EP0484512A4 - Human peripheral blood cells in an immunocompromised host - Google Patents

Human peripheral blood cells in an immunocompromised host

Info

Publication number
EP0484512A4
EP0484512A4 EP19910911224 EP91911224A EP0484512A4 EP 0484512 A4 EP0484512 A4 EP 0484512A4 EP 19910911224 EP19910911224 EP 19910911224 EP 91911224 A EP91911224 A EP 91911224A EP 0484512 A4 EP0484512 A4 EP 0484512A4
Authority
EP
European Patent Office
Prior art keywords
host
human
bone marrow
cells
murine
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Withdrawn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
EP19910911224
Other versions
EP0484512A1 (en
Inventor
Charles M. Baum
Ann Tsukamoto
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.)
Systemix Inc
Original Assignee
Systemix 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 Systemix Inc filed Critical Systemix Inc
Publication of EP0484512A1 publication Critical patent/EP0484512A1/en
Publication of EP0484512A4 publication Critical patent/EP0484512A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0271Chimeric vertebrates, e.g. comprising exogenous cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/26Lymph; Lymph nodes; Thymus; Spleen; Splenocytes; Thymocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/407Liver; Hepatocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates

Definitions

  • the field of this invention is the production of human hematopoietic cells.
  • peripheral blood cells it would be desirable to have a relatively high proportion of the peripheral blood cells as human cells' and desirably have a significant proportion of the total number of hematopoietic cells being human cells. Further, it would be desirable to have circulatory myelomonocytic cells and red blood cells, besides lymphoid cells.
  • Xenogeneic bone marrow is introduced into an endogenous bone marrow depleted genetically immunocompromised mammalian host, where the bone marrow may be present as bone tissue or dispersed bone marrow, particularly from a fetal source.
  • the fetal bone tissue may be conveniently introduced into the peritoneal cavity or the bone marrow may be injected into the long bone of the immunocompromised host.
  • Non-human mammals particularly small mammals under 25kg, are provided having human peripheral blood cells present as at least two percent (2%) of the total peripheral blood cells, preferably at least five percent (5%) of the total peripheral blood cells and more preferably at least about fifteen percent (15%) .
  • the enhanced level of human cells may be achieved by depletion of endogenous bone marrow, particularly stem cells, conveniently by irradiating the host with at least a sublethal dosage of radiation, generally being at least about 200 rad and varying with the size and nature of the host.
  • the blood cells are then reconstituted with human bone marrow, conveniently as human bone or bone marrow cell suspensions, particularly from a fetal source.
  • the human bone marrow source may be introduced before and after ablation to enhance the survival rate of the host upon ablation, particularly by irradiation.
  • Various immunocompromised hosts may be employed, where the host being immunocompromised is as a result of a natural mutation, breeding, transformation of embryonic cells to provide a transgenic mouse, or the like.
  • hosts which lack a functional lymphoid lineage The lack of the immune system may be as a result of a lack of a particular organ, a genetic defect, such as an incompetent reco binase or regulatory gene regulating the expression of the recombinase, transport of slg and T cell receptors to the surface, non-functional major histocompatibility antigens, or the like.
  • CB-17 scid/scid mice and mice derived therefrom which may have further enhanced immunodeficiency, e.g. lack of natural killer (NK) cells.
  • the host is irradiated or treated in any convenient manner to substantially ablate the endogenous bone marrow.
  • X-irradiation may be employed where the level of radiation will be selected to provide at least substantially complete ablation of the hematopoietic cell system, while maintaining a reasonable, preferably a high, proportion of viable hosts. With each type of host, the particular level of irradiation may be selected by screening for the level of remaining viable hematopoietic cells for the efficiency of the ablation.
  • Other techniques for bone marrow ablation which may be employed by themselves or in combination include cytotoxic drugs, immunotoxins, antibodies to lymphokines and growth factors, antibodies to natural killer cells, etc. The selection of cytotoxic drugs would be based on their ability to clear quickly from the lymphatic and blood circulatory systems, so as to rapidly be reduced to a non-cytotoxic level, prior to or shortly after the administration of the human bone marrow.
  • the human bone marrow will normally be fetal bone marrow and may be employed as bone marrow slices, fragments, chunks, or the like, coming from various parts of the fetus, such as the femur, tibia, humerus or rib.
  • the longest dimensions of human bone will generally be in the size range of about 8mm to 2cm, where the bone may be fragmented along the long axis, usually in not more than about four parts.
  • the total volume of fetal bone which is introduced into the host will vary with the size of the host. For mice, the volume will generally be in the range of about 40mm 3 to 1,200mm 3 .
  • human fetal bone marrow cell suspensions may be injected into long bones, such as the femur or tibia.
  • long bones such as the femur or tibia.
  • the volume of the bone marrow suspension will be in the range of about 10ml to 50ml for a mouse.
  • the age of the host will generally be at least neonatal and usually not more than about six months, more usually not more than three months.
  • the particular age is not critical to this invention, but younger animals are more convenient to handle and are more economical.
  • the site of introduction of the fetal bone portions may be interperitoneal, subcutaneous, mammary fat pad, or other convenient site.
