WO2006067920A1 - Pharmaceutical for bone marrow stem cell transplantation therapy - Google Patents

Pharmaceutical for bone marrow stem cell transplantation therapy Download PDF

Info

Publication number
WO2006067920A1
WO2006067920A1 PCT/JP2005/020697 JP2005020697W WO2006067920A1 WO 2006067920 A1 WO2006067920 A1 WO 2006067920A1 JP 2005020697 W JP2005020697 W JP 2005020697W WO 2006067920 A1 WO2006067920 A1 WO 2006067920A1
Authority
WO
WIPO (PCT)
Prior art keywords
bone marrow
marrow stem
ctack
cells
stem cells
Prior art date
Application number
PCT/JP2005/020697
Other languages
French (fr)
Japanese (ja)
Inventor
Hiroshi Shimizu
Riichiro Abe
Daisuke Inokuma
Original Assignee
National University Corporation Hokkaido University
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 National University Corporation Hokkaido University filed Critical National University Corporation Hokkaido University
Priority to JP2006548722A priority Critical patent/JPWO2006067920A1/en
Publication of WO2006067920A1 publication Critical patent/WO2006067920A1/en

Links

Classifications

    • 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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/195Chemokines, e.g. RANTES
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • the present invention opens to the use of chemokines in a new treatment method by skin cell regeneration for congenital skin diseases that have been reported as intractable diseases, such as congenital epidermolysis bullosa.
  • EB congenital epidermolysis bullosa
  • EB congenital epidermolysis bullosa
  • EB is congenitally fragile and is understood as a collective term for congenital diseases that easily cause blistering on the skin, even when weak external force causes dissociation of the epidermal dermis (see, for example, Toshiaki Saida et al.
  • EB is a genetically diverse disease, and blister formation is based on the force generated at any site under electron microscope observation.
  • Epidermis cell blister formation simple type
  • blister formation junction type
  • dermal blister formation nutrient disorder type directly below
  • Hiroshi Shimizu “Classification of epidermolysis bullosa”, previous training course required A course materials, Japan Dermatological Association resident member Issued by the association, 2002 'Non-patent document 2).
  • keratin 5 or keratin 14 gene in the junction type in laminin 5 gene, in the nutritional disorder type VII collagen gene, mutation or injury is considered to be the cause of the onset
  • Non-Patent Document 1 edited by Toshiaki Saida et al., “Today's skin disease treatment policy”, third edition, published by Medical School, pages 308-309.
  • Non-patent document 2 Hiroshi Shimizu, “Classification of epidermolysis bullosa”, previous training course required A course material, published by the Japan Dermatological Association Members Committee, 2002.
  • Non-Patent Document 3 Daisuke Sawamura et al., “Possibility of gene therapy for epidermolysis bullosa”, 2002 West Japan Dermatology, separate volume, Vol. 64, No. 3, pp. 277-280.
  • the present invention particularly relates to a method for effectively treating or ameliorating a hereditary disease without effective pharmaceutical treatment, such as congenital epidermolysis bullosa, by a method other than gene therapy, It provides a useful medicine.
  • CTACK T-cell attracting chemo kine
  • BMDK bone marrow derived-keratinocytes
  • the present invention relates to a regenerative therapeutic drug using bone marrow stem cells containing CTACK as an active ingredient, and particularly to a regenerative therapeutic drug using transplanted bone marrow stem cells.
  • the present invention relates to a pharmaceutical comprising CTACK as an active ingredient that differentiates transplanted bone marrow stem cells into epidermal cells.
  • Regenerative treatment using bone marrow stem cells is expected as a treatment for various diseases such as angiogenesis therapy, heart failure, and liver regeneration, and the bone marrow transplant promotion foundation (http: ⁇ www.jmdp.or. jp / index.html) establishes so-called bone marrow bank to promote effective use of bone marrow stem cells is doing. Attempts have also been made to treat bone diseases by regenerating the bone marrow stem cell skin for skin diseases. However, CTACK's chemotolatato effect on bone marrow stem cells and the differentiation induction effect on epidermal cells and its bone marrow There is no report on its use for stem cell transplantation therapy.
  • the present invention uses CTACK in order to improve the efficiency in skin regenerative medicine as a treatment for skin diseases.
  • CTACK in the present invention is a protein having a total of 112 amino acid residues described in International Patent Publication WO98Z23750, or a force in which some of the amino acid residues are different from those described in the same publication. It means a protein that retains chemotratat activity against stem cells and BM DK-inducing activity.
  • CTACK functions have been known to include the selective attraction of CLA + memory T cells, skin dendritic cells (Langelnos cells), and their effects on their precursors. It has been reported that a certain healing effect is expected for skin disorders involving the immune system, such as psoriasis, skin cancer, inflammation, allergy, dermatitis, wound healing, infection, etc. Publication WO98Z23750, Janine Morales, et al., Proc. Natl. Ac ad. Sci. No. 157-165, 2002).
  • CTACK binds to CCR10, which is a chemokine receptor, and chemostratates cells such as CLA + cells, T cells, rod cells or rod precursors, and these are dermis of the skin. It has been reported that by moving into the layer and into the Z or epidermal layer, it shows therapeutic effects on skin diseases associated with immune system cells. However, it has been reported that CTA CK has the function of chemostratating bone marrow stem cells different from immune system cells and further inducing differentiation into epidermal cells! Wow! /.
  • the present invention uses CTACK's differentiation-inducing function for bone marrow stem cells, so that the use of CTACK can be applied to the treatment of skin diseases related to immune cells that have been proposed in the past, and to the application frame. It expands to the treatment of new skin diseases located outside.
  • the present invention can induce differentiation of bone marrow stem cells into epidermal cells in the field of skin damage, and is a congenital skin disease that has been considered to be particularly difficult to cure, such as congenital skin diseases. It is possible to provide a new treatment method and a pharmaceutical for the treatment of sexual epidermolysis bullosa.
  • FIG. 1 shows chemotract activity of CTACK against bone marrow stem cells by in vitro test.
  • FIG. 2 shows the activity of inducing differentiation of CTACK into bone marrow stem cells into epidermal cells in an in vivo test.
  • FIG. 3 shows the differentiation-inducing activity of CTACK to epidermal cells against bone marrow stem cells in mice pretreated with G-CSF.
