WO1998052598A1 - Nouveau procede d'administration de thrombopoietine - Google Patents

Nouveau procede d'administration de thrombopoietine Download PDF

Info

Publication number
WO1998052598A1
WO1998052598A1 PCT/US1998/010475 US9810475W WO9852598A1 WO 1998052598 A1 WO1998052598 A1 WO 1998052598A1 US 9810475 W US9810475 W US 9810475W WO 9852598 A1 WO9852598 A1 WO 9852598A1
Authority
WO
WIPO (PCT)
Prior art keywords
tpo
dose
administered
cells
treatment
Prior art date
Application number
PCT/US1998/010475
Other languages
English (en)
Inventor
Robert L. Cohen
Dan L. Eaton
Andrew J. S. Jones
Dennie V. Jones
Michael F. Powell
Theresa D. Sweeney
Griffith R. Thomas
Gerard Wagemaker
Original Assignee
Genentech, Inc.
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 Genentech, Inc. filed Critical Genentech, Inc.
Priority to JP55066298A priority Critical patent/JP4562815B2/ja
Priority to AU75898/98A priority patent/AU7589898A/en
Priority to CA002288964A priority patent/CA2288964A1/fr
Priority to EP98923662A priority patent/EP0983082A1/fr
Publication of WO1998052598A1 publication Critical patent/WO1998052598A1/fr

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
    • A61K38/18Growth factors; Growth regulators
    • 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
    • A61K38/196Thrombopoietin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents

