WO2014207009A2 - Methods and kits for determining whether a nk cell is activated and is able to proliferate - Google Patents

Methods and kits for determining whether a nk cell is activated and is able to proliferate Download PDF

Info

Publication number
WO2014207009A2
WO2014207009A2 PCT/EP2014/063329 EP2014063329W WO2014207009A2 WO 2014207009 A2 WO2014207009 A2 WO 2014207009A2 EP 2014063329 W EP2014063329 W EP 2014063329W WO 2014207009 A2 WO2014207009 A2 WO 2014207009A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
population
cell
cd45ra
cd45ro
Prior art date
Application number
PCT/EP2014/063329
Other languages
French (fr)
Other versions
WO2014207009A3 (en
Inventor
Martin VILLALBA GONZALEZ
Amélie CORNILLON
Ewelina KRZYWINSKA
Jean-François ROSSI
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université de Montpellier I
Centre Hospitalier Universitaire De Montpellier
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Université de Montpellier I, Centre Hospitalier Universitaire De Montpellier filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2014207009A2 publication Critical patent/WO2014207009A2/en
Publication of WO2014207009A3 publication Critical patent/WO2014207009A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/42Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving phosphatase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46434Antigens related to induction of tolerance to non-self
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56972White blood cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70589CD45
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to methods and kits for determining whether a NK cell is activated and is able to proliferate.
  • Natural Killer (NK) cells are members of the lymphocyte lineage and belong to the innate immune system. They show natural cytotoxicity and produce cytokines l ' 2 . The majority of human NK cells in peripheral blood are CD3 ⁇ CD56 dim cells while the minority shows a CD3 " CD56 bngth phenotype. The last population shines at cytokine production whereas CD56 dim cells show mainly cytotoxic activity 3 . In vitro evidence indicates that CD56 bnght cells are precursors of CD56 dim cells, which can also be the case in vivo .
  • NK cells mostly target cells lacking major histocompatibility complex-I (MHC-I), which include transformed or virus-infected cells, which downregulate MHC-I expression to evade recognition by cytotoxic T lymphocytes (CTL).
  • MHC-I major histocompatibility complex-I
  • CTL cytotoxic T lymphocytes
  • the main NK cell inhibitory receptors recognize MHC-I complexes and include NKG2A, which recognizes HLA-E, and Killer-cell Immunoglobulin- like Receptors (KIRs), which recognize the self classical class I molecules HLA-A, -B and -C.
  • NK cell receptors perceive stress and/or non-self ligands on cells, i.e. the stress-induced ligands UL16-binding protein (ULBP) and MHC class I polypeptide-related sequence (MIC) are recognized by the activating receptor NKG2D.
  • ULBP stress-induced ligands UL16-binding protein
  • MIC MHC class I polypeptide-related sequence
  • NK cells are an interesting option to treat these patients because clinical-grade production of NK cells has proven efficient 6 , and NK cell-mediated therapy after hematopoietic cell transplantation seems safe 7"9 .
  • NK cells are not a homogenous population, there are different subsets keeping different physiological activities. It would be interesting to identify the populations with higher anti-tumor activity and select them for expansion and/or patient infusion.
  • CD45 is a protein tyrosine phosphatase (PTP) encoded by the PTPRC gene, which is specifically expressed in hematopoietic cells 10 .
  • PTPs regulate cellular processes including differentiation, mitotic cycle, cell growth and oncogenic transformation n .
  • CD45 regulates receptor signaling by direct interaction with components of the receptor complexes or by activating and dephosphorylating various Src family kinases (SFK) i.e. Lck 12 . But it can inhibit cytokine receptor signaling by inhibiting JAK kinases 13 or by dephosphorylating the activating residues of Src 12 .
  • SFK Src family kinases
  • JAK kinases 13 i.e. Lck 12
  • CD45 levels increase with cell maturation 14 .
  • the CD45 family comprises several members derived of a single complex gene 14 .
  • Naive T lymphocytes are usually positive for the long CD45RA isoform.
  • Activated and memory T cells express CD45RO, the shortest CD45 isoform by activation-induced alternative splicing of CD45 pre- mRNA 14 ⁇ 17 .
  • CD45RO the shortest CD45 isoform by activation-induced alternative splicing of CD45 pre- mRNA 14 ⁇ 17 .
  • Most studies in CD45 function have been developed in T cells. Much less is known about its function on NK cells, although it is commonly accepted that CD45 positively regulates the activation of these cells through its ability to dephosphorylate the inhibitory site of SFKs. This is particularly true for the activation that leads to the production of cytokines and chemokines, whereas cytotoxicity is only slightly impaired in NK cells derived from CD45- deficient mice 18 ⁇ 20 .
  • CD45 -deficient NK cells show increased basal phosphorylation of multiple phosphoproteins suggesting that CD45 may also dephosphorylate other substrates in NK cells, including the activating tyrosine residue of SFKs 20 .
  • the role of CD45 in NK cells is an open issue, although it could depend on the type and strength of the activation.
  • CD45 -deficient NK cells do not protect mice from cytomegalovirus infection due to impaired function of all immunoreceptor tyrosine-based activation motif (ITAM)-dependent NK cell functions, including degranulation 21 .
  • ITAM immunoreceptor tyrosine-based activation motif
  • the present invention relates to methods and kits for determining whether a NK cell is activated and is able to proliferate.
  • NK cells The use of alloreactive NK cells seems a promising co-treatment or an alternative to allogenic HSCT, which has greatly increased the clinical interest for these lymphocytes. However, it is important to identify the different NK cell subpopulations, in particular those with superior antitumor activity.
  • CD45 the ubiquitous lymphocyte marker CD45 to identify different ex vivo and in vitro expanded NK cell populations from PBL and UCBL.
  • NK cells from healthy donors express high CD45 levels that correlate with the expression of maturation markers.
  • healthy donor NK cells are exclusively CD45RA and in vitro IL- 2-mediated activation induces CD45RO expression with down regulation of CD45RA. This effect is enhanced when NK cells encounter their targets.
  • NK cells After in vitro activation, NK cells increase in size and granularity, mainly in a newly identified CD45RA + RO + population, which shows the higher CD 16 expression and degranulating activity.
  • Some blood borne cancer patients and all tested bone marrow transplanted patients show CD45RO RA NK cells and a new population of CD45RA dim RO + NK cells.
  • Some cancer patients with normal CD45RA expression in blood show CD45RA dim RO dim/+ and/or CD45RO + RA ⁇ NK cells.
  • long term survivors of hematological malignancies that show abnormally elevated numbers of NK cells show almost exclusively CD45RA NK cells. Taking these data together suggest that CD45RO is mainly a marker of activation/proliferation and not a marker of memory NK cells; or that memory NK cells do not exist in vivo.
  • an aspect of the invention relates to a method for determining whether a NK cell is activated and is able to proliferate comprising the steps consisting of i) determining the expression level of CD45RO at the cell surface of the NK cell, ii) comparing the expression level determined in step i) with a predetermined reference value and iii) concluding that the NK cell is activated and is able to proliferate when the expression level determined at step i) is higher than the predetermined reference value.
  • the term "NK cell” has its general meaning in the art and refers to natural killer (NK) cell.
  • NK natural killer
  • NK cells typically are prepared from a blood sample.
  • blood sample means a whole blood sample obtained from a subject (e.g. an individual for which it is interesting whether a population of NK cells is activated and is able to proliferate).
  • the NK cell population is prepared from a PBMC sample.
  • PBMC peripheral blood mononuclear cells
  • unfractionated PBMC refers to whole PBMC, i.e. to a population of white blood cells having a round nucleus, which has not been enriched for a given sub-population.
  • the PBMC sample may have been subjected to a selection step to contain non-adherent PBMC (which contain T cells, B cells, natural killer (NK) cells, NK T cells and DC precursors).
  • a PBMC sample according to the invention therefore contains lymphocytes (B cells, T cells, NK cells, NKT cells).
  • lymphocytes B cells, T cells, NK cells, NKT cells.
  • these cells can be extracted from whole blood using Ficoll, a hydrophilic polysaccharide that separates layers of blood, with the PBMC forming a cell ring under a layer of plasma.
  • PBMC can be extracted from whole blood using a hypotonic lysis buffer, which will preferentially lyse red blood cells. Such procedures are known to the expert in the art.
  • the NK cells can be prepared by Percoll density gradients, by negative depletion methods or by FACS sorting methods. These cells can also be isolated by column immunoadsorption using an avidine-biotin system or by immunoselection using microbeads grafted with antibodies. It is also possible to use combinations of these different techniques, optionally combined with plastic adherence methods.
  • the NK cells can be prepared by providing blood mononuclear cells depleted of T cells from the donor, activating said cells with phytohemagglutinin (PHA) and culturing said cells with interleukin (IL)-2 and irradiated feeder cells.
  • PHA phytohemagglutinin
  • IL interleukin
  • the population of NK cells is prepared from a subject suffers from a disease, for example from a cancer or an infectious disease.
  • the population of NK cells is prepared from a transplanted subject.
  • grafts include, but are not limited to transplanted heart, kidney, lung, liver, pancreas, pancreatic islets, brain tissue, stomach, large intestine, small intestine, cornea, skin, trachea, bone, bone marrow, muscle, or bladder.
  • CD45 has its general meaning in the art and refers to the protein tyrosine phosphatase (PTP) encoded by the PTPRC gene, which is specifically expressed in hematopoietic cells 10 .
  • CD45 regulates receptor signalling by direct interaction with components of the receptor complexes or by activating and dephosphorylating various Src family kinases (SFK) i.e. Lck 12. But it can inhibit cytokine receptor signalling by inhibiting JAK kinases or by dephosphorylating the activating residues of Src 12 .
  • SFK Src family kinases
  • JAK JAK kinases
  • CD45RO Standard methods for detecting the expression of specific surface markers at cell surface (e.g.
  • the step consisting of determining the expression levels of CD45RO at the NK cell surface may consist in collecting a NK cell population from a subject and using at least one differential binding partner directed against CD45RO, wherein said NK cells are bound by said binding partners to said CD45RO.
  • binding partner directed against the CD45RO refers to any molecule (natural or not) that is able to bind the CD45RO with high affinity.
  • Said binding partners include but are not limited to antibodies, aptamer, and peptides.
  • the binding partners may be antibodies that may be polyclonal or monoclonal, preferably monoclonal, specifically directed against said CD45RO. In another embodiment, the binding partners may be a set of ap tamers.
  • Polyclonal antibodies of the invention or a fragment thereof can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • Various adjuvants known in the art can be used to enhance antibody production.
  • antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
  • Monoclonal antibodies of the invention or a fragment thereof can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture.
  • Techniques for production and isolation include but are not limited to the hybridoma technique originally; the human B-cell hybridoma technique; and the EBV-hybridoma technique.
  • the binding partners may be aptamers.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA or RNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
  • Peptide aptamers consist of conformationally constrained antibody variable regions displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods.
  • binding partners of the invention such as antibodies or aptamers may be labelled with a detectable molecule or substance, such as preferentially a fluorescent molecule, or a radioactive molecule or any others labels known in the art.
  • Labels are known in the art that generally provide (either directly or indirectly) a signal.
  • the term "labelled", with regard to the antibody or aptamer, is intended to encompass direct labelling of the antibody or aptamer by coupling (i.e., physically linking) a detectable substance, such as a fluorophore [e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)]) or a radioactive agent to the antibody or aptamer, as well as indirect labelling of the probe or antibody by reactivity with a detectable substance.
  • FITC fluorescein isothiocyanate
  • PE phycoerythrin
  • Indocyanine Cy5
  • An antibody or aptamer of the invention may be labelled with a radioactive molecule by any method known in the art.
  • radioactive molecules include but are not limited radioactive atom for scintigraphic studies such as 1123, 1124, Inl 11, Rel86, Rel88.
  • the antibodies against the CD45RO are already conjugated to a fluorophore (e.g. FITC-conjugated and/or PE-conjugated).
  • a fluorophore e.g. FITC-conjugated and/or PE-conjugated.
  • the aforementioned assays may involve the binding of the binding partners (ie. antibodies or aptamers) to a solid support.
  • the solid surface could a microtitration plate coated with the binding partner.
  • NK cells specifically bound to the binding partner may be detected with an antibody to a common NK cell marker
  • the solid surfaces may be beads, such as activated beads, magnetically responsive beads. Beads may be made of different materials, including but not limited to glass, plastic, polystyrene, and acrylic.
  • the beads are preferably fluorescently labelled. In a preferred embodiment, fluorescent beads are those contained in TruCount(TM) tubes, available from Becton Dickinson Biosciences, (San Jose, California).
  • methods of flow cytometry are preferred methods for measuring the level of CD45RO at the NK cell surface.
  • Said methods are well known in the art.
  • fluorescence activated cell sorting FACS
  • a FACS method such as described in Example here below may be used to measuring the level of CD45RO at the NK cell surface.
