EP1049485A1 - Enhancement of hematopoietic cells - Google Patents

Enhancement of hematopoietic cells

Info

Publication number
EP1049485A1
EP1049485A1 EP98903533A EP98903533A EP1049485A1 EP 1049485 A1 EP1049485 A1 EP 1049485A1 EP 98903533 A EP98903533 A EP 98903533A EP 98903533 A EP98903533 A EP 98903533A EP 1049485 A1 EP1049485 A1 EP 1049485A1
Authority
EP
European Patent Office
Prior art keywords
prolactin
hematopoietic
composition
cells
interleukin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98903533A
Other languages
German (de)
English (en)
French (fr)
Inventor
Susan M. Richards
William J. Murphy
Dan L. Longo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genzyme Corp
Original Assignee
Genzyme Corp
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 Genzyme Corp filed Critical Genzyme Corp
Publication of EP1049485A1 publication Critical patent/EP1049485A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2257Prolactin
    • 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

Definitions

  • the red marrow that is found in these bones consists of a sponge-like reticular framework located between long trabeculae. The spaces in this framework are filled with fat cells, which mature and exit via the dense network of vascular sinuses to become part of the circulatory system.
  • All blood cells originate from a common stem cell that becomes committed to differentiate along particular lineages (i.e., erythroid, megakaryocytic, granulocytic, monocytic, and lymphocytic).
  • the proliferation and maturation of precursor cells in the bone marrow are stimulated by certain cytokines.
  • Many of these cytokines are also called "colony-stimulating factors" because they are assayed by their ability to stimulate the growth and development of various leukocyte colonies from marrow cells. While it is known that different cytokines promote the proliferation and maturation of different lineages of bone marrow precursor cells, little is known about the nature of the self- renewing pluripotent stem cell or the mechanisms that regulate its commitment to specific lineages.
  • the invention relates to a method for enhancing hematopoiesis by contacting hematopoietic pluripotent stem cells or progenitor cells with a composition containing prolactin.
  • the prolactin used is recombinant prolactin. Stimulation of hematopoesis can serve to replace hematopoietic cells as they become ablated because of a therapeutic drug or treatment.
  • the enhancement can also function to recruit new or additional cell lineages to a depleted or poorly-functional repertoire of cells.
  • the invention further relates to a method for treating an animal to improve hematopoiesis or prevent hematopoietic-suppression by administering a pharmaceutically acceptable composition containing prolactin.
  • the invention further relates to a composition comprising a cytokine that can enhance hematopoiesis and prolactin.
  • the invention further relates to a composition comprising a therapeutic that can cause hematopoietic-suppression and a prolactin.
  • Figure 1A shows a graph demonstrating that recombinant human prolactin promotes the growth of hematopoietic progenitor cells in long term bone marrow culture as verified by improved cumulative cellularity.
  • Figure IB graphically illustrates that recombinant human prolactin promotes the growth of hematopoietic progenitor cells in long term bone marrow cultures as measured by colony forming unit-culture assay.
  • Figure 1C shows a graph demonstrating that recombinant human prolactin promotes the growth of hematopoietic progenitor cells in long term bone marrow cultures as measured by burst forming unit-erythroid assay.
  • Figure 2 graphically illustrates that azidothymidine (AZT) significantly lowers hematopoietic progenitor content in the bone marrow cells and that prolactin counteracts the effect as measured by hematocrit.
  • AKT azidothymidine
  • Figure 3A shows a graph illustrating that prolactin counteracts the myelosuppressive effects of AZT lowering the hematopoietic progenitor content in the bone marrow cells as verified by improved cumulative cellularity.