  • FIG. 4 shows C TACK differentiation-inducing activity into epidermal cells against bone marrow stem cells in mice with an increased number of CD34-positive bone marrow stem cells in peripheral blood.
  • CTACK used in the present invention can be prepared recombinantly in accordance with the description of International Patent Publication No. WO98Z23750 or the literature of Morales et al.
  • CTACK which is commercially available from R & D System of Minneapolis, USA or other institutions, may also be used.
  • a bone marrow stem cell comprising an amino acid sequence in which one or more amino acids are substituted, deleted, and Z or added.
  • Polypeptides having chemoretorato activity against BMDK and activity to induce BMDK can also be used in the present invention.
  • the mutation is highly conserved, such as glycine (Gly) and proline (Pro), Gly and alanine ( Ala) or valine (Val), leucine (Leu) and isoleucine (lie), glutamic acid (Glu) and glutamine (Gin), aspartic acid (Asp) and asparagine (Asn), cysteine (Cys) and threonine (Thr),
  • Gly proline
  • Ala valine
  • Val leucine
  • Glu glutamine
  • Gin glutamic acid
  • Asp aspartic acid
  • Asparagine Asparagine
  • cysteine cysteine
  • Thr threonine
  • Examples include substitution between Thr and serine (Ser) or Ala, lysine (Lys) and arginine (Arg).
  • a gene encoding a powerful CTACK or a variant thereof can also be used for recombinant production of CTACK and the like.
  • a vector holding a DNA encoding CTACK, a host to be transformed with the vector, and the like may be used by selecting a combination described in the preceding literature or other general-purpose combinations.
  • various viruses such as baculovirus, retrovirus, vaccinia virus and the like can also be used.
  • Expression may be performed under the control of a promoter sequence specific to the CTACK gene, or another appropriate expression promoter may be used by connecting or replacing it with a promoter sequence specific to the CTACK gene.
  • the promoter used in this case may be appropriately selected according to the host and the purpose of expression.
  • the 1S host such as T7 promoter, lac promoter, trp promoter, ⁇ PL promoter is yeast.
  • PH05 promoter, GAP promoter, ADH promoter and the like can be exemplified by SV40-derived promoter, retrovirus promoter and the like when the host is an animal cell, but of course, it is not limited thereto.
  • CTACK is administered to patients who have undergone bone marrow stem cell transplantation in the form of a solution in which CTACK is dissolved in an appropriate buffer, for example, physiological saline, an emulsion in which an appropriate emulsifier is added, and the like.
  • an appropriate buffer for example, physiological saline, an emulsion in which an appropriate emulsifier is added, and the like.
  • Either a solution or a sheet form such as a nonwoven fabric holding emulsion can be used. These can be used by directly applying to or pasting the affected area.
  • Bone marrow cells were isolated from the bone marrow of mice (C57BL6) according to the method of Wiley et al. (Current Protocols of Immunology, ISS N 0-471-52276-7). Then expressed on the cell surface Antibody staining using an antibody specific for CD34 antigen (PharMingen) was performed, and CD34 positive bone marrow stem cells were separated using FACSVantage (BD Bioscience).
  • CD34 positive bone marrow stem cells suspended in DMEM medium in the upper insert of a 3 ⁇ m pore insert (Kurashiki Boseki Co., Ltd.) dissolved in the same medium in the lower plate 0 to 500 ngZml CTACK (Manufactured by R & D System) or stromal cell derived factor-1 (stroma cell derived factor-1, SDF-1) was added and incubated at 37 ° C. After 4 hours, the number of CD34-positive bone marrow stem cells that had migrated to the lower plate was counted with a hemocytometer.
  • transgenic mouse (GFP mouse, distributed by Jackson Laboratories, USA) prepared using a recombinant gene obtained by placing the GFP protein gene under the control of the ⁇ -actin promoter, the same procedure as in Example 1 was performed. Bone marrow stem cells were isolated.
  • the chimeric mice were divided into 4 groups, and immediately after the creation of a skin defect wound with a diameter of 6 mm on the back of each chimeric mouse, 1 group was CTACK (R & D System, IngZ phosphate buffered saline 30 1) SDF—l (lngZ phosphate buffered saline 30 1) in group 2, secondary lymphoid tissue chemokine (IngZ phosphate buffered saline 30 / zl) in group 3, In group 4, only the same buffer was administered locally around the wound, and the animals were raised under standard conditions until the wound was completely healed (about 28 days).
  • CTACK R & D System, IngZ phosphate buffered saline 30 1
  • SDF—l lngZ phosphate buffered saline 30 1
  • secondary lymphoid tissue chemokine IngZ phosphate buffered saline 30 / zl
  • the wound is excised, embedded in OCTcompound, and a 5 ⁇ l thick tissue piece is prepared.
  • Antibodies and fluorescent dyes against keratin 14, CTACK, and other chemokines (FITC, RI TC) Each antibody reaction was diluted 100-fold with a secondary antibody bound to Fluorescent tissue staining was performed at room temperature for 1 hour.
  • the stained tissue pieces were observed using a confocal laser microscope (Laser Scanning Confocal Imaging System MRC 1024, manufactured by Bio-Rad), and GFP positive and keratin 14 positive cells were detected. The percentage of total epidermal cells was measured as the epidermal cells.
  • Example 1 Prior to CTACK administration, Example 1 except that 150 g / kg / day of G-CSF was administered for 3 consecutive days to increase the number of CD34-positive bone marrow stem cells in peripheral blood (approximately 100 times). In the same manner as above, the ratio of epidermal cells differentiated from bone marrow stem cells in the total epidermal cells was measured (Fig. 3).
  • CD34 positive cells were separated from bone marrow cells of GFP mice using MACS (magnetic activated cell sorting, Miltenyl Biotec), and these cells (5 x 10 5 cells) were separated from normal mice.
  • Tail force was also injected intravenously to increase the number of CD34-positive bone marrow stem cells in peripheral blood, increasing the number of epidermal cells differentiated from bone marrow stem cells in the total epidermal cells. Measured ( Figure 4).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dermatology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Materials For Medical Uses (AREA)

Abstract

A pharmaceutical comprising as an active ingredient CTACK (T-cell attracting chemokine) that is effective in the regenerative therapy utilizing bone marrow stem cells, especially the regenerative therapy of skin diseases utilizing transplanted bone marrow stem cells. In particular, the pharmaceutical provides a novel therapeutic method for congenital skin diseases whose fundamental treatment has been regarded as being difficult, for example, congenital epidermolysis bullosa.