  • the method further comprises the steps consisting of i) determining the expression level of CD45RA at the cell surface of the NK cell, ii) comparing the expression level determined in step i) with a predetermined reference value and iii) concluding that NK cell is activated and is able to proliferate when the expression level determined at step i) is lower than the predetermined reference value.
  • the techniques described for determining the expression of CD45RO may be applied mutatis mutandis for determining the expression of CD45RA in the population of NK cells.
  • the present invention also relates to a method for isolating a population of NK cells capable of activation and proliferation comprising the steps consisting of i) providing a population of NK cells and ii) selecting the population of CD45RO + NK cells.
  • the present invention also relates to a method for isolating a population of NK cells capable of activation, proliferation and high level of cytotoxicity comprising the steps consisting of i) providing a population of NK cells and ii) selecting the population of CD45RA + RO + NK cells.
  • NK cells typically are isolated from a blood sample obtained from a subject.
  • the methods are particularly suitable to be implemented in in vitro protocols of expansion of NK cells.
  • the protocol of expansion may consist in contacting a population of NK cells with an amount of IL-2.
  • the protocol of expansion may also consist of the protocol described in WO2012146702. Briefly the protocol comprise the steps consisting of (i) contacting a population of NK cells with at least one accessory cell (i.e. a cell wherein the expression of one gene encoding for a Killer-Cell Immunoglobulin-like Receptor(s) (KIR) ligand is inhibited) under conditions and for a duration sufficient to induce activation of the population of NK cells; (ii) recovering said activated population NK cells.
  • KIR Killer-Cell Immunoglobulin-like Receptor
  • the method of the invention may be performed for determining whether the protocol leads to a population of NK cells that is activated and that is capable to proliferate, and/or for isolating at the end of the protocol the population of NK cells that that is activated and that is capable to proliferate.
  • the methods of the invention may also suitable for determining whether an agent is able to induce activation and proliferation of NK cells.
  • said agent may represent a pharmaceutical agent for the treatment of a disease (e.g. a cancer or an infectious disease), and the methods of the invention may thus be useful for determining whether said pharmaceutical agent is able to induce the development of population of NK cells that is activated and that is able to proliferate.
  • said pharmaceutical agent may be selected from the group of recombinant interleukins, recombinant cytokines, recombinant growth factors, and antibodies...
  • an aspect of the invention relates to a method for monitoring the treatment of a subject with a pharmaceutical agent comprising the steps consisting of administering the subject with the pharmaceutical agent, providing a blood sample or PBMC sample from the subject after said treatment, and determining whether said blood or PBMC sample contain a population of CD45RO+ NK cells (or CD45RA+RO+ NK cells).
  • the methods of the invention are also particularly suitable for the immunopro filing of a subject, typically suffering from an infectious disease or a cancer.
  • a subject harbours a population of CD45RO+ NK cells, more particularly a population of CD45RO+RA- NK cells or a population of CD45RA+RO+ NK cells, then it can be concluded that the patient harbours an efficient immune response and the subject can have a good prognostic.
  • a further aspect of the invention relates to a method for determining the survival time of a subject suffering of a cancer or an infectious disease comprising the steps consisting of i) detecting in a blood sample obtained from the subject the presence of a population of CD45RO+ NK cells, more particularly a population of CD45RO+RA- NK cells or a population of CD45RA+RO+ NK cells and ii) providing a good prognosis when the population is detected at step i).
  • a further aspect of the invention also relates to a method for determining whether a transplanted subject is at risk of graft rejection comprising the steps consisting of i) detecting in a blood sample obtained from the subject the presence of a population of CD45RO+ NK cells, more particularly a population of CD45RO+RA- NK cells or a population of CD45RA+RO+ NK cells and ii) concluding that the transplanted subject is at risk of graft rejection when the population is detected at step i).
  • the method may be used to evaluate survival of a variety of different types of grafts.
  • Grafts of interest include, but are not limited to: transplanted heart, kidney, lung, liver, pancreas, pancreatic islets, brain tissue, stomach, large intestine, small intestine, cornea, skin, trachea, bone, bone marrow, muscle, or bladder.
  • the population of NK cells isolated by the method of the invention may be suitable for testing whether a therapeutic antibody is capable to induce an antibody dependent cellular cytotoxicity (ADCC).
  • ADCC antibody dependent cellular cytotoxicity
  • Antibody-dependent cell-mediated cytotoxicity refers to a form of cytotoxicity in which secreted antibodies bound onto Fc receptors (FcRs) present on NK cells enable these cytotoxic effector cells to bind specifically to an antigen-bearing target cell and subsequently kill the target cell.
  • FcRs Fc receptors
  • the antibody is incubated with a population of cells expressing the antigen (i.e. the population of "target cells”) that is recognized by the antibody in presence of an isolated population of NK cells according to the invention. Then the cytoxicity on the population of target cells may be then evaluated.
  • the population of NK cells isolated by the method of the invention may be then used in therapeutic protocols such as methods for treating cancers.
  • the population that is more interesting, although not exclusively, for a therapeutic purpose is the population of CD45RA+RO+ NK cells.
  • a method of transplanting allogeneic graft into a patient in need thereof may comprise the steps consisting of a) administering to said patient an effective amount of the activated NK cells as described above; and, b) transplanting the allogeneic graft into the recipient.
  • This method of transplanting allogeneic graft, more particularly hematopoietic graft can be applied for reducing the GVHD, for decreasing the intensity of the conditioning regimen, for treating a subject having hematologic disorder, more particularly leukemia, for treating or preventing an infection in a recipient of allogeneic graft, for enhancing immune reconstitution in an allogeneic graft recipient, for proceeding a hematopoietic graft with a greater T cell content, for increasing the engraftment, for reducing the graft rejection, for avoiding the tumor relapse and/or for conditioning a patient in need of a hematopoietic graft.
  • Hematologic disorder includes neoplastic proliferation of hematopoietic cells.
  • said hematologic disorder is selected from the group consisting of lymphoblastic leukemia, acute or chronic myelogenous leukemia, Hodgkin's lymphoma, Non-Hodgkin's lymphoma, myelodysplasia syndrome, multiple myeloma, and chronic lymphocytic leukemia.
  • said hematologic disorder is a leukemia, more preferably myeloid leukemia.
  • Hematologic disorder also includes non-malignant disorders such as an inherited erythrocyte abnormalities, an inherited immune system disorders or a hemoglobinopathy, e.g.
  • the isolated population of NK cells of the invention e.g. CD45RA+RO+ NK cells
  • KIR Killer-Cell Immunoglobulin- like Receptor
  • the physicians may perform HLA genotyping of patients. They could then order the isolated population of NK cells of the invention missing expression of the required KIRs (e.g. an AML patient has tumor cells that do not express ligands for KIR2DL1).
  • NK cells expressing only one KIR may then be provided and used in the therapeutic protocol.
  • said allogeneic graft is a hematopoietic graft.
  • said hematopoietic graft is a bone marrow transplant.
  • said patient is treated for leukemia, more preferably myeloid leukemia, optionally an acute or chronic myeloid leukemia.
  • the isolated population of NK cells according to the invention and allogeneic graft are administered simultaneously.
  • the isolated population of NK cells of the invention are administered prior to the allogeneic graft.
  • the efficient amount of the isolated population of NK cells of the invention administered to the recipient can be between about 0.05 10 6 and about 100 10 6 cells/kg of recipient's body weight.
  • the efficient amount of hematopoietic cells administered to the recipient can be between about 0.2 10 6 and about 10 10 6 CD34+ cells/kg of recipient's body weight,
  • the graft comprises a maximum of 1 10 5 CD3+ cells/kg of recipient's body weight.
  • NK cells of the invention and hematopoietic cells are typically administered to the recipient in a pharmaceutically acceptable carrier by intravenous infusion.
  • Carriers for these cells can include but are not limited to solutions of phosphate buffered saline (PBS) containing a mixture of salts in physiologic concentrations.
  • PBS phosphate buffered saline
  • the hematopoietic cells can be provided by bone marrow cells, mobilized peripheral blood cells or cord blood cells.
  • the bone marrow cells can be obtained from the donor by standard done bone marrow aspiration techniques know in the art, for example by aspiration of marrow from the iliac crest.
  • Peripheral blood stem cells are obtained after stimulation of the donor with a single or several doses of a suitable cytokine, such as granulocyte colony- stimulating factor (G-CSF), granulocyte/macrophage colony- stimulating factor (GM-CSF) and interleukin-3 (IL-3).
  • G-CSF granulocyte colony- stimulating factor
  • GM-CSF granulocyte/macrophage colony- stimulating factor
  • IL-3 interleukin-3
  • the donor is stimulated with G-CSF.
  • the hematopoietic cells can be T-cell depleted.
  • T-cell depletion of bone marrow or of peripheral blood cell may be carried out by any known technique, for example, by soybean agglutination and E-rosetting with sheep red blood cells as described.
  • the host patient is conditioned prior to the transplantation of the allogeneic graft. Conditioning may be carried out under sublethal, lethal or supralethal conditions, for example by total body irradiation (TBI) and/or by treatment with myelo- reductive or myelo-ablative and immunosuppressive agents.
  • TBI total body irradiation
  • myelo- reductive or myelo-ablative and immunosuppressive agents e.g., a lethal dose of irradiation is within the range of 7-9,5 Gy TBI
  • a sublethal dose is within the range of 3-7 Gy TBI
  • a supralethal dose is within the range of 9,5-16 Gy TBI.
  • immunosuppressive agent used in transplantation to control the rejection can be used according to the invention, such as prednisone, methyl prednisolone, azathioprine, cyclophosphamide, cyclosporine, monoclonal antibodies against T-cells, e.g. OKT3, and antisera to human lymphocytes (antilymphocyte globulin - ALS) or to thymus cells (antithymocyte globulin - ATG).
  • myelo-ablative agents that can be used according to the invention are busulphan, dimethyl myleran and thiotepa.
  • the advantage of the administration of the isolated population of NK cells of the invention is the possibility to reduce the intensity of the conditioning regimen.
  • the conditioning regimen can be reduced to the intensity of conditioning regimen adopted for matched human transplants.
  • a reduced version of a high- intensity regimen according to the present invention includes Fludarabine at the total dose of 200 mg/M2, Thiotepa 5 mg/Kg, and Melphalan 70 mg/M2, plus anti-T cell antibodies such as ATG, 20 mg/Kg.
  • the doses of Thiotepa and Melphalan can be increased by 50%.
  • such conditioning regimen is highly toxic and some patients are unable to withstand such toxicity. Therefore, the present invention makes possible the allogeneic graft for these patients.
  • the isolated population of NK cells of the invention as prepared according to the invention may be used as first line of treatment in combination with standard total body irradiation (TBI) and/or by treatment with myelo -reductive or myelo-ablative and immunosuppressive agents; or used to control the residual disease after total body irradiation (TBI) and/or by treatment with myelo -reductive or myelo-ablative and immunosuppressive agents.
  • TBI total body irradiation
  • myelo -reductive or myelo-ablative and immunosuppressive agents or used to control the residual disease after total body irradiation (TBI) and/or by treatment with myelo -reductive or myelo-ablative and immunosuppressive agents.
  • the isolated population of NK cells of the invention as prepared according to the invention may also be used for the treatment of other NK-sensitive malignancies including pediatric cancers such as the sarcomas Ewing sarcoma and rhabdomyosarcoma (Cho...Campana clinical cancer Research 16:3901 (2010), neuroblastoma, malignant glioma and, possibly, prostate cancer (Cho, D. & Campana, D. Expansion and activation of natural killer cells for cancer immunotherapy. Korean J Lab Med 29, 89-96 (2009).
  • the isolated population of NK cells of the invention as prepared according to the invention may also be used for the treatment of aggressive cancer with no major drugs in order to improve the response rate and PFS.
  • Typical said cancers include but are not limited to pancreatic cancer, lung cancer, breast and colon cancer.
  • the isolated population of NK cells according to the invention is used in combination with a therapeutic antibody, typically a therapeutic antibody capable of ADCC (e.g. a CD20 antibody).
  • the isolated population of NK cells of the invention as prepared according to the invention may also useful for the treatment of infectious diseases or dysimmune diseases.
  • PBMC and UCB mononuclear cells were respectively collected from peripheral blood samples and UCB units using Histopaque (Invitrogen). Briefly, thirteen ml Histopaque were put in 50 ml centrifugation tubes and 20-30 ml of 1/2 diluted blood in RPMI, (Invitrogen) were slowly added at the top. Tubes were centrifugated 30 minutes at 1600 rpm and 20 °C without break. Mononuclear cells were collected from the interlayer white ring. After washing in RPMI, cells were suspended in complete RPMI medium supplemented with 10% FBS (Invitrogen).
  • cells were stained with 7AAD (Beckman) to identify viable cells and antibodies against surface markers, CD25-FITC, CD45RO-FITC, CD69-PE, CD62L- PE, CD19-PE, CD3-PE, CD19-ECD, CD56-PECy7, CD56-APC, CD3-APC, CD45- APCAlexaFluor750, CD45RA-APCAlexaFluor750, CD16-PacificBlue, CD57-PacificBlue, CD45-KromeOrange, CD16-KromeOrange (Beckman), CD158b-FITC, CD158a-PE, CD107a- HV500 (BD Biosciences), CD158e-Vioblue (Miltenyi), by incubating Ixl0 5 -3xl0 5 cells with different antibodies in PBS 2,5% FBS for 20-30 minutes at 37 °C. Cells were then washed and suspended in 200-250 ⁇ PBS 2,5% FBS. Staining was analyzed
  • Alive lymphocytes were gated using FS/SS and 7AAD staining.
  • B lymphocytes were gated using FS/SS and 7AAD staining.
