetitive mechanical loading below the failure threshold and concurrent inflammatory responses.
  • the most common of these injuries are of the anterior cruciate and medial collateral ligaments of the knee, the supraspinatus tendon of the rotator cuff, the Achilles tendon, and the flexor tendons of the hand.
  • full healing of a torn ligament or tendon fails to take place, and replacement tissues or grafts are required.
  • When healing does occur typically the mechanical properties do not return to normal, owing to the development of scar tissues at the injured site, the biomechanical properties of which are inferior to uninjured ligament or tendon.
  • the loss of mechanical competence is mainly due to a distorted extra-cellular matrix (ECM) composition and a misalignment of collagen fibrils in the scar tissue.
  • ECM extra-cellular matrix
  • the amelogenins are a major component of the developing extracellular enamel matrix proteins, produced by the ameloblast cells and play a major role in the biomineralization and structural organization of enamel (Robinson et al. 1998).
  • the human amelogenin gene contains 7 exons, which undergo alternative mRNA splicing.
  • the most abundant amelogenin lacks the internal region encoded by exon 4, is termed HX175 in human, which corresponds to isoform Ml 80 in mice.
  • HX175 The relatively large number of amelogenin alternatively spliced mRNA translated polypeptides and the fact that amelogenin is expressed in different tissues (calcifying and soft tissues) and of different embryonic origin, possibly reflect different functions of amelogenin.
  • Amelogenin was shown to be expressed in periodontal ligament (PDL) cells, in long bone cells; osteocytes, osteoblasts and osteoclasts, in cartilage chondrocytes and differentially in growth plate cells.
  • PDL periodontal ligament
  • osteocytes, osteoblasts and osteoclasts in cartilage chondrocytes and differentially in growth plate cells.
  • amelogenin was identified in long bone marrow stromal cells, some of which are multi-potent stem cells (Haze et al. 2007).
  • amelogenin expression is increased at sites of high activity and remodeling of ligaments and bones (Haze et al. 2009).
  • Amelogenin expression was also identified in cells of non-mineralizing tissues such as brain and eye in embryonic and postnatal tissues (Deutsch et al. 2006, Gruenbaum-Cohen et al. 2008).
  • amelogenin has been shown to be beneficial for the treatment of periodontitis (Haze et al. 2009).
  • International Patent Application WO2011/030185 teaches a cell guiding scaffold which according to one embodiment may comprise amelogenin as one of its active agents for inducing periodontal tissue regeneration.
  • the scaffold may be used for joint ligament regeneration as well.
  • U.S. Patent Applications 20100093632 and 20030077291 teach the use of amelogenin for the treatment of inflammatory disorders.
  • Additional background material includes European Patent Publication Nos. 0337967, 1862143 and 0053197 and US Patent Application No. 20110003745.
  • a method of treating an injury to or a disease of a skeletal joint ligament or tendon in a subject in need thereof comprising contacting the skeletal joint ligament or tendon of the subject with a therapeutically effective amount of amelogenin, wherein the amelogenin is not comprised in a scaffold, thereby treating the injury to or disease of the skeletal joint ligament or tendon.
  • a method of treating an injury to or a disease of a skeletal muscle ligament or tendon in a subject in need thereof comprising contacting the skeletal muscle ligament or tendon of the subject with a therapeutically effective amount of mesenchymal stem cells (MSCs) which have been genetically modified to express amelogenin, thereby treating the injury to or disease of the skeletal joint ligament or tendon.
  • MSCs mesenchymal stem cells
  • a method of treating osteoarthritis in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of amelogenin, thereby treating the osteoarthritis.
  • a method of enhancing regeneration of cardiac tissue the method comprising contacting the cardiac tissue with amelogenin, thereby enhancing regeneration of the cardiac tissue.
  • a method of treating a disease associated with cardiac tissue degeneration in a subject in need thereof comprising administering to the subject a therapeutically effective amount of amelogenin, thereby treating the disease associated with cardiac tissue degeneration.
  • a method of treating an injury to, or a disease of, mesenchymal tissue in a subject in need thereof comprising administering to the subject a therapeutically effective amount of mesenchymal stem cells (MSCs) which have been genetically modified to express amelogenin, with the proviso that the mesenchymal tissue does not comprise periodontal tissue, thereby treating the injury to or disease of the mesenchymal tissue.
  • MSCs mesenchymal stem cells
  • a method of treating an injury associated with a meniscus, labrum and spinal intervertebral disc in a subject in need thereof comprising contacting the meniscus, labrum or disc of the subject with a therapeutically effective amount of amelogenin, thereby treating the injury to or disease of the meniscus, labrum and spinal disc.
  • the amelogenin is expressed in a population of MSCs.
  • the amelogenin is human amelogenin.
  • the amelogenin comprises an amino acid sequence as set forth in SEQ ID NO: 1.
  • the mesenchymal tissue is selected from the group consisting of ligament, tendon, cartilage, bone, muscle and fat.
  • the MSCs are isolated from bone marrow tissue.
  • the amelogenin is expressed from an adenoviral vector.
  • FIGs. 1A-D are photographs illustrating transected medial collateral ligament (MCL) two weeks after operation, the dotted yellow lines mark the borders of the ligament.
  • MCL medial collateral ligament
  • FIGs. 1A-D are photographs illustrating transected medial collateral ligament (MCL) two weeks after operation, the dotted yellow lines mark the borders of the ligament.
  • A Transacted MCL, treated with PGA carrier only (control) as compared with (B) non-transected MCL, from the contra-leg.
  • C Transacted MCL, treated with rHAM + dissolved in PGA (experimental) as compared with (D) non-transected MCL, from the contra-leg.
  • the dotted line marks the transection zone.
  • the results indicate closure of the gap between the stumps, already two weeks following treatment with rHAM + dissolved in PGA (experimental), while no such recovery was demonstrated after treatment with PGA carrier only (control).
  • FIGs. 2A-B are graphs illustrating maximal force comparison (A) and stiffness (slope) comparison (B) between the transected MCL to the non-transected normal MCL from the contra-lateral leg of each rat.
  • the difference between the experimental and control groups was significant (A. p ⁇ 0.03, B. p ⁇ 0.01).
  • FIGs. 5A-C are photographs illustrating immunohistochemical staining of the mesenchymal stem cell marker CD 105 (brown staining) in the ligaments 4 days following transection.
  • the dotted yellow line marks the borders of the transected ligament, all the other tissue is inflammatory tissue filling the gap between the ligament stumps.
  • the photographs show that there are a high number of cells expressing CD 105, indicating recruitment of MSCs in the filled gap of the experimental ligament, while almost no CD 105 recruited MSCs are detected in the filled gap of the control ligament.
  • FIGs. 6A-D are photographs illustrating the size of the osteochondreal defect (OCD) at the medial femoral condyle 6 weeks after application of different concentration of rHAM + dissolved in PGA (experimental) or PGA only (control).
  • the dotted red circle marks the size of the OCD 6 weeks after the treatment.
  • the size of the defect decreased in all concentrations of rHAM .
  • the control group treated with PGA carrier only
  • the size of the defect was increased and degenerative changes were seen in large areas of the articular surface.
  • FIGs. 7A-D are photographs illustrating the size of the osteochondreal defect (OCD) at the medial femoral condyle 12 weeks after application of different concentration of rHAM + dissolved in PGA (experimental) or PGA only (control).
  • the dotted red circle marks the size of the OCD 12 weeks after the treatment.
  • the size of the defect decreased in all concentrations of rHAM .
  • the control group treated with PGA carrier only
  • the size of the defect was increased and degenerative changes were seen in large areas of the articular surface.
  • FIGs. 8A-D are photographs illustrating the size of the osteochondreal defect (OCD) in the femoral trochlea (marked by dotted yellow line); A-B- at the time of the operation, C- 12 weeks after application of 1 ⁇ / ⁇ 1 rHAM + dissolved in PGA (experimental) and D- 12 weeks after application of PGA only (control). C and D represent the defect area in a higher magnification than A and B, respectively, 12 weeks after the beginning of the experiment. In the control group (treated with PGA carrier only), the size of the defect was increased and degenerative changes were seen in large areas of the articular surface, while in the experimental knee the changes were much more limited and the OCD was filled with tissue.
  • OCD osteochondreal defect
  • control application of PGA carrier only
  • a fibrous tissue, not resembling hyaline cartilage was formed.
  • the experimental application of rHAM + dissolved in PGA carrier
  • a multilayered cartilaginous tissue is seen, its structure resembles the structure of the hyaline cartilage.
  • FIG. 11 is a photograph illustrating immunohistochemical analysis of rat skeletal muscle with monoclonal anti-human ameloganin antibody. The brown staining indicates amelogenin expression in the fibers of rat skeletal muscle.
  • FIG. 12 is a photograph illustrating immunohistochemical analysis of mouse E9 (embryonic day 9) heart with anti-human amelogenin antibody. The red staining indicates amelogenin expression in the mouse E9 embryo heart. Arrow indicates the myocardium.
  • the present invention in some embodiments thereof, relates to the use of amelogenin for enhancing the regeneration of mesenchymal tissue, and more specifically ligaments and tendons.
  • the amelogenins are a major component of the developing extracellular enamel matrix proteins, produced by the ameloblast cells and play a major role in the biomineralization and structural organization of enamel (Robinson et al. 1998).
  • the human amelogenin gene contains 7 exons, which undergo alternative mRNA splicing.
  • the most abundant amelogenin lacks the internal region encoded by exon 4, is termed HX175 in human, which corresponds to isoform Ml 80 in mice.
  • HX175 The relatively large number of amelogenin alternatively spliced mRNA translated polypeptides and the fact that amelogenin is expressed in different tissues (calcifying and soft tissues) and of different embryonic origin, possibly reflect different functions of amelogenin.
  • amelogenin may be used to enhance regeneration of mesenchymal tissue, including ligaments and tendons.
  • Local administration of recombinant human amelogenin following injury to the ligament enhanced regeneration of the tissue in a rat model ( Figures 1A-D).
  • the ligament regenerated in the presence of amelogenin showed improved mechanical properties as compared with a ligament regenerated in the absence of amelogenin ( Figures 2A-B).
  • a method of treating an injury to, or a disease of, mesenchymal tissue in a subject in need thereof comprising administering to the subject a therapeutically effective amount of recombinant amelogenin protein and /or mesenchymal stem cells (MSCs) which have been genetically modified to express amelogenin, with the proviso that the mesenchymal tissue does not comprise periodontal tissue, thereby treating the injury to or disease of the mesenchymal tissue.
  • MSCs mesenchymal stem cells
  • mesenchymal stem cell or “MSC” is used interchangeably for adult cells which are not terminally differentiated, which can divide to yield cells that are either stem cells, or which, irreversibly differentiate to give rise to cells of a mesenchymal cell lineage e.g., adipose, osseous, cartilaginous, elastic and fibrous connective tissues, myoblasts, as well as to tissues other than those originating in the embryonic mesoderm (e.g., neural cells) depending upon various influences from bioactive factors such as cytokines.
  • a mesenchymal cell lineage e.g., adipose, osseous, cartilaginous, elastic and fibrous connective tissues, myoblasts, as well as to tissues other than those originating in the embryonic mesoderm (e.g., neural cells) depending upon various influences from bioactive factors such as cytokines.