  • Figure 3B graphically illustrates that prolactin counteracts the myelosuppressive effects of AZT lowering the hematopoietic progenitor content in the bone marrow cells as measured by colony forming unit-culture assay.
  • Figure 3C shows a graph demonstrating that prolactin counteracts the myelosuppressive effects of AZT lowering the hematopoietic progenitor content in the bone marrow cells as measured by burst forming unit-erythroid assay
  • Figure 4 graphically illustrates that prolactin prevents the myelosuppressive effects of AZT in pretreated mice as measured by hematocrit.
  • Figure 5 shows a graph demonstrating that prolactin can reverse the myelosuppressive effects of AZT as measured by hematocrit.
  • Figure 6A graphically illustrates that prolactin increases platelet content.
  • Figure 6B shows a graph demonstrating that prolactin increases white blood cell count.
  • Figure 7A and 7B graphically demonstrates through differential analysis that the lymphocyte and neutrophil percentage in blood was significantly increased, suggesting prolactin improved the peripheral lymphocyte and neutrophil development.
  • Figure 8 shows a graph illustrating that prolactin influenced B-cell progenitor cells by improving responsiveness to keyhole limpet hemocyanin (KLH) as measured by increased production of KLH-specific IgG and IgM.
  • KLH keyhole limpet hemocyanin
  • Figure 9 graphically illustrates that prolactin increases natural killer function, as assessed by cytotoxicity.
  • prolactin refers to a polypeptide obtained from tissue cultures or by recombinant techniques and other techniques known to those of skill in the art, exhibiting the spectrum of activities characterizing this protein.
  • the word includes not only human prolactin (hPRL), but also other mammalian prolactin such as, e.g., mouse, rat, rabbit, primate, pig (ovine) and cow (bovine) prolactin.
  • the recombinant PRL (r-PRL) includes any active fragment or active prolactin sequence.
  • recombinant prolactin designated as r-PRL, preferably human prolactin, refers to prolactin having comparable biological activity to native prolactin prepared by recombinant DNA techniques known by those of skill in the art.
  • hematopoiesis or “hemopoiesis” refers to the conventional meaning of the word which encompasses the formation and development of various types of cells including pluripotent stem cells, myeloid progenitor cells and lymphoid progenitor cells as well as blood products derived therefrom such as platelets.
  • composition refers to any formulation or preparation that when administered to an animal will be tolerated by said animal. Administration includes oral administration and injection including subcutaneous, intraperitoneal, intravenous, intradermal, intramuscular, etc.
  • hematopoietic-suppression include myelosuppression or lymphoid- suppression as caused by such treatment as AZT, irradiation, cytoreductive treatment, chemotherapy, cytolytic therapy, immunocytolytic, or combinations thereof.
  • cytokine or cytokines as used herein means any cytokine or growth factor or colony-stimulating factor that can stimulate the expansion and differentiation of stem cells or progenitor cells.
  • Cytokines include interleukin-1, interleukin -2, interleukin 3, interleukin-4, interleukin-6, interleukin-7, interleukin-9, interleukin- 1 1 , interleukin -15, c-Kit ligand, granulocyte-monocyte colony-stimulating factor, monocyte-colony-stimulating factor, granulocyte-colony-stimulating factor, Flt3 ligand, Mpl ligand, erythropoietin (Epo), thrombopoietin, (Tpo), growth hormone, (GH), insulin-growth factor, (IGF), transforming-growth factor- ⁇ , (TGF- ⁇ ), and mixtures thereof.
  • Example 1 Effect of prolactin on hematopoietic progenitor content in vivo.
  • mice 8-12 weeks of age
  • mice 8-12 weeks of age
  • mice 8-12 weeks of age
  • mice 8-12 weeks of age
