WO2014048920A1 - Method for polyclonal stimulation of t cells by mobile nanomatrices - Google Patents

Method for polyclonal stimulation of t cells by mobile nanomatrices Download PDF

Info

Publication number
WO2014048920A1
WO2014048920A1 PCT/EP2013/069854 EP2013069854W WO2014048920A1 WO 2014048920 A1 WO2014048920 A1 WO 2014048920A1 EP 2013069854 W EP2013069854 W EP 2013069854W WO 2014048920 A1 WO2014048920 A1 WO 2014048920A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
nanomatrix
cell
matrix
nanomatrices
Prior art date
Application number
PCT/EP2013/069854
Other languages
French (fr)
Inventor
Alexander Scheffold
Mario Assenmacher
Original Assignee
Miltenyi Biotec Gmbh
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 Miltenyi Biotec Gmbh filed Critical Miltenyi Biotec Gmbh
Priority to JP2015532453A priority Critical patent/JP6216793B2/en
Priority to CA2878997A priority patent/CA2878997C/en
Priority to EP13766319.1A priority patent/EP2900809B1/en
Priority to CN201380045752.3A priority patent/CN104619831A/en
Priority to DK13766319.1T priority patent/DK2900809T3/en
Priority to ES13766319.1T priority patent/ES2632450T3/en
Priority to US14/430,875 priority patent/US20150240204A1/en
Publication of WO2014048920A1 publication Critical patent/WO2014048920A1/en

Links

Classifications

    • 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
    • C12N5/0636T lymphocytes
    • C12N5/0637Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • 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
    • A61K39/46449Melanoma antigens
    • A61K39/464491Melan-A/MART
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • C12N5/0068General culture methods using substrates
    • C12N5/0075General culture methods using substrates using microcarriers
    • 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
    • C12N5/0636T lymphocytes
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/51B7 molecules, e.g. CD80, CD86, CD28 (ligand), CD152 (ligand)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • 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
    • C12N2539/00Supports and/or coatings for cell culture characterised by properties

