WO2016189104A1 - Nouveau procédé pour produire des lymphocytes t - Google Patents

Nouveau procédé pour produire des lymphocytes t Download PDF

Info

Publication number
WO2016189104A1
WO2016189104A1 PCT/EP2016/061941 EP2016061941W WO2016189104A1 WO 2016189104 A1 WO2016189104 A1 WO 2016189104A1 EP 2016061941 W EP2016061941 W EP 2016061941W WO 2016189104 A1 WO2016189104 A1 WO 2016189104A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
specific
seq
cell
melanoma
Prior art date
Application number
PCT/EP2016/061941
Other languages
English (en)
Inventor
Nathalie Labarriere
François LANG
Sylvain SIMON
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université de Nantes
Université d'Angers
Centre National De La Recherche Scientifique (Cnrs)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Université de Nantes, Université d'Angers, Centre National De La Recherche Scientifique (Cnrs) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2016189104A1 publication Critical patent/WO2016189104A1/fr

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
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • 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
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the invention relates to an in vitro or ex vivo method to produce high avidity T cells for one or several antigens wherein said T cell obtained from a subject are cultivated with said one or several specific antigens and an anti-PDl antibody.
  • PD-1 The first function assigned to PD-1 was its involvement in immunological peripheral tolerance, maintaining T cell homeostasis by the control of auto-reactive T cells [Nishimura H et al., 1999 and Nishimura H et al., 2001].
  • PD-1 is expressed on thymocytes and its interaction with its ligand PD-L1, also expressed in the thymus, modulates both positive and negative selection.
  • PD-1 is inducible on many immune cell types, such as T cells, natural killer T cells and B cells. It has two natural ligands: PD-L1, expressed on activated T cells, monocytes and dendritic cells, and PD-L2, whose expression is restricted to dendritic cells and macrophages.
  • PD-1 Ligation of PD-1 with one of its ligands results in dampening early TCR signaling, through the recruitment of the phosphatases SHP-1 and SHP-2, resulting in direct dephosphorylation of signaling intermediates. Besides its role in maintenance of physiologic self-tolerance, PD-1 is also implicated in the down-regulation of anti-tumor immunity. Indeed, PD-L1 is commonly expressed on a variety of solid tumors including melanomas [Dong H et al., 2002] contributing to immune escape and is often associated with poor prognosis [Zitvogel L et al., 2012].
  • melanoma infiltrating lymphocytes are often enriched in PD-1 expressing CD8 T cells, which are functionally impaired [Ahmadzadeh M et al., 2009].
  • blocking PD-1/PD-L1 pathway appears to be a promising strategy to increase the efficiency of anti-tumor T cell responses.
  • Several clinical trials using blocking anti-PD-1 antibody reported unparalleled effectiveness for cancer immunotherapy, including melanoma, in terms of clinical response rates [Hamid O et al., 2013; Topalian SL et al., 2012; Topalian SL et al., 2014 and Wolchok JD et al., 2013].
  • long-term tumor regression using this treatment cannot be achieved in most patients, and several issues require further improvements such as the therapy to combine with anti-PD-1 treatment and the characterization of biomarkers unequivocally associated with clinical benefit.
  • TIL tumor infiltrating lymphocytes
  • this approach is evolving towards an increased specificity of infused T cells that can be reached either with the use of genetically modified T cells, such as TCR or CAR-transduced T cells [Kalos M. et al, 2011; Morgan RA et al., 2006 and Robbins et al., 2011] or with enriched or cloned T cells specific for a given HLA-peptide complex [Hunder NN et al., 2008; Khammari A et al., 2009; Mackensen A et al, 2006; Meidenbauer N et al, 2003; Vignard V et al, 2005 and Yee C et al, 2002]. All these approaches could be further improved by the use of specific T cells with optimized functions, such as the avidity of infused T cells and the modulation of inhibitory receptors' expression.
  • the invention relates to an in vitro or ex vivo method to produce high avidity T cells for one or several antigens wherein said T cell obtained from a subject are cultivated with said one or several specific antigens and an anti-PDl antibody.
  • DETAILED DESCRIPTION OF THE INVENTION The invention relates to an in vitro or ex vivo method to produce high avidity T cells for one or several antigens wherein said T cell obtained from a subject are cultivated with said one or several specific antigens and an anti-PDl antibody.
  • the term "high avidity T cells for one or several antigens” denotes T cells which recognize a given specific antigen with an EC50 below 0.2 nM. T cells with high avidity are particularly sought and useful for immunotherapeutic strategies, because of their related strong reactivity against tumor cells expressing the target antigens.
  • the invention relates to an in vitro or ex vivo method to produce T cells which recognize one or several antigens wherein said T cells obtained from a subject are cultivated with said one or several antigens and an anti-PDl antibody.
  • the invention also relates to an in vitro or ex vivo method to produce high avidity and high lytic T cells for one or several antigens wherein said T cell obtained from a subject are cultivate with said one or several specific antigens and an anti-PDl antibody.
  • the T cells are obtained from Peripheral Blood Mononuclear Cell
  • PBMC derived from HLA-A2 donors or patients.
  • the T cells are T CD8 + cells.
  • the T cell obtained from a subject are cultivate with interleukin-2 (IL-2) and/or human serum.
  • IL-2 interleukin-2
  • an anti-PD-1 antibody is used to obtain high avidity T cells for one or several antigens.
  • Antibodies directed against the PD-1 protein can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • Various adjuvants known in the art can be used to enhance antibody production.
  • antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
  • Monoclonal antibodies against PD-1 protein can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture.
  • Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975); the human B-cell hybridoma technique (Cote et al, 1983); and the EBV-hybridoma technique (Cole et al. 1985).
  • techniques described for the production of single chain antibodies can be adapted to produce anti-PD-1 protein single chain antibodies.
  • Compounds useful in practicing the present invention also include anti-PD-1 protein, antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule
  • Fab fragments which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to PD-1 protein.
  • Humanized anti-PD-1 protein antibodies and antibody fragments therefrom can also be prepared according to known techniques.
  • “Humanized antibodies” are forms of non-human (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • any type of antigen and particularly peptide antigen can be used to obtain to obtain high avidity T cells for said antigen.