Description

明 細 書  Specification
骨髄幹細胞移植治療用医薬  Drugs for bone marrow stem cell transplantation
技術分野  Technical field
[0001] 本発明は、これまで難病として報告されてきた先天性皮膚疾患、例えば先天性表 皮水疱症に対する、皮膚細胞再生による新たな治療法における、ケモカインの利用 に開する。  [0001] The present invention opens to the use of chemokines in a new treatment method by skin cell regeneration for congenital skin diseases that have been reported as intractable diseases, such as congenital epidermolysis bullosa.
背景技術  Background art
[0002] 先天性すなわち遺伝性の疾患は、その殆どが、正常な生体の維持に必要とされる 蛋白質あるいは遺伝子に、その本来の機能が損なわれるような変異あるいは欠失が 存在することで発症する疾患である。そのため、一般に、外科的治療あるいは薬物等 の投与による薬学的治療によって根本的に治癒することは難しいとされている。  [0002] Most congenital or inherited diseases are caused by the presence of mutations or deletions in proteins or genes that are required for the maintenance of normal living organisms that impair their original functions. Disease. Therefore, it is generally considered difficult to cure fundamentally by surgical treatment or pharmaceutical treatment by administration of drugs or the like.
[0003] 皮膚組織におけるこの様な先天性疾患の一つに、先天性表皮水疱症(epidermoysi s bullosa、以下 EBとする)がある。 EBは、先天的に皮膚が脆弱であり、弱い外力で も表皮真皮の結合が解離し、皮膚に容易に水疱をきたす先天性疾患の総称として理 解されている (例えば斎田俊明ら編集、「今日の皮膚疾患治療方針」、第 3版、医学 書院発行、第 308〜309頁,非特許文献 1)。  [0003] One of such congenital diseases in skin tissue is congenital epidermolysis bullosa (hereinafter referred to as EB). EB is congenitally fragile and is understood as a collective term for congenital diseases that easily cause blistering on the skin, even when weak external force causes dissociation of the epidermal dermis (see, for example, Toshiaki Saida et al. Today's skin disease treatment policy ", 3rd edition, published by medical school, pages 308-309, non-patent literature 1).
[0004] EBは遺伝子性の非常に多様な疾患であり、水疱形成が電子顕微鏡観察下でどの 部位に生ずる力を基に、表皮細胞内水疱形成 (単純型)、水疱形成 (接合部型)およ び直下の真皮内水疱形成 (栄養障害型)に大別される (清水 宏、「表皮水疱症の分 類」、前実績研修講習会必須 Aコース用資料、日本皮膚科学会研修医委員会発行、 2002年'非特許文献 2)。単純型ではケラチン 5あるいはケラチン 14遺伝子に、接合 部型ではラミニン 5遺伝子に、栄養障害型では VII型コラーゲン遺伝子に、それぞれ 変異あるいは傷害が生じて 、ることが発症原因であると考えられて 、る。  [0004] EB is a genetically diverse disease, and blister formation is based on the force generated at any site under electron microscope observation. Epidermis cell blister formation (simple type), blister formation (junction type) And dermal blister formation (nutrient disorder type) directly below (Hiroshi Shimizu, “Classification of epidermolysis bullosa”, previous training course required A course materials, Japan Dermatological Association resident member Issued by the association, 2002 'Non-patent document 2). In the simple type, keratin 5 or keratin 14 gene, in the junction type in laminin 5 gene, in the nutritional disorder type VII collagen gene, mutation or injury is considered to be the cause of the onset, The
[0005] この EBと 、う遺伝性疾患に対して、これまでには有効な根治的治療法はなぐ対症 的な外用療法として抗生物質あるいは抗炎症剤を与える程度の療法しかないのが現 状である。そのため、有効な治療法としては、異常が確認された遺伝子の機能を復 活させる、いわゆる遺伝子治療の開発が持たれている(澤村大輔ら、「表皮水疱症に 対する遺伝子治療の可能性」、 2002年、西日本皮膚科別冊、第 64卷、第 3号、第 2 77〜280頁'非特許文献 3)。 [0005] For EB and hereditary diseases, there are currently only therapies that give antibiotics or anti-inflammatory agents as symptomatic external treatments that are not effective radical treatments so far. It is. For this reason, effective treatments include the development of so-called gene therapy that restores the function of genes that have been identified as abnormal (Daisuke Sawamura et al., “ "Possibility of gene therapy against", 2002, West Japan Dermatology, separate volume, Vol. 64, No. 3, pp. 277-280 (Non-patent Document 3).
[0006] 非特許文献 1 :斎田俊明ら編集、「今日の皮膚疾患治療方針」、第 3版、医学書院発 行、第 308〜309頁。 [0006] Non-Patent Document 1: edited by Toshiaki Saida et al., “Today's skin disease treatment policy”, third edition, published by Medical School, pages 308-309.
非特許文献 2 :清水 宏、「表皮水疱症の分類」、前実績研修講習会必須 Aコース用 資料、 日本皮膚科学会研修医委員会発行、 2002年。  Non-patent document 2: Hiroshi Shimizu, “Classification of epidermolysis bullosa”, previous training course required A course material, published by the Japan Dermatological Association Residents Committee, 2002.
非特許文献 3 :澤村大輔ら、「表皮水疱症に対する遺伝子治療の可能性」、 2002年、 西日本皮膚科別冊、第 64卷、第 3号、第 277〜280頁。  Non-Patent Document 3: Daisuke Sawamura et al., “Possibility of gene therapy for epidermolysis bullosa”, 2002 West Japan Dermatology, separate volume, Vol. 64, No. 3, pp. 277-280.
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0007] し力しながら、遺伝子治療は日常的に行!、得る治療法であるとは言 、難ぐより汎 用性の高!、、新 、EB治療法の開発が望まれて 、る。 [0007] However, gene therapy is routinely performed, and although it is a treatment that can be obtained, it is difficult to be more versatile! New development of EB therapy is desired. .
