WO1999050637A2 - Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof - Google Patents

Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof Download PDF

Info

Publication number
WO1999050637A2
WO1999050637A2 PCT/US1999/006615 US9906615W WO9950637A2 WO 1999050637 A2 WO1999050637 A2 WO 1999050637A2 US 9906615 W US9906615 W US 9906615W WO 9950637 A2 WO9950637 A2 WO 9950637A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
seq
peptide
complex
cell
Prior art date
Application number
PCT/US1999/006615
Other languages
French (fr)
Other versions
WO1999050637A8 (en
WO1999050637A3 (en
Inventor
Pedro Romero
Rod Dunbar
Danila Valmori
Mikael Pittet
Daniel Speiser
Vincenzo Cerundolo
Graham Ogg
Jean-Charles Cerrotini
Original Assignee
Ludwig Institute For Cancer Research
The Chancellor, Masters And Scholars Of The University Of Oxford
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 Ludwig Institute For Cancer Research, The Chancellor, Masters And Scholars Of The University Of Oxford filed Critical Ludwig Institute For Cancer Research
Priority to AU33654/99A priority Critical patent/AU3365499A/en
Publication of WO1999050637A2 publication Critical patent/WO1999050637A2/en
Publication of WO1999050637A8 publication Critical patent/WO1999050637A8/en
Publication of WO1999050637A3 publication Critical patent/WO1999050637A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56977HLA or MHC typing
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • 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
    • 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
    • C07K16/2809Immunoglobulins [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 the T-cell receptor (TcR)-CD3 complex
    • 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
    • C07K16/2815Immunoglobulins [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 CD8
    • 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
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • 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
    • C07K16/289Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD45