  • the antigen used according to the invention may be the immunogenic tumor antigen NY-ESO-1 (see for example Gnjatic S et al, 2006).
  • antigens are melanoma antigens and especially melanoma antigen peptides.
  • the melanoma antigens peptides can be the melanoma antigens Melan-A (SEQ ID NO: 1), MELOE-1 (SEQ ID NO: 2), MELOE-2 (SEQ ID NO: 3) and HLa- A2 restricted peptides derived from these antigens. These antigens allow obtaining T cell specific melanoma.
  • melanoma antigens peptides comprising the amino acids motif derived from Melan-A:
  • X2 is leucine, methionine, valine, isoleucine or glutamine
  • the melanoma antigens peptides has the sequence SEQ ID NO: 4. In one embodiment, melanoma antigens peptides comprising the amino acids motif derived from MELOE-1 :
  • X2 is leucine, methionine, valine, isoleucine or glutamine and X9 is alanine, valine or leucine,
  • melanoma antigens peptides comprising the amino acids motif derived from MELOE-2:
  • X2 is cysteine, leucine, methionine, valine, isoleucine or glutamine and X9 is alanine, valine or leucine.
  • peptide refers to an amino acid sequence having less than 50 amino acids.
  • peptide encompasses amino acid sequences having less than 50 amino acids, less than 40 amino acids, less than 30 amino acids, less than 25 amino acids, less than 20 amino acids, less than 15 amino acids or less than 10 amino acids.
  • melanoma antigen peptide a peptide capable of binding to HLA (particularly HLA-A2) molecule and causing a cellular response in a subject against melanoma.
  • said melanoma antigen peptide may comprise a specific motif such that the polypeptide binds an HLA molecule and induces a CTL response.
  • said melanoma antigen peptide may comprise a specific motif such that the polypeptide binds an HLA molecule and induces a helper T cell response.
  • said melanoma antigen peptides as described here above are HLA-A2 restricted.
  • said melanoma antigen peptide is an amino acid sequence of less than 50 amino acids long that comprises the amino acid motif SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above. In another embodiment of the invention, said melanoma antigen peptide is an amino acid sequence of less than 45 amino acids long that comprises the amino acid motif SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above.
  • said melanoma antigen peptide is an amino acid sequence of less than 40 amino acids long that comprises the amino acid SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above.
  • said melanoma antigen peptide is an amino acid sequence of less than 30 amino acids long that comprises the amino acid motif SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above.
  • said melanoma antigen peptide is an amino acid sequence of less than 20 amino acids long that comprises the amino acid motif SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above.
  • said melanoma antigen peptide is an amino acid sequence of less than 15 amino acids long that comprises the amino acid motif SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above.
  • said melanoma antigen peptide is an amino acid sequence of 9, 10 or 11 amino acids long that comprises the amino acid motif SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as defined here above.
  • said melanoma antigen peptide is selected in the group consisting of peptides derives from MELOE-1 having the sequence SEQ ID NO: 7 to SEQ ID NO: 21, peptides derives from MELOE-2 having the sequence SEQ ID NO: 22 to SEQ ID NO: 39 and peptide derives from Melan-A having the sequence SEQ ID NO: 40.
  • the T cells are cultivated with at least one of the melanoma antigen peptide of SEQ ID NO: 7 and SEQ ID NO: 40.
  • the invention also encompasses peptides that are function-conservative variants of melanoma antigen peptides comprising SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6as described here above.
  • the invention encompasses peptides substantially identical to melanoma antigen peptides comprising SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 in which one or more residues have been conservatively substituted with a functionally similar residue and which displays the functional aspects of the melanoma antigen peptides comprising SEQ ID NO: 4, SEQ ID NO: 5 or SEQ ID NO: 6 as described here above, i.e. being still able to bind to an HLA molecule in substantially the same way as a peptide consisting of the given amino acid sequence.
  • hydrophobic residue such as isoleucine, valine, leucine or methionine for another, the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, between glycine and serine, the substitution of one basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue, such as aspartic acid or glutamic acid or another.
  • “conservative substitution” also includes the use of a chemically derivatized residue in place of a non-derivatized residue.
  • “Chemical derivative” refers to a subject peptide having one or more residues chemically derivatized by reaction of a functional side group. Examples of such derivatized molecules include for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides.
  • Free hydroxyl groups may be derivatized to form O-acyl or O-alkyl derivatives.
  • the imidazole nitrogen of histidine may be derivatized to form N-im- benzylhistidine.
  • Chemical derivatives also include peptides which contain one or more naturally-occurring amino acid derivatives of the twenty standard amino acids. For examples: 4-hydroxyproline may be substituted for proline; 5 -hydroxy lysine may be substituted for lysine; 3-methylhistidine may be substituted for histidine; homoserine may be substituted for serine; and ornithine may be substituted for lysine.
  • the melanoma antigen peptide consists essentially of an amino acid sequence according to SEQ ID NO: 7 to 40 or a variant thereof.
  • a peptide according to the present invention in addition to the sequence according to any of SEQ ID No. 7 to SEQ ID No. 40 or a variant thereof, contains additional N- and/or C-terminally located stretches of amino acids that are not necessarily forming part of the peptide that functions as core sequence of the peptide comprising the binding motif and as an immunogenic epitope.
  • the melanoma antigen peptides of the invention can be obtained by synthesizing the peptides according to the method for peptide synthesis known in the art.
  • the present invention also relates to a culture medium comprising anti-PDl antibody and one or several antigens.
  • the culture medium of the present invention is suitable for producing high avidity T cells for said antigens.
  • culture medium refers to a liquid medium suitable for the in vitro culture of T cell, particularly manufactured at clinical grade.
  • the culture medium of the invention contains:
  • a source of carbon as energy substrate such as glucose, galactose or sodium pyruvate
  • vitamins such as biotin, folic acid, B12...;
  • the culture medium may also contain pH buffers in order to maintain the pH of the medium at a value suitable for cell growth.
  • the culture medium of the invention may be based on a commercially available medium such as RPMI 1640 supplemented with foetal calf serum.
  • culture medium of the invention may contain interleukin-2
  • IL-2 IL-2 and/or human serum.