  • CD 19+ T lymphocytes (CD3+CD56-) and NK cells (CD56+CD3-) were distinguished using respectively CD 19, CD3 and CD56 antibodies.
  • NK cells were separated in 3 distinct populations CD45RA+RO-, CD45RA-RO+ and CD45RA+RO+. These different populations were then analyzed for CD 16, CD69, CD62L, CD57, IFNy and CD 107a expression or size/granularity (FSC/SSC) parameters.
  • PBMC and UCBMC were stimulated for NK amplification and activation for 10 to 20 days.
  • Two different protocols were compared, starting with PBMC/UCBMC 1.10 6 cells/ml. Firstly, stimulation with high dose of IL-2 (1000 U/ml, eBiosciences) added to complete medium. Secondly, co-stimulation with the NK cell target EBV cell line C30, developed in our laboratory, together with IL-2 100 U/ml + IL-15 (5 ng/ml, Miltenyi).
  • Cells were incubated over night at 37°C 5%C02. After incubation, living cells in the wells were counted using a cytometer Muse (Millipore) with the count and viability kit (Millipore). Cells were analyzed by FACS using 7AAD, CD45RO-FITC, CD19-PE, CD56- PECy7, CD3-APC, CD45RA-APCAlexaFluor750, and CD16-PacificBlue for cytotoxicity assay; or 7AAD, CD45RO-FITC, CD19-PE, CD56-PECy7, CD3-APC, CD45RA- APCAlexaFluor750, CD16-KromeOrange and CD107a-HV500 (BD Biosciences) for degranulation assay.
  • 7AAD CD45RO-FITC, CD19-PE, CD56-PECy7, CD3-APC, CD45RA- APCAlexaFluor750, CD16-KromeOrange and CD107a-
  • FACS results were analyzed using the Kaluza software.
  • target cells were gated in living cells (7AAD-) using FS/CD45 plots and FS/CD3 (Jurkat) or FS/CD19 plots.
  • the numbers of living target cells after incubation with effector cells were subtracted to the number of living target cells when incubated alone to give the percentage of target cell lysis.
  • the percentages of CD107a + NK cells were analyzed.
  • NK cell expansion is required for clinical means.
  • the source of these cells can be PBL or UCBL. It is expected that lymphocytes for the second source would be na ' ive and/or less committed into differentiation.
  • CD45 a marker of maturation 14
  • T three main types of human lymphocytes: T, B and NK cells derived from PBL and UCBL.
  • B and NK cells express similar CD45 levels in PBL and UCBL.
  • NK cell population can be divided in two regarding CD56 expression.
  • CD56 bnght are relatively immature, with low cytotoxic activity and high cytokine production and CD56 dim , which are more mature with high cytolytic activity and lower cytokine production 3 .
  • CD56 bnght cells showed lower CD45 expression in NK cells, according to the observation that CD45 levels increased with cell maturation 14 .
  • the median percentage of CD56 bnght was similar in UCBL and PBL 22 .
  • T cells derived from UCBL showed lower CD45 expression than other lymphocytes and than T cells derived from PBL.
  • the lower CD45 expression was probably linked to the stage of maturity because we showed that T cells derived from UCBL expressed lower levels of CD45RO and higher levels of CD45RA , a sign of immaturity 14 ⁇ 17 .
  • B and NK cells were basically equivalent in PBL and UCBL regarding expression of the CD45 isoforms.
  • naive NK cells expressed almost exclusively the isoform CD45RA, although the CD56 bnght NK cells at lower level than other NK cells.
  • NK cells of both origins increased CD45RO expression and decreased CD45RA expression.
  • UCBL T cells also showed the same changes although less pronounced than PBL T cells. In UCBL T cells, these changes were mainly observed at day 10, whereas 3 days after stimulation the changes were minimum and a 20-day stimulation did not increase further the CD45 changes. In contrast, these changes started 3 days after activation in UCBL NK cells. In agreement with previous descriptions 15 , 20 days after stimulation some NK cells recovered CD45RA expression without loosing CD45RO expression. Hence, a new population CD45RA RO + (from now on called CD45RARO) appeared that was mainly absent in T cells.
  • CD45 isoforms during in vitro activation, we compared IL-2 and co-stimulation conditions. The latter reflected incubation with a prototypical NK cell target, in this case an EBV cell line, together with low concentrations of two NK cell activating cytokines: IL-2 and IL-15 26 . After 10 days of co-stimulation NK cells expressed higher CD45RO and lower CD45RA levels compared to IL-2 stimulation. In addition, CD45 isoforms changes appeared faster during co-stimulation although the CD45RARO population was similar after 20-day stimulation.
  • CD69 is used as a general marker of NK cell activation including in vivo 5 .
  • in vitro activation with IL-2 induced a larger increase in CD69 than co- stimulation.
  • co-stimulation induced a larger decrease in CD45RA expression that IL-2.
  • both protocols, CD69 up regulation and CD45RA down regulation were compatible to study NK cell activation and could be used depending of the NK cell activation stimulus.
  • the CD45RA/RO detection method has the advantage of couple two phenomena: CD45RA down regulation and CD45RO up regulation. This should decrease the number of false positives and the artifacts.
  • CD45RARO CD45RA RO
  • CD45RORO showed a biphasic curve with a first peak around day 3 and a second increase around day 12. Co-stimulation induces a faster and more complete decline of CD45 cells and increase in CD45RO cells. The biphasic curve of CD45RARO was more apparent. Whereas we cannot prove it, we believe that the first peak is related to acquisition of CD45RO expression without totally loosing CD45RA expression. The CD56 bngth probably respond faster to stimulus i.e.
  • cytokines could be responsible of this peak. However, we could not study this fact, because NK cell activation increased CD56 levels, and therefore we could not distinguish anymore the two populations CD56 bnght and CD56 dim .
  • the second peak is probably due to expression of CD45RA in the CD45RO population generated after in vitro activation.
  • CD45RA NK cells were mainly small cells with low granularity. After activation, NK cells increased in size and granularity. This was particularly true for CD45RO and, mainly, CD45RARO populations. These results were in agreement with the notion that CD45RO expression was associated to stimulation and that re-acquisition of CD45RA could be an additional sign of activation.
  • CD56 bright cells originates transitory CD56 dim CD62L + CD57 ⁇ cells that started generating higher levels of perforin while keeping high IFN- ⁇ production in response to cytokines 3 ' 27 .
  • CD56 dim CD62L ⁇ CD57 + cells show low response to cytokines but increased cytotoxic capacity 3 ' 28 .
  • UCBL NK cells expressed low CD62L levels and almost no expression of CD57 compared to PBL NK cells in agreement with previous reports 2 ' .
  • In vitro activated UCBL NK cells increased CD62L expression in all CD45 subpopulations; however, the expression decreased with time and even disappeared by day 20 in cells activated by co-stimulation.
  • NK cell activation induced a significantly increase only in the CD45RARO population.
  • PBL NK cells expressed higher levels of both CD62L and CD57. After activation, CD62L barely increased whereas CD57 decreased.
  • in vitro activation of UCBL NK cells did not induce an in vivo model of NK cell differentiation; probably the CD56 dim cells are already fully differentiated even in UCBL.
  • Transplanted patients showed both CD45RA down regulation and CD45RO up regulation, generating a comet-like figure, similar to our observation during in vitro NK cells activation, although less pronounced.
  • CD45RA dim cells which were the first step for the lost of CD45RA before acquisition of CD45RO, and represented the start of the comet tail.
  • CD45RA resting NK cells clearly diminished in these patients.
  • CD56 bnght cells which were CD 16 " suggesting that these were de novo produced NK cells and not CD56 dim cells that gain CD56 expression.
  • CD56 bnght cells were CD62L + CD57 ⁇ and showed a more immature phenotype than the CD56 dim cells, which were CD62L ⁇ CD57 + .
  • CD56 bnght cells were mainly CD45RO cells, whereas CD56 dim cells were mainly CD45RA.
  • CD45RO cells were mainly CD45RO cells, whereas CD56 dim cells were mainly CD45RA.
  • the CD45RO population was mainly CD56 bright CD62L + CD57 " , in contrast to the CD45RA population that shows a CD56 dim CD62L " CD57 + phenotype.
  • CD56 dim cells are usually CD57 + whereas CD56 bnght cells usually express CD62L 27 ' 28 .
  • the increase in CD56 bnght cells compared to healthy donors was probably due to recovery after lymphopenia, which induced a large production of na ' ive cells.
  • NK cell subpopulations in patients with hematological cancers including B-CLL and MM. Five out of 12 of these patients showed down regulation of the CD45RA population and CD45 dim and/or CD45RO up regulation.
  • NK cells could be diluted in the blood of hematological cancer patients preventing their detection.
  • NK cells that were in contact with tumor cells in patients with low numbers of activated NK cells in blood.
  • MM multiple myeloma
  • NK cells in the bone marrow show activated cells in the tumor site, in our case in the bone marrow.
  • the pC9 expressed a low reduction of CD45RA cells.
  • This patient showed a gammopathy and NK cells in the bone marrow showed a higher activation with reduction of CD45RA levels and increase in CD45RO.
  • NK cells in the bone marrow show a decline in the number of CD45RA cells and increased in CD45RA dim and CD45RO cells.
  • CD45RO expression depended on the contact with tumor cells, at least in certain patients.
  • CD45 isoforms in the tumor ganglion of 2 B-CLL patients.
  • CD45RA expression in the tumor site.
  • expression of different CD45 isoforms in vivo can identify NK cell activation and/or proliferation.
  • CD45RARO NK cells identified by co-expression of CD45RO and CD45RA molecules. This CD45RARO population was a minority and we knew if in certain individuals this population could be enlarged after activation.
  • UCB439 which after activation with IL-2 or co-stimulation, showed the atypical phenotype of keeping CD45RA after CD45RO expression, generating a large CD45RARO population. This was more remarkable after co-stimulation.
  • CD45RA hlgh /RO + population which in the other UCBs, corresponded to common CD45RARO population in FSC/SSC behavior.
  • NK alloreactive natural killer
  • UCBT umbilical cord blood transplantation
  • NK cells 1) are not responsible of GvHD; 2) can be injected as "differentiated" cells without the need of long time survival on patient's body; 3) protect from opportunist infections 33 , probably through their immunoregulatory effects on B cells, T cells, macrophages, and more importantly on polymorphonuclear cells (PMNs 35 ).
  • PMNs 35 polymorphonuclear cells
  • CD45 activity is regulated by dimerization and CD45 spontaneously homodimerizes at the plasma membrane, which inhibits its activity 38 .
  • the size of the CD45 extracellular domain is inversely proportional to the extent of CD45 dimerization and auto inhibition 38 . Larger CD45 isoforms such as CD45RA dimerize less efficiently and, accordingly, and perhaps they are more efficient at promoting TCR signaling than are smaller isoforms such as CD45RO 12 .
  • CD45 activity also depends on its plasma membrane localization that depends on the extracellular domain n ' 12 . At least in T cells, too high CD45 activity leads to dephosphorylation of the activating residues in the Src kinases, whereas too low would leave phosphorylated the inhibitory residues.
  • CD45 activity is between this window 14 , and the amount and the specific CD45 isoform would regulate the final activity.
  • NK cell decision to kill depends on activating and inhibitory signals.
  • CD45RA + /RO + NK cells show the maximal cytolytic activity.
  • the presence of both CD45RA and RO could give the NK cells the appropriate level of CD45 activity for efficient signaling to boost the cytolytic activity.
  • CD45 activity is critical for an efficient immune response, because its deficiency results in a severe combined immunodeficiency (SCID) phenotype in both mice 39 41 and humans 42 ' 43 . This could explain the high expression of this phosphatase and its complex regulation.
  • SCID severe combined immunodeficiency
  • IL-2 stimulation induces CD45RA down regulation and CD45RO up regulation in PBL and this occurs first in NK cells.
  • UCBL show a similar behavior.
  • IL-2 was the molecule that drives those changes 16 .
  • other signals independent to IL-2 contribute to CD45RO up regulation and CD45RA down regulation because high IL-2 doses do not reach the same level of changes than co-stimulation with target cells.
  • T cells derived from PBL show more CD45RO expression than T cells derived from UCBL. This suggests that in PBL there are more activated and/or memory cells than in UCBL, what is intuitive. It is fair to assume that in these situations CD45RO cells are memory cells.
  • CD45RO cells are memory cells.
  • an extremely low number of NK cells express CD45RO ex vivo in both PBL and UCBL. At least in vitro, NK cell activation clearly induces CD45RO expression. Hence, it is surprising that ex vivo we find so low numbers of NK cells expressing CD45RO in PBL.
  • CD45RO is not a marker of memory NK cells or CD45RO is specifically lost during ex vivo manipulations in NK cells, although the last possibility is unlikely because NK cells express slightly higher levels of total CD45 than other lymphocyte.
  • CD45RO is mainly a marker of NK cell proliferation in this specific setting and degranulation activity is mostly related to CD45RARO cells, which are however, a minority population.
  • Domaica CI Fuertes MB, Uriarte I, et al. Human natural killer cell maturation defect supports in vivo CD56(bright) to CD56(dim) lineage development. PLoS One. 2012;7(12):e51677.
  • CD45 is a JAK phosphatase and negatively regulates cytokine receptor signalling. Nature. 2001;409(6818):349-354.
  • Hermiston ML, Zikherman J, Zhu JW. CD45, CD148, and Lyp/Pep critical phosphatases regulating Src family kinase signaling networks in immune cells. Immunol Rev. 2009;228(1):288-311.
  • Hesslein DG Takaki R, Hermiston ML, Weiss A, Lanier LL. Dysregulation of signaling pathways in CD45 -deficient NK cells leads to differentially regulated cytotoxicity and cytokine production. Proc Natl Acad Sci USA. 2006;103(18):7012-7017.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Oncology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention relates to methods and kits for determining whether a NK cell is activated and is able to proliferate. In particular, the present invention relates to a method for determining whether a NK cell is activated and is able to proliferate comprising the steps consisting of i) determining the expression level of CD45RO at the cell surface of the NK cell, ii) comparing the expression level determined in step i) with a predetermined reference value and iii) concluding that the NK cell is activated and is able to proliferate when the expression level determined at step i) is higher than the predetermined reference value. The present invention also relates to a population of CD45RA+RO+ NK cells.