Definitions

  • the present invention relates generally to the field of immunology, in particular to processes for polyclonal stimulation of T cells by nanomatrices.
  • Antibodies against CD3 are a central element in many T cell proliferation protocols. Immobilized on a surface, anti-CD3 delivers an activating and proliferation-inducing signal by crosslinking of the T cell receptor complex on the surface of T cells. By immobilizing anti- CD3 and anti-CD28 to simultaneously deliver a signal and a co-stimulatory signal, proliferation can be increased (Baroja et al (1989), Cellular Immunology, 120: 205-217). In WO09429436A1 solid phase surfaces such as culture dishes and beads are used to immobilize the anti-CD3 and anti-CD28 antibodies. Regularly, the immobilization on beads is performed on DynaBeads ® M-450 having a size of 4.5 ⁇ in diameter.
  • EP01257632B1 describes a method for stimulating a population of T-cells by simultaneous T- cell concentration and cell surface moiety ligation that comprises providing a population of cells wherein at least a portion thereof comprises T-cells, contacting the population of cells with a surface, wherein the surface has attached thereto one or more agents that ligate a cell surface moiety of at least a portion of the T-cell and stimulates at least that portion of T cells or a subpopulation thereof and applying a force that predominantly drives T-cell concentration and T-cell surface moiety ligation, thereby inducing T-cell stimulation.
  • force refers to a force used to drive the cells and may include a variety of forces that function similarly, and include a force greater than gravitational force, a hydraulic force, a filtration force generated by transmembrane pressure, a centrifugal force, or a magnetic force.
  • EP1257632B1 describes that ratios of particles to cells can vary, however certain preferred values include at least 1:4 to 6: 1, with one preferred ratio being at least 2: 1 beads per T-cell.
  • DynaBeads ® M-450 having a size of 4.5 ⁇ in diameter coupled to anti-CD3 and anti-CD28 antibodies were used in experiments in a bead/T-cell ratio of 3: 1.
  • T cell stimulation agents such as anti-CD3 and anti-CD28 antibodies.
  • These surfaces are cell-sized and comparable with the T cells themselves.
  • US2008/0317724A1 discloses that the spatial presentation of signal molecules can dramatically affect the response of T cells to those signal molecules. For example, when anti- CD3 and anti-CD28 antibodies are placed on separate predefined regions of a substrate, T cells incubated on the substrate secrete different amounts of interleukin-2 and/or exhibit spikes in calcium, depending not only on the types but also on the spacing of these signal molecules.
  • a pattern was generated with anti-CD3 and anti-CD28 antibodies, where anti-CD3 antibodies occupied a central feature surrounded by satellite features of anti- CD28 antibodies that were spaced about 1 to 2 microns from the central anti-CD3 feature.
  • anti-CD28 antibody features were spaced about 1 to 2 microns apart, the T cell secretion of interleukin-2 (IL-2) was enhanced compared to when the anti-CD3 and anti- CD28 antibodies were presented together to the T cells in "co-localized" features.
  • IL-2 interleukin-2
  • US8,012,750B2 discloses a biodegradable device for activating T-cells.
  • the biodegradable support is first formulated into a shape, such as a microsphere.
  • the biodegradable supports then coated with a first material providing a reactive surface which is capable of binding to second materials.
  • the second materials have a reactive surface which permits binding to surface structures on a cell.
  • the biodegradable support can be formulated into various shapes. Microspheres are a preferred formulation because of the simplicity of manufacture and the spherical shape allows an increased surface area for interaction with cellular receptors.
  • nanospheres do not provide enough cross-linking to activate naive T-cells and thus can only be used with previously activated T-cells.
  • experimental data were generated with spheres co-immobilized with anti-CD3 and anti-CD28 antibodies ranging in size from 4 to 24 microns with a mean of 7 microns.
  • US2012/121649A1 discloses methods of expanding antigen- specific anti-tumorigenic T cells comprising administering to a subject or to cells in vitro an antigen/MHC/co- stimulatory molecule/nanoparticle complex in an amount sufficient to stimulate expansion of an antigen- specific anti-tumorigenic T cell.
  • the nanoparticle is from about 1 nm to 10 nm. The abilities of the pMHC and pMHC/anti-CD28 mAb-conjugated nanoparticles to stimulate and activate cognate naive CD8+ T cells were compared.
  • the nanoparticle can further comprise one or more of a biodegradable coating formed from dextran and other molecules. E.g.
  • US2012/121649A1 does not disclose a polyclonal stimulation of T cells, e.g. with anti CD3 and antiCD28 mAbs immobilized on nanoparticles.
  • US2010/284965 A 1 discloses methods for activating T cells by administering to the cells polymeric nano- or microparticles which have an anti-CD28 antibody attached.
  • the microparticles are in the range of between 0.5 and 1000 microns and nanoparticles in the range of between 50 nm to less than 0.5 nm.
  • the particles consist of PLGA and thus represent spherically shaped solid surface particles.
  • US2010/284965 A 1 does not disclose polyclonal stimulation with the nanoparticles.
  • beads or microspheres used in the state of the art for polyclonal T cell activation via immobilized T cell stimulatory antibodies regularly are cell-sized (mostly 1 to 10 ⁇ in size), uniformly round-shaped particles. Beads of this size have several disadvantages with regard to their potential to interact with T cells as well as their production, handling and safety in clinical T cell therapy procedures.
  • T cell stimulation strength is mostly determined by the density of antibodies on the cell surface and not by the number of beads/cell
  • polyclonal T cell stimulatory agents such as antibodies, e.g. against CD3 and CD28, attached to nanomatrices, which are characterised by a mobile polymeric matrix backbone (non-solid surface), which may have embedded within the matrix additional functional compounds, such as magnetic nanocrystals, can be used to stimulate naive and memory T cells in vitro, although their diameter is smaller than 1 ⁇ , preferentially smaller than 500 nm, more preferentially smaller than 200 nm.
  • the nanomatrix is biodegradable and non-toxic to living cells, i.e. the nanomatrix is a biologically inert entity with regard to alterations of the cell function.
  • the nanomatrix used in the method of the present invention improves the in-vitro stimulation of T-cells by saving the viability of the cells.
  • sterile filtration of the small nanomatrices is possible which is an important feature for long term T cell in vitro expansion under conditions which are compliant with rigorous GMP standards and is a valuable option for clinical application of the in vitro expanded T cells.
  • these nanomatrices are well suited for use in sterile and closed cell culture system such as described e.g. in WO2009/072003 (CliniMACS ® Prodigy, Miltenyi Biotec GmbH, Germany).
  • the T cell stimulatory agents such as antibodies, e.g. against CD3 and CD28, attached to nanomatrices may be conjugated to separate nanomatrices (instead of conjugating to the same nanomatrix), which can be mixed hereafter for optimised use.
  • the ratio of nanomatrices to cells is larger than 100: 1, preferentially larger than 500: 1, most preferentially larger than 1000: 1.
  • the present invention provides the use of the nanomatrices disclosed herein for the polyclonal in-vitro stimulation of T cells.
  • the present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, wherein the nanomatrix comprises a matrix of mobile polymer chains, and has attached thereto one or more agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; and wherein the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size.
  • the nanomatrix is biologically inert with regard to alterations of the cell function.
  • the nanomatrix is biodegradable.
  • the stimulated and optionally expanded T cells achieved with the present invention can be used in subsequent therapeutic or non-therapeutic applications without the need for eliminating or removing the nanomatrix due to the property of the nanomatrix of being biologically inert with regard to alterations of the cell function
  • the nanomatrices can easily be diluted by repeated washing steps to effective concentrations below the T cell activation threshold after the T-cell stimulation process.
  • the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm in size.
  • the nanomatrix is a mobile matrix consisting of a polymeric material but has no solid phase surface in contrast to beads or microspheres.
  • Agents such as anti-CD3 and/or anti-CD28 antibodies which allow for polyclonal stimulation of T cells are attached to the mobile polymer chains of the matrix.
  • additional substances such as magnetic nanocrystals, fluorescent dyes, etc., can be embedded and add additional functions to the nanomatrix without altering its basic mobile structure, surface features, or cell interaction parameters of the nanomatrix.
  • this typical feature of polysaccharide embedded superparamagnetic nanocrystals is best described by the discrepancy in the size values obtained by different methods used for size determination.
  • transmission electron microscopy uses dried samples and thus determines mainly the size of the embedded nanocyrstals, which are always in the range of 10 nm.
  • contrast dynamic laser scattering which also takes into consideration the size of the surrounding matrix in aqueous solution, determines much larger diameters (e.g. 5-10 nm versus 80-150nm for AMI-25 a clinical contrast reagent, consisting of a dextran matrix and embedded ironoxide crystals.
  • these examples indicate the current common knowledge about the behaviour of mobile, flexible polymer matrices in aqueous solution (prerequisite for all cell culture applications) and highlight the fact that also the biophysical properties of polymer coated colloids, such as the ironoxide containing polysaccharide matrices used in this study but not limited to those, are determined mainly by the feature of polymer material, independent of the embedded substances.
  • the present invention provides an in vitro method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
  • the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size, and wherein the majority (i.e. more than 50%), preferentially more than 80 % and more preferentially more than 90% and most preferentially more than 99% of the total volume of the nanomatrix in aqueous solution consists of mobile polymer chains.
  • the resulting complex consists mainly of mobile polymer chains, making up more than 50%, preferentially more than 80 % and more preferentially more than 90% and most preferentially more than 99% of the volume in aqueous solution.
  • FIG 1 CD3/CD28 Nanomatrices for naive T cell expansion.
  • Sorted human naive CD4 and CD8 T cells were stimulated with CD3/CD28 conjugated nanomatrices at the indicated concentrations (effective CD3 concentration) in the presence of IL-2 for 7 days. Nanomatrices conjugated with CD3 and CD28 in various indicated ratios were compared. As a high control CD3/CD28 conjugated MACSiBeads were used. The absolute number of viable cells in the culture at day 7 is given.
  • FIG 2 A-C Nano-sized solid particles versus Nanomatrix for stimulation of T cells.
  • As activation parameters either CD3 down-regulation (FIG 2A, day 3) or induction of the early activation markers CD25 (day 3 FIG 2B, day 5 FIG2C) were analysed by flow- cytometry. The values were normalized to the value of the same cells stimulated via CD3/CD28 nanomatrix (100 ng/ml CD3). The values from 4 different donors are given.
  • FIG 3 Expansion of sorted human T cell populations: Human T cells were sorted into various subpopulations (total T cells, total naive T cells, naive CD4+ T cells, naive CD8+ T cells) and stimulated with CD3/CD28 conjugated nanomatrices at the indicated concentrations (effective CD3 concentration) and a CD3/CD28 ratio of 1: 1 in the presence of IL-2 for 7 days. As a high control CD3/CD28 conjugated MACSiBeads were used. The absolute number of viable cells in the culture at day 7 is given.
  • FIG 4 Expansion of human Treg: CD25+ Treg were isolated from PBMC by magnetic CD25 selection and expanded in the presence or absence of 100 nM Rapamycin for 14 days using CD3/CD28 nanomatrix (200 ng/ml CD3) and high dose IL-2. On day 7 the cells were restimulated by adding fresh nanomatrix + IL-2. On day 14 the number of viable Treg was determined. The frequency of Foxp3 expressing cells was determined by intracellular immunofluorescence. Each dot represents an individual healthy donor.
  • FIG5 Comparison of CD3 and CD28 conjugated to different nanomatrices versus conjugated to the same nanomatrix.
  • Sorted human naive CD4 and CD8 T cells were stimulated either with CD3/CD28 conjugated nanomatrices or CD3 and CD28 conjugated to different nanomatrices at the indicated concentrations (effective CD3 concentration) in the presence of IL-2 for 7 days.
  • CD3/CD28 conjugated MACSiBeads were used as a high control CD3/CD28 conjugated MACSiBeads. The absolute number of viable cells in the culture at day 7 is given (A). Results from two donors are depicted. In addition the cells were labelled with CFSE and the proliferative activity measured on day 5 after activation (1 representative donor) (B).
  • FIG 6 Comparison of soluble CD3 or CD28 antibodies to CD3 or CD28 antibodies conjugated to nanomatrices.
  • Naive CD4 and CD8 T cells were isolated and stimulated in vitro in the presence of IL-2 using soluble CD3 (0-lOOOOng/mL) w/o CD28 (A) or in the presence of soluble CD28 (200 ng/ml) (B) and compared to CD28 conjugated to nanomatrix (200 ng/ml) and analysed for early activation markers CD25/CD69 on day 5 or for expansion (C) on day 7. 2 donors were analysed in duplicates.
  • FIG 7 Transduction efficiency of isolated T cell subsets stimulated with various stimulation agents.
  • T cell subsets naive (T N , CD62L+ CD45RA+), central memory (T CM , CD62L+CD45RA-) and effector (T EM , CD62L-CD45RA-) T cells activated using CD3/CD28 nanomatrices, plate-bound CD3 + soluble CD28 or CD3/CD28 conjugated MACSibeads and transduced them using a retroviral vector expressing a TCR specific for MART-1.
  • a standard total PBMC were activated using soluble CD3/CD28.
  • the frequency of transduced cells expressing the MART-1 TCR was determined using a fluorescently labelled MART- 1/HLA-A2 tetramer.
  • FIG 8 Enriched T cell subsets expand at least like PBMC or better.
  • CD8 + T cell subsets from freshly isolated PBMC from melanoma patients were isolated and the stimulated with coated CD3 plus soluble CD28 (CD3+CD28 in the graph) or with CD3/CD28/CD2 coated MACSiBeads (MACSiBeads in the graph) or with CD3/CD28 Nanomatrix in the presence of IL2. After 2 days of stimulation cells have been transduced to express MART-1 TCR. In the graph is reported the fold expansion of each culture at day 13- 15 after stimulation.
  • FIG 9 Naive and central memory cells stimulated with MACSiBeads or CD3/CD28 nanomatrix show a less terminal differentiated phenotype.
  • PBMC were freshly isolated from leukapheresis of melanoma patients.
  • CD8+ T cell subsets were enriched and then stimulated with CD3+CD28 or MACSiBeads or CD3/CD28 nanomatrix in the presence of IL2.
  • PBMC instead were stimulated with soluble CD3 and IL2.
  • Cells were transduced to express MART-1 TCR 48h after stimulation. The data here were obtained 13-15 days after stimulation of each culture.
  • FIG 10 Cytokine secretion upon MART-1 restimulation in CD8 + T cell subsets.
  • CD8 + T cell subsets from freshly isolated PBMC from melanoma patients were isolated and the stimulated with CD3+CD28 MACSiBeads or with CD3/CD28 nanomatrix in the presence of IL2.
  • PBMC from the same melanoma patient were stimulated with soluble CD3 and IL2.
  • After 2 days of stimulation cells have been transduced to express MART-1 TCR and cultured for a total 13- 15 days. Afterwards cells were washed out from IL2 and rested for further 2 days and then restimulated with MART-1 + HLA-A2 + melanoma cell line for 6h.
  • the cytokine production was determined by intracellular staining.
  • Graphs show the frequencies of A) IFNy + ; B) IL2 + and C) TNFcc + CD8+ T cells.
  • beads or microspheres with solid phase surfaces used to stimulate polyclonally T cells are always larger than 1 ⁇ in size in the state of the art, regularly they are cell-sized.
  • nanomatrices being smaller than 1 ⁇ , preferentially smaller than 500 nm, more preferentially smaller than 200 nm, having a mobile matrix and having attached thereto polyclonal stimulatory agent(s) are convenient to stimulate T cells. It is essential to the present invention that the nanomatrix being smaller than 500 nm has no solid phase surface (resulting in a flexible and mobile phase) in contrast to beads or microspheres of the same size (see Example 3).
  • the nanomatrix is like a mesh or net consisting of a mobile polymeric material, preferentially dextran.
  • the nanomatrix is very plastic resulting in the ability to snuggle to the cell surface membrane of target cells, i.e. the T cells which shall be activated.
  • the nanomatrix binds with its agents attached to the mobile matrix to the respective receptors (antigens) on the cell surface, whereby the flexibility of the matrix allows optimal interaction with the binding partners.
  • the shape of the nanomatrix adapts to the target cell surface thereby extending the contacting surface between nanomatrix and target cell. Due to the size of the nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm, they are too small to cause perturbance in the cell, i.e. the nanomatrix is biologically inert with regard to alterations of the cell function. Such perturbances triggered by direct cell/bead contact is problematic if beads or microspheres of 1 ⁇ or larger in size are used.
  • the nanomatrix is biodegradable and non-toxic to the cells due to the composition consisting of biodegradable polymeric material such as a polymer of dextran.
  • the nanomatrix is a completely biologically inert entity with regard to alterations of the cell function but biodegradable. Therefore there is no need to remove the nanomatrix after contacting it with the T cells for stimulation and proliferation. No disturbing effects occur due to the presence of the nanomatrices in an activated T cell composition for subsequent analysis, experiments and/or clinical applications of these cells.
  • the unbound nanomatrices can easily be diluted by repeated washing steps to effective concentrations below the T cell activation threshold after the T-cell stimulation process.
  • the mobile matrix of the nanomatrix has attached thereto one or more stimulatory agents which provide activation signal(s) to the T cells, thereby activating and inducing the T cells to proliferate.
  • the agents are molecules which are capable of binding to a cell surface structure and induce the polyclonal stimulation of the T cells.
  • agents attached to the mobile matrix of the nanomatrix is anti-CD3 monoclonal antibody (mAb) in combination with a co- stimulatory protein such as anti-CD28 mAb.
  • mAb monoclonal antibody
  • Other examples are anti-CD2, anti- CD137, anti-CD134, Notch-ligands, e.g. Delta-like 1/4, Jaggedl/2 either alone or in various combinations with anti-CD3.
  • T cells to be stimulated are e.g.
  • the stimulatory agent attached to the mobile matrix of the nanomatrix is anti-CD3 monoclonal antibody (mAb) in combination with the co- stimulatory protein anti-CD28 mAb.
  • the present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
  • the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size.
  • the nanomatrix may be biologically inert with regard to alteration of the cell function.
  • the nanomatrix may be biodegradable.
  • the nanomatrix may be of cGMP quality for clinical applications. Sterility can be achieved e.g. by sterile filtration using filters with suitable pore size (200 nm) or by other methods well known by the person skilled in the art. GMP processing of cell products is frequently performed in closed cell culture systems. Use of the nanomatrix in GMP cultivation systems is of advantage due to:
  • the feature of the nanomatrix of being able to pass sterile filters allows the addition to closed cell culture systems being equipped or equipable with sterile filters, e.g. cell cultivation bags (Miltenyi Biotec, Baxter, CellGenics), G-Rex devices (Wilson Wolf manufacturing), WAVE Bioreactors (GE Healthcare), Quantum Cell Expansion System (Terumo BCT), CliniMACS® Prodigy (Miltenyi Biotec, see Apel et al. 2013, Chemie Ingenieurtechnik 85: 103-110).
  • the nanomatrix can be added to closed cell culture systems using a syringe to push the nanomatrix through the filter or a pump to pull the nanomatrix through the filter from a bag or a vial (connected to a vented vial adapter).
  • Rigid cell sized magnetic particles used to induce T cell proliferation have a higher density than lymphocytes including T cells (1.07 g/1) and thus sediment faster than the nanomatrix.
  • Agitated cell culture systems such as the WAVE Bioreactor (GE Healthcare) avoid complete sedimentation of the cells by movement of the container used for cultivation. Movement of the container applies different forces to cells / particles of different size and density, resulting in a relative movement. Induction of proliferation is reduced within the WAVE Bioreactor compared to static culture in cell culture flasks.
  • CD3CD28 MACSiBeads (“ExpAct T reg kit”, “T cell expansion kit”, Miltenyi Biotec) utilizing rigid cell sized magnetic particles has been optimized to be used in a given ratio of beads per cell (e.g. 1:2), requiring determination of the number of T cells within the culture container and defined cell density, i.e. an optimized number of cells and beads per surface area, enabling optimal interaction of beads and cells, which both sediment to the bottom of the culture vessel within minutes. For a clinical application of expanded T cells this results in an additional sampling step, possibly affected sterility of the product.
  • the nanomatrix of the present invention allows for dosing of the T cell proliferation inducing agent based on the volume of the culture, since the reagent can freely diffuse in the medium and does not sediment.
  • the volume of the T cell containing cell suspension can be determined by a balance or a camera system without affecting the sterility barrier. Dosing of the agent based on volume determination allows for a completely automated cell product manufacturing process (e.g. using the CliniMACS® Prodigy system) without manual cell counting steps.
  • the contacting can occur e.g. in vitro in any container capable of holding cells, preferably in a sterile environment.
  • containers may be e.g. culture flasks, culture bags, bioreactors or any device that can be used to grow cells (e.g. the sample processing system of WO2009072003, i.e. the CliniMACS® Prodigy system).
  • the present inventions provides a method for polyclonal stimulation of T cells in a closed cell culture system, the method comprising contacting a cell culture comprising a population of T cells within said closed cell culture system with a dosage of nanomatrix, the nanomatrix comprising
  • nanomatrix is 1 to 500 nm in size
  • said dosage of nanomatrix is applied sterile to said closed cell culture system, and wherein said dosage depends on the volume of the cell culture in said closed cell culture system.
  • Said volume of the cell culture can be determined by a balance or a camera system without affecting the sterility barrier.
  • Determining and applying said dosage may be performed automatically.
  • the nanomatrix used in the present invention can be a nanomatrix wherein at least one first agent and one second agent are attached to the same mobile matrix. Nanomatrices of this kind are contacted with T cells, thereby activating and inducing the T cells to proliferate.
  • the ratio of the first and the second agent attached to the same flexible matrix may be in the range of the ratios of 100: 1 to 1: 100, preferentially between 10: 1 and 1: 10, most preferentially between 2:1 and 1:2.
  • the nanomatrix of the present invention also can be a nanomatrix wherein at least one first agent and one second agent are attached to separate mobile matrices. A mixture of these nanomatrices is contacted with T cells, thereby activating and inducing the T cells to proliferate (see Example 6).
  • the ratio and/or concentration of the mobile matrix having attached thereto the first agent and the mobile matrix having attached thereto the second agent may vary to yield optimal stimulation results depending on the kind of T cells used and/or agents used. This facilitates the optimisation of the activation conditions for specialised T cell subsets by titrating various concentrations and ratios of the mobile matrix having attached thereto the first agent and the mobile matrix having attached thereto the second agent.
  • the ratio of nanomatrices to cells is larger than 100: 1, preferentially larger than 500: 1, most preferentially larger than 1000: 1.
  • the large amount of nanomatrices per cell allows for a fine-tuning of the separate labelled nanomatrices which would be impossible with lower ratios of 1: 10 to 10: 1 commonly used by stimulation of T cells using cell-sized beads.
  • the nanomatrix used in the present invention is a nanomatrix wherein the mobile matrix consists of a polymeric, preferentially biodegradable material which is non-toxic to cells.
  • the nanomatrix used in the present invention is a nanomatrix wherein the mobile matrix consists of a polymer of dextran.
  • polymeric material such as dextran for generating the matrix results in a mobile, flexible matrix.
  • the mobility (flexibility or plasticity) of the matrix is superior compared to more rigid particles (having a solid phase surface) or beads of the same size, i.e. nanostructures, for proliferation of cells.
  • the nanomatrix used in the present invention can be a nanomatrix wherein the mobile matrix is the only or at least main component of the nanomatrix regardless the agents which are attached thereto.
  • the nanomatrix used in the present invention also can be a nanomatrix wherein the nanomatrix carries magnetic, paramagnetic, superparamagnetic nano-crystals, or fluorescent dyes embedded into the flexible matrix, preferentially embedded into the polymer of dextran.
  • the nanomatrix used in the present invention can be used in a method for stimulating T-cells with this nanomatrix wherein the nanomatrix is not removed in subsequent applications of the stimulated T cells.
  • the nanomatrix used in the present invention also can be used in a method for stimulating T-cells with this nanomatrix wherein the nanomatrix is removed before subsequent applications of the stimulated T cells.
  • the present invention also provides a composition comprising
  • nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size
  • the nanomatrix may be biologically inert with regard to alteration of the cell function.
  • the nanomatrix may be biodegradable.
  • Agents are attached to the same or separate nanomatrices at high density, with more than 25 ⁇ g per mg nanomatrix, preferentially with more than 50 ⁇ g per mg nanomatrix.
  • composition may be convenient for generating T cell therapeutics for in vivo use (pharmaceutical composition).
  • composition also can be used in other subsequent analyses and experiments.
  • the present invention also provides a composition or a pharmaceutical composition comprising a population of stimulated and (optionally expanded) T cells produced according to the method of the present invention.
  • the pharmaceutical composition may comprise a population of stimulated T cells produced by the method of the present invention, wherein the method is performed in a closed cell culture system such as the sample processing system of WO2009072003.
  • Nanomatrices can be prepared by various methods known in the art, including solvent evaporation, phase separation, spray-drying, or solvent extraction at low temperature. The process selected should be simple, reproducible and scalable. The resulting nanomatrices should be free-flowing and not aggregate in order to produce a uniform syringeable suspension. The nanomatrix should also be sterile. This can be ensured by e.g. filtration, a terminal sterilization step and/or through aseptic processing. A preparation of nanomatrices is described in Example 1.
  • matrix of mobile polymer chains and “mobile matrix” as used herein have an interchangeable meaning.
  • the term “bulmobile” refers to the common and well described feature of organic biopolymers such as dextran or others on nanoparticles (see Bertholon et al. Langmuir 2006, pp45485-5490). These polymers consists of mobile (motile), preferentially highly mobile (motile) chains, so the matrix is characterised by the absence of a solid surface as the attachment point for the stimulating agents such as antibodies, and which is in strong contrast to currently used beads or microspheres which regularly have an inflexible, stiff surface. As a result the nanomatrix comprising a matrix of mobile polymer chains is flexible and adjustable to the form of the surface of the cells.