  • Another aspect of the invention relates to an in vitro method for producing T cells with a high avidity for one or several antigens wherein said method comprises the step of culturing of T cells with the culture medium as described above.
  • the step of culturing of T cells with the culture medium of the invention shall be carried out for the necessary time required for the production of functional T cells. Typically, the culture of T cells with the medium of the invention shall be carried out for.
  • the method according to the invention has three culture steps.
  • the first culture step is called "stimulation step”.
  • PBMC from HLA-A2 donor are seeded in 96 well-plates (0.2106/well) in RPMI medium containing antibody anti-PDl, the stimulating peptide, IL-2 (50U/mL) and human serum.
  • This stimulation step is a 10 to 20, particularly, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 days-culture period.
  • Cultured cells are regularly splitted on the basis of cell concentration (when > 106/mL), with fresh medium containing IL-2 and human serum. Typically 2, 3, 4 or 5 splitting during 14, 15 or 16 days.
  • the second culture step is a "sorting step" with HLA-peptide coated beads (Labarriere N et al 2013).
  • the third step is an "amplification step" on irradiated feeder cells, with anti-PDl, PHA and IL-2, of sorted T cells, with anti-PD-1 antibody.
  • This third step is also a 14-16 days- culture period, with regular splitting.
  • the antibody anti-PD-1 is added to culture medium in the three steps presented above.
  • Another aspect of the invention is a kit comprising: (i) anti-PDl antibody and (ii) one or several antigens according to the inventions.
  • anti-PD-1 antibody used for the culture of T cells may be added to culture medium several times during the time of culture, to be maintained at a concentration of 10 ⁇ / ⁇ ., at each cell splitting or medium replacement.
  • the anti-PDl antibody may be added 1 time, 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 8 times, 9 times or 10 times during the culture.
  • the concentration of anti-PD-1 antibody is 1 ⁇ g/mL, 2 ⁇ g/mL, 3 ⁇ g/mL, 4 ⁇ g/mL, 5 ⁇ g/mL, 6 ⁇ g/mL, 7 ⁇ g/mL, 8 ⁇ g/mL, 9 ⁇ g/mL, 10 ⁇ g/mL, 11 ⁇ g/mL or 12 ⁇ g/mL.
  • the concentration of anti-PD-1 antibody is selected between 1 ⁇ g/mL and 12 ⁇ g/mL.
  • PHA-L and/or Interleukin 2 may be added to the culture medium.
  • T cell therapeutic uses and pharmaceutical composition
  • the invention relates to T cell obtainable by the method as above described.
  • T cells obtained by the method of the invention are useful to treat cancer in a subject in need thereof and specially melanoma when melanoma antigens are used.
  • T cells obtained by the method of the invention are useful to treat cancer selected from the group consisting of bile duct cancer (e.g. periphilar cancer, distal bile duct cancer, intrahepatic bile duct cancer), bladder cancer, bone cancer (e.g. osteoblastoma, osteochrondroma, hemangioma, chondromyxoid fibroma, osteosarcoma, chondrosarcoma, fibrosarcoma, malignant fibrous histiocytoma, giant cell tumor of the bone, chordoma, lymphoma, multiple myeloma), brain and central nervous system cancer (e.g.
  • bile duct cancer e.g. periphilar cancer, distal bile duct cancer, intrahepatic bile duct cancer
  • bladder cancer e.g. osteoblastoma, osteochrondroma, hemangioma, chondromyxoid fibroma, osteosarcoma,
  • meningioma astocytoma, oligodendrogliomas, ependymoma, gliomas, medulloblastoma, ganglioglioma, Schwannoma, germinoma, craniopharyngioma), breast cancer (e.g. ductal carcinoma in situ, infiltrating ductal carcinoma, infiltrating, lobular carcinoma, lobular carcinoma in, situ, gynecomastia), Castleman disease (e.g. giant lymph node hyperplasia, angiofollicular lymph node hyperplasia), cervical cancer, colorectal cancer, endometrial cancer (e.g.
  • lung cancer e.g. small cell lung cancer, non-small cell lung cancer
  • mesothelioma plasmacytoma, nasal cavity and paranasal sinus cancer (e.g. esthesioneuroblastoma, midline granuloma), nasopharyngeal cancer, neuroblastoma, oral cavity and oropharyngeal cancer, ovarian cancer, pancreatic cancer, penile cancer, pituitary cancer, prostate cancer, retinoblastoma, rhabdomyosarcoma (e.g.
  • the cancer is a colorectal cancer.
  • melanoma includes, but is not limited to, melanomas, metastatic melanomas, melanomas derived from either melanocytes or melanocytes related nevus cells, melanocarcinomas, melanoepitheliomas, melanosarcomas, melanoma in situ, superficial spreading melanoma, nodular melanoma, lentigo maligna melanoma, acral lentiginous melanoma, ocular melanoma invasive melanoma or familial atypical mole and melanoma (FAM-M) syndrome.
  • melanomas in mammals may be caused by, chromosomal abnormalities, degenerative growth and developmental disorders, mitogenic agents, ultraviolet radiation (UV), viral infections, inappropriate tissue expression of a gene, alterations in expression of a gene, or carcinogenic agents.
  • treating refers to reversing, alleviating or inhibiting the process of one or more symptoms of such disorder or condition.
  • preventing refers to preventing one or more symptoms of such disorder or condition.
  • the term "subject” denotes a mammal, such as a rodent, a feline, a canine, and a primate. Particularly a subject according to the invention is a human.
  • a “therapeutically effective amount” as used herein is intended for a minimal amount of active agent which is necessary to impart therapeutic benefit to a subject.
  • a “therapeutically effective amount of the active agent” to a subject is an amount of the active agent that induces, ameliorates or causes an improvement in the pathological symptoms, disease progression, or physical conditions associated with the disease affecting the subject.
  • a further aspect of the present invention provides an ex vivo and/or in vivo method for treating or preventing cancer.
  • a further aspect of the invention relates to an ex vivo method of treating cancer comprising
  • the invention relates to T cell obtainable by the method as described above for use in the treatment and prevention of cancer.
  • Another aspect of the invention relates to an in vivo method for treating or preventing cancer comprising administering to a subject in need thereof a therapeutically effective amount of T cells as described above.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising T cells as described above and optionally a pharmaceutically acceptable carrier and the use of this pharmaceutical composition in therapy of cancer.
  • the therapeutic ingredients of the invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • “Pharmaceutically” or “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • compositions for example, the route of administration, the dosage and the regimen naturally depend upon the condition to be treated, the severity of the illness, the age, weight, and sex of the patient, etc.
  • compositions of the invention can be formulated for a topical, oral, intranasal, parenteral, intraocular, intravenous, intramuscular or subcutaneous administration and the like.
  • the total daily usage of the T cells and composition of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
  • the cytokine T cells and the composition are administered into the subject simultaneously or sequentially.