Description

METHODS AND KITS FOR DETERMINING WHETHER A NK CELL IS
ACTIVATED AND IS ABLE TO PROLIFERATE
FIELD OF THE INVENTION:
The present invention relates to methods and kits for determining whether a NK cell is activated and is able to proliferate.
BACKGROUND OF THE INVENTION:
Natural Killer (NK) cells are members of the lymphocyte lineage and belong to the innate immune system. They show natural cytotoxicity and produce cytokines l'2. The majority of human NK cells in peripheral blood are CD3~CD56dim cells while the minority shows a CD3" CD56bngth phenotype. The last population shines at cytokine production whereas CD56dim cells show mainly cytotoxic activity 3. In vitro evidence indicates that CD56bnght cells are precursors of CD56dim cells, which can also be the case in vivo . NK cells mostly target cells lacking major histocompatibility complex-I (MHC-I), which include transformed or virus-infected cells, which downregulate MHC-I expression to evade recognition by cytotoxic T lymphocytes (CTL). The "missing self hypothesis proposes that NK cells distinguish target cells from other healthy "self cells based on MHC-I expression. However, NK cell activation depends on a complex signaling mechanism mediated by both activating and inhibitory receptors. The main NK cell inhibitory receptors recognize MHC-I complexes and include NKG2A, which recognizes HLA-E, and Killer-cell Immunoglobulin- like Receptors (KIRs), which recognize the self classical class I molecules HLA-A, -B and -C. The first clinical trial using an anti-KIR that can block KIR-mediated inhibition of natural killer (NK) cells has recently been published 5. It shows absence of toxicity and favorably overall and relapse-free survival compared to reports in comparable patient populations of acute myeloid leukemia (AML). Activating NK cell receptors perceive stress and/or non-self ligands on cells, i.e. the stress-induced ligands UL16-binding protein (ULBP) and MHC class I polypeptide-related sequence (MIC) are recognized by the activating receptor NKG2D.
Long-term survival is limited in a significant number of patients with hematological cancers. NK cells are an interesting option to treat these patients because clinical-grade production of NK cells has proven efficient 6, and NK cell-mediated therapy after hematopoietic cell transplantation seems safe 7"9. However, NK cells are not a homogenous population, there are different subsets keeping different physiological activities. It would be interesting to identify the populations with higher anti-tumor activity and select them for expansion and/or patient infusion.
CD45 is a protein tyrosine phosphatase (PTP) encoded by the PTPRC gene, which is specifically expressed in hematopoietic cells 10. PTPs regulate cellular processes including differentiation, mitotic cycle, cell growth and oncogenic transformation n. CD45 regulates receptor signaling by direct interaction with components of the receptor complexes or by activating and dephosphorylating various Src family kinases (SFK) i.e. Lck 12. But it can inhibit cytokine receptor signaling by inhibiting JAK kinases 13 or by dephosphorylating the activating residues of Src 12. Usually, CD45 levels increase with cell maturation 14. The CD45 family comprises several members derived of a single complex gene 14. Naive T lymphocytes are usually positive for the long CD45RA isoform. Activated and memory T cells express CD45RO, the shortest CD45 isoform by activation-induced alternative splicing of CD45 pre- mRNA 14~17. Most studies in CD45 function have been developed in T cells. Much less is known about its function on NK cells, although it is commonly accepted that CD45 positively regulates the activation of these cells through its ability to dephosphorylate the inhibitory site of SFKs. This is particularly true for the activation that leads to the production of cytokines and chemokines, whereas cytotoxicity is only slightly impaired in NK cells derived from CD45- deficient mice 18~20. However, in contrast to T and B cells, CD45 -deficient NK cells show increased basal phosphorylation of multiple phosphoproteins suggesting that CD45 may also dephosphorylate other substrates in NK cells, including the activating tyrosine residue of SFKs 20. Hence, the role of CD45 in NK cells is an open issue, although it could depend on the type and strength of the activation. However, in vivo studies show that CD45 -deficient NK cells do not protect mice from cytomegalovirus infection due to impaired function of all immunoreceptor tyrosine-based activation motif (ITAM)-dependent NK cell functions, including degranulation 21. However, most of the results cited above were obtained in mouse and could not reflect the human sketch.
SUMMARY OF THE INVENTION:
The present invention relates to methods and kits for determining whether a NK cell is activated and is able to proliferate.
DETAILED DESCRIPTION OF THE INVENTION:
The use of alloreactive NK cells seems a promising co-treatment or an alternative to allogenic HSCT, which has greatly increased the clinical interest for these lymphocytes. However, it is important to identify the different NK cell subpopulations, in particular those with superior antitumor activity. Here, we use the ubiquitous lymphocyte marker CD45 to identify different ex vivo and in vitro expanded NK cell populations from PBL and UCBL. NK cells from healthy donors express high CD45 levels that correlate with the expression of maturation markers. Ex vivo, healthy donor NK cells are exclusively CD45RA and in vitro IL- 2-mediated activation induces CD45RO expression with down regulation of CD45RA. This effect is enhanced when NK cells encounter their targets. After in vitro activation, NK cells increase in size and granularity, mainly in a newly identified CD45RA+RO+ population, which shows the higher CD 16 expression and degranulating activity. Some blood borne cancer patients and all tested bone marrow transplanted patients show CD45RO RA NK cells and a new population of CD45RAdimRO+ NK cells. Some cancer patients with normal CD45RA expression in blood show CD45RAdimROdim/+ and/or CD45RO+RA~ NK cells. However, long term survivors of hematological malignancies that show abnormally elevated numbers of NK cells show almost exclusively CD45RA NK cells. Taking these data together suggest that CD45RO is mainly a marker of activation/proliferation and not a marker of memory NK cells; or that memory NK cells do not exist in vivo.
Accordingly an aspect of the invention relates to a method for determining whether a NK cell is activated and is able to proliferate comprising the steps consisting of i) determining the expression level of CD45RO at the cell surface of the NK cell, ii) comparing the expression level determined in step i) with a predetermined reference value and iii) concluding that the NK cell is activated and is able to proliferate when the expression level determined at step i) is higher than the predetermined reference value. As used herein, the term "NK cell" has its general meaning in the art and refers to natural killer (NK) cell. One skilled in the art can easily identify NK cells by determining for instance the expression of specific phenotypic marker (e.g. CD56) and the ability to express different kind of cytokines or the ability to induce cytotoxicity. Typically a population of NK cells is generally prepared from a blood sample. The term "blood sample" means a whole blood sample obtained from a subject (e.g. an individual for which it is interesting whether a population of NK cells is activated and is able to proliferate). Preferably, the NK cell population is prepared from a PBMC sample. The term "PBMC" or "peripheral blood mononuclear cells" or "unfractionated PBMC", as used herein, refers to whole PBMC, i.e. to a population of white blood cells having a round nucleus, which has not been enriched for a given sub-population. Typically, the PBMC sample may have been subjected to a selection step to contain non-adherent PBMC (which contain T cells, B cells, natural killer (NK) cells, NK T cells and DC precursors). A PBMC sample according to the invention therefore contains lymphocytes (B cells, T cells, NK cells, NKT cells). Typically, these cells can be extracted from whole blood using Ficoll, a hydrophilic polysaccharide that separates layers of blood, with the PBMC forming a cell ring under a layer of plasma. Additionally, PBMC can be extracted from whole blood using a hypotonic lysis buffer, which will preferentially lyse red blood cells. Such procedures are known to the expert in the art. The NK cells can be prepared by Percoll density gradients, by negative depletion methods or by FACS sorting methods. These cells can also be isolated by column immunoadsorption using an avidine-biotin system or by immunoselection using microbeads grafted with antibodies. It is also possible to use combinations of these different techniques, optionally combined with plastic adherence methods. For example, the NK cells can be prepared by providing blood mononuclear cells depleted of T cells from the donor, activating said cells with phytohemagglutinin (PHA) and culturing said cells with interleukin (IL)-2 and irradiated feeder cells.
In a particular embodiment, the population of NK cells is prepared from a subject suffers from a disease, for example from a cancer or an infectious disease.
In a particular embodiment, the population of NK cells is prepared from a transplanted subject. Example of grafts include, but are not limited to transplanted heart, kidney, lung, liver, pancreas, pancreatic islets, brain tissue, stomach, large intestine, small intestine, cornea, skin, trachea, bone, bone marrow, muscle, or bladder.
As used herein the term "CD45" has its general meaning in the art and refers to the protein tyrosine phosphatase (PTP) encoded by the PTPRC gene, which is specifically expressed in hematopoietic cells 10. CD45 regulates receptor signalling by direct interaction with components of the receptor complexes or by activating and dephosphorylating various Src family kinases (SFK) i.e. Lck 12. But it can inhibit cytokine receptor signalling by inhibiting JAK kinases or by dephosphorylating the activating residues of Src 12. Typically it is possible to distinguish two iso forms of CD45: CD45RA and CD45RO. Standard methods for detecting the expression of specific surface markers at cell surface (e.g. NK cell surface) are well known in the art. Typically, the step consisting of determining the expression levels of CD45RO at the NK cell surface may consist in collecting a NK cell population from a subject and using at least one differential binding partner directed against CD45RO, wherein said NK cells are bound by said binding partners to said CD45RO.
As used herein, the term "binding partner directed against the CD45RO" refers to any molecule (natural or not) that is able to bind the CD45RO with high affinity. Said binding partners include but are not limited to antibodies, aptamer, and peptides. The binding partners may be antibodies that may be polyclonal or monoclonal, preferably monoclonal, specifically directed against said CD45RO. In another embodiment, the binding partners may be a set of ap tamers.
Polyclonal antibodies of the invention or a fragment thereof can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others. Various adjuvants known in the art can be used to enhance antibody production. Although antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
Monoclonal antibodies of the invention or a fragment thereof can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique originally; the human B-cell hybridoma technique; and the EBV-hybridoma technique.
In another embodiment, the binding partners may be aptamers. Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity. Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library. The random sequence library is obtainable by combinatorial chemical synthesis of DNA or RNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence. Peptide aptamers consist of conformationally constrained antibody variable regions displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods.
The binding partners of the invention such as antibodies or aptamers may be labelled with a detectable molecule or substance, such as preferentially a fluorescent molecule, or a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal.
As used herein, the term "labelled", with regard to the antibody or aptamer, is intended to encompass direct labelling of the antibody or aptamer by coupling (i.e., physically linking) a detectable substance, such as a fluorophore [e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)]) or a radioactive agent to the antibody or aptamer, as well as indirect labelling of the probe or antibody by reactivity with a detectable substance. An antibody or aptamer of the invention may be labelled with a radioactive molecule by any method known in the art. For example radioactive molecules include but are not limited radioactive atom for scintigraphic studies such as 1123, 1124, Inl 11, Rel86, Rel88.
Preferably, the antibodies against the CD45RO are already conjugated to a fluorophore (e.g. FITC-conjugated and/or PE-conjugated).
The aforementioned assays may involve the binding of the binding partners (ie. antibodies or aptamers) to a solid support. The solid surface could a microtitration plate coated with the binding partner. After incubation of the NK cell sample, NK cells specifically bound to the binding partner may be detected with an antibody to a common NK cell marker Alternatively, the solid surfaces may be beads, such as activated beads, magnetically responsive beads. Beads may be made of different materials, including but not limited to glass, plastic, polystyrene, and acrylic. In addition, the beads are preferably fluorescently labelled. In a preferred embodiment, fluorescent beads are those contained in TruCount(TM) tubes, available from Becton Dickinson Biosciences, (San Jose, California).
According to the invention, methods of flow cytometry are preferred methods for measuring the level of CD45RO at the NK cell surface. Said methods are well known in the art. For example, fluorescence activated cell sorting (FACS) may be therefore used. Typically, a FACS method such as described in Example here below may be used to measuring the level of CD45RO at the NK cell surface.
In a particular embodiment, the method further comprises the steps consisting of i) determining the expression level of CD45RA at the cell surface of the NK cell, ii) comparing the expression level determined in step i) with a predetermined reference value and iii) concluding that NK cell is activated and is able to proliferate when the expression level determined at step i) is lower than the predetermined reference value. The techniques described for determining the expression of CD45RO may be applied mutatis mutandis for determining the expression of CD45RA in the population of NK cells.
The present invention also relates to a method for isolating a population of NK cells capable of activation and proliferation comprising the steps consisting of i) providing a population of NK cells and ii) selecting the population of CD45RO+ NK cells.
The present invention also relates to a method for isolating a population of NK cells capable of activation, proliferation and high level of cytotoxicity comprising the steps consisting of i) providing a population of NK cells and ii) selecting the population of CD45RA+RO+ NK cells.
Typically the population of NK cells as above mentioned are isolated from a blood sample obtained from a subject.
The methods are particularly suitable to be implemented in in vitro protocols of expansion of NK cells. For example the protocol of expansion may consist in contacting a population of NK cells with an amount of IL-2. The protocol of expansion may also consist of the protocol described in WO2012146702. Briefly the protocol comprise the steps consisting of (i) contacting a population of NK cells with at least one accessory cell (i.e. a cell wherein the expression of one gene encoding for a Killer-Cell Immunoglobulin-like Receptor(s) (KIR) ligand is inhibited) under conditions and for a duration sufficient to induce activation of the population of NK cells; (ii) recovering said activated population NK cells. Then the method of the invention may be performed for determining whether the protocol leads to a population of NK cells that is activated and that is capable to proliferate, and/or for isolating at the end of the protocol the population of NK cells that that is activated and that is capable to proliferate.
The methods of the invention may also suitable for determining whether an agent is able to induce activation and proliferation of NK cells. For example said agent may represent a pharmaceutical agent for the treatment of a disease (e.g. a cancer or an infectious disease), and the methods of the invention may thus be useful for determining whether said pharmaceutical agent is able to induce the development of population of NK cells that is activated and that is able to proliferate. Typically, said pharmaceutical agent may be selected from the group of recombinant interleukins, recombinant cytokines, recombinant growth factors, and antibodies... Accordingly, an aspect of the invention relates to a method for monitoring the treatment of a subject with a pharmaceutical agent comprising the steps consisting of administering the subject with the pharmaceutical agent, providing a blood sample or PBMC sample from the subject after said treatment, and determining whether said blood or PBMC sample contain a population of CD45RO+ NK cells (or CD45RA+RO+ NK cells).
The methods of the invention are also particularly suitable for the immunopro filing of a subject, typically suffering from an infectious disease or a cancer. For example, if a subject harbours a population of CD45RO+ NK cells, more particularly a population of CD45RO+RA- NK cells or a population of CD45RA+RO+ NK cells, then it can be concluded that the patient harbours an efficient immune response and the subject can have a good prognostic. Thus, a further aspect of the invention relates to a method for determining the survival time of a subject suffering of a cancer or an infectious disease comprising the steps consisting of i) detecting in a blood sample obtained from the subject the presence of a population of CD45RO+ NK cells, more particularly a population of CD45RO+RA- NK cells or a population of CD45RA+RO+ NK cells and ii) providing a good prognosis when the population is detected at step i).
A further aspect of the invention also relates to a method for determining whether a transplanted subject is at risk of graft rejection comprising the steps consisting of i) detecting in a blood sample obtained from the subject the presence of a population of CD45RO+ NK cells, more particularly a population of CD45RO+RA- NK cells or a population of CD45RA+RO+ NK cells and ii) concluding that the transplanted subject is at risk of graft rejection when the population is detected at step i).
The method may be used to evaluate survival of a variety of different types of grafts. Grafts of interest include, but are not limited to: transplanted heart, kidney, lung, liver, pancreas, pancreatic islets, brain tissue, stomach, large intestine, small intestine, cornea, skin, trachea, bone, bone marrow, muscle, or bladder. The population of NK cells isolated by the method of the invention may be suitable for testing whether a therapeutic antibody is capable to induce an antibody dependent cellular cytotoxicity (ADCC). "Antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to a form of cytotoxicity in which secreted antibodies bound onto Fc receptors (FcRs) present on NK cells enable these cytotoxic effector cells to bind specifically to an antigen-bearing target cell and subsequently kill the target cell. Typically the antibody is incubated with a population of cells expressing the antigen (i.e. the population of "target cells") that is recognized by the antibody in presence of an isolated population of NK cells according to the invention. Then the cytoxicity on the population of target cells may be then evaluated.
The population of NK cells isolated by the method of the invention may be then used in therapeutic protocols such as methods for treating cancers. Typically the population that is more interesting, although not exclusively, for a therapeutic purpose is the population of CD45RA+RO+ NK cells.
