JP2021505172A5 - - Google Patents

Download PDF

Info

Publication number
JP2021505172A5
JP2021505172A5 JP2020531468A JP2020531468A JP2021505172A5 JP 2021505172 A5 JP2021505172 A5 JP 2021505172A5 JP 2020531468 A JP2020531468 A JP 2020531468A JP 2020531468 A JP2020531468 A JP 2020531468A JP 2021505172 A5 JP2021505172 A5 JP 2021505172A5
Authority
JP
Japan
Prior art keywords
donor
disease
cells
population
hematopoietic stem
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
JP2020531468A
Other languages
Japanese (ja)
Other versions
JP2021505172A (en
Filing date
Publication date
Priority claimed from US15/834,017 external-priority patent/US10058573B1/en
Application filed filed Critical
Priority claimed from PCT/US2018/064335 external-priority patent/WO2019113375A2/en
Publication of JP2021505172A publication Critical patent/JP2021505172A/en
Publication of JP2021505172A5 publication Critical patent/JP2021505172A5/ja
Priority to JP2023188499A priority Critical patent/JP2024023226A/en
Pending legal-status Critical Current

Links

Claims (15)

哺乳動物のドナーの骨髄から末梢血中に造血幹細胞の集団を動員させる方法であって、(i) Gro-β、Gro-β T、及びそれらのバリアントからなる群より選択されるCXCR2アゴニスト、並びに(ii)CXCR4アンタゴニスト、を前記ドナーに投与することを含む方法。 A method of mobilizing a population of hematopoietic stem cells from the bone marrow of a mammalian donor into peripheral blood, (i) a CXCR2 agonist selected from the group consisting of Gro-β, Gro-β T, and variants thereof, and (ii) A method comprising administering a CXCR4 antagonist, to said donor. 請求項1に記載の方法、ここで:(a) 前記CXCR2アゴニストは:(i) Gro-β Tである;(ii) 約100 μg/kg〜約250 μg/kgの用量で前記ドナーに投与する;(iii) 約125 μg/kg〜約225 μg/kgの用量で前記ドナーに投与する;(iv) 約150 μg/kgの用量で前記ドナーに投与する;及び/若しくは(v) 前記ドナーに静脈内投与する;並びに/又は、(b) 前記CXCR4アンタゴニストは:(i) 前記ドナーに皮下投与する;(ii) プレリキサホル又はその薬学的に許容可能な塩である、任意選択的にここで、前記プレリキサホル又はその薬学的に許容可能な塩を、約50 μg/kg〜約500 μg/kgの用量で、例えば、約200 μg/kg〜約300 μg/kgの用量で、例えば、約240 μg/kgの用量で前記ドナーに投与する。 The method of claim 1, wherein : (a) the CXCR2 agonist is : (i) Gro-β T ; (ii) administered to the donor at a dose of about 100 μg / kg to about 250 μg / kg. (Iii) administer to the donor at a dose of about 125 μg / kg to about 225 μg / kg ; (iv) administer to the donor at a dose of about 150 μg / kg ; and / or (v) the donor. And / or (b) the CXCR4 antagonist : (i) subcutaneously administered to the donor ; (ii) prelixaform or a pharmaceutically acceptable salt thereof , optionally here. , The prelyxaform or a pharmaceutically acceptable salt thereof, at a dose of about 50 μg / kg to about 500 μg / kg , for example, at a dose of about 200 μg / kg to about 300 μg / kg , for example, about 240. at a dose of [mu] g / kg, it is administered to the donor. 哺乳動物のドナーの骨髄から末梢血中に造血幹細胞の集団を動員させる方法であって、前記方法は、以下を含む:
a.動員させる量のCXCR2アゴニスト及びCXCR4アンタゴニストを前記ドナーに投与すること;
b.前記ドナーの末梢血のサンプルを特徴付ける、表2に列挙した1つ以上のパラメータの各々についての入力値を取得すること;並びに、
c.前記1つ以上のパラメータの各々についての入力値が前記1つ以上のパラメータの各々についての対応する参照基準を満たす場合、前記サンプルを、哺乳動物の患者における1種以上の幹細胞障害を治療する際に使用するための造血幹細胞をex vivoで増殖させることに、供すること。
A method of mobilizing a population of hematopoietic stem cells from the bone marrow of a mammalian donor into peripheral blood, the method comprising:
a. Administering mobilized doses of CXCR2 agonist and CXCR4 antagonist to said donor;
b. Obtaining input values for each of the one or more parameters listed in Table 2, which characterizes the donor's peripheral blood sample;
c. When the input value for each of the one or more parameters meets the corresponding reference criteria for each of the one or more parameters, the sample is used to treat one or more stem cell disorders in a mammalian patient. the hematopoietic stem cells for use in be grown in ex vivo, subjecting it to.