  • the nanomatrix is a nanomatrix wherein the majority (i.e. more than 50%), preferentially more than 80 % and more preferentially more than 90% and most preferentially more than 99% of the total volume of the nanomatrix in aqueous solution consists of mobile polymer chains.
  • the contact between nanomatrix which has coupled thereto one or more stimulatory agents and the cells to be stimulated benefit from the fact that the nanomatrix has not a fixed, stiff or rigid surface allowing the nanomatrix to snuggle to the cells.
  • the matrix consists of a polymeric, preferentially biodegradable or biocompatible inert material which is non-toxic to cells.
  • the matrix is composed of hydrophilic polymer chains, which obtain maximal mobility in aqueous solution due to hydration of the chains.
  • the mobile matrix is the only or at least main component of the nanomatrix regardless the agents which are attached thereto.
  • the mobile matrix may be of collagen, purified proteins, purified peptides, polysaccharides, glycosaminoglycans, or extracellular matrix compositions.
  • a polysaccharide may include for example, cellulose ethers, starch, gum arabic, agarose, dextran, chitosan, hyaluronic acid, pectins, xanthan, guar gum or alginate.
  • polymers may include polyesters, polyethers, polyacrylates, polyacrylamides, polyamines, polyethylene imines, polyquaternium polymers, polyphosphazenes, polyvinylalcohols, polyvinylacetates, polyvinylpyrrolidones, block copolymers, or polyurethanes.
  • the mobile matrix is a polymer of dextran.
  • the mobile matrix defines the property of the nanomatrix of being very plastic, i.e. flexible, leading to the ability to snuggle to the cell surface membrane of target cells, i.e. the T cells which shall be activated and proliferated. Therefore, the nanomatrix tightly binds with its agents attached to the mobile matrix to the cells because the mobility of the matrix provides optimal access of the attached ligands or antibodies to their cell surface receptors or antigens. Due to this property the nanomatrix has the ability to provide enough cross-linking to activate T cells regardless of the small size of the structure, i.e. smaller than 1 ⁇ , preferentially smaller than 500 nm, more preferentially smaller than 200 nm, in size.
  • the adaptability of the nanomatrix caused by the mobile, flexible nanostructure extends the contacting area between cell surface membrane and the nanomatrix resulting in more efficient bindings between cell surface molecules and agents attached to the mobile matrix.
  • the mobile matrix may embed magnetic, paramagnetic or superparamagnetic nano-crystals or other substances which add additional functional properties such as fluorescent dyes without altering the basic mobile structure and/or surface features, i.e. interaction with target cells.
  • stimulatory agent which is attached to the mobile matrix of the nanomatrix as used herein refers to molecules which are capable of binding to a cell surface structure and contribute to a polyclonal stimulation of the T cells.
  • the terms "stimulatory agent” and “polyclonal stimulatory agent” as used herein have an interchangeable meaning.
  • suitable agents for use in the present invention include agents such as synthesized compounds, nucleic acids and proteins, including polyclonal or monoclonal antibodies, and fragments or derivatives thereof, and bioengineered proteins, such as fusion proteins. In one example, the agents are mitogenic proteins.
  • Mito genie proteins are two or more proteins that are able to deliver the requisite minimum of two signals to T-cells in order to cause the T-cells to become activated.
  • mitogenic proteins are anti-CD3 and anti-CD2 monoclonal antibodies (mAb) in combination with a co-stimulatory protein such as and including proteins specific for one or more of the following T-cell surface molecules: CD28, CD5, CD4, CD8, MHCI, MHCII, CTLA-4, ICOS, PD-1, OX40, CD27L (CD70), 4-1BBL, CD30L and LIGHT, including the corresponding ligands to these surface structures, or fragments thereof.
  • mAb monoclonal antibodies
  • Suitable agents include agents capable of delivering a signal to T-cells through cytokine receptors such as IL-2R, IL-12R, IL-1R, IL-15R; IFN-gammaR, TNF-alphaR, IL-4R, and IL- 10R, including monoclonal antibodies (mAbs) to these receptors, fusion proteins with a reactive end specific for these receptors and the corresponding ligands to these receptors or fractions thereof.
  • mAbs monoclonal antibodies
  • Other suitable agents include any agent capable of binding to cellular adhesion molecules on T-cells such as mAbs, fusion proteins and the corresponding ligands or fractions thereof to adhesion molecules in the following categories: cadherins, ICAM, integrins, and selectins.
  • adhesion molecules on T-cells are: CD44, CD31, CD18/CDl la (LFA-1), CD29, CD54 (ICAM-1), CD62L (L-selectin), and CD29/CD49d (VLA-4).
  • suitable agents include any agents capable of binding to chemokine receptors, including those in the C-C and C-X-C categories.
  • chemokine receptors associated with T-cell function include CCR1, CCR2, CCR3, CCR4, CCR5, and CXCR3.
  • An agent may be attached or coupled to the mobile matrix by a variety of methods known and available in the art.
  • the attachment may be covalent or noncovalent, electrostatic, or hydrophobic and may be accomplished by a variety of attachment means, including for example, chemical, mechanical, enzymatic, or other means whereby an agent is capable of stimulating the cells.
  • the antibody to a cell surface structure first may be attached to the matrix, or avidin or strep tavidin may be attached to the matrix for binding to a biotinylated agent.
  • the antibody to the cell surface structure may be attached to the matrix directly or indirectly, e.g. via an anti-isotype antibody.
  • Another example includes using protein A or protein G, or other non-specific antibody binding molecules, attached to matrices to bind an antibody.
  • the agent may be attached to the matrix by chemical means, such as cross-linking to the matrix.
  • antibody is intended to include polyclonal and monoclonal antibodies, chimeric antibodies, haptens and antibody fragments, and molecules which are antibody equivalents in that they specifically bind to an epitope on the antigen.
  • antibody includes polyclonal and monoclonal antibodies of any isotype (IgA, IgG, IgE, IgD, IgM), or an antigen-binding portion thereof, including, but not limited to, F(ab) and Fv fragments such as sc Fv, single chain antibodies, chimeric antibodies, humanized antibodies, and a Fab expression library.
  • biologically inert refers to the properties of the nanomatrix, that it is non-toxic to living cells and does not induce strong alterations of the cell function via physical interaction with the cell surface, due to its small size, except the specific ligand/receptor triggering function of the attached ligands or antibodies.
  • the nanomatrices may be biodegradable, e.g. degraded by enzymatic activity or cleared by phagocytic cells.
  • the biodegradable material can be derived from natural or synthetic materials that degrade in biological fluids, e.g. cell culture media and blood. The degradation may occur using enzymatic means or may occur without enzymatic means.
  • the biodegradable material degrades within days, weeks or few months, which may depend on the environmental conditions it is exposed to.
  • the biodegradable material should be non-toxic and non-antigenic for living cells and in humans.
  • the degradation products must produce non-toxic by-products.
  • An important aspect in the context of being biologically inert is the fact that the nanomatrix does not induce strong alteration in structure, function, activity status or viability of labelled cells, i.e. it does not cause perturbance of the cells and does not interfere with subsequent experiments and therapeutic applications of the stimulated cells.
  • the mechanical or chemical irritation of the cell is decreased due to the properties of the nanomatrix of being very small, i.e. nano-scale range, and having a mobile matrix which rather snuggles to the cell surface than altering the shape of the cell surface or exerting strong shearing force to the cells, e.g. resulting in membrane rupture.
  • PBMC peripheral blood mononuclear cells isolated from human peripheral blood preparations e.g. by use of a density gradient (e.g. Ficoll, PanColl). "PBMC” consist of lymphocytes and monocytes.
  • T cells refers to cell populations with an increased content of T cells.
  • T cells may be purified by different methods, including FACS® sorting, enrichment by magnetic beads (e.g. CD4 and CD8 MicroBeads, Miltenyi Biotec), magnetic bead depletion of non-T cells, Ficoll based methods (e.g. RosetteSep®, Stem Cell Technologies) or whole blood isolation methods (MACSxpress®, Miltenyi Biotec), as well as panning on antibody coated surfaces.
  • FACS® sorting enrichment by magnetic beads (e.g. CD4 and CD8 MicroBeads, Miltenyi Biotec), magnetic bead depletion of non-T cells, Ficoll based methods (e.g. RosetteSep®, Stem Cell Technologies) or whole blood isolation methods (MACSxpress®, Miltenyi Biotec), as well as panning on antibody coated surfaces.
  • CD4 and CD8 MicroBeads e.g. CD4 and CD8 MicroBeads
  • a first nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto one agent, e.g. anti CD3 mAb.
  • a second nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto another agent, e.g. anti CD28mAb.
  • the nanomatrix of the present invention is a nanomatrix wherein at least one first agent and one second agent are attached to separate mobile matrices.
  • a mixture of these nanomatrices is contacted with T cells, thereby activating and inducing the T cells to proliferate.
  • Fine-tuning of nanomatrices for the stimulation of the T cells is easily performed due to the high ratio of nanomatrices to cells (normally larger than 500: 1).
  • a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto one agent, e.g. anti CD3 mAb.
  • the nanomatrix of the present invention is a nanomatrix wherein at least one first agent is attached to mobile matrices. This nanomatrix is contacted with T cells, thereby activating and inducing the T cells to proliferate.
  • a second or more (multiple) co- stimulating agents may be added as soluble agents to optimize or support the activation induced by the nanomatrix with the first agent attached thereto.
  • a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto two agents which provide activation signals to the cell, e.g. anti CD3mAb and anti CD28 mAb.
  • the nanomatrix of the present invention is a nanomatrix wherein at least one first agent and one second agent are attached to the same flexible matrix.
  • Nanomatrices of this kind are contacted with T cells, thereby activating and inducing the T cells to proliferate.
  • a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto multiple agents which provide activation signals to the cell, e.g. anti-CD3mAb and anti-CD28 mAb, anti ICOS, anti-CD137 or other known co- stimulatory molecules.
  • the nanomatrix of the present invention is a nanomatrix wherein at least one first agent and multiple other agents are attached to the same mobile matrix.
  • Nanomatrices of this kind are contacted with T cells, thereby activating and inducing the T cells to proliferate.
  • a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto one or more agents which provide activation signals to the cells, e.g. anti CD3mAb and/or anti CD28 mAb.
  • the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the polymer.
  • the nanomatrix is contacted with T cells, thereby activating and inducing the T cells to proliferate.
  • the unbound magnetic, paramagnetic or superparamagnetic nanomatrix may be removed by applying a magnetic field gradient.
  • the cells labelled with the magnetic nanomatrices may be separated by applying a magnetic field gradient, in particular a high-gradient magnetic field, and subsequent expansion of the purified T cells.
  • the nanomatrix Although there is no need to remove the nanomatrix after activation and proliferation of the population of T cells due to their property of being biologically inert with regard to alteration of the cell function one might optionally remove the nanomatrix with mild washing conditions, which are sufficient to wash way the nanomatrices from the cells or cell culture.
  • the nanomatrices can easily be diluted by repeated washing steps to effective concentrations below the T cell activation threshold. This optionally removing step is much easier performed with the nanomatrices than with beads or microspheres well known in the state of the art due to their small size. If the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the polymer than optionally the removal step can be performed by applying a magnetic field gradient to the cell/nanomatrix mixture.
  • the method of the invention can be used to expand selected T cell populations for use in treating an infectious disease or cancer.
  • the resulting T cell population can be genetically transduced and used for immunotherapy or can be used for in vitro analysis of infectious agents such as HIV.
  • Proliferation of a population of CD4+ cells obtained from an individual or patient, e.g. infected with HIV, can be achieved and the cells rendered resistant, e.g. to HIV infection.
  • the expanded T cells are re-infused into the individual or patient.
  • a population of tumor-infiltrating lymphocytes can be obtained from an individual afflicted with cancer and the T cells stimulated to proliferate to sufficient numbers and restored to the individual.
  • supernatants from cultures of T cells expanded in accordance with the method of the invention are a rich source of cytokines and can be used to sustain T cells in vivo or ex vivo.
  • a nanomatrix as described in any proceeding embodiment may be used in a closed cell culture system, e.g. the sample processing system of WO2009072003 or cell cultivation bags (Miltenyi Biotec, Baxter, CellGenics), G-Rex devices (Wilson Wolf manufacturing), WAVE Bioreactors (GE Healthcare), Quantum Cell Expansion System (Terumo BCT).
  • the nanomatrices have optimal connectivity to such a closed cell culture system, they can be easily sterile filtrated and integrated into the closed cell culture system.
  • the nanomatrix can be added in relation to the volume of the culture rather than the T cell number.
  • Culture volume can be assesses by a balance or a camera system (e.g. in the sample processing system of WO2009072003).
  • the use of the method of the present invention within a closed cell culture system such as the sample processing system of WO2009072003 results in a safe and easy way to produce a pharmaceutical composition of stimulated T cells due to the reduced risk of e.g. contaminating agents such as other eukaryotic cells, bacteria or viruses (safer and faster handling by the operator).
  • cultivation of T cells by use of the nanomatrix is performed within a magnetic field.
  • the nanomatrix consists of superparamagnetic cores embedded into the mobile matrix.
  • magnetic forces are induced in the nanomatrix and ligands triggering activating T cell receptors can be concentrated for improved induction of proliferation.
  • Cultivation of T cells within a magnetic field can be performed in a column consisting of a ferromagnetic matrix (e.g. of WO2009072003) or in bag, flask or chamber in close proximity to strong permanent magnet.
  • the present invention has broad applicability to any cell type having a cell surface moiety that may be stimulated.
  • many cell signalling events can be enhanced by the method of the present invention.
  • Such methodologies can be used therapeutically in an ex vivo setting to activate and stimulate cells for infusion into a patient or could be used in vivo, to induce cell signalling events on a target cell population.
  • the target cells of the method are T cells, but are in no way limited thereto.
  • the T cells Prior to stimulation of T cells by the present invention the T cells may be directly identified and/or separated or isolated from blood, peripheral mononuclear blood cells (PBMC), body tissue or cells from tissue fluids.
  • PBMC peripheral mononuclear blood cells
  • the cells are normally identified and/or separated from cell samples from mammals such as humans, mouse, or rat, but especially from humans and preferably from test subject and/or patients.
  • the separation is performed by well known sorting methods in the art. This includes for example affinity chromatography or any other antibody-dependent separation technique known in the art. Any ligand-dependent separation technique known in the art may be used in conjunction with both positive and negative separation techniques that rely on the physical properties of the cells.
  • An especially potent sorting technology is magnetic cell sorting. Methods to separate cells magnetically are commercially available e.g. from Invitrogen, Stemcell Technologies, Cellpro, Advanced Magnetics, or Miltenyi Biotec.
  • T cell subpopulations such as CD4+ T cells, CD8+ T cells, NKT cells, ⁇ / ⁇ T cells, ⁇ ⁇ T cells, CD4+CD25+Foxp3+ regulatory T cells, naive T cells (CD45RA+CCR7+ and/or CD62L+) or central memory T cells (CD45R0+CCR7+), effector memory T cells (CD45R0+CCR7-) or terminal effector T cells (CD45RA+CCR7-).
  • T cell subpopulations such as CD4+ T cells, CD8+ T cells, NKT cells, ⁇ / ⁇ T cells, ⁇ ⁇ T cells, CD4+CD25+Foxp3+ regulatory T cells, naive T cells (CD45RA+CCR7+ and/or CD62L+) or central memory T cells (CD45R0+CCR7+), effector memory T cells (CD45R0+CCR7-) or terminal effector T cells (CD45RA+CCR7-).
  • Target cell populations such as the T cell populations obtained via the present disclosure may be administered either alone, or as a pharmaceutical composition in combination with diluents and/or with other components such as IL-2 or other cytokines or cell populations.
  • pharmaceutical compositions of the present disclosure may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • a pharmaceutical composition may comprise a) a population of T cells, wherein said T cells are proliferated to therapeutically effective amounts according to the present invention; and b) one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • compositions of the present disclosure are preferably formulated for intravenous administration.
  • Pharmaceutical compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
  • Magnetic nanomatrices were produced by a modification of the procedure of Molday and MacKenzie.
  • Ten grams of Dextran T40 (Pharmacia Uppsala, Sweden), 1.5 g FeCl 3 ⁇ 6 H 2 0 and 0.64 g FeCl 2 ⁇ 4 H 2 0 are dissolved in 20 ml H 2 0, and heated to 40°C. While stirring, 10 ml 4N NaOH are added slowly and the solution is heated to 70°C for 5 min. The particle suspension is neutralized with acetic acid. To remove aggregates the suspension is centrifuged for 10 min at 2,000 g and filtrated through a 0.22 ⁇ pore-size filter (Millex GV, Millipore, Molsheim, France).
  • HGMF washing of magnetic nanomatrices is performed in steelwool columns made as described below and placed in a magnetic field of approx. 0.6 Tesla (MACS permanent magnet, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany).
  • MCS permanent magnet Miltenyi Biotec GmbH, Bergisch Gladbach, Germany.
  • Ten milliliters of nanomatrix suspension are applied to a 15 x 40 mm column of 2 g steelwool.
  • the loaded column is washed with 30 ml 0.05 M sodium acetate.
  • the magnetic nanomatrices are eluted with 0.05 M sodium acetate.
  • the nanomatrices form a brown suspension.
  • the relative particle concentration is given as optical density at 450 nm.
  • the size of the nanomatrices was determined by electron microscopy and dynamic light scattering to be 30 + 20nm (e.m.) and 65 + 20 nm (DLS). Dextran content and magnetite content of the matrix within the colloidal solution were determined to be in the range of about 3.5 mg/ml and 4.8 mg/ml, respectively, resulting approximately in a 40:60 w/w ratio. Based on the density of the dried nanomatrix of 2,5g/mL determined by pycnometer and the known densities of dextran and magnetite of 1.6 g/ml and 5.2 g/ml, respectively, the volume of dextran can be calculated to be about 70% for the dried nanomatrix. The nanomatrices show superparamagnetic behavior, as determined by susceptibility measurements. The size of the trapped ferrit microcrystals was determined from magnetic measurements to be approximately 10 nm.
  • CD3 antibodies (clone OKT3) and CD28 antibodies (clone 15E8) (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) were conjugated to the same or separate nanomatrices by standard bioconjugation chemistry (Bioconjugate Techniques, 2nd Edition, By Greg T. Hermanson, Published by Academic Press, Inc., 2008).
  • Example 2 Expansion of T cells using nanomatrices at various CD3/CD28 concentrations and ratios versus CD3/CD28 MACSiBeads
  • the current state-of-the-art reagents for activation of highly purified T cells comprise activating antibodies against CD3/CD28 immobilized either on the surfaces of a cell culture dish or large cell-sized (4-5 ⁇ ) particles. Both techniques are error prone and technically difficult to realize and standardize, especially under GMP-compatible production conditions. In contrast nanomatrices can be easily prepared and conveniently be used for cell culture under GMP-conditions. Therefore we compared the T cell activation potential by analysing the expansion potential of the CD3/CD28 coated nanomatrices at various concentrations and CD3/CD28 ratios with commercially available cell stimulation beads (MACSiBeads, ⁇ 4,5 ⁇ , Miltenyi Biotec GmbH).
  • MCSiBeads commercially available cell stimulation beads
  • the nanomatrices expand T cells efficiently even at very low CD3 concentrations (20 - 100 ng/ml) which are also typically used for soluble CD3/CD28 in the presence of accessory cells which provide crosslinking. Besides the antibody concentration the CD3/CD28 ratio can also influence the cell activation and provides an additional means to optimize the T cell culture.
  • the expansion at optimal doses (20 - 300 ng/ml) was similar or better than the standard reagent (MACSiBeads). At higher doses the expansion was reduced due to overstimulation of the T cells (activation- induced cell death), a phenomenon known to occur at a too high degree of TCR stimulation. Taken together, these results show that CD3/CD28 coated nanomatrices can efficiently activate and expand T cells at very low antibody concentrations and without the need for additional crosslinking.
  • Example 3 Comparison CD3/CD28 conjugated to nanomatrices versus 200 nm and 300 nm solid particles
  • Soluble antibodies stimulating e.g. against CD3 and CD28
  • an accessory cells e.g. Immunoglobulin Fc-receptors.
  • Reagents which do not depend on an extra crosslinking step to be used for T cell activation e.g. to stimulate highly purified T cells in the absence of accessory cells, are based on cell-sized particles ( ⁇ 4-5 ⁇ ) coated with stimulating CD3 and CD28 antibodies. It is known that solid particles below a critical diameter of about 1 ⁇ are not suitable to properly expand T cells.
  • TCR stimulation Another direct result of TCR stimulation is the downregulation of the T cell receptor, which can be analysed via loss of CD3 expression on the cell surface.
  • Highly purified T cells were cultured with CD3/CD28 coated nanomatrices (100 ng/ml CD3) or solid particles with a diameter of 4.5 ⁇ (MACSiBeads) or 200 nm (Ademtech beads) both covalently coated with CD3 and CD28 antibodies.
  • MACSiBeads were used at an optimal 1: 1 ratio whereas 200 nm particles were titrated to achieve an active dose of CD3 and CD28 ranging from 25-3000 ng/ml CD3.
  • the frequency of CD25+ T cells and on day 3 the expression intensity of CD3 was measured.
  • B and C nanomatrices lead to strong activation at the optimal dose (100 ng/ml) as shown by up-regulation of CD25 [FIG 2B (day 3), FIG 2C (day5)] and downregulation of CD3 (FIG 2A) which occurred at similar levels like with the cell-sized MACSiBeads.
  • up-regulation of CD25 [FIG 2B (day 3), FIG 2C (day5)]
  • downregulation of CD3 (FIG 2A) which occurred at similar levels like with the cell-sized MACSiBeads.
  • no CD25 up-regulation and almost no CD3 down-regulation was seen for 200 nm solid particles even at 30 fold higher CD3/CD28 concentrations.
  • Even on day five 200 nm solid particles were not able to induce CD25 expression to a similar level like the nanomatrix. Only at high concentrations (5-30 fold higher than for the nanomatrix) there was a slight upregulation observed achieving about 50-70% of the levels of the nanomatrix.
  • T cell subsets can have different activation requirements.
  • naive T cells are difficult to activate in the absence of accessory cells.
  • CD4 and CD8 T cells may have different needs when activated alone or in presence of additional cell types.
  • Treg are of particular interest for therapeutic applications for transplantation, autoimmunity and chronic inflammation and Treg are difficult to expand in vitro without loss of regulatory activity, i.e. Foxp3 expression. Therefore we also analysed whether CD25 selected Treg cells (Foxp3 purity typically 60-90%) from various donors can be expanded using nanomatrices. To support growth of Treg versus conventional T cells the expansion was done in the presence of 100 nM Rapamycine, a well described drug inhibiting conventional T cell growth. As shown in FIG 4 following 14 days of culture Treg can be expanded 10-20 times (w/o Rapa) or 5-10 times (with Rapa). As described before without Rapa the Foxp3 purity was highly variable (10-75%) whereas in the presence of Rapa the purity was always >50%.
  • Example 6 Comparison of T cell activation by CD3/CD28-conjugated to the same nanomatrix versus CD3 and CD28 conjugated to separate nanomatrices.
  • the stimulation with the CD3 nanomatrix alone did not induce significant expansion and only few cell divisions can be observed, as it is expected for naive T cells which depend on a costimulatory signal.
  • the CD28 nanomatrix already at 10-50 ng/ml induced full cell dividing activity and also expansion of T cell numbers, which was similar to the CD3/CD28 control nanomatrix or the standard MACSiBeads.
  • both antibodies may be conjugated to separate nanomatrices, which can be mixed hereafter for optimised use. This facilitates the production process and quality control of the single nanomatrices and improves the flexibility of the reagent, e.g. facilitating the optimisation of the activation conditions for specialised T cell subsets by titrating various CD3 and CD28 concentrations and ratios (fine-tuning).
  • Example 7 The effect of conjugation of soluble CD3 or CD28 to the nanomatrix.
  • soluble CD3 In the presence of a saturating amount (200 ng/ml) of soluble CD28 as costimulator (FIG 6B) soluble CD3 also induced CD25/CD69 expression in about 20-40% of the cells at the highest tested doses (100-10000 ng/ml). In contrast the CD3 coated nanomatrix (100 ng/ml CD3) induced CD25/CD69 expression in 40-70% of the cells.
  • CD28 conjugated to the nanomatrix was tested. Since the effects of costimulation are best visualized under suboptimal CD3 stimulation, we titrated CD28 either soluble or conjugated to the nanomatrix in the presence of soluble CD3 to a culture of naive T cells. As shown in FIG 6C soluble CD3 alone similar to the induction of CD25/CD69 as shown above did not induce any expansion of the naive T cells. In the presence of soluble CD28 however a 2-6 fold expansion was detectable but only at the highest tested dose of CD28 (10000 ng/ml). In contrast to this, CD28 conjugated to nanomatrix induced a similar degree of expansion already at a 1000 fold lower concentration (10 ng/ml).
  • Example 8 Nanomatrices can be used to activate T cells for introduction of TCR genes by viral transduction
  • T N purified naive
  • T CM central memory
  • T EM effector
  • T CM and T EM CD8 + T cell subsets compare to T N in all stimulatory conditions but this was not statistically significant (FIG 10 top panel). Focussing on the IL-2 production (FIG 10 middle panel) we observed that a higher percentage of T cells produces IL-2 when they have been stimulated with MACSiBeads/nanomatrices when compared to coated ccCD3+aCD28 stimulation. The same is true for TNFcc producing cells detected in T N subset when stimulated with MACSiBeads (FIG 10 bottom panel). These results indicate cells of T derived cells stimulated with beads showed diminished effector cell differentiation, suggesting less progress toward terminal differentiation.
  • CD3/CD28 nanomatrices can be used to efficiently activate and transduce purified T cell subsets to generate fully functional T cell transplants, e.g. for tumor therapy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising a) a matrix of mobile polymer chains, and b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; wherein the nanomatrix is 1 to 500 nm in size. At least one first and one second stimulatory agents are attached to the same or to separate mobile matrices. If the stimulatory agents are attached to separate nanomatrices, fine-tuning of nanomatrices for the stimulation of the T cells is possible. Closed cell culture systems also benefit from this method.