Definitions

  • the present invention relates to a new method of using thrombopoietin, and biologically active derivatives and isoforms thereof, for the treatment of immune and/or hematopoietic disorders including thrombocytopenia.
  • the use contemplates the co-administration of such materials together with a cytokine, especially a colony stimulating factor or interleukin.
  • the use includes and is included within a method for treating a mammal having or at risk for thrombocytopenia by administering to the mammal in need of such treatment a therapeutically effective amount of the material(s).
  • the hematopoietic system produces the mature highly specialized blood cells known to be necessary for survival of all mammals. These mature cells include erythrocytes, specialized to transport oxygen and carbon dioxide, T- and B-lymphocytes, responsible for cell- and antibody- mediated immune responses, platelets or thrombocytes, specialized to form blood clots, and granulocytes and macrophages, specialized as scavengers and as accessory cells to combat infection. All of these specialized mature blood cells are derived from a single common primitive ceTl type referred to as the pluripotent stem cell found primarily in bone marrow.
  • the mature highly specialized blood cells must be produced in large numbers continuously throughout the life of a mammal.
  • the vast majority of these specialized blood cells are destined to remain functionally active for only a few hours to weeks.
  • continuous renewal of these mature blood cells, the primitive stem cells themselves, as well as any intermediate or lineage, committed progenitor cell lines lined between the primitive and mature cells is necessary in order to maintain the normal steady state blood cell needs for continued life of the mammal.
  • pluripotent stem cell(s) At the heart of the hematopoietic system lies the pluripotent stem cell(s). These cells are relatively few in number and undergo self-renewal by proliferation to produce daughter stem cells, or they are transformed in a series of differentiation steps into increasingly mature lineage-restricted progenitor cells, ultimately forming the highly specialized mature blood cell(s).
  • the underlying principal of the normal hematopoietic cell system appears to be decreased capacity for self-renewal as multipotency is lost and lineage-restriction and maturity is acquired.
  • the pluripotent stem cell possessing the capacity for self-renewal and differentiation into all the various lineage-specific committed progenitor cells.
  • the highly lineage-restricted progenitors and their progeny which have lost the ability of self renewal but have acquired mature functional activity.
  • hematopoietic growth factors may influence growth and differentiation of one or more lineages, may overlap with other growth factors in affecting a single progenitor cell-line, or may act synergistically with other factors.
  • novel hematopoietic growth factors that effect survival, proliferation, differentiation or maturation of any of the blood cells or predecessors thereof would be useful, especially to assist in the re-establishment of a diminished hematopoietic system caused by disease or after radiation- or chemo-therapy.
  • Platelets are critical elements of the blood clotting mechanism. Depletion of the circulating level of platelets, called thrombocytopenia, occurs and is manifested in various clinical conditions and disorders. Clinical thrombocytopenia is commonly defined as a condition wherein the platelet count is below about 150 X 10 ⁇ per liter.
  • thrombocytopenia can be broadly divided into three categories on the basis of platelet life span, namely: 1) impaired production of platelets by the bone marrow, e.g., thrombocytopenia brought about by chemo- and radiation-therapy, 2) platelet sequestration in the spleen (splenomegaly) and 3) increased destruction of platelets in the peripheral circulation, e.g., thrombocytopenia brought about by autoimmune disorders. Additionally, in patients receiving large volumes of rapidly administered platelet-poor blood products, thrombocytopenia may develop due to dilution factors.
  • thrombocytopenia A more detailed description of thrombocytopenia and its causes, may be found in Schafner, "Thrombocytopenia and Disorders of Platelet Disfunction", Internal Medicine, John J. Hutton et al. Eds., Little, Brown & Co., Boston/Toronto/London, Third Ed. (1990) as well as International Patent Application No. PCT US94/14553 (International Publication No. W095/18858).
  • the therapeutic approach to the treatment of patients with thrombocytopenia is dictated by the severity and urgency of the clinical situation.
  • the treatment is similar for HIV-associated and non- HlV-related thrombocytopenia, and although a number of different therapeutic approaches have been used, the therapy remains clinically controversial.
  • TPO thrombopoietin
  • MK-CSF megakaryocyte colony-stimulating factor
  • MGDF megakaryocyte growth and development factor
  • mpl ligand megakaryocyte stimulating factor
  • the PCT US94/14553 application includes various aspects of associated embodiments of TPO, including a method of treating a mammal having or at risk for a hematopoietic disorder, notably thrombocytopenia, comprising administering a therapeutically effective amount of TPO materials to the mammal.
  • TPO is administered as such or in combination with a cytokine, especially a colony stimulating factor or interleukin.
  • cytokine especially a colony stimulating factor or interleukin.
  • TPO is broadly defined as including TPO itself or various variants, derivatives or isoforms thereof, including fragments that share at least one biological property in common with intact TPO for the treatment of thrombocytopenia.
  • Bio property when used in conjunction with the definition of the various TPO materials useful as described in the patent application, means that they have thrombopoietic activity or an in vivo effector or antigenic function or activity that is directly or indirectly caused or performed by the TPO material.
  • the TPO materials are therein described for administration in admixture with a pharmaceutically acceptable carrier via any of several administrative modes.
  • the daily regimen is described as ranging from about 0.1 to 100 microgram/kilogram body weight, preferably from about 0.1 to 50 microgram kg body weight, preferably at an initial dosage ranging from about 1 to 5 microgram/kg per day.
  • Implicit within the teachings of the patent application is a regimen of administering such a dosage rate over a period of several to many days following a projected or actual state of reduced platelet count.
  • thrombopoietin indicates a dosage and administration regimen consisting of the administration of thrombopoietin, subcutaneously at dosages of 0.03 to 5.0 microgram/kg body weight once per day over a period often days for a condition marked by thrombocytopenia. See Abstract 1977, Blood 86 (1995). See also Abstracts 1012, 1014 and 1978, Blood 86 (1995).
  • PEG pegylated murine megakarocyte growth and development factor
  • the compound epoetin alfa which is a given name for erythropoietin (marketed as EPOGEN by Amgen, Inc.), is a glycoprotein indicated for stimulation of red blood cell production. It is indicated in a dosage and administration regimen consisting of starting doses over a range of 150 to 300 units per kg three times weekly for a period of many weeks in order to stimulate the proliferation of red blood cells in patients suffering from a depletion however realized.
  • G-CSF granulocyte colony stimulating factor
  • GM-CSF has also been studied clinically for use in conjunction with chemotherapy. In contrast to G-CSF, GM-CSF has a relatively short effective half-life. Administration of GM-CSF is followed by a rapid increase in the proliferative activity of the hematopoietic precursors. However, within 72 hours after suspension of administration, a negative feedback is established resulting in a reduction of the proliferative activity of the marrow to values below baseline. The short half-life of GM-CSF has enabled this cytokine to be administered prior to chemotherapy. Cancer, 1993, vol. 72, No. 10.
  • the conventional regimen in administering materials for the proliferation of red blood cells or other primary blood cells to reverse the effects of thrombocytopenia is continuous administration of therapeutically effective amounts of the biological material daily over a period of many days to patients in need of such therapy following chemotherapy resulting in thrombocytopenia. While GM-CSF may have limited effectiveness when administered prior to chemotherapy, G-CSF worsens patient thrombocytopenia when administered prior to chemotherapy.
  • cytokines having a relatively long half-life and which start hematopoietic progenitor cells cycling and proliferating, prior to treatment with radiation or a chemotherapeutic agent has generally been contra-indicated since these cytoreductive treatments kill not only malignant cells, but also the proliferating progenitor cells as well.
  • Another approach to the treatment of patients with thrombocytopenia or who are at risk of thrombocytopenia as a result of a medical procedure, e.g. radiation and/or chemotherapy, is to rescue the patient with an autologous hematopoietic implant.
  • patients are administered a compound which mobilizes peripheral blood hematopoietic progenitor cells prior to the medical treatment which will induce thrombocytopenia.
  • the mobilized progenitor cells are harvested by known leukapheresis procedures and then retransplanted into the patient after the onset of thrombocytopenia in order to reestablish the patients autologous hematopoietic cells in the bone marrow.
  • One object of the present invention is to provide a method of administering a thrombopoietin which provides improved recovery from thrombocytopenia and overcomes the deficiencies noted above for existing methods of administering cytokines.
  • Another object is to provide a method of administering a thrombopoietin to a mammal or patient receiving radiation and/or chemotherapy treatment which minimizes thrombocytopenia associated with such treatment and reduces the need for platelet transfusions in the mammal.
  • thrombopoietin materials are growth factors for and are believed to act directly on early bone marrow stem cells and megakaryocyte progenitor cells, in contrast to G-CSF and GM-CSF which are thought to act on progenitor cells later in the hematopoietic cell lineage.
  • the materials of the invention are capable of causing megakaryocyte differentiation of stem cells and increasing platelet count following administration. They induce proliferation and differentiation of bone marrow hematopoietic cells, increasing the number of mature megakaryocytes, which yield increased numbers of circulating platlets.
  • the present invention is directed to a method of treating a mammal having or at risk for thrombocytopenia comprising administering to a mammal in need of such treatment a single or low- multiple daily dose of a therapeutically effective amount of a thrombopoietin.
  • the present invention is directed to the single administration of a therapeutically effective amount of a thrombopoietin to such a mammal.
  • the invention concerns the administration of a thrombopoietin to a mammal which receives at least one cycle of radiation and/or chemotherapeutic agent in need of such a cycle.
  • the mammal will need one or more of such a cycle for the treatment of a tumor, malignancy, etc.
  • the invention is directed to a method of reducing the number of platelet transfusions in a thrombocytopenic patient.
  • the invention is directed to a method of mobilizing progenitor cells by the administration of a single or low-multiple daily dose of an effective amount of a thrombopoietin.
  • Figure 1 Animals rendered pancytopenic, by a combination of 5.0 Gy of ⁇ -irradiation and carboplatin (1.2 mg), were injected subcutaneously with 0.1 microgram rmTPO(335) for 1,2, 4, or 8 days.
  • Panel A shows the platelet response to the treatment regimens while panels B and C represent the erythrocyte and leukocyte responses respectively over a 28 day period.
  • the key set forth in panel B refers to all three panels.
  • Figure 2 Animals rendered pancytopenic, by a combination of 5.0 Gy of ⁇ -irradiation and carboplatin (1.2 mg), were injected subcutaneously with a single dose at various levels of rmTP0(335) 24 hours after the initiation of the experiment.
  • Panel A shows the platelet response to the treatment regimens while panels B and C represent the erythrocyte and leukocyte responses respectively over a 28 day period.
  • the key set forth in panel B refers to all three panels.
  • Figure 3 Log-linear representations of the platelet (panel A) and erythrocyte (panel B) responses to single administrations of rmTP0(335) given either subcutaneously or intravenously in animals rendered pancytopenic by a combination of 5.0 Gy of ⁇ - irradiation and carboplatin (1.2 mg).
  • the cell numbers plotted are those measured on day 14 after initiation of the experiment.
  • Q is base line zero level.
  • Figure 4 Animals rendered pancytopenic, by a combination of 5.0 Gy of ⁇ -irradiation and carboplatin (1.2 mg), were injected intravenously with a single dose at various levels of rmTP0(335) 24 hours after the initiation of the experiment.
  • Panel A shows the platelet response to the treatment regimens while panels B and C represent the erythrocyte and leukocyte responses respectively over a 28 day period.
  • the key set forth in panel B refers to all three panels.
  • Figure 5 Animals rendered pancytopenic, by a combination of 5.0 Gy of ⁇ -irradiation and carboplatin (1.2 mg), were injected subcutaneously with a single dose at 24 hours after the initiation of the experiment with various forms of rmTP0(153) conjugated to polyethylene glycol (peg) of either 20K or 40K molecular weight.
  • Panel A shows the platelet response to the treatment regimens while panels B and C represent the erythrocyte and leukocyte responses respectively over a 28 day period.
  • the key set forth in panel B refers to all three panels.
  • Figure 6 Animals rendered pancytopenic, by a combination of 5.0 Gy of ⁇ -irradiation and carboplatin (1.2 mg), were injected subcutaneously with a single dose at 24 hours after the initiation of the experiment with either rmTP0(335) or rmTPO(l 53) conjugated to polyethylene glycol (peg) of 40K molecular weight.
  • Panel A shows the platelet response to the treatment regimens while panels B and C represent the erythrocyte and leukocyte responses respectively over a 28 day period.
  • the key set forth in panel B refers to all three panels.
  • Figure 7 Animals rendered pancytopenic, by a combination of 5.0 Gy of ⁇ -irradiation and carboplatin (1.2 mg), were injected intravenously with a single dose at 24 hours after the initiation of the experiment with either rmTP0(335) or rmTPO(153) conjugated to polyethylene glycol (peg) of 40K molecular weight.
  • Panel A shows the platelet response to the treatment regimens while panels B and C represent the erythrocyte and leukocyte responses respectively over a 28 day period.
  • the key set forth in panel B refers to all three panels.
  • Figure 8 - Figure 8 shows the thrombocyte level of 6 Gy irradiated mice at the time of nadir in placebo controls as a function of the time of administration of a single i.p. dose (0.3 microgram) of TPO at each of the various time points indicated in the Figure.
  • Figure 9 - Figure 9 shows the thrombocyte level of 6 Gy irradiated mice at the time of nadir in placebo controls as a function of the time of administration of a single i.p. dose (30 microgram) of TPO at two hours before (-2h) irradiation.
  • FIG 10 In order to model a protracted form of cytoreductive treatment very similar to radiation or chemotherapy, total body irradiation (TBI) was given to mice in three equal fractions of 3 Gy separated by 24 hours each. TPO was given in a total dose of 0.9 microgram in three different dosing regimen; 3 x 0.3 microgram at +2h from irradiation, 0.9 microgram at +2h from irradiation, and 0.9 microgram at -2h from irradiation. The resulting thrombocyte levels are shown vs a placebo.
  • FIG 11 This Figure shows hemopoietic progenitor cell data of the femur for the regimen of Figure 10.
  • Figure 12 This Figure shows hemopoietic progenitor cell data of the spleen for the regimen of Figure 10.
  • Figure 13 - Figure 13 shows pharmacokinetic data following three doses of 0.3 microgram or a single dose of 0.9 microgram of TPO.
  • Figure 14 - Figure 14 shows median platelet counts averaged over dose levels by study arm for Example 7, cycle 1 (chemotherapy alone).
  • Figure 15 - Figure 15 shows median platelet counts averaged over dose levels by study arm for
  • Example 7, cycle 2 (chemotherapy and rhTPO).
  • Figure 16 - Figure 16 shows median platelet counts averaged over dose levels by study arm for Example 7, cycle 3 (chemotherapy and rhTPO).
  • Figure 17 - Figure 17 shows median platelet counts averaged over dose levels by study arm for Example 7, cycle 4 (chemotherapy and rhTPO).
  • Figure 18 - Figure 18 shows median platelet count by rhTPO dose level for arm C, cycle 2 of
  • Figure 19 - Figure 19 shows median platelet count by rhTPO dose level for arm D, cycle 2 of Example 7.
  • Figure 21 Platelet counts in mice exposed to multiple inhalations of rhTPO; see Example 8.
  • Figure 22 Expansion of CD34 + cells by TPO/FL/KL.
  • Figure 22 is shown the expansion of CD34 + cells over 8 weeks in cultures containing TPO, Flt-3 and c-kit ligand. An expansion of over a 10e6 fold is observed; see Example 14.
  • Figure 23 - Expansion of CD34+CD38" cells by TPO/KL/FL.
  • the subpopulation of CD34 + CD38 " cells is also expanded. At week one this subpopulation only makes up only 8% of the culture but by week 8 comprises 33% of the culture, indicating a 4 fold expansion.