  • MSC cultures utilized by some embodiments of the invention preferably include three groups of cells which are defined by their morphological features: small and agranular cells (referred to as RS-1, hereinbelow), small and granular cells (referred to as RS-2, hereinbelow) and large and moderately granular cells (referred to as mature MSCs, hereinbelow).
  • RS-1 small and agranular cells
  • RS-2 small and granular cells
  • mature MSCs large and moderately granular cells
  • MSCs When MSCs are cultured under the culturing conditions of some embodiments of the invention they exhibit negative staining for the hematopoietic stem cell markers CD34, CD11B, CD43 and CD45. A small fraction of cells (less than 10 %) are dimly positive for CD31 and/or CD38 markers.
  • the mesenchymal stem cells are human.
  • the mesenchymal stem cells are isolated from newborn humans.
  • the mesenchymal stem cells are autologous to the patient being treated.
  • the mesenchymal stem cells are non-autologous (allergenic) to the patient being treated.
  • the mesenchymal stem cells may be derived from various tissues including but not limited to bone marrow, peripheral blood, placenta (e.g. fetal side of the placenta), cord blood, umbilical cord, amniotic fluid and from adipose tissue.
  • placenta e.g. fetal side of the placenta
  • cord blood e.g. umbilical cord
  • amniotic fluid e.g., amniotic fluid and from adipose tissue.
  • a method of enriching for mesenchymal stem cells from peripheral blood is described by Kassis et al [Bone Marrow Transplant. 2006 May; 37(10): 967-76].
  • a method of isolating mesenchymal stem cells from placental tissue is described by Zhang et al [Chinese Medical Journal, 2004, 117 (6):882-887].
  • Methods of enriching for adipose tissue, placental and cord blood mesenchymal stem cells are described by Kern et al [Stem Cells, 2006; 24: 1294-1301].
  • Bone marrow can be isolated from the iliac crest (or other bone) of an individual by aspiration.
  • Low-density BM mononuclear cells BMMNC
  • BMMNC Low-density BM mononuclear cells
  • Hetastarch hydroxy ethyl starch
  • mesenchymal stem cell cultures are generated by diluting BM aspirates (usually 20 ml) with equal volumes of Hank's balanced salt solution (HBSS; GIBCO Laboratories, Grand Island, NY, USA) and layering the diluted cells over about 10 ml of a Ficoll column (Ficoll-Paque; Pharmacia, Piscataway, NJ, USA).
  • MSC mononuclear cell layer
  • FCS fetal calf serum
  • Adipose tissue-derived MSCs can be obtained by liposuction and mononuclear cells can be isolated manually by removal of the fat and fat cells, or using the Celution System (Cytori Therapeutics) following the same procedure as described above for preparation of MSCs.
  • the MSCs are at least 50 % purified, more preferably at least 75 % purified and even more preferably at least 90 % purified.
  • Methods of purifying MSCs include for example culturing (in vitro or ex vivo) on polystyrene plastic surfaces (e.g. in a flask) by removing non-adherent cells (i.e. non-mesenchymal stem cells).
  • MSCs are known in the art including for example positive selection against mesenchymal stem cell markers (e.g. CD 105) and/or negative selection against hematopoietic stem and progenitor markers such as CD34, CD133, CD8, etc.
  • Methods of determining protein cell-surface expression are well known in the art. Examples include immunological methods, such as, FACS analysis as well as biochemical methods (cell-surface labeling, e.g., radioactive, fluorescence, avidin- biotin).
  • the proliferation medium may be DMEM, alpha-MEM or DMEM/F12.
  • the proliferation medium is DMEM.
  • the proliferation medium further comprises SPN, L-glutamine and a serum (such as fetal calf serum or horse serum).
  • mesenchymal stem cells are effected so that they express the polypeptide amelogenin. This is preferably effected by transforming such cells with an expression construct which is designed for expression of amelogenin.
  • amelogenin refers to any one of the alternatively spliced variants of the mammalian amelogenin polypeptide (e.g., human, rat, mouse amelogenin) which exhibits an amelogenin activity, e.g. enhancement of mesenchymal tissue regeneration.
  • the GenPept REFSEQ numbers for the 3 alternative protein isoforms of amelogenin are set forth in NP_001133.1, NP_872621.1, NP_872622.1.
  • the GeneBank REFSEQ transcripts for amelogenin for the 3 human alternative transcripts are set fourth in NM_001142.2, NMJ82680.1 and NM_182681.1. Additional cDNA sequences (also including protein sequence) include: GeneBank AF436849.1; BC069118.1; BC074951.2; M86932.1 and S67147.1.
  • An amelogenin of the present invention also refers to homologs (e.g., polypeptides which are at least 50 %, at least 55 %, at least 60 %, at least 65 %, at least 70 %, at least 75 %, at least 80 %, at least 85 %, at least 87 %, at least 89 %, at least 91 %, at least 93 %, at least 95 % or more say 100 % homologous to the amelogenin sequences described above (e.g. SEQ ID NO: 1) as determined using BlastP software of the National Center of Biotechnology Information (NCBI) using default parameters).
  • the homolog may also refer to a deletion, insertion, or substitution variant, including an amino acid substitution, thereof and biologically active polypeptide fragments thereof.
  • the expression construct of the present invention preferably includes a polynucleotide sequence encoding the amelogenin under control of a transcriptional regulatory sequence (e.g. a promoter).
  • a transcriptional regulatory sequence e.g. a promoter
  • an isolated polynucleotide refers to a single or double stranded nucleic acid sequence which is isolated and provided in the form of an RNA sequence, a complementary polynucleotide sequence (cDNA), a genomic polynucleotide sequence and/or a composite polynucleotide sequences (e.g., a combination of the above).
  • complementary polynucleotide sequence refers to a sequence, which results from reverse transcription of messenger RNA using a reverse transcriptase or any other RNA dependent DNA polymerase. Such a sequence can be subsequently amplified in vivo or in vitro using a DNA dependent DNA polymerase.
  • genomic polynucleotide sequence refers to a sequence derived (isolated) from a chromosome and thus it represents a contiguous portion of a chromosome.
  • composite polynucleotide sequence refers to a sequence, which is at least partially complementary and at least partially genomic.
  • a composite sequence can include some exonal sequences required to encode the polypeptide of the present invention, as well as some intronic sequences interposing therebetween.
  • the intronic sequences can be of any source, including of other genes, and typically will include conserved splicing signal sequences. Such intronic sequences may further include cis acting expression regulatory elements.
  • the expression construct can be designed as a gene knock-in construct in which case it will lead to genomic integration of construct sequences, or it can be designed as an episomal expression vector.
  • the expression construct can be generated using standard ligation and restriction techniques, which are well known in the art (see Maniatis et al., in: Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1982). Isolated plasmids, DNA sequences, or synthesized oligonucleotides are cleaved, tailored, and religated in the form desired.
  • Promoters suitable for use with the present invention may be constitutive, tissue specific or regulatable (e.g. comprise response elements capable for directing transcription of the polynucleotide sequence so as to confer regulatable synthesis of the amelogenin).
  • Constitutive promoters suitable for use with some embodiments of the invention are promoter sequences which are active under most environmental conditions and most types of cells such as the cytomegalovirus (CMV) and Rous sarcoma virus (RSV).
  • CMV cytomegalovirus
  • RSV Rous sarcoma virus
  • a suitable response element for use in regulatable promoters can be, for example, a tetracycline response element (such as described by Gossen and Bujard (Proc. Natl. Acad. Sci. USA 89:5547-551, 1992); an ectysone-inducible response element (No D et al., Proc Natl Acad Sci U S A. 93:3346-3351, 1996) a metal-ion response element such as described by Mayo et al. (Cell. 29:99-108, 1982); Brinster et al. (Nature 296:39-42, 1982) and Searle et al. (Mol. Cell. Biol.
  • a heat shock response element such as described by Nouer et al. (in: Heat Shock Response, ed. Nouer, L., CRC, Boca Raton, Fla., ppl67-220, 1991); or a hormone response element such as described by Lee et al. (Nature 294:228-232, 1981); Hynes et al. (Proc. Natl. Acad. Sci. USA 78:2038-2042, 1981); Klock et al. (Nature 329:734-736, 1987); and Israel and Kaufman (Nucl. Acids Res. 17:2589-2604, 1989).
  • a heat shock response element such as described by Nouer et al. (in: Heat Shock Response, ed. Nouer, L., CRC, Boca Raton, Fla., ppl67-220, 1991); or a hormone response element such as described by Lee et al. (Nature 294:228-232, 1981); Hynes et al.
  • Enhancer elements can stimulate transcription up to 1,000 fold from linked homologous or heterologous promoters. Enhancers are active when placed downstream or upstream from the transcription initiation site. Many enhancer elements derived from viruses have a broad host range and are active in a variety of tissues. For example, the SV40 early gene enhancer is suitable for many cell types. Other enhancer/promoter combinations that are suitable for the present invention include those derived from polyoma virus, human or murine cytomegalovirus (CMV), the long term repeat from various retroviruses such as murine leukemia virus, murine or Rous sarcoma virus and HIV. See, Enhancers and Eukaryotic Expression, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. 1983, which is incorporated herein by reference.
  • CMV cytomegalovirus
  • Polyadenylation sequences can also be added to the expression construct in order to increase the translation efficiency of the enzyme expressed from the expression construct of the present invention.
  • Two distinct sequence elements are required for accurate and efficient polyadenylation: GU or U rich sequences located downstream from the polyadenylation site and a highly conserved sequence of six nucleotides, AAUAAA, located 11-30 nucleotides upstream.
  • Termination and polyadenylation signals that are suitable for the present invention include those derived from SV40.
  • the expression construct of the present invention may typically contain other specialized elements intended to increase the level of expression of cloned polynucleotides or to facilitate the identification of cells that carry the recombinant DNA.
  • a number of animal viruses contain DNA sequences that promote the extra chromosomal replication of the viral genome in permissive cell types. Plasmids bearing these viral replicons are replicated episomally as long as the appropriate factors are provided by genes either carried on the plasmid or with the genome of the host cell.
  • the expression construct may or may not include a eukaryotic replicon. If a eukaryotic replicon is present, then the vector is amplifiable in eukaryotic cells using the appropriate selectable marker. If the construct does not comprise a eukaryotic replicon, no episomal amplification is possible. Instead, the recombinant DNA integrates into the genome of the engineered cell, where the promoter directs expression of the desired polynucleotide.
  • mammalian expression constructs include, but are not limited to, pcDNA3, pcDNA3.1 (+/-), pGL3, pZeoSV2(+/-), pSecTag2, pDisplay, pEF/myc/cyto, pCMV/myc/cyto, pCR3.1, pSinRep5, DH26S, DHBB, pNMTl, pNMT41, pNMT81 , which are available from Invitrogen, pCI which is available from Promega, pMbac, pPbac, pBK-RSV and pBK-CMV which are available from Strategene, pTRES which is available from Clontech, and their derivatives.
  • SV40 vectors include pSVT7 and pMT2.
  • Vectors derived from bovine papilloma virus include pBV- 1MTHA, and vectors derived from Epstein Bar virus include pHEBO, and p205.
  • exemplary vectors include pMSG, pAV009/A + , pMTO10/A + , pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV-40 early promoter, SV-40 later promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.
  • Viruses are specialized infectious agents that have evolved, in many cases, to elude host defense mechanisms. Typically, viruses infect and propagate in specific cell types.
  • the targeting specificity of viral vectors utilizes its natural specificity to specifically target predetermined cell types and thereby introduce a recombinant gene into the infected cell.
  • the type of vector used by the present invention will depend on the cell type transformed. The ability to select suitable vectors according to the cell type transformed is well within the capabilities of the ordinary skilled artisan and as such no general description of selection consideration is provided herein.