  • the host After introduction of the bone marrow, the host is grown in accordance with conventional ways, feeding the host as appropriate, either with or without the presence of antibiotics in the feed. Conveniently, about 0.2% to 1.2% of sulfamethoxazole/trimethoprim (Septra) may be provided in the feed. As appropriate, additional introductions of the human fetal bone marrow may be made, usually not more than about two additional explants, generally at four to twenty week intervals.
  • the host may receive a variety of growth factors and cytokines, either native to the host or human. Interleukins, colony stimulating factors, hormones, or the like may be added, such as IL-1, IL-3, IL-6, and G-CSF. Generally, the amount of the additive will vary, depending upon the particular additive, the host, and the like. For example, in the case of IL-3 0.5 - 5 ⁇ g/day may be administered. In the subject host, it is found that the bone tissue shows variable cellularity from 10 to 80%.
  • the human cells comprise a significant proportion of the blood cells in circulation in the peripheral blood. The cells may include members of the lymphoid, myelomonocytic and erythroid lineages.
  • the cells may be detected by employing a wide variety of antibodies which are commercially available for the detection of human markers on blood cells, such as CD-3, -A , -8, -10, -13, -15, -19, -20, -33, -41, -45, -59, HLA, or the like.
  • a blood sample may be taken and assayed employing a fluorescence activated cell sorter.
  • the cells may be lysed and a Western blot employed.
  • Other techniques which may find use for the quantitative determination of a particular cell type include the polymerase chain reaction, gel electrophoresis, HPLC, or the like.
  • lymph node In addition to the bone marrow, other human organs may also be present, particularly thy us, lymph node, skin, liver, pancreas, tonsil, appendix, epithelium, kidney, etc.
  • One or more mesenteric or peripheral lymph nodes may be introduced.
  • the various organs may be introduced conveniently in the kidney capsule, mammary fat pad, particularly the fourth mammary fat pad for a mouse, the popliteal fossa, or other convenient site where the organ may be vascularized, lymphatic vessels connected and maintained by the host for a reasonable period of time, usually at least four weeks, preferably at least six weeks.
  • the subject host may find a wide variety of uses by virtue of the significant presence of circulating human blood cells in the non-human host.
  • the host may be employed for the production of human monoclonal antibodies or immortalized T lymphocytes.
  • the host may be immunized in accordance with conventional ways with an appropriate immunogen, which may be injected intravascularly at an appropriate site. Of particular interest is injection at a site which is drained by a human lymph node introduced into the host.
  • the immunized host may receive one or more booster shots, followed by removal of a lymphoid organ, e.g. lymph node, followed by immortalization, conveniently by fusion with an appropriate yeloid cell or transfection with Epstein-Barr virus.
  • the cells may be cloned by any convenient means, at limiting dilution to provide for individual clones, and the supernatants of the individual clones may be screened for the binding affinity of the antibodies present in the host.
  • the host may be used for studying T and B lymphocyte interactions, in determining the manner of stimulation of T and B lymphocytes and the mechanism for the immune reaction.
  • Individual lymphocytes may be cloned to evaluate their role in disease protection against disease, identify slg or T cell receptor variable regions, isolate the DNA encoding the variable regions, or the like.
  • Various immunogens may be screened for their abilities as vaccines in producing antibodies which may be effective in neutralizing pathogens and the cells or DNA provide a source of the variable regions associated with the response.
  • Pathogens of interest are those particularly associated with humans, such as HIV, HTLV-I, and - II, human papilloma virus, cytomegalovirus, Epstein- Barr virus, hepatitis B virus, non-A, non-B hepatitis, chlamydia, malaria (Falciparum) etc.
  • the subject system may be used in the study of cancer in attempting to develop cytotoxic cells specific for a particular cancer type.
  • mice were transplanted into either untreated mice (6-10 weeks old) or mice (6-10 weeks old) pre-treated with radiation from a cesium 137 source.
  • the mice are treated with either whole body irradiation or irradiation of the long bones.
  • Mice treated with whole body irradiation receive 200 to 400 rads on a single dose.
  • the mice are treated with 600 rads after shielding of the thorax and abdomen with a lead shield.
  • the mice are anesthetized with Nembutol prior to shielded irradiation.
  • the mice were CB-17.5 scid/scid mice.
  • some of the mice were treated with exogenous human IL-3 in two l ⁇ g doses per day.
  • the percentage of human cells in the peripheral blood is determined by employing monoclonal antibodies specific for human hematopoietic cell markers.
  • a low level of human cells is observed in the peripheral blood by FACScan.
  • the positive cells vary from about 0 - 1.6 percent.
  • injection of IL-3 does not significantly affect the percentage of positive cells.
  • the mice irradiated before marrow transplantation showed between 1.5 and 30 percent human cells in the peripheral blood. These cells appear to be of the myelomonocytic lineage. They show no staining with T or B cell markers (CD 3, 4, 8, 19), although they do stain for the myeloid cell marker (CD33) .
  • the human cells have a high side scatter profile indicating that they are of the myelomonocytic lineage.
  • two of the four mice which were irradiated before implantation show low but significant levels of serum immunoglobulin after six weeks.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Environmental Sciences (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nutrition Science (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Physiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • External Artificial Organs (AREA)