請求項3に記載の方法、ここで:(a) 前記1つ以上の基準パラメータが、表3〜6の何れか1つに列挙したパラメータの組合せである;及び/若しくは、(b) 前記サンプルを、ex vivoで増殖させることに供する場合、前記方法は、ex vivoで前記造血幹細胞を増殖させることを更に含む;並びに/又は、(c) 前記サンプルを、1種以上の幹細胞障害を治療する際に使用するために供する場合、前記方法は、前記造血幹細胞又はその子孫を、前記1種以上の幹細胞障害に罹患している哺乳動物に注入することを更に含む。 3. The method of claim 3, wherein (a) the one or more reference parameters are a combination of the parameters listed in any one of Tables 3-6 ; and / or (b) the sample. and when subjected to be grown in ex vivo, the method further comprises growing said hematopoietic stem cells ex vivo; and / or, (c) a said sample, treating one or more stem cell disorders When used in an ex vivo manner , the method further comprises injecting the hematopoietic stem cells or their progeny into a mammal suffering from one or more of the stem cell disorders. 哺乳動物のドナーから単離した造血幹細胞又はその子孫の集団を含む医薬組成物、ここで:(a) 前記集団中の白血球に対するCD34+細胞の比率は約0.0008〜約0.0021である、若しくは、約0.0010〜約0.0018である;又は(b) 前記集団中の好中球に対するCD34+細胞の比率は約0.0018〜約0.0058である、若しくは、約0.0026〜約0.0046である;又は(c) 前記集団中のリンパ球に対するCD34+細胞の比率は約0.0021〜約0.0094である、若しくは、約0.0025〜約0.0035である;又は(d) 前記集団中の単球に対するCD34+細胞の比率は約0.0071〜約0.0174である、若しくは、約0.0100〜約0.0140である;又は(e) 前記集団中のCD34+細胞の頻度は約0.051%〜約0.140%である、若しくは、約0.080%〜約0.120%である;又は(f) 前記集団中の白血球に対するCD34+ CD90+ CD45RA- 細胞の比率は約0.0003〜約0.0016である、若しくは、約0.0006〜約0.0012である;又は(g) 前記集団中の好中球に対するCD34+ CD90+ CD45RA- 細胞の比率は約0.0007〜約0.0043である、若しくは、約0.0014〜約0.0034である;又は(h) 前記集団中のリンパ球に対するCD34+ CD90+ CD45RA- 細胞の比率は約0.0008〜約0.0069である、若しくは、約0.0011〜約0.0031である;又は(i) 前記集団中の単球に対するCD34+ CD90+ CD45RA- 細胞の比率は約0.0028〜約0.0130である、若しくは、約0.0063〜約0.0083である;又は(j) 前記集団中のCD34+細胞に対するCD34+ CD90+ CD45RA- 細胞の比率は約0.393〜約0.745である、若しくは、約0.625〜約0.725である;又は(k) 前記集団中のCD34+ CD90+ CD45RA- 細胞の頻度は約0.020%〜約0.110%である、若しくは、約0.046%〜約0.086%である。 A pharmaceutical composition comprising a population of hematopoietic stem cells or their progeny isolated from a mammalian donor, wherein (a) the ratio of CD34 + cells to leukocytes in the population is from about 0.0008 to about 0.0021 , or about 0.0010. ~ About 0.0018 ; or (b) the ratio of CD34 + cells to neutrophils in the population is about 0.0018 to about 0.0058 , or about 0.0026 to about 0.0046 ; or (c) lymph in the population. The ratio of CD34 + cells to spheres is about 0.0021 to about 0.0094 , or about 0.0025 to about 0.0035 ; or (d) the ratio of CD34 + cells to monospheres in the population is about 0.0071 to about 0.0174 , or , About 0.0100 to about 0.0140 ; or (e) the frequency of CD34 + cells in the population is about 0.051% to about 0.140% , or about 0.080% to about 0.120% ; or (f) the population. The ratio of CD34 + CD90 + CD45RA-cells to leukocytes in the population is about 0.0003 to about 0.0016 , or about 0.0006 to about 0.0012 ; or (g) the ratio of CD34 + CD90 + CD45RA-cells to neutrophils in the population. It is about 0.0007 to about 0.0043 , or about 0.0014 to about 0.0034 ; or (h) the ratio of CD34 + CD90 + CD45RA-cells to lymphocytes in the population is about 0.0008 to about 0.0069 , or about 0.0011 to. Or (i) the ratio of CD34 + CD90 + CD45RA-cells to monospheres in the population is about 0.0028 to about 0.0130 , or about 0.0063 to about 0.0083 ; or (j) in the population. The ratio of CD34 + CD90 + CD45RA-cells to CD34 + cells is about 0.393 to about 0.745 , or about 0.625 to about 0.725 ; or (k) the frequency of CD34 + CD90 + CD45RA-cells in the population is about 0.020% to about. It is 0.110% , or about 0.046% to about 0.086%. 哺乳動物の患者における幹細胞障害を治療する方法であって、前記方法は以下を含む:請求項1〜の何れか一項に記載の方法に従って、哺乳動物のドナーにおいて造血幹細胞の集団を動員させること;及び、治療的有効量の造血幹細胞又はその子孫を前記患者に注入すること、任意選択的にここで、前記幹細胞障害は、以下のうちの何れか1種である:
異常ヘモグロビン症障害鎌状赤血球貧血、サラセミア、ファンコニー貧血、再生不良性貧血、及びウィスコット‐アルドリッチ症候群からなる群より選択される)、
骨髄異形成障害
先天性免疫不全症
後天性免疫不全症ヒト免疫不全ウイルス又は後天性免疫不全症候群からなる群より選択される)、
代謝性障害グリコーゲン蓄積症, ムコ多糖症、ゴーシェ病、ハーラー病、スフィンゴ脂質投与量、及び異染性白質ジストロフィーからなる群より選択される)、
がん白血病、リンパ腫、多発性骨髄腫、神経芽細胞腫、急性骨髄性白血病、急性リンパ性白血病、慢性骨髄性白血病、慢性リンパ性白血病、多発性骨髄腫、びまん性大細胞型B-細胞リンパ腫、又は非‐ホジキン・リンパ腫(non-Hodgkin's lymphoma)からなる群より選択される)、
アデノシン・デアミナーゼ欠損症及び重度複合免疫不全症、高免疫グロブリンM症候群、チェディアック‐東病、遺伝性リンパ組織球症、大理石骨病、骨形成不全症、貯蔵疾患、サラセミア・メジャー、全身性硬化症、全身性エリテマトーデス(systemic lupus erythematosus)、多発性硬化症、及び若年性関節リウマチからなる群より選択される障害、又は、
自己免疫障害多発性硬化症、ヒト全身性エリテマトーデス、関節リウマチ、炎症性腸疾患、乾癬の治療、1型糖尿病、急性散在性脳脊髄炎、アジソン病、汎発性脱毛症、強直性脊椎炎、抗リン脂質抗体症候群、再生不良性貧血、自己免疫性溶血性貧血、自己免疫性肝炎、自己免疫性内耳疾患、自己免疫性リンパ増殖症候群、自己免疫性卵巣炎、バロー病、ベーチェット病、水疱性類天疱瘡、心筋症、シャーガス病、慢性疲労免疫不全症候群、慢性炎症性脱髄性多発ニューロパチー、クローン病、瘢痕性類天疱瘡、セリアック・スプルー疱疹状皮膚炎(coeliac sprue-dermatitis herpetiformis)、寒冷凝集素症、CREST症候群、デゴス病、円板状エリテマトーデス、自律神経障害、子宮内膜症、本態性混合型クリオグロブリン血症、線維筋痛症−線維筋炎、グッドパスチャー症候群、グレーブス病、ギラン‐バレー症候群、橋本甲状腺炎、化膿性汗腺炎、特発性及び/又は急性血小板減少性紫斑病、特発性肺線維症、IgAニューロパチー、間質性膀胱炎、若年性関節炎、川崎病、扁平苔癬、ライム病、メニエール病、混合性結合組織病、重症筋無力症、ニューロミオトニア、オプソクローヌス・ミオクローヌス症候群、視神経炎、オルド甲状腺炎(Ord's thyroiditis)、尋常性天疱瘡、悪性貧血、多発軟骨炎、多発性筋炎及び皮膚筋炎、原発性胆汁性肝硬変、結節性多発動脈炎、多内分泌腺症候群、リウマチ性多発筋痛症、原発性無ガンマグロブリン血症(primary agammaglobulinemia)、レイノー現象、ライター症候群、リウマチ熱、サルコイドーシス、強皮症、シェーグレン症候群、スティッフ・パーソン症候群、高安動脈炎、側頭動脈炎、潰瘍性大腸炎、ぶどう膜炎、血管炎、白斑、外陰部痛、及びヴェーゲナー肉芽腫症、からなる群より選択される
A method of treating a stem cell disorder in a mammalian patient, said method comprising: mobilizing a population of hematopoietic stem cells in a mammalian donor according to the method according to any one of claims 1-4. And, injecting a therapeutically effective amount of hematopoietic stem cells or their progeny into the patient , optionally here the stem cell disorder is any one of the following:
Hemoglobinopathy disorders (sickle cell anemia, thalassemia, Fanconi anemia, aplastic anemia, and Wiskott - is selected from the group consisting of Aldrich syndrome),
Myelodysplastic disorder ,
Congenital immunodeficiency ,
Acquired immunodeficiency ( selected from the group consisting of human immunodeficiency virus or acquired immunodeficiency syndrome),
Metabolic disorders ( selected from the group consisting of glycogen accumulation, mucopolysaccharidosis, Gaucher's disease, Harler's disease, sphingolipid doses, and metachromatic leukodystrophy ),
Cancer ( leukemia, lymphoma, multiple myeloma, neuroblastoma, acute myeloid leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, diffuse large cell type B-cell Lymphoma, or selected from the group consisting of non-Hodgkin's lymphoma),
Adenosine deaminase deficiency and severe combined immunodeficiency, hyperimmunoglobulin M syndrome, Chediac-East disease, hereditary lymphohistiocytosis, marble bone disease, osteodysplasia, storage disease, salacemia major, systemic sclerosis Disorders selected from the group consisting of disease, systemic lupus erythematosus, multiple sclerosis, and juvenile lupus erythematosus , or
Autoimmunity disorders ( multiple sclerosis, human systemic erythematosus, rheumatoid arthritis, inflammatory bowel disease, treatment of psoriasis, type 1 diabetes, acute diffuse encephalomyelitis, Addison's disease, generalized alopecia, tonic spondylitis , Antiphospholipid antibody syndrome, poor regeneration anemia, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune lymphoproliferative syndrome, autoimmune ovarian inflammation, Barrow disease, Bechet's disease, blisters Chronic fatigue immunodeficiency syndrome, chronic inflammatory demyelinating polyneuropathy, Crohn's disease, scarring psoriasis, coeliac sprue-dermatitis