Description

Title
Method for polyclonal stimulation of T cells by mobile nanomatrices Field of invention
The present invention relates generally to the field of immunology, in particular to processes for polyclonal stimulation of T cells by nanomatrices.
Background of the invention
Antibodies against CD3 are a central element in many T cell proliferation protocols. Immobilized on a surface, anti-CD3 delivers an activating and proliferation-inducing signal by crosslinking of the T cell receptor complex on the surface of T cells. By immobilizing anti- CD3 and anti-CD28 to simultaneously deliver a signal and a co-stimulatory signal, proliferation can be increased (Baroja et al (1989), Cellular Immunology, 120: 205-217). In WO09429436A1 solid phase surfaces such as culture dishes and beads are used to immobilize the anti-CD3 and anti-CD28 antibodies. Regularly, the immobilization on beads is performed on DynaBeads®M-450 having a size of 4.5 μιη in diameter.
EP01257632B1 describes a method for stimulating a population of T-cells by simultaneous T- cell concentration and cell surface moiety ligation that comprises providing a population of cells wherein at least a portion thereof comprises T-cells, contacting the population of cells with a surface, wherein the surface has attached thereto one or more agents that ligate a cell surface moiety of at least a portion of the T-cell and stimulates at least that portion of T cells or a subpopulation thereof and applying a force that predominantly drives T-cell concentration and T-cell surface moiety ligation, thereby inducing T-cell stimulation. The term force as used herein refers to a force used to drive the cells and may include a variety of forces that function similarly, and include a force greater than gravitational force, a hydraulic force, a filtration force generated by transmembrane pressure, a centrifugal force, or a magnetic force. EP1257632B1 describes that ratios of particles to cells can vary, however certain preferred values include at least 1:4 to 6: 1, with one preferred ratio being at least 2: 1 beads per T-cell. Regularly, DynaBeads®M-450 having a size of 4.5 μιη in diameter coupled to anti-CD3 and anti-CD28 antibodies were used in experiments in a bead/T-cell ratio of 3: 1. Again, these methods use solid phase surfaces to co-immobilize T cell stimulation agents such as anti-CD3 and anti-CD28 antibodies. These surfaces are cell-sized and comparable with the T cells themselves. US2008/0317724A1 discloses that the spatial presentation of signal molecules can dramatically affect the response of T cells to those signal molecules. For example, when anti- CD3 and anti-CD28 antibodies are placed on separate predefined regions of a substrate, T cells incubated on the substrate secrete different amounts of interleukin-2 and/or exhibit spikes in calcium, depending not only on the types but also on the spacing of these signal molecules. For example, a pattern was generated with anti-CD3 and anti-CD28 antibodies, where anti-CD3 antibodies occupied a central feature surrounded by satellite features of anti- CD28 antibodies that were spaced about 1 to 2 microns from the central anti-CD3 feature. When the anti-CD28 antibody features were spaced about 1 to 2 microns apart, the T cell secretion of interleukin-2 (IL-2) was enhanced compared to when the anti-CD3 and anti- CD28 antibodies were presented together to the T cells in "co-localized" features.
The publication of Erin R Steenblock and Tarek M Fahmy (Molecular Therapy vol. 16 no. 4, 765-772 April 2008) uses solid-surface nanoparticles (130 nm) and show that these nanoparticle stimulate T cells weaker than microparticles (8 μιη). The authors stated that these findings are supported by those of previous reports (Mescher, MF (1992). J Immunol 149: 2402-2405.), demonstrating that micron-sized particles, which are close in size to T cells, provide optimal T-cell stimulation. Mesher" s study demonstrated the critical importance of a large, continuous surface contact area for effective CTL activation. Using class I alloantigen immobilized on latex microspheres, particle sizes of 4 to 5 microns were found to provide an optimum stimulus. Below 4 microns, responses decreased rapidly with decreasing particle size, and large numbers of small particles could not compensate for suboptimal size. US8,012,750B2 discloses a biodegradable device for activating T-cells. The biodegradable support is first formulated into a shape, such as a microsphere. The biodegradable supports then coated with a first material providing a reactive surface which is capable of binding to second materials. The second materials have a reactive surface which permits binding to surface structures on a cell. The biodegradable support can be formulated into various shapes. Microspheres are a preferred formulation because of the simplicity of manufacture and the spherical shape allows an increased surface area for interaction with cellular receptors. According to US8,012,750B2 nanospheres do not provide enough cross-linking to activate naive T-cells and thus can only be used with previously activated T-cells. Again, experimental data were generated with spheres co-immobilized with anti-CD3 and anti-CD28 antibodies ranging in size from 4 to 24 microns with a mean of 7 microns.
US2012/121649A1 discloses methods of expanding antigen- specific anti-tumorigenic T cells comprising administering to a subject or to cells in vitro an antigen/MHC/co- stimulatory molecule/nanoparticle complex in an amount sufficient to stimulate expansion of an antigen- specific anti-tumorigenic T cell. The nanoparticle is from about 1 nm to 10 nm. The abilities of the pMHC and pMHC/anti-CD28 mAb-conjugated nanoparticles to stimulate and activate cognate naive CD8+ T cells were compared. The nanoparticle can further comprise one or more of a biodegradable coating formed from dextran and other molecules. E.g. iron (III) oxide and PLGA can be combined to form nanoparticle resulting in a solid surface particle. US2012/121649A1 does not disclose a polyclonal stimulation of T cells, e.g. with anti CD3 and antiCD28 mAbs immobilized on nanoparticles. US2010/284965 A 1 discloses methods for activating T cells by administering to the cells polymeric nano- or microparticles which have an anti-CD28 antibody attached. The microparticles are in the range of between 0.5 and 1000 microns and nanoparticles in the range of between 50 nm to less than 0.5 nm. The particles consist of PLGA and thus represent spherically shaped solid surface particles. In addition, US2010/284965 A 1 does not disclose polyclonal stimulation with the nanoparticles.
Taken together, beads or microspheres used in the state of the art for polyclonal T cell activation via immobilized T cell stimulatory antibodies regularly are cell-sized (mostly 1 to 10 μιη in size), uniformly round-shaped particles. Beads of this size have several disadvantages with regard to their potential to interact with T cells as well as their production, handling and safety in clinical T cell therapy procedures.
1. Due to the solid surface of the bead the size of interaction area between the bead and cells is limited.
2. Their preparation is complex and costly as compared to soluble antibodies and it is especially inconvenient to generate them in cGMP quality, e.g. due to their size no sterile filtration is possible, sedimentation complicates handling, i.e. constant particle number/volume during filling and antibody loading.
3. They are inconvenient to use for in vitro processes to generate T cell therapeutics for in vivo use, since they have to be added to cells in defined cell/bead ratios at defined density cell/beads per surface area,
adaption of stimulation strength is only possible to some extent, since the T cell stimulation strength is mostly determined by the density of antibodies on the cell surface and not by the number of beads/cell
aliquoting is inaccurate due to sedimentation,
sterile filtration is not possible
due to their size they might affect cell viability and function and they cannot simply be removed from cells by centrifugation. Therefore either special protocols for "bead removal" or biodegradable particles have been developed. However both methods suffer from inaccuracies with regard to the actual number of residual beads after the removal process, leaving behind a certain risk for toxic effects if T cell stimulatory beads are injected into patients. This problem is particularly relevant because of the size of the particles, since each single particle on its own might still have retained the capacity to activate T cells in vivo.
Therefore, there is a need for an improved in- vitro method for polyclonal T cell stimulation.
Summary of the invention
Surprisingly, it was found that polyclonal T cell stimulatory agents such as antibodies, e.g. against CD3 and CD28, attached to nanomatrices, which are characterised by a mobile polymeric matrix backbone (non-solid surface), which may have embedded within the matrix additional functional compounds, such as magnetic nanocrystals, can be used to stimulate naive and memory T cells in vitro, although their diameter is smaller than 1 μιη, preferentially smaller than 500 nm, more preferentially smaller than 200 nm. Contrary thereto, it was found that beads with solid surfaces of the same size as the nanomatrices used herein are not able to stimulate T cells at all or to a similar level like the nanomatrices which is in accordance with the well established opinion of the person skilled in the art. Due to their small size the nanomatrices per se, without antibodies attached thereto, do not alter structure, function, activity status or viability of cells, i.e. they do not cause perturbance in the cells and do not interfere with subsequent analyses, experiments and therapeutic applications of the stimulated cells. In addition, preferentially, the nanomatrix is biodegradable and non-toxic to living cells, i.e. the nanomatrix is a biologically inert entity with regard to alterations of the cell function. Therefore the nanomatrix used in the method of the present invention improves the in-vitro stimulation of T-cells by saving the viability of the cells. In addition sterile filtration of the small nanomatrices is possible which is an important feature for long term T cell in vitro expansion under conditions which are compliant with rigorous GMP standards and is a valuable option for clinical application of the in vitro expanded T cells. Moreover these nanomatrices are well suited for use in sterile and closed cell culture system such as described e.g. in WO2009/072003 (CliniMACS®Prodigy, Miltenyi Biotec GmbH, Germany).
In addition surprisingly, it was found that the T cell stimulatory agents such as antibodies, e.g. against CD3 and CD28, attached to nanomatrices may be conjugated to separate nanomatrices (instead of conjugating to the same nanomatrix), which can be mixed hereafter for optimised use. In general, the ratio of nanomatrices to cells is larger than 100: 1, preferentially larger than 500: 1, most preferentially larger than 1000: 1. This results in the possibility of fine-tuning of the nanomatrices used for stimulation of the target T cells, e.g. it facilitates the production process and quality control of the single nanomatrices and improves the flexibility of the reagent, e.g. facilitating the optimisation of the activation conditions for specialised T cell subsets by titrating various CD3 and CD28 concentrations and ratios.
In a first aspect the present invention provides the use of the nanomatrices disclosed herein for the polyclonal in-vitro stimulation of T cells. In a further aspect the present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, wherein the nanomatrix comprises a matrix of mobile polymer chains, and has attached thereto one or more agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; and wherein the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size. Preferentially, the nanomatrix is biologically inert with regard to alterations of the cell function. In addition preferentially, the nanomatrix is biodegradable.
The stimulated and optionally expanded T cells achieved with the present invention can be used in subsequent therapeutic or non-therapeutic applications without the need for eliminating or removing the nanomatrix due to the property of the nanomatrix of being biologically inert with regard to alterations of the cell function
Alternatively, due to being soluble or colloidal the nanomatrices can easily be diluted by repeated washing steps to effective concentrations below the T cell activation threshold after the T-cell stimulation process. The nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm in size. The nanomatrix is a mobile matrix consisting of a polymeric material but has no solid phase surface in contrast to beads or microspheres. Agents such as anti-CD3 and/or anti-CD28 antibodies which allow for polyclonal stimulation of T cells are attached to the mobile polymer chains of the matrix. Within the matrix additional substances, such as magnetic nanocrystals, fluorescent dyes, etc., can be embedded and add additional functions to the nanomatrix without altering its basic mobile structure, surface features, or cell interaction parameters of the nanomatrix.
This mobility, i.e. flexibility of polymeric matrices in aqueous solutions even when attached to solid surfaces is well known in the field. For example, a quantitative estimation of the flexibility or mobility of dextran and other polymers used for biological or medical applications is given in Bertholon et al. Langmuir 2006, pp45485-5490.
The mobility introduced to the stimulation matrix by the used polymer is well described in multiple publications for polysaccharides and embedded superparamagnetic nanocrystals, which are frequently used for in vivo imaging technologies in clinical situations. These reagents are very similar to the type of reagent which was used here in several of the examples given. It is well known that despite the term "particle" these reagents are actually not comparable to spherical solid surface particles:
"Most of the colloidal (so called) nanoparticles used for medical purpose are coated with polymer or polyelectrolytes and they cannot be really considered as rigid spheres (Di Marco et al. Int J. Nanomed. 2007, p618, line 5 ff. )".
To exemplify directly: this typical feature of polysaccharide embedded superparamagnetic nanocrystals is best described by the discrepancy in the size values obtained by different methods used for size determination. Typically transmission electron microscopy uses dried samples and thus determines mainly the size of the embedded nanocyrstals, which are always in the range of 10 nm. However, in contrast dynamic laser scattering which also takes into consideration the size of the surrounding matrix in aqueous solution, determines much larger diameters (e.g. 5-10 nm versus 80-150nm for AMI-25 a clinical contrast reagent, consisting of a dextran matrix and embedded ironoxide crystals. This means that >99 of the total volume of the complex in aqueous solution is made up by the mobile matrix (Wang et al. Eur. Radiol. 2001: page 2323). For the matrices which were used for example in Example 1, size measurement revealed 30 nm (TEM) and 65 nm (DLS), thus about 80-90 % of the complex in aqueous solution consists of the mobile matrix. Therefore in these types of mobile matrices, the mobile polymer determines the biophysical behaviour during interaction with cells, rather than a stiff particle or solid surface covered, which might in addition be coated with a thin layer of polymer used for attachment. Thus the well-known flexibility or mobility (motility) of the polymer chains in the described matrix is the determining feature and critical difference to solid surface attached antibodies on microspheres classically used for stimulation.
In summary, these examples indicate the current common knowledge about the behaviour of mobile, flexible polymer matrices in aqueous solution (prerequisite for all cell culture applications) and highlight the fact that also the biophysical properties of polymer coated colloids, such as the ironoxide containing polysaccharide matrices used in this study but not limited to those, are determined mainly by the feature of polymer material, independent of the embedded substances.
Therefore the present invention provides an in vitro method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains; and
b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate, wherein the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size, and wherein the majority (i.e. more than 50%), preferentially more than 80 % and more preferentially more than 90% and most preferentially more than 99% of the total volume of the nanomatrix in aqueous solution consists of mobile polymer chains.
If additional substances, such as magnetic nano-crystals, fluorescent dyes, etc. are embedded into the matrix, the resulting complex consists mainly of mobile polymer chains, making up more than 50%, preferentially more than 80 % and more preferentially more than 90% and most preferentially more than 99% of the volume in aqueous solution.
Brief description of the drawings
FIG 1: CD3/CD28 Nanomatrices for naive T cell expansion.
Sorted human naive CD4 and CD8 T cells were stimulated with CD3/CD28 conjugated nanomatrices at the indicated concentrations (effective CD3 concentration) in the presence of IL-2 for 7 days. Nanomatrices conjugated with CD3 and CD28 in various indicated ratios were compared. As a high control CD3/CD28 conjugated MACSiBeads were used. The absolute number of viable cells in the culture at day 7 is given.
FIG 2 A-C: Nano-sized solid particles versus Nanomatrix for stimulation of T cells. Nano- sized solid particles (AdemtechBeads, diameter = 200nm) were conjugated with CD3/CD28 (1: 1) and used for stimulation of naive CD4 and CD8 T cells at the indicated concentrations of CD3. As activation parameters either CD3 down-regulation (FIG 2A, day 3) or induction of the early activation markers CD25 (day 3 FIG 2B, day 5 FIG2C) were analysed by flow- cytometry. The values were normalized to the value of the same cells stimulated via CD3/CD28 nanomatrix (100 ng/ml CD3). The values from 4 different donors are given.
FIG 3: Expansion of sorted human T cell populations: Human T cells were sorted into various subpopulations (total T cells, total naive T cells, naive CD4+ T cells, naive CD8+ T cells) and stimulated with CD3/CD28 conjugated nanomatrices at the indicated concentrations (effective CD3 concentration) and a CD3/CD28 ratio of 1: 1 in the presence of IL-2 for 7 days. As a high control CD3/CD28 conjugated MACSiBeads were used. The absolute number of viable cells in the culture at day 7 is given.
FIG 4: Expansion of human Treg: CD25+ Treg were isolated from PBMC by magnetic CD25 selection and expanded in the presence or absence of 100 nM Rapamycin for 14 days using CD3/CD28 nanomatrix (200 ng/ml CD3) and high dose IL-2. On day 7 the cells were restimulated by adding fresh nanomatrix + IL-2. On day 14 the number of viable Treg was determined. The frequency of Foxp3 expressing cells was determined by intracellular immunofluorescence. Each dot represents an individual healthy donor. FIG5: Comparison of CD3 and CD28 conjugated to different nanomatrices versus conjugated to the same nanomatrix. Sorted human naive CD4 and CD8 T cells were stimulated either with CD3/CD28 conjugated nanomatrices or CD3 and CD28 conjugated to different nanomatrices at the indicated concentrations (effective CD3 concentration) in the presence of IL-2 for 7 days. As a high control CD3/CD28 conjugated MACSiBeads were used. The absolute number of viable cells in the culture at day 7 is given (A). Results from two donors are depicted. In addition the cells were labelled with CFSE and the proliferative activity measured on day 5 after activation (1 representative donor) (B).
FIG 6: Comparison of soluble CD3 or CD28 antibodies to CD3 or CD28 antibodies conjugated to nanomatrices. Naive CD4 and CD8 T cells were isolated and stimulated in vitro in the presence of IL-2 using soluble CD3 (0-lOOOOng/mL) w/o CD28 (A) or in the presence of soluble CD28 (200 ng/ml) (B) and compared to CD28 conjugated to nanomatrix (200 ng/ml) and analysed for early activation markers CD25/CD69 on day 5 or for expansion (C) on day 7. 2 donors were analysed in duplicates.
FIG 7: Transduction efficiency of isolated T cell subsets stimulated with various stimulation agents. T cell subsets, naive (TN, CD62L+ CD45RA+), central memory (TCM, CD62L+CD45RA-) and effector (TEM, CD62L-CD45RA-) T cells activated using CD3/CD28 nanomatrices, plate-bound CD3 + soluble CD28 or CD3/CD28 conjugated MACSibeads and transduced them using a retroviral vector expressing a TCR specific for MART-1. As a standard total PBMC were activated using soluble CD3/CD28. The frequency of transduced cells expressing the MART-1 TCR was determined using a fluorescently labelled MART- 1/HLA-A2 tetramer.
FIG 8: Enriched T cell subsets expand at least like PBMC or better.
CD8+ T cell subsets from freshly isolated PBMC from melanoma patients were isolated and the stimulated with coated CD3 plus soluble CD28 (CD3+CD28 in the graph) or with CD3/CD28/CD2 coated MACSiBeads (MACSiBeads in the graph) or with CD3/CD28 Nanomatrix in the presence of IL2. After 2 days of stimulation cells have been transduced to express MART-1 TCR. In the graph is reported the fold expansion of each culture at day 13- 15 after stimulation. The fold expansion values are relative to soluble CD3 stimulated PBMC that shows a fold expansion of 57.61+17.75 FIG 9: Naive and central memory cells stimulated with MACSiBeads or CD3/CD28 nanomatrix show a less terminal differentiated phenotype. PBMC were freshly isolated from leukapheresis of melanoma patients. CD8+ T cell subsets were enriched and then stimulated with CD3+CD28 or MACSiBeads or CD3/CD28 nanomatrix in the presence of IL2. PBMC instead were stimulated with soluble CD3 and IL2. Cells were transduced to express MART-1 TCR 48h after stimulation. The data here were obtained 13-15 days after stimulation of each culture. Frequencies of A) MART-1 tetramer+ CD62L+and B) MART-1 tetramer+CCR7+cells among CD8+ T cells are shown. After IL2 withdrawal cells were stained for CD27, CD57 markers or stimulated with a MART-1+ HLA-A2+melanoma cell line in the presence of CD 107a antibody and Monensin for 5 h. Statistical analysis of C) MART- 1 tetramer+ CD27+; D) MART-1 tetramer+ CD57+ and E) CD107a+ cells among CD8+ T cells frequencies.
FIG 10: Cytokine secretion upon MART-1 restimulation in CD8+ T cell subsets. CD8+ T cell subsets from freshly isolated PBMC from melanoma patients were isolated and the stimulated with CD3+CD28 MACSiBeads or with CD3/CD28 nanomatrix in the presence of IL2. PBMC from the same melanoma patient were stimulated with soluble CD3 and IL2. After 2 days of stimulation cells have been transduced to express MART-1 TCR and cultured for a total 13- 15 days. Afterwards cells were washed out from IL2 and rested for further 2 days and then restimulated with MART-1+ HLA-A2+ melanoma cell line for 6h. The cytokine production was determined by intracellular staining. Graphs show the frequencies of A) IFNy+; B) IL2+and C) TNFcc+ CD8+ T cells.
Detailed description of the invention
It was a well established opinion in the scientific community that particles smaller than 1 μιη are not convenient to stimulate T cells effectively because such small particles do not provide enough cross-linking to activate T cells. Therefore, generally, beads or microspheres with solid phase surfaces used to stimulate polyclonally T cells are always larger than 1 μιη in size in the state of the art, regularly they are cell-sized.