Abstract

Human peripheral blood cell circulation is achieved in xenogeneic immunocompromised host with introduction of human fetal bone marrow into the host after irradiation of the host to substantially deplete the host of endogenous hematopoietic cells. The host remain viable for extended periods of time while demonstrating substantial levels of human blood cells in the peripheral blood.

Description

HUMAN PERIPHERAL BLOOD CELLS
IN AN IMMUNOCOMPROMISED HOST
INTRODUCTION Technical Field The field of this invention is the production of human hematopoietic cells.
Background
The blood cell system comprising the diverse cells emanating from a single stem cell is essential to the survival and well being of mammals. The cells serve to carry oxygen, cleanse the body of debris, inhibit the proliferation of pathogens, monitor for neoplastic cells, and provide a wide variety of factors essential for their own and the growth of other cells. Recently, chimeric mice were developed, where human fetal tissue was introduced into CB-17 scid/scid mice to provide the chimeric SCID-hu mouse. The mice were shown to maintain for extended periods of time, human fetal lymph node and thymus and provide for peripheral blood cells, when supplied with a source of hematopoietic stem cells from fetal liver. While in some instances, the maintenance of human peripheral blood cells could be extended for long periods of time, in all instances the human cells were only a very small proportion of the total number of peripheral blood cells, frequently substantially fewer than one percent. Furthermore, the circulating cells in these animals were shown to be T Cells.
In many instances, it would be desirable to have a relatively high proportion of the peripheral blood cells as human cells' and desirably have a significant proportion of the total number of hematopoietic cells being human cells. Further, it would be desirable to have circulatory myelomonocytic cells and red blood cells, besides lymphoid cells.
For many applications, it would be of substantial advantage to have a high absolute number of human cells. These include the opportunity for immunization to obtain a strong immune response from the human cells, studies of various diseases and their effect on the various hematopoietic cells and studies of drugs against various diseases and their effect on the hematopoietic cells. There is, therefore, substantial interest in providing for ways in which a non-human mammal may be provided with human blood cells for testing, research, and as a source of blood cells of the various lineages.
Relevant Literature EPA 0 322 240 describes the introduction of human fetal tissue in a CB-17 scid/scid mouse. See references cited therein.
SUMMARY OF THE INVENTION Xenogeneic bone marrow is introduced into an endogenous bone marrow depleted genetically immunocompromised mammalian host, where the bone marrow may be present as bone tissue or dispersed bone marrow, particularly from a fetal source. The fetal bone tissue may be conveniently introduced into the peritoneal cavity or the bone marrow may be injected into the long bone of the immunocompromised host.
DESCRIPTION OF THE SPECIFIC EMBODIMENTS
Non-human mammals, particularly small mammals under 25kg, are provided having human peripheral blood cells present as at least two percent (2%) of the total peripheral blood cells, preferably at least five percent (5%) of the total peripheral blood cells and more preferably at least about fifteen percent (15%) . The enhanced level of human cells may be achieved by depletion of endogenous bone marrow, particularly stem cells, conveniently by irradiating the host with at least a sublethal dosage of radiation, generally being at least about 200 rad and varying with the size and nature of the host. The blood cells are then reconstituted with human bone marrow, conveniently as human bone or bone marrow cell suspensions, particularly from a fetal source. The human bone marrow source may be introduced before and after ablation to enhance the survival rate of the host upon ablation, particularly by irradiation.