Definitions

  • This invention relates to multicomponent complexes which are useful in analysis of T
  • Tumor Antigens Using Fluorogenic MHC Class I/Peptide Complexes
  • the family of turn " antigen presenting cell lines are immunogenic variants obtained by
  • antigens are obtained by mutating tumor cells which do not generate an immune response in
  • immunosuppressive amounts of radiation are administered with the following challenge of cells
  • tumor rejection antigen a so-called "tumor rejection antigen” in a tumor which is a target for a syngeneic rejection response. Similar results have been obtained when foreign genes have been
  • a class of antigens has been recognized which are presented on the surface of tumor cells
  • TRAs tumor rejection antigens
  • TRAs may or may not elicit antibody
  • CTL cytolytic T cell
  • a tumor exemplary of the subject matter described supra is known as P815. See DePlaen
  • the P815 tumor is a mastocytoma, induced in a DBA/2 mouse with
  • the P815 line has generated many turn " variants following mutagenesis, including variants referred to as P91A
  • antigens are only present after the tumor cells
  • Tumor rejection antigens are present on cells of a given tumor without
  • a cell line can be tu ⁇ T, such as the line
  • PI phenotype
  • PI 98 differ from their normal alleles by point mutations in the coding regions of the gene.
  • P91 A is presented by L d , P35 by D d and PI 98 by K d .
  • tumor cells are expressed in tumor cells, and can serve as markers for the diagnosis of such tumors.
  • nonapeptide amino acids long
  • positions 5 and 8 of an octamer, for H-2D b they are positions 5 and 9 of a nonapeptide while the
  • anchor residues for HLA-A1 are positions 3 and 9 of a nonamer.
  • HLA molecules For HLA molecules,
  • positions 2 and 9 are anchors.
  • CTLs cytolytic T cells
  • Patent No. 5,571,711 as well as many other issued U.S. patents found that when comparing
  • nonapeptides are useful for various purposes including their use as immunogens, either alone or
  • Nonapeptides are of sufficient size to constitute an antigenic epitope
  • melanoma antigens which are recognized by cytolytic T cells are now divided into three broad
  • antigens are the expression product of normal genes which are usually silent in normal tissues.
  • a second family of melanoma antigens includes antigens which are derived from mutant
  • MUM-1 Manton, et al., Proc. Natl. Acad.
  • a third category also discussed, supra, includes the differentiation
  • antigens which are expressed by both melanoma and melanocytes.
  • antigens which are expressed by both melanoma and melanocytes.
  • exemplary are tyrosinase
  • CTLs Cytolytic T cells
  • lymphocytes and tumor infiltrating lymphocytes, of melanoma patients such as those who are
  • SEQ ID NO:2 (SEQ ID NO:2), is actually a better target than SEQ ID NO: 1. See U.S. Patent Application
  • the invention relies on the known avidity of two binding
  • the invention involves
  • multiple targets for cytolytic T cells i.e., a plurality of immune complexes which comprise an
  • MHC molecule such as an HLA molecule, a B2 microglobulin, and a peptide which binds to the
  • the complex is labelled, and can be used to isolate, or to determine, cytolytic
  • Figure 1 shows a correlation between frequency of cells positive for particular peptide containing
  • Figure 2 shows that CD8 + cells, isolated from bulk cultures and stimulated and expanded without
  • antigen had high tumoricidal activity.
  • Figure 3 presents data on determination of relative activity of peptides.
  • Figure 4 sets forth data showing that complexes of HLA-A2 and SEQ ID NO: 4 are more stable
  • FIG. 5 presents data contributing to analysis of both polyclonal and monoclonal T cell
  • Figure 6 presents phenotype data for CD8 + cells specific for particular peptide containing
  • Figure 8 summarizes data on stimulation of cytokine production by polyclonal and monoclonal
  • Figure 9 summarizes results of experiments designed to test PBL activity in the presence of NK
  • HLA-A * 0201 positive cells and HLA-A * 0201 was then cloned, using specific primers for the
  • extracellular coding portion of the molecule was amplified, again using specific primers.
  • inclusion bodies were purified and the HLA and B2 microglobulin recombinant proteins were
  • refolding solution contained 100 mM Tris, at pH 8.0, L-arginine, 400 mM, EDTA, 2 mM,
  • BirA reaction buffer (Tris 100 mM, pH 7.5, NaCl 200 mM, Mg Cl 2 5mM, PMSF 100 ⁇ M,
  • SEQ ID NO: 5 Gly He Leu Gly Phe Val Phe Thr Leu (SEQ ID NO: 5) are all known to bind to HLA-A2 molecules, and stimulate CTLs.
  • SEQ ID NO: 2 is derived
  • SEQ ID NO: 3 is an analogue of SEQ ID NO: 2
  • SEQ ID NO: 4 is derived from
  • SEQ ID NO: 5 is a control peptide
  • influenza matrix protein (Gotch et al., 1987, Nature 326; 881). Hence, they were used in
  • TILNS tumor infiltrated lymph nodes
  • TILNS were surgically removed from ten melanoma patients, who had been typed as
  • TILNS HLA- A * 0201 positive.
  • the TILNS were treated in the same manner as the TILNS in Romero