For example, the populations of NK cells isolated according to the method of the invention are particularly suitable to enhance the efficacy and safety of allogeneic grafts. By a way of example, a method of transplanting allogeneic graft into a patient in need thereof may comprise the steps consisting of a) administering to said patient an effective amount of the activated NK cells as described above; and, b) transplanting the allogeneic graft into the recipient. This method of transplanting allogeneic graft, more particularly hematopoietic graft, can be applied for reducing the GVHD, for decreasing the intensity of the conditioning regimen, for treating a subject having hematologic disorder, more particularly leukemia, for treating or preventing an infection in a recipient of allogeneic graft, for enhancing immune reconstitution in an allogeneic graft recipient, for proceeding a hematopoietic graft with a greater T cell content, for increasing the engraftment, for reducing the graft rejection, for avoiding the tumor relapse and/or for conditioning a patient in need of a hematopoietic graft. Hematologic disorder includes neoplastic proliferation of hematopoietic cells. Optionally, said hematologic disorder is selected from the group consisting of lymphoblastic leukemia, acute or chronic myelogenous leukemia, Hodgkin's lymphoma, Non-Hodgkin's lymphoma, myelodysplasia syndrome, multiple myeloma, and chronic lymphocytic leukemia. Preferably said hematologic disorder is a leukemia, more preferably myeloid leukemia. Hematologic disorder also includes non-malignant disorders such as an inherited erythrocyte abnormalities, an inherited immune system disorders or a hemoglobinopathy, e.g. sickle cell anemia, aplastic anemia or thalassemia. In a particular embodiment the isolated population of NK cells of the invention (e.g. CD45RA+RO+ NK cells) expressing only one type of KIR (Killer-Cell Immunoglobulin- like Receptor). For example, when shall be used in the treatment of hematological cancers, the physicians may perform HLA genotyping of patients. They could then order the isolated population of NK cells of the invention missing expression of the required KIRs (e.g. an AML patient has tumor cells that do not express ligands for KIR2DL1). NK cells expressing only one KIR (e.g. KIR2DL1) may then be provided and used in the therapeutic protocol.
Preferably, said allogeneic graft is a hematopoietic graft. Optionally, said hematopoietic graft is a bone marrow transplant. In a particularly interesting embodiment of the methods according to the present invention, said patient is treated for leukemia, more preferably myeloid leukemia, optionally an acute or chronic myeloid leukemia.
In one embodiment of the methods according to the present invention, the isolated population of NK cells according to the invention and allogeneic graft are administered simultaneously. In an alternative embodiment, the isolated population of NK cells of the invention are administered prior to the allogeneic graft.
The efficient amount of the isolated population of NK cells of the invention administered to the recipient can be between about 0.05 106 and about 100 106 cells/kg of recipient's body weight. The efficient amount of hematopoietic cells administered to the recipient can be between about 0.2 106 and about 10 106 CD34+ cells/kg of recipient's body weight, In a preferred embodiment, the graft comprises a maximum of 1 105 CD3+ cells/kg of recipient's body weight.
The isolated population of NK cells of the invention and hematopoietic cells are typically administered to the recipient in a pharmaceutically acceptable carrier by intravenous infusion. Carriers for these cells can include but are not limited to solutions of phosphate buffered saline (PBS) containing a mixture of salts in physiologic concentrations.
The hematopoietic cells can be provided by bone marrow cells, mobilized peripheral blood cells or cord blood cells. The bone marrow cells can be obtained from the donor by standard done bone marrow aspiration techniques know in the art, for example by aspiration of marrow from the iliac crest. Peripheral blood stem cells are obtained after stimulation of the donor with a single or several doses of a suitable cytokine, such as granulocyte colony- stimulating factor (G-CSF), granulocyte/macrophage colony- stimulating factor (GM-CSF) and interleukin-3 (IL-3). In a preferred embodiment of the invention, the donor is stimulated with G-CSF. In order to harvest desirable amounts of stem cells from the peripheral blood cells, leukapheresis is performed by conventional techniques and the final product is tested for mononuclear cells. Optionally, the hematopoietic cells can be T-cell depleted. T-cell depletion of bone marrow or of peripheral blood cell may be carried out by any known technique, for example, by soybean agglutination and E-rosetting with sheep red blood cells as described.
According to the invention the host patient is conditioned prior to the transplantation of the allogeneic graft. Conditioning may be carried out under sublethal, lethal or supralethal conditions, for example by total body irradiation (TBI) and/or by treatment with myelo- reductive or myelo-ablative and immunosuppressive agents. According to standard protocols, a lethal dose of irradiation is within the range of 7-9,5 Gy TBI, a sublethal dose is within the range of 3-7 Gy TBI and a supralethal dose is within the range of 9,5-16 Gy TBI.
Any immunosuppressive agent used in transplantation to control the rejection, or a combination of such agents, can be used according to the invention, such as prednisone, methyl prednisolone, azathioprine, cyclophosphamide, cyclosporine, monoclonal antibodies against T-cells, e.g. OKT3, and antisera to human lymphocytes (antilymphocyte globulin - ALS) or to thymus cells (antithymocyte globulin - ATG). Examples of myelo-ablative agents that can be used according to the invention are busulphan, dimethyl myleran and thiotepa.
The advantage of the administration of the isolated population of NK cells of the invention is the possibility to reduce the intensity of the conditioning regimen. For example, the conditioning regimen can be reduced to the intensity of conditioning regimen adopted for matched human transplants. A reduced version of a high- intensity regimen according to the present invention includes Fludarabine at the total dose of 200 mg/M2, Thiotepa 5 mg/Kg, and Melphalan 70 mg/M2, plus anti-T cell antibodies such as ATG, 20 mg/Kg. Optionally, the doses of Thiotepa and Melphalan can be increased by 50%. Indeed, such conditioning regimen is highly toxic and some patients are unable to withstand such toxicity. Therefore, the present invention makes possible the allogeneic graft for these patients.
The isolated population of NK cells of the invention as prepared according to the invention may be used as first line of treatment in combination with standard total body irradiation (TBI) and/or by treatment with myelo -reductive or myelo-ablative and immunosuppressive agents; or used to control the residual disease after total body irradiation (TBI) and/or by treatment with myelo -reductive or myelo-ablative and immunosuppressive agents.
The isolated population of NK cells of the invention as prepared according to the invention may also be used for the treatment of other NK-sensitive malignancies including pediatric cancers such as the sarcomas Ewing sarcoma and rhabdomyosarcoma (Cho...Campana clinical cancer Research 16:3901 (2010), neuroblastoma, malignant glioma and, possibly, prostate cancer (Cho, D. & Campana, D. Expansion and activation of natural killer cells for cancer immunotherapy. Korean J Lab Med 29, 89-96 (2009).
The isolated population of NK cells of the invention as prepared according to the invention may also be used for the treatment of aggressive cancer with no major drugs in order to improve the response rate and PFS. Typical said cancers include but are not limited to pancreatic cancer, lung cancer, breast and colon cancer. In particular, the isolated population of NK cells according to the invention is used in combination with a therapeutic antibody, typically a therapeutic antibody capable of ADCC (e.g. a CD20 antibody).
The isolated population of NK cells of the invention as prepared according to the invention may also useful for the treatment of infectious diseases or dysimmune diseases.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
EXAMPLE: Material & Methods PBMC and UCBMC purification
PBMC and UCB mononuclear cells (UCBMC) were respectively collected from peripheral blood samples and UCB units using Histopaque (Invitrogen). Briefly, thirteen ml Histopaque were put in 50 ml centrifugation tubes and 20-30 ml of 1/2 diluted blood in RPMI, (Invitrogen) were slowly added at the top. Tubes were centrifugated 30 minutes at 1600 rpm and 20 °C without break. Mononuclear cells were collected from the interlayer white ring. After washing in RPMI, cells were suspended in complete RPMI medium supplemented with 10% FBS (Invitrogen).
FACS analysis
For phenotype analysis, cells were stained with 7AAD (Beckman) to identify viable cells and antibodies against surface markers, CD25-FITC, CD45RO-FITC, CD69-PE, CD62L- PE, CD19-PE, CD3-PE, CD19-ECD, CD56-PECy7, CD56-APC, CD3-APC, CD45- APCAlexaFluor750, CD45RA-APCAlexaFluor750, CD16-PacificBlue, CD57-PacificBlue, CD45-KromeOrange, CD16-KromeOrange (Beckman), CD158b-FITC, CD158a-PE, CD107a- HV500 (BD Biosciences), CD158e-Vioblue (Miltenyi), by incubating Ixl05-3xl05 cells with different antibodies in PBS 2,5% FBS for 20-30 minutes at 37 °C. Cells were then washed and suspended in 200-250 μΐ PBS 2,5% FBS. Staining was analyzed on a flow cytometer Gallios (Beckman) using the Kaluza software.
Alive lymphocytes were gated using FS/SS and 7AAD staining. B lymphocytes
(CD 19+), T lymphocytes (CD3+CD56-) and NK cells (CD56+CD3-) were distinguished using respectively CD 19, CD3 and CD56 antibodies. For special NK analysis depending CD45RA/RO phenotypes, NK cells were separated in 3 distinct populations CD45RA+RO-, CD45RA-RO+ and CD45RA+RO+. These different populations were then analyzed for CD 16, CD69, CD62L, CD57, IFNy and CD 107a expression or size/granularity (FSC/SSC) parameters.
Stimulation protocol
PBMC and UCBMC were stimulated for NK amplification and activation for 10 to 20 days. Two different protocols were compared, starting with PBMC/UCBMC 1.106 cells/ml. Firstly, stimulation with high dose of IL-2 (1000 U/ml, eBiosciences) added to complete medium. Secondly, co-stimulation with the NK cell target EBV cell line C30, developed in our laboratory, together with IL-2 100 U/ml + IL-15 (5 ng/ml, Miltenyi).
Cytotoxicity assay
Ex vivo or stimulated UCBMC cytotoxic capacities were tested against the target cell line K562 by using a degranulation assay based on CD 107a labelling. Briefly, 50 thousand target cells per well were put in RPMI 10%FBS IL-2 lOOU/ml with monensin (BD Biosciences) in a 96-V bottom-plate. Effector cells (UCBMC) were added at different effectontarget ratios.
Cells were incubated over night at 37°C 5%C02. After incubation, living cells in the wells were counted using a cytometer Muse (Millipore) with the count and viability kit (Millipore). Cells were analyzed by FACS using 7AAD, CD45RO-FITC, CD19-PE, CD56- PECy7, CD3-APC, CD45RA-APCAlexaFluor750, and CD16-PacificBlue for cytotoxicity assay; or 7AAD, CD45RO-FITC, CD19-PE, CD56-PECy7, CD3-APC, CD45RA- APCAlexaFluor750, CD16-KromeOrange and CD107a-HV500 (BD Biosciences) for degranulation assay. FACS results were analyzed using the Kaluza software. For cytotoxicity assay, target cells were gated in living cells (7AAD-) using FS/CD45 plots and FS/CD3 (Jurkat) or FS/CD19 plots. The numbers of living target cells after incubation with effector cells were subtracted to the number of living target cells when incubated alone to give the percentage of target cell lysis. For degranulation assay, the percentages of CD107a+ NK cells were analyzed. Statistics
All the experiments were performed at least three times with similar results.
Results NK cell expansion is required for clinical means. In general, the source of these cells can be PBL or UCBL. It is expected that lymphocytes for the second source would be na'ive and/or less committed into differentiation. We compared the expression of CD45, a marker of maturation 14, in the three main types of human lymphocytes: T, B and NK cells derived from PBL and UCBL. We show that B and NK cells express similar CD45 levels in PBL and UCBL. NK cell population can be divided in two regarding CD56 expression. CD56bnght are relatively immature, with low cytotoxic activity and high cytokine production and CD56dim, which are more mature with high cytolytic activity and lower cytokine production 3. CD56bnght cells showed lower CD45 expression in NK cells, according to the observation that CD45 levels increased with cell maturation 14. However, the median percentage of CD56bnght was similar in UCBL and PBL22. T cells derived from UCBL showed lower CD45 expression than other lymphocytes and than T cells derived from PBL. The lower CD45 expression was probably linked to the stage of maturity because we showed that T cells derived from UCBL expressed lower levels of CD45RO and higher levels of CD45RA , a sign of immaturity 14~17. In contrast, B and NK cells were basically equivalent in PBL and UCBL regarding expression of the CD45 isoforms.
Next, we analyzed if total CD45 was associated to the expression of NK cell activation or maturation markers. Killer-Inhibitory Receptors (KIRs) and CD 16 expression are markers of maturation 23'24, and during in vitro expansion of NK cells, proliferation is associated to CD25 expression whereas cytolytic activity is associated to CD69 expression 25. CD45 showed a positive correlation with CD 16 and CD69, although in the last the differences were very small. There were a negative correlation between CD45 and CD25 and CD56 expression. Regarding expression of the KIRs, we observed that there was a positive relationship of CD45 with CD158a and CD158b. These results showed that total CD45 expression mainly correlated with maturation markers such as high CD 16, CD69 and KIRs and low CD56 and CD25 in NK cells derived from both UCBL and PBL.
Finally, independently of their origin and CD56 level, naive NK cells expressed almost exclusively the isoform CD45RA, although the CD56bnght NK cells at lower level than other NK cells.
After 10 days of in vitro activation with a high dose of IL-2, NK cells of both origins increased CD45RO expression and decreased CD45RA expression. UCBL T cells also showed the same changes although less pronounced than PBL T cells. In UCBL T cells, these changes were mainly observed at day 10, whereas 3 days after stimulation the changes were minimum and a 20-day stimulation did not increase further the CD45 changes. In contrast, these changes started 3 days after activation in UCBL NK cells. In agreement with previous descriptions 15, 20 days after stimulation some NK cells recovered CD45RA expression without loosing CD45RO expression. Hence, a new population CD45RA RO+ (from now on called CD45RARO) appeared that was mainly absent in T cells.
To study more in detail the expression of CD45 isoforms during in vitro activation, we compared IL-2 and co-stimulation conditions. The latter reflected incubation with a prototypical NK cell target, in this case an EBV cell line, together with low concentrations of two NK cell activating cytokines: IL-2 and IL-15 26. After 10 days of co-stimulation NK cells expressed higher CD45RO and lower CD45RA levels compared to IL-2 stimulation. In addition, CD45 isoforms changes appeared faster during co-stimulation although the CD45RARO population was similar after 20-day stimulation.
This co-stimulation was not optimized for T cell activation; however we still observed an increase of CD45RO compared to cells treated with a high dose of IL-2. These results showed that UCBL T cells responded to this co-stimulation, which induced a faster change from CD45RA to CD45RO isoform. However, the CD45RARO population was mainly absent at day 20.
CD69 is used as a general marker of NK cell activation including in vivo 5. We observed that in vitro activation with IL-2 induced a larger increase in CD69 than co- stimulation. In contrast co-stimulation induced a larger decrease in CD45RA expression that IL-2. These data suggested that both protocols, CD69 up regulation and CD45RA down regulation, were compatible to study NK cell activation and could be used depending of the NK cell activation stimulus. In our opinion, the CD45RA/RO detection method has the advantage of couple two phenomena: CD45RA down regulation and CD45RO up regulation. This should decrease the number of false positives and the artifacts. A more detailed time course of in vitro NK cell stimulation showed that IL-2 treatment induced a large declined of CD45RA RO" (from herein called CD45RA) population with increase of CD45RA~RO+ (from now on called CD45RO) population. In contrast, CD45RARO showed a biphasic curve with a first peak around day 3 and a second increase around day 12. Co-stimulation induces a faster and more complete decline of CD45 cells and increase in CD45RO cells. The biphasic curve of CD45RARO was more apparent. Whereas we cannot prove it, we believe that the first peak is related to acquisition of CD45RO expression without totally loosing CD45RA expression. The CD56bngth probably respond faster to stimulus i.e. cytokines could be responsible of this peak. However, we could not study this fact, because NK cell activation increased CD56 levels, and therefore we could not distinguish anymore the two populations CD56bnght and CD56dim. The second peak is probably due to expression of CD45RA in the CD45RO population generated after in vitro activation.
Resting CD45RA NK cells were mainly small cells with low granularity. After activation, NK cells increased in size and granularity. This was particularly true for CD45RO and, mainly, CD45RARO populations. These results were in agreement with the notion that CD45RO expression was associated to stimulation and that re-acquisition of CD45RA could be an additional sign of activation.
During in vivo maturation CD56bright cells originates transitory CD56dimCD62L+CD57~ cells that started generating higher levels of perforin while keeping high IFN-γ production in response to cytokines 3'27. In the next step, CD56dimCD62L~CD57+ cells show low response to cytokines but increased cytotoxic capacity 3'28. UCBL NK cells expressed low CD62L levels and almost no expression of CD57 compared to PBL NK cells in agreement with previous reports 2 '. In vitro activated UCBL NK cells increased CD62L expression in all CD45 subpopulations; however, the expression decreased with time and even disappeared by day 20 in cells activated by co-stimulation. Regarding CD57, in vitro NK cell activation induced a significantly increase only in the CD45RARO population. PBL NK cells expressed higher levels of both CD62L and CD57. After activation, CD62L barely increased whereas CD57 decreased. In summary, in vitro activation of UCBL NK cells did not induce an in vivo model of NK cell differentiation; probably the CD56dim cells are already fully differentiated even in UCBL.
Hence, we investigated which population expressed the maturation marker CD 16 after in vitro UCBL activation. As an additional marker of activation, we studied the degranulating capacity of NK cells on K562 targets by analyzing CD 107a expression, which is commonly used as a marker of degranulation. During in vitro expansion, the pool of CD45RA cells partially lost CD 16 expression. This was less evident in CD45RO cells, whereas CD45RARO cells showed the higher expression. Regarding degranulation, CD45RARO cells displayed the higher levels, with CD45RA and CD45RO showing similar levels. In summary we identified in in vitro expanded NK cells the population with the higher degranulation activity and CD 16 expression.
However, our in vitro studies could not reflect the real populations in vivo because in PBL and UCBL from healthy donors, we found almost exclusively CD45RA expression. We hypothesized that the other two populations CD45RO and CD45RARO could be present in a framework of NK cell expansion and/or activation. As a model of NK cell activation we chose patients of hematological cancers where NK cells could be in contact with their target tumor cells 29. As a model of NK cell proliferation we chose patients who have received a bone marrow transplant and show a rapid reconstitution of their NK cell pool 30.