  • mice were injected with lO ⁇ g of recombinant human prolactin (r-hPRL, Genzyme Corporation) that was resuspended in 0.2 mL Hanks' Balanced Salt Solution (HBSS) Mediatech, Inc. Herndon, VA.
  • HBSS Hanks' Balanced Salt Solution
  • VA Herndon, VA.
  • the animals received i.p. injections every other day for 10 days (a total of five injections).
  • mice were weighed weekly. Blood was collected from the mice via the lateral tail vein, using EDTA as an anticoagulant.
  • IMDM Iscove's modified Dulbecco's medium
  • FBS fetal bovine serum
  • L-glutamine 1% L-glutamine
  • LTBMC Long term bone marrow cultures
  • BMC from the mouse femur were cultured at an initial concentration of 1X10 cells/mL in 24-well plates. Every third day, the cultures had half of their volume exchanged with fresh medium. When the cell concentration was more than 2x10 /mL, the culture was diluted and separated into two wells. Every 10 days the cultures were evaluated for their cellularity and by colony assays for CFU-c and BFU-e.
  • CFU-c colony forming unit-culture
  • GM-CSF murine granulocyte macrophage stimulating factor
  • IL-3 murine interleukin-3
  • the burst forming unit-erythroid (BFU-e) assay was performed as described by Stephenson JR, et.al (1971) Proc Natl Acad USA, 68: 1542.
  • a 5-mL volume of the suspension included: 1.5 mL cells, 1.5 mL FBS, 0.5 mL 10% BSA, 0.5 mL of 1.0 mM 2-mercaptoethanol, 0.5 mL Epo, and 0.5 mL recombinant murine DL-3 (rmIL-3).
  • the final concentration of erythropoietin (Epo, Stem Cell Technologies, British Columbia, Canada) was 2 U/mL, rmLL-3 was 20 ng/mL and the cells were at lxlO ⁇ /mL.
  • BFU-e were scored after 12 days of incubation. A BFU-e was defined as a group containing 50 or more benzidine-positive cells. All assays had at least three mice per group and were performed at least three times, with a representative experiment being shown.
  • Example ⁇ Effect of prolactin on hematopoietic progenitor content in vitro.
  • CFU-c and BFU-e short-term colony culture system
  • LTBMC long term suspension culture system
  • Example JJJ Effects of prolactin on hematopoietic progenitor content in mice administered AZT as a means of inducing mvelosuppression.
  • mice were placed on AZT (2.5 mg/mL in drinking water) for several weeks. Upon analysis, these mice exhibited significantly lower (p ⁇ 0.01) BMC hematopoietic progenitor content as measured with CFU-c and BFU-e (Table 3 shown below) as well as significantly lower hematocrit (HCT) than normal mice ( Figure 2). These effects became more pronounced the longer the mice were placed on AZT, with most hematologic values approaching nearly half the control values.
  • mice concurrently received 1, 10, or lOO ⁇ g r-hPRL ip every other day for 20 days.
  • Cellularity and colony assays (CFU-c and BFU-e) . were determined after 14 or 28 days.
  • the CFU-c/Femur or BFU-e/Femur were calculated as: colony number/2xl0 x cellularity of femur.
  • the hematopoietic progenitor cell content (CFU-c and BFU-e) fully recovered to normal or even higher.
  • the HCT value also increased in response to r-hPRL treatment (Figure 2), increasing from 29.5+/-1.3% to 40.3+/-3.2% with mice administered AZT and examined at day 14 after concurrent r-hPRL treatment. Similar results were obtained after 28 days. Additionally, the mice exhibited no apparent pathologic effects from repeated r-hPRL injections. The mice appeared to be in good health throughout the study. They maintained a constant weight and mice sacrificed at the end of the study showed no gross pathologic abnormality.
  • Example IV Early administration of prolactin prevents AZT-induced mvelosuppression in mice.
  • mice were injected with 10- ⁇ g of r-hPRL ip every other day for 14 days and then administered AZT in their drinking water (2.5 mg/mL in drinking water), for another 14 days without r-hPRL injections.
  • Cellularity and progenitor cell content were determined at various time points.
  • Significant protection of myelosuppression induced by AZT was observed.
  • the HCT value was significantly (p ⁇ 0.01) enhanced at day 29 to day 34 ( Figure 4).