  • NTNs normal lymph nodes
  • samples containing from 0.5-l.OxlO 6 were stained with fluorescent, anti-CD3, anti-CD8
  • the anti-CD3 antibody was labelled with PerCP (peridinin chlorophyll protein).
  • anti-CD8 antibody was labelled with FITC. A total of 1-2 ug of the tetramer being tested, in 20
  • lymphocytes (0.12 to 21% of CD3 + and CD8 + lymph node
  • the binding lymphocytes were CD3 + CD8 + lymph node cells. There were also high numbers of TILN cells which bound to the tetramers containing SEQ ID NO: 4, ranging from
  • the levels ofSEQ ID NO: 3 tetramer binding lymphocytes ranged from 0.12 to 1.29%.
  • lymph node a heavily tumor infiltrated lymph node and a normal lymph node ("NLN") from the same patient.
  • the suspensions were cultured, as described supra, overnight, for 15 days, or for 21 days and
  • Tetramer positive cells rose from 0.12 to
  • CD8 + cells rose from 210 to about 1.8x10 5 after 15 days, and 5.38x 10 5 after 21 days, representing
  • TILN cells from a patient were cultured, for 21 days, as described supra, using
  • tetramer positive T lymphocytes were able to kill T2 target cells sensitized with the peptide of
  • TILNs which were sorted as
  • lymphocytes which were specified to the tetramers containing the
  • chlorophyll protein anti-CD45RO, labelled with FITC, and the tetramers described supra.
  • vivo cells suspensions were prepared from two patients, and then studied, with a tumor
  • infiltrated lymph node being tested with an unfiltrated node used for comparison.
  • tetramers could be used to isolated complex specific CTLs from circulating lymphocytes.
  • cryopreserved peripheral blood lymphocytes were thawed (37°C, 3 minutes), then
  • CD8 T cells following standard techniques. The cells were then analyzed, using CD45RA " specific antibodies labelled with cytochrome, and CD28 labelled with FITC, following the
  • CD45RA ⁇ This shows that uncultured, unstimulated blood lymphocytes can be assayed for
  • tetramer was then used to study PBMCs of individuals who were not known to be suffering from
  • influenza infection to determine if the tetramers could be used to assay for low frequency CTLs
  • PBMCs were stained, sorted into sets of 15 cells and duplicate
  • CD8 + cells failed to respond at all. Further, one of the clones was tested in a chromium
  • tetramer staining followed by FACS sorting, can be used to generate highly effective
  • the frequency of tetramer positive cells ranged from 0.03% to 0.09% of the total CD8 +
  • PBMCs were taken from 3 of the 7
  • IL-2 1 OU/ml
  • IL-7 1 Ong/ml
  • the assay described in example 15 was carried out on the samples 7 days after each
  • T2 cells in the absence or presence of each of the two peptides.
  • T2 cells are known as cells
  • the assays were carried out by labelling the T2 cells
  • the effector cells had been preincubated for at least 20 minutes at 37 °C in the presence
  • lymphocytes in peripheral blood. Their stimulatory capacity appears to be similar. Notwithstanding this point, the increase in proportion of tetramer positive lymphocytes
  • lymphocytes using flourescent tetramers, and to separate them from bulk cultures. They suggest the possibility of early stage, in vitro stimulation with peptide, and continued expansion in
  • Me 290 is illustrative of additional Melan-A positive tumor cell lines which
  • target cells 50 ⁇ ls
  • varying concentrations of peptides 50 ⁇ l
  • E26A (SEQ ID NO: 37) 0.09 0.14 0.12 0.19 1 Melan-A Melan-A E26A A27L E26A/A27
  • E26A/A27L (SEQ ID NO: 38) 0.02 0.01 0.008 0.005 0.5
  • E26A (SEQ ID NO: 37) 300 157 183 194 65 A27L (SEQ ID NO: 3) 180 110 110 231 162 E26A/A27L (SEQ ID NO: 38) 1350 2200 2750 7400 130
  • the peptide presented supra as SEQ ID NO: 4 is derived from the tyrosinase molecule
  • NO: 4 functioning as a tumor rejection antigen. See, e.g., Brichard, et al., J. Exp. Med. 178:489
  • test peptides SEQ ID NOS: 39-47 were added, in 50 ⁇ l
  • influenza matrix peptide was used.
  • SEQ ID NO: 4 displays intermediate competitor activity, i.e., to secure
  • Tyrosinase 368-376 YMDGTMSQV (SEQ ID NO: 4) 102
  • H YADGTMSQV (SEQ ID NO: 40) 300 0.3
  • H YMDGTASQV (SEQ ID NO: 44) 200 0.6 d YMDGTMAQV (SEQ ID NO: 45) 50 r 2.0 w YMDGTMSAV (SEQ ID NO: 46) 30 3.3 t YMDGTMSQA (SEQ ID NO: 47) 80 1.3
  • Influenza A matrix 58-66 GILGFVFTL (SEQ ID NO: 5) 17
  • SEQ ID NO: 2 are unstable, dissociating completely in under one hour when incubated at
  • T2 cells were loaded with saturating concentrations (lO ⁇ M) of one of SEQ ID NO: 4,
  • T2 cells were treated with
  • the CD8 ⁇ , tetramer positive fraction lysed the T2 cells in the presence of SEQ ID NO: 4, but not
  • PBMCs irradiated allogeneic PBMCs, EBV transformed B lymphocytes, PHA and recombinant IL-2.
  • NA8-MEL is an HLA-A*0201 positive melanoma cell line
  • NA8-MEL tyr is a cell line derived by transfecting NA8
  • Melan-A + cell line is also tyrosinase + .
  • the melanoma cell lines and T cells were combined, at varying ratios, either with or
  • Example 27 Fine antigenic specificity of the T cell populations discussed in example 26 was analyzed