  • a "mammal having or at risk for thrombocytopenia” means a mammal, including a human, which is experiencing thrombocytopenia, that is, a platelet count which is below the platelet count for average normal individuals in the mammal population.
  • thrombocytopenia is defined as a condition where the platelet count is below about 150 x 10 ⁇ per liter of blood.
  • the mammal may, however, also be at risk for thrombocytopenia, meaning that the mammal may forseeably experience a thrombocytopenic condition as a result of a specific treatment which is known to cause thrombocytopenia.
  • a mammal is at risk for thrombocytopenia if the mammal will be administered a radiation and/or chemotherapeutic treatment which is known to induce thrombocytopenia in the treated mammal.
  • the mammal is at risk for or has a high probability of experiencing thrombocytopenia as a result of the treatment which is known to induce thrombocytopenia.
  • Such mammals at risk for thrombocytopenia may be treated with the method of the present invention. Included within the scope of this invention are mammals having or at risk for thrombocytopenia as a result of a disfunctional liver, e.g. liver cirrhosis, and mammals undergoing progenitor cell mobilization therapy and apheresis, generally prior to radiation and/or chemotherapy treatment.
  • cytokine is a generic term for proteins released by one cell population which act on another cell as intercellular mediators.
  • lymphokines include lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone, insulin-like growth factors, human growth hormone including N-methionyl human growth hormone, bovine growth hormone, parathyroid hormone, thyroxine, insulin, proinsulin, relaxin, prorelaxin, glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and leutinizing hormone (LH), hematopoietic growth factor, hepatic growth factor, fibroblast growth factor, prolactin, placental lactogen, tumor necrosis factors (TNF- ⁇ and TNF- ⁇ ), mullerian-inhibiting substance, mouse gonadotropin-associated peptide, inhibin, activin, vascular endothelial growth factor, integrin, nerve growth factors (NGFs) such as
  • the foregoing terms are meant to include proteins from natural sources or from recombinant cell culture. Similarly, the terms are intended to include biologically active equivalents; e.g., differing in amino acid sequence by one or more amino acids or in type or extent of glycosylation.
  • biologically active when used in conjunction with a thrombopoietin (TPO) means thrombopoietin or a thrombopoietic polypeptide that exhibits thrombopoietic activity or shares an effector function of the mpl ligand isolated from aplastic porcine plasma or expressed in recombinant cell culture.
  • TPO thrombopoietin
  • Another known effector function of the mpl ligand or polypeptide herein is the ability to stimulate the incorporation of -"S into circulating platelets in a mouse platelet rebound assay.
  • Yet another known effector function of mpl ligand is the ability to stimulate in vitro human megakaryocytopoiesis that may be quantitated by using a radio labeled monoclonal antibody specific to the megakaryocyte glycoprotein GPIIbIII a .
  • mpl ligand mpl ligand polypeptide
  • ML mpl ligand polypeptide
  • TPO thrombopoietin
  • An exemplary biological property is the ability to stimulate the incorporation of labeled nucleotides (e.g. ⁇ H-thymidine) into the DNA of IL-3 dependent Ba/F3 cells transfected with human mpl.
  • Another exemplary biological property is the ability to stimulate the incorporation of 35 S into circulating platelets in a mouse platelet rebound assay.
  • This definition encompasses a polypeptide isolated from a mpl ligand source such as aplastic porcine plasma described herein or from another source, such as another animal species, including humans, or prepared by recombinant or synthetic methods.
  • a mpl ligand source such as aplastic porcine plasma described herein or from another source, such as another animal species, including humans, or prepared by recombinant or synthetic methods.
  • Examples include TPO(332) and rhTP0332-
  • TPO thrombopoietic ligand described in WO 95/28907 having a molecular weight of about 31 ,000 daltons (31 kd) as determined by SDS gel under reducing conditions and 28,000 daltons (28kd) under non-reducing conditions.
  • TPO includes variant forms, such as fragments, alleles, isoforms, analogues, chimera thereof and mixtures of these forms. For convenience, all of these ligands will be referred to below simply as "TPO"
  • the TPO is a compound having thrombopoietic activity or being capable of increasing serum platelet counts in a mammal.
  • the TPO is preferably capable of increasing endogenous platelet counts by at least 10%, more preferably by 50%, and most preferably capable of elevating platelet counts in a human to greater than about 150 X 10 ⁇ per liter of blood.
  • the TPO of this invention preferably has at least 70% overall sequence identity with the amino acid sequence of the highly purified substantially homogeneous porcine mpl ligand polypeptide and at least 80% sequence identity with the "EPO-domain" of the porcine mpl ligand polypeptide.
  • the TPO of this invention may be a mature human mpl ligand (hML), or a variant or post- transcriptionally modified form thereof or a protein having about 80% sequence identity with mature human mpl ligand.
  • the TPO may be a fragment, especially an amino-terminus or "EPO- domain" fragment, of the mature human mpl ligand.
  • the amino terminus fragment retains substantially all of the human ML sequence between the first and fourth cysteine residues but may contain substantial additions, deletions or substitutions outside that region.
  • the fragment polypeptide may be represented by the formula:
  • hTPO(7-151) represents the human TPO (hML) amino acid sequence from Cys 7 through Cys 151 inclusive
  • X represents the amino group of Cys 7 or one or more of the amino-terminus amino acid residue(s) of the mature TPO or amino acid residue extensions thereto such as Met, Lys, Tyr or amino acid substitutions thereof such as arginine to lysine or leader sequences containing, for example, proteolytic cleavage sites (e.g. Factor Xa or thrombin)
  • Y represents the carboxy terminal group of Cys ⁇ l or one or more carboxy-terminus amino acid residue(s) of the mature TPO or extensions thereto.
  • a "TPO fragment” means a portion of a naturally occurring mature full length mpl ligand or TPO sequence having one or more amino acid residues or carbohydrate units deleted.
  • the deleted amino acid residue(s) may occur anywhere in the peptide including at either the N-terminal or C- terminal end or internally, so long as the fragment shares at least one biological property in common with mpl ligand.
  • Mpl ligand fragments typically will have a consecutive sequence of at least 10, 15, 20, 25, 30 or 40 amino acid residues that are identical to the sequences of the mpl ligand isolated from a mammal including the ligand isolated from aplastic porcine plasma or the human or murine ligand, especially the EPO-domain thereof.
  • Representative examples of N-terminal fragments are TPO(153), hML 153 or TPO(Mer 1 1-153).
  • TPO isoform(s) and TPO sequence isoform(s) or the term “derivatives” in association with TPO, etc. as used herein means a biologically active material as defined below having less than 100% sequence identity with the TPO isolated from recombinant cell culture, aplastic porcine plasma or the human mpl ligand.
  • a biologically active mpl ligand or TPO isoform will have an amino acid sequence having at least about 70% amino acid sequence identity with the mpl ligand/TPO isolated from aplastic porcine plasma or the mature murine, human mpl ligand or fragments thereof, preferably at least about 75%, more preferably at least about 80%, still more preferably at least about 85%, even more preferably at least about 90%, and most preferably at least about 95%.
  • the TPO may have the formula:
  • Xaa at position 37 is Thr, Asp or Glu
  • Xaa at position 46 is Phe, Ala, Val, Leu, He, Pro, Tip, or Met;
  • Xaa at position 47 is Ser, Asp or Glu
  • Xaa at position 112 is deleted or Leu, Ala, Val, He, Pro, Phe, Tip, or Met;
  • Xaa at position 113 is deleted or Pro, Phe, Ala, Val, Leu, He, Tip, or Met;
  • Xaa at position 114 is deleted or Pro, Phe, Ala, Val, Leu, He, Tip, or Met;
  • Xaa at position 122 is Lys, Arg, His, Glu, or Asp;
  • Xaa at position 200 is Tip, Ala, Val, Leu, He, Pro, Phe, Met, Arg and Lys, or His.
  • Xaa at position 37 is Thr, Asp or Glu
  • Xaa at position 46 is Phe, Ala, Val, Leu, He, Pro, Tip, or Met;
  • Xaa at position 47 is Ser, Asp or Glu;
  • Xaa at position 112 is deleted or Leu, Ala, Val, He, Pro, Phe, Tip, or Met;
  • Xaa at position 113 is deleted or Pro, Phe, Ala, Val, Leu, He, Tip, or Met;
  • Xaa at position 114 is deleted or Pro, Phe, Ala, Val, Leu, He, Tip, or Met;
  • Xaa at position 115 is deleted or Gin, Gly, Ser, Thr, Tyr, or Asn; Xaa at position 122 is Lys, Arg, His, Glu, or Asp; and with the proviso that at least one of the amino acids designated by Xaa is different from the corresponding amino acids of the native TPO (1-332).
  • These variants may have an improved biological profile, such as increased proliferative activity and/or decreased side-effects, and/or improved physical properties, such as improved half-life, stability, and/or re-fold efficiencies.
  • the preparation of the polypeptides of this embodiment is described in W096/23888.
  • a "chimeric polypeptide” or “chimera” as used herein is a polypeptide containing full length parent ligand (TPO or mpl ligand) or one or more fragments thereof fused or bonded to a second heterologous polypeptide or one or more fragments thereof.
  • the chimera will share at least one biological property in common with TPO.
  • the second polypeptide will typically be a cytokine, for example the cytokines noted above, immunoglobin or fragment thereof.
  • the two polypeptides may be directly bonded together or may be bonded together through a linker, for example a peptide linker which may have 2-50, generally 2-20 amino acid units.
  • TPO/G-CSF TPO/GM-CSF
  • TPO/IL-3 TPO/IL-6
  • Preparation of chimeric proteins may be accomplished using methods well-known in the art.
  • biological property when used in conjunction with either the "mpl ligand" or
  • isolated mpl ligand or “TPO” means having thrombopoietic activity or having an in vivo effector or antigenic function or activity that is directly or indirectly caused or performed by a mpl ligand or “TPO” (whether in its native or denatured conformation) or a fragment thereof.
  • Effector functions include mpl binding and any carrier binding activity, agonism or antagonism of mpl, especially transduction of a proliferative signal including replication, DNA regulatory function, modulation of the biological activity of other cytokines, receptor (especially cytokine) activation, deactivation, up-or down regulation, cell growth or differentiation and the like.
  • An antigenic function means possession of an epitope or antigenic site that is capable of cross-reacting with antibodies raised against the native mpl ligand or TPO.
  • the principal antigenic function of a mpl ligand or TPO polypeptide is that it binds with an affinity of at least about 10 ⁇ 1/mole to an antibody raised against the mpl ligand or TPO isolated from aplastic porcine plasma. Ordinarily, the polypeptide binds with an affinity of at least about 10 7 1/mole.
  • the antigenically active mpl ligand or TPO polypeptide is a polypeptide that binds to an antibody raised against the mpl ligand or TPO having one of the above described effector functions.
  • the antibodies used to define "biological property" are rabbit polyclonal antibodies raised by formulating the mpl ligand or TPO isolated from recombinant cell culture or aplastic porcine plasma in Freund's complete adjuvant, subcutaneously injecting the formulation, and boosting the immune response by intraperitoneal injection of the formulation until the titer of mpl ligand or TPO antibody plateaus.
  • Thrombopoietic activity is defined as biological activity characterized by accelerating the proliferation, differentiation and/or maturation of megakaryocytes or megakaryocyte precursors into the platelet producing form of these cells. This activity may be measured in various assays including an in vivo mouse platelet rebound synthesis assay, induction of platelet cell surface antigen assay as measured by an anti-platelet immunoassay (anti-GPIIbIII a ) for a human leukemia megakaryoblastic cell line (CMK), and induction of polyploidization in a megakaryoblastic cell line (DAMI).
  • an in vivo mouse platelet rebound synthesis assay induction of platelet cell surface antigen assay as measured by an anti-platelet immunoassay (anti-GPIIbIII a ) for a human leukemia megakaryoblastic cell line (CMK), and induction of polyploidization in a megakaryoblastic cell line (DAMI).
  • low-multiple in connection with the dosing is meant the administration of multiple doses of therapeutically effective amounts over a short period of time.
  • a low-multiple dose may include 2 to about 6 doses per day, preferably 2-4 doses per day.
  • the present invention is directed to the mere single administration of a therapeutically effective amount of a thrombopoietin. It has been found that a single administration produces a therapeutic effect equivalent to that realized when a therapeutically effective amount of the same material is administered over the conventional multiple many day regimen suggested and taught by the art.
  • a typical treatment phase may run 1-10 days, preferably 1-6 or 1-4 days, during which time the radiation or chemotherapeutic agent is administered continuously or portion-wise.
  • a typical recovery phase may run 5-60 days, preferably 14-24 days, during which time the mammal or patient is observed, evaluated and allowed to recover from the treatment.
  • more than one treatment cycle may be given, typically 2 to about 6 cycles depending on the particular treatment regimen and the purpose of the treatment.
  • TPO has pharmacokinetic properties which are suprisingly different than the properties of cytokines such as G-CSF and GM-CSF and which allow TPO to be administered prior to and/or concurrently with radiation and/or chemotherapy.
  • cytokines such as G-CSF and GM-CSF
  • Prior and/or concurrent administration of TPO has been found to reduce the depth of the nadir of thrombocytopenia and to shorten the time for platelet titer recovery in patients receiving radiation and/or chemotherapeutic treatment. This difference in properties of TPO is believed to derive from the effect of TPO on early progenitor cells in the hematopoietic lineage.
  • TPO tumor necrosis factor
  • This effect appears to result in a delay in the appearance of more mature cells in the lineage following administration of TPO, allowing radiation and/or chemotherapy treatments to be given with little or clinically insignificant loss of proliferating cells during cytoreductive treatments.
  • the discovery of these unique properties of TPO is part of the present invention.
  • the method of the invention hastens the recovery of platelet titers to baseline levels and even to substantially elevated levels following radiation and/or chemotherapy.
  • the generation of elevated platelet titers is useful in preparing a patient for subsequent cycles of radiation and/or chemotherapy treatment.
  • a patient entering a subsequent cycle of treatment with elevated platelet levels is better able to withstand the cytoreductive effects of the treatment.
  • the invention therefore, is effective to increase patient tolerance to a radiation and/or chemotherapeutic regimen relative to the patient tolerance for the regimen without administration of TPO according to the invention.
  • the method of the invention is also useful in mobilization therapy.
  • the peripheral blood progenitor cells are mobilized from the bone marrow to reduce or eliminate neutropenia and/or thrombocytopenia.
  • TPO is administered as a single or low-multiple daily dose to mobilize peripheral blood progenitor cells.
  • the mammal is a human patient having or at risk for thrombocytpenia, for example, as a result of radiation and/or chemotherapy or liver disease.
  • TPO is administered to the patient prior to or concurrent with radiation or chemotherapy treatment.
  • TPO may also be administered subsequent to radiation or chemotherapy treatment to restore platelet blood titer in conjunction with the prior or concurrent administration noted above.
  • the TPO may also be administered together with another cytokine, e.g. G-CSF, IL-3, IL-6, GM-CSF, etc.
  • the progenitor cells which are mobilized by the method of the invention may be collected by standard leukapheresis, optionally frozen, and retransplanted into the patient after radiation and/or chemotherapy.
  • the additional cytokine is generally administered in an amount similar to the amount of TPO.
  • the TPO might be administered in an amount from about 0.1-10 microgram/kg alone or together with an additional cytokine in a similar amount.
  • cytokine e.g. G-CSF, IL-3, IL-6, GM-CSF, etc.
  • the progenitor cells which are mobilized by the method of the invention may be collected by standard leukapheresis, optionally frozen, and retransplanted into the patient after radiation and/or chemotherapy.
  • the additional cytokine is generally administered in an amount similar to the amount of TPO
  • TPO optionally in combination with another cytokine as discussed above, may be administered to a mammal including a human patient, for the purpose of mobilizing peripheral blood progenitor cells which may then be harvested by leukapheresis, optionally frozen and transplanted into a mammal having or at risk for thrombocytopenia.