Now unexpectedly, the inventors found that nanomatrices being smaller than 1 μιη, preferentially smaller than 500 nm, more preferentially smaller than 200 nm, having a mobile matrix and having attached thereto polyclonal stimulatory agent(s) are convenient to stimulate T cells. It is essential to the present invention that the nanomatrix being smaller than 500 nm has no solid phase surface (resulting in a flexible and mobile phase) in contrast to beads or microspheres of the same size (see Example 3). The nanomatrix is like a mesh or net consisting of a mobile polymeric material, preferentially dextran. The nanomatrix is very plastic resulting in the ability to snuggle to the cell surface membrane of target cells, i.e. the T cells which shall be activated. Therefore, the nanomatrix binds with its agents attached to the mobile matrix to the respective receptors (antigens) on the cell surface, whereby the flexibility of the matrix allows optimal interaction with the binding partners. To a certain degree the shape of the nanomatrix adapts to the target cell surface thereby extending the contacting surface between nanomatrix and target cell. Due to the size of the nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm, they are too small to cause perturbance in the cell, i.e. the nanomatrix is biologically inert with regard to alterations of the cell function. Such perturbances triggered by direct cell/bead contact is problematic if beads or microspheres of 1 μιη or larger in size are used. In addition, preferentially, the nanomatrix is biodegradable and non-toxic to the cells due to the composition consisting of biodegradable polymeric material such as a polymer of dextran. In consequence, the nanomatrix is a completely biologically inert entity with regard to alterations of the cell function but biodegradable. Therefore there is no need to remove the nanomatrix after contacting it with the T cells for stimulation and proliferation. No disturbing effects occur due to the presence of the nanomatrices in an activated T cell composition for subsequent analysis, experiments and/or clinical applications of these cells.
In addition, due to being soluble or colloidal the unbound nanomatrices can easily be diluted by repeated washing steps to effective concentrations below the T cell activation threshold after the T-cell stimulation process.
The mobile matrix of the nanomatrix has attached thereto one or more stimulatory agents which provide activation signal(s) to the T cells, thereby activating and inducing the T cells to proliferate. The agents are molecules which are capable of binding to a cell surface structure and induce the polyclonal stimulation of the T cells. One example for agents attached to the mobile matrix of the nanomatrix is anti-CD3 monoclonal antibody (mAb) in combination with a co- stimulatory protein such as anti-CD28 mAb. Other examples are anti-CD2, anti- CD137, anti-CD134, Notch-ligands, e.g. Delta-like 1/4, Jaggedl/2 either alone or in various combinations with anti-CD3. T cells to be stimulated are e.g. naive T cells, memory T cells, CD4 Treg and CD8 Treg cells. Preferentially, the stimulatory agent attached to the mobile matrix of the nanomatrix is anti-CD3 monoclonal antibody (mAb) in combination with the co- stimulatory protein anti-CD28 mAb.
Therefore, in one aspect the present invention provides a method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains; and
b) attached to said matrix of polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate; and wherein the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size.
The nanomatrix may be biologically inert with regard to alteration of the cell function.
In addition, or alternatively, the nanomatrix may be biodegradable. The nanomatrix may be of cGMP quality for clinical applications. Sterility can be achieved e.g. by sterile filtration using filters with suitable pore size (200 nm) or by other methods well known by the person skilled in the art. GMP processing of cell products is frequently performed in closed cell culture systems. Use of the nanomatrix in GMP cultivation systems is of advantage due to:
- feature of nanomatrix to pass a sterile filter
- a low particle density or colloidal behaviour that avoids faster sedimentation of matrix compared to T cells
- dosing of the nanomatrix in relation to the volume of the culture rather than the T cell number.
The feature of the nanomatrix of being able to pass sterile filters allows the addition to closed cell culture systems being equipped or equipable with sterile filters, e.g. cell cultivation bags (Miltenyi Biotec, Baxter, CellGenics), G-Rex devices (Wilson Wolf manufacturing), WAVE Bioreactors (GE Healthcare), Quantum Cell Expansion System (Terumo BCT), CliniMACS® Prodigy (Miltenyi Biotec, see Apel et al. 2013, Chemie Ingenieur Technik 85: 103-110). The nanomatrix can be added to closed cell culture systems using a syringe to push the nanomatrix through the filter or a pump to pull the nanomatrix through the filter from a bag or a vial (connected to a vented vial adapter).
Other agents used to induce T cell proliferation (cell sized magnetic beads, cell lines) cannot pass a sterile filter. Use of those reagents would require to connect agents to the closed cell culture system using a sterile tubing welder (e.g. Terumo, GE, Genesis). Not all of those tubing welders are designed for operation within a GMP clean room. Thus adding an agent to the closed cell culture system through a sterile filter provides a significant advantage.
Rigid cell sized magnetic particles used to induce T cell proliferation have a higher density than lymphocytes including T cells (1.07 g/1) and thus sediment faster than the nanomatrix. Agitated cell culture systems such as the WAVE Bioreactor (GE Healthcare) avoid complete sedimentation of the cells by movement of the container used for cultivation. Movement of the container applies different forces to cells / particles of different size and density, resulting in a relative movement. Induction of proliferation is reduced within the WAVE Bioreactor compared to static culture in cell culture flasks.
The use of the CD3CD28 MACSiBeads ("ExpAct T reg kit", "T cell expansion kit", Miltenyi Biotec) utilizing rigid cell sized magnetic particles has been optimized to be used in a given ratio of beads per cell (e.g. 1:2), requiring determination of the number of T cells within the culture container and defined cell density, i.e. an optimized number of cells and beads per surface area, enabling optimal interaction of beads and cells, which both sediment to the bottom of the culture vessel within minutes. For a clinical application of expanded T cells this results in an additional sampling step, possibly affected sterility of the product. Use of the nanomatrix of the present invention allows for dosing of the T cell proliferation inducing agent based on the volume of the culture, since the reagent can freely diffuse in the medium and does not sediment. The volume of the T cell containing cell suspension can be determined by a balance or a camera system without affecting the sterility barrier. Dosing of the agent based on volume determination allows for a completely automated cell product manufacturing process (e.g. using the CliniMACS® Prodigy system) without manual cell counting steps.
The contacting can occur e.g. in vitro in any container capable of holding cells, preferably in a sterile environment. Such containers may be e.g. culture flasks, culture bags, bioreactors or any device that can be used to grow cells (e.g. the sample processing system of WO2009072003, i.e. the CliniMACS® Prodigy system).
Therefore, in one aspect the present inventions provides a method for polyclonal stimulation of T cells in a closed cell culture system, the method comprising contacting a cell culture comprising a population of T cells within said closed cell culture system with a dosage of nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains; and
b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells;
wherein the nanomatrix is 1 to 500 nm in size, and
wherein said dosage of nanomatrix is applied sterile to said closed cell culture system, and wherein said dosage depends on the volume of the cell culture in said closed cell culture system.
Said volume of the cell culture can be determined by a balance or a camera system without affecting the sterility barrier.
Determining and applying said dosage may be performed automatically.
The nanomatrix used in the present invention can be a nanomatrix wherein at least one first agent and one second agent are attached to the same mobile matrix. Nanomatrices of this kind are contacted with T cells, thereby activating and inducing the T cells to proliferate. The ratio of the first and the second agent attached to the same flexible matrix may be in the range of the ratios of 100: 1 to 1: 100, preferentially between 10: 1 and 1: 10, most preferentially between 2:1 and 1:2.
In addition surprisingly, it was found that the nanomatrix of the present invention also can be a nanomatrix wherein at least one first agent and one second agent are attached to separate mobile matrices. A mixture of these nanomatrices is contacted with T cells, thereby activating and inducing the T cells to proliferate (see Example 6). The ratio and/or concentration of the mobile matrix having attached thereto the first agent and the mobile matrix having attached thereto the second agent may vary to yield optimal stimulation results depending on the kind of T cells used and/or agents used. This facilitates the optimisation of the activation conditions for specialised T cell subsets by titrating various concentrations and ratios of the mobile matrix having attached thereto the first agent and the mobile matrix having attached thereto the second agent. It is advantageous that generally the ratio of nanomatrices to cells is larger than 100: 1, preferentially larger than 500: 1, most preferentially larger than 1000: 1. The large amount of nanomatrices per cell allows for a fine-tuning of the separate labelled nanomatrices which would be impossible with lower ratios of 1: 10 to 10: 1 commonly used by stimulation of T cells using cell-sized beads.
The nanomatrix used in the present invention is a nanomatrix wherein the mobile matrix consists of a polymeric, preferentially biodegradable material which is non-toxic to cells. Preferentially, the nanomatrix used in the present invention is a nanomatrix wherein the mobile matrix consists of a polymer of dextran. Using polymeric material such as dextran for generating the matrix results in a mobile, flexible matrix. The mobility (flexibility or plasticity) of the matrix is superior compared to more rigid particles (having a solid phase surface) or beads of the same size, i.e. nanostructures, for proliferation of cells.
The nanomatrix used in the present invention can be a nanomatrix wherein the mobile matrix is the only or at least main component of the nanomatrix regardless the agents which are attached thereto.
But the nanomatrix used in the present invention also can be a nanomatrix wherein the nanomatrix carries magnetic, paramagnetic, superparamagnetic nano-crystals, or fluorescent dyes embedded into the flexible matrix, preferentially embedded into the polymer of dextran. The nanomatrix used in the present invention can be used in a method for stimulating T-cells with this nanomatrix wherein the nanomatrix is not removed in subsequent applications of the stimulated T cells.
Alternatively, the nanomatrix used in the present invention also can be used in a method for stimulating T-cells with this nanomatrix wherein the nanomatrix is removed before subsequent applications of the stimulated T cells. In another aspect the present invention also provides a composition comprising
i) the nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains, and
b) attached to said matrix of polymer chains one or more agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate;
and wherein the nanomatrix is 1 to 500 nm, preferentially 10 to 200 nm, in size
ii) a population of T cells which are activated and induced to proliferate triggered by the contact between said nanomatrix and cells.
The nanomatrix may be biologically inert with regard to alteration of the cell function.
The nanomatrix may be biodegradable.
Agents are attached to the same or separate nanomatrices at high density, with more than 25 μg per mg nanomatrix, preferentially with more than 50 μg per mg nanomatrix.
This composition may be convenient for generating T cell therapeutics for in vivo use (pharmaceutical composition). The composition also can be used in other subsequent analyses and experiments.
In another aspect the present invention also provides a composition or a pharmaceutical composition comprising a population of stimulated and (optionally expanded) T cells produced according to the method of the present invention. The pharmaceutical composition may comprise a population of stimulated T cells produced by the method of the present invention, wherein the method is performed in a closed cell culture system such as the sample processing system of WO2009072003. Nanomatrices can be prepared by various methods known in the art, including solvent evaporation, phase separation, spray-drying, or solvent extraction at low temperature. The process selected should be simple, reproducible and scalable. The resulting nanomatrices should be free-flowing and not aggregate in order to produce a uniform syringeable suspension. The nanomatrix should also be sterile. This can be ensured by e.g. filtration, a terminal sterilization step and/or through aseptic processing. A preparation of nanomatrices is described in Example 1.
Definitions
The terms "matrix of mobile polymer chains" and "mobile matrix" as used herein have an interchangeable meaning. The term„mobile" refers to the common and well described feature of organic biopolymers such as dextran or others on nanoparticles (see Bertholon et al. Langmuir 2006, pp45485-5490). These polymers consists of mobile (motile), preferentially highly mobile (motile) chains, so the matrix is characterised by the absence of a solid surface as the attachment point for the stimulating agents such as antibodies, and which is in strong contrast to currently used beads or microspheres which regularly have an inflexible, stiff surface. As a result the nanomatrix comprising a matrix of mobile polymer chains is flexible and adjustable to the form of the surface of the cells. In addition as a result the nanomatrix is a nanomatrix wherein the majority (i.e. more than 50%), preferentially more than 80 % and more preferentially more than 90% and most preferentially more than 99% of the total volume of the nanomatrix in aqueous solution consists of mobile polymer chains.
The contact between nanomatrix which has coupled thereto one or more stimulatory agents and the cells to be stimulated benefit from the fact that the nanomatrix has not a fixed, stiff or rigid surface allowing the nanomatrix to snuggle to the cells. The matrix consists of a polymeric, preferentially biodegradable or biocompatible inert material which is non-toxic to cells. Preferentially the matrix is composed of hydrophilic polymer chains, which obtain maximal mobility in aqueous solution due to hydration of the chains. The mobile matrix is the only or at least main component of the nanomatrix regardless the agents which are attached thereto.
The mobile matrix may be of collagen, purified proteins, purified peptides, polysaccharides, glycosaminoglycans, or extracellular matrix compositions. A polysaccharide may include for example, cellulose ethers, starch, gum arabic, agarose, dextran, chitosan, hyaluronic acid, pectins, xanthan, guar gum or alginate. Other polymers may include polyesters, polyethers, polyacrylates, polyacrylamides, polyamines, polyethylene imines, polyquaternium polymers, polyphosphazenes, polyvinylalcohols, polyvinylacetates, polyvinylpyrrolidones, block copolymers, or polyurethanes. Preferentially the mobile matrix is a polymer of dextran.
The mobile matrix defines the property of the nanomatrix of being very plastic, i.e. flexible, leading to the ability to snuggle to the cell surface membrane of target cells, i.e. the T cells which shall be activated and proliferated. Therefore, the nanomatrix tightly binds with its agents attached to the mobile matrix to the cells because the mobility of the matrix provides optimal access of the attached ligands or antibodies to their cell surface receptors or antigens. Due to this property the nanomatrix has the ability to provide enough cross-linking to activate T cells regardless of the small size of the structure, i.e. smaller than 1 μιη, preferentially smaller than 500 nm, more preferentially smaller than 200 nm, in size. The adaptability of the nanomatrix caused by the mobile, flexible nanostructure extends the contacting area between cell surface membrane and the nanomatrix resulting in more efficient bindings between cell surface molecules and agents attached to the mobile matrix.
In some embodiments the mobile matrix may embed magnetic, paramagnetic or superparamagnetic nano-crystals or other substances which add additional functional properties such as fluorescent dyes without altering the basic mobile structure and/or surface features, i.e. interaction with target cells.
The term "stimulatory agent" which is attached to the mobile matrix of the nanomatrix as used herein refers to molecules which are capable of binding to a cell surface structure and contribute to a polyclonal stimulation of the T cells. The terms "stimulatory agent" and "polyclonal stimulatory agent" as used herein have an interchangeable meaning. Examples of suitable agents for use in the present invention include agents such as synthesized compounds, nucleic acids and proteins, including polyclonal or monoclonal antibodies, and fragments or derivatives thereof, and bioengineered proteins, such as fusion proteins. In one example, the agents are mitogenic proteins. Mito genie proteins are two or more proteins that are able to deliver the requisite minimum of two signals to T-cells in order to cause the T-cells to become activated. Examples of mitogenic proteins are anti-CD3 and anti-CD2 monoclonal antibodies (mAb) in combination with a co-stimulatory protein such as and including proteins specific for one or more of the following T-cell surface molecules: CD28, CD5, CD4, CD8, MHCI, MHCII, CTLA-4, ICOS, PD-1, OX40, CD27L (CD70), 4-1BBL, CD30L and LIGHT, including the corresponding ligands to these surface structures, or fragments thereof. Other suitable agents include agents capable of delivering a signal to T-cells through cytokine receptors such as IL-2R, IL-12R, IL-1R, IL-15R; IFN-gammaR, TNF-alphaR, IL-4R, and IL- 10R, including monoclonal antibodies (mAbs) to these receptors, fusion proteins with a reactive end specific for these receptors and the corresponding ligands to these receptors or fractions thereof. Other suitable agents include any agent capable of binding to cellular adhesion molecules on T-cells such as mAbs, fusion proteins and the corresponding ligands or fractions thereof to adhesion molecules in the following categories: cadherins, ICAM, integrins, and selectins. Examples of adhesion molecules on T-cells are: CD44, CD31, CD18/CDl la (LFA-1), CD29, CD54 (ICAM-1), CD62L (L-selectin), and CD29/CD49d (VLA-4). Other suitable agents include any agents capable of binding to chemokine receptors, including those in the C-C and C-X-C categories. Examples of chemokine receptors associated with T-cell function include CCR1, CCR2, CCR3, CCR4, CCR5, and CXCR3.
An agent may be attached or coupled to the mobile matrix by a variety of methods known and available in the art. The attachment may be covalent or noncovalent, electrostatic, or hydrophobic and may be accomplished by a variety of attachment means, including for example, chemical, mechanical, enzymatic, or other means whereby an agent is capable of stimulating the cells. For example, the antibody to a cell surface structure first may be attached to the matrix, or avidin or strep tavidin may be attached to the matrix for binding to a biotinylated agent. The antibody to the cell surface structure may be attached to the matrix directly or indirectly, e.g. via an anti-isotype antibody. Another example includes using protein A or protein G, or other non-specific antibody binding molecules, attached to matrices to bind an antibody. Alternatively, the agent may be attached to the matrix by chemical means, such as cross-linking to the matrix.
As used herein, the term "antibody" is intended to include polyclonal and monoclonal antibodies, chimeric antibodies, haptens and antibody fragments, and molecules which are antibody equivalents in that they specifically bind to an epitope on the antigen. The term "antibody" includes polyclonal and monoclonal antibodies of any isotype (IgA, IgG, IgE, IgD, IgM), or an antigen-binding portion thereof, including, but not limited to, F(ab) and Fv fragments such as sc Fv, single chain antibodies, chimeric antibodies, humanized antibodies, and a Fab expression library.
The term "biologically inert" as used herein refers to the properties of the nanomatrix, that it is non-toxic to living cells and does not induce strong alterations of the cell function via physical interaction with the cell surface, due to its small size, except the specific ligand/receptor triggering function of the attached ligands or antibodies. The nanomatrices, in addition, may be biodegradable, e.g. degraded by enzymatic activity or cleared by phagocytic cells. The biodegradable material can be derived from natural or synthetic materials that degrade in biological fluids, e.g. cell culture media and blood. The degradation may occur using enzymatic means or may occur without enzymatic means. The biodegradable material degrades within days, weeks or few months, which may depend on the environmental conditions it is exposed to. The biodegradable material should be non-toxic and non-antigenic for living cells and in humans. The degradation products must produce non-toxic by-products. An important aspect in the context of being biologically inert is the fact that the nanomatrix does not induce strong alteration in structure, function, activity status or viability of labelled cells, i.e. it does not cause perturbance of the cells and does not interfere with subsequent experiments and therapeutic applications of the stimulated cells. The mechanical or chemical irritation of the cell is decreased due to the properties of the nanomatrix of being very small, i.e. nano-scale range, and having a mobile matrix which rather snuggles to the cell surface than altering the shape of the cell surface or exerting strong shearing force to the cells, e.g. resulting in membrane rupture.
The term "PBMC" as used herein refers to peripheral blood mononuclear cells isolated from human peripheral blood preparations e.g. by use of a density gradient (e.g. Ficoll, PanColl). "PBMC" consist of lymphocytes and monocytes.
The term "purified T cells" as used herein refers to cell populations with an increased content of T cells. T cells may be purified by different methods, including FACS® sorting, enrichment by magnetic beads (e.g. CD4 and CD8 MicroBeads, Miltenyi Biotec), magnetic bead depletion of non-T cells, Ficoll based methods (e.g. RosetteSep®, Stem Cell Technologies) or whole blood isolation methods (MACSxpress®, Miltenyi Biotec), as well as panning on antibody coated surfaces.
Embodiments
In one embodiment of the present invention a first nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto one agent, e.g. anti CD3 mAb. A second nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto another agent, e.g. anti CD28mAb. In this case the nanomatrix of the present invention is a nanomatrix wherein at least one first agent and one second agent are attached to separate mobile matrices.
A mixture of these nanomatrices is contacted with T cells, thereby activating and inducing the T cells to proliferate.
Fine-tuning of nanomatrices for the stimulation of the T cells is easily performed due to the high ratio of nanomatrices to cells (normally larger than 500: 1).
In another embodiment of the present invention a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto one agent, e.g. anti CD3 mAb. In this case the nanomatrix of the present invention is a nanomatrix wherein at least one first agent is attached to mobile matrices. This nanomatrix is contacted with T cells, thereby activating and inducing the T cells to proliferate.
A second or more (multiple) co- stimulating agents, e.g. anti CD28mAb, may be added as soluble agents to optimize or support the activation induced by the nanomatrix with the first agent attached thereto.
In another embodiment of the present invention a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto two agents which provide activation signals to the cell, e.g. anti CD3mAb and anti CD28 mAb. In this case the nanomatrix of the present invention is a nanomatrix wherein at least one first agent and one second agent are attached to the same flexible matrix.