herpetiformis, Cold agglutinosis, CREST syndrome, Degos's disease, discoid erythematosus, autonomic neuropathy, endometriosis, essential mixed cryoglobulinemia, fibromyalgia-fibromyitis, Good Pasture syndrome, Graves' disease, Gillan -Valley syndrome, Hashimoto thyroiditis, purulent sweat adenitis, idiopathic and / or acute thrombocytopenic purpura, idiopathic pulmonary fibrosis, IgA neuropathy, interstitial cystitis, juvenile arthritis, Kawasaki disease, squamous lichen , Lime's disease, Meniere's disease, mixed connective tissue disease, severe myasthenia, neuromiotonia, opsocronus-myoclonus syndrome, optic neuritis, Ord's thyroiditis, cystitis vulgaris, malignant anemia, polychondritis, Polymyopathy and dermatomyitis, primary biliary cirrhosis, nodular polyarteritis, polyendocrine gland syndrome, rheumatic polymyopathy, primary agammaglobulinemia, Raynaud's phenomenon, Reiter's syndrome, rheumatism From fever, sarcoidosis, scleroderma, Schegren's syndrome, Stiff-Person's syndrome, hyperan arteritis, temporal arteritis, ulcerative colitis, vasculitis, vasculitis, leukoplakia, genital pain, and Wegener's granulomatosis. (Selected from the group of ) .
請求項に記載の方法、ここで、前記造血幹細胞は:(a) 前記患者に対して自己である;(b) 前記患者に対して同種異系である、任意選択的に、前記患者に対してHLA適合である;及び/又は(c) 内因性遺伝子を破壊するように遺伝的に改変されている、任意選択的にここで、前記内因性遺伝子が主要組織適合性遺伝子複合体タンパク質をコードする。 6. The method of claim 6, wherein the hematopoietic stem cells are : (a) self to the patient ; (b) allogeneic to the patient , optionally to the patient. In contrast, it is HLA compatible ; and / or (c) genetically modified to disrupt the endogenous gene , optionally where the endogenous gene is a major histocompatibility gene complex protein. Code. 同種異系造血幹細胞移植を、それを必要とする患者において実施する方法であって、治療的有効量の同種異系造血幹細胞を前記患者に注入することを含み、ここで、前記造血幹細胞を、(i) Gro-β、Gro-β T、及びそれらのバリアントからなる群より選択されるCXCR2アゴニスト、並びに(ii) CXCR4アンタゴニスト、を前記ドナーに投与することを含む方法によって、ヒトのドナーの骨髄からヒトのドナーの末梢血中に動員させる、方法。 A method of performing allogeneic hematopoietic stem cell transplantation in a patient in need thereof, comprising injecting a therapeutically effective amount of allogeneic hematopoietic stem cells into the patient, wherein the hematopoietic stem cells are expressed. Bone marrow of human donors by a method comprising administering to said donor (i) a CXCR2 agonist selected from the group consisting of Gro-β, Gro-β T, and variants thereof, and (ii) a CXCR4 antagonist. A method of mobilizing from to the peripheral blood of a human donor. 移植後感染の予防、発症のリスクの低減、又は重症度の低減を、それを必要とする患者において実施する方法であって、前記患者に治療的有効量の造血幹細胞を注入することを含み、ここで、前記造血幹細胞を、(i) Gro-β、Gro-β T、及びそれらのバリアントからなる群より選択されるCXCR2アゴニスト、並びに(ii) CXCR4アンタゴニスト、を前記ドナーに投与することを含む方法によって、ヒトのドナーの骨髄からヒトのドナーの末梢血中に動員させる、方法。 A method of preventing post-transplant infection, reducing the risk of developing, or reducing the severity in a patient in need thereof, comprising injecting a therapeutically effective amount of hematopoietic stem cells into the patient. Here, the hematopoietic stem cells are administered to the donor with (i) a CXCR2 agonist selected from the group consisting of Gro-β, Gro-β T, and variants thereof, and (ii) a CXCR4 antagonist. A method of mobilizing from the bone marrow of a human donor into the peripheral blood of a human donor by method. 移植片対宿主病(GVHD)の予防、発症のリスクの低減、又は重症度の低減を、それを必要とする患者において実施する方法であって、前記患者に治療的有効量の造血幹細胞を注入することを含み、ここで、前記造血幹細胞を、(i) Gro-β、Gro-β T、及びそれらのバリアントからなる群より選択されるCXCR2アゴニスト、並びに(ii) CXCR4アンタゴニスト、を前記ドナーに投与することを含む方法によって、ヒトのドナーの骨髄からヒトのドナーの末梢血中に動員させる、方法。 