[0008] 本発明は、特に、有効な薬学的治療法のない遺伝性疾患、例えば先天性表皮水 疱症を、遺伝子治療以外の方法によって有効に治療あるいは症状を改善させる方法 と、該方法に有用な医薬を提供するものである。 [0008] The present invention particularly relates to a method for effectively treating or ameliorating a hereditary disease without effective pharmaceutical treatment, such as congenital epidermolysis bullosa, by a method other than gene therapy, It provides a useful medicine.
課題を解決するための手段  Means for solving the problem
[0009] 本発明者らは、皮膚で発現するケモカインである CTACK(T-cell attracting chemo kine)が、これを投与された皮膚組織に骨髄幹細胞を誘引(ケモアトラタト)し、最終的 に表皮細胞(bone marrow derived- keratinocytes、 BMDK)へと分化させる機能を有 していることを見出し、これを骨髄幹細胞の移植と組み合わせることで、皮膚疾患、例 えば EBの治療が可能となり得ること等を見出し、本発明を完成した。  [0009] The present inventors have found that CTACK (T-cell attracting chemo kine), which is a chemokine expressed in the skin, attracts bone marrow stem cells to the skin tissue to which this is administered (chemoatratat), and finally epidermal cells ( bone marrow derived-keratinocytes (BMDK), and it has been found that combining this with bone marrow stem cell transplantation can treat skin diseases such as EB, The present invention has been completed.
[0010] すなわち本発明は、 CTACKを有効成分とする骨髄幹細胞を利用した再生治療用 医薬、特に移植された骨髄幹細胞を利用した再生治療用医薬に関する。特に、移植 された骨髄幹細胞を表皮細胞に分化させる CTACKを有効成分とする医薬に関する  That is, the present invention relates to a regenerative therapeutic drug using bone marrow stem cells containing CTACK as an active ingredient, and particularly to a regenerative therapeutic drug using transplanted bone marrow stem cells. In particular, the present invention relates to a pharmaceutical comprising CTACK as an active ingredient that differentiates transplanted bone marrow stem cells into epidermal cells.
[0011] 骨髄幹細胞を用いた再生治療は、血管新生療法、心不全、肝臓再生など、様々な 疾患の治療法として期待されており、財団法人骨髄移植推進財団 (http:〃 www.jmdp .or.jp/index.html)がいわゆる骨髄バンクを設立して、骨髄幹細胞の有効利用を推進 している。皮膚疾患に対しても、骨髄幹細胞力 皮膚を再生させることで治療を行お うとする試みがなされているが、 CTACKの骨髄幹細胞に対するケモアトラタト作用な らびに表皮細胞への分化誘導作用、ならびにその骨髄幹細胞移植治療への利用に ついては報告がない。 [0011] Regenerative treatment using bone marrow stem cells is expected as a treatment for various diseases such as angiogenesis therapy, heart failure, and liver regeneration, and the bone marrow transplant promotion foundation (http: 〃 www.jmdp.or. jp / index.html) establishes so-called bone marrow bank to promote effective use of bone marrow stem cells is doing. Attempts have also been made to treat bone diseases by regenerating the bone marrow stem cell skin for skin diseases. However, CTACK's chemotolatato effect on bone marrow stem cells and the differentiation induction effect on epidermal cells and its bone marrow There is no report on its use for stem cell transplantation therapy.
[0012] 本発明は、皮膚疾患治療としての皮膚再生医療においてその効率を向上させるた めに CTACKを利用するものである。  [0012] The present invention uses CTACK in order to improve the efficiency in skin regenerative medicine as a treatment for skin diseases.
[0013] 本発明における CTACKは、国際特許公開公報 WO98Z23750号に記載された 全 112アミノ酸残基力 なる蛋白質、あるいはそのアミノ酸残基の一部が同公報記載 のものとは異なっている力 なお骨髄幹細胞に対するケモアトラタト活性ならびに BM DKへの分ィ匕誘導活性を保持している蛋白質を意味する。  [0013] CTACK in the present invention is a protein having a total of 112 amino acid residues described in International Patent Publication WO98Z23750, or a force in which some of the amino acid residues are different from those described in the same publication. It means a protein that retains chemotratat activity against stem cells and BM DK-inducing activity.
[0014] これまでに、 CTACKの機能としては、 CLA+記憶 T細胞の選択的な誘引活性、 皮膚榭状細胞 (ランゲルノヽンス細胞)およびこれらの前駆体への作用などが知られて おり、特に免疫系が関与する皮膚障害、例えば乾癬、皮膚癌、炎症、アレルギー、皮 膚炎、創傷治癒、感染などに対して、一定の治癒効果が期待されることが報告されて いる(前記国際特許公開公報 WO98Z23750号、 Janine Moralesら、 Proc. Natl. Ac ad. Sci. U. S.A.ゝ第 96卷、第 25号、第 14470〜14475頁、 1999年、 Bernhard Ho meyら、 Nature Medicine,第 8卷、第 2号、第 157〜165頁、 2002年)。  [0014] To date, CTACK functions have been known to include the selective attraction of CLA + memory T cells, skin dendritic cells (Langelnos cells), and their effects on their precursors. It has been reported that a certain healing effect is expected for skin disorders involving the immune system, such as psoriasis, skin cancer, inflammation, allergy, dermatitis, wound healing, infection, etc. Publication WO98Z23750, Janine Morales, et al., Proc. Natl. Ac ad. Sci. No. 157-165, 2002).