Various immunocompromised hosts may be employed, where the host being immunocompromised is as a result of a natural mutation, breeding, transformation of embryonic cells to provide a transgenic mouse, or the like. Of particular interest are hosts which lack a functional lymphoid lineage. The lack of the immune system may be as a result of a lack of a particular organ, a genetic defect, such as an incompetent reco binase or regulatory gene regulating the expression of the recombinase, transport of slg and T cell receptors to the surface, non-functional major histocompatibility antigens, or the like. Of particular interest are CB-17 scid/scid mice and mice derived therefrom which may have further enhanced immunodeficiency, e.g. lack of natural killer (NK) cells.
The host is irradiated or treated in any convenient manner to substantially ablate the endogenous bone marrow. Conveniently, X-irradiation may be employed where the level of radiation will be selected to provide at least substantially complete ablation of the hematopoietic cell system, while maintaining a reasonable, preferably a high, proportion of viable hosts. With each type of host, the particular level of irradiation may be selected by screening for the level of remaining viable hematopoietic cells for the efficiency of the ablation. Other techniques for bone marrow ablation which may be employed by themselves or in combination include cytotoxic drugs, immunotoxins, antibodies to lymphokines and growth factors, antibodies to natural killer cells, etc. The selection of cytotoxic drugs would be based on their ability to clear quickly from the lymphatic and blood circulatory systems, so as to rapidly be reduced to a non-cytotoxic level, prior to or shortly after the administration of the human bone marrow.
The human bone marrow will normally be fetal bone marrow and may be employed as bone marrow slices, fragments, chunks, or the like, coming from various parts of the fetus, such as the femur, tibia, humerus or rib. The longest dimensions of human bone will generally be in the size range of about 8mm to 2cm, where the bone may be fragmented along the long axis, usually in not more than about four parts. Usually, the total volume of fetal bone which is introduced into the host will vary with the size of the host. For mice, the volume will generally be in the range of about 40mm3 to 1,200mm3. Alternatively, human fetal bone marrow cell suspensions may be injected into long bones, such as the femur or tibia. Generally the volume of the bone marrow suspension will be in the range of about 10ml to 50ml for a mouse.
The age of the host will generally be at least neonatal and usually not more than about six months, more usually not more than three months. The particular age is not critical to this invention, but younger animals are more convenient to handle and are more economical.
The site of introduction of the fetal bone portions may be interperitoneal, subcutaneous, mammary fat pad, or other convenient site.
After introduction of the bone marrow, the host is grown in accordance with conventional ways, feeding the host as appropriate, either with or without the presence of antibiotics in the feed. Conveniently, about 0.2% to 1.2% of sulfamethoxazole/trimethoprim (Septra) may be provided in the feed. As appropriate, additional introductions of the human fetal bone marrow may be made, usually not more than about two additional explants, generally at four to twenty week intervals.
The host may receive a variety of growth factors and cytokines, either native to the host or human. Interleukins, colony stimulating factors, hormones, or the like may be added, such as IL-1, IL-3, IL-6, and G-CSF. Generally, the amount of the additive will vary, depending upon the particular additive, the host, and the like. For example, in the case of IL-3 0.5 - 5 μg/day may be administered. In the subject host, it is found that the bone tissue shows variable cellularity from 10 to 80%. In addition, the human cells comprise a significant proportion of the blood cells in circulation in the peripheral blood. The cells may include members of the lymphoid, myelomonocytic and erythroid lineages. The cells may be detected by employing a wide variety of antibodies which are commercially available for the detection of human markers on blood cells, such as CD-3, -A , -8, -10, -13, -15, -19, -20, -33, -41, -45, -59, HLA, or the like. Conveniently, a blood sample may be taken and assayed employing a fluorescence activated cell sorter. Alternatively, the cells may be lysed and a Western blot employed. Other techniques which