Nanomatrices of this kind are contacted with T cells, thereby activating and inducing the T cells to proliferate. In another embodiment of the present invention a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto multiple agents which provide activation signals to the cell, e.g. anti-CD3mAb and anti-CD28 mAb, anti ICOS, anti-CD137 or other known co- stimulatory molecules. In this case the nanomatrix of the present invention is a nanomatrix wherein at least one first agent and multiple other agents are attached to the same mobile matrix.
Nanomatrices of this kind are contacted with T cells, thereby activating and inducing the T cells to proliferate. In another embodiment of the present invention a nanomatrix of 1 to 500 nm, preferentially 10 to 200 nm in size consists of a mobile matrix of a polymer of dextran and has attached thereto one or more agents which provide activation signals to the cells, e.g. anti CD3mAb and/or anti CD28 mAb. In addition the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the polymer.
The nanomatrix is contacted with T cells, thereby activating and inducing the T cells to proliferate. Optionally, after stimulating of T cells the unbound magnetic, paramagnetic or superparamagnetic nanomatrix may be removed by applying a magnetic field gradient. Alternatively, the cells labelled with the magnetic nanomatrices may be separated by applying a magnetic field gradient, in particular a high-gradient magnetic field, and subsequent expansion of the purified T cells.
Although there is no need to remove the nanomatrix after activation and proliferation of the population of T cells due to their property of being biologically inert with regard to alteration of the cell function one might optionally remove the nanomatrix with mild washing conditions, which are sufficient to wash way the nanomatrices from the cells or cell culture. The nanomatrices can easily be diluted by repeated washing steps to effective concentrations below the T cell activation threshold. This optionally removing step is much easier performed with the nanomatrices than with beads or microspheres well known in the state of the art due to their small size. If the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the polymer than optionally the removal step can be performed by applying a magnetic field gradient to the cell/nanomatrix mixture.
The method of the invention can be used to expand selected T cell populations for use in treating an infectious disease or cancer. The resulting T cell population can be genetically transduced and used for immunotherapy or can be used for in vitro analysis of infectious agents such as HIV. Proliferation of a population of CD4+ cells obtained from an individual or patient, e.g. infected with HIV, can be achieved and the cells rendered resistant, e.g. to HIV infection. Following expansion of the T cell population to sufficient numbers, the expanded T cells are re-infused into the individual or patient. Similarly, a population of tumor-infiltrating lymphocytes can be obtained from an individual afflicted with cancer and the T cells stimulated to proliferate to sufficient numbers and restored to the individual. In addition, supernatants from cultures of T cells expanded in accordance with the method of the invention are a rich source of cytokines and can be used to sustain T cells in vivo or ex vivo. In another embodiment of the present invention a nanomatrix as described in any proceeding embodiment may be used in a closed cell culture system, e.g. the sample processing system of WO2009072003 or cell cultivation bags (Miltenyi Biotec, Baxter, CellGenics), G-Rex devices (Wilson Wolf manufacturing), WAVE Bioreactors (GE Healthcare), Quantum Cell Expansion System (Terumo BCT). The nanomatrices have optimal connectivity to such a closed cell culture system, they can be easily sterile filtrated and integrated into the closed cell culture system. They ease the processes of the closed cell culture system, i.e. stimulation of the T cells or other target cells) because no removal of the nanomatrices after the stimulation (and expanding) process is necessary as described herein. The nanomatrix can be added in relation to the volume of the culture rather than the T cell number. Culture volume can be assesses by a balance or a camera system (e.g. in the sample processing system of WO2009072003).The use of the method of the present invention within a closed cell culture system such as the sample processing system of WO2009072003 results in a safe and easy way to produce a pharmaceutical composition of stimulated T cells due to the reduced risk of e.g. contaminating agents such as other eukaryotic cells, bacteria or viruses (safer and faster handling by the operator).
In another embodiment of the present invention cultivation of T cells by use of the nanomatrix is performed within a magnetic field. In this embodiment the nanomatrix consists of superparamagnetic cores embedded into the mobile matrix. When applied to a magnetic field magnetic forces are induced in the nanomatrix and ligands triggering activating T cell receptors can be concentrated for improved induction of proliferation. Cultivation of T cells within a magnetic field can be performed in a column consisting of a ferromagnetic matrix (e.g. of WO2009072003) or in bag, flask or chamber in close proximity to strong permanent magnet.
The present invention has broad applicability to any cell type having a cell surface moiety that may be stimulated. In this regard, many cell signalling events can be enhanced by the method of the present invention. Such methodologies can be used therapeutically in an ex vivo setting to activate and stimulate cells for infusion into a patient or could be used in vivo, to induce cell signalling events on a target cell population. Preferentially the target cells of the method are T cells, but are in no way limited thereto. Prior to stimulation of T cells by the present invention the T cells may be directly identified and/or separated or isolated from blood, peripheral mononuclear blood cells (PBMC), body tissue or cells from tissue fluids. The cells are normally identified and/or separated from cell samples from mammals such as humans, mouse, or rat, but especially from humans and preferably from test subject and/or patients. The separation is performed by well known sorting methods in the art. This includes for example affinity chromatography or any other antibody-dependent separation technique known in the art. Any ligand-dependent separation technique known in the art may be used in conjunction with both positive and negative separation techniques that rely on the physical properties of the cells. An especially potent sorting technology is magnetic cell sorting. Methods to separate cells magnetically are commercially available e.g. from Invitrogen, Stemcell Technologies, Cellpro, Advanced Magnetics, or Miltenyi Biotec. In addition to mixtures of T cells with other cells, such as monocytes, macrophages, dendritic cells, B cells or other cells which are part of hematologic cell samples, such as blood or leukapheresis, highly purified T cell populations can be used for contacting with the presented invention, including T cell subpopulations, such as CD4+ T cells, CD8+ T cells, NKT cells, γ/δ T cells, α β T cells, CD4+CD25+Foxp3+ regulatory T cells, naive T cells (CD45RA+CCR7+ and/or CD62L+) or central memory T cells (CD45R0+CCR7+), effector memory T cells (CD45R0+CCR7-) or terminal effector T cells (CD45RA+CCR7-). Nanomatrices provide sufficient crosslinking activity to the T cell receptor, therefore additional crosslinking, e.g. via Fc-receptor expressing cells such as monocytes or dendritic cells is not required for activation.
Target cell populations, such as the T cell populations obtained via the present disclosure may be administered either alone, or as a pharmaceutical composition in combination with diluents and/or with other components such as IL-2 or other cytokines or cell populations. Briefly, pharmaceutical compositions of the present disclosure may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. A pharmaceutical composition may comprise a) a population of T cells, wherein said T cells are proliferated to therapeutically effective amounts according to the present invention; and b) one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such a composition may contain traces of nanomatrices which are biologically inert with regard to alteration of the cell function but may be biodegradable and which are non-toxic and non-antigenic to humans. Compositions of the present disclosure are preferably formulated for intravenous administration. Pharmaceutical compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
Examples
Example 1 : Preparation of nanomatrices
Magnetic nanomatrices were produced by a modification of the procedure of Molday and MacKenzie. Ten grams of Dextran T40 (Pharmacia Uppsala, Sweden), 1.5 g FeCl3 · 6 H20 and 0.64 g FeCl2 · 4 H20 are dissolved in 20 ml H20, and heated to 40°C. While stirring, 10 ml 4N NaOH are added slowly and the solution is heated to 70°C for 5 min. The particle suspension is neutralized with acetic acid. To remove aggregates the suspension is centrifuged for 10 min at 2,000 g and filtrated through a 0.22 μιη pore-size filter (Millex GV, Millipore, Molsheim, France). Unbound Dextran is removed by washing in a high-gradient magnetic field (HGMF). HGMF washing of magnetic nanomatrices is performed in steelwool columns made as described below and placed in a magnetic field of approx. 0.6 Tesla (MACS permanent magnet, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany). Ten milliliters of nanomatrix suspension are applied to a 15 x 40 mm column of 2 g steelwool. The loaded column is washed with 30 ml 0.05 M sodium acetate. After removing the column from the external magnetic field, the magnetic nanomatrices are eluted with 0.05 M sodium acetate. The nanomatrices form a brown suspension. The relative particle concentration is given as optical density at 450 nm. The size of the nanomatrices was determined by electron microscopy and dynamic light scattering to be 30 + 20nm (e.m.) and 65 + 20 nm (DLS). Dextran content and magnetite content of the matrix within the colloidal solution were determined to be in the range of about 3.5 mg/ml and 4.8 mg/ml, respectively, resulting approximately in a 40:60 w/w ratio. Based on the density of the dried nanomatrix of 2,5g/mL determined by pycnometer and the known densities of dextran and magnetite of 1.6 g/ml and 5.2 g/ml, respectively, the volume of dextran can be calculated to be about 70% for the dried nanomatrix. The nanomatrices show superparamagnetic behavior, as determined by susceptibility measurements. The size of the trapped ferrit microcrystals was determined from magnetic measurements to be approximately 10 nm.
CD3 antibodies (clone OKT3) and CD28 antibodies (clone 15E8) (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) were conjugated to the same or separate nanomatrices by standard bioconjugation chemistry (Bioconjugate Techniques, 2nd Edition, By Greg T. Hermanson, Published by Academic Press, Inc., 2008).
Example 2: Expansion of T cells using nanomatrices at various CD3/CD28 concentrations and ratios versus CD3/CD28 MACSiBeads
The current state-of-the-art reagents for activation of highly purified T cells comprise activating antibodies against CD3/CD28 immobilized either on the surfaces of a cell culture dish or large cell-sized (4-5 μιη) particles. Both techniques are error prone and technically difficult to realize and standardize, especially under GMP-compatible production conditions. In contrast nanomatrices can be easily prepared and conveniently be used for cell culture under GMP-conditions. Therefore we compared the T cell activation potential by analysing the expansion potential of the CD3/CD28 coated nanomatrices at various concentrations and CD3/CD28 ratios with commercially available cell stimulation beads (MACSiBeads, ø 4,5μιη, Miltenyi Biotec GmbH). As can be seen in FIG 1 the nanomatrices expand T cells efficiently even at very low CD3 concentrations (20 - 100 ng/ml) which are also typically used for soluble CD3/CD28 in the presence of accessory cells which provide crosslinking. Besides the antibody concentration the CD3/CD28 ratio can also influence the cell activation and provides an additional means to optimize the T cell culture. The expansion at optimal doses (20 - 300 ng/ml) was similar or better than the standard reagent (MACSiBeads). At higher doses the expansion was reduced due to overstimulation of the T cells (activation- induced cell death), a phenomenon known to occur at a too high degree of TCR stimulation. Taken together, these results show that CD3/CD28 coated nanomatrices can efficiently activate and expand T cells at very low antibody concentrations and without the need for additional crosslinking.
Example 3: Comparison CD3/CD28 conjugated to nanomatrices versus 200 nm and 300 nm solid particles
As outlined above currently available reagents for activation of T cells can be split into two groups. Soluble antibodies stimulating, e.g. against CD3 and CD28, require immobilisation either on a surface of the cell culture dish or via receptors an accessory cells, e.g. Immunoglobulin Fc-receptors. Reagents which do not depend on an extra crosslinking step to be used for T cell activation, e.g. to stimulate highly purified T cells in the absence of accessory cells, are based on cell-sized particles (ø 4-5 μιη) coated with stimulating CD3 and CD28 antibodies. It is known that solid particles below a critical diameter of about 1 μιη are not suitable to properly expand T cells. To show the unique activating capacity of the CD3/CD28 coated nanomatrices (ø 50-200 nm), we compared their activating capacity with solid particles of similar size (200 nm, and 300 nm) versus cell sized particles (ø 4,5 μιη). Since small solid particles do not usually lead to expansion of T cells we analysed early T cell activation markers (CD25 up-regulation and loss of CD3 expression) to have a sensitive screen for T cell activation. CD25 is up-regulated within the first 24-48 hours following T cell stimulation. Because the TCR induced CD25 up-regulation is further supported by IL-2, we also added IL-2 to the culture conditions to maximize the sensitivity of the assay. Another direct result of TCR stimulation is the downregulation of the T cell receptor, which can be analysed via loss of CD3 expression on the cell surface. Highly purified T cells were cultured with CD3/CD28 coated nanomatrices (100 ng/ml CD3) or solid particles with a diameter of 4.5 μιη (MACSiBeads) or 200 nm (Ademtech beads) both covalently coated with CD3 and CD28 antibodies. MACSiBeads were used at an optimal 1: 1 ratio whereas 200 nm particles were titrated to achieve an active dose of CD3 and CD28 ranging from 25-3000 ng/ml CD3. On day 3 and 5 the frequency of CD25+ T cells and on day 3 the expression intensity of CD3 was measured.
As can be seen in FIG 2A, B and C nanomatrices lead to strong activation at the optimal dose (100 ng/ml) as shown by up-regulation of CD25 [FIG 2B (day 3), FIG 2C (day5)] and downregulation of CD3 (FIG 2A) which occurred at similar levels like with the cell-sized MACSiBeads. In sharp contrast no CD25 up-regulation and almost no CD3 down-regulation was seen for 200 nm solid particles even at 30 fold higher CD3/CD28 concentrations. Even on day five 200 nm solid particles were not able to induce CD25 expression to a similar level like the nanomatrix. Only at high concentrations (5-30 fold higher than for the nanomatrix) there was a slight upregulation observed achieving about 50-70% of the levels of the nanomatrix.
These data show that despite their small size mobile nanomatrices indeed have a unique potential to activate T cells when compared to similarly sized particles with a solid surface. The titration experiment also shows that the lack of activation by CD3/CD28 coated 200-300 nm-sized solid particles cannot simply be compensated by higher doses of particles but obviously there is a different quality of activation signal induced by the nanomatrix.
Example 4: expansion of purified T cell subsets
As indicated above various T cell subsets can have different activation requirements. In particular naive T cells are difficult to activate in the absence of accessory cells. Furthermore CD4 and CD8 T cells may have different needs when activated alone or in presence of additional cell types. To show that all T cell subsets can be expanded equally well by nanomatrices, we activated purified CD4 and CD8 naive T cells, total naive T cells or total T cells with either nanomatrices at the indicated dose and composition or MACSiBeads and compared their expansion. As shown in FIG 3 all subsets can be efficiently expanded by nanomatrices and at comparable level to the standard MACSiBead culture.
Example 5: Expansion of CD25+Foxp3+ regulatory T cells (Treg)
Treg are of particular interest for therapeutic applications for transplantation, autoimmunity and chronic inflammation and Treg are difficult to expand in vitro without loss of regulatory activity, i.e. Foxp3 expression. Therefore we also analysed whether CD25 selected Treg cells (Foxp3 purity typically 60-90%) from various donors can be expanded using nanomatrices. To support growth of Treg versus conventional T cells the expansion was done in the presence of 100 nM Rapamycine, a well described drug inhibiting conventional T cell growth. As shown in FIG 4 following 14 days of culture Treg can be expanded 10-20 times (w/o Rapa) or 5-10 times (with Rapa). As described before without Rapa the Foxp3 purity was highly variable (10-75%) whereas in the presence of Rapa the purity was always >50%.
Taken together these results show that nanomatrices can even be used to activate and expand Treg in culture.
Example 6: Comparison of T cell activation by CD3/CD28-conjugated to the same nanomatrix versus CD3 and CD28 conjugated to separate nanomatrices.
It is described in various applications of CD3 and CD28 based T cell activation reagents that both antibodies have to be immobilized onto the same surface for optimal activation. Therefore we also tested whether this is also required for CD3 and CD28 conjugated to nanomatices. We compared expansion of purified naive T cells activated by a CD3/CD28 nanomatrix versus CD3 nanomatrix + CD28 nanomatrix mixed at different ratios/concentrations. Expansion (day 5) and cell division (day 7), as measured by Violetye dilution was analysed. As shown in FIG 5A, B the stimulation with the CD3 nanomatrix alone did not induce significant expansion and only few cell divisions can be observed, as it is expected for naive T cells which depend on a costimulatory signal. However addition of the CD28 nanomatrix, already at 10-50 ng/ml induced full cell dividing activity and also expansion of T cell numbers, which was similar to the CD3/CD28 control nanomatrix or the standard MACSiBeads. These data clearly show that both antibodies may be conjugated to separate nanomatrices, which can be mixed hereafter for optimised use. This facilitates the production process and quality control of the single nanomatrices and improves the flexibility of the reagent, e.g. facilitating the optimisation of the activation conditions for specialised T cell subsets by titrating various CD3 and CD28 concentrations and ratios (fine-tuning).
Example 7: The effect of conjugation of soluble CD3 or CD28 to the nanomatrix.
To rule out the possibility that similar results as with the CD3 and/or CD28 coated nanomatrix could be achieved by use of the respective soluble antibodies, we compared the stimulating effects of CD3 or CD28 coated the nanomatrix with soluble antibodies at various concentrations to demonstrate that indeed the conjugation of the antibodies to the matrix is the critical step to obtain good T cell activation. IL-2 was added to all cultures. As shown in FIG 6A a soluble CD3 alone did not induce any significant up-regulation of the early activation markers CD25 and CD69 in naive T cells over a wide concentration range (10-10000 ng/ml) whereas CD3 coated Nanomatrix (100 ng/ml CD3) induced CD25/CD69 expression in 20- 60% of the cells. In the presence of a saturating amount (200 ng/ml) of soluble CD28 as costimulator (FIG 6B) soluble CD3 also induced CD25/CD69 expression in about 20-40% of the cells at the highest tested doses (100-10000 ng/ml). In contrast the CD3 coated nanomatrix (100 ng/ml CD3) induced CD25/CD69 expression in 40-70% of the cells.
We also tested the effect of conjugation of CD28 antibodies to the nanomatrix. Since the effects of costimulation are best visualized under suboptimal CD3 stimulation, we titrated CD28 either soluble or conjugated to the nanomatrix in the presence of soluble CD3 to a culture of naive T cells. As shown in FIG 6C soluble CD3 alone similar to the induction of CD25/CD69 as shown above did not induce any expansion of the naive T cells. In the presence of soluble CD28 however a 2-6 fold expansion was detectable but only at the highest tested dose of CD28 (10000 ng/ml). In contrast to this, CD28 conjugated to nanomatrix induced a similar degree of expansion already at a 1000 fold lower concentration (10 ng/ml). These data show again the strong crosslinking and T cell activating capacity of nanomatrix versus soluble antibodies which explains why CD3CD28 conjugated nanomatrices in contrast to soluble antibodies can be used to activate and expand even naive human T cells in vitro.
Example 8: Nanomatrices can be used to activate T cells for introduction of TCR genes by viral transduction
One important application for activating and expanding T cells and in particular purified cell subsets is their genetic manipulation, e.g. to introduce a certain antigen receptor with specificity for tumor antigens. We have used nanomatrices to activate purified naive (TN, CD62L+ CD45RA+), central memory (TCM, CD62L+CD45RA-) and effector (TEM, CD62L- CD45RA-) T cells and transduced them using a retroviral vector expressing a TCR specific for MART-1, a tumor antigen. To test the relative frequency of transgene expression in these T cell subsets we performed MHC-peptide Class I tetramer staining. All T cell subsets are efficiently transduced (>50%) independent on the stimulatory conditions we tested (FIG.7). We also compared the in vitro expansion of the transduced T cells. As shown in FIG 8 after 10 days we observed no differences with regard to expansion of the three subsets under all conditions. All activation regimens for the isolated T cell subsets were equal or better to the „standard" stimulation of total PBMC with soluble CD3 (all values were normalized to this standard to allow better comparison between different donors). We observed a trend (not statistically significant) for better expansion when T cell subsets are stimulated with MACSiBeads or nanomatrices when compared to coated ccCD3+CcCD28. We further investigated the functional activity of the introduced MART-1 TCR and the differentiation status of transduced cells looking at surface markers and cytokine production upon re- stimulation with a MART-1+HLA-A2+ tumor cell line. As shown in FIG 9 nanomatrix- and MACSiBead- stimulated TCM and TN cells seem to have a higher expression of CD62L and CCR7, two molecules facilitating migration of the T cells into peripheral lymph nodes. This capacity is regarded as beneficial to promote long term persistence and functional activity of transferred T cells in vivo and thus is thought to increase therapeutic efficacy. The percentage of MART-1 reactive IFNy+ cells tend to be higher in TCM and TEM CD8+ T cell subsets compare to TN in all stimulatory conditions but this was not statistically significant (FIG 10 top panel). Focussing on the IL-2 production (FIG 10 middle panel) we observed that a higher percentage of T cells produces IL-2 when they have been stimulated with MACSiBeads/nanomatrices when compared to coated ccCD3+aCD28 stimulation. The same is true for TNFcc producing cells detected in TN subset when stimulated with MACSiBeads (FIG 10 bottom panel). These results indicate cells of T derived cells stimulated with beads showed diminished effector cell differentiation, suggesting less progress toward terminal differentiation.
Taken together, the results indicate that CD3/CD28 nanomatrices can be used to efficiently activate and transduce purified T cell subsets to generate fully functional T cell transplants, e.g. for tumor therapy.