  • Hematopoietic progenitor cells were also significantly (p ⁇ 0.01) increased in r-hPRL pretreated mice during AZT administration (Table 5 shown below). These results suggest that r-hPRL may protect the progenitor cells in vivo and increase their ability to resist myelosuppression. Table 5
  • Example V Later administration of prolactin reverses AZT-induced mvelosuppression in mice.
  • mice were administered AZT in drinking water (2.5 mg/mL in drinking water) for 14 days. After this two week period, the mice were evaluated to confirm they were myelosuppressed. The animals subsequently received lO ⁇ g r-hPRL ip administered every other day for another 20 days (total of 10 injections). The animals were evaluated for cellularity and progenitor cell content at various time points. Significant improvement of BMC hematopoietic cell content was noted in r-hPRL treated mice (Table 6 shown below) at day 29 (7 r-hPRL injections).
  • the CFU-c/Femur or BFU-e/Femur were calculated as: colony number/2xl0 x cellularity of femur. Values are representative of three experiments containing three to four mice per group. HCT values also recovered to nearly normal levels by day 24 (10 days after r-hPRL treatment) and were significantly higher than HBSS control (Figure 5). These findings suggest that r-hPRL can reverse myelosuppression induced by AZT. Table 6
  • Example VI Prolactin accelerates hematopoietic reconstitution after lethal irradiation followed by bone marrow transplantation.
  • Recipient BALB/c mice (8-12 weeks of age) were exposed to a 137 Cs irradiation source in order to lethally irradiate the animals for cellular reconstitution studies. The mice received
  • mice 850 cGy total body irradiation. These mice then received 1x10 syngeneic BMC intravenously (iv). This procedure is referred to as syngeneic bone marrow transplantation (SBMT). There were five mice per group and each experiment was performed 3-4 times.
  • SBMT syngeneic bone marrow transplantation
  • mice At day 1 after syngeneic bone marrow transplantation (SBMT) the mice started their treatment of either lO ⁇ g r-hPRL or Hanks Balanced Salt Solution (HBSS) as control. r-hPRL was resuspended in 0.2 mL HBSS and injected i.p. every other day until the mice were assayed or received a total of 10 injections over 20 days. Mice were weighed weekly.
  • SBMT syngeneic bone marrow transplantation
  • spleen cells and bone marrow cells were obtained from one tibia and washed and resuspended in Iscove's modified Dulbecco's medium (LMDM) with 10% fetal bovine serum (FBS), 1 % L-glutamine, and antibiotics.
  • LMDM Iscove's modified Dulbecco's medium
  • FBS fetal bovine serum
  • CFU-c and BFU-e colony forming assays
  • mice receiving r-hPRL every other day until day 14 or 21 were put into four groups while the control animals receiving HBSS are put into four other groups.
  • Single cell suspensions from a tibia (BM) were taken from two of the groups of the r-hPRL treated mice and from two of the groups of the HBSS treated mice.
  • Single cell suspensions from the spleen were taken from the two remaining groups from the r-hPRL and HBSS treated groups.
  • Both BM and spleens cells were analyzed by double-color cytometric analysis as described previously (Murphy et.al.1992. J. Immunol 148: 3799-3805).
  • the cells were stained with rat FITC-labeled anti-5E6 (natural killer (NK) cell) antibody and rat PE-labeled 8C5 (granulocyte) antibody obtained from Becton Dickinson (Mountain View, CA).
  • the cells were fixed in 100% paraformaldehyde and analyzed on a EPCIS flow cytometer.
  • FITC or PE-labeled normal rat immunoglobulin (NRIg) was used as a control and each group had 3-4 mice per group.
  • 8C5 (granulocyte marker) cell content was increased in both BM and spleen. Because the cellularity of both BM and spleen also increased (BM group vs.
  • BM+r- hPRL group 7.8xl0 6 vs. 12.6xl0 6 for BM; 54.5xl0 6 vs. 78.5xl0 6 for spleen), the absolute number of 8C5 granulocytes in BM and spleen increased 2.24 fold and 2.85 fold at day 14. This cell population increased 2.03 fold and 1.92 fold at day 21.