[0015] これらの報告によれば、 CTACKは、ケモカインレセプターである CCR10に結合し 、 CLA+細胞、 T細胞、榭状細胞または榭状細胞前駆体等の細胞をケモアトラタトし て、これらを皮膚の真皮層内および Zまたは表皮層内に移動させることで、免疫系細 胞に関連した皮膚疾患の治療効果を示すことが報告されている。し力しながら、 CTA CKが、免疫系細胞とは異なる骨髄幹細胞をケモアトラタトし、さらにこれを表皮細胞 へと分化誘導する機能を有して 、ることにつ 、ては、何ら報告されて!、な!/、。  [0015] According to these reports, CTACK binds to CCR10, which is a chemokine receptor, and chemostratates cells such as CLA + cells, T cells, rod cells or rod precursors, and these are dermis of the skin. It has been reported that by moving into the layer and into the Z or epidermal layer, it shows therapeutic effects on skin diseases associated with immune system cells. However, it has been reported that CTA CK has the function of chemostratating bone marrow stem cells different from immune system cells and further inducing differentiation into epidermal cells! Wow! /.
[0016] 本発明は、 CTACKの骨髄幹細胞に対する分化誘導機能を活用することで、 CTA CKについての用途を、従来提唱されていた免疫系細胞が関与する皮膚疾患の治 療と 、う適用枠の外に位置する新たな皮膚疾患の治療へと、拡大するものである。  [0016] The present invention uses CTACK's differentiation-inducing function for bone marrow stem cells, so that the use of CTACK can be applied to the treatment of skin diseases related to immune cells that have been proposed in the past, and to the application frame. It expands to the treatment of new skin diseases located outside.
[0017] 本発明は、骨髄幹細胞の表皮細胞への分化誘導を、皮膚損傷の場において行うこ とができ、特に根本的治癒が困難であるとされてきた先天性皮膚疾患、例えば先天 性表皮水疱症に対して、新たな治療法ならびに該治療のための医薬を提供すること ができる。 [0017] The present invention can induce differentiation of bone marrow stem cells into epidermal cells in the field of skin damage, and is a congenital skin disease that has been considered to be particularly difficult to cure, such as congenital skin diseases. It is possible to provide a new treatment method and a pharmaceutical for the treatment of sexual epidermolysis bullosa.
図面の簡単な説明  Brief Description of Drawings
[0018] [図 1]図 1は、インビトロ試験による、 CTACKの骨髄幹細胞に対するケモアトラクト活 性を示す。  [0018] FIG. 1 shows chemotract activity of CTACK against bone marrow stem cells by in vitro test.
[図 2]図 2は、インビボ試験による、 CTACKの骨髄幹細胞に対する表皮細胞への分 化誘導活性を示す。  [FIG. 2] FIG. 2 shows the activity of inducing differentiation of CTACK into bone marrow stem cells into epidermal cells in an in vivo test.
[図 3]図 3は、 G— CSFで前処理したマウスにおける、 CTACKの骨髄幹細胞に対す る表皮細胞への分化誘導活性を示す。  [FIG. 3] FIG. 3 shows the differentiation-inducing activity of CTACK to epidermal cells against bone marrow stem cells in mice pretreated with G-CSF.
[図 4]図 4は、末梢血中の CD34陽性骨髄幹細胞数を増加させたマウスにおける、 C TACKの骨髄幹細胞に対する表皮細胞への分化誘導活性を示す。  FIG. 4 shows C TACK differentiation-inducing activity into epidermal cells against bone marrow stem cells in mice with an increased number of CD34-positive bone marrow stem cells in peripheral blood.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0019] 本発明で使用する CTACKは、国際特許公開公報 WO98Z23750号あるいは前 記 Moralesらの文献の記載に従って、組換え的に調製することができる。  [0019] CTACK used in the present invention can be prepared recombinantly in accordance with the description of International Patent Publication No. WO98Z23750 or the literature of Morales et al.
[0020] 例えば、国際特許公開公報 WO98Z23750号に記載された CTACKをコードす る遺伝子の塩基配列の全部または一部力もなるプローブを用いて、同公報の記載に 従ってクロー-ングすればよい。また、 CTACKの全長をコードする DNAを DNA合 成機などを用いて合成し、これを用いて組換え的に調製することができる。また、米 国ミネアポリスの R&D System社あるいはその他の機関から市販されて!、る CTACK を利用しても良い。  [0020] For example, using a probe having the whole or part of the base sequence of the gene encoding CTACK described in International Patent Publication No. WO98Z23750, cloning may be performed according to the description in the publication. Also, DNA encoding the full length of CTACK can be synthesized using a DNA synthesizer or the like, and can be prepared recombinantly using this. CTACK, which is commercially available from R & D System of Minneapolis, USA or other institutions, may also be used.
[0021] また、同国際公報等に具体的に記載されたアミノ酸配列あるいは DNA配列以外で あっても、例えば 1以上のアミノ酸が置換、欠失、および Z若しくは付加したアミノ酸 配列からなる、骨髄幹細胞に対するケモアトラタト活性ならびに BMDKへの分ィ匕誘 導活性を有するポリペプチドも、本発明において利用することができる。  [0021] Further, even if the amino acid sequence or DNA sequence specifically described in the international publication or the like is used, for example, a bone marrow stem cell comprising an amino acid sequence in which one or more amino acids are substituted, deleted, and Z or added. Polypeptides having chemoretorato activity against BMDK and activity to induce BMDK can also be used in the present invention.
[0022] 例えば、アミノ酸残基の電荷、大きさ、疎水性等の物理ィ匕学的性質にっ 、て保存 性の高 、変異、例えばグリシン(Gly)とプロリン (Pro)、 Glyとァラニン (Ala)またはバ リン (Val)、ロイシン(Leu)とイソロイシン(lie)、グルタミン酸(Glu)とグルタミン(Gin) 、ァスパラギン酸 (Asp)とァスパラギン (Asn)、システィン(Cys)とスレオニン (Thr)、 Thrとセリン (Ser)または Ala、リジン(Lys)とアルギニン (Arg)との間の、それぞれの置 換等が挙げられる。 [0022] For example, due to the physical properties such as the charge, size, and hydrophobicity of amino acid residues, the mutation is highly conserved, such as glycine (Gly) and proline (Pro), Gly and alanine ( Ala) or valine (Val), leucine (Leu) and isoleucine (lie), glutamic acid (Glu) and glutamine (Gin), aspartic acid (Asp) and asparagine (Asn), cysteine (Cys) and threonine (Thr), Examples include substitution between Thr and serine (Ser) or Ala, lysine (Lys) and arginine (Arg).