  • the mammal or patient donating the heterologous bone marrow graft (progenitor cells) and the transplant recipient may be tissue-typed according to known procedures.
  • the TPO and other cytokine are generally administered in the amounts discussed above for autologous transplants.
  • Immunogenicity is reduced or eliminated due to less frequent dosing, including single doses, relative to continuous daily dosing known in the art.
  • Dosing is preferably intravenous.
  • hybrid regimen in which IV dosing is combined with SC dosing is also contemplated by this invention.
  • This initial dose of TPO might be followed by one or more SC doses of TPO after the treatment to maintain the platelet levels.
  • TPO parenteral or subcutaneous delivery of TPO
  • IV intravenous
  • SC subcutaneous
  • proteins e.g. insulin, hGH, BSA, and LHRH
  • a TPO e.g. recombinant human thrombopoietin (rhTPO)
  • rhTPO recombinant human thrombopoietin
  • rhTPO recombinant human thrombopoietin
  • Therapeutic serum concentrations can be achieved by delivering rhTPO to the lungs as a liquid or powder aerosol.
  • Solutions of TPO may be nebulized using conventional nebulizers and administered to a mammal or human patient through the nose or mouth as an aerosol.
  • TPO may also be dried to a powder, e.g. by spray-drying, and administered using a conventional dry powder inhaler.
  • a higher dose of rhTPO is required to achieve a similar therapeutic effect when given as an aerosol as compared to IV.
  • the dose by aerosol should be about 100-fold higher for aerosol administration as compared to IV administration.
  • a suitable dose range for aerosol administration is about 5-1000 microgram/kg, preferably 50-750 microgram/kg, as a single inhalation dose or as multiple inhalations on a single day or on multiple days, preferably 2-10 or 2-6, sequential or non-sequential days.
  • the method of the invention may be used with any radiation and/or chemotherapy regimen in which a mammal or human patient is having or is at risk of thrombocytopenia.
  • the method may be used with conventional chemotherapeutic agents used in conventional amounts including, but not limited to asparaginase, bleomycin, calcium leucovorin, carmustine, carboplatin, cisplatin, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin, epirubicin, etoposide, fluorouracil, fluoxymesterone, flutamide, hexamethylmelamine, hydroxyurea, ifosfamide, leuprolide, levamisole, leuprolide depot, lomustine, mechlorethamine, melphalan, mercaptopurine, methotrexate, methyl-CCNU (semustine), methylprednisolone,
  • chemotherapeutic agent metabolites e.g. ifosfamide metabolites.
  • the chemotherapeutic agents are given in combination to maximize tumor cell kill with minimal or at least acceptable toxicity to the mammal or patient.
  • the method of the invention further reduces this toxicity.
  • Suitable non-limiting chemotherapy regimen with which the method of the invention may be used are listed below using conventional acronyms and indicating specific tumors/cancers for which the regimen is contemplated.
  • chemotherapeutic agents may be administered in conventional amounts and according to conventional treatment times and regimen. See, for example, "The Cerenex Handbook", Robert S. Benjamin, Ed., Cerenex Pharmaceuticals, Research Triangle Park, N.C. (1993); “Combination Cancer Chemotherapy Regimens", Roger W. Anderson and William J. Dana, Eds., Laderley Laboratories (1991). Any cytoreductive regimen which induces thrombocytopenia is considered to within the scope of the invention, however.
  • CMFP - cyclophosphamide methotrexate, fluorouracil, prednisolone
  • FMV - fluorouracil methyl-CCNU, vincristine Gastric Cancer
  • FAM - fluorouracil doxorubicin, mitomycin C
  • MVAC - methotrexate vinblastine, doxorubicin, cisplatin
  • MV - mitoxantrone etoposide Acute Non-Lymphocytic Leukemia (A.N.L.L.)
  • MOPP mechlorethamine, vincristine, procarbazine, prednisone
  • MVPP mechlorethamine, vinblastine, procarbazine, prednisone
  • VDP - vinblastine dacarbazine, cisplatin Multiple Myeloma AC - doxorubicin, carmustine
  • MAID - mesna, doxorubicin, ifosfamide, dacarbazine Selected regimens particularly associated with thrombocytopenia which may be treated with the method of the invention include:
  • BCNU carmustine Glioblastoma procarbazine/CCNU/vincristine Brain cyclophaphamide/etoposide +/- Salvage Hodgkin's disease, breast, cisplatin or carboplatin ovary, non-Hodgkin's lymphoma, head and neck, lung, myeloma, sarcoma dexamethasone/THDAC/cisplatin salvage non-Hodgkin's lymphoma mesna ifosfamide/doxorubicin/+ DTIC Soft tissue sarcoma cyclophophamide/vincristine/DTIC Soft tissue sarcoma doxorubicin
  • the single or low multiple dose of the invention may be given prior to the first treatment time of radiation and/or chemotherapeutic agent in a treatment cycle, during or concurrent with a treatment time in a treatment cycle or following one or more individual treatment times of radiation or chemotherapeutic agent in a treatment cycle.
  • a cycle may constitute a single treatment time of radiation or chemotherapeutic agent.
  • the single or low-multiple dose of TPO would be administered before, during or after this treatment time.
  • the cycle may constitute multiple treatment times, for example 2-10 or more treatment times, of radiation or chemotherapeutic agent.
  • the invention contemplates administering TPO before, during or after any one treatment time or before, during or after each individual treatment time.
  • the cycle may constitute three treatments with a chemotherapeutic agent.
  • TPO might be administered before each of the three treatment times or might be administered after each of the three treatment times.
  • the invention also includes administration of a single daily dose of TPO before the first treatment time of the chemotherapeutic agent in the cycle and after the last treatment time in the cycle.
  • the mammal receives at least one treatment cycle of radiation and/or chemotherapeutic agent, where the treatment cycle has a first treatment time TQ and a last treatment time Tp for administering radiation and/or chemotherapeutic agent.
  • the dose of TPO is preferably administered at TQ plus or minus 24 hours, more preferably TQ plus or minus 10 hours, still more preferably To plus or minus 6 hours, and most preferably T Q plus or minus 2 hours.
  • the dose is administered at T Q or prior to T Q , but not more than seven days prior to TQ, preferably not more than one day prior to TQ.
  • T Q Tp.
  • the dose is administered prior to Tp, but not more than seven days prior to Tp.
  • TPO may also be administered after Tp.
  • the dose is preferably administered not more than 24 hours after Tp.
  • the mammal or patient may, of course, receive multiple treatment cycles, generally 2-6 cycles, but as many cycles as is medically necessary to reduce the size of or to completely irradicate a cancer or tumor.
  • a tumor is reduced in size relative to the size of the tumor prior to radiation and/or chemotherapy treatment, and then surgery is utilized to remove the remaining malignant tissue of the tumor.
  • the method of the invention may be used in these regimen as well.
  • the invention also includes co-administering a therapeutically effective amount of a cytokine, a colony stimulating factor and an interleukin, generally after administration of the TPO dose, preferably after administration of the last TPO dose in a treatment cycle.
  • the cytokine is preferably KL, LIF, G-CSF, GM-CSF, M-CSF, EPO, FLT-3, IL-1, IL-2, IL-3, IL-5, IL-6, IL-7, IL-8, IL-9 or IL- 1 1, in particular, G-CSF or GM-CSF.
  • the single or low multiple administration regimen of the present invention is effective at daily dosage rates on the order of about 0.1 to 50, preferably about 0.1 to 10, more preferably about 1 to 5, or preferably about 1 to 3 microgram/kg body weight of the patient.
  • preferred would be the total administration of about 2 ⁇ 1.5 microgram/kg of body weight.
  • low-multiple dosing preferred would be the administration of from about 0.5 to 1.5 microgram kg body weight per dose.
  • the above dosages are predicated on preferred intravenous administration. In administration via the subcutaneous route, the total amount administered would be in the range of about one to three times the amount administered via the intravenous route, preferably about two times. Further, the doses for administration via the lung are higher as noted above. Specific therapeutically effective dosages for individual patients may be determined by conventional methods.
  • the method of the invention also preferably provides a dose of TPO sufficient to maintain a blood TPO level in the mammal of 35 x 10 ⁇ 12 M or greater during the radiation and/or chemotherapy -12 treatment cycle.
  • the dose is sufficient to maintain a blood TPO level of 100 x 10 M or greater, more preferably about 35 x 10" 12 M to about 3500 x 10" 12 M during the treatment cycle.
  • the optimal dosage rate and regimen will be determined by the attending physician taking into consideration various factors known to modify the action of drugs including severity and type of disease, body weight, sex, diet, time and route of administration, other medications and other relevant clinical factors.
  • thrombopoietin thrombopoietin to a patient has been found to be therapeutically effective for the treatment of thrombocytopenia, it can be appreciated that a low-multiple (daily) regimen may be employed. It has been found that a single dose stimulates the onset of therapeutic response, and although multiple dosing is contemplated herein, termination of dosing after a single or low-multiple administration is independent of therapeutic response.
  • the biologically active thrombopoietin materials of the present invention can be administered, in accord herewith, in various routes including via the nose or lung, subcutaneously, and preferably intravenously. In all events, depending upon the route of administration, the biologically active thrombopoietin materials of the present invention are preferably administered in combination with an appropriate pharmaceutically acceptable carrier or excipient.
  • the therapeutic composition should be pyrogen-free and in a parenterally acceptable solution having due regard for physiological pH isotonicity and stability. These conditions are generally well known and accepted to those of skill in the appropriate art.
  • dosage formulations of the materials of the present invention are prepared for storage or administration by mixing the compound having the desired degree of purity with physiologically acceptable carriers, excipients and/or stabilizers.
  • Such materials are non-toxic to the recipients at the dosages and concentrations employed and include buffers such as phosphate, citrate, acetate and other organic acid salts; antioxidants such as ascorbic acid; low molecular weight peptides such as polyarginine, proteins such as serum albumen, gelatin or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidinone; amino acids such as glycine, glutamic acid, aspartic acid or arginine; monosaccharides, disaccharides and other carbohydrates including cellulose or its derivatives, glucose, mannose or dextrins; chelating agents such as EDTA; sugar alcohol such as mannitol or sorbitol; counter-ions such as sodium; and/or non-ionic surfactants such as TWEEN, PLURONICS or poly
  • the biologically active thrombopoietin materials hereof can be administered as the free acid or base form or as a pharmaceutically acceptable salt and are compounded with a physiologically acceptable vehicle, carrier, excipient, binder, preservative, stabilizer, flavoring agent, etc. as called for by accepted pharmaceutical practice.
  • atenle compositions for injection can be formulated according to conventional pharmaceutical or pharmacological practice. For example, dissolution or suspension of the active material in a vehicle such as water or naturally occurring vegetable oil like sesame, peanut, or cottonseed oil or a synthetic fatty vehicle like ethyl oleate or the like may be desired.
  • buffers, preservatives, anti-oxidants and the like can be incorporated according to accepted pharmaceutical practice.
  • the biologically active thrombopoietin materials of the present invention may be employed alone or administered in combination with other cytokines, hematapoietins, interleukins, growth factors, or antibodies in the treatment of the above identified disorders and conditions marked by thrombocytopenia.
  • the present active materials may be employed in combination with other protein or peptide having thrombopoietic activity including: G-CSF, GM-CSF, LIF, M-CSF, IL-2, IL-3, erythropoietin (EPO), Kit ligand, IL-6, IL-11, FLT-3 ligand, and so forth.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (e.g. poly(2-hydroxyethyl-methacrylate) as described by Langer et al. J. Biomed. Mater. Res., 15: 167- 277 (1981) and Langer, Chem. Tec, 72:98-105 (1982) or poly(vinylalcohol)), polylactides (U.S. Patent No.
  • Sustained-release thrombopoietic protein compositions also include liposomally entrapped megakaryocytopoietic protein.
  • Liposomes containing megakaryocytopoietic protein are prepared by methods knew per se: DE, 3,218,121; Epstein et al.Proc. Natl. Acad. Sci. USA, 52:3688-3698 (1985); Hwang et al.Proc. Natl. Acad. Sci. USA, 77:4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EP 143,949; EP 142,641 ; Japanese patent application 83- 118008; U.S. Patent Nos.4,485,045 and
  • the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. % cholesterol, the selected proportion being adjusted for the optimal megakaryocytopoietic protein therapy.
  • a type of covalent modification of TPO or mpl ligand comprises linking the TPO polypeptide to one of a variety of nonproteinaceous polymers, e.g. polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144;
  • TPO polypeptides covalently linked to the forgoing polymers are referred to herein as pegylated TPO.
  • TPO variant some screening of the recovered TPO variant will be needed to select the optimal variant for binding to a mpl and having the immunological and/or biological activity defined above.
  • a change in the immunological character of the TPO polypeptide is measured by a competitive-type immunoassay.
  • Human genomic DNA clones of the TPO gene were isolated by screening a human genomic library in 8-Geml2 with pR45, under low stringency conditions or under high stringency conditions with a fragment corresponding to the 3N half of human cDNA coding for the mpl ligand. Two overlapping lambda clones spanning 35 kb were isolated. Two overlapping fragments (BamHI and
  • the structure of the human gene is composed of 6 exons within 7 kb of genomic DNA. The boundaries of all exon/intron junctions are consistent with the consensus motif established for mammalian genes (Shapiro, M.B. et al, Nucl. Acids. Res. 75:7155 (1987)). Exon 1 and exon 2 contain
  • a second vector coding for the EPO homologous domain was generated the same but using different PCR primers to obtain the final construct called pRK5-tkneoEPO-D.
  • ⁇ ML332 Purification of ⁇ ML332 was conducted as described in Example 1. Briefly, 293- ⁇ ML332 conditioned media was applied to a BLUE-SEPHAROSE (Pharmacia) column that was subsequently washed with a buffer containing 2M urea, the column was eluted with a buffer containing 2M urea and 1 M NaCl. The BLUE-SEPHAROSE elution pool was then directly applied to a WGA-SEPHAROSE column, washed with 10 column volumes of buffer containing 2M urea and IM NaCl and eluted with the same buffer containing 0.5M N-acetyl-D-glucosamine.
  • the WGA-SEPHAROSE eluate was applied to a C4-HPLC column (Synchrom, Inc.) and eluted with a discontinuous propanol gradient.
  • C4-HPLC column Synchronization, Inc.
  • the purified 293-fhML332 migrates as a broad band in the 68-80 kDa region of the gel.
  • Purification of ⁇ ML 153 was also conducted as described in Example 1. Briefly, 293-rhML j 53 conditioned media was resolved on BLUE-SEPHAROSE as described for rhML332- The BLUE- SEPHAROSE eluate was applied directly to a w ⁇ p/-affinity column as described above.
  • R 1ML153 eluted from the m/./-affinity column was purified to homogeneity using a C4-HPLC column run under the same conditions used for rhML332- By SDS-PAGE the purified rhML ⁇ resolves into 20 major and 2 minor bands with Mr of ⁇ 18,000-22,000.
  • the expression vectors used to transfect CHO cells are designated: pSVI5.ID.LL.MLORF (full length of TPO332), and pSVI5.ID.LL.MLEPO-D (truncated or TPO 153).
  • cDNA corresponding to the entire open reading frame of TPO was obtained by PCR.
  • the PCR product was purified and cloned between two restriction sites (Clal and Sail) of the plasmid pSVI5.ID.LL to obtain the vector pSVI5.ID.LL.MLORF.
  • a second construct corresponding to the EPO homologous domain was generated the same way but using a different reverse primer (EPOD.Sal).
  • the final construct for the vector coding for the EPO homologous domain of TPO is called pSVI5.ID.LL.MLEPO-D.
  • These two constructs were linearized with Notl and transfected into Chinese Hamster Ovary cells (CHO-DP12 cells, EP 307,247 published 15 March 1989) by electroporation. 10 7 cells were electroporated in a BRL electroporation apparatus (350 Volts, 330 mF, low capacitance) in the presence of 10, 25 or 50 mg of DNA as described (Andreason, G.L. J.Tissue Cult. Meth, 75:56 (1993)).
  • DHFR selective media High glucose DMEM-F12 50:50 without glycine, 2mM glutamine, 2-5% dialyzed fetal calf serum. 10 to 15 days later individual colonies were transferred to 96 well plates and allowed to grow to confluency. Expression of ML 1 53 or ML332 in the conditioned media from these clones was assessed using the