  • Example VII Treatment with prolactin after BMT improves B-cell lineage development.
  • B-cell progenitor content and B-cell mitogen responses as a B-cell function assay were evaluated in mice after SBMT.
  • Six mouse groups received r-hPRL treatment (lO ⁇ g/injection, every other day until day 14 or day 21) while 6 groups of control mice, groups received HBSS.
  • the B-cell progenitor content was determined by flow cytometry using the dual-labeling method described above and FITC-labeled anti-B220 and PE-labeled slgM, both obtained from Becton Dickinson.
  • B-cell progenitors would stain positive for the B220 marker and negative for surface
  • the B-cell progenitor (B220 slgM " cell) content increased after r-hPRL treatment in BM and lymph node (LN ) cells, but not in the spleen at days 14 and 21.
  • the absolute number of B-cell progenitors in BM and LN increased 2.56 and 3.78 fold respectively (cellularity x positive cells), suggesting that r-hPRL accelerated the B-cell lineage engraftment and development.
  • the mature B-cell (B220 /slgM cells) content increased in both spleen and LN at day 14 and day 21 after SBMT.
  • mice receiving r-hPRL demonstrated an enhanced proliferative response to the B-cell mitogen, with the CPM of the r-hPRL treatment group significantly higher than the control (p ⁇ 0.01 for any dose of LPS) and the stimulating index (SI) enhanced at each dose of LPS.
  • the BMC-transplanted mice were further evaluated for B cell function by immunizing the animals with KLH and measuring their IgG and IgM response over time.
  • T-cell progenitor content (CD4 CD8 cell) in thymus was increased when examined at day 14 and day 21 after SBMT.
  • the mature T cell content (CD4 + CD8 " or CD4 " CD8 + cell) in the spleen and lymph node were also increased.
  • the effect of r-hPRL administration on T-cell function was also evaluated.
  • the effect of PRL on antigen-specific T cells during a primary immune response was evaluated by examining the splenic T-cell proliferation to KLH in KLH-immunized mice after SBMT.
  • mice were immunized subcutaneously with lOO ⁇ g of KLH in complete Freund's adjuvant at day 7 after SBMT.
  • day 21 i.e. two weeks after KLH immunization
  • the spleens were harvested and the cell suspension was used to assess KLH-specific proliferation.
  • the data demonstrate that r-hPRL administration exerted significant immunopotentiating effects as demonstrated by the significantly increased in vitro proliferation to KLH in the mice receiving r-hPRL treatment.
  • the data verify that r-hPRL may improve the development and function of T-cell lineage from hematopoietic progenitor cells after SBMT.
  • Example LX Prolactin improves NK recovery after BMT
  • NK cells are lymphoid cells that mediate MHC-unrestricted killing of tumors and virally- infected cells. Recently, it was reported that NK cells play an important role in hematopoiesis. Studies were therefore undertaken to examine the development and function of NK cells after
  • NK cells The functionality of the NK cells was also examined by assessing NK cytotoxicity.
  • YAC- 1 NK sensitive target cells
  • Na GO4 New England Nuclear, Boston, MA, specific activity approximately 400 ⁇ Ci/ ⁇ g.
  • the target cells were washed 3 times in RPMI 1640 supplemented with 2% FCS before used in the assay.
  • Effector cells splenocytes
  • target cells were added to round bottomed 96-well plates (Costar) to obtain effector/target (E/T) cell ratio of 40/1, 20/1, 10/1, and 5/1. Four replicate wells were used.
  • %specific lysis CPMexp - CPMspontaneous/ CPMmaximun - CPMspontaneous x 100%.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Endocrinology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
EP98903533A 1997-01-21 1998-01-20 Enhancement of hematopoietic cells Withdrawn EP1049485A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3587597P 1997-01-21 1997-01-21
PCT/US1998/000887 WO1998031385A1 (en) 1997-01-21 1998-01-20 Enhancement of hematopoietic cells