may find use for the quantitative determination of a particular cell type include the polymerase chain reaction, gel electrophoresis, HPLC, or the like. In addition to the bone marrow, other human organs may also be present, particularly thy us, lymph node, skin, liver, pancreas, tonsil, appendix, epithelium, kidney, etc. One or more mesenteric or peripheral lymph nodes may be introduced. The various organs may be introduced conveniently in the kidney capsule, mammary fat pad, particularly the fourth mammary fat pad for a mouse, the popliteal fossa, or other convenient site where the organ may be vascularized, lymphatic vessels connected and maintained by the host for a reasonable period of time, usually at least four weeks, preferably at least six weeks. Thus, the non-human mammal may provide for a significant portion of the human hematopoietic system with the continued regeneration of stem cells and generation of more than fifty percent (50%) of the different types of committed or mature cells. In this manner cells of the myelomonocytic lineage, such as macrophages, mast cells, neutrophils (PMNs) , eosinophils, basophils, monocytes, megakaryocytes, etc. or the lymphoid lineage, B lymphocytes, T lymphocytes, NK cells, ADCC cells, LAK cells, TIL cells, etc. may be propagated, studied or used for various purposes. The subject host may find a wide variety of uses by virtue of the significant presence of circulating human blood cells in the non-human host. The host may be employed for the production of human monoclonal antibodies or immortalized T lymphocytes. The host may be immunized in accordance with conventional ways with an appropriate immunogen, which may be injected intravascularly at an appropriate site. Of particular interest is injection at a site which is drained by a human lymph node introduced into the host. The immunized host may receive one or more booster shots, followed by removal of a lymphoid organ, e.g. lymph node, followed by immortalization, conveniently by fusion with an appropriate yeloid cell or transfection with Epstein-Barr virus. Alternatively, the cells may be cloned by any convenient means, at limiting dilution to provide for individual clones, and the supernatants of the individual clones may be screened for the binding affinity of the antibodies present in the host.
Alternatively, the host may be used for studying T and B lymphocyte interactions, in determining the manner of stimulation of T and B lymphocytes and the mechanism for the immune reaction. Individual lymphocytes may be cloned to evaluate their role in disease protection against disease, identify slg or T cell receptor variable regions, isolate the DNA encoding the variable regions, or the like. Various immunogens may be screened for their abilities as vaccines in producing antibodies which may be effective in neutralizing pathogens and the cells or DNA provide a source of the variable regions associated with the response.
Pathogens of interest are those particularly associated with humans, such as HIV, HTLV-I, and - II, human papilloma virus, cytomegalovirus, Epstein- Barr virus, hepatitis B virus, non-A, non-B hepatitis, chlamydia, malaria (Falciparum) etc. Also, the subject system may be used in the study of cancer in attempting to develop cytotoxic cells specific for a particular cancer type.
The following examples are offered by way of illustration and not by way of limitation.
EXPERIMENTAL Bone pieces were transplanted into either untreated mice (6-10 weeks old) or mice (6-10 weeks old) pre-treated with radiation from a cesium 137 source. The mice are treated with either whole body irradiation or irradiation of the long bones. Mice treated with whole body irradiation receive 200 to 400 rads on a single dose. Alternatively, the mice are treated with 600 rads after shielding of the thorax and abdomen with a lead shield. The mice are anesthetized with Nembutol prior to shielded irradiation. The mice were CB-17.5 scid/scid mice. In addition, some of the mice were treated with exogenous human IL-3 in two lμg doses per day. The fetal bone pieces are prepared from fetuses of 18-22 week gestation by first removing all of the soft tissues and cartilagenous portions from the bone. The bones are then cross sectioned at 2 - 5 mm intervals. The mice are prepared by injection ip of Nembutol. Once the mice are fully anesthetized, a 1 cm incision is made in the skin as well as the underlying peritoneum. The bone fragments are then placed into the peritoneal cavity randomly. Approximately 25mm to 1.5cm of a femur, tibia or humerus is implanted into each mouse. The mice which are irradiated are irradiated within 24 hours of the subsequent implantation of the fetal bone.