[0023] 同様に、力かる CTACKあるいはその変異体をコードする遺伝子も、 CTACK等の 組み換え的生産に利用することができる。  [0023] Similarly, a gene encoding a powerful CTACK or a variant thereof can also be used for recombinant production of CTACK and the like.
[0024] また CTACKをコードする DNAを保持するベクター、該ベクターを用いて形質転換 させる宿主等も、先行する文献に記載の組み合わせあるいはその他の汎用の組み 合わせを選択して使用すればよい。また、プラスミドの他に、バキュロウィルス、レトロ ウィルス、ワクシニアウィルス等の種々のウィルスを用いることも可能である。  [0024] In addition, a vector holding a DNA encoding CTACK, a host to be transformed with the vector, and the like may be used by selecting a combination described in the preceding literature or other general-purpose combinations. In addition to plasmids, various viruses such as baculovirus, retrovirus, vaccinia virus and the like can also be used.
[0025] 発現は、 CTACK遺伝子固有のプロモーター配列の制御下に発現させてもよぐあ るいは、別の適当な発現プロモーターを CTACK遺伝子固有のプロモーター配列に 接続あるいは置き換えて使用することもできる。この場合に使用するプロモーターは、 宿主及び発現の目的に応じて適宜選択すればよぐ例えば宿主が大腸菌である場 合には T7プロモーター、 lacプロモーター、 trpプロモーター、 λ PLプロモーターなど 1S 宿主が酵母である場合には PH05プロモーター、 GAPプロモーター、 ADHプロ モーター等が、宿主が動物細胞である場合には SV40由来プロモーター、レトロウイ ルスプロモーター等を例示できるが、当然ながらこれらには限定されな 、。  [0025] Expression may be performed under the control of a promoter sequence specific to the CTACK gene, or another appropriate expression promoter may be used by connecting or replacing it with a promoter sequence specific to the CTACK gene. The promoter used in this case may be appropriately selected according to the host and the purpose of expression. For example, when the host is E. coli, the 1S host such as T7 promoter, lac promoter, trp promoter, λ PL promoter is yeast. In some cases, PH05 promoter, GAP promoter, ADH promoter and the like can be exemplified by SV40-derived promoter, retrovirus promoter and the like when the host is an animal cell, but of course, it is not limited thereto.
[0026] 骨髄幹細胞移植を受けた患者への CTACKの投与形態としては、 CTACKを適当 な緩衝液、例えば生理食塩水等に溶解した溶液の形態、適当な乳化剤を添加した ェマルジヨンの形態、これらの溶液あるいはェマルジヨンを保持した不織布などのシ ートの形態など、いずれも採ることができる。これらは患部に直接塗布あるいは貼付し て使用することができる。  [0026] CTACK is administered to patients who have undergone bone marrow stem cell transplantation in the form of a solution in which CTACK is dissolved in an appropriate buffer, for example, physiological saline, an emulsion in which an appropriate emulsifier is added, and the like. Either a solution or a sheet form such as a nonwoven fabric holding emulsion can be used. These can be used by directly applying to or pasting the affected area.
[0027] 以下、実施例により本発明を具体的に説明するが、本発明はこれら実施例により何 ら限定されるものではない。  Hereinafter, the present invention will be specifically described with reference to examples, but the present invention is not limited to these examples.
実施例 1  Example 1
[0028] CTACKの骨髄幹細胞に対するケモアトラタト活性を、インビトロ試験により確認し た。  [0028] The chemotolatato activity of CTACK on bone marrow stem cells was confirmed by an in vitro test.
[0029] マウス(C57BL6)の骨髄から Wileyらの方法(Current Protocols of Immunology, ISS N 0-471-52276-7)に従って骨髄細胞を分離した。次いで、細胞表面に発現している CD34抗原に特異的な抗体を用いた抗体染色(PharMingen社製)を行 、、 CD34陽 性骨髄幹細胞を FACSVantage (BD Bioscience社製)を用いて分離した。 [0029] Bone marrow cells were isolated from the bone marrow of mice (C57BL6) according to the method of Wiley et al. (Current Protocols of Immunology, ISS N 0-471-52276-7). Then expressed on the cell surface Antibody staining using an antibody specific for CD34 antigen (PharMingen) was performed, and CD34 positive bone marrow stem cells were separated using FACSVantage (BD Bioscience).
[0030] 孔径 3 μ mのインサート (倉敷紡績社製)の上部インサートに DMEM培地に懸濁し た CD34陽性骨髄幹細胞 1 X 105個を、下部プレートに同培地に溶解した 0〜500n gZmlの CTACK(R&D System社製)またはストロマ細胞由来因子 1 (stroma cell d erived factor- 1、 SDF— 1)をそれぞれ加え、 37°Cでインキュベートした。 4時間後、 下部プレートに遊走した CD34陽性骨髄幹細胞の個数をへモサイトメーターで計測 した。 [0030] 1 to 10 5 CD34 positive bone marrow stem cells suspended in DMEM medium in the upper insert of a 3 μm pore insert (Kurashiki Boseki Co., Ltd.) dissolved in the same medium in the lower plate 0 to 500 ngZml CTACK (Manufactured by R & D System) or stromal cell derived factor-1 (stroma cell derived factor-1, SDF-1) was added and incubated at 37 ° C. After 4 hours, the number of CD34-positive bone marrow stem cells that had migrated to the lower plate was counted with a hemocytometer.
[0031] この結果、 lOngZml以上の濃度において、 CTACKは濃度依存的に CD34陽性 骨髄幹細胞をケモアトラタトした(図 1)。  [0031] As a result, CTACK chemostratated CD34-positive bone marrow stem cells in a concentration-dependent manner at concentrations of lOngZml or higher (Fig. 1).