  • bone marrow cells can be targeted using the human T cell leukemia virus type I (HTLV- I)-
  • Recombinant viral vectors are useful for in vivo expression of transgenic polynucleotides since they offer advantages such as lateral infection and targeting specificity.
  • Lateral infection is inherent in the life cycle of, for example, retrovirus and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is that a large area becomes rapidly infected, most of which was not initially infected by the original viral particles. This is in contrast to vertical-type of infection in which the infectious agent spreads only through daughter progeny.
  • Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells.
  • transformed cells are generated, they are tested (in culture) for their ability to express and synthesize amelogenin analyzed using standard chemical analytical methods such as, for example, HPLC, ELISA or GC-MS.
  • the cultures are comparatively analyzed for expression of the recombinant enzyme (e.g., amelogenin), using biochemical analytical methods such as immunoassays, Western blot and Realtime PCR.
  • the genetically modified cells of this aspect of the present invention may be seeded on a scaffold prior to use.
  • the term "scaffold” refers to a 3D matrix upon which cells may be cultured (i.e., survive and preferably proliferate for a predetermined time period).
  • Static seeding includes incubation of a cell-medium suspension in the presence of the scaffold under static conditions and results in non-uniformity cell distribution (depending on the volume of the cell suspension); filtration seeding results in a more uniform cell distribution; and centrifugation seeding is an efficient and brief seeding method (see for example EP 19980203774).
  • the cells may be seeded directly onto the scaffold, or alternatively, the cells may be mixed with a gel which is then absorbed onto the interior and exterior surfaces of the scaffold and which may fill some of the pores of the scaffold. Capillary forces will retain the gel on the scaffold before hardening, or the gel may be allowed to harden on the scaffold to become more self-supporting.
  • the cells may be combined with a cell support substrate in the form of a gel optionally including extracellular matrix components.
  • the present invention also contemplates treating subjects with amelogenin alone.
  • the amelogenin when used to enhance mesenchymal tissue repair without the use of MSCs, the amelogenin is not incorporated into a scaffold.
  • the amelogenin is a recombinant amelogenin.
  • prokaryotic or eukaryotic cells can be used as host-expression systems to express the amelogenin of the present invention. These include, but are not limited to, microorganisms, such as bacteria transformed with a recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vector containing the polypeptide coding sequence; yeast transformed with recombinant yeast expression vectors containing the polypeptide coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors, such as Ti plasmid, containing the polypeptide coding sequence.
  • microorganisms such as bacteria transformed with a recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vector containing the polypeptide coding sequence
  • non-bacterial expression systems e.g. mammalian expression systems such as CHO cells
  • polypeptides of the present invention are glycosylated.
  • a number of expression vectors can be advantageously selected.
  • vectors that direct the expression of high levels of the protein product possibly as a fusion with a hydrophobic signal sequence, which directs the expressed product into the periplasm of the bacteria or the culture medium where the protein product is readily purified may be desired.
  • Certain fusion protein engineered with a specific cleavage site to aid in recovery of the polypeptide may also be desirable.
  • Such vectors adaptable to such manipulation include, but are not limited to, the pET series of E. coli expression vectors [Studier et al, Methods in Enzymol. 185:60-89 (1990)].
  • yeast a number of vectors containing constitutive or inducible promoters can be used, as disclosed in U.S. Pat. Application No: 5,932,447.
  • vectors can be used which promote integration of foreign DNA sequences into the yeast chromosome.
  • the expression of the polypeptide coding sequence can be driven by a number of promoters.
  • viral promoters such as the 35S RNA and 19S RNA promoters of CaMV [Brisson et al., Nature 310:511-514 (1984)], or the coat protein promoter to TMV [Takamatsu et al., EMBO J. 3:17-311 (1987)] can be used.
  • plant promoters can be used such as, for example, the small subunit of RUBISCO [Coruzzi et al., EMBO J.
  • the expression construct of the present invention can also include sequences engineered to optimize stability, production, purification, yield or activity of the expressed polypeptide.
  • Transformed cells are cultured under effective conditions, which allow for the expression of high amounts of recombinant polypeptide.
  • Effective culture conditions include, but are not limited to, effective media, bioreactor, temperature, pH and oxygen conditions that permit protein production.
  • An effective medium refers to any medium in which a cell is cultured to produce the recombinant polypeptide of the present invention.
  • Such a medium typically includes an aqueous solution having assimilable carbon, nitrogen and phosphate sources, and appropriate salts, minerals, metals and other nutrients, such as vitamins.
  • Cells of the present invention can be cultured in conventional fermentation bioreactors, shake flasks, test tubes, microtiter dishes and petri plates. Culturing can be carried out at a temperature, pH and oxygen content appropriate for a recombinant cell. Such culturing conditions are within the expertise of one of ordinary skill in the art.
  • the ameogenin of the present invention may either remain within the recombinant cell, secreted into the fermentation medium, secreted into a space between two cellular membranes, such as the periplasmic space in E. coli; or retained on the outer surface of a cell or viral membrane.
  • the phrase "recovering the amelogenin" used herein refers to collecting the whole fermentation medium containing the amelogenin and need not imply additional steps of separation or purification.
  • amelogenins of the present invention can be purified using a variety of standard protein purification techniques, such as, but not limited to, affinity chromatography, ion exchange chromatography, filtration, electrophoresis, hydrophobic interaction chromatography, gel filtration chromatography, reverse phase chromatography, concanavalin A chromatography, chromatofocusing and differential solubilization.
  • standard protein purification techniques such as, but not limited to, affinity chromatography, ion exchange chromatography, filtration, electrophoresis, hydrophobic interaction chromatography, gel filtration chromatography, reverse phase chromatography, concanavalin A chromatography, chromatofocusing and differential solubilization.
  • the expressed coding sequence can be engineered to encode the amelogenin fused to a cleavable moiety.
  • a fusion protein can be designed so that the polypeptide can be readily isolated by affinity chromatography; e.g., by immobilization on a column specific for the cleavable moiety.
  • the polypeptide can be released from the chromatographic column by treatment with an appropriate enzyme or agent that specifically cleaves the fusion protein at this site [e.g., see Booth et al., Immunol. Lett. 19:65-70 (1988); and Gardella et al., J. Biol. Chem.
  • the amelogenin is preferably retrieved in "substantially pure" form.
  • the phrase "substantially pure” refers to a purity that allows for the effective use of the amelogenin in the applications described herein.
  • the amelogenin can also be synthesized using in vitro expression systems. These methods are well known in the art and the components of the system are commercially available.
  • the genetically modified cells of this aspect of the present invention may be used for enhancing mesenchymal tissue repair or regeneration.
  • mesenchymal tissue examples include, but are not limited to ligament, tendon, cartilage, meniscus, intervertebral disc, bone, fat, and muscle.
  • enhancing repair and or regeneration refers one or more of the following: increase in the rate of production of new tissue, improvement in the functionality, amount or quality of the new tissue produced as compared to untreated control.
  • this term can refer to increase in rate of new bone formation, to formation of bone where without treatment no bone would have formed, or to increase in the mass and mechanical parameters of the new bone formed.
  • cartilage after an injury the body usually produces a scar tissue with some characteristics of cartilage (fibrocartilage), which in some cases may enable functional joint movement (may be painful), but eventually the joint will degenerate and osteoarthritis will develop. Few types of cartilage are known.
  • the articular hyaline cartilage, the menisceal tissue, the labrum tissue (acetabular labrum and glenoid labrum) and the intervertebral disc generally do not naturally regenerate. Regeneration of hyaline cartilage means preferential formation of functional cartilage (articular cartilage in joints) sufficient enough to enable joint function and/or to prevent further pain and destruction e.g. osteoarthritis.
  • menisci most of the meanisceal tears are at the white-white zone, which is an area without blood supply. Tears in this area and most of the tears in the red- white zone will not heal and partial menisectomy should be performed. Regeneration of menisci refers to formation of new menisceal cartilaginous tissue connection between the sides of a tear. It can also refer to renewal of degenerative menisceal tissue.
  • the acetabular and glenoid labrums are fibrocartilagenous structures, which serve as static stabilizers of the hip and shoulder joints. Tears or detachment of the labrum from the bone, causes pain, disability and joint instability (mainly in the shoulder).
  • the intervertebral discs are fibrocartilagenous, with surrounding annulus fibrosus composed mainly of collagen type 1 and a softer central nucleus pulposus made mainly of type 2 collagen.
  • the nucleus pulposus has a high content of polysaccharide and is approximately 88% water. Aging results in loss of water and conversion to fibrocartilage.
  • Regeneration of intervertebral disc refers to closure or decrease in the size of the annulus fibrosus tear, and/or to renewal of the nucleus pulposus composition. As regards to ligament and tendon, these tissues also generally do not normally regenerate. In part, scar tissue forms, which has significantly inferior mechanical properties.
  • Regeneration of ligament and tendon means formation of new connection between the edges of the stumps, with similar mechanical properties to the tissue prior to the injury.
  • Muscle regeneration refers to formation of new functional connection between the edges of a torn muscle, or to renewal of functional muscular units after an insult e.g. ischemic heart attack.
  • the method of the invention is intended to treat conditions where ligament, tendon, cartilage, intervertebral disc, menisci, bone, cardiac muscle and skeletal muscle are damaged due to trauma or pathological conditions, including degeneration due to normal age and exercise.
  • Examples of conditions that may be treated in accordance with the above two methods include:
  • Extra-articular ligament rapture as the anterior talo-fibular and deltoid ligaments of the ankle, which are torn in severe ankle sprain.
  • Intra-articular ligament rapture as the shoulder capsular ligaments (e.g. inferior glenohumeral ligament, which prevents anterior shoulder dislocation), the anterior and posterior cruciate ligaments of the knee.
  • Traumatic or degenerative tear of skeletal muscle Traumatic or degenerative tear of skeletal muscle.
  • Enhancement of bone integration between implants and the patient bone e.g. orthopaedic implants and dental implants.
  • the present invention can be advantageously used to treat disorders associated with, for example, necrotic, apoptotic, damaged, dysfunctional or morphologically abnormal myocardium.
  • disorders include, but are not limited to, ischemic heart disease, cardiac infarction, congestive heart failure, rheumatic heart disease, endocarditis, autoimmune cardiac disease, valvular heart disease, congenital heart disorders, cardiac rhythm disorders, impaired myocardial conductivity and cardiac insufficiency. Since the majority of cardiac diseases involve necrotic, apoptotic, damaged, dysfunctional or morphologically abnormal myocardium, the genetically modified MSCs (and amelogenin alone) described herein can be used to treat the majority of instances of cardiac disorders.