After two to six weeks, the percentage of human cells in the peripheral blood is determined by employing monoclonal antibodies specific for human hematopoietic cell markers. In un-irradiated mice a low level of human cells is observed in the peripheral blood by FACScan. The positive cells vary from about 0 - 1.6 percent. Furthermore, injection of IL-3 does not significantly affect the percentage of positive cells. In contrast, the mice irradiated before marrow transplantation showed between 1.5 and 30 percent human cells in the peripheral blood. These cells appear to be of the myelomonocytic lineage. They show no staining with T or B cell markers (CD 3, 4, 8, 19), although they do stain for the myeloid cell marker (CD33) . Furthermore, the human cells have a high side scatter profile indicating that they are of the myelomonocytic lineage. In addition, two of the four mice which were irradiated before implantation show low but significant levels of serum immunoglobulin after six weeks.
The above results demonstrate that one can obtain high levels of human cells in an immunocompromised mouse at least partially ablated of endogenous bone marrow, by implanting relatively large amounts of human bone marrow, particularly with the stromal bone tissue. The human bone marrow implanted after irradiation provides a mouse which results in substantial numbers of cells of human myelomonocytic lineage and can provide for cells of other lineages as well, such as lymphoid. By appropriate combinations of tissue in conjunction with the bone marrow one can provide inside the immunocompromised mouse a hematopoietic system mimicking the human hematopoietic system. Thus, one may study various processes, compounds, drugs, or the like, and their effect on the human hematopoietic system by cytologic or histologic analysis, selection, staining, or the like.
All publications and patent applications mentioned in this specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A non-primate genetically immunocompromised mammalian host comprising at least two percent circulating human blood cells of the total number of circulating blood cells in the blood stream of said host, said host being produced by substantially ablating endogenous bone marrow and implanting human bone marrow.
2. A non-primate host according to Claim 1, wherein said ablating is by at least sublethal irradiation.
3. A non-primate host according to Claim 1, wherein said human bone marrow is bone tissue.
4. A non-primate host according to Claim 1, wherein said human bone marrow is dispersed bone marrow.
5. A non-primate host according to Claim 1, wherein said host is murine.
6. A murine genetically immunocompromised host, wherein said host lacks functional B and T lymphocytes as a result of a genetic defect, said host comprising at least five percent circulating human blood cells of the total number of circulating blood cells in the blood stream of said host, said host being produced by substantially ablating endogenous bone marrow and implanting human bone marrow.
7. A murine host according to Claim 6, wherein said host is a CB-17 scid/scid mouse or progeny thereof.
8. A murine host according to Claim 7, comprising at least twenty-five percent of circulating human blood cells.
9. A.murine host according to Claim 6 , comprising human fetal bone marrow.
10. A murine host according to Claim 6 , comprising at least one of human lymph node, liver or thymus tissue.
11. A murine host according to Claim 6, wherein said host is at least six weeks old from the time of implantation of said human bone marrow.
12. A murine host according to Claim 6 comprising circulating human immunoglobulins.
13. A murine host according to Claim 6, wherein said murine host is irradiated with at least 200 rad.
EP19910911224 1990-05-25 1991-05-13 Human peripheral blood cells in an immunocompromised host Withdrawn EP0484512A4 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US52921790A 1990-05-25 1990-05-25
US529217 1990-05-25