  • BalF3-mpl proliferation assay The process for purifying and isolating TPO from harvested CHO cell culture fluid is described in Example 2. Briefly, harvested cell culture fluid (HCCF) is applied to a BLUE-SEPHAROSE column (Pharmacia) at a ratio of approximately 100L of HCCF per liter of resin. The column is then washed with 3 to 5 column volumes of buffer followed by 3 to 5 column volumes of a buffer containing 2.0M urea. TPO is then eluted with 3 to 5 column volumes of buffer containing both 2.0M urea and 1.0M NaCl.
  • HCCF harvested cell culture fluid
  • Pharmacia BLUE-SEPHAROSE column
  • the BLUE-SEPHAROSE eluate pool containing TPO is then applied to a wheat germ lectin SEPHAROSE column (Pharmacia) equilibrated in the BLUE-SEPHAROSE eluting buffer at a ratio of from 8 to 16 ml of BLUE-SEPHAROSE eluate per ml of resin.
  • the column is then washed with 2 to 3 column volumes of equilibration buffer.
  • TPO is then eluted with 2 to 5 column volumes of a buffer containing 2.0M urea and 0.5M N-acetyl-D-glucosamine.
  • the wheat germ lectin eluate containing TPO is then acidified and C ⁇ Eg is added to a final concentration of 0.04%.
  • the resulting pool is applied to a C4 reversed phase column equilibrated in 01% TFA, 0.04% C ⁇ Eg at a load of approximately 0.2 to 0.5 mg protein per ml of resin.
  • the protein is eluted in a two phase linear gradient of acetonitrile containing 0.1% TFA and 0.04% C 12 E 8 and a pool is made on the basis of SDS-PAGE.
  • the C4 Pool is then diluted and diafiltered versus approximately 6 volumes of buffer on an AMICON YM or like ultrafiltration membrane having a 10,000 to 30,000 Dalton molecular weight cutoff.
  • the resulting diafiltrate may be then directly processed or further concentrated by ultrafiltration.
  • the diafiltrate/concentrate is usually adjusted to a final concentration of 0.01% TWEEN-80. All or a portion of the diafiltrate/concentrate equivalent to 2 to 5% of the calculated column volume is then applied to a SEPHACRYL S-300 HR column (Pharmacia) equilibrated in a buffer containing 0.01% TWEEN-80 and chromatographed.
  • TPO containing fractions which are free of aggregate and proteolytic degradation products are then pooled on the basis of SDS-PAGE.
  • the resulting pool is filtered and stored at 2-8°C.
  • E. coli TPO expression vectors Construction of E. coli TPO expression vectors is described in detail in Example 3. Briefly, plasmids pMP21 , pMP 151, pMP41 , pMP57 and pMP202 were all designed to express the first 155 amino acids of TPO downstream of a small leader which varies among the different constructs. The leaders provide primarily for high level translation initiation and rapid purification.
  • the plasmids pMP210-l, -T8, -21, 22, -24, -25 are designed to express the first 153 amino acids of TPO downstream of an initiation methionine and differ only in the codon usage for the first 6 amino acids of TPO, while the plasmid pMP251 is a derivative of pMP210-l in which the carboxy-terminal end of TPO is extended by two amino acids. All of the above plasmids will produce high levels of intracellular expression of TPO in E. coli upon induction of the tryptophan promoter (Yansure, D. G.
  • the plasmids pMPl and pMP172 are intermediates in the construction of the above TPO intracellular expression plasmids.
  • the above TPO expression plasmids were used to transform the E. coli using the CaCl 2 heat shock method (Mandel, M. et al. , J. Mol. Biol, 53: 159- 162, ( 1970)) and other procedures described in Example 3. Briefly, the transformed cells were grown first at 37°C until the optical density (600 nm) of the culture reached approximately 2-3. The culture was then diluted and, after growth with aeration, acid was added. The culture was then allowed to continue growing with aeration for another 15 hours after which time the cells were harvested by centrifugation.
  • Example 4 The isolation, purification and refolding procedures given below for production of biologically active, refolded human TPO or fragments thereof is described in Example 4 can be applied for the recovery of any TPO variant including N and C terminal extended forms.
  • Other procedures suitable for refolding recombinant or synthetic TPO can be found in the following patents: Builder et al, US 4,511,502; Jones et al., US 4,512,922; Olson, US 4,518,526 and Builder et al., US 4,620,948; for a general description of the recovery and refolding process for a variety of recombinant proteins expressed in an insoluble form in E. coli .
  • Thrombopoietic activity may be measured in various assays including the Ba/F3 mpl ligand assay.
  • CMK human leukemia megakaryoblastic cell line
  • DAMI megakaryoblastic cell line
  • Maturation of megakaryocytes from immature, largely non-DNA synthesizing cells, to morphologically identifiable megakaryocytes involves a process that includes appearance of cytoplasmic organelles, acquisition of membrane antigens (GPIIbIII a ), endoreplication and release of platelets as described in the background.
  • a lineage specific promoter (i.e. the mpl ligand) of megakaryocyte maturation would be expected to induce at least some of these changes in immature megakaryocytes leading to platelet release and alleviation of thrombocytopenia.
  • assays were designed to measure the emergence of these parameters in immature megakaryocyte cell lines, i.e., CMK and DAMI cells.
  • the CMK assay measures the appearance of a specific platelet marker, GPII
  • the DAMI assay measures endoreplication since increases in ploidy are hallmarks of mature megakaryocytes. Recognizable megakaryocytes have ploidy values of 2N, 4N, 8N, 16N, 32N, etc.
  • the in vivo mouse platelet rebound assay is useful in demonstrating that administration of the test compound (here the mpl ligand) results in elevation of platelet numbers. Two additional in vitro assays have been developed to measure TPO activity.
  • the first is a kinase receptor activation (KIRA) ELISA in which CHO cells are transfected with a mpl-Rse chimera and tyrosine phosphorylation of Rse is measured by ELISA after exposure of the mpl portion of the chimera to mpl ligand.
  • the second is a receptor based ELISA in which ELISA plate coated rabbit anti- human IgG captures human chimeric receptor mpl-lgG which binds the mpl ligand being assayed.
  • a biotinylated rabbit polyclonal antibody to mpl ligand TPO155 is used to detect bound mpl ligand which is measured using streptavidin-peroxidase.
  • the biologically active thrombopoietic protein may be used in a sterile pharmaceutical preparation or formulation to stimulate megakaryocytopoietic or thrombopoietic activity in patients suffering from thrombocytopenia due to impaired production, sequestration, or increased destruction of platelets.
  • thrombocytopenia-associated bone marrow hypoplasia e.g.
  • aplastic anemia following chemotherapy or bone marrow transplant may be effectively treated with the compounds of this invention as well as disorders such as disseminated intravascular coagulation (DIC), immune thrombocytopenia (including HIV-induced ITP and non HIV-induced ITP), chronic idiopathic thrombocytopenia, congenital thrombocytopenia, myelodysplasia, and thrombotic thrombocytopenia.
  • DIC disseminated intravascular coagulation
  • immune thrombocytopenia including HIV-induced ITP and non HIV-induced ITP
  • chronic idiopathic thrombocytopenia congenital thrombocytopenia, myelodysplasia
  • thrombotic thrombocytopenia include myeloproliferative thrombocytotic diseases and iron deficiency.
  • the method of the invention is also useful to treat mammals or human patients which have suffered from exposure to ionizing radiation sufficient to cause thrombocytopenia, for example, persons exposed to nuclear accidents such as the well-known accident which occurred at Chernobyl.
  • TPO is well tolerated by patients and this may justify the rapid administration of TPO within a few hours after a nuclear accident to all persons affected by radiation.
  • the TPO responsiveness of progenitor cells appears to be very large shortly after exposure of a person to radiation and/or chemotherapy, the method of the invention may also be used as a radioprotective procedure by administering TPO prophylactically to a person who will be exposed to ionizing radiation.
  • an emergency worker may be required to enter highly contaminated areas in cases of a major nuclear accident.
  • Administration of a prophylactic dose of TPO prior to exposure according to the dosing methods of the present invention will ensure that the worker has high levels of early multilineage progenitor cells in order to reduce the degree of thrombocytopenia induced by the exposure to radiation.
  • TPO thrombocytopoietic protein
  • Still other disorders usefully treated with the thrombopoietin proteins of this invention include defects or damage to platelets resulting from drugs, poisoning or activation on artificial surfaces.
  • the instant compounds may be employed to stimulate "shedding" or new "undamaged” platelets.
  • hmpII-R 5N GCTAGC TCT AGA CAG GGA AGG GAG CTG TAC ATG AGA 3N
  • prk5-Hwp/ was used as a template for the reaction in the presence of pfu DNA polymerase (Stratagene). Initial denaturation was for 7 min. at 94°C followed by 25 cycles of amplification (1 min. at 94°C, 1 min. at 55°C and 1 min. at 72°C). Final extension was for 15 min. at 72°C.
  • the PCR product was purified and cloned between the restriction sites Clal and Xbal of the plasmid pRK5tkneo, a pRK5 derived vector modified to express a neomycin resistance gene under the control of the thymidine kinase promote, to obtain the vector pRK5tkneo.ORF.
  • a second construct corresponding to the epo homologous domain was generated the same way but using Cla.FL.F as forward primer and the following reverse primer:
  • 392-rhML332 conditioned media was applied to a BLUE-SEPHAROSE (Pharmacia) column that was equilibrated in 10 mM sodium phosphate pH 7.4 (buffer A). The column was subsequently washed with 10 column volumes each of buffer A and buffer A containing 2M urea. The column was then eluted with buffer A containing 2M urea and IM NaCl. The BLUE-SEPHAROSE elution pool was then directly applied to a WGA-SEPHAROSE column equilibrated in buffer A.
  • the WGA- SEPHAROSE column was then washed with 10 column volumes of buffer A containing 2M urea and 1 M NaCl and eluted with the same buffer containing 0.5M N-acetyl-D-glucosamine.
  • the WGA- SEPHAROSE eluate was applied to a C4-HPLC column (Synchrom, Inc.) equilibrated in 0.1% TFA.
  • the C4-HPLC column was eluted with discontinuous propanol gradient (0-25%, 25-35%, 35-70%).
  • rhML332 was found to elute in the 28-30% propanol region of the gradient, by SDS-PAGE the purified rhML332 migrates as a broad band in the 68-8- kDa region of the gel.
  • 392-rhML ⁇ 53 conditioned media was resolved on BLUE-SEPHAROSE as described for rhML332-
  • the BLUE-SEPHAROSE eluate was applied directly to a mpl-affinity column as described above.
  • RI1ML153 eluted from the mpl-afRnity column was purified to homogeneity using a C4-HPLC column run under the same conditions as described for rhML332-
  • SDS-PAGE the purified rhML j 53 resolves into 2 major and 2 minor bands with Mr of -18,000-21,000.
  • a cDNA corresponding to the hTPO entire open reading frame was obtained by PCR using the oligonucleotide primes of the following Table.
  • PRK5-/WK/?/ 1 was used as template for the reaction in the presence of pfu DNA polymerase (Stratagene). Initial denaturation was for 7 min. at 94°C followed by 25 cycles of amplification (1 min. at 94°C, 1 min. at 55°C and 1 min. at 72°C). Final extension was for 15 min. at 72°C.
  • the PCR product was purified and cloned between the restriction sites Clal and Sail of the plasmid pSV15.ID.LL to obtain the vector pSV15.ID.LL.MLORF.
  • a second construct corresponding to the EPO homologous domain was generated the same way but using Cla.FL.F2 as forward primer and the following reverse primer: EPOD.Sal 5' AGT CGA CGT CGA CTC ACC TGA CGC AGA GGG TGG ACC 3' (SEQ ID NO:6).
  • the final construct is called pSV 15. ID.LL.MLEPO-D. The sequence of both constructs was verified.
  • the coding sequences for the full length and truncated ligand were introduced into the multiple cloning site of the CHO expression vector pSV15.ID.LL.
  • This vector contains the SV40 early promoter/enhancer region, a modified splice unit containing the mouse DHFR cDNA, a multiple cloning site for the introduction of the gene of interest (in this case the TPO sequences described) an SV40 polyadenylation signal and origin of replication and the beta-lactamase gene for plasmid selection and amplification in bacteria.
  • the host CHO (Chinese Hamster Ovary) cell line used for the expression of the TPO molecules described herein is known as CHO-DP12 (see EP 307,247 published 15 March 1989).
  • This mammalian cell line was clonally selected from a transfection of the parent line (CHO-K1 DUX-B11 (DHFR-)- obtained from Dr. Frank Lee of Stanford University with the permission of Dr. L. Chasin) with a vector expressing preproinsulin to obtain clones with reduced insulin requirements.
  • DHFR minus and clones can be selected for the presence of DHFR cDNA vector sequences by growth on medium devoid of nucleoside supplements (glycine, hypoxanthine, and thymidine). This selection system for stably expressing CHO cell lines is commonly used.
  • b. Transfection method electroporation
  • Suspension growing DP 12 cells were collected, washed one time in the medium described for resuspending the DNA and finally resuspended in the same medium at a concentration of 10 7 cells per 750 microliters. Aliquots of cells (750 microliters) and each linearized DNA mix were incubated together at room temperature for one hour and then transferred to a BRL electroporation chamber.
  • Each reaction mix was then electroporated in a standard BRL electroporation apparatus at 350 volts set at 330 micro F and low capacitance. After electroporation, the cells were allowed to sit in the apparatus for 5 minutes and then on ice for an additional 10 minute incubation period. The electroporated cells were transferred to 60mm cell culture dishes containing 5 ml of standard, complete growth medium for CHO cells (High glucose DMEM-F12 50:50 without glycine supplemented with IX GHT, 2mM glutamine, and 5% fetal calf serum) and grown overnight in a 5% CO2 cell culture incubator. c. Selection and screening method
  • DHFR selective medium Ham's DMEM-F12, 1 : lmedium described above supplemented with either 2% or 5% dialyzed fetal calf serum but devoid of glycine, hypoxanthine and thymidine this is the standard DHFR selection medium we use.
  • Cells from each 60mm dish were subsequently replated into 5/150 mm dishes. Cells were then incubated for 10 to 15 days( with one medium change) at 37 degrees/15% CO2 until clones began to appear and reached sizes amenable to transfer to 96 well dishes.
  • cell lines were transferred to 96 well dishes using sterile yellow tips on a pipettman set at 50 ml.
  • the cells were allowed to grow to confluency (usually 3-5 days) and then the trays were trypsinized and 2 copies of the original tray were reproduced. Two of these copies were short term stored in the freezer with cells in each well diluted into 50 microliter pf 10% FCS in DMSO. 5 day conditioned serum free medium samples were assayed from confluent wells in the third tray for TPO expression via the Ba/F cell based activity assay.
  • the highest expressing clones based on this assay were revived from storage and scaled up to 2 confluent 150mm T-flasks for transfer to the cell culture group for suspension adaptation, re-assay and banking.
  • telomere clones are expanded and plated in 10cm dishes at 4 concentrations of methotrexate (i.e 50 nM, 100 nM, 200 nM and 400 nM) at two or three cell numbers ( 10 5 , 5x10 5 , and 10 6 cells per dish). These cultures are then incubated at 37 degree/5% CO2 until clones are established and amenable to transfer to 96 well dishes for further assay.
  • methotrexate i.e 50 nM, 100 nM, 200 nM and 400 nM
  • Banked cells are thawed and the cell population is expanded by standard cell growth methods in either serum free or serum containing medium. After expansion to sufficient cell density, cells are washed to remove spent cell culture media. Cells are then cultured by any standard method including; batch, fed-batch or continuous culture at 25-40°C, neutral pH, with a dissolved O2 content of at least
  • the TPO is then eluted with 3 to 5 column volumes of 0.01 M Na phosphate pH 7.4, 2.0M urea, l.OM NaCl.
  • the BLUE-SEPHAROSE pool containing TPO is then applied to a wheat germ lectin SEPHAROSE 6 MB column (Pharmacia) equilibrated in 0.01 M Na phosphate pH 7.4, 2.0M urea, and l.OM NaCI at a ratio of from 8 to 16 ml of BLUE-SEPHAROSE pool per ml of resin at flow rate of approximately 50 ml/hr/cm 2 .
  • the column is then washed with 2 to 3 column volumes of equilibration buffer.
  • the TPO is then eluted with 2 to 5 column volumes of 0.01 M Na phosphate pH 7.4, 2.0M urea, 0.5 M N-acetyl-D-glucosamine.
  • the wheat germ lectin pool is then adjusted to a final concentration of 0.04%C j 2Eg and 0.1% trifluroacetic acid (TFA).
  • TFA trifluroacetic acid
  • the resulting pool is applied to a C4 reverse phase column (Vydac 214TP1022) equilibrated in 0.1% TFA, 0.04% C 12 E 8 at a load of approximately 0.2 to 0.5 mg protein per ml of resin at a flow rate of 157 ml/hr/cm 2 .
  • the protein is eluted in a two phase linear gradient of acetonitrile containing 0.1% TFA, 0.04% C ⁇ Eg.
  • the first phase is composed of a linear gradient from O to 30% acetonitrile in 15 minutes
  • the second phase is composed of a linear gradient from 30 to 60% acetonitrile in 60 minutes.
  • the TPO elutes at approximately 50% acetonitrile.
  • a pool is made on the basis of SDS-PAGE.