  • the method according to this aspect of the present invention can be advantageously used to efficiently reverse, inhibit or prevent cardiac damage caused by ischemia resulting from myocardial infarction.
  • the genetically modified MSCs (or the amelogenin alone) can be administered either per se or, preferably as a part of a pharmaceutical composition that further comprises a pharmaceutically acceptable carrier.
  • a "pharmaceutical composition” refers to a preparation of amelogenin or cells genetically engineered to express amelogenin, with other chemical components such as pharmaceutically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to a subject.
  • the term "pharmaceutically acceptable carrier” refers to a carrier or a diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compound.
  • examples, without limitations, of carriers are propylene glycol alginate, saline, emulsions and mixtures of organic solvents with water.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • the pharmaceutical carrier is an aqueous solution of saline.
  • Suitable routes of administration include direct administration into the tissue or organ of interest (local administration).
  • the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays.
  • a dose is formulated in an animal model to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition, (see e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p. l).
  • the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • Dosage amount and interval may be adjusted individually so that sufficient amount of amelogenin reach the appropriate cells. Dosages necessary to achieve the desired effect will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
  • dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or diminution of the disease state is achieved.
  • the amount of a composition to be administered will, of course, be dependent on the individual being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • the dosage and timing of administration will be responsive to a careful and continuous monitoring of the individual changing condition.
  • the protein and/or the engineered cells may be administered to the desired site by direct application either by an injection, by arthroscopic device or during open surgery.
  • the cells may be placed in an isolated form, or placed in a suitable medium, or in a suitable matrix including scaffold matrixes that may incorporate the cells.
  • the cells may be administered alone or together with other compounds intended to promote activity or proliferation of MSC, or with other compounds known to enhance regeneration and/or repair of these tissues.
  • non-autologous cells are likely to induce an immune reaction when administered to the body
  • approaches have been developed to reduce the likelihood of rejection of non-autologous cells. These include either suppressing the recipient's immune system, providing anti-inflammatory treatment and/or encapsulating the non-autologous cells in immunoisolating, semipermeable membranes before transplantation.
  • Encapsulation techniques are generally classified as microencapsulation, involving small spherical vehicles and macroencapsulation, involving larger flat-sheet and hollow-fiber membranes (Uludag, H. et al. Technology of mammalian cell encapsulation. Adv Drug Deliv Rev. 2000; 42: 29-64).
  • Methods of preparing microcapsules are known in the arts and include for example those disclosed by Lu MZ, et al., Cell encapsulation with alginate and alpha- phenoxycinnamylidene-acetylated poly(allylamine). Biotechnol Bioeng. 2000, 70: 479- 83, Chang TM and Prakash S.
  • microcapsules are prepared by complexing modified collagen with a ter-polymer shell of 2-hydroxyethyl methylacrylate (HEMA), methacrylic acid (MAA) and methyl methacrylate (MMA), resulting in a capsule thickness of 2-5 ⁇ .
  • HEMA 2-hydroxyethyl methylacrylate
  • MAA methacrylic acid
  • MMA methyl methacrylate
  • Such microcapsules can be further encapsulated with additional 2-5 ⁇ ter-polymer shells in order to impart a negatively charged smooth surface and to minimize plasma protein absorption (Chia, S.M. et al. Multi-layered microcapsules for cell encapsulation Biomaterials. 2002 23: 849-56).
  • microcapsules are based on alginate, a marine polysaccharide (Sambanis,
  • microcapsules can be prepared by the polyelectrolyte complexation between the polyanions sodium alginate and sodium cellulose sulphate with the polycation poly(methylene-co-guanidine) hydrochloride in the presence of calcium chloride.
  • immunosuppressive agents include, but are not limited to, methotrexate, cyclophosphamide, cyclosporine, cyclosporin A, chloroquine, hydroxychloroquine, sulfasalazine (sulphasalazopyrine), gold salts, D-penicillamine, leflunomide, azathioprine, anakinra, infliximab (REMICADETM), etanercept, TNF. alpha, blockers, a biological agent that targets an inflammatory cytokine, and Nonsteroidal Anti-Inflammatory Drug (NSAIDs).
  • methotrexate cyclophosphamide
  • cyclosporine cyclosporin A
  • chloroquine hydroxychloroquine
  • sulfasalazine sulphasalazopyrine
  • gold salts gold salts
  • D-penicillamine leflunomide
  • azathioprine anakinr
  • NSAIDs include, but are not limited to acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors and tramadol.
  • compositions, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
  • method refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • treating includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical symptoms of a condition.
  • Human amelogenin cDNA was amplified by PCR from a recombinant plasmid containing human amelogenin X cDNA (GeneBank accession no. M86932), representing the most abundant amelogenin mRNA transcript, which lacks exon 4 and codes for a 175 amino acid protein.
  • the human cDNA was subcloned into the pFastBacTM HTb vector (Invitrogen). This system adds a hexa-histidine tag to the amino terminus of the expressed protein, enabling effective one-step purification by Ni 2+ -NTA affinity chromatography.
  • rHAM + The recombinant protein was expressed in Spodoptera frugiperda (Sf9) eukaryotic insect cells and the yield of purified human amelogenin (rHAM + ) was up to 10 mg/L culture. rHAM + was characterized by SDS-PAGE, Western blot, ESI- TOF spectrometry, peptide mapping and MS/MS sequencing (Taylor et al. 2006).
  • CD105 + - hMSCs CD105 + - hBMSC from human femoral bone marrow were isolated using ficoll gradient, immunomagnetic methodology (MACS) and culturing. Analysis of mononuclear cells and fresh CD105 + cells, obtained from the same source was performed by flow cytometry (FACS) and in vitro differentiation assays (chondrogenic, osteogenic, and adipogenic differentiation).
  • Luciferase, Beta- galactosidase, FLAG, and GFP amelogenin adenovirus were produced using the Gateway cloning system (Invitrogen), under the CMV promoter.
  • HEK-293 cells were infected with each of the different recombinant adenoviruses (for propagation), followed by transduction into the CD105 + -hMSCs.
  • the engineered hMSCs were serially diluted to achieve 50 % effective titer, for determination of viral titer (using FACS to determine the ratio of GFP-positive/negative cells, blue count for beta-gal positive/negative cells, ⁇ -gal with a nuclear localization signal, anti-adenovirus antibodies, or by plaque forming unit as described in Taylor et al. 2006.
  • the two knees (treated & untreated) from experimental rats and control rats were dissected out and prepared for mechanical testing, histological, cross polarization microscopy, immunohistochemical, confocal microscopy, in-situ hybridization and electron microscopy studies.
  • OCD osteochondreal defect
  • the bone- ligament-bone unit was gently isolated under dissecting-microscope to assure that the MCL is the only connection between the femur and tibia.
  • the specimen was wrapped in cotton gauze soaked in normal saline solution and stored at -20 °C before testing, or was tested immediately.
  • the bone-ligament-bone unit was fastened in a clamping device which was attached to an electrohydraulic-materials testing machine at room temperature in normal saline solution.
  • Force-displacement curves were recorded and analyzed. Load to failure (N) (peak of the curve) was measured and was compared to the normal non-transected ligament of the same animal.
  • the slope of the force- displacement curve is a measure of the ligament stiffness; a higher slope represents stiffer ligament and a lower slope represents a more lax ligament.
  • Tissue preparation for histology, cross polarization microscopy, immunohistochemical analysis and confocal microscopy For histology and immunohistochemistry the entire ligament tissue was cut from the bone and fixed in 4% Para-Formaldehyde (PFA) for 24 hours at 4 °C. The region of the regenerating tissues was studied. More details are described in Haze et al. 2007; Haze et al. 2009.
  • Determination of the degree of regeneration and the characterization of the regenerated tissues and ectopically formed tissues with time was carried out using micro-CT, histology, immunohistochemistry, in-situ hybridization, quantitative- PCR, confocal microscopy, Western blot, and various biochemical assays (e.g. alkaline phosphatase etc.), and molecular biology techniques aimed at identification of phenotypes of the regenerated tissues.
  • the spatio-temporal distribution of the engineered CD105 + -hMSCs in the regenerating tissues and neighboring tissues was monitored by the detection of the various fused amelogenin proteins. Markers for the specific cells such as bone, PDL, cementum, fat, muscle were tested.
  • Indirect Immunohistochemistry Performed using the Zymed laboratories inc. kit protocol. Controls comprise of pre -immune sera or PBS. Various amelogenin antibodies (monoclonal and polyclonal) and antibodies against other proteins (e.g. specific mesenchymal cell markers (such as CD 105, CD73, CD90, STRO-1, osteocalcin, BMP2, BSP, etc.) were used.
  • amelogenin antibodies monoclonal and polyclonal
  • antibodies against other proteins e.g. specific mesenchymal cell markers (such as CD 105, CD73, CD90, STRO-1, osteocalcin, BMP2, BSP, etc.) were used.
  • RNA-probes antisense and sense
  • ISH was performed using DIG RNA labeling kit and InnoGenex universal ISH protocol.
  • Regenerating tissues were dissected, RNA samples isolated, and RT is performed using Cells-to-cDNA Kit. PCR reactions were performed using specific amelogenin primers and primers for various cell markers.
  • Quantitative PCR To compare specific patterns of gene expression at different stages of regeneration/ectopic tissue formation, quantitative PCR was employed, using Taqman probes and 7300 Real time PCR system (Applied Biosystems).
  • Dil and DiO two different fluorescent colors was preformed during the operation prior to application of rHAM + or MSCs engineered to overexpress amelogenin.
  • the purpose of adding Dil/DiO was to mark the exact location of the defect.
  • the other fluorescent label was used to mark migration of MSCs from distant locations or to colocalize amelogenin or other osteogenic/chondrogenic etc. markers. Colocalization was preformed using confocal microscopy.
  • the IVIS kinetic (Caliper) was used for in-vivo tracking of cell recruitment to the defect site, induced by recombinant human amelogenin (rHAM + ).
  • osteochondreal defect OCD
  • morphometric analysis demonstrated that the size of the defect was reduced significantly in the groups treated with various concentrations of recombinant human amelogenin (rHAM + ) dissolved in PGA carrier (experimental), as compared to the size of the defect in the group treated with PGA carrier only (control; Figures 6A-D, 7A-D).
  • the size of the osteochondreal defect (OCD) in the femoral trochlea of the control group (treated with PGA carrier only) was increased and degenerative changes were seen in large areas of the articular surface, while in the experimental knee the changes were much more limited and the OCD was filled with tissue that we are now testing its histological structure and composition.
  • Figures 9A-F illustrate the results of a histological analysis (Hematoxylin & Eosin) comparing the normal cartilage structure in the lateral femoral condyle (LFC, A and D) to the tissue filling the OCD in the medial femoral condyle of the experimental knee (1 ⁇ /1 ⁇ 1 of rHAM + dissolved in PGA, B and E) and control (PGA carrier, C and F), 12 weeks after creation of OCD.
  • PGA carrier application of PGA carrier only