実施例 2  Example 2
[0032] CTACKの皮膚損傷部位における骨髄幹細胞の誘導分化活性を、インビボ試験に より確認した。  [0032] The induced differentiation activity of bone marrow stem cells at the site of CTACK skin injury was confirmed by in vivo tests.
[0033] βァクチンプロモーターの制御下に GFP蛋白質遺伝子を置 、た組換え遺伝子を用 いて作成されたトランスジエニックマウス(GFPマウス、米国 Jackson Laboratoriesより分 与)から、実施例 1と同様にして骨髄幹細胞を分離した。  [0033] From a transgenic mouse (GFP mouse, distributed by Jackson Laboratories, USA) prepared using a recombinant gene obtained by placing the GFP protein gene under the control of the β-actin promoter, the same procedure as in Example 1 was performed. Bone marrow stem cells were isolated.
[0034] この細胞を、 850cradの放射線処理によって骨髄細胞を破壊したマウス野生株(C5 7BI/6)の骨髄内に注射した後、 4週間、標準的な条件下で飼育して、骨髄由来細胞 のみが GFPの蛍光を発する、 GFPキメラマウスを作成した。  [0034] After these cells were injected into the bone marrow of a wild-type mouse strain (C5 7BI / 6) whose bone marrow cells had been destroyed by 850crad radiation treatment, the cells were raised under standard conditions for 4 weeks. Only GFP chimeric mice that emit GFP fluorescence were created.
[0035] キメラマウスを 4群に分け、それぞれのキメラマウスの背部に直径 6mmの皮膚欠損 創を作成した直後に、 1群には CTACK (R&D System社製、 IngZリン酸緩衝食塩 水 30 1)を、 2群には SDF— l (lngZリン酸緩衝食塩水 30 1)を、 3群には 2次リン パ球組織ケモカイン(secondary lymphoid tissue chemokine、 IngZリン酸緩衝食塩 水 30 /z l)を、 4群には同緩衝液のみをそれぞれ創部周辺に局所投与し、さらに創傷 が完全に治癒するまで (約 28日)標準的な条件下で飼育した。  [0035] The chimeric mice were divided into 4 groups, and immediately after the creation of a skin defect wound with a diameter of 6 mm on the back of each chimeric mouse, 1 group was CTACK (R & D System, IngZ phosphate buffered saline 30 1) SDF—l (lngZ phosphate buffered saline 30 1) in group 2, secondary lymphoid tissue chemokine (IngZ phosphate buffered saline 30 / zl) in group 3, In group 4, only the same buffer was administered locally around the wound, and the animals were raised under standard conditions until the wound was completely healed (about 28 days).
[0036] 飼育後、創部を切除し、 OCTcompoundに包埋し、厚さ 5 μ 1の組織片を作成し、ケラ チン 14、 CTACK,ならびに他のケモカインに対する抗体および蛍光色素(FITC、 RI TC)が結合した 2次抗体を用いて、いずれの抗体反応も濃度 100倍希釈、作用時間 1時間、室温の条件下で行って蛍光組織染色を行った。 [0036] After breeding, the wound is excised, embedded in OCTcompound, and a 5 μl thick tissue piece is prepared. Antibodies and fluorescent dyes against keratin 14, CTACK, and other chemokines (FITC, RI TC) Each antibody reaction was diluted 100-fold with a secondary antibody bound to Fluorescent tissue staining was performed at room temperature for 1 hour.
[0037] 染色した組織片を、共焦点レーザー顕微鏡 (Laser Scanning Confocal Imaging Syst em MRC 1024、 Bio- Rad社製)を用いて観察し、 GFP陽性かつケラチン 14陽性である 細胞を骨髄幹細胞力 分ィ匕した表皮細胞として、全表皮細胞中の割合を計測した。 [0037] The stained tissue pieces were observed using a confocal laser microscope (Laser Scanning Confocal Imaging System MRC 1024, manufactured by Bio-Rad), and GFP positive and keratin 14 positive cells were detected. The percentage of total epidermal cells was measured as the epidermal cells.
[0038] その結果、 CTACK投与を行った群において、有意に骨髄幹細胞由来の表皮細 胞の割合が増カロした(図 2)。 [0038] As a result, the proportion of bone marrow stem cell-derived epidermal cells significantly increased in the group receiving CTACK (Fig. 2).
実施例 3  Example 3
[0039] CTACK投与前に、 150 g/kg/日の G— CSFを 3日間連続投与して、末梢血 中の CD34陽性骨髄幹細胞数を増加させた (約 100倍)以外は、実施例 1と同様にし て行!、、骨髄幹細胞から分化した表皮細胞の全表皮細胞中の割合を計測した(図 3 [0039] Prior to CTACK administration, Example 1 except that 150 g / kg / day of G-CSF was administered for 3 consecutive days to increase the number of CD34-positive bone marrow stem cells in peripheral blood (approximately 100 times). In the same manner as above, the ratio of epidermal cells differentiated from bone marrow stem cells in the total epidermal cells was measured (Fig. 3).
) o ) o
[0040] また、 GFPマウスの骨髄細胞内から、 MACS法 (magnetic activated cell sorting, Mil tenyl Biotec社)を用いて CD34陽性細胞を分離し、この細胞(5 X 105個)を正常マウ スの尾部力も静脈内注射して、末梢血中の CD34陽性骨髄幹細胞数を増カロさせた 以外は、実施例 1と同様にして行い、骨髄幹細胞から分化した表皮細胞の全表皮細 胞中の割合を計測した (図 4)。 [0040] In addition, CD34 positive cells were separated from bone marrow cells of GFP mice using MACS (magnetic activated cell sorting, Miltenyl Biotec), and these cells (5 x 10 5 cells) were separated from normal mice. Tail force was also injected intravenously to increase the number of CD34-positive bone marrow stem cells in peripheral blood, increasing the number of epidermal cells differentiated from bone marrow stem cells in the total epidermal cells. Measured (Figure 4).
[0041] その結果、実施例 1の場合よりも、より多い数の骨髄幹細胞由来表皮細胞を確認す ることがでさた。  As a result, it was possible to confirm a larger number of bone marrow stem cell-derived epidermal cells than in Example 1.