  • the C4 pool is then diluted with 2 volumes of 0.01 M Na phosphate pH 7.4, 0.15 M NaCI and diafiltered versus approximately 6 volumes of 0.01 M Na phosphate pH 7.4, 0.15 M NaCI on an AMICOM YM or like ultrafiltration membrane having a 10,000 to 30,000 Dalton molecular weight cut-off.
  • the resulting diafiltrate may be then directly processed or further concentrated by ultrafiltration.
  • the diafiltrate/concentrate is adjusted to a final concentration of 0.01% TWEEN-80.
  • All or a portion of the diafiltrate/concentrate equivalent to 2 to 5% of the calculated column volume is then applied to a SEPHACRYL S-300 HR column (Pharmacia) equilibrated in 0.01 M Na phosphate pH 7.4, 0.15M NaCI, 0.01% TWEEN-80 and chromatographed at a flow rate of approximately 17 ml/hr/cm 2 .
  • the TPO containing fractions which are free of aggregate and proteolytic degradation products are pooled on the basis of SDS-PAGE. The resulting pool is filtered on a 0.22 micron filter, MILLEX-GV or like, and stored at 2-8°C.
  • EXAMPLE 3 Transformation and Induction of TPO Protein Synthesis In E. coli 1. Construction of E. coli TPO expression vectors
  • the plasmids pMP21, pMP151, pMP41, pMP57 and pMP202 are all designed to express the first 155 amino acids of TPO downstream of a small leader which varies among the different constructs.
  • the leaders provide primarily for high level translation initiation and rapid purification.
  • the plasmids pMP210-l, -T8, -21, -22, -24, -25 are designed to express the first 153 amino acids of TPO downstream of an initiation methionine and differ only in the codon usage for the first 6 amino acids of TPO, while the plasmid pMP251 is a derivative of pMP210-l in which the carboxy terminal end of TPO is extended by two amino acids.
  • All of the above plasmids will produce high levels of intracellular expression of TPO in E. coli upon induction of the tryptophan promoter (Yansura, D. G. et. al. Methods in Enzymology (Goeddel, D. V., Ed.) 755:54-60, Academic Press, San Diego (1990)).
  • the plasmids pMPl and pMP172 are intermediates in the construction of the above TPO intracellular expression plasmids.
  • Plasmid pMPl is a secretion vector for the first 155 amino acids of TPO, and was constructed by ligating together 5 fragments of DNA. The first of these was the vector pPho21 in which the small Mlul-BamHI fragment had been removed.
  • pPho21 is a derivative of phGHl (Chang, C. N. et. al, Gene 55:189-196 (1987) in which the human growth hormone gene has been replaced with the E. coli phoA gene, and a Mlul restriction site has been engineered into the coding sequence for the STII signal sequence at amino acids 20-21.
  • the fourth was a 152 base pair Pstl-Haelll fragment from pRK5hmpII encoding amino acids 104-155 of TPO.
  • the last was a 412 base pair Stul-BamHI fragment from pdhl08 containing the lambda to transcriptional terminator as previously described (Scholtissek, S. et. al., NAR 75:3185 (1987)).
  • the plasmid pMP21 is designed to express the first 155 amino acids of TPO with the aid of a 13 amino acid leader comprising part of the STII signal sequence. It was constructed by ligating together three (3) DNA fragments, the first of these being the vector pVEG31 in which the small Xbal-Sphl fragment had been removed.
  • the vector pVEG31 is a derivative of pHGH207-l (de Boer, H. A. et. al., in Promoter Structure and Function (Rodriguez, R. L. and Chamberlain, M.
  • the second part in the ligation was a synthetic DNA duplex with the following sequence:
  • the last piece was a 1072 base pair Mlul-Sphl fragment from pMPl encoding 155 amino acids of TPO.
  • Plasmid pMPl 51 The plasmid pMP151 is designed to express the first 155 amino acids of TPO downstream of a leader comprising 7 amino acids of the STII signal sequence, 8 histidines, and a factor Xa cleavage site. pMP151 was constructed by ligating together three DNA fragments, the first of these being the previously described vector pVEG31 from which the small Xbal-Sphl fragment had been removed. The second was a synthetic DNA duplex with the following sequence: 5'-CTAGAATTATGAAAAAGAATATCGCATTTCATCACCATCACCATCACCATCACATCGAA GGTCGTAGCC (SEQ ID NO:l 1)
  • the plasmid pMPl 1 is identical to pMPl with the exception of a few codon changes in the STII signal sequence( this fragment can be obtained from pMPl).
  • the plasmid pMP202 is very similar to the expression vector pMPl 51 with the exception that the factor Xa cleavage site in the leader has been replaced with a thrombin cleavage site. As shown in Fig. 36, pMP202 was constructed by ligating together three DNA fragments. The first of these was the previously described pVEG31 in which the small Xbal-Sphl fragment had been removed. The second was a synthetic DNA duplex with the following sequence:
  • CCACGTAGCC (SEQ ID NO: 13) TTAATACTTTTTCTTATAGCGTAAAGTAGTGGTAGTGGTAGTGTAGCTTGGTGCA
  • the last piece was a 1064 base pair Bgll-Sphl fragment from the previously described plasmid pMPl l.
  • Plasmid pMP 172 is a secretion vector for the first 153 amino acids of TPO, and is an intermediate for the construction of pMP210.
  • pMP 172 was prepared by ligating together three DNA fragments, the first of which was the vector pLS321amB in which the small EcoRI-Hindl section had been removed. The second was a 946 base pair EcoRI-Hgal fragment from the previously described plasmid pMPl 1. The last piece was a synthetic DNA duplex with the following sequence: 5'-TCCACCCTCTGCGTCAGGT (SEQ ID NO: 15) GGAGACGCAGTCCATCGA-5' (SEQ ID NO: 16) (f) Plasmid pMP210
  • the plasmid pMP210 is designed to express the first 153 amino acids of TPO after a translational initiation methionine.
  • This plasmid was actually made as a bank of plasmids in which the first 6 codons of TPO were randomized in the third position of each codon, and was constructed by the ligation of three DNA fragments. The first of these was the previously described vector pVEG31 in which the small Xbal-Sphl fragment had been removed. The second was a synthetic DNA duplex shown below treated first with DNA polymerase (Klenow) followed by digestion with Xbal and Hinl, and encoding the initiation methionine and the randomized first 6 codons of TPO. 5'-GCAGCAGTTCTAGAATTATGTCNCCNGCNCCNCCNGCNTGTGACCTCCGAACACTGGAG GCTGTTCTCAGTAAA (SEQ ID NO: 17)
  • the third was a 890 base pair Hinfl-Sphl fragment from pMP172 encoding amino acids 19-153 of TPO.
  • Plasmid pMP41 The plasmid pMP41 is designed to express the first 155 amino acids of TPO fused to a leader consisting of 7 amino acids of the STII signal sequence following by a factor Xa cleavage site.
  • the plasmid was constructed by ligating together three pieces of DNA, the first of which was the previously described vector pVEG31 in which the small Xbal-Sphl fragment had been removed. The second was the following synthetic DNA duplex: 5'-CTAGAATTATGAAAAAGAATATCGCATTTATCGAAGGTCGTAGCC (SEQ ID NO: 19) TTAATACTTTTTCTTATAGCGTAAATAGCTTCCAGCAT-5N (SEQ ID NO:20)
  • Plasmid pMP57 expresses the first 155 amino acids of TPO downstream of a leader consisting of 9 amino acids of the Stll signal sequence and the dibasic site Lys- Arg. This dibasic site provides for a means of removing the leader with the protease ArgC.
  • This plasmid was constructed by ligating together three DNA pieces. The first of these was the previously described vector pVEG31 in which the small Xbal-Sphl fragment had been removed. The second was the following synthetic DNA duplex:
  • Plasmid pMP251 The plasmid pMP251 is a derivative of pMP210-l in which two additional amino acids of TPO are included on the carboxy terminal end. This plasmid was constructed by ligating together two pieces of DNA, the first of these being the previously described pMP21 in which the small Xbal-Apal fragment had been removed. The second part of the ligation was a 316 base pair Xbal-Apal fragment from pMP210-1. 2. Transformation and Induction of E. co/i with TPO expression vectors
  • the above TPO expression plasmids were used to transform the E. coli strain 44C6 (w3110 tonA ⁇ rpoHts Ion ⁇ cip ⁇ galE) using the CaCl 2 heat shock method (Mandel, M. et al, J. Mol. Biol, 55:159-162, (1970)).
  • the transformed cells were grown first at 37°C in LB media containing 50 pg/ml carbenicillin until the optical density (600nm) of the culture reached approximately 2-3.
  • the LB culture was then diluted 20x into M9 media containing 0.49% casamino acids (w/v) and 50 pg/ml carbenicillin.
  • indole-3 -acrylic acid was added to a final concentration of 50 llg/ml. The culture was then allowed to continue growing at 30°C with aeration for another 15 hours at which time the cells were harvested by centrifugation.
  • the washed cell pellet is again resuspended in 1 L cell disruption buffer with the Polytron homogenizer and the cell suspension is passed through an LH CELL DISRUPTER (LH Inceltech, Inc.) or through a MICROFLUIDIZER (Microfluidics
  • the suspension is centrifuged at 5,000 x g for 30 min. and resuspended and centrifuged a second time to make a washed retractile body pellet.
  • the washed pellet is used immediately or stored frozen at -70°C
  • TPO protein eluted with 20 mM Na phosphate, pH 6.0, with 10 mM DTT fractions containing monomeric, denatured TPO protein eluting between 160 and 200 ml are pooled.
  • the TPO protein is further purified on a semi-preparative C4 reversed phase column (2 x 20 cm VYDAC). The sample is applied at 5 ml min. to a column equilibrated in 0.1% TFA(trifluoroacetic acid) with 30% acetonitrile. The protein is eluted with a linear gradient of acetonitrile (30-60% in 60 min.). The purified reduced protein elutes at approximately 50% acetonitrile. This material is used for refolding to obtain biologically active TPO variant. 3. Generation of biologically active TPO (Met" 1 1-153)
  • the refolding buffer is gently stirred at 4°C for 12-48 hr to effect maximal refolding yields of the correct disulfide-bonded form of TPO (see below).
  • the solution is then acidified with TFA to a final concentration of 0.2%, filtered through a 0.45 or 0.22 micron filter, and 1 10 volume of acetonitrile added. This solution is then pumped directly onto a C4 reversed phase column and the purified, refolded TPO (Met" 1 1-153) eluted with the same gradient program as above.
  • TPO Refolded, biologically active TPO is eluted at approximately 45% acetonitrile under these conditions. Improper disulfide-bonded versions of TPO are eluted earlier.
  • the final purified TPO (Met" 1 1-153) is greater than 95% pure as assessed by SDS gels and analytical C4 reversed phase chromatography. For animal studies, the C4 purified material was dialyzed into physiologically compatible buffers. Isotonic buffers (10 mM Na acetate, pH 5.5, 10 mM Na succinate, pH 5.5 or 10 mM Na phosphate, pH 7.4) containing 150 mM NaCI and 0.01% TWEEN-80 were utilized.
  • CHAPS CHAPS family
  • CHAPS CHAPS
  • Sodium chloride was required for best yields, with the optimal levels between 0.1 M and 0.5M.
  • EDTA 1-5 mM
  • Glycerol concentrations of greater than 15% produced the optimal refolding conditions.
  • Organic solvents e.g. ethanol, acetonitrile, methanol
  • Tris and phosphate buffers were generally useful.
  • Incubation at 4°C also produced higher levels of properly folded TPO.
  • Refolding yields of 40-60% are typical for preparations of TPO that have been purified through the first C4 step. Active material can be obtained when less pure preparations (e.g. directly after the Superdex 200 column or after the initial refractile body extraction) although the yields are less due to extensive precipitation and interference of non-TPO proteins during the TPO refolding process.
  • TPO (Met" 1 1-153) contains 4 cysteine residues
  • version 1 disulfides between cysteine residues 1 -4 and 2-3
  • version 2 disulfides between cysteine residues 1-2
  • 3-4 version 3 disulfides between cysteine residues 1-3 and 2-4.
  • C4 reversed phase chromatography Only one of these peaks had significant biological activity as determined using the Ba/F3 assay. Subsequently, the refolding conditions were optimized to yield preferentially that version.
  • the disulfide pattern for the biologically active TPO has been determined to be 1-4 and 2-3 by mass spectrometry and protein sequencing (i.e. version 1). Aliquots of the various C4-resolved peaks (5-10 nmoles) were digested with trypsin (1:25 mole ratio of trypsin to protein). The digestion mixture was analyzed by matrix assisted laser desorption mass spectrometry before and after reduction with DTT. After reduction, masses corresponding to most of the larger tryptic peptides of TPO were detected. In the un-reduced samples, some of these masses were missing and new masses were observed.
  • rmTPO murine thrombopoietin
  • CBC complete blood count
  • Treatment Regimens Experiment 1 In order to determine the response to recombinant murine thrombopoietin
  • rmTP0335aa in animals rendered thrombocytopenic, groups of animals were treated for 1, 2, 4, or 8 consecutive days with 0.1 microgram/day (5 microgram/kg/day approx.). Treatment with rmTPO (335aa) was started 24 hours after the initiation of the model and was given as a daily 100 microliter subcutaneous injection.
  • Experiment 2 In order to determine the nature of the dose-response relationship for rmTPO(335) in this model, animals were given a single injection of rmTPO (335) 24 hours after the initiation of the model. Groups of animals received one of 0.01, 0.03, 0.1 or 0.3 microgram of rmTPO (335) as a single 100 microliter subcutaneous injection. In order to compare two routes of administration, a contemporaneous experiment used 4 groups of animals receiving identical doses of rmTPO (335) but via an intravenous route (lateral tail vein).
  • Experiment 1 A single dose of 0.1 microgram rmTPO(335) given 24 hours after the initiation of the model accelerated the recovery of platelet numbers in this murine model.
  • This single administration of rmTPO(335) elevated the nadir of the response from 196xl0 3 ⁇ 33xl0 3 /microliter on day 10 to 434x10 3 ⁇ 7xl0 3 /microliter on day 7.
  • the initial rate of decline in the platelet numbers remained unchanged but the recovery phase was much more rapid with platelet numbers returning to normal by day 14 as opposed to day 21 in the control group.
  • Some further improvement in the rate of recovery was seen by giving 0.1 microgram/day on day 1 and day 2 but this was marginal.
  • EXAMPLE 7A A Phase I Study To Determine The Safety, Tolerance, Pharmacokinetics And Pharmacodynamics Of Recombinant Human Thrombopoietin (rhTPO) In Subjects With Sarcoma Receiving Adriamycin And Ifosfamide Treatment Plan
  • rhTPO This was a single-center, open-label, dose-escalation, study of single and multiple IV doses of rhTPO with major safety endpoints.
  • rhTPO was administered to subjects with histologically diagnosed sarcoma to determine whether its administration helped to prevent, delay, ameliorate, or shorten the duration of the known thrombocytopenic effects of doxorubicin and ifosfamide.
  • 71 subjects have been enrolled on to this study which addresses the safety and the activity, of various rhTPO dosing schedule in conjuction with G-CSF and GM-CSF.
  • the study begins with a 21-day pre-chemotherapy cycle (cycle 0) to assess safety, activity, and pharmacokinetics in patients with cancer.
  • the data indicates a dose-dependent increase in peripheral blood platelet counts and bone marrow megakaryocytes in response to either a single or multiple IV doses of rhTPO, with doses ranging from 0.3-3.6 mg/kg.
  • rhTPO dosing regimens in which all of the rhTPO was administered after the completion of chemotherapy were safe, but demonstrated only modest activity.
  • One patient with a normal platelet count developed an uncomplicated deep venous thrombosis in the leg which resolved with conservative therapy, and neutralizing antibodies have not been observed.
  • Subjects had either metastatic or unresectable sarcoma.
  • Cycle 0 rhTPO on Day 0
  • Cycle 1 no rhTPO Cycle 2+: rhTPO on Day 4 Arm B
  • Cycle 0 rhTPO on Days 0 and 3
  • Cycle 1 no rhTPO Cycle 2+: rhTPO on Days 4 and 7 Arm C
  • Cycle 1 no rhTPO Cycle 2+: daily rhTPO on Days 4 through 10, or until the post-nadir platelet count is 3 100,000/ ⁇ L Arm D
  • Cycle 2+ rhTPO as per Arm D regimen, but with GM-CSF replacing G-CSF Cycle 0: This phase of the study evaluates safety, pharmacokinetics, and pharmacodynamics of single and multiple dosing.
  • Subjects in Arm A received a single IV injection of rhTPO (Day 0).
  • Subjects in Arm B received an IV injection of rhTPO on Days 0 and 3.
  • Cycle 1 Subjects received doxorubicin and ifosfamide. There was no rhTPO administered during Cycle 1. This provides a control cycle for comparison of each subject's response to subsequent cycles with rhTPO. This control cycle also clarifies chemotherapy-related adverse events in the absence of rhTPO dosing.
  • Cycle 2+ Subjects received doxorubicin and ifosfamide. Subjects in Arm A received a single
  • IV injection of rhTPO (Day 4).
  • Subjects in Arm B received IV injections of rhTPO on Days 4 and 7.
  • Subjects in Arm C received up to seven daily IV injections of rhTPO on Days 4-10, or until the post-nadir platelet count was greater than or equal to 100,000/ ⁇ L.
  • Subjects in Arm D received IV injections of rhTPO on Day -1 (1 day prior to chemotherapy) and on Days 4 and 7.
  • GM-CSF replaced G-CSF in Arm E.
  • GM-CSF was administered in Cycles 1 and 2, and in subsequent cycles where there is proven benefit.
  • Subjects in Arm E received rhTPO according to the Arm D regimen.
  • Doxorubicin was obtained from commercial sources and stored and administered in accordance with the manufacturer's guidelines. Doxorubicin (a total of 90 mg/m 2 ) was given as a continuous infusion for the first 3 days of each chemotherapy cycle (30 mg/m 2 daily for 3 days, (Days 0-2).