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention porte sur un procédé de traitement d'une lésion au niveau d'un ligament d'articulation squelettique ou d'une maladie d'un ligament d'articulation squelettique. Le procédé consiste à mettre en contact le ligament ou tendon d'articulation squelettique du sujet avec une quantité thérapeutiquement efficace d'amélogénine, l'amélogénine n'étant pas comprise dans un échafaudage, permettant ainsi de traiter la lésion au niveau du ligament ou tendon d'articulation squelettique ou la maladie du ligament ou tendon d'articulation squelettique.
PCT/IL2012/050163 2011-05-09 2012-05-09 Régénération et réparation de tissu mésenchymateux par utilisation d'amélogénine WO2012153333A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/116,796 US20140073765A1 (en) 2011-05-09 2012-05-09 Regeneration and repair of mesenchymal tissue using amelogenin
US14/471,262 US9957314B2 (en) 2011-05-09 2014-08-28 Regeneration and repair of mesenchymal tissue using amelogenin
US15/899,621 US11034753B2 (en) 2011-05-09 2018-02-20 Regeneration and repair of mesenchymal tissue using amelogenin

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161483909P 2011-05-09 2011-05-09
US61/483,909 2011-05-09

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/116,796 A-371-Of-International US20140073765A1 (en) 2011-05-09 2012-05-09 Regeneration and repair of mesenchymal tissue using amelogenin
US14/471,262 Continuation-In-Part US9957314B2 (en) 2011-05-09 2014-08-28 Regeneration and repair of mesenchymal tissue using amelogenin