A method for preventing graft-versus-host disease (GVHD), reducing the risk of developing it, or reducing its severity in patients in need thereof, injecting a therapeutically effective amount of hematopoietic stem cells into the patient. Here, the hematopoietic stem cells are fed with (i) a CXCR2 agonist selected from the group consisting of Gro-β, Gro-β T, and variants thereof, and (ii) a CXCR4 antagonist. A method of mobilizing from the bone marrow of a human donor into the peripheral blood of a human donor by a method comprising administration. 請求項1~4及び6〜10の何れか一項に記載の方法、ここで:(a) 前記CD34dim細胞は、前記造血幹細胞を前記CXCR4アンタゴニスト単独を使用して動員させた場合よりも、少なくとも2〜10倍高い量で末梢血中に存在する;及び/又は(b) 前記CD34dim細胞は、レシピエントに投与した場合に、同種反応性T-リンパ球が増殖することを抑制することができる。 The method according to any one of claims 1 to 4 and 6 to 10 , wherein (a) the CD34 dim cells are more than the hematopoietic stem cells mobilized using the CXCR4 antagonist alone. It is present in peripheral blood in at least 2-10 fold higher amounts ; and / or (b) the CD34 dim cells suppress the proliferation of allogeneic T-lymphocytes when administered to the recipient. Can be done. 請求項1~4及び6〜11の何れか一項に記載の方法、ここで、前記CXCR4アンタゴニストを、哺乳動物のドナーに、前記CXCR2アゴニストを投与する、約30分から約180分前、約40分から約160分前、約50分から約150分前、約60分から約140分前、約70分から約130分前、又は約120分前、に投与する。The method according to any one of claims 1 to 4 and 6 to 11, wherein the CXCR4 antagonist is administered to a mammalian donor about 30 to about 180 minutes before, about 40 minutes. Administer about 160 minutes to about 160 minutes, about 50 to about 150 minutes, about 60 to about 140 minutes, about 70 to about 130 minutes, or about 120 minutes before. Gro-β Tを生産する方法、ここで、前記方法は、Gro-β Tを化学的合成することを含む、ここで、5%未満、1%未満、又は0.1%未満の前記Gro-β Tは、脱アミド化したAsn65残基を含む。A method for producing Gro-β T, wherein the method comprises chemically synthesizing Gro-β T, wherein less than 5%, less than 1%, or less than 0.1% of the Gro-β T. Contains deamidated Asn65 residues. 哺乳動物のドナーの骨髄から末梢血中に造血幹細胞の集団を動員させる方法であって、(i) Gro-β、Gro-β T、及びそれらのバリアントからなる群より選択されるCXCR2アゴニスト、並びに(ii)CXCR4アンタゴニスト、ここで、10%未満、5%未満、1%未満、又は0.1%未満の前記Gro-β Tは、脱アミド化したAsn65残基を含む、を前記ドナーに投与することを含む方法。A method of mobilizing a population of hematopoietic stem cells from the bone marrow of a mammalian donor into peripheral blood, (i) a CXCR2 agonist selected from the group consisting of Gro-β, Gro-β T, and variants thereof, and (ii) Administer to the donor a CXCR4 antagonist, wherein the Gro-β T <10%, <5%, <1%, or <0.1% contains a deamidated Asn65 residue. How to include. 請求項14に記載の方法、ここで、前記CXCR4アンタゴニストは:(i) 前記ドナーに皮下投与する;(ii) プレリキサホル又はその薬学的に許容可能な塩である、任意選択的にここで、前記プレリキサホル又はその薬学的に許容可能な塩を、約50 μg/kg〜約500 μg/kgの用量で、例えば、約200 μg/kg〜約300 μg/kgの用量で、例えば、約240 μg/kgの用量で前記ドナーに投与する、及び任意選択的にここで、前記CXCR4アンタゴニストを、哺乳動物のドナーに、前記CXCR2アゴニストを投与する、約30分から約180分前、に投与する。 14. The method of claim 14, wherein the CXCR4 antagonist is: (i) administered subcutaneously to the donor ; (ii) prelixaform or a pharmaceutically acceptable salt thereof , optionally said herein. Prelixaform or a pharmaceutically acceptable salt thereof, at a dose of about 50 μg / kg to about 500 μg / kg , for example, at a dose of about 200 μg / kg to about 300 μg / kg , for example , about 240 μg / kg. at a dose of kg, it is administered to the donor, and optionally wherein the CXCR4 antagonist, the donor mammal, administering the CXCR2 agonist is administered from about 30 minutes to about 180 minutes before, to.
JP2020531468A 2017-12-06 2018-12-06 Dosing regimen to mobilize hematopoietic stem cells and progenitor cells Pending JP2021505172A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2023188499A JP2024023226A (en) 2017-12-06 2023-11-02 Dosing regimens for mobilization of hematopoietic stem and progenitor cells