  • Ifosfamide was obtained from commercial sources and stored and administered in accordance with the manufacturer's guidelines. Ifosfamide (a total of 10 g/m 2 ) was given as four separate daily infusions during the first 4 days of each chemotherapy cycle (2.5 g/m 2 over 3 hours daily for 4 days, (Days 0-3).
  • Mesna was obtained from commercial sources and stored and administered in accordance with the manufacturer's guidelines. Mesna (500 mg/m 2 or 20% of the dose of ifosfamide) was administered rv over 3 hours with the initial dose of ifosfamide on Day 0 of each chemotherapy cycle. Mesna administration was maintained as a continuous IV infusion (1500 mg/m 2 /day for a total of 6 g/m 2 ) until 24 hours after the final ifosfamide administration of each chemotherapy cycle (Days 0-4).
  • G-CSF was obtained from commercial sources and stored and administered in accordance with the manufacturer's guidelines. G-CSF (5 ⁇ g/kg) was administered daily beginning on Day 4 of Cycle 1 and any subsequent chemotherapy cycles. Subjects were instructed to self-administer G-CSF. All injections of G-CSF should be administered at -8:00 p.m. Injections on the same day as rhTPO administration should be given -12 hours after rhTPO administration to help define any injection-related phenomena. G-CSF administration continued on a daily basis until the absolute neutrophil count was >1500/ ⁇ L for at least two consecutive measurements post-nadir.
  • GM-CSF GM-CSF is obtained from commercial sources and stored and administered in accordance with the manufacture's guidelines. GM-CSF (250 mg/m 2 ) is administered subcutaneously daily beginning on Day 4 of Cycle 1 and any subsequent chemotherapy cycles. Subjects are instructed to self-administer GM-CSF. All injections of GM-CSF should be administered at -8:00 p.m. Injections on the same day as rhTPO administrations should be given -12 hours after rhTPO administration to help define any injection-related phenomena. GM-CSF administration continued on a daily basis until the post-nadir absolute neutrophil count is >1500/ ⁇ L for at least two consecutive measurements. The results of this study are shown in Figures 14 - 19.
  • This study addresses the safety of rhTPO administered subcutaneously and the activity of rhTPO dosing in an every other day schedule.
  • the study begins with a 21 -day pre-chemotherapy cycle (cycle 0) to assess safety, activity, and pharmacokinetics in patients with cancer.
  • cycle 0 pre-chemotherapy cycle
  • 16 subjects have been enrolled on to this trial.
  • the data indicates a dose-dependent increase in peripheral blood platelet counts in cycle 0, though the platelet responses, and the pharmacokjinetics, are blunted when compared to similar doses of IV rhTPO.
  • mice were exposed to rhTPO in a nose-only inhalation (CH Technologies) with simultaneous measurement of breathing pattern (body plethysmograph) for a total duration of 60 minutes.
  • a PARI- IS2 nebulizer was used to aerosolize rhTPO at 22 psi and a flow rate of 5.1 LPM. Filter samples were taken to estimate the aerosol concentration for each exposure.
  • a vehicle control group and 3 different concentrations (0.05, 0.5, and 5.0 mg/ml rhTPO) were nebulized. The dose groups are expressed as the estimated amount per kg that deposited in the mouse lungs for each group.
  • Serum and blood were collected at -4, 3, 6, 8, 10, 14, 21, 30, and 43 days post aerosol inhalation for hematology (platelet counts) and measurement of serum anti-TPO antibodies.
  • mice Female (C57BLxCBA)Fl (BCBA) mice approximately 12 weeks old were bred at the Experimental Animal Facility of the Erasmus University, Rotterdam, The Netherlands, and maintained under SPF conditions. Housing, experiments and all other conditions were approved by an ethical committee in accordance with legal regulations in The Netherlands.
  • TBI TBI was given at day 0 using a two opposing 13 ?Cs sources (Gammacell 40, Atomic Energy of Canada, Ottawa, Canada) at a dose rate between 0.92 and 0.94 Gy/min. Doses used were 6 Gy for the single dose irradiation and a total dose of 9 Gy was split in three doses of 3 Gy given with 24 hour intervals. For each data point groups of three mice were killed. All parameters were collected for individual mice.
  • Test Drug Recombinant full length murine TPO produced by CHO cells (Genentech Inc., South San Francisco, CA was used throughout the experiments, diluted in PBS/0.01% TWEEN-20 and administered intraperitoneally in a volume of 0.5 ml.
  • Pharmacokinetic parameters were estimated after converting TCA-precipitable cpm/mL and fitting the concentration versus time data to a two-compartment model with first order absorption using nonlinear least-squares regression analysis (WIN-NONLIN; Statistical Consultants, Lexington, KY). Area under the concentration time curves (AUC), maximum concentration (Cmax), terminal half-lives (tl/2), and clearance (mL/hr/kg) were calculated using coefficients and exponents obtained from the model fits. Hematological examinations: After either-anesthesia the mice were bled by retro-orbital puncture and killed by cervical dislocation. Blood was collected in EDTA tubes.
  • Appropriate numbers of bone marrow cells were suspended in Dulbecco's modified Eagle's medium (Dulbecco's MEM) obtained from GIBCO (Life Technologies LTD, Paisley, Scotland) supplemented with the amino acids L-alanine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid and L-proline (Sigma), vitamin B12, biotin, Na-pyruvate, glucose, NaHC0 3 , and antibiotics (penicillin and streptomycin) at an osmolarity of 300 mOsm/1 (oc-medium).
  • Dulbecco's MEM Dulbecco's modified Eagle's medium obtained from GIBCO (Life Technologies LTD, Paisley, Scotland) supplemented with the amino acids L-alanine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid and L-proline (Sigma), vitamin B
  • Granulocyte/macrophage colony formation was stimulated by a saturating concentration of M-
  • CSF purified from pregnant mouse uteri extract (PMUE) essentially as described before, supplemented with 100 ng/ml murine stem cell factor (SCF, a kind gift from Immunex Corporation, Seattle, WA) and
  • BFU-E growth was stimulated by 100 ng/ml DCF and 4 U/ml murine erythropoietin (EPO,
  • Behringwerke, Marburg, F.R.G. purified from the serum of phenylhydrazine treated mice, titrated to an optimal concentration. Colonies were counted after 10 days of culture.
  • the culture medium of the erythroid progenitors also contained hermine (bovine, type I, Sigma) at a concentration of 2x 10" 4 mol/1.
  • CFU-Meg Megakaryocyte progenitor cells
  • Colony numbers represent the mean ⁇ standard deviation of bone marrow samples of individual mice.
  • mice were injected with 5 x 10 ⁇ BM cells or 5 x 10 ⁇ spleen cells in HH one day after TBI.
  • mice Thirteen days later, mice were sacrificed, and spleens were excised and fixed in Tellyesniczsky's solution (64% ethanol, 5% acetic acid and 2% formaldehyde) in H2O.
  • Tellyesniczsky's solution (64% ethanol, 5% acetic acid and 2% formaldehyde) in H2O.
  • TPO 2 h before TBI a very high dose of TPO 2 h before TBI.
  • the effect originated from multilineage cells, as shown in the peripheral blood by equivalent effects of TPO on red cells, white cells (mainly neutrophils) and platelets. See Table 1 below.
  • Table 1 Major peripheral blood cell counts 10 days after 6 GY TBI and TPO administration
  • TBI was given in three equal fractions of 3 Gy separated by 24 h each (experimental design 2).
  • TPO was given in a total dose of 0.9 microgram as indicated in the legend of Fig. 10.
  • the thrombocyte response was optimal when this dose of TPO was given in three equal fractions of 0.3 microgram TPO, 2 h after each fraction of TBI. Similar effects were shown for red cells and white cells. Table 2 shows that in this experimental setting also, a very high dose of TPO 2 h before the first fraction of TBI was equally effective.
  • Figure 13 shows the pharmacokinetic data following three doses of 0.3 microgram of TPO or a single dose of 0.9 microgram. Peak levels relevant occur about 2 h after i.p. injection. An effective level is 30 ng/ml plasma. However, the minimum effective TPO level has not yet been determined by titration experiments.
  • TPO appeared to be highly effective in mice exposed to a total dose of 9 Gy TBI in three equal fractions separated by 24 h each, if administered intraperitoneally 2 h after each TBI fraction.
  • TPO administration prevented the severe reduction of thrombocyte numbers observed in the placebo control group, stimulated the recovery of granulocytes and also fully prevented the development of anemia. Similar to the 6 Gy single TBI experiments, the effect appeared to be mediated by accelerated reconstitution of progenitor cells of multiple blood cell differentiation lineages. This implies that the efficacy of TPO is dependent on residual immature multilineage cells, which need to be stimulated by TPO in a time window following TBI.
  • the small time window to achieve optimal efficacy of TPO is peculiar in view of the slow phase of the wash-out of TPO levels, which has a terminal half-life of approximately 20 h and would result in approximately similar levels at all time points within the first twelve h after administration with the exception of the initial rise due to the distribution in plasma.
  • the latter hypothesis was tested in two ways, i.e., by pharmacokinetic measurements and by administration of a very high dose of TPO before TBI to examine whether an efficacy could be reached similar to that obtained by TPO administration after each fraction of radiation.
  • Thrombocyte counts 10 days after the last dose of TBI in mice treated with 30 micrograms before the first dose of TBI were not significantly differently from those of the mice treated with the most effective schedule of 0.3 microgram TPO 2 h after each TBI fraction.
  • a similar efficacy was obtained by the high dose of TPO 2 h before a single TBI dose of 6 Gy.
  • Table 3 Effects of TPO treatment following 9 Gy TBI (3x3Gy, 24 hours apart) on CFU-S day 13 and progenitor cell content of bone marrow 24 hours after the last dose of TBI in mice treated with TPO 2 hours after each dose of TBI vs control
  • Platelet recovery was defined as the first day of an unsupported platelet count > 25,000/ ⁇ l
  • rhTPO in all Dose Levels was administered Q3D with the exception of Dose Level 5 where the rhTPO was administered QD.
  • rhTPO in combination with G-CSF appears to be safe and well-tolerated in patients undergoing myeloablative therapy and rescue with autologous bone marrow.
  • TPO thrombopoietin
  • CVP Cyclophosphamide 1.5 mg/m 2 /d IV+ mesna; etoposide 250 mg/m 2 dl-3; cisplatin 40 mg/m 2 dl-3, followed by single dose thrombpoietin 0.6-2.4 ⁇ g/kg IV d4 G-CSF 6 ⁇ g/kg ql2h.
  • CBT Cyclophosphamide2.0 gm/m 2 IV, Thiotepa 240 mg/m 2 /d, BCNU 150 mg/m 2 /d days -8, -1, -6 with reinfusion of the cryopreserved cells on day 0. G-CSF 5 ⁇ g/kg/d SC until recovery of granulocytes.
  • Thrombopoietin was well tolerated when given with G-CSF following CVP chemotherapy for PBPC mobilization.
  • Mobilization was enhanced compared to historical controls. A median of one apheresis was required to reach target cell dose. Hematopoietic recovery post transplant was rapid.
  • TPO thrombopoietin
  • HDCT high dose chemotherapy
  • cisplatin cisplatin
  • VP-16 cyclophosphamide
  • the HDCT treatment scheme was as follows:
  • the TPO dosing scheme was as follows:
  • Mobilization consisted of TPO and G-CSF 10 microgram/kg (5 microgram/kg BID) in group A. G-CSF 10 microgram/kg once a day in group B. G-CSF 10 microgram/kg (5 microgram/kg BID) in group C.
  • PBPC-s were cryopreserved in a solution containing 5% DMSO and were frozen by simple immersion into a -130 °C freezer.
  • Group A vs B vs C comparisons were performed using the Kruskal-Wallis test.
  • Group A vs B were compared using Wilcoxon rank-sum test.
  • Data on PBPC apheresis, hematopoietic recovery and transfusion requirements are presented as median (range).
  • PLT Platelet transfusions were provided for ⁇ 20,000 PLT/ul or as clinically indicated. Platelet independence is defined as the day after the last platelet transfusion. Single pheresis products and pooled platelet products are counted as 1 PLT transfusion.
  • TPO in the dose ranges tested is well tolerated.
  • TPO in combination with G-CSF increases the efficacy of PBPC mobilization and CD34 + yield.
  • TPO and G-CSF mobilized PBPC accelerate both platelet and granulocyte recovery.
  • the invention specifically contemplates treatment cycles in which the radiation or chemotherapy agent is administered on multiple consecutive days, for example on 4, 5, 6 or 7 consecutive days, where the TPO dose is administered prior to the first of the consecutive days and/or concurrent with one or more of the consecutive days of the treatment cycle.
  • TPO For a treatment cycle of 5 consecutive days, TPO might be given on day -1 and on days 6, 9, 12, 15, etc.
  • TPO for a treatment cycle of 7 consecutive days, TPO might be given on day -1 and on days 2, 4, 6, 8, 10, 12, etc. or on day -1 and on days 4, 6, 8, 10, 12, etc.
  • the radiation or chemotherapy agent will be given on alternate days of the treatment cycle, e.g. days 1, 3, 5. In this embodiment, TPO might be given on day -1 and on days 2, 4, 6, 8, 10, etc.
  • TPO tissue-derived neurotrophic factor
  • progenitor cells obtained, for example, from mammalian bone marrow, peripheral blood, or umbilical cord blood.
  • Progenitors expanded in such a manner can be used for allogeneic stem cell transplant for patients who have been treated with chemotherapy or radiation treatment.
  • a progenitor population that is enriched for stem cell activity is the CD34 + population and this population can be fiither enriched by selecting for CD34 + CD38 " cells. This population has the ability to generate multilineage hematopoietic colonies in vitro and multipotential hematopoietic engraftment in vivo.
  • Progenitor stem cells for expansion can be obtained from peripheral blood by pheresis and from bone man-row aspirates by standard techniques. Hematopoietic progenitors can also be obtained from umbilical cord blood. Hematopoietic stem cells having the CD34 + phenotype may then be isolated from the blood or marrow using an immunomagnetic enrichment column. The isolated cells may then be cultured in a cell growth medium containing a cocktail of the growth factors TPO, Flt-3 and c-kit ligand in order to expand or increase the number of cells in the stem cell population. The growth factors are added in amounts sufficient to stimulate growth of the progenitor stem cells.
  • each of the growth factors is added in an amount of about 10 to about 100 ng/ml to growth media containing about 10 2 to about 10 ⁇ stem cells/ml.
  • the cells are cultured using standard culture techniques, for example, about 35 - 40°C in aproximately 5% CO2 for about 1 - 8 weeks.
  • the cultures are exchanged into fresh media containing growth factors every week.
  • the growth media may contain other conventional nutrients, fetal serum, etc in standard amounts.
  • the expanded cells are then readministered as an allogeneic stem cell transplant according to known procedures.
  • Hematopoietic progenitor cell populations were isolated from human bone marrow. Briefly, the mononuclear fraction was enriched for CD34 cells using animmunomagnetic enrichment column. (Miltenyi Biotech, Auburn, CA). Purity was routinely >90% by FACS.
  • Suspension culture assays Hematopoietic stem cell populations were seeded at 2e4 cells/mL in IMDM Gibco BRL (Grand Island, NY), plus 10 % fetal bovine serum (Gibco BRL), 10 "5 M 2- mercaptoethanol, 10 " M hydrocortisone , and 2 mM L-glutamine (Gibco BRL). Growth factors were added at the following concentrations: Flt-3 ligand (Immunex, Seattle WA) 50 ng/mL, TPO (Genentech, S. San Francisco, CA) 50 ng/mL, and c-kit ligand (R and D Systems) 50 ng/mL.
  • Flt-3 ligand Immunex, Seattle WA
  • TPO Geneentech, S. San Francisco, CA
  • c-kit ligand R and D Systems
  • Flow cytometric analysis For FACS analysis, cells were resuspended in PBS/2% FBS at le6 cells /mL and stained with mouse anti human CD34 FITC, CD38 PE (Becton Dickenson). Viable cells were selected by propidium iodide exclusion and analyzed on a FACscan (Becton Dickenson).
  • Methylcellulose colony assays were performed using "complete" myeloid methylcellulose media (Stem Cell Technologies, Vancouver, B.C.). Cells were seeded in methylcellulose at 1,000 cells/mL and plated in 4 x 35 mm gridded dishes. Colonies were counted and visually phenotyped on an inverted phase contrast microscope after 14 days in culture.
  • SCID-hu mouse reconstitution assay CB- 17 scid/scid mice were implanted with fetal bone marrow as described previously. Mice were used so that the grafts and cells were mismatched for major histocompatibility complex (MHC) class I antigens.
  • MHC major histocompatibility complex
  • mice received 250 rads whole body ⁇ irradiation and then followed by injection of 30,000 cultured human bone marrow cells into the bone graft. Eight weeks after the injection of the cells, the bone grafts were harvested and analyzed for donor HLA contribution to FITC conjugated anti human CD34 (progenitor), anti human CD33
  • Donor HLA positive cells were then sorted by FACS. Thirty thousand donor HLA positive cells were then injected into secondary recipients in the same manner as the primary recipients. Eight weeks after injection the secondary bone grafts were removed and analyzed for engraftment of CD34, CD19, and CD33.
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO: 3:
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO: 7:
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO:10:
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO: 12:
  • MOLECULE TYPE DNA (genomic)
  • CACATCGAAC CACGT 65 INFORMATION FOR SEQ ID NO: 14:
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • Leu Asn Arg lie His Glu Leu Leu Asn Gly Thr Arg Gly Leu Phe 230 235 240