Publications (2)

Publication Number Publication Date
EP0484512A1 EP0484512A1 (en) 1992-05-13
EP0484512A4 true EP0484512A4 (en) 1993-05-05

Family

ID=24108994

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19910911224 Withdrawn EP0484512A4 (en) 1990-05-25 1991-05-13 Human peripheral blood cells in an immunocompromised host

Country Status (4)

Country Link
EP (1) EP0484512A4 (en)
JP (1) JPH05500617A (en)
CA (1) CA2064075A1 (en)
WO (1) WO1991018615A1 (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5652373A (en) * 1990-01-15 1997-07-29 Yeda Research And Development Co. Ltd. Engraftment and development of xenogeneic cells in normal mammals having reconstituted hematopoetic deficient immune systems
US5849288A (en) * 1990-01-15 1998-12-15 Yeda Research And Development Co. Ltd. Method for production of monoclonal antibodies in chimeric mice or rats having xenogeneic antibody-producing cells
CA2063408A1 (en) * 1990-05-03 1991-11-04 Susan Mayo Human lymphoid tissue in an immunocompromised host
US5804160A (en) * 1991-06-04 1998-09-08 Yeda Research And Development Co. Ltd Animal model for hepatitis virus infection
US5858328A (en) * 1991-06-04 1999-01-12 Yeda Research And Development Co. Ltd. Animal model for hepatitis virus infection
US5849987A (en) * 1992-06-02 1998-12-15 Yeda Research And Development Co. Ltd. Animal model for hepatitis virus infection
EP0539970B1 (en) * 1991-10-30 1999-05-26 Idemitsu Kosan Company Limited Methods for producing human lymphocytes and human monoclonal antibodies, and human monoclonal antibodies produced thereby
US6353150B1 (en) * 1991-11-22 2002-03-05 Hsc Research And Development Limited Partnership Chimeric mammals with human hematopoietic cells
US5866757A (en) * 1992-06-02 1999-02-02 Yeda Research And Development Co. Ltd. Engraftment and development of xenogeneic cells in normal mammals having reconstituted hematopoetic deficient immune systems
EP0659046A1 (en) * 1992-09-11 1995-06-28 The Regents of The University of Michigan Non-human animal with xenograft of on airway populated by human cells
ATE221382T1 (en) * 1993-05-24 2002-08-15 Ximerex Inc THE GENERATION OF TOLERANCE AGAINST FOREIGN TRANSPLANTS THROUGH TOLEROGENESIS WITH THE HELP OF SUBSTITUTE LIVING BEINGS
DK2485583T3 (en) 2009-10-06 2016-10-03 Regeneron Pharma GENETICALLY MODIFIED MICE AND inoculation
EP4233537A3 (en) 2011-02-15 2023-09-13 Regeneron Pharmaceuticals, Inc. Humanized m-csf mice and uses thereof
CN108782459B (en) 2012-09-07 2021-11-05 再生元制药公司 Genetically modified non-human animals and methods of use thereof
EP3556206B1 (en) 2012-11-05 2021-06-02 Regeneron Pharmaceuticals, Inc. Genetically modified non-human animals and methods of use thereof
MX2016014995A (en) 2014-05-19 2017-03-31 Regeneron Pharma Genetically modified non-human animals expressing human epo.
RU2730599C2 (en) 2015-04-13 2020-08-24 Ридженерон Фармасьютикалз, Инк. Humanised mice with sirpa-il15 nokin and methods for using them

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0322240A2 (en) * 1987-12-23 1989-06-28 The Board Of Trustees Of The Leland Stanford Junior University Chimeric immunocompromised mammals and their use
WO1989012823A1 (en) * 1988-06-14 1989-12-28 Medical Biology Institute Human immune system in non-human animal