Transplanted patients showed both CD45RA down regulation and CD45RO up regulation, generating a comet-like figure, similar to our observation during in vitro NK cells activation, although less pronounced. We identified a new population in vivo, the CD45RAdim cells, which were the first step for the lost of CD45RA before acquisition of CD45RO, and represented the start of the comet tail. We considered these cells as "activated" cells and were not included as part of the resting CD45RA population. We show that the number of CD45RA resting NK cells clearly diminished in these patients. These patients expressed higher percentages of CD56bnght cells, which were CD 16" suggesting that these were de novo produced NK cells and not CD56dim cells that gain CD56 expression. We next analyzed the maturation state of these cells. Independently of CD 16 expression, CD56bnght cells were CD62L+CD57~ and showed a more immature phenotype than the CD56dim cells, which were CD62L~CD57+.
In transplanted patients and independently of CD 16 expression, CD56bnght cells were mainly CD45RO cells, whereas CD56dim cells were mainly CD45RA. These data showed that the more differentiated cells displayed a lower level of CD45RO than the immature cells. A possible explanation is that in bone marrow transplanted patients there is a boost of cytokines that preferably activate CD56bnght cells, which are more immature 3 and that are shown in bigger numbers in transplanted patients than in healthy donors. Activation and/or proliferation of these cells would provide them with the CD45RO marker. The CD45RARO population was mainly absent in most of these individuals, although 2 out of 6 patients showed around 5% of their NK cells with this phenotype. In summary, the CD45RO population was mainly CD56brightCD62L+CD57", in contrast to the CD45RA population that shows a CD56dimCD62L" CD57+ phenotype. In this context, it should be noted that CD56dim cells are usually CD57+ whereas CD56bnght cells usually express CD62L 27'28. The results strongly supported that the CD56bright cells that appeared in these patients were initially CD56bright cells and not CD56dim cells that could increase CD56 expression. The increase in CD56bnght cells compared to healthy donors was probably due to recovery after lymphopenia, which induced a large production of na'ive cells.
In the second model we investigated NK cell subpopulations in patients with hematological cancers including B-CLL and MM. Five out of 12 of these patients showed down regulation of the CD45RA population and CD45dim and/or CD45RO up regulation.
We were surprised for the lack of activated NK cells in 7 of 12 cancer patients. Recently, it has been shown that tumor infiltrating NK cells show a specific phenotype that distinguish them from circulating NK cells 29'31. We hypothesized that the activated, CD45RAdim and/or CD45RO, NK cells could be diluted in the blood of hematological cancer patients preventing their detection. Hence, we investigated NK cells that were in contact with tumor cells in patients with low numbers of activated NK cells in blood. We focused in patients with bone marrow disorders where it was easy to identify the NK cell population in contact with tumor cells i.e. multiple myeloma (MM). We observed that several patients that did not show activated NK cells in blood showed activated cells in the tumor site, in our case in the bone marrow. The pC9 expressed a low reduction of CD45RA cells. This patient showed a gammopathy and NK cells in the bone marrow showed a higher activation with reduction of CD45RA levels and increase in CD45RO. We next analyzed pC2, suffering from multiple myeloma (MM), and with CD45RA levels similar to healthy donors. NK cells in the bone marrow show a decline in the number of CD45RA cells and increased in CD45RAdim and CD45RO cells. In contrast, two patients suffering from a B-CLL did not show a decrease in CD45RA number in his bone marrow, suggesting that CD45RO expression depended on the contact with tumor cells, at least in certain patients. We next analyzed expression of CD45 isoforms in the tumor ganglion of 2 B-CLL patients. We found a large decrease in CD45RA expression in the tumor site. In summary, expression of different CD45 isoforms in vivo can identify NK cell activation and/or proliferation.
We have identified a new population of NK cells identified by co-expression of CD45RO and CD45RA molecules. This CD45RARO population was a minority and we wondered if in certain individuals this population could be enlarged after activation. We analyzed 14 UCB and found one, UCB439, which after activation with IL-2 or co-stimulation, showed the atypical phenotype of keeping CD45RA after CD45RO expression, generating a large CD45RARO population. This was more remarkable after co-stimulation. We identified a new CD45RAhlgh/RO+ population, which in the other UCBs, corresponded to common CD45RARO population in FSC/SSC behavior. Gain of CD45RO expression is similar in time and intensity to other UCBs. However, NK cell amplification was significantly larger in this UCB compared to other UCBs. These results suggested that individuals keeping CD45RA in their CD45RO NK cell compartment would develop a larger number of NK cells after sensing "danger" and could show improved anti-tumor activity.
We next try identifying other individuals with distinct CD45 expression after activation. We focused on patients with long-term survival after an hematological cancer and with elevated NK cell levels. We hypothesized that in these patients there was a direct relationship between their survival and high NK cell levels, which could be due to a specific NK cell phenotype. We found 3 patients with these characteristics (pCSNK), which express very low levels of CD56brigth and the CD56dim cells were predominantly CD57+/CD16+. Although CD57 expressing cells increased with age, they rarely overpassed 50% even in very old donors 22. We did not observe this high numbers in cancer patients of similar age, suggesting that high CD57+ population could be related to cancer survival. However, ex vivo, we did not find differences in CD45RA expression.
One of these patients suffered of cancer and has survived more than 20 years and is considered now cured. This individual showed high NK cell numbers during a long period of years and when we finally analyzed him, he showed 15.2 % of NK cell lymphocytes, more than double than a healthy donor. His NK cells showed a resting phenotype similar to healthy donors regarding CD25, CD 16 and CD69 expression. Conversely, these cells showed the classical phenotype of healthy donors: absence of CD45RO expression. However, they expressed higher levels of CD57 than healthy donors. After stimulation, NK cells from this patient showed and increase of CD45RARO cells. However, the amplification and the cytotoxic activity of the amplified NK cells were similar to a healthy donor.
Discussion:
Current clinical protocols fail inducing long-term survival in a significant number of patients with hematological cancers. Those refractory to standard treatment i.e. radio- and/or chemotherapy are often subjected to a myeloablative regimen followed by allogenic hematopoietic stem cell transplantation (HSCT). However, the mortality linked to this treatment is closed to 20%. Moreover, Graft versus Host (GvH) and HvG diseases and opportunistic infections hamper this procedure. Relapse has become the leading cause of death following allogeneic HSCT: the relapse rate has not decrease over the past 20 years 32. In general, prognosis is poor for patients who relapsed to an allograft since effective treatment options are limited. These include donor lymphocyte infusions, withdrawal of immunosuppressive medication and second allogeneic HSCT. However, new specific cellular approaches are under investigation. In particular, the use of alloreactive natural killer (NK) cells after umbilical cord blood transplantation (UCBT) seems promising. Killer cell immunoglobulin-like receptors ( 7i?)-ligand incompatibility in the GvH direction improves outcomes after UCBT in the clinic 33'34. Moreover, NK cells: 1) are not responsible of GvHD; 2) can be injected as "differentiated" cells without the need of long time survival on patient's body; 3) protect from opportunist infections 33, probably through their immunoregulatory effects on B cells, T cells, macrophages, and more importantly on polymorphonuclear cells (PMNs 35). We, and others, have shown the requirement of NK cells to eradicate tumors in several mouse models 26'36. NK cell infiltration gives a good prognostic in several cancers
29 31 37
( ' ' . However, some unresolved problems remain before the standard use of allogeneic NK cells in the clinic. The most important is engraftment of an adequate number of cells and that these cells show clinically efficient anti-tumor activity. For the last purpose, it is basic to identify the different NK cell populations and their cytolytic activity, which is believed to be the most essential component on the anti-tumor NK cell function 3. Here, we identify that the expression of the CD45 isoforms CD45RA and RO distinguishes NK cell populations with diverse cytolytic activity, in particular the CD45RARO population shows improved cytotoxicity. Hence, expansion protocols keeping this population could have a better clinical effect.
CD45 activity is regulated by dimerization and CD45 spontaneously homodimerizes at the plasma membrane, which inhibits its activity 38. The size of the CD45 extracellular domain is inversely proportional to the extent of CD45 dimerization and auto inhibition 38. Larger CD45 isoforms such as CD45RA dimerize less efficiently and, accordingly, and perhaps they are more efficient at promoting TCR signaling than are smaller isoforms such as CD45RO 12. However, CD45 activity also depends on its plasma membrane localization that depends on the extracellular domain n'12. At least in T cells, too high CD45 activity leads to dephosphorylation of the activating residues in the Src kinases, whereas too low would leave phosphorylated the inhibitory residues. Therefore, it is important for efficient activation that CD45 activity is between this window 14, and the amount and the specific CD45 isoform would regulate the final activity. NK cell decision to kill depends on activating and inhibitory signals. We have found that CD45RA+/RO+ NK cells show the maximal cytolytic activity. The presence of both CD45RA and RO could give the NK cells the appropriate level of CD45 activity for efficient signaling to boost the cytolytic activity.
It is important to note that regulation of CD45 activity is critical for an efficient immune response, because its deficiency results in a severe combined immunodeficiency (SCID) phenotype in both mice 39 41 and humans 42'43. This could explain the high expression of this phosphatase and its complex regulation. In agreement to previous results 16 , IL-2 stimulation induces CD45RA down regulation and CD45RO up regulation in PBL and this occurs first in NK cells. We show here that UCBL show a similar behavior. It was proposed that IL-2 was the molecule that drives those changes 16. In contrast, we have found that other signals independent to IL-2 contribute to CD45RO up regulation and CD45RA down regulation because high IL-2 doses do not reach the same level of changes than co-stimulation with target cells.
Ex vivo T cells derived from PBL show more CD45RO expression than T cells derived from UCBL. This suggests that in PBL there are more activated and/or memory cells than in UCBL, what is intuitive. It is fair to assume that in these situations CD45RO cells are memory cells. However, an extremely low number of NK cells express CD45RO ex vivo in both PBL and UCBL. At least in vitro, NK cell activation clearly induces CD45RO expression. Hence, it is surprising that ex vivo we find so low numbers of NK cells expressing CD45RO in PBL. Several non-exhaustive explanations are that the amount of memory NK cells is extremely low, CD45RO is not a marker of memory NK cells or CD45RO is specifically lost during ex vivo manipulations in NK cells, although the last possibility is unlikely because NK cells express slightly higher levels of total CD45 than other lymphocyte. In vivo we found a significant CD45RO population in bone marrow transplanted patients. These cells that are CD57" are probably proliferating cells and are predominantly CD56bnght. Hence, our results suggest that CD45RO is mainly a marker of NK cell proliferation in this specific setting and degranulation activity is mostly related to CD45RARO cells, which are however, a minority population. This is in agreement with results in mice where CD45 is related to ITAM-dependent cytokine production, but is not related to ITAM-dependent cytotoxicity in vitro 18~20, although it is required for full cytotoxic NK cell activity in vivo 21. This suggest that in NK cells the expression of different CD45 isoforms plays a different role that in T cells where it plays a main role in activity. In this context, long-term cancer survival patients with high percentages of NK cells do not show activated, or memory, NK cells, at least regarding to CD45 RA/RO expression. This could reflect the difference between the innate immune system that is based in the number of cells protecting the host; and the adaptive immune system where a small population of memory cells can originate a strong response by clonal expansion of the selected TCR-expressing cells.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
1. Velardi A. Role of KIRs and KIR ligands in hematopoietic transplantation. Curr Opin Immunol. 2008;20(5):581-587.
2. Lanier LL. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol. 2008;9(5):495-502.
3. Bryceson YT, Chiang SC, Darmanin S, et al. Molecular mechanisms of natural killer cell activation. J Innate Immun. 2011;3(3):216-226.
4. Domaica CI, Fuertes MB, Uriarte I, et al. Human natural killer cell maturation defect supports in vivo CD56(bright) to CD56(dim) lineage development. PLoS One. 2012;7(12):e51677.
5. Vey N, Bourhis JH, Boissel N, et al. A phase 1 trial of the anti- inhibitory KIR mAb IPH2101 for AML in complete remission. Blood. 2012; 120(22) :4317-4323.
6. McKenna DH, Jr., Sumstad D, Bostrom N, et al. Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience. Transfusion. 2007;47(3):520-528.
7. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105(8):3051-3057.
8. Ruggeri L, Mancusi A, Burchielli E, Aversa F, Martelli MF, Velardi A. Natural killer cell alloreactivity in allogeneic hematopoietic transplantation. Curr Opin Oncol. 2007;19(2): 142-147.
9. Rubnitz JE, Inaba H, Ribeiro RC, et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol. 2010;28(6):955-959.
10. Kaplan R, Morse B, Huebner K, et al. Cloning of three human tyrosine phosphatases reveals a multigene family of receptor- linked protein-tyrosine-phosphatases expressed in brain. Proc Natl Acad Sci USA. 1990;87(18):7000-7004. 11. Mustelin T, Vang T, Bottini N. Protein tyrosine phosphatases and the immune response. Nat Rev Immunol. 2005;5(l):43-57.
12. Pvhee I, Veillette A. Protein tyrosine phosphatases in lymphocyte activation and autoimmunity. Nat Immunol. 2012;13(5):439-447.
13. Irie-Sasaki J, Sasaki T, Matsumoto W, et al. CD45 is a JAK phosphatase and negatively regulates cytokine receptor signalling. Nature. 2001;409(6818):349-354.
14. Hermiston ML, Zikherman J, Zhu JW. CD45, CD148, and Lyp/Pep: critical phosphatases regulating Src family kinase signaling networks in immune cells. Immunol Rev. 2009;228(1):288-311.
15. Warren HS, Skipsey LJ. Loss of activation- induced CD45RO with maintenance of CD45RA expression during prolonged culture of T cells and NK cells.
Immunology. 1991;74(l):78-85.
16. Roth MD. Interleukin 2 induces the expression of CD45RO and the memory phenotype by CD45RA+ peripheral blood lymphocytes. J Exp Med. 1994;179(3):857-864.
17. Lynch KW, Weiss A. A model system for activation- induced alternative splicing of CD45 pre-mRNA in T cells implicates protein kinase C and Ras. Mol Cell Biol.
2000;20(l):70-80.
18. Huntington ND, Xu Y, Nutt SL, Tarlinton DM. A requirement for CD45 distinguishes Ly49D-mediated cytokine and chemokine production from killing in primary natural killer cells. J Exp Med. 2005;201(9): 1421-1433.
19. Hesslein DG, Takaki R, Hermiston ML, Weiss A, Lanier LL. Dysregulation of signaling pathways in CD45 -deficient NK cells leads to differentially regulated cytotoxicity and cytokine production. Proc Natl Acad Sci USA. 2006;103(18):7012-7017.
20. Mason LH, Willette-Brown J, Taylor LS, McVicar DW. Regulation of Ly49D/DAP12 signal transduction by Src-family kinases and CD45. J Immunol.
2006;176(l l):6615-6623.
21. Hesslein DG, Palacios EH, Sun JC, et al. Differential requirements for CD45 in NK-cell function reveal distinct roles for Syk-family kinases. Blood. 2011;117(11):3087- 3095.
22. Le Garff-Tavernier M, Beziat V, Decocq J, et al. Human NK cells display major phenotypic and functional changes over the life span. Aging Cell. 2010;9(4):527-535.
23. Beziat V, Descours B, Parizot C, Debre P, Vieillard V. NK cell terminal differentiation: correlated stepwise decrease of NKG2A and acquisition of KIRs. PLoS One. 2010;5(8):el l966. 24. Moretta L. Dissecting CD56dim human NK cells. Blood. 2010;116(19):3689-
3691.
25. Clausen J, Vergeiner B, Enk M, Petzer AL, Gastl G, Gunsilius E. Functional significance of the activation-associated receptors CD25 and CD69 on human NK-cells and NK-like T-cells. Immunobiology. 2003;207(2):85-93.
26. Anel A, Aguilo JI, Catalan E, et al. Protein Kinase C-theta (PKC-theta) in Natural Killer Cell Function and Anti-Tumor Immunity. Front Immunol. 2012;3: 187.
27. Juelke K, Killig M, Luetke-Eversloh M, et al. CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood. 2010;116(8): 1299-1307.
28. Lopez-Verges S, Milush JM, Pandey S, et al. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 2010;116(19):3865-3874.
29. Mamessier E, Pradel LC, Thibult ML, et al. Peripheral blood NK cells from breast cancer patients are tumor-induced composite subsets. J Immunol. 2013;190(5):2424- 2436.
30. Giebel S, Dziaczkowska J, Czerw T, et al. Sequential recovery of NK cell receptor repertoire after allogeneic hematopoietic SCT. Bone Marrow Transplant. 2010;45(6): 1022-1030.
31. Mamessier E, Bertucci F, Sabatier R, Birnbaum D, Olive D. "Stealth" tumors: Breast cancer cells shun NK-cells anti-tumor immunity. Oncoimmunology. 2012;1(3):366-
368.
32. Kroger N. Approaches to relapse after allogeneic stem cell transplantation. Curr Opin Oncol. 2011;23(2):203-208.
33. Willemze R, Rodrigues CA, Labopin M, et al. KIR-ligand incompatibility in the graft-versus-host direction improves outcomes after umbilical cord blood transplantation for acute leukemia. Leukemia. 2009;23(3):492-500.
34. Stern M, Ruggeri L, Mancusi A, et al. Survival after T cell-depleted haploidentical stem cell transplantation is improved using the mother as donor. Blood. 2008;112(7):2990-2995.
35. Bhatnagar N, Hong HS, Krishnaswamy JK, et al. Cytokine-activated NK cells inhibit PMN apoptosis and preserve their functional capacity. Blood. 2010; 116(8): 1308-1316.
36. Aguilo JI, Garaude J, Pardo J, Villalba M, Anel A. Protein kinase C-theta is required for NK cell activation and in vivo control of tumor progression. J Immunol. 2009; 182(4): 1972- 1981. 37. Senovilla L, Vacchelli E, Galon J, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology. 2012;1(8): 1323-1343.
38. Xu Z, Weiss A. Negative regulation of CD45 by differential homodimerization of the alternatively spliced iso forms. Nat Immunol. 2002;3(8):764-771.
39. Kishihara K, Penninger J, Wallace VA, et al. Normal B lymphocyte development but impaired T cell maturation in CD45-exon6 protein tyrosine phosphatase- deficient mice. Cell. 1993;74(1): 143-156.
40. Byth KF, Conroy LA, Howlett S, et al. CD45-null transgenic mice reveal a positive regulatory role for CD45 in early thymocyte development, in the selection of CD4+CD8+ thymocytes, and B cell maturation. J Exp Med. 1996;183(4): 1707-1718.
41. Mee PJ, Turner M, Basson MA, Costello PS, Zamoyska R, Tybulewicz VL. Greatly reduced efficiency of both positive and negative selection of thymocytes in CD45 tyrosine phosphatase-deficient mice. Eur J Immunol. 1999;29(9):2923-2933.
42. Kung C, Pingel JT, Heikinheimo M, et al. Mutations in the tyrosine phosphatase CD45 gene in a child with severe combined immunodeficiency disease. Nat Med.
2000;6(3):343-345.
43. Tchilian EZ, Wallace DL, Wells RS, Flower DR, Morgan G, Beverley PC. A deletion in the gene encoding the CD45 antigen in a patient with SCID. J Immunol. 2001;166(2): 1308-1313.