Claims

請求の範囲 The scope of the claims
[1] CTACKを有効成分とする、骨髄幹細胞を利用した再生治療用医薬。  [1] A regenerative treatment drug using bone marrow stem cells containing CTACK as an active ingredient.
[2] 移植された骨髄幹細胞を利用した再生治療である、請求項 1に記載の医薬。 [2] The medicament according to claim 1, which is a regenerative treatment using transplanted bone marrow stem cells.
[3] 再生治療が皮膚再生治療である、請求項 1又は 2に記載の医薬。 [3] The medicament according to claim 1 or 2, wherein the regenerative treatment is a skin regenerative treatment.
[4] 移植された骨髄幹細胞を表皮細胞に分化させて皮膚再生を行う治療である、請求項 3に記載の医薬。 [4] The medicament according to claim 3, which is a treatment for regenerating skin by differentiating transplanted bone marrow stem cells into epidermal cells.
PCT/JP2005/020697 2004-12-21 2005-11-11 Pharmaceutical for bone marrow stem cell transplantation therapy WO2006067920A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2006548722A JPWO2006067920A1 (en) 2004-12-21 2005-11-11 Drugs for bone marrow stem cell transplantation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2004-369272 2004-12-21
JP2004369272 2004-12-21

Publications (1)

Publication Number Publication Date
WO2006067920A1 true WO2006067920A1 (en) 2006-06-29

Family

ID=36601525

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2005/020697 WO2006067920A1 (en) 2004-12-21 2005-11-11 Pharmaceutical for bone marrow stem cell transplantation therapy

Country Status (2)

Country Link
JP (1) JPWO2006067920A1 (en)
WO (1) WO2006067920A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020160024A1 (en) * 1998-12-24 2002-10-31 Schering Corpration, A New Jersey Corporation Chemokine and receptor uses; compositions; methods
WO2003024404A2 (en) * 2001-09-20 2003-03-27 Schering Corporation Chemokines as adjuvants of immune response

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020160024A1 (en) * 1998-12-24 2002-10-31 Schering Corpration, A New Jersey Corporation Chemokine and receptor uses; compositions; methods
WO2003024404A2 (en) * 2001-09-20 2003-03-27 Schering Corporation Chemokines as adjuvants of immune response

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
HOMEY B ET AL: "CCL27-CCR10 interactions regulate T cell-mediated skin inflammation.", NATURE MEDICINE., vol. 8, no. 2, 2002, pages 157 - 165, XP001097778 *
JUN FENG JI ET AL: "Interactions of Chemokines and Chemokine Receptors Mediated the Migration of Mesenchymal Stem Cells to the Impaired Site in the Brain After Hypoglossal Nerve Injury.", STEM CELLS., vol. 22, May 2004 (2004-05-01), pages 415 - 427, XP009055260 *
MORALES J ET AL: "CTACK, a skin-associated chemokine that preferentially attracts skin-homing memory T cells.", PNAS., vol. 96, no. 25, 1999, pages 14470 - 14475, XP002137995 *
REISS Y ET AL: "CC Chemokine Receptor (CCR)4 and the CCR10 Ligand Cutaneous T Cell-attracting Chemokine (CTACK) in Lymphocyte Trafficking to Inflamed Skin.", J EXP MED., vol. 194, no. 10, 2001, pages 1541 - 1547, XP002995901 *
VON LUTTICHAU I ET AL: "Human Adult CD34() Progenitor Cells Functionally Express the Chemokine Receptors CCR1, CCR4, CCR7, CXCR5, and CCR10 but Not CXCR4.", STEM CELLS AND DEVELOPMENT., vol. 14, 2005, pages 329 - 336, XP002995300 *

Also Published As

Publication number Publication date
JPWO2006067920A1 (en) 2008-06-12

Similar Documents

Publication Publication Date Title
AU2019210647B2 (en) Peptide for inducing regeneration of tissue and use thereof
Armour et al. Cellular and molecular pathology of HTS: basis for treatment
Vorotnikova et al. Extracellular matrix-derived products modulate endothelial and progenitor cell migration and proliferation in vitro and stimulate regenerative healing in vivo
Kohara et al. Angiogenesis induced by controlled release of neuropeptide substance P
JP5676253B2 (en) Highly efficient collection of functional cells in vivo
Bloise et al. Engineering immunomodulatory biomaterials for regenerating the infarcted myocardium
CN104768567A (en) Combination treatments and compositions for wound healing
KR20190128622A (en) Perinatal Tissue-derived Mesenchymal Stem Cells: Methods and Their Uses
Hwang et al. VEGF-encoding, gene-activated collagen-based matrices promote blood vessel formation and improved wound repair
JP2019524753A (en) Use of IL-12 as a replacement immunotherapeutic
Zhang et al. Bone marrow stromal cells transplantation promotes the resolution and recanalization of deep vein thrombosis in rabbits through regulating macrophage infiltration and angiogenesis
WO2006067920A1 (en) Pharmaceutical for bone marrow stem cell transplantation therapy
JP5777886B2 (en) Peptide composition and use thereof
He et al. Mechanical stretch preconditioned adipose‐derived stem cells elicit polarization of anti‐inflammatory M2‐like macrophages and improve chronic wound healing
Rai et al. In-silico Functional Prediction of Proteomic Data Obtained from Perinatal Human Amniotic Membrane Used as Tissue Regeneration Transplant
TW200925165A (en) A composition to enhance wound healing
Sadtler et al. Integrating tissue microenvironment with scaffold design to promote immune-mediated regeneration
Wang et al. The role of fibrocytes in post-burn hypertrophic scarring
Armour et al. Cellular and molecular pathology of HTS: basis for treatment [corrected][published erratum appears in WOUND REPAIR REGENERATION 2008 Jul-Aug; 16 (4): 582].
JP2004269423A (en) Hgf(hepatocyte growth factor) derivative

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006548722

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05805913

Country of ref document: EP

Kind code of ref document: A1