Publications (1)

Publication Number Publication Date
WO2012153333A1 true WO2012153333A1 (fr) 2012-11-15

Family

ID=46262119

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2012/050163 WO2012153333A1 (fr) 2011-05-09 2012-05-09 Régénération et réparation de tissu mésenchymateux par utilisation d'amélogénine

Country Status (1)

Country Link
WO (1) WO2012153333A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017056092A1 (fr) 2015-09-30 2017-04-06 Hadasit Medical Research Services And Development Ltd. Utilisation de l'amélogénine pleine longueur pour favoriser la croissance ou la régénération nerveuse
US9957314B2 (en) 2011-05-09 2018-05-01 Hadasit Medical Research Services And Development Ltd. Regeneration and repair of mesenchymal tissue using amelogenin

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3839153A (en) 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
EP0053197A1 (fr) 1980-11-29 1982-06-09 Georg Fischer Aktiengesellschaft Procédé de désoxydation d'un métal en fusion
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
EP0337967A2 (fr) 1988-03-17 1989-10-18 Bioventures N.V. Compositions induisant la liaison entre des parties de tissu vivant minéralisé
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US5464764A (en) 1989-08-22 1995-11-07 University Of Utah Research Foundation Positive-negative selection methods and vectors
US5932447A (en) 1994-05-17 1999-08-03 Bristol-Myers Squibb Company Cloning and expression of a gene encoding bryodin 1 from Bryonia dioica
WO2000006734A1 (fr) 1998-07-29 2000-02-10 Northwestern University Molecule induisant la chondrogenese et l'osteogenese
US20030077291A1 (en) 2001-09-14 2003-04-24 Stina Gestrelius Matrix protein compositions for modulating immune response
EP1862143A1 (fr) 2006-05-18 2007-12-05 Marcello Marchesi Procédé pour la régénération guidée de tissus périodontiques ou osseux et dispositif correspondant
US20110003745A1 (en) 2006-02-20 2011-01-06 Straumann Holding Ag Granulate-matrix
WO2011030185A1 (fr) 2009-09-12 2011-03-17 Inanc Buelend Échafaudages fibro-inducteurs et angiogènes de guidage de cellules pour le modelage de tissu parodontal

Patent Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3839153A (en) 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
EP0053197A1 (fr) 1980-11-29 1982-06-09 Georg Fischer Aktiengesellschaft Procédé de désoxydation d'un métal en fusion
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683202B1 (fr) 1985-03-28 1990-11-27 Cetus Corp
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
EP0337967A2 (fr) 1988-03-17 1989-10-18 Bioventures N.V. Compositions induisant la liaison entre des parties de tissu vivant minéralisé
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5464764A (en) 1989-08-22 1995-11-07 University Of Utah Research Foundation Positive-negative selection methods and vectors
US5487992A (en) 1989-08-22 1996-01-30 University Of Utah Research Foundation Cells and non-human organisms containing predetermined genomic modifications and positive-negative selection methods and vectors for making same
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US5932447A (en) 1994-05-17 1999-08-03 Bristol-Myers Squibb Company Cloning and expression of a gene encoding bryodin 1 from Bryonia dioica
WO2000006734A1 (fr) 1998-07-29 2000-02-10 Northwestern University Molecule induisant la chondrogenese et l'osteogenese
US20030077291A1 (en) 2001-09-14 2003-04-24 Stina Gestrelius Matrix protein compositions for modulating immune response
US20100093632A1 (en) 2001-09-14 2010-04-15 Institut Straumann Ag Enamel matrix protein composition for treatment of systemic inflammatory response
US20110003745A1 (en) 2006-02-20 2011-01-06 Straumann Holding Ag Granulate-matrix
EP1862143A1 (fr) 2006-05-18 2007-12-05 Marcello Marchesi Procédé pour la régénération guidée de tissus périodontiques ou osseux et dispositif correspondant
WO2011030185A1 (fr) 2009-09-12 2011-03-17 Inanc Buelend Échafaudages fibro-inducteurs et angiogènes de guidage de cellules pour le modelage de tissu parodontal

Non-Patent Citations (79)