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US15/834,017 US10058573B1 (en) 2017-12-06 2017-12-06 Dosing regimens for the mobilization of hematopoietic stem cells
US15/834,017 2017-12-06
US201762596056P 2017-12-07 2017-12-07
US62/596,056 2017-12-07
US16/101,676 2018-08-13
US16/101,676 US20190167726A1 (en) 2017-12-06 2018-08-13 Dosing Regimens for the Mobilization of Hematopoietic Stem Cells
US201862753656P 2018-10-31 2018-10-31
US62/753,656 2018-10-31
US201862773954P 2018-11-30 2018-11-30
US62/773,954 2018-11-30
PCT/US2018/064335 WO2019113375A2 (en) 2017-12-06 2018-12-06 Dosing regimens for the mobilization of hematopoietic stem and progenitor cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
JP2023188499A Division JP2024023226A (en) 2017-12-06 2023-11-02 Dosing regimens for mobilization of hematopoietic stem and progenitor cells

Publications (2)

Publication Number Publication Date
JP2021505172A JP2021505172A (en) 2021-02-18
JP2021505172A5 true JP2021505172A5 (en) 2022-01-11

Family

ID=66749948

Family Applications (2)

Application Number Title Priority Date Filing Date
JP2020531468A Pending JP2021505172A (en) 2017-12-06 2018-12-06 Dosing regimen to mobilize hematopoietic stem cells and progenitor cells
JP2023188499A Pending JP2024023226A (en) 2017-12-06 2023-11-02 Dosing regimens for mobilization of hematopoietic stem and progenitor cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
JP2023188499A Pending JP2024023226A (en) 2017-12-06 2023-11-02 Dosing regimens for mobilization of hematopoietic stem and progenitor cells

Country Status (12)

Country Link
EP (1) EP3720494A2 (en)
JP (2) JP2021505172A (en)
KR (1) KR20200096942A (en)
CN (1) CN111712262A (en)
AU (1) AU2018378804A1 (en)
BR (1) BR112020011186A2 (en)
CA (1) CA3083783A1 (en)
CO (1) CO2020007275A2 (en)
IL (1) IL275077A (en)
MX (1) MX2020005878A (en)
SG (1) SG11202004913TA (en)
WO (1) WO2019113375A2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11260079B2 (en) 2017-12-06 2022-03-01 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progenitor cells
WO2021087406A1 (en) * 2019-11-01 2021-05-06 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progentor cells
EP4143302A1 (en) * 2020-04-27 2023-03-08 Magenta Therapeutics, Inc. Methods and compositions for transducing hematopoietic stem and progenitor cells in vivo
WO2022197776A1 (en) * 2021-03-16 2022-09-22 Magenta Therapeutics, Inc. Dosing regimens for hematopoietic stem cell mobilization for stem cell transplants in multiple myeloma patients