Claims

Claims
1) The use of a nanomatrix for in- vitro polyclonal stimulation of T cells, the nanomatrix comprising
a) a matrix of mobile polymer chains, and
b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells;
wherein the nanomatrix is 1 to 500 nm in size.
2) An in vitro method for polyclonal stimulation of T cells, the method comprising contacting a population of T cells with a nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains, and
b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells; thereby activating and inducing the T cells to proliferate;
wherein the nanomatrix is 1 to 500 nm in size.
3) The method according to claim 2, wherein at least one first and one second stimulatory agents are attached to the same matrix of mobile polymer chains. 4) The method according to claim 2, wherein at least one first and one second stimulatory agents are attached to separate matrices of mobile polymer chains.
5) The method according to claim 4, wherein the ratio of nanomatrices to cells is larger than 500: 1 allowing fine-tuning of T cell stimulation.
6) The method according to any one of claims 2 to 5, wherein one stimulatory agent is an anti-CD3 antibody or fragment thereof.
7) The method according to any one of claims 1 to 6, wherein the second stimulatory agent is an anti-CD28 antibody.
8) The method according to any one of claims 2 to 7, wherein the matrix of mobile polymer chains consists of a polymer of dextran. 9) The method according to any one of claims 2 to 8, wherein the nanomatrix carries magnetic, paramagnetic or superparamagnetic nano-crystals, embedded into the matrix of mobile polymer chains. 10) The method according to any one of claims 2 to 9, wherein the stimulatory agent is attached at high density with more than 25 μg per mg nanomatrix.
11) The method according to any one of claims 2 to 10, wherein the stimulated T cells are Treg cells.
12) A method for polyclonal stimulation of T cells in a closed cell culture system, the method comprising contacting a cell culture comprising a population of T cells within said closed cell culture system with a dosage of nanomatrix, the nanomatrix comprising
a) a matrix of mobile polymer chains; and
b) attached to said matrix of mobile polymer chains one or more stimulatory agents which provide activation signals to the T cells;
wherein the nanomatrix is 1 to 500 nm in size, and
wherein said dosage of nanomatrix is applied sterile to said closed cell culture system, and wherein said dosage depends on the volume of the cell culture in said closed cell culture system.
13) A method according to claim 12, wherein said volume of the cell culture can be determined by a balance or a camera system without affecting the sterility barrier. 14) A method according to claims 12 and 13, wherein determining and applying said dosage are performed automatically.
15) A pharmaceutical composition comprising a population of stimulated T cells produced according to the method of claims 12-14.
PCT/EP2013/069854 2012-09-25 2013-09-24 Method for polyclonal stimulation of t cells by mobile nanomatrices WO2014048920A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2015532453A JP6216793B2 (en) 2012-09-25 2013-09-24 Polyclonal stimulation of T cells using mobile nanomatrix
CA2878997A CA2878997C (en) 2012-09-25 2013-09-24 Method for polyclonal stimulation of t cells by mobile nanomatrices
EP13766319.1A EP2900809B1 (en) 2012-09-25 2013-09-24 Method for polyclonal stimulation of t cells by mobile nanomatrices
CN201380045752.3A CN104619831A (en) 2012-09-25 2013-09-24 Method for polyclonal stimulation of t cells by mobile nanomatrices
DK13766319.1T DK2900809T3 (en) 2012-09-25 2013-09-24 PROCEDURE FOR POLYCLONAL STIMULATION OF T-CELLS USING MOBILE NANOMATRICES
ES13766319.1T ES2632450T3 (en) 2012-09-25 2013-09-24 Method for polyclonal stimulation of T cells by mobile nanomatrices
US14/430,875 US20150240204A1 (en) 2012-09-25 2013-09-25 Method for polyclonal stimulation of t cells by mobile nanomatrices

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12185939.1 2012-09-25
EP12185939.1A EP2711418B1 (en) 2012-09-25 2012-09-25 Method for polyclonal stimulation of T cells by flexible nanomatrices

Publications (1)

Publication Number Publication Date
WO2014048920A1 true WO2014048920A1 (en) 2014-04-03

Family

ID=46940385

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/069854 WO2014048920A1 (en) 2012-09-25 2013-09-24 Method for polyclonal stimulation of t cells by mobile nanomatrices

Country Status (9)