Claims

CLAIMS:
1. A method for isolating a population of NK cells capable of activation, proliferation and high level of cytotoxicity comprising the steps consisting of i) providing a population of NK cells and ii) selecting the population of CD45RA+RO+ NK cells.
2. The method of claim 1 wherein the population of NK cells is isolated from a blood sample obtained from a subject.
3. An isolated population of CD45RA+RO+ NK cells obtainable by the method of claim 1.
4. A method for monitoring the treatment of a subject with a pharmaceutical agent comprising the steps consisting of administering the subject with the pharmaceutical agent, providing a blood sample or PBMC sample from the subject after said treatment, and determining whether said blood or PBMC sample contain a population of CD45RA+RO+ NK cells.
5. The method of claim 4 wherein the pharmaceutical agent is be selected from the group of recombinant interleukins, recombinant cytokines, recombinant growth factors, and antibodies.
6. A method for determining the survival time of a subject suffering of a cancer or an infectious disease comprising the steps consisting of i) detecting in a blood sample obtained from the subject the presence of a population of CD45RA+RO+ NK cells and ii) providing a good prognosis when the population is detected at step i).
7. The isolated population of NK cells isolated by the method of claim 1 for use in a method for treating cancer or an infectious disease.
8. A pharmaceutical composition comprising a population of NK cells isolated by the method of claim 1.
PCT/EP2014/063329 2013-06-28 2014-06-25 Methods and kits for determining whether a nk cell is activated and is able to proliferate WO2014207009A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13305915.4 2013-06-28
EP13305915 2013-06-28

Publications (2)

Publication Number Publication Date
WO2014207009A2 true WO2014207009A2 (en) 2014-12-31
WO2014207009A3 WO2014207009A3 (en) 2015-03-19

Family

ID=48748122

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/063329 WO2014207009A2 (en) 2013-06-28 2014-06-25 Methods and kits for determining whether a nk cell is activated and is able to proliferate

Country Status (1)