* Cited by examiner, † Cited by third party
Title
"Enhancers and Eukaryotic Expression", 1983, COLD SPRING HARBOR PRESS
"Immobilized Cells and Enzymes", 1986, IRL PRESS
"Methods in Enzymology", vol. 1-317, ACADEMIC PRESS
"PCR Protocols: A Guide To Methods And Applications", 1990, ACADEMIC PRESS
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
"Vectors: A Survey of Molecular Cloning Vectors and Their Uses", 1988, BUTTERWORTHS
ASLAN H; ZILBERMAN Y; KANDEL L; LIEBERGALL M; OSKOUIAN R.J; GAZIT D; GAZIT Z: "Osteogenic Differentiation of Noncultured Immunoisolated Bone Marrow-Derived CD105+ Cells", STEM CELLS, vol. 24, no. 7, 2006, pages 1728 - 1737, XP009073405
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1989, JOHN WILEY AND SONS
AUSUBEL, R. M.,: "Current Protocols in Molecular Biology", vol. I-III, 1994
BHOSALE AM; RICHARDSON JB: "Articular cartilage: structure, injuries and review of management", BRITISH MEDICAL BULLETIN, vol. 87, 2008, pages 77 - 95, XP055262494, DOI: doi:10.1093/bmb/ldn025
BI Y; EHIRCHIOU D; KILTS TM; INKSON CA; EMBREE MC; SONOYAMA W; LI L; LEET AI; SEO BM; ZHANG L: "Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche", NAT MED, vol. 13, 2007, pages 1219 - 1227, XP002496557, DOI: doi:10.1038/nm1630
BIRREN ET AL.: "Genome Analysis: A Laboratory Manual Series", vol. 1-4, 1998, COLD SPRING HARBOR LABORATORY PRESS
BOOTH ET AL., IMMUNOL. LETT., vol. 19, 1988, pages 65 - 70
BRINSTER ET AL., NATURE, vol. 296, 1982, pages 39 - 42
BRISSON ET AL., NATURE, vol. 310, 1984, pages 511 - 514
BROGLI ET AL., SCIENCE, vol. 224, 1984, pages 838 - 843
CANAPLE L. ET AL.: "Improving cell encapsulation through size control", J BIOMATER SCI POLYM ED., vol. 13, 2002, pages 783 - 96
CELLIS, J. E.,: "Cell Biology: A Laboratory Handbook", vol. I-III, 1994
CELLIS, J. E.,: "Current Protocols in Immunology", vol. I-III, 1994
CHANG ET AL.: "Somatic Gene Therapy", 1995, CRC PRESS
CHANG TM; PRAKASH S: "Procedures for microencapsulation of enzymes, cells and genetically engineered microorganisms", MOL BIOTECHNOL., vol. 17, 2001, pages 249 - 60
CHIA, S.M. ET AL.: "Multi-layered microcapsules for cell encapsulation", BIOMATERIALS, vol. 23, 2002, pages 849 - 56, XP004322709, DOI: doi:10.1016/S0142-9612(01)00191-0
CORUZZI ET AL., EMBO J., vol. 3, 1984, pages 1671 - 1680
DESAI, T.A.: "Microfabrication technology for pancreatic cell encapsulation", EXPERT OPIN BIOL THER., vol. 2, 2002, pages 633 - 46
DEUTSCH D; HAZE-FILDERMAN A; BLUMENFELD A; DAFNI L; LEISER Y; SHAY B; GRUENBAUM-COHEN Y; ROSENFELD E; FERMON E; ZIMMERMANN B: "Amelogenin, a major structural protein in mineralizing enamel, is also expressed in soft tissues: brain and cells of the hematopoietic system", EUR J ORAL SCI., vol. 114, 2006, pages 183 - 9
FINGL ET AL.: "The Pharmacological Basis of Therapeutics", 1975, pages: 1
FRESHNEY, R. I.,: "Animal Cell Culture", 1986
FRESHNEY: "Culture of Animal Cells - A Manual of Basic Technique", 1994, WILEY-LISS
GAIT, M. J.,: "Oligonucleotide Synthesis", 1984
GARDELLA ET AL., J. BIOL. CHEM., vol. 265, 1990, pages 15854 - 15859
GILBOA, BIOTECHNIQUES, vol. 4, no. 6, 1986, pages 504 - 512
GOSSEN; BUJARD, PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 5547 - 551
GRUENBAUM-COHEN Y; TUCKER AS; HAZE A; SHILO D; TAYLOR AL; SHAY B; SHARPE PT; MITSIADIS TA; ORNOY A; BLUMENFELD A: "Amelogenin in cranio-facial development: the tooth as a model to study the role of amelogenin during embryogenesis", J EXP ZOOLOG B MOL DEV EVOL., vol. 15, 2008, pages 445 - 57
GURLEY ET AL., MOL. CELL. BIOL., vol. 6, 1986, pages 559 - 565
HAMES, B. D., AND HIGGINS S. J.,: "Nucleic Acid Hybridization", 1985
HAMES, B. D., AND HIGGINS S. J.,: "Transcription and Translation", 1984
HAZE A; TAYLOR AL; BLUMENFELD A; ROSENFELD E; LEISER Y; DAFNI L; SHAY B; GRUENBAUM-COHEN Y; FERMON E; HAEGEWALD S: "Amelogenin expression in long bone and cartilage cells and in bone marrow progenitor cells", ANAT REC (HOBOKEN, vol. 290, 2007, pages 455 - 60
HAZE A; TAYLOR AL; HAEGEWALD S; LEISER Y; SHAY B; ROSENFELD E; GRUENBAUM-COHEN Y; DAFNI L; ZIMMERMAN B; HEIKINHEIMO K: "Regeneration of bone and periodontal ligament induced by Recombinant amelogenin after periodontitis", J CELL MOL MED., vol. 13, 2009, pages 1110 - 24, XP002683976, DOI: doi:10.1111/j.1582-4934.2009.00700.x
HAZE AMIR ET AL: "Regeneration of bone and periodontal ligament induced by recombinant amelogenin after periodontitis", JOURNAL OF CELLULAR AND MOLECULAR MEDICINE, vol. 13, no. 6, June 2009 (2009-06-01), pages 1110 - 1124, XP002683976, ISSN: 1582-1838 *
HOFFMANN A; GROSS G: "Tendon and ligament engineering in the adult organism: mesenchymal stem cells and gene-therapeutic approaches", INT ORTHOP, vol. 31, 2007, pages 791 - 797, XP019561109, DOI: doi:10.1007/s00264-007-0395-9
HYNES ET AL., PROC. NATL. ACAD. SCI. USA, vol. 78, 1981, pages 2038 - 2042
ISRAEL; KAUFMAN, NUCL. ACIDS RES., vol. 17, 1989, pages 2589 - 2604
KASSIS ET AL., BONE MARROW TRANSPLANT, vol. 37, no. 10, May 2006 (2006-05-01), pages 967 - 76
KAWAGUCHI H; HIRACHI A; HASEGAWA N; IWATA T; HAMAGUCHI H; SHIBA H; TAKATA T; KATO Y; KURIHARA H: "Enhancement of periodontal tissue regeneration by transplantation of bone marrow mesenchymal stem cells", JPERIODONTOL., vol. 75, 2004, pages 1281 - 1287
KAWASAKY K; SUGIHARA S; NISHIDA K; OZAKI T; YOSHIDA A; OHTSUKA A; INOUE A: "Hoechst 33342 is a useful cell tracer for long-term investigation of articular cartilage repair", ARCH HISTOL CYTOL, vol. 67, 2004, pages 13 - 19
KERN ET AL., STEM CELLS, vol. 24, 2006, pages 1294 - 1301
KLOCK ET AL., NATURE, vol. 329, 1987, pages 734 - 736
LEE ET AL., NATURE, vol. 294, 1981, pages 228 - 232
LU MZ ET AL.: "A novel cell encapsulation method using photosensitive poly(allylamine alpha- cyanocinnamylideneacetate", J MICROENCAPSUL., vol. 17, 2000, pages 245 - 51
LU MZ ET AL.: "Cell encapsulation with alginate and alpha- phenoxycinnamylidene-acetylated poly(allylamine", BIOTECHNOL BIOENG., vol. 70, 2000, pages 479 - 83, XP008025769, DOI: doi:10.1002/1097-0290(20001205)70:5<479::AID-BIT1>3.0.CO;2-E
MANIATIS ET AL.: "Molecular Cloning: A Laboratory Manual", 1982, COLD SPRING HARBOR LABORATORY
MARSHAK ET AL.: "Strategies for Protein Purification and Characterization - A Laboratory Course Manual", 1996, CSHL PRESS
MAYO ET AL., CELL, vol. 29, 1982, pages 99 - 108
MISHELL AND SHIIGI: "Selected Methods in Cellular Immunology", 1980, W. H. FREEMAN AND CO.
MOUTSATSOS IK; TURGEMAN G; ZHOU S; KURKALLI BG; PELLED G; TZUR L; KELLEY P; STUMM N; MI S; MULLER R: "Exogenously regulated stem cell- mediated gene therapy for bone regeneration", MOL THER., vol. 3, 2001, pages 449 - 461, XP003018665, DOI: doi:10.1006/mthe.2001.0291
NO D ET AL., PROC NATL ACAD SCI USA., vol. 93, 1996, pages 3346 - 3351
NOUER, L.,: "Heat Shock Response", 1991, CRC, pages: 167 - 220
PERBAL, B.: "A Practical Guide to Molecular Cloning", 1984
PERBAL: "A Practical Guide to Molecular Cloning", 1988, JOHN WILEY & SONS
ROBINSON C; BROOKES SJ; SHORE RC; KIRKHAM J: "The developing enamel matrix: nature andfunction", EUR J ORAL SCI, vol. 106, no. 1, 1998, pages 282 - 291, XP009075109
SAMBANIS, A.: "Encapsulated islets in diabetes treatment", DIABETES TECHNOL. THER., vol. 5, 2003, pages 665 - 8
SAMBROOK ET AL.: "Molecular Cloning: A laboratory Manual", 1989
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRINGS HARBOR LABORATORY
SEARLE ET AL., MOL. CELL. BIOL., vol. 5, 1985, pages 1480 - 1489
SHAPIRO JL; WEN X; OKAMOTO CT; WANG HJ; LYNGSTADAAS SP; GOLDBERG M; SNEAD ML; PAINE ML: "Cellular uptake of amelogenin, and its localization to CD63, and Lanipl-positive vesicles", CELL MOL LIFE SCI., vol. 64, 2007, pages 244 - 56, XP019471740
STITES ET AL.: "Basic and Clinical Immunology(8th Edition)", 1994, APPLETON & LANGE
STUDIER ET AL., METHODS IN ENZYMOL., vol. 185, 1990, pages 60 - 89
TAKAMATSU ET AL., EMBO J., vol. 6, 1987, pages 307 - 311
TAYLOR AL; HAZE-FILDERMAN A; BLUMENFELD A; SHAY B; DAFNI L; ROSENFELD E; LEISER Y; FERMON E; GRUENBAUM-COHEN Y; DEUTSCH D: "High yield of biologically active recombinant human amelogenin using the baculovirus expression system", PROTEIN EXPR PURIF, vol. 45, 2006, pages 43 - 53, XP024908591, DOI: doi:10.1016/j.pep.2005.05.010
TAYLOR ET AL., PROTEIN EXPRESSION AND PURIFICATION, vol. 45, 2006, pages 43 - 53
TEI K; MATSUMOTO T; MIFUNE Y; ISHIDA K; SASAKI K; SHOJI T; KUBO S; KAWAMOTO A; ASAHARA T; KUROSAKA M: "Administrations of peripheral blood CD34- positive cells contribute to medial collateral ligament healing via vasculogenesis", STEM CELLS, vol. 26, 2008, pages 819 - 830
ULUDAG, H. ET AL.: "Technology of mammalian cell encapsulation", ADV DRUG DELIV REV., vol. 42, 2000, pages 29 - 64, XP002302229, DOI: doi:10.1016/S0169-409X(00)00053-3
VEGA ET AL.: "Gene Targeting", 1995, CRC PRESS
WATSON ET AL.: "Recombinant DNA", SCIENTIFIC AMERICAN BOOKS
WEISSBACH; WEISSBACH: "Methods for Plant Molecular Biology", 1988, ACADEMIC PRESS, pages: 421 - 463
WILLIAMS D: "Small is beautiful: microparticle and nanoparticle technology in medical devices", MED DEVICE TECHNOL., vol. 10, 1999, pages 6 - 9
YU-CHING HUANG ET AL: "Effects of human full-length amelogenin on the proliferation of human mesenchymal stem cells derived from bone marrow", CELL AND TISSUE RESEARCH, SPRINGER, BERLIN, DE, vol. 342, no. 2, 22 October 2010 (2010-10-22), pages 205 - 212, XP019860977, ISSN: 1432-0878, DOI: 10.1007/S00441-010-1064-7 *
ZHANG ET AL., CHINESE MEDICAL JOURNAL, vol. 117, no. 6, 2004, pages 882 - 887
ZOU Y; WANG H; SHAPIRO JL; OKAMOTO CT; BROOKES SJ; LYNGSTADAAS SP; SNEAD ML; PAINE ML: "Determination of protein regions responsible for interactions of amelogenin with CD63 and LAMP i.", BIOCHEM J., vol. 408, 2007, pages 347 - 54, XP008092160, DOI: doi:10.1042/BJ20070881