  • the T cells may be administered only as a single dose to the individual.
  • the T cells is administered in multiple doses, the administration of successive doses of the T cells being separated by at least 2, 3 or 4 or more weeks.
  • compositions of the invention may further be combined with other active ingredients, for example chemotherapeutics, anti-metastatic or anti-cancer or antiproliferative agents.
  • such compound may be combined with cT cells of the invention for cancer therapy, for example, drugs selected from the group consisting of: immunotherapeutic drugs (Imids), therapeutic monoclonal antibodies, and biological therapeutics.
  • drugs selected from the group consisting of: immunotherapeutic drugs (Imids), therapeutic monoclonal antibodies, and biological therapeutics.
  • FIGURES are a diagrammatic representation of FIGURES.
  • FIG. 1 Amplification rates and Melan-A specific T cell diversity in presence of anti-PD-1 blocking antibody.
  • A Absolute number of Melan-A specific T cells after the step of PBMC-peptide stimulation. 107 PBMC from two healthy donors and three HLA-A2 melanoma patients were stimulated in 96-well plates (2 x 105 cells/well) during 14 days with 1 ⁇ of Melan-AA27Lpeptide, in presence of 10 ⁇ g/mL of anti-PD-1 Ab (hatched bars) or with 10 ⁇ g/mL of control IgG (white bars). At the end of the stimulation period, the absolute number of Melan-A specific T cells was calculated from the total number of expanded T lymphocytes and the percentage of tetramer positive cells.
  • FIG. 2 Avidities of the different VB subfamilies specifically expanded in the two culture conditions.
  • Avidities of specific VB subfamilies amplified in the control condition (dotted lines) or in the presence of anti-PD-1 antibody (solid lines) were evaluated by measuring IFN- ⁇ (A, D), TNF-a production (B, E) and CD 107a membrane expression (C, F) in response to T2 cells loaded with a range of Melan-AA27L peptide, at an E:T ratio of 1 :2.
  • Cytokine production and CD 107a membrane expression were evaluated by double staining with specific anti-VB antibodies and intracellular or membrane labeling.
  • EC50 were determined after activation of Melan-A specific T cell lines by T2 cells loaded with a range of Melan-AA27L peptide (5 hours). The fraction of IFN- ⁇ , TNF-a and CD 107a positive cells among a specific VB subfamily was evaluated by flow cytometry, by double labeling. The % indicates the proportion of each VB subfamily among all Melan-A specific T cells.
  • PBMC Peripheral blood mononuclear cells
  • the melanoma cell line Ml 13 was established from metastatic tumor fragments in the Unit of Cell therapy of France and are registered in the BiocoUection PC-U892-NL (CHUée).
  • the human TAP deficient cell line T2 (174 x CEM.T2) used as a presenting cell was purchased from the ATCC (CRL-1992).
  • Stable cell lines expressing human PD-Ll were established from Ml 13 and T2 cell lines. Briefly, Ml 13 and T2 cells were transfected using lipofectamine, according to the manufacturer's recommendation (Life technologies, France) with an eukaryotic expression vector (pCDNA3) bearing human PD-Ll gene (NM 14143.2), purchased from Sino Biological (Beijing, China). Stable transfectants expressing PD-Ll were selected and cultured in medium containing O ⁇ g/mL of G418 antibiotic.
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • HS human serum
  • IL-2 Proleukin, Novartis, France
  • 10 ⁇ g/mL of either anti-PD-1 antibody Clone EH12.2H7, Biolegend, France
  • 10 ⁇ g/mL of IgGl control istotype Biolegend, France
  • PBMC were stimulated by adding 1 ⁇ of Melan-AA27L peptide (ELAGIGILTV, SEQ ID NO: 40) or 10 ⁇ of MELOE-l 36 -44 peptide (TLNDECWPA, SEQ ID NO: 7).
  • Peptides were purchased from Proteogenix (Schiltigheim, France). Following stimulation, each microculture was evaluated for the percentage of specific CD8 T lymphocytes by double staining with the relevant HLA-peptide tetramer (from the SFR Sante recombinant protein facility) and anti-CD8 mAb (clone BW135/80, Miltenyi Biotec, France) using a FACS Canto HTS. Microcultures that contained at least 1% of Melan-AA27L or MELOE-136-44 specific T cells were selected, pooled and sorted with the relevant multimer-coated beads (35).
  • Sorting of Melan-A and MELOE-1 specific T cells was performed as previously described (35, 40). Sorted specific T cells were seeded at 1000 T cells/well in 96 well plates for polyclonal amplification on feeder cells with 1 ⁇ g/mL of PHA-L (Sigma, France) and 150 IU/mL of IL-2 (Novartis) as previously described (35). To isolate and expand Melan-A and MELOE-1 specific T cell clones from specific sorted T cells, we used a limiting dilution cloning method previously described (49).
  • T cell clones from microcultures showing greater than 95% probability of monoclonality according to the single-hit Poisson law, were selected on the basis on specific tetramer labeling. T cell clones were further expanded into new plates with freshly irradiated feeder cells, IL-2 and PHA-L.
  • Phenotypic and functional analyses were performed on resting or activated T cells.
  • Antigen specific T cells were activated 6 hours in 96 well plates with either coated anti-CD3 antibody (clone OKT3, CRL-8001, ATCC) at ⁇ g/mL, addition of 1 ⁇ of Melan-AA27L peptide (ELAGIGILTV, SEQ ID NO: 40) or 10 ⁇ of MELOE-136-44 peptide (TLNDECWPA, SEQ ID NO: 7), co-culture with the Ml 13 melanoma cell line presenting specific peptides at two effector/target ratios (1/1 and 1/2) or with addition of ⁇ g/mL of phorbol myristate acetate and calcium ionophore (PMA-Cal) (Sigma Aldrich, USA).
  • coated anti-CD3 antibody clone OKT3, CRL-8001, ATCC
  • ELAGIGILTV Melan-AA27L peptide
  • TLNDECWPA MELOE-136-44 peptide
  • PMA-Cal phorbol myristate
  • the specificity of stimulated microcultures, sorted T cell lines and T cell clones was assessed by double labeling with MELOE-1 and Melan-A tetramers (10 ⁇ g/mL) (Recombinant protein facility, SFR Sante, France) and anti-CD8 specific antibody (clone BW135/80, Miltenyi Biotec, France).
  • PD-1 expression was tested on specific T cell clones or sorted T cells at rest and after activation either by double labeling with anti-CD25 (clone M-A251, BD Biosciences, France), as activation marker, and anti-PD-1 antibody (Clone EH12, BD Biosciences), or by a quadruple labeling with specific tetramers, anti-CD8, anti-PD-1 and anti-CD25 antibodies. All the antibodies were used at a concentration of 5 ⁇ g/mL. Vbeta diversity of sorted Melan-A specific T cell lines was analyzed by labeling with 24 anti-Vb mAbs included in the IOTest Beta Mark TCR V Kit (Beckman-Coulter, Marseille, France). The staining protocol includes a one-step procedure with directly conjugated antibody mixes (45 min at 4°C) and a wash step with PBS, 0.1%BSA. All the cytometric analyses were performed on a Facs Canto II (BD Biosciences).
  • RNA was retrotranscribed using Superscript III reverse transcriptase and oligodT (Life technologies, France).
  • Relative quantification of PD-1 and house keeping genes RPLPO and Cyclophilin-A was performed using brilliant SYBR Green qPCR with an Mx4000 machine (Agilent Technologies France). lOng of each cDNA sample were added to RT2 Sybr Green Master Mix (Agilent Technologies) with 200nM of specific primers.