Country Link
US (2) US10513687B2 (en)
EP (2) EP2711418B1 (en)
JP (1) JP6216793B2 (en)
CN (1) CN104619831A (en)
CA (1) CA2878997C (en)
DK (1) DK2900809T3 (en)
ES (1) ES2632450T3 (en)
PT (1) PT2900809T (en)
WO (1) WO2014048920A1 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015162211A1 (en) 2014-04-24 2015-10-29 Miltenyi Biotec Gmbh Method for automated generation of genetically modified t cells
EP3091032A1 (en) 2015-05-08 2016-11-09 Miltenyi Biotec GmbH Humanized antibody or fragment thereof specific for cd3
WO2017035577A1 (en) * 2015-08-28 2017-03-09 Ctm@Crc Ltd. Products and methods for activating and/or expanding t cells
EP3315511A1 (en) 2016-10-29 2018-05-02 Miltenyi Biotec GmbH Adapter chimeric antigen receptor expressing cells for targeting of multiple antigens
EP3336107A1 (en) 2016-12-15 2018-06-20 Miltenyi Biotec GmbH Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
WO2018158775A1 (en) 2017-03-02 2018-09-07 Yeda Research And Development Co. Ltd. Methods of culturing t cells and uses of same
EP3415617A1 (en) * 2017-06-16 2018-12-19 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. A method and a device for increasing ex vivo expansion of t cells by using adhesive nanostructured surfaces and costimulatory signals
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3134095B1 (en) 2014-04-25 2020-04-22 Bluebird Bio, Inc. Improved methods for manufacturing adoptive cell therapies
WO2020239866A1 (en) 2019-05-28 2020-12-03 Miltenyi Biotec B.V. & Co. KG Method for generation of genetically modified t cells
WO2021046134A1 (en) * 2019-09-03 2021-03-11 Allogene Therapeutics, Inc. Methods of preparing t cells for t cell therapy
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021164959A1 (en) * 2020-02-17 2021-08-26 Miltenyi Biotec B.V. & Co. KG Method for providing personalized cells with chimeric antigen receptors (car) against tumor microenvironment cells
WO2021173964A1 (en) * 2020-02-28 2021-09-02 KSQ Therapeutics, Inc. Methods for activation and expansion of tumor infiltrating lymphocytes
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11248238B2 (en) 2015-10-22 2022-02-15 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
US11351236B2 (en) 2014-12-12 2022-06-07 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US11466253B2 (en) 2015-10-22 2022-10-11 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2022215718A1 (en) 2021-04-08 2022-10-13 武田薬品工業株式会社 Method for activating t-cells
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
US11560547B2 (en) 2014-06-06 2023-01-24 2Seventy Bio, Inc. Methods of making T cell compositions
WO2023021477A1 (en) 2021-08-20 2023-02-23 Novartis Ag Methods of making chimeric antigen receptor–expressing cells
WO2023079524A2 (en) 2021-11-08 2023-05-11 Life Technologies As Polymer-interaction molecule conjugates and methods of use
US11866465B2 (en) 2017-04-27 2024-01-09 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
US11913024B2 (en) 2015-10-22 2024-02-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2024068760A1 (en) 2022-09-27 2024-04-04 King's College London Compositions comprising nkg2d, cxcr2, and dap10/dap12 fusion polypeptides and methods of use thereof
US12006369B2 (en) 2019-07-18 2024-06-11 2Seventy Bio, Inc. BCMA chimeric antigen receptors

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2612918T3 (en) * 2013-07-10 2017-05-19 Miltenyi Biotec Gmbh Method to induce the proliferation of natural killer cells by mobile nanomatrices
EP3998278A1 (en) 2014-04-25 2022-05-18 2seventy bio, Inc. Mnd promoter chimeric antigen receptors
JP6588084B2 (en) 2014-08-19 2019-10-09 ミルテニイ バイオテック ゲゼルシャフト ミット ベシュレンクテル ハフツング Chimeric antigen receptor specific for SSEA4 antigen
WO2016033690A1 (en) 2014-09-04 2016-03-10 Stemcell Technologies Inc. Soluble antibody complexes for t cell or nk cell activation and expansion
US10201606B2 (en) 2014-11-26 2019-02-12 Miltenyi Biotec Gmbh Combination immunotherapy of antigen-recognizing receptors and hematopoietic cells for the treatment of diseases
EP3184548A1 (en) 2015-12-23 2017-06-28 Miltenyi Biotec GmbH Chimeric antigen receptor with cytokine receptor activating or blocking domain
US10294454B2 (en) 2016-08-24 2019-05-21 General Electric Company Methods and kits for cell activation
US10617720B2 (en) 2016-10-20 2020-04-14 Miltenyi Biotech, GmbH Chimeric antigen receptor specific for tumor cells
TWI699218B (en) 2017-12-22 2020-07-21 財團法人工業技術研究院 Method for in vitro activation and/or expansion of immune cells
EP3735425A1 (en) 2018-01-04 2020-11-11 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor specific for bdca2 antigen
AU2019226392A1 (en) * 2018-03-01 2020-09-24 Fundación Para La Investigación Biomédica Del Hospital Gregorio Marañón Method for obtaining regulatory T cells derived from thymic tissue and use of said cells as cell immunotherapy in immune system disorders
AU2019323839A1 (en) * 2018-08-22 2021-03-11 Cellect Biotherapeutics Ltd. Modulation of apoptosis susceptible cells
WO2020080475A1 (en) 2018-10-18 2020-04-23 武田薬品工業株式会社 Method for activation/proliferation of t cells
WO2020126634A2 (en) * 2018-12-21 2020-06-25 Lavision Biotec Gmbh Light sheet microscope with movable container
AU2020392094A1 (en) * 2019-11-25 2022-06-23 KSQ Therapeutics, Inc. Methods for activation and expansion of tumor infiltrating lymphocytes
WO2022096425A2 (en) 2020-11-04 2022-05-12 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor comprising an antigen binding domain specific for msln having a specificity for tumor cells
CN112773905B (en) * 2021-02-08 2023-08-01 暨南大学 Macrophage knapsack system and preparation method and application thereof
WO2023147505A2 (en) * 2022-01-28 2023-08-03 Cellular Biomedicine Group, Inc. Method for enriching tumor infiltrating lymphocytes

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0125763B1 (en) 1983-03-18 1987-05-06 Fuji Photo Film Co., Ltd. Medium for electrophoresis
WO1994029436A1 (en) 1993-06-04 1994-12-22 The United States Of America Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of t cells
US6117982A (en) * 1991-04-19 2000-09-12 Tanox, Inc. Conjugates of microbeads and antibodies specific for T lymphocytes and their use as in vivo immune modulators
US20070086996A1 (en) * 2004-02-26 2007-04-19 Michael Har-Noy Biodegradable T-cell activation device and methods
EP1257632B1 (en) 2000-02-24 2007-09-12 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US20080317724A1 (en) 2007-01-18 2008-12-25 Lance Kam Micropatterned T cell stimulation
WO2009072003A2 (en) 2007-12-07 2009-06-11 Miltenyi Biotec Gmbh Sample processing system and methods
US20100151031A1 (en) * 2007-03-23 2010-06-17 Desimone Joseph M Discrete size and shape specific organic nanoparticles designed to elicit an immune response
US20100284965A1 (en) 2008-01-15 2010-11-11 Yale University Compositions and methods for adoptive and active immunotherapy
US20120121649A1 (en) 2010-11-12 2012-05-17 Uti Limited Partnership Compositions and methods for the prevention and treatment of cancer

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5872222A (en) * 1991-04-19 1999-02-16 Tanox Biosystems, Inc. Conjugates of polymers and antibodies specific for T lymphocytes, and their use as adjuvants
WO1994012196A1 (en) * 1992-11-25 1994-06-09 Tanox Biosystems, Inc. Conjugates and constructs including anti-cd28 and anti-cd3 binding molecules
CA2164646A1 (en) * 1993-06-11 1994-12-22 Wade E. Bolton Anti-cd3 antibody-aminodextran conjugates for induction of t-cell activation and proliferation
US6797514B2 (en) * 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
AU2003216436A1 (en) * 2002-02-08 2003-09-02 Life Technologies Corporation Compositions and methods for restoring immune responsiveness in patients with immunological defects
US8658178B2 (en) * 2008-03-19 2014-02-25 Yale University Carbon nanotube compositions and methods of use thereof
US9790467B2 (en) 2015-09-22 2017-10-17 Qt Holdings Corp Methods and compositions for activation or expansion of T lymphocytes

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0125763B1 (en) 1983-03-18 1987-05-06 Fuji Photo Film Co., Ltd. Medium for electrophoresis
US6117982A (en) * 1991-04-19 2000-09-12 Tanox, Inc. Conjugates of microbeads and antibodies specific for T lymphocytes and their use as in vivo immune modulators
WO1994029436A1 (en) 1993-06-04 1994-12-22 The United States Of America Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of t cells
EP1257632B1 (en) 2000-02-24 2007-09-12 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US20070086996A1 (en) * 2004-02-26 2007-04-19 Michael Har-Noy Biodegradable T-cell activation device and methods
US20090291498A1 (en) * 2004-02-26 2009-11-26 Immunovative Therapies Ltd. Biodegradable T-cell activation method
US8012750B2 (en) 2004-02-26 2011-09-06 Immunovative Therapies Ltd. T-cell activation device
US20080317724A1 (en) 2007-01-18 2008-12-25 Lance Kam Micropatterned T cell stimulation
US20100151031A1 (en) * 2007-03-23 2010-06-17 Desimone Joseph M Discrete size and shape specific organic nanoparticles designed to elicit an immune response
WO2009072003A2 (en) 2007-12-07 2009-06-11 Miltenyi Biotec Gmbh Sample processing system and methods
US20100284965A1 (en) 2008-01-15 2010-11-11 Yale University Compositions and methods for adoptive and active immunotherapy
US20120121649A1 (en) 2010-11-12 2012-05-17 Uti Limited Partnership Compositions and methods for the prevention and treatment of cancer

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
APEL ET AL.: "Miltenyi Biotec", CHEMIE INGENIEUR TECHNIK, vol. 85, 2013, pages 103 - 110
BAROJA ET AL., CELLULAR IMMUNOLOGY, vol. 120, 1989, pages 205 - 217
BERTHOLON ET AL., LANGMUIR, 2006, pages 45485 - 5490
DEMENTO S ET AL: "Biomimetic approaches to modulating the T cell immune response with nano- and micro- particles", PROCEEDINGS OF THE 31ST ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY: ENGINEERING THE FUTURE OF BIOMEDICINE, EMBC 2009, IEEE, 3 September 2009 (2009-09-03), pages 1161 - 1166, XP031881178, ISBN: 978-1-4244-3296-7, DOI: 10.1109/IEMBS.2009.5332625 *
DI MARCO ET AL., INT J. NANOMED., 2007, pages 618
DINAUER N ET AL: "Selective targeting of antibody-conjugated nanoparticles to leukemic cells and primary T-lymphocytes", BIOMATERIALS, ELSEVIER SCIENCE PUBLISHERS BV., BARKING, GB, vol. 26, no. 29, 1 October 2005 (2005-10-01), pages 5898 - 5906, XP027767546, ISSN: 0142-9612, [retrieved on 20051001] *
ERIN R STEENBLOCK; TAREK M FAHMY, MOLECULAR THERAPY, vol. 16, no. 4, April 2008 (2008-04-01), pages 765 - 772
FAHMY ET AL: "A nanoscopic multivalent antigen-presenting carrier for sensitive detection and drug delivery to T Cells", NANOMEDICINE: NANOTECHNOLOGY, BIOLOGY AND MEDICINE, ELSEVIER, NL, vol. 3, no. 1, 28 March 2007 (2007-03-28), pages 75 - 85, XP005928987, ISSN: 1549-9634, DOI: 10.1016/J.NANO.2006.11.010 *
GREG T. HERMANSON: "Bioconjugate Techniques, 2nd Edition,", 2008, ACADEMIC PRESS, INC.
MESCHER, MF, J IMMUNOL, vol. 149, 1992, pages 2402 - 2405
STEENBLOCK E R ET AL: "A comprehensive platform for ex vivo T-cell expansion based on biodegradable polymeric artificial antigen-presenting cells", MOLECULAR THERAPY, ACADEMIC PRESS, SAN DIEGO, CA, US, vol. 16, no. 4, 1 April 2008 (2008-04-01), pages 765 - 772, XP002551496, ISSN: 1525-0016, [retrieved on 20080304], DOI: 10.1038/MT.2008.11 *
WANG ET AL., EUR. RADIOL., 2001, pages 2323

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015162211A1 (en) 2014-04-24 2015-10-29 Miltenyi Biotec Gmbh Method for automated generation of genetically modified t cells
US10131876B2 (en) 2014-04-24 2018-11-20 Miltenyi Biotec Gmbh Method for automated generation of genetically modified T cells
EP3134095B1 (en) 2014-04-25 2020-04-22 Bluebird Bio, Inc. Improved methods for manufacturing adoptive cell therapies
US11560547B2 (en) 2014-06-06 2023-01-24 2Seventy Bio, Inc. Methods of making T cell compositions
US11351236B2 (en) 2014-12-12 2022-06-07 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US11382965B2 (en) 2014-12-12 2022-07-12 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US11633463B2 (en) 2014-12-12 2023-04-25 2Seventy Bio, Inc. BCMA chimeric antigen receptors
EP3091032A1 (en) 2015-05-08 2016-11-09 Miltenyi Biotec GmbH Humanized antibody or fragment thereof specific for cd3
CN107889492A (en) * 2015-05-08 2018-04-06 美天旎生物技术有限公司 To the specific humanized antibodies of CD3 or its fragment
CN107889492B (en) * 2015-05-08 2022-01-18 美天施生物科技有限两合公司 Humanized antibodies or fragments thereof specific for CD3
WO2017035577A1 (en) * 2015-08-28 2017-03-09 Ctm@Crc Ltd. Products and methods for activating and/or expanding t cells
US20180371413A1 (en) * 2015-08-28 2018-12-27 Ctm@Crc Ltd. Products and methods for activating and/or expanding t cells
US11466253B2 (en) 2015-10-22 2022-10-11 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
US11913024B2 (en) 2015-10-22 2024-02-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
US11248238B2 (en) 2015-10-22 2022-02-15 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
WO2018078066A1 (en) 2016-10-29 2018-05-03 Miltenyi Biotec Gmbh Adapter chimeric antigen receptor expressing cells for targeting of multiple antigens
EP3315511A1 (en) 2016-10-29 2018-05-02 Miltenyi Biotec GmbH Adapter chimeric antigen receptor expressing cells for targeting of multiple antigens
WO2018108766A1 (en) 2016-12-15 2018-06-21 Miltenyi Biotec Gmbh Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
EP3336107A1 (en) 2016-12-15 2018-06-20 Miltenyi Biotec GmbH Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
WO2018158775A1 (en) 2017-03-02 2018-09-07 Yeda Research And Development Co. Ltd. Methods of culturing t cells and uses of same
US11932871B2 (en) 2017-03-02 2024-03-19 Yeda Research And Development Co. Ltd. Methods of culturing t cells and uses of same
US11866465B2 (en) 2017-04-27 2024-01-09 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
EP3415617A1 (en) * 2017-06-16 2018-12-19 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. A method and a device for increasing ex vivo expansion of t cells by using adhesive nanostructured surfaces and costimulatory signals
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020239866A1 (en) 2019-05-28 2020-12-03 Miltenyi Biotec B.V. & Co. KG Method for generation of genetically modified t cells
US12006369B2 (en) 2019-07-18 2024-06-11 2Seventy Bio, Inc. BCMA chimeric antigen receptors
WO2021046134A1 (en) * 2019-09-03 2021-03-11 Allogene Therapeutics, Inc. Methods of preparing t cells for t cell therapy
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021164959A1 (en) * 2020-02-17 2021-08-26 Miltenyi Biotec B.V. & Co. KG Method for providing personalized cells with chimeric antigen receptors (car) against tumor microenvironment cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173964A1 (en) * 2020-02-28 2021-09-02 KSQ Therapeutics, Inc. Methods for activation and expansion of tumor infiltrating lymphocytes
WO2022215718A1 (en) 2021-04-08 2022-10-13 武田薬品工業株式会社 Method for activating t-cells
WO2023021477A1 (en) 2021-08-20 2023-02-23 Novartis Ag Methods of making chimeric antigen receptor–expressing cells
WO2023079524A2 (en) 2021-11-08 2023-05-11 Life Technologies As Polymer-interaction molecule conjugates and methods of use
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
WO2024068760A1 (en) 2022-09-27 2024-04-04 King's College London Compositions comprising nkg2d, cxcr2, and dap10/dap12 fusion polypeptides and methods of use thereof

Also Published As

Publication number Publication date
EP2900809B1 (en) 2017-05-31
EP2711418A1 (en) 2014-03-26
ES2632450T3 (en) 2017-09-13
EP2900809A1 (en) 2015-08-05
CA2878997A1 (en) 2014-04-03
US20150240204A1 (en) 2015-08-27
JP2015533493A (en) 2015-11-26
JP6216793B2 (en) 2017-10-18
PT2900809T (en) 2017-07-13
CN104619831A (en) 2015-05-13
EP2711418B1 (en) 2017-08-23
US10513687B2 (en) 2019-12-24
CA2878997C (en) 2019-06-11
DK2900809T3 (en) 2017-07-24
US20140087462A1 (en) 2014-03-27

Similar Documents

Publication Publication Date Title
CA2878997C (en) Method for polyclonal stimulation of t cells by mobile nanomatrices
JP7204719B2 (en) Reagents and methods for identifying, enriching and/or expanding antigen-specific T cells
US20210115401A1 (en) Methods and Materials for the Generation of Regulatory T Cells
EP2824112B1 (en) Method for inducing proliferation of Natural Killer cells by mobile nanomatrices
CN115305229A (en) Antigen presenting cell mimetic scaffolds and methods of making and using same
EP3368659A1 (en) Selective expansion of different subpopulations of t cells by the alteration of cell surfacing signals and signal ratio
WO2010099205A1 (en) Methods for treating progressive multifocal leukoencephalopathy (pml)
WO2019217661A1 (en) Compositions and systems for ex vivo cell modulation and methods of use thereof
US20180142209A1 (en) Natural killer cell priming composition
WO2023049619A1 (en) Beads for targeted signal delivery
AU2022378730A1 (en) Beads for targeted signal delivery

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: 13766319

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2878997

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015532453

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14430875

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2013766319

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2013766319

Country of ref document: EP