  • Relative avidity was determined by comparison to the concentration of the peptide of SEQ ID NO: 1
  • the net result is a showing of wide heterogeneity in the recognition of epitopes
  • Example 15 showed how tetramers of HLA-A* 0201 and a Melan-A derived
  • peptide could be used to measure the frequency of CD8 + T lymphocytes in a population. Similar
  • IL-2 100 U/ml
  • IL-7 10 ng/ml
  • lymphocytes were detectable in six out of 10 samples after only short term culture. (The other
  • the values refer to the percentage of A2/SEQ ID NO:4 tetramer positive T cells, relative to the
  • these PBLs were pulsed with lO ⁇ M of SEQ ID NO. 9 in "CTL medium” (Iscove's medium, 5% human serum, IL-2 at 100 U/ml), plus IL-7 (lOng/ml).
  • CTL medium Iscove's medium, 5% human serum, IL-2 at 100 U/ml
  • IL-7 ILOng/ml
  • IL-7 (lOng/ml). Any proliferating blasts were expanded in the CTL medium for 14 days (for
  • Tetramer staining was carried out as described supra, using appropriate tetramers.
  • patient MM15 showed very few CD8 + cells in a PHA stimulated culture from skin metastasis; however, a high portion of these
  • CTLs are cloned, directly, from these CD8 + populations.
  • the cloning plates were incubated at 37°C in 5% CO 2 for 10-14 days, without any
  • SK-Mel-29 and SK-Mel 23 both of which are HLA-A2.T, and express both Melan-A and
  • the lysis percentage was calculated by the formula:
  • Example 32 The expanded CTLs were then tested for surface phenotytpes via FACS, using FITC
  • CD 49a-e., inclusive integrated-Bl-6, VLA 1-6
  • goat-antimouse Ig goat-antimouse Ig. Also, anti-
  • CLA cutaneous lymphocyte antigen
  • FITC labelled mouse-antirat IgM labelled with FITC
  • CD45RO CD45RA ⁇ CD44 + and CD62L " , which is consistent with previous antigenic
  • the clones were CD1 lb and CD49d positive, which is
  • VCAM-1 - a phenotype associated with metastatic melanoma (Rice, et al., Science 246:1303
  • clones with the best proliferative potential might be pre-selected by only
  • PBMCs were isolated from samples, using standard methods. The
  • CD8 + cells in these samples were purified via two rounds of positive selection, using magnetic
  • CD8 + cells were then stained with tetramers of HLA-A*0201 and SEQ ID NO: 3,
  • control peptide SEQ ID NO: 5 were present in both melanoma patients and healthy subjects.
  • CD28, CD45RA, and CD45RO were phenotyped for CD28, CD45RA, and CD45RO.
  • CD8 + lymphocyte populations were purified, as described supra. The highly
  • CD45RA and negative for CD45RO when the cells were positive for tetramers of SEQ ID NO:
  • CD8 + T cells which were positive for SEQ ID NO: 5 tetramers were CD28 and
  • CD45RO positive, and CD45RA negative i.e., CD45RA low . This suggests that the CD8" T cells
  • SEQ ID NO: 5 The phenotype of the CD8 + T cells from
  • CD45RO negative a phenotype identical to that of healthy donors' CD8 + T cells.
  • CD45RA negative positive, CD45RA negative
  • CD28 negative CD45RA
  • CD45RA * cells were by a factor of 3.6 for CD45RA " (CD45RA ,0W ) cells, but 11.4 for CD45RA * cells.
  • SEQ ID NO: 3 specific spots were only detectable in one melanoma
  • ELISPOT is restricted to memory phenotype, antigen specific cells.
  • amino acids 368-378 of tyrosinase i.e., SEQ ID NO: 4
  • TILN tumor infiltrated lymph nodes
  • HLA-A T 0201 positive The single cells were cultured in medium (2 ml of
  • FITC peridinin chlorophyll protein
  • TILN were found to contain more than 90% CD37CD8 + cells. Of these, 19.4% were positive
  • T2 cells labelled with the 51 Cr as a target T2 cells labelled with the 51 Cr as a target.
  • T2 cells were incubated with the peptides for 15 minutes at room
  • Chromium release was measured after 4 hours at 37°C.
  • T cell receptor down regulation is one of the earliest T cell activation events induced upon
  • T2 cells were pulsed for 1 hour at 37°C with varying concentrations of
  • T cells were loaded with a commercially available fluorescent dye
  • SEQ ID NO: 2 triggered Ca 2+ influx in only 60% of the cells of the CTL
  • SEQ ID NO: 3 mobilized Ca 2* in
  • SEQ ID NO: 3 also induced a more rapid Ca 2* response as compared to the
  • cytokine synthesis events were also studied, i.e., cytokine synthesis, and induction of cytolytic effector cell function.
  • the former was done via intracellular staining of cytokines, which is described by Jung, et al,
  • gamma, GM-CSF, and IL-4 were used. All mAbs were labelled with phycoerythrin. In brief,
  • the saponin permeabilizes the cell, allowing entry of the mAbs.
  • ID NOS: 1 and 2 only stimulated small fractions of the CTLs (e.g., only 30% o of the cells of the
  • SEQ ID NO: 3 stimulated the totality of CTLs able to
  • the polyclonal population was distinguishable from the monoclonal population in that
  • SEQ ID NOS: 1 and 2 stimulated low fractions of these cells
  • cytolytic effector function Such functions require strongly agonistic peptides, and/or higher peptide concentrations, in order to maximize induction of expansion and cytolytic effector
  • PBMCs stimulator cells
  • peptide pulsed PBMCs were washed, twice, irradiated (3000 rads), and then adjusted to