  • PD-1 specific primers were purchased from Qiagen (catalog number PPH13086G, USA). Thermal cycling was one step at 95°C for 10', followed by 40 cycles at 95°C for 30"and 60°C for 1 '.
  • DNA from specific T cells was extracted using QiaAmp DNA mini kit (Qiagen, France). Methyl-Collector Bisulfite modification kit (Active Motif, Belgium) was used for DNA conversion. DNA converted samples were amplified by two successive PCR with specific primers. Thermal cycles for PCR1 were one step at 95°C for 5', followed by 20 cycles at 95°C for 30", 63°C for 2' and 72°C for 1 '30. Thermal cycles for PCR2 were one step at 95°C for 5', followed by 20 cycles at 95°C for 30", 57°C for 1 ' and 72°C for 1 '30. Amplimers were cloned into pSC-B-Amp/Kan vector (Agilent Technologies France) and a minimum of twelve clones for each sample were sequenced (Eurofms scientific, France).
  • IFN- ⁇ secretion of activated T cells was measured by a specific ELISA assay (Human
  • T cells were labeled with PE-conjugated specific anti-VB antibodies (Beckman Coulter). Cells were then fixed for 10 min at room temperature in PBS 4% paraformaldehyde (Sigma, France). Fixed lymphocytes were stained for cytokine production using APC conjugated anti-TNF-a (clone cA2, Miltenyi Biotec) and anti-IFN- ⁇ (clone 45-15, Miltenyi Biotec).
  • CD 107a mobilization experiment specific T cells were stimulated at a E/T ratio of 1/2 with peptide loaded T2 cells for 4 h at 37°C in the presence of APC- conjugated mAb specific for CD 107a (clone H4A3, BD Biosciences, France). The T cells were then stained with selected anti-VB antibodies (Beckman coulter) and analyzed by flow cytometry.
  • PD-1 is differentially expressed on melanoma specific T cells clones
  • Produced T lymphocytes were fully specific and reactive against melanoma cell lines expressing these two widely shared melanoma antigens [Godet Y et al., 2008 and Kawakami Y et al, 1994].
  • T cell clones We tested the ability of these T cell clones to express PD-1 when stimulated by various stimuli: specific peptides, anti-CD3 antibody (OKT3), melanoma cell lines expressing Melan-A and MELOE- 1 antigens or PMA-Cal.
  • specific peptides specific peptides
  • OKT3 anti-CD3 antibody
  • melanoma cell lines expressing Melan-A and MELOE- 1 antigens or PMA-Cal.
  • the fraction of PD-1 expressing T cells increased upon stimulation for PD-l pos T cell clones, regardless of the stimulation mode, whereas PD-l neg T cell clones remained unable or poorly able to express this molecule even when bypassing TCR signaling using PMA-Cal stimulation.
  • PD-1 expression on melanoma specific T cell clones is regulated by epigenetic mechanisms
  • Results obtained show the methylation status of each CpG position for individual clonotypes, and shows that most CpG nucleotides displayed differences in methylation status between PD-lneg and PD-lpos clonotypes, especially from positions 15 to 21 (data not shown).
  • the methylation status of the regulatory region only slightly decreased in one PD-lneg T cell clones (1D12, data not shown), an observation consistent with the low PD-1 expression observed by qPCR in this T cell clone after stimulation (data not shown).
  • the selected cell line stably expressed PD-L1 (data not shown) and similar levels of antigens, together with similar levels of co -stimulation molecules (HLA-A2, ICAM-1, LFA-3) as compared to their non transfected counterparts (data not shown).
  • the reactivity of T cell clones was measured against wild type (data not shown) and transfected cell lines (data not shown) by an IFN- ⁇ specific ELISA test, after a 6hr activation period.
  • results showed that both clones produced IFN- ⁇ upon stimulation with peptide-pulsed T2 cells and that only the reactivity of 4D1 T cell clone (PD-l pos ) was affected by PD-L1 expression on T2 cells. Furthermore, as observed for Melan-A specific T cell clones, the PD-l pos T cell clone (4D1) was slightly more reactive than the PD-l neg one (2A1), in terms of global IFN- ⁇ production on loaded wild type T2 cells. Taken together, these results suggest that PD-l pos specific T cell clones may be of higher avidity than PD-l neg ones.
  • the procedure used to grow melanoma-specific T cells is a two-step process including a first step of peptide-stimulation of melanoma patient's PBMC, and a second step of sort and amplification of specific T lymphocytes.
  • the absolute number of Melan-A specific T cells (calculated from the total number of T cells and the fraction of tetramer-positive lymphocytes at the end of the peptide stimulation step) was higher when the PD-1 blocking antibody was added (Figure 1A). This absolute number was from 2 to 9 times greater in this new culture condition, as compared to the control condition.
  • PD-1 blockade enhances the specific T cell expansion induced by peptide stimulation of patients' PBMC.
  • PD-1 blocking antibody modifies the Melan-A specific T cell repertoire expanded in vitro
  • VB subfamilies selected in the presence of anti-PD-1 mAb exhibited better functional avidities than those amplified in the control condition with a difference in EC50 ranging from 2 to 15 for each tested function, reaching statistical significance for VB16 (IFNg and CD 107a) and VB7.1 (CD 107a) subfamilies from HD49 and for VB7.2 (for the three tested functions) from HD52.
  • Concerning patient P2 the VB14 subpopulation (amplified in the presence of anti-PD-1 antibody and representing 78% of Melan-A specific T cells) exhibited a slightly better EC50 than the other VB families, both in terms of TNF-a and IFN- ⁇ production ( Figure 2 and Table I).
  • PD-1/PD-L1 blockade had a less pronounced effect on PD-1 expression in terms of percentage of positive cells (56%> with anti-PD-1 vs 67%> without) although we observed a marked difference in the two culture conditions in terms of fluorescence intensity suggesting a decreased density of PD-1 molecules on T cells expanded with the blocking antibody.
  • the reactivity of Melan-A specific T cells produced with anti-PD-1 antibody is less or not affected by PD-L1 expression on target cells
  • Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793-800.
  • MELOE-1 is a new antigen overexpressed in melanomas and involved in adoptive T cell transfer efficiency. J Exp Med. 2008;205(11):2673-82.
  • T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 201 l;3(95):95ra73.
  • Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma in vivo persistence, migration, and antitumor effect of transferred T cells.
  • Cutting edge Prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells. J Immunol. 2013;191(2):540-4.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Mycology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Genetics & Genomics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne un procédé in vitro ou ex vivo pour produire des lymphocytes T à avidité élevée pour un ou plusieurs antigènes dans lequel les lymphocytes T obtenus d'un sujet sont cultivés avec ce ou ces antigènes spécifiques et un anticorps anti-PD1.
PCT/EP2016/061941 2015-05-27 2016-05-26 Nouveau procédé pour produire des lymphocytes t WO2016189104A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15305797 2015-05-27
EP15305797.1 2015-05-27