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des matières de thrombopoïétine pouvant être administrées de façon à obtenir un effet sensiblement thérapeutique en une seule prise quotidienne ou en plusieurs prises à faible dose. On obtient une inversion de la thrombocytopénie en administrant à un patient atteint de cette maladie ou nécessitant un tel traitement une seule dose quotidienne ou de multiples faibles doses d'une quantité thérapeutiquement efficace d'une thrombopoïétine.
PCT/US1998/010475 1997-05-21 1998-05-21 Nouveau procede d'administration de thrombopoietine WO1998052598A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP55066298A JP4562815B2 (ja) 1997-05-21 1998-05-21 トロンボポエチンの新規な投与
AU75898/98A AU7589898A (en) 1997-05-21 1998-05-21 Novel administration of thrombopoietin
CA002288964A CA2288964A1 (fr) 1997-05-21 1998-05-21 Nouveau procede d'administration de thrombopoietine
EP98923662A EP0983082A1 (fr) 1997-05-21 1998-05-21 Nouveau procede d'administration de thrombopoietine

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US85976797A 1997-05-21 1997-05-21
US08/859,767 1997-05-21
US1501698A 1998-01-28 1998-01-28
US09/015,016 1998-01-28

Publications (1)

Publication Number Publication Date
WO1998052598A1 true WO1998052598A1 (fr) 1998-11-26

Family

ID=26686844

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/010475 WO1998052598A1 (fr) 1997-05-21 1998-05-21 Nouveau procede d'administration de thrombopoietine

Country Status (5)

Country Link
EP (1) EP0983082A1 (fr)
JP (1) JP4562815B2 (fr)
AU (1) AU7589898A (fr)
CA (1) CA2288964A1 (fr)
WO (1) WO1998052598A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000044398A2 (fr) * 1999-01-28 2000-08-03 Board Of Regents, The University Of Texas System Procedes d'augmentation de plaquettes circulantes pour prelevement et cryoconservation avec des compositions de thrombopoietine
DE19956156A1 (de) * 1999-11-23 2001-06-13 Univ Eberhard Karls Verfahren zur Aufbereitung eines Präparates von Thrombozyten und Thrombozytenpräparat
US6673580B2 (en) 2000-10-27 2004-01-06 Genentech, Inc. Identification and modification of immunodominant epitopes in polypeptides
EP1670821A1 (fr) * 2003-10-09 2006-06-21 Daewoong Co., Ltd. Procede de purification de la thrombopoietine humaine a teneur en acide sialique elevee
JP2009114212A (ja) * 2000-11-06 2009-05-28 Pharma Mar Sa 効果的な抗腫瘍治療
WO2013064672A3 (fr) * 2011-11-03 2013-07-04 Ulf Niemeyer Oxazaphosphorines en dose unique utilisées pour le traitement de maladies
US8895557B2 (en) 2004-10-29 2014-11-25 Pharma Mar, S.A., Sociedad Unipersonal Pharmaceutical formulations of ecteinascidin compounds
US9192568B2 (en) 2005-10-31 2015-11-24 Pharma Mar, S.A. Formulations comprising jorumycin-, renieramycin-, safracin- or saframycin-related compounds for treating proliferative diseases
EP3045123A1 (fr) * 2008-06-16 2016-07-20 University Of Rochester Analogues de facteur de croissance de fibroblaste (fgf) et leurs utilisations
US9427456B2 (en) 2009-06-14 2016-08-30 Biokine Therapeutics Ltd. Peptide therapy for increasing platelet levels
US9439942B2 (en) 2012-04-24 2016-09-13 Biokine Therapeutics Ltd. Peptides and use thereof in the treatment of large cell lung cancer
US10682390B2 (en) 2015-07-16 2020-06-16 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US10993985B2 (en) 2016-02-23 2021-05-04 BioLmeRx Ltd. Methods of treating acute myeloid leukemia

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3025724B1 (fr) * 2009-05-13 2018-07-11 The University of North Carolina At Chapel Hill Inhibiteurs de kinases cycline-dépendantes et leurs procédés d'utilisation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0675201A1 (fr) * 1994-03-31 1995-10-04 Amgen Inc. Compositions et procédés pour la stimulation de la croissance et la différenciation des mégacaryocytes
WO1997026907A1 (fr) * 1996-01-25 1997-07-31 Genentech, Inc. Nouvelle administration de thrombopoietine

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG47030A1 (en) * 1994-01-03 1998-03-20 Genentech Inc Thrombopoietin
JP2991630B2 (ja) * 1994-02-14 1999-12-20 麒麟麦酒株式会社 ヒトtpo活性を有するタンパク質をコードするdna

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0675201A1 (fr) * 1994-03-31 1995-10-04 Amgen Inc. Compositions et procédés pour la stimulation de la croissance et la différenciation des mégacaryocytes
WO1997026907A1 (fr) * 1996-01-25 1997-07-31 Genentech, Inc. Nouvelle administration de thrombopoietine

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BASSER R L ET AL: "Randomized, blinded, placebo -controlled phase I trial of pegylated recombinant human megakaryocyte growth and development factor with filgrastim after dose-intensive chemotherapy in patients with advanced cancer [published erratum appears in Blood 1997 Sep 15;90(6):2513].", BLOOD, (1997 MAY 1) 89 (9) 3118-28, XP002076300 *
NEELIS K J ET AL: "Prevention of thrombocytopenia by thrombopoietin in myelosuppressed rhesus monkeys accompanied by prominent erythropoietic stimulation and iron depletion.", BLOOD, (1997 JUL 1) 90 (1) 58-63, XP002076301 *
NEELIS K J ET AL: "Simultaneous administration of TPO and G-CSF after cytoreductive treatment of rhesus monkeys prevents thrombocytopenia, accelerates platelet and red cell reconstitution, alleviates neutropenia, and promotes the recovery of immature bone marrow cells.", EXPERIMENTAL HEMATOLOGY, (1997 SEP) 25 (10) 1084-93, XP002076303 *
NEELIS K J ET AL: "The efficacy of single-dose administration of thrombopoietin with coadministration of either granulocyte/macrophage or granulocyte colony-stimulating factor in myelosuppressed rhesus monkeys.", BLOOD, (1997 OCT 1) 90 (7) 2565-73, XP002076302 *

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000044398A3 (fr) * 1999-01-28 2001-03-15 Univ Texas Procedes d'augmentation de plaquettes circulantes pour prelevement et cryoconservation avec des compositions de thrombopoietine
EP1374890A2 (fr) * 1999-01-28 2004-01-02 Board Of Regents, The University Of Texas System Compositions de thrombopoiétine pour l'augmentation de plaquettes circulantes
EP1374890A3 (fr) * 1999-01-28 2004-08-25 Board Of Regents, The University Of Texas System Compositions de thrombopoiétine pour l'augmentation de plaquettes circulantes
WO2000044398A2 (fr) * 1999-01-28 2000-08-03 Board Of Regents, The University Of Texas System Procedes d'augmentation de plaquettes circulantes pour prelevement et cryoconservation avec des compositions de thrombopoietine
DE19956156A1 (de) * 1999-11-23 2001-06-13 Univ Eberhard Karls Verfahren zur Aufbereitung eines Präparates von Thrombozyten und Thrombozytenpräparat
US6673580B2 (en) 2000-10-27 2004-01-06 Genentech, Inc. Identification and modification of immunodominant epitopes in polypeptides
JP2009114212A (ja) * 2000-11-06 2009-05-28 Pharma Mar Sa 効果的な抗腫瘍治療
EP1670821A1 (fr) * 2003-10-09 2006-06-21 Daewoong Co., Ltd. Procede de purification de la thrombopoietine humaine a teneur en acide sialique elevee
EP1670821A4 (fr) * 2003-10-09 2006-11-29 Daewoong Co Ltd Procede de purification de la thrombopoietine humaine a teneur en acide sialique elevee
US8895557B2 (en) 2004-10-29 2014-11-25 Pharma Mar, S.A., Sociedad Unipersonal Pharmaceutical formulations of ecteinascidin compounds
US10322183B2 (en) 2004-10-29 2019-06-18 Pharma Mar, S.A., Sociedad Unipersonal Pharmaceutical formulations of ecteinascidin compounds
US9192568B2 (en) 2005-10-31 2015-11-24 Pharma Mar, S.A. Formulations comprising jorumycin-, renieramycin-, safracin- or saframycin-related compounds for treating proliferative diseases
EP3045123A1 (fr) * 2008-06-16 2016-07-20 University Of Rochester Analogues de facteur de croissance de fibroblaste (fgf) et leurs utilisations
US9427456B2 (en) 2009-06-14 2016-08-30 Biokine Therapeutics Ltd. Peptide therapy for increasing platelet levels
WO2013064672A3 (fr) * 2011-11-03 2013-07-04 Ulf Niemeyer Oxazaphosphorines en dose unique utilisées pour le traitement de maladies
US9439942B2 (en) 2012-04-24 2016-09-13 Biokine Therapeutics Ltd. Peptides and use thereof in the treatment of large cell lung cancer
US11590200B2 (en) 2015-07-16 2023-02-28 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US10682390B2 (en) 2015-07-16 2020-06-16 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11648293B2 (en) 2015-07-16 2023-05-16 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11534478B2 (en) 2015-07-16 2022-12-27 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11554159B2 (en) 2015-07-16 2023-01-17 Blokine Therapeutics Ltd. Compositions and methods for treating cancer
US11559562B2 (en) 2015-07-16 2023-01-24 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US10786547B2 (en) 2015-07-16 2020-09-29 Biokine Therapeutics Ltd. Compositions, articles of manufacture and methods for treating cancer
US11596666B2 (en) 2015-07-16 2023-03-07 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11638743B2 (en) 2015-07-16 2023-05-02 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11612638B2 (en) 2015-07-16 2023-03-28 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11607444B2 (en) 2015-07-16 2023-03-21 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11638742B2 (en) 2015-07-16 2023-05-02 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US11642393B2 (en) 2015-07-16 2023-05-09 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US10993985B2 (en) 2016-02-23 2021-05-04 BioLmeRx Ltd. Methods of treating acute myeloid leukemia

Also Published As

Publication number Publication date
EP0983082A1 (fr) 2000-03-08
JP2001526689A (ja) 2001-12-18
JP4562815B2 (ja) 2010-10-13
CA2288964A1 (fr) 1998-11-26
AU7589898A (en) 1998-12-11

Similar Documents

Publication Publication Date Title
ES2314999T3 (es) Factor celular madre.
AU711639B2 (en) Novel administration of thrombopoietin
US6248319B1 (en) Method for increasing hematopoietic progenitor cells by stem cell factor polypeptides
JPH04501421A (ja) 血小板産生刺激薬剤のためのil―7の使用
US7144731B2 (en) SCF antibody compositions and methods of using the same
Alexander Cytokines in hematopoiesis
US6013067A (en) Methods for increasing hematopoietic cells
WO1998052598A1 (fr) Nouveau procede d'administration de thrombopoietine
CA2212006A1 (fr) Nouveaux ligands se fixant a c-mpl
Hartung Immunomodulation by colony-stimulating factors
US5498599A (en) Methods for stimulating platelet production
US6207454B1 (en) Method for enhancing the effciency of gene transfer with stem cell factor (SCF) polypeptide
JPH05247095A (ja) 新規な巨核球増幅因子とその製造方法および医薬組成物
AU2006201129B2 (en) Novel administration of thrombopoietin
US6852313B1 (en) Method of stimulating growth of melanocyte cells by administering stem cell factor
Kuter Thrombopoietins and thrombopoiesis: a clinical perspective
EP1033997B1 (fr) Technique visant a mobiliser des cellules souches hematopoietiques
Murthy Studies into the mechanism of action of interleukin 3: purification of interleukin 3 and characterization of its cell surface receptor
JPH07118163A (ja) β2−ミクログロブリンを含有する組成物
MXPA97009244A (en) Methods to increase hematopoyeti cells
JPH05255396A (ja) ヒトインターロイキン11様蛋白とその製造方法および医薬組成物

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 09180028

Country of ref document: US

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
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: 1998923662

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2288964

Country of ref document: CA

Ref country code: CA

Ref document number: 2288964

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1998 550662

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 75898/98

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1998923662

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642