Publications (1)

Publication Number Publication Date
EP1049485A1 true EP1049485A1 (en) 2000-11-08

Family

ID=21885308

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98903533A Withdrawn EP1049485A1 (en) 1997-01-21 1998-01-20 Enhancement of hematopoietic cells

Country Status (5)

Country Link
EP (1) EP1049485A1 (ja)
JP (1) JP2001516341A (ja)
AU (1) AU6028298A (ja)
CA (1) CA2277482A1 (ja)
WO (1) WO1998031385A1 (ja)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6183756B1 (en) 1997-05-01 2001-02-06 Dynagen, Inc. Methods for prevention and/or treatment of thrombocytopenia

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL82885A (en) * 1987-06-15 1991-07-18 Migal Galilee Technology Cente Method and composition containing a peptide hormone for stimulating growth in poultry
US4837202A (en) * 1987-09-14 1989-06-06 Pitman-Moore, Inc. Method for stimulating the immune system
US5696128A (en) * 1994-07-07 1997-12-09 The Board Of Supervisors Of Louisiana University And Agricultural And Mechanical College Method of regulating immune function
CA2183260A1 (en) * 1994-02-14 1995-08-17 Susan Richards Prolactin as a vaccine adjuvant
US5888980A (en) * 1994-06-30 1999-03-30 Bio-Logic Research And Development Corporation Compositions for enhancing immune function

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9831385A1 *

Also Published As

Publication number Publication date
WO1998031385A1 (en) 1998-07-23
AU6028298A (en) 1998-08-07
CA2277482A1 (en) 1998-07-23
JP2001516341A (ja) 2001-09-25

Similar Documents

Publication Publication Date Title
CA2087525C (en) Adoptive immunotherapy with interleukin-7
US5632983A (en) Method for treating secondary immunodeficiency
Geissler et al. Recombinant human interleukin-3 expands the pool of circulating hematopoietic progenitor cells in primates--synergism with recombinant human granulocyte/macrophage colony-stimulating factor
EP0831888B1 (en) Methods for increasing hematopoietic cells
ES2321189T3 (es) Estimulacion de la hematopoyesis por medio de celulas inmunitarias activadas ex vivo.
WO1995006475A1 (en) Increase in hematopoietic progenitor cells in peripheral blood by transforming growth factor beta
Sun et al. Recombinant human prolactin improves antitumor effects of murine natural killer cells in vitro and in vivo
WO1998031385A1 (en) Enhancement of hematopoietic cells
WO1998031385A9 (en) Enhancement of hematopoietic cells
WO1994018991A1 (en) EXPANSION OF STEM CELLS IN LONG TERM BONE MARROW CULTURES BY NEUTRALIZATION OF TGF-$g(b)
Sun et al. Human prolactin improves engraftment and reconstitution of human peripheral blood lymphocytes in SCID mice
Sun et al. Immunologic and hematopoietic effects of recombinant human prolactin after syngeneic bone marrow transplantation in mice
EP0473696B1 (en) Avian Interleukin-2 for enhancing growth in birds
Li et al. Cytokines transduced bone marrow stromal cell lines promote immunohematopoietic reconstitution in mice after allogeneic bone marrow transplantation
EP1871872B1 (en) Method for activating cd8 t cells
US20050004037A1 (en) Osteogenic growth oligopeptides as stimulants of hematopoiesis
Frasca et al. Activation of gp130 signaling in vivo by the IL‐6 super‐agonist K‐7/D‐6 accelerates repopulation of lymphoid organs after irradiation
RU2272810C2 (ru) Средство для активации стволовых клеток
Togni Hematopoietic regulation by catecholamines
AU2001282436A1 (en) Osteogenic growth oligopeptides as stimulants of hematopoiesis
Roncarolo et al. Effects of Cytokine Administration on Human Hematopoiesis in SCID-hu Mice
Khanna et al. Hematopoietic growth factors in transplantation: Biology and applications
WO1993005801A1 (en) Use of il-5 to treat solid tumors
MXPA97009244A (en) Methods to increase hematopoyeti cells

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19990812

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20020630