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9957314B2 (en) 2011-05-09 2018-05-01 Hadasit Medical Research Services And Development Ltd. Regeneration and repair of mesenchymal tissue using amelogenin
US11034753B2 (en) 2011-05-09 2021-06-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Regeneration and repair of mesenchymal tissue using amelogenin
WO2017056092A1 (fr) 2015-09-30 2017-04-06 Hadasit Medical Research Services And Development Ltd. Utilisation de l'amélogénine pleine longueur pour favoriser la croissance ou la régénération nerveuse
US10478476B2 (en) 2015-09-30 2019-11-19 Hadasit Medical Research Services And Development Ltd. Use of full-length amelogenin for promoting nerve growth or regeneration

Similar Documents

Publication Publication Date Title
Cucchiarini et al. Gene therapy for cartilage defects
ES2654428T3 (es) Aislamiento de células multipotenciales adultas por fosfatasa alcalina no específica de tejido
RU2709780C2 (ru) Адгезивные клетки жировой ткани или плаценты и их использование в лечебных целях
Su et al. Enhancement of periodontal tissue regeneration by transplantation of osteoprotegerin-engineered periodontal ligament stem cells
US8545888B2 (en) Tendon stem cells
US20080292599A1 (en) Chondrogenic Compositions and Methods of Use
JP2010539123A (ja) 変性疾患の処置のための神経内分泌因子
Kofron et al. Orthopaedic applications of gene therapy
US20140073765A1 (en) Regeneration and repair of mesenchymal tissue using amelogenin
KR20210040908A (ko) 인간 유도 만능 줄기세포로부터 연골세포의 펠렛을 제조하는 방법 및 이의 용도
US7160725B2 (en) Hedgehog signaling promotes the formation of three dimensional cartilage matrices
WO2012153333A1 (fr) Régénération et réparation de tissu mésenchymateux par utilisation d&#39;amélogénine
Komur et al. Can a biodegradable implanted bilayered drug delivery system loaded with BMP-2/BMP-12 take an effective role in the biological repair process of bone–tendon injuries? A preliminary report
Zhu et al. Effects of Sox9 gene therapy on the healing of bone-tendon junction: An experimental study
US9981010B2 (en) Methods and compositions for bone formation
US11034753B2 (en) Regeneration and repair of mesenchymal tissue using amelogenin
EP3355911B1 (fr) Utilisation de l&#39;amélogénine pleine longueur pour favoriser la croissance ou la régénération nerveuse
WO2005007098A2 (fr) Traitement a l&#39;acide nucleique pour stimulation de la reparation des cartilages
AU2020201937A1 (en) Isolation of adult multipotential cells by tissue non-specific alkaline phosphatase
Komur et al. Research Article Can a Biodegradable Implanted Bilayered Drug Delivery System Loaded with BMP-2/BMP-12 Take an Effective Role in the Biological Repair Process of Bone–Tendon Injuries? A Preliminary Report
Hung Toward a novel tissue engineering method for repairing critically sized craniofacial bone defects
Bush Gene-induced chondrogenesis of mesenchymal stem cells through viral gene delivery
Franceschi Gene Therapy Approaches for Musculoskeletal Tissue Regeneration

Legal Events

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

Ref document number: 12727171

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14116796

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 12727171

Country of ref document: EP

Kind code of ref document: A1