Publications (1)

Publication Number Publication Date
WO2016189104A1 true WO2016189104A1 (fr) 2016-12-01

Family

ID=53267280

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/061941 WO2016189104A1 (fr) 2015-05-27 2016-05-26 Nouveau procédé pour produire des lymphocytes t

Country Status (1)

Country Link
WO (1) WO2016189104A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3775167A4 (fr) * 2018-04-13 2022-01-19 Syz Cell Therapy Co. Procédés de traitement du cancer à l'aide de lymphocytes t spécifiques d'un antigène tumoral

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
WO2001014557A1 (fr) * 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, recepteur de b7-4, et son utilisation
WO2008083174A2 (fr) * 2006-12-27 2008-07-10 Emory University Compositions et procédés pour le traitement d'infections et de tumeurs
WO2014127917A1 (fr) * 2013-02-22 2014-08-28 Curevac Gmbh Combinaison d'une vaccination et de l'inhibition de la voie de pd-1

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
WO2001014557A1 (fr) * 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, recepteur de b7-4, et son utilisation
WO2008083174A2 (fr) * 2006-12-27 2008-07-10 Emory University Compositions et procédés pour le traitement d'infections et de tumeurs
WO2014127917A1 (fr) * 2013-02-22 2014-08-28 Curevac Gmbh Combinaison d'une vaccination et de l'inhibition de la voie de pd-1

Non-Patent Citations (34)