Country Link
WO (1) WO2014207009A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020168254A1 (en) * 2019-02-14 2020-08-20 Research Institute At Nationwide Children's Hospital Use of plasma membrane particles, liposomes, and exosomes to assay immune cell potency
CN113588934A (en) * 2021-06-18 2021-11-02 创模生物科技(北京)有限公司 Detection method for immune system reconstruction capability of PBMC (peripheral blood mononuclear cell) and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012146702A1 (en) 2011-04-28 2012-11-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for preparing accessory cells and uses thereof for preparing activated nk cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012146702A1 (en) 2011-04-28 2012-11-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for preparing accessory cells and uses thereof for preparing activated nk cells

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
AGUILO JI; GARAUDE J; PARDO J; VILLALBA M; ANEL A.: "Protein kinase C-theta is required for NK cell activation and in vivo control of tumor progression", J IMMUNOL., vol. 182, no. 4, 2009, pages 1972 - 1981
ANEL A; AGUILO JI; CATALAN E ET AL.: "Protein Kinase C-theta (PKC-theta) in Natural Killer Cell Function and Anti-Tumor Immunity", FRONT IMMUNOL., vol. 3, 2012, pages 187
BEZIAT V; DESCOURS B; PARIZOT C; DEBRE P; VIEILLARD V.: "NK cell terminal differentiation: correlated stepwise decrease of NKG2A and acquisition of KIRs", PLOS ONE, vol. 5, no. 8, 2010, pages EL 1966
BHATNAGAR N; HONG HS; KRISHNASWAMY JK ET AL.: "Cytokine-activated NK cells inhibit PMN apoptosis and preserve their functional capacity", BLOOD, vol. 116, no. 8, 2010, pages 1308 - 1316
BRYCESON YT; CHIANG SC; DARMANIN S ET AL.: "Molecular mechanisms of natural killer cell activation.", J INNATE IMMUN, vol. 3, no. 3, 2011, pages 216 - 226
BYTH KF; CONROY LA; HOWLETT S ET AL.: "CD45-null transgenic mice reveal a positive regulatory role for CD45 in early thymocyte development, in the selection of CD4+CD8+ thymocytes, and B cell maturation", J EXP MED., vol. 183, no. 4, 1996, pages 1707 - 1718
CHO, D.; CAMPANA, D.: "Expansion and activation of natural killer cells for cancer immunotherapy", KOREAN J LAB MED, vol. 29, 2009, pages 89 - 96
CHO...CAMPANA CLINICAL CANCER RESEARCH, vol. 16, 2010, pages 3901
CLAUSEN J; VERGEINER B; ENK M; PETZER AL; GASTL G; GUNSILIUS E.: "Functional significance of the activation-associated receptors CD25 and CD69 on human NK-cells and NK-like T-cells.", IMMUNOBIOLOGY, vol. 207, no. 2, 2003, pages 85 - 93
DOMAICA CI; FUERTES MB; URIARTE I ET AL.: "Human natural killer cell maturation defect supports in vivo CD56(bright) to CD56(dim) lineage development", PLOS ONE, vol. 7, no. 12, 2012, pages E51677
GIEBEL S; DZIACZKOWSKA J; CZERW T ET AL.: "Sequential recovery of NK cell receptor repertoire after allogeneic hematopoietic SCT", BONE MARROW TRANSPLANT., vol. 45, no. 6, 2010, pages 1022 - 1030
HERMISTON ML; ZIKHERMAN J; ZHU JW.: "CD45, CD148, and Lyp/Pep: critical phosphatases regulating Src family kinase signaling networks in immune cells", IMMUNOL REV., vol. 228, no. 1, 2009, pages 288 - 311
HESSLEIN DG; PALACIOS EH; SUN JC ET AL.: "Differential requirements for CD45 in NK-cell function reveal distinct roles for Syk-family kinases", BLOOD, vol. 117, no. 11, 2011, pages 3087 - 3095
HESSLEIN DG; TAKAKI R; HERMISTON ML; WEISS A; LANIER LL.: "Dysregulation of signaling pathways in CD45-deficient NK cells leads to differentially regulated cytotoxicity and cytokine production", PROC NATL ACAD SCI USA., vol. 103, no. 18, 2006, pages 7012 - 7017
HUNTINGTON ND; XU Y; NUTT SL; TARLINTON DM: "A requirement for CD45 distinguishes Ly49D-mediated cytokine and chemokine production from killing in primary natural killer cells", J EXP MED., vol. 201, no. 9, 2005, pages 1421 - 1433
IRIE-SASAKI J; SASAKI T; MATSUMOTO W ET AL.: "CD45 is a JAK phosphatase and negatively regulates cytokine receptor signalling", NATURE, vol. 409, no. 6818, 2001, pages 349 - 354
JUELKE K; KILLIG M; LUETKE-EVERSLOH M ET AL.: "CD62L expression identifies a unique subset ofpolyfunctional CD56dim NK cells", BLOOD, vol. 116, no. 8, 2010, pages 1299 - 1307
KAPLAN R; MORSE B; HUEBNER K ET AL.: "Cloning of three human tyrosine phosphatases reveals a multigene family of receptor-linked protein-tyrosine-phosphatases expressed in brain", PROC NATL ACAD SCI US A., vol. 87, no. 18, 1990, pages 7000 - 7004
KISHIHARA K; PENNINGER J; WALLACE VA ET AL.: "Normal B lymphocyte development but impaired T cell maturation in CD45-exon6 protein tyrosine phosphatase-deficient mice", CELL, vol. 74, no. 1, 1993, pages 143 - 156
KROGER N.: "Approaches to relapse after allogeneic stem cell transplantation", CURR OPIN ONCOL., vol. 23, no. 2, 2011, pages 203 - 208
KUNG C; PINGEL JT; HEIKINHEIMO M ET AL.: "Mutations in the tyrosine phosphatase CD45 gene in a child with severe combined immunodeficiency disease", NAT MED., vol. 6, no. 3, 2000, pages 343 - 345
LANIER LL.: "Up on the tightrope: natural killer cell activation and inhibition", NAT IMMUNOL, vol. 9, no. 5, 2008, pages 495 - 502
LE GARFF-TAVEMIER M; BEZIAT V; DECOCQ J ET AL.: "Human NK cells display major phenotypic and functional changes over the life span", AGING CELL, vol. 9, no. 4, 2010, pages 527 - 535
LOPEZ-VERGES S; MILUSH JM; PANDEY S ET AL.: "CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset.", BLOOD, vol. 116, no. 19, 2010, pages 3865 - 3874
LYNCH KW; WEISS A.: "A model system for activation-induced alternative splicing of CD45 pre-mRNA in T cells implicates protein kinase C and Ras", MOL CELL BIOL., vol. 20, no. 1, 2000, pages 70 - 80
MAMESSIER E; BERTUCCI F; SABATIER R; BIRNBAUM D; OLIVE D: "Stealth'' tumors: Breast cancer cells shun NK-cells anti-tumor immunity", ONCOIMMUNOLOGY, vol. 1, no. 3, 2012, pages 366 - 368
MAMESSIER E; PRADEL LC; THIBULT ML ET AL.: "Peripheral blood NK cells from breast cancer patients are tumor-induced composite subsets", J IMMUNOL., vol. 190, no. 5, 2013, pages 2424 - 2436
MASON LH; WILLETTE-BROWN J; TAYLOR LS; MCVICAR DW.: "Regulation of Ly49D/DAP12 signal transduction by Src-family kinases and CD45.", J IMMUNOL., vol. 176, no. 11, 2006, pages 6615 - 6623
MCKENNA DH, JR.; SUMSTAD D; BOSTROM N ET AL.: "Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience", TRANSFUSION, vol. 47, no. 3, 2007, pages 520 - 528
MEE PJ; TURNER M; BASSON MA; COSTELLO PS; ZAMOYSKA R; TYBULEWICZ VL.: "Greatly reduced efficiency of both positive and negative selection of thymocytes in CD45 tyrosine phosphatase-deficient mice", EUR J IMMUNOL., vol. 29, no. 9, 1999, pages 2923 - 2933
MILLER JS; SOIGNIER Y; PANOSKALTSIS-MORTARI A ET AL.: "Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer", BLOOD, vol. 105, no. 8, 2005, pages 3051 - 3057
MORETTA L.: "Dissecting CD56dim human NK cells", BLOOD, vol. 116, no. 19, 2010, pages 3689 - 3691
MUSTELIN T; VANG T; BOTTINI N.: "Protein tyrosine phosphatases and the immune response.", NAT REV IMMUNOL, vol. 5, no. 1, 2005, pages 43 - 57
RHEE I; VEILLETTE A.: "Protein tyrosine phosphatases in lymphocyte activation and autoimmunity", NAT IMMUNOL, vol. 13, no. 5, 2012, pages 439 - 447
ROTH MD: "Interleukin 2 induces the expression of CD45RO and the memory phenotype by CD45RA+ peripheral blood lymphocytes.", J XP MED., vol. 179, no. 3, 1994, pages 857 - 864
RUBNITZ JE; INABA H; RIBEIRO RC ET AL.: "NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia", J CLIN ONCOL, vol. 28, no. 6, 2010, pages 955 - 959
RUGGERI L; MANCUSI A; BURCHIELLI E; AVERSA F; MARTELLI MF; VELARDI A.: "Natural killer cell alloreactivity in allogeneic hematopoietic transplantation", CURR OPIN ONCOL., vol. 19, no. 2, 2007, pages 142 - 147
SENOVILLA L; VACCHELLI E; GALON J ET AL.: "Trial watch: Prognostic and predictive value of the immune infiltrate in cancer", ONCOIMMUNOLOGY, vol. 1, no. 8, 2012, pages 1323 - 1343
STERN M; RUGGERI L; MANCUSI A ET AL.: "Survival after T cell-depleted haploidentical stem cell transplantation is improved using the mother as donor", BLOOD, vol. 112, no. 7, 2008, pages 2990 - 2995
TCHILIAN EZ; WALLACE DL; WELLS RS; FLOWER DR; MORGAN G; BEVERLEY PC.: "A deletion in the gene encoding the CD45 antigen in a patient with SCID", J IMMUNOL., vol. 166, no. 2, 2001, pages 1308 - 1313
VELARDI A.: "Role of KIRs and KIR ligands in hematopoietic transplantation", CURR OPIN IMMUNOL., vol. 20, no. 5, 2008, pages 581 - 587
VEY N; BOURHIS JH; BOISSEL N ET AL.: "A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission", BLOOD, vol. 120, no. 22, 2012, pages 4317 - 4323
WARREN HS; SKIPSEY LJ.: "Loss of activation-induced CD45RO with maintenance of CD45RA expression during prolonged culture of T cells and NK cells", IMMUNOLOGY, vol. 74, no. 1, 1991, pages 78 - 85
WILLEMZE R; RODRIGUES CA; LABOPIN M ET AL.: "KIR-ligand incompatibility in the graft-versus-host direction improves outcomes after umbilical cord blood transplantation for acute leukemia", LEUKEMIA, vol. 23, no. 3, 2009, pages 492 - 500
XU Z; WEISS A.: "Negative regulation ofCD45 by differential homodimerization of the alternatively spliced isoforms", NAT IMMUNOL., vol. 3, no. 8, 2002, pages 764 - 771

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020168254A1 (en) * 2019-02-14 2020-08-20 Research Institute At Nationwide Children's Hospital Use of plasma membrane particles, liposomes, and exosomes to assay immune cell potency
CN113588934A (en) * 2021-06-18 2021-11-02 创模生物科技(北京)有限公司 Detection method for immune system reconstruction capability of PBMC (peripheral blood mononuclear cell) and application thereof

Also Published As

Publication number Publication date
WO2014207009A3 (en) 2015-03-19

Similar Documents

Publication Publication Date Title
JP7057669B2 (en) Methods and Compositions for Dosing in Adoptive Cell Therapy
Lee et al. Haploidentical natural killer cells infused before allogeneic stem cell transplantation for myeloid malignancies: a phase I trial
US20200239845A1 (en) Expanded nk cells
JP6215394B2 (en) A population of natural killer cells cultured ex vivo
Lamb et al. Natural killer cell therapy for hematologic malignancies: successes, challenges, and the future
US9657269B2 (en) Regulatory B cells (tBREGS) and their use
KR20180086204A (en) Composition for use in immunotherapy
Locatelli et al. At the Bedside: Innate immunity as an immunotherapy tool for hematological malignancies
Alexia et al. Polyoxidonium® activates cytotoxic lymphocyte responses through dendritic cell maturation: clinical effects in breast cancer
WO2015154012A1 (en) Clonogenic natural killer (nk) cell populations and methods of producing and using such populations
CA2878928A1 (en) Toxicity management for anti-tumor activity of cars
JP2017526938A (en) Method for diagnosing hematological cancer
Bayati et al. The therapeutic potential of regulatory T cells: challenges and opportunities
US11447747B2 (en) Methods for allogenic hematopoietic stem cell transplantation
EP3786287A1 (en) Cd3-negative cell population expressing chemokine receptor and cell adhesion molecule, use thereof, and method for producing same
WO2020172328A1 (en) Expansion of natural killer and chimeric antigen receptor-modified cells
EP3834849A1 (en) Method for treating tumor using immune effector cell
Killig et al. Tracking in vivo dynamics of NK cells transferred in patients undergoing stem cell transplantation
US20210008110A1 (en) Activated lymphocytic cells and methods of using the same to treat cancer and infectious conditions
WO2019084234A1 (en) Methods and compositions for treating cd33+ cancers and improving in vivo persistence of chimeric antigen receptor t cells
CN115003698A (en) anti-TCR antibody molecules and uses thereof
Meehan et al. Adoptive cellular therapy using cells enriched for NKG2D+ CD3+ CD8+ T cells after autologous transplantation for myeloma
Roshandel et al. NK cell therapy in relapsed refractory multiple myeloma
WO2014207009A2 (en) Methods and kits for determining whether a nk cell is activated and is able to proliferate
KR20080091776A (en) Method of using hepatic progenitors in treating liver dysfunction

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14732878

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 14732878

Country of ref document: EP

Kind code of ref document: A2