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0322240A2 (en) * 1987-12-23 1989-06-28 The Board Of Trustees Of The Leland Stanford Junior University Chimeric immunocompromised mammals and their use
WO1989012823A1 (en) * 1988-06-14 1989-12-28 Medical Biology Institute Human immune system in non-human animal

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BLOOD vol. 74, no. 5, October 1989, USA, pages 1537 - 1544 FULOP, G.M. & PHILIPS, R.A. 'Use of scid mice to identify and quantitate lymphoid-restricted stem cells in long-term bone marrow cultures' *
SCIENCE vol. 241, 23 September 1988, LANCASTER, PA US pages 1581 - 1583 YANCOPOULOS, G.D. & ALT, F.W. 'Reconstruction of an immune system' *
See also references of WO9118615A1 *

Also Published As

Publication number Publication date
JPH05500617A (en) 1993-02-12
WO1991018615A1 (en) 1991-12-12
EP0484512A1 (en) 1992-05-13
CA2064075A1 (en) 1991-11-26

Similar Documents

Publication Publication Date Title
EP0484512A4 (en) Human peripheral blood cells in an immunocompromised host
Mueller et al. Potential of the scid mouse as a host for human tumors
JP3753321B2 (en) Method for producing mouse suitable for engraftment, differentiation and proliferation of heterologous cells, mouse produced by the method and use of the mouse
Martinov et al. Building the next generation of humanized hemato-lymphoid system mice
US5633426A (en) In vivo use of human bone marrow for investigation and production
Naito et al. Severe osteopetrosis, defective interleukin‐1 signalling and lymph node organogenesis in TRAF6‐deficient mice
DK2448967T3 (en) NON-HUMAN MAMMALS MODEL OF CANCER human hematopoietic
Mosier Immunodeficient mice xenografted with human lymphoid cells: new models for in vivo studies of human immunobiology and infectious diseases
WO2011002727A1 (en) Methods of producing humanized non-human mammals
US20100115642A1 (en) Xenogenic immune system in a non-human mammal
Murphy et al. Recombinant human growth hormone promotes human lymphocyte engraftment in immunodeficient mice and results in an increased incidence of human Epstein Barr virus-induced B-cell lymphoma
JP4906059B2 (en) Screening method for human platelet number regulator
US5434341A (en) Xenogeneic lymph node in mammary fat pad
US6455756B1 (en) Long term xenogeneic myeloid and lymphoid cell production in chimeric immunocompromised mice
GRÖNVIK et al. Monoclonal Antibodies against Murine Neonatal and Pregnancy‐Associated Natural Suppressor Cells Induce Resorption of the Fetus
Tyan Thymus: role in maturation of fetal lymphoid precursors
Leblond et al. The SCID mouse mutant: definition and potential use as a model for immune and hematological disorders
Eswaraka et al. Humanized mice and PDX models
US7259290B1 (en) Animal model of human hematopoietic tumor
US9173383B2 (en) Recombinant non-human mammalian model for hepatitis infection and immunopathogenesis
Camacho et al. Characterization of the NOD/scid-[Tg] DR1 mouse expressing HLA-DRB1∗ 01 transgene: a model of SCID-hu mouse for vaccine development
Boniver et al. Target cells and thymus microenvironment in the pathogenesis of thymic lymphomas in C57BL/Ka mice
Bubbers Single-gene abrogation of spontaneous pleomorphic SJL/J mouse reticulum cell sarcoma expression
Wagner et al. c-fos and polyoma middle T oncogene expression in transgenic mice and embryonal stem cell chimaeras
Riesenfeld et al. Lymphoma development from transplanted murine leukemia virus infected organ cultured thymuses: Inhibitory effect of in vitro interferon treatment

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

AK Designated contracting states

Kind code of ref document: A1

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

17P Request for examination filed

Effective date: 19920519

A4 Supplementary search report drawn up and despatched

Effective date: 19930316

AK Designated contracting states

Kind code of ref document: A4

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

17Q First examination report despatched

Effective date: 19950829

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: 19960110