* Cited by examiner, † Cited by third party
Title
AHMADZADEH M; JOHNSON LA; HEEMSKERK B; WUNDERLICH JR; DUDLEY ME; WHITE DE; ROSENBERG SA.: "Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired", BLOOD, vol. 114, no. 8, 2009, pages 1537 - 44
BOUQUIE R; BONNIN A; BERNARDEAU K; KHAMMARI A; DRENO B; JOTEREAU F; LABARRIERE N; LANG F.: "A fast and efficient HLA multimer-based sorting procedure that induces little apoptosis to isolate clinical grade human tumor specific T lymphocytes", CANCER IMMUNOL IMMUNOTHER., vol. 58, no. 4, 2009, pages 553 - 66, XP019706296
DONG H; STROME SE; SALOMAO DR; TAMURA H; HIRANO F; FLIES DB; ROCHE PC; LU J; ZHU G; TAMADA K ET AL.: "Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion", NAT MED., vol. 8, no. 8, 2002, pages 793 - 800, XP002397368
DRENO B; NGUYEN JM; KHAMMARI A; PANDOLFINO MC; TESSIER MH; BERCEGEAY S; CASSIDANIUS A; LEMARRE P; BILLAUDEL S; LABARRIERE N ET AL.: "Randomized trial of adoptive transfer of melanoma tumor-infiltrating lymphocytes as adjuvant therapy for stage III melanoma", CANCER IMMUNOL IMMUNOTHER., vol. 51, no. 10, 2002, pages 539 - 46
GNJATIC SL; NISHIKAWA H; JUNGBLUTH AA; GURE AO; RITTER G; JAGER E; KNUTH A; CHEN YT; OLD LJ.: "NY-ESO-1: review of an immunogenic tumor antigen", ADV CANCER RES., vol. 95, 2006, pages 1 - 30, XP008073732, DOI: doi:10.1016/S0065-230X(06)95001-5
GODET Y; MOREAU-AUBRY A; GUILLOUX Y; VIGNARD V; KHAMMARI A; DRENO B; JOTEREAU F; LABARRIERE N.: "MELOE-1 is a new antigen overexpressed in melanomas and involved in adoptive T cell transfer efficiency", J EXP MED., vol. 205, no. 11, 2008, pages 2673 - 82, XP009126548, DOI: doi:10.1084/jem.20081356
GODET YANN ET AL: "MELOE-1 is a new antigen overexpressed in melanomas and involved in adoptive T cell transfer efficiency", THE JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 205, no. 11, 18 October 2008 (2008-10-18), pages 2673 - 2682, XP009126548, ISSN: 0022-1007, DOI: 10.1084/JEM.20081356 *
HAMID 0; ROBERT C; DAUD A; HODI FS; HWU WJ; KEFFORD R; WOLCHOK JD; HERSEY P; JOSEPH RW; WEBER JS ET AL.: "Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma", N ENGL J MED., vol. 369, no. 2, 2013, pages 134 - 44, XP055182016, DOI: doi:10.1056/NEJMoa1305133
HIRANO FUMIYA ET AL: "Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 65, no. 3, 1 February 2005 (2005-02-01), pages 1089 - 1096, XP002419626, ISSN: 0008-5472 *
HUNDER NN; WALLEN H; CAO J; HENDRICKS DW; REILLY JZ; RODMYRE R; JUNGBLUTH A; GNJATIC S; THOMPSON JA; YEE C.: "Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1", N ENGL J MED., vol. 358, no. 25, 2008, pages 2698 - 703, XP055187197, DOI: doi:10.1056/NEJMoa0800251
KALOS M; LEVINE BL; PORTER DL; KATZ S; GRUPP SA; BAGG A; JUNE CH.: "T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia", SCI TRANSL MED., vol. 3, no. 95, 2011, pages 95RA73, XP002667262, DOI: doi:10.1126/scitranslmed.3002842
KAWAKAMI Y; ELIYAHU S; DELGADO CH; ROBBINS PF; RIVOLTINI L; TOPALIAN SL; MIKI T; ROSENBERG SA: "Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor", PROC NATL ACAD SCI U S A., vol. 91, no. 9, 1994, pages 3515 - 9, XP002628354, DOI: doi:10.1073/pnas.91.9.3515
KHAMMARI A; KNOL AC; NGUYEN JM; BOSSARD C; DENIS MG; PANDOLFINO MC; QUEREUX G; BERCEGEAY S; DRENO B: "Adoptive TIL transfer in the adjuvant setting for melanoma: long-term patient survival", J IMMUNOL RES., 2014, pages 186212
KHAMMARI A; LABARRIERE N; VIGNARD V; NGUYEN JM; PANDOLFINO MC; KNOL AC; QUEREUX G; SAIAGH S; BROCARD A; JOTEREAU F ET AL.: "Treatment of metastatic melanoma with autologous Melan-A/MART-1-specific cytotoxic T lymphocyte clones", J INVEST DERMATOL, vol. 129, no. 12, 2009, pages 2835 - 42
KHAMMARI A; NGUYEN JM; PANDOLFINO MC; QUEREUX G; BROCARD A; BERCEGEAY S; CASSIDANIUS A; LEMARRE P; VOLTEAU C; LABARRIERE N ET AL.: "Long-term follow-up of patients treated by adoptive transfer of melanoma tumor-infiltrating lymphocytes as adjuvant therapy for stage III melanoma", CANCER IMMUNOL IMMUNOTHER., vol. 56, no. 11, 2007, pages 1853 - 60, XP019539120, DOI: doi:10.1007/s00262-007-0340-1
LABARRIERE N; FORTUN A; BELLEC A; KHAMMARI A; DRENO B; SAIAGH S; LANG F.: "A full GMP process to select and amplify epitope-specific T lymphocytes for adoptive immunotherapy of metastatic melanoma", CLIN DEV IMMUNOL, 2013, pages 932318
MACKENSEN A; MEIDENBAUER N; VOGL S; LAUMER M; BERGER J; ANDREESEN R.: "Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma", J CLIN ONCOL., vol. 24, no. 31, 2006, pages 5060 - 9
MACKENSEN ANDREAS ET AL: "Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma", AMERICAN JOURNAL OF CLINICAL ONCOLOGY (CANCER CLINICAL TRIALS), RAVEN PRESS LTD., NEW YORK NY, US, vol. 24, no. 31, 1 November 2006 (2006-11-01), pages 5060 - 5069, XP002538948, ISSN: 0277-3732, DOI: 10.1200/JCO.2006.07.1100 *
MEIDENBAUER N; MARIENHAGEN J; LAUMER M; VOGL S; HEYMANN J; ANDREESEN R; MACKENSEN A.: "Survival and tumor localization of adoptively transferred Melan-A-specific T cells in melanoma patients", J IMMUNOL., vol. 170, no. 4, 2003, pages 2161 - 9
MORGAN RA; DUDLEY ME; WUNDERLICH JR; HUGHES MS; YANG JC; SHERRY RM; ROYAL RE; TOPALIAN SL; KAMMULA US; RESTIFO NP ET AL.: "Cancer regression in patients after transfer of genetically engineered lymphocytes", SCIENCE, vol. 314, no. 5796, 2006, pages 126 - 9, XP002478784, DOI: doi:10.1126/science.1129003
OMID HAMID ET AL: "Safety and Tumor Responses with Lambrolizumab (Anti-PD-1) in Melanoma", NEW ENGLAND JOURNAL OF MEDICINE, vol. 369, no. 2, 11 July 2013 (2013-07-11), pages 134 - 144, XP055182016, ISSN: 0028-4793, DOI: 10.1056/NEJMoa1305133 *
ROBBINS PF; MORGAN RA; FELDMAN SA; YANG JC; SHERRY RM; DUDLEY ME; WUNDERLICH JR; NAHVI AV; HELMAN LJ; MACKALL CL ET AL.: "Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1", J CLIN ONCOL., vol. 29, no. 7, 2011, pages 917 - 24, XP002705979, DOI: doi:10.1200/JCO.2010.32.2537
ROSENBERG SA: "Cell transfer immunotherapy for metastatic solid cancer--what clinicians need to know", NAT REV CLIN ONCOL, vol. 8, no. 10, 2011, pages 577 - 85, XP009170930, DOI: doi:10.1038/nrclinonc.2011.116
S. L. TOPALIAN ET AL: "Survival, Durable Tumor Remission, and Long-Term Safety in Patients With Advanced Melanoma Receiving Nivolumab", JOURNAL OF CLINICAL ONCOLOGY, vol. 32, no. 10, 3 March 2014 (2014-03-03), US, pages 1020 - 1030, XP055218601, ISSN: 0732-183X, DOI: 10.1200/JCO.2013.53.0105 *
SOPHIE R. SIERRO ET AL: "Combination of lentivector immunization and low-dose chemotherapy or PD-1/PD-L1 blocking primes self-reactive T cells and induces anti-tumor immunity", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 41, no. 8, 1 August 2011 (2011-08-01), pages 2217 - 2228, XP055053701, ISSN: 0014-2980, DOI: 10.1002/eji.201041235 *
TOPALIAN SL; HODI FS; BRAHMER JR; GETTINGER SN; SMITH DC; MCDERMOTT DF; POWDERLY JD; CARVAJAL RD; SOSMAN JA; ATKINS MB ET AL.: "Safety, activity, and immune correlates of anti-PD-1 antibody in cancer", N ENGL J MED., vol. 366, no. 26, 2012, pages 2443 - 54, XP055098235, DOI: doi:10.1056/NEJMoa1200690
TOPALIAN SL; SZNOL M; MCDERMOTT DF; KLUGER HM; CARVAJAL RD; SHARFMAN WH; BRAHMER JR; LAWRENCE DP; ATKINS MB; POWDERLY JD ET AL.: "Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab.", J CLIN ONCOL., vol. 32, no. 10, 2014, pages 1020 - 30, XP055218601, DOI: doi:10.1200/JCO.2013.53.0105
VIGNARD V; LEMERCIER B; LIM A; PANDOLFINO MC; GUILLOUX Y; KHAMMARI A; RABU C; ECHASSERIEAU K; LANG F; GOUGEON ML ET AL.: "Adoptive transfer of tumor-reactive Melan-A-specific CTL clones in melanoma patients is followed by increased frequencies of additional Melan-A-specific T cells", J IMMUNOL., vol. 175, no. 7, 2005, pages 4797 - 805, XP002538944
W. WANG ET AL: "PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+CD25Hi regulatory T cells", INTERNATIONAL IMMUNOLOGY, vol. 21, no. 9, 1 September 2009 (2009-09-01), pages 1065 - 1077, XP055217859, ISSN: 0953-8178, DOI: 10.1093/intimm/dxp072 *
WOLCHOK JD; KLUGER H; CALLAHAN MK; POSTOW MA; RIZVI NA; LESOKHIN AM; SEGAL NH; ARIYAN CE; GORDON RA; REED K ET AL.: "Nivolumab plus ipilimumab in advanced melanoma", N ENGL J MED., vol. 369, no. 2, 2013, pages 122 - 33, XP055182024, DOI: doi:10.1056/NEJMoa1302369
YEE C; THOMPSON JA; BYRD D; RIDDELL SR; ROCHE P; CELIS E; GREENBERG PD.: "Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells", PROC NATL ACAD SCI U S A., vol. 99, no. 25, 2002, pages 16168 - 73, XP002505178, DOI: doi:10.1073/PNAS.242600099
YOUNGBLOOD B; NOTO A; PORICHIS F; AKONDY RS; NDHLOVU ZM; AUSTIN JW; BORDI R; PROCOPIO FA; MIURA T; ALLEN TM ET AL.: "Cutting edge: Prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells", J IMMUNOL., vol. 191, no. 2, 2013, pages 540 - 4
YOUNGBLOOD B; OESTREICH KJ; HA SJ; DURAISWAMY J; AKONDY RS; WEST EE; WEI Z; LU P; AUSTIN JW; RILEY JL ET AL.: "Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8(+) T cells", IMMUNITY, vol. 35, no. 3, 2011, pages 400 - 12, XP028298665, DOI: doi:10.1016/j.immuni.2011.06.015
ZITVOGEL L; KROEMER G.: "Targeting PD-1/PD-L1 interactions for cancer immunotherapy", ONCOIMMUNOLOGY, vol. 1, no. 8, 2012, pages 1223 - 5, XP002716013, DOI: doi:10.4161/onci.21335