Abstract

The invention involves multicomponent complexes, which use useful in analysis of T cells. The complexes contain at least first and second binding partners, which bind to each other. The second binding partner engages a plurality of immune complexes, which comprise an NHC molecule, a β2 microglobulin molecule, and a peptide. Preferably, there are at least four of these immune complexes per multicomponent complex. These can be used to determine or to isolate cytolytic T cells.

Description

ISOLATED MULTIMERIC COMPLEXES USEFUL IN ANALYSIS OF T CELLS, PEPTIDES USEFUL IN MAKING THE COMPLEXES, AND USES THEREOF
RELATED APPLICATION
This application is a continuation in part of Serial No. 09/049,850, filed on March 27,
1998, and incorporated by reference in its entirety.
FIELD OF THE INVENTION
This invention relates to multicomponent complexes which are useful in analysis of T
cell populations.
RELATED ART
Certain aspects of this invention will be found in published materials involving some of
the inventors. See Dunbar et al., "Characterization of Human Cytotoxic T Lymphocyte (CTL)
responses To Tumor Antigens Using Fluorogenic MHC Class I/Peptide Complexes", Tumor
Immunology 92 Suppl. Dec. 1997, reporting abstract 3.3 of the 5th Annual Congress of the BSI.
Also see Dunbar et al., "Direct isolation, phenotyping and cloning of law- frequency antigen
specific cytotoxic T lymphocytes from peripheral blood", Curr. Biol. (in press), but possibly
available via Internet on March 16, 1998. BACKGROUND AND PRIOR ART
The study of the recognition or lack of recognition of cancer cells by a host organism has
proceeded in many different directions. Understanding of the field presumes some
understanding of both basic irnmunology and oncology.
Early research on mouse tumors revealed that these displayed molecules which led to
rejection of tumor cells when transplanted into syngeneic animals. These molecules are
"recognized" by T-cells in the recipient animal, and provoke a cytolytic T-cell response with
lysis of the transplanted cells. This evidence was first obtained with tumors induced in vitro by
chemical carcinogens, such as methylcholanthrene. The antigens expressed by the tumors and
which elicited the T-cell response were found to be different for each tumor. See Prehn, et al.,
J. Natl. Cane. Inst. 18: 769-778 (1957); Klein et al., Cancer Res. 20: 1561-1572 (1960); Gross,
Cancer Res. 3: 326-333 (1943), Basombrio, Cancer Res. 30: 2458-2462 (1970) for general
teachings on inducing tumors with chemical carcinogens and differences in cell surface antigens.
This class of antigens has come to be known as "tumor specific transplantation antigens" or
"TSTAs". Following the observation of the presentation of such antigens when induced by
chemical carcinogens, similar results were obtained when tumors were induced in vitro via
ultraviolet radiation. See Kripke, J. Natl. Cane. Inst. 53: 333-1336 (1974).
While T-cell mediated immune responses were observed for the types of tumor described
supra, spontaneous tumors were thought to be generally non-immunogenic. These were
therefore believed not to present antigens which provoked a response to the tumor in the tumor
carrying subject. See Hewitt, et al., Brit. J