Family Cites Families (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5233044A (en) 1989-06-08 1993-08-03 Millipore Corporation Active esters for solid phase peptide synthesis
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
US5021409A (en) 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
AU633698B2 (en) 1990-01-12 1993-02-04 Amgen Fremont Inc. Generation of xenogeneic antibodies
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
ATE275198T1 (en) 1991-12-02 2004-09-15 Medical Res Council PRODUCTION OF ANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES.
GB9126677D0 (en) 1991-12-16 1992-02-12 Johnson Matthey Plc Improvements in chemical compounds
US6080398A (en) 1993-06-08 2000-06-27 Smithkline Beecham Corporation Truncated gro and KC chemokines having enhanced bioactivity
US6447766B1 (en) 1993-06-08 2002-09-10 Smithkline Beecham Corporation Method of mobilizing hematopoietic stem cells
GB9400411D0 (en) 1994-01-11 1994-03-09 Johnson Matthey Plc Improvements in chemical compounds
EP1978033A3 (en) 1995-04-27 2008-12-24 Amgen Fremont Inc. Human antibodies derived from immunized xenomice
CA2219486A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6506770B1 (en) 1996-06-06 2003-01-14 Anormed, Inc. Antiviral compounds
US5801030A (en) 1995-09-01 1998-09-01 Genvec, Inc. Methods and vectors for site-specific recombination
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
CA2616914C (en) 1996-12-03 2012-05-29 Abgenix, Inc. Egfr-binding antibody
NZ332174A (en) 1997-02-11 1999-03-29 Mallinckrodt Chemical Inc Reactor and method for solid peptide synthesis
ATE525477T1 (en) 1998-03-30 2011-10-15 Northwest Biotherapeutics Inc THERAPEUTIC AND DIAGNOSTIC APPLICATIONS BASED ON THE ROLE OF CXCR-4 IN TUMORGENESIS
MXPA01009674A (en) 1999-03-24 2003-07-21 Anormed Inc Chemokine recpetor binding heterocyclic compounds.
JP2003516984A (en) 1999-12-17 2003-05-20 アノーメッド・インコーポレイテッド Heterocyclic compounds that bind chemokine receptors
DE60103052T2 (en) 2000-05-09 2005-03-03 The University Of British Columbia, Vancouver USE OF CXCR4 ANTAGONISTS FOR THE TREATMENT OF CANCER AND AUTOIMMUNE DISEASES
JP5414958B2 (en) 2000-06-05 2014-02-12 ザ・トラスティーズ・オブ・コランビア・ユニバーシティー・イン・ザ・シティー・オブ・ニューヨーク Identification of human bone marrow-derived endothelial progenitor cells and use of human bone marrow-derived endothelial progenitor cells to improve the function of cardiomyocytes after ischemic injury
MXPA04000982A (en) * 2001-07-31 2004-04-20 Anormed Inc Methods to mobilize progenitor/stem cells.
US7169750B2 (en) * 2001-07-31 2007-01-30 Anormed, Inc. Methods to mobilize progenitor/stem cells
DE60236520D1 (en) 2001-08-16 2010-07-08 Univ Pennsylvania SYNTHESIS AND USE OF REAGENTS FOR IMPROVED DNA LIPOFEKTION AND / OR PRODRUG AND MEDICAMENT THERAPIES WITH SLOW RELEASE
US20030199464A1 (en) 2002-04-23 2003-10-23 Silviu Itescu Regeneration of endogenous myocardial tissue by induction of neovascularization
US7683036B2 (en) 2003-07-31 2010-03-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
JP2008509928A (en) * 2004-08-13 2008-04-03 アノーメド インコーポレイテッド Chemokine combination to mobilize progenitor / stem cells
US20070033666A1 (en) 2005-06-24 2007-02-08 Harris Reuben S Using cytosine deaminases to diminish retroelement transfer from pigs to humans
US20090221683A1 (en) * 2005-10-18 2009-09-03 Caritas St.Elizabeth Medical Center Of Boston, Inc Combination of CXCR4 Antagonist and Morphogen to Increase Angiogenesis
EP2484758B1 (en) 2005-10-18 2013-10-02 Precision Biosciences Rationally-designed meganucleases with altered sequence specificity and DNA-binding affinity
RU2009107030A (en) * 2006-08-02 2010-09-10 Джензим Корпорейшн (Us) COMBINED THERAPY
MX2009001445A (en) * 2006-08-07 2009-02-18 Genzyme Corp Combination therapy.
EP1995316A1 (en) 2007-05-25 2008-11-26 Qiagen GmbH Method for gentle purification of cells, cell production and cell transfection
EP2009095A1 (en) 2007-06-28 2008-12-31 Innovalor AG Method of generating glucose-responsive cells
EP3098309B1 (en) 2007-10-31 2019-04-10 Precision Biosciences, Inc. Rationally-designed single-chain meganucleases with non-palindromic recognition sequences
PE20100362A1 (en) 2008-10-30 2010-05-27 Irm Llc PURINE DERIVATIVES THAT EXPAND HEMATOPOYETIC STEM CELLS
EP2270025A1 (en) 2009-06-29 2011-01-05 Centre National pour la Recherche Scientifique (CNRS) Solid phase peptide synthesis of peptide alcohols
WO2011100505A2 (en) 2010-02-11 2011-08-18 Recombinetics, Inc. Methods and materials for producing transgenic artiodactyls
CA2828239C (en) 2011-02-25 2020-10-06 Recombinetics, Inc. Genetically modified animals and methods for making the same
US9388212B2 (en) 2013-02-21 2016-07-12 Chemical & Biopharmaceutical Laboratories Of Patras S.A. Solid phase peptide synthesis via side chain attachment
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
KR20160023638A (en) * 2013-02-28 2016-03-03 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Methods and compositions for mobilizing stem cells
US9169287B2 (en) 2013-03-15 2015-10-27 Massachusetts Institute Of Technology Solid phase peptide synthesis processes and associated systems
WO2017147610A1 (en) * 2016-02-26 2017-08-31 President And Fellows Of Harvard College Highly engraftable hematopoietic stem cells
US10058573B1 (en) * 2017-12-06 2018-08-28 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem cells