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3775167A4 (fr) * 2018-04-13 2022-01-19 Syz Cell Therapy Co. Procédés de traitement du cancer à l'aide de lymphocytes t spécifiques d'un antigène tumoral
US11471519B2 (en) 2018-04-13 2022-10-18 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific T cells

Similar Documents

Publication Publication Date Title
JP7349365B2 (ja) 液性腫瘍からの腫瘍浸潤リンパ球の拡大培養及びその治療的使用
JP2022028750A (ja) Cd19に対するヒト化抗原結合ドメイン及び使用方法
JP2020202867A (ja) キメラ受容体及びその使用方法
EP4327878A2 (fr) Combinaison d'une thérapie cellulaire et d'un composé immunomodulateur
AU2016321256A1 (en) NY-ESO-1 specific TCRs and methods of use thereof
KR20210111247A (ko) 치료 및 t 세포 조절을 위한 방법 및 조합
JP7277388B2 (ja) 方法
US20190358262A1 (en) Methods for modulation of car-t cells
JP2019513008A (ja) Btlaに対して特異性を有する抗体及びその使用
JP2020502259A (ja) アルギナーゼ阻害剤併用療法
US20230374453A1 (en) Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
EP4378530A2 (fr) Utilisation de lymphocytes infiltrant les tumeurs pour traiter les patients souffrant de nsclc réfractaires à un anticorps anti-pd-1
JP2021512637A (ja) サイクリンa1特異的t細胞受容体およびその使用
KR20230020421A (ko) Cd70 특이적 융합 단백질을 사용하는 tcr 재프로그래밍을 위한 조성물 및 방법
US11697677B2 (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2016189104A1 (fr) Nouveau procédé pour produire des lymphocytes t
CA3195023A1 (fr) Traitement de patients souffrant de cpnpc avec des therapies lymphocytaires infiltrant les tumeurs
RU2776890C2 (ru) Клеточная терапия, основанная на улучшенных клетках-естественных киллерах
JP6716801B2 (ja) 抗腫瘍剤及びその評価方法
Wang Development of class II-agnostic CAR T cell therapy targeting WT1 Peptide Presented by Diverse HLA class II Molecules
TW202216752A (zh) 用於過繼細胞療法之提供靶向共刺激之嵌合分子
KR20210143779A (ko) 면역 기능을 향상시키기 위한 세포, 조성물 및 방법

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: 16727372

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16727372

Country of ref document: EP

Kind code of ref document: A1