Similar Documents

Publication Publication Date Title
JP2021505172A5 (en)
Liu et al. Busulfan plus fludarabine as a myeloablative conditioning regimen compared with busulfan plus cyclophosphamide for acute myeloid leukemia in first complete remission undergoing allogeneic hematopoietic stem cell transplantation: a prospective and multicenter study
Gao et al. Phase II multicenter, randomized, double-blind controlled study of efficacy and safety of umbilical cord–derived mesenchymal stromal cells in the prophylaxis of chronic graft-versus-host disease after HLA-haploidentical stem-cell transplantation
Zhao et al. Immunomodulatory properties of mesenchymal stromal cells and their therapeutic consequences for immune-mediated disorders
Jurewicz et al. Congenic mesenchymal stem cell therapy reverses hyperglycemia in experimental type 1 diabetes
Kim et al. The potential use of mesenchymal stem cells in hematopoietic stem cell transplantation
Wang et al. Umbilical cord mesenchymal stem cell transplantation in active and refractory systemic lupus erythematosus: a multicenter clinical study
US9301979B2 (en) Method for attenuating immune response using mesenchymal stem cells and cytokines
Fang et al. Favorable response to human adipose tissue-derived mesenchymal stem cells in steroid-refractory acute graft-versus-host disease
Tang et al. The therapeutic effect of ICAM-1-overexpressing mesenchymal stem cells on acute graft-versus-host disease
Lu et al. Unmanipulated haploidentical hematopoietic stem cell transplantation achieved outcomes comparable with matched unrelated donor transplantation in young acquired severe aplastic anemia
Chang et al. Granulocyte colony-stimulating factor-primed unmanipulated haploidentical blood and marrow transplantation
Bacigalupo Alternative donor transplants for severe aplastic anemia
Prabahran et al. Clinical features, pathophysiology, and therapy of poor graft function post–allogeneic stem cell transplantation
Luo et al. CMV infection and CMV-specific immune reconstitution following haploidentical stem cell transplantation: an update
Murphy et al. Mesenchymal stem cell therapy to promote corneal allograft survival: current status and pathway to clinical translation
Picardi et al. Brentuximab vedotin followed by bendamustine supercharge for refractory or relapsed Hodgkin lymphoma
Jaiswal et al. Higher CD45RA+ Regulatory T Cells in the Graft Improves Outcome in Younger Patients Undergoing T Cell–Replete Haploidentical Transplantation: Where Donor Age Matters
JP2021502824A5 (en)
Ringdén et al. Mesenchymal stromal cells in pediatric hematopoietic cell transplantation a review and a pilot study in children treated with decidua stromal cells for acute graft-versus-host disease
Tyndall et al. Adult stem cell transplantation in autoimmune disease
Yolcu et al. Immunomodulation with donor regulatory T cells armed with Fas-ligand alleviates graft-versus-host disease
Wang et al. Clinical application of mesenchymal stem cells in rheumatic diseases
Lin et al. Role of memory T cells and perspectives for intervention in organ transplantation
Fu et al. Reduced intensity conditioning and co-transplantation of unrelated peripheral stem cells combined with umbilical cord mesenchymal stem/stroma cells for young patients with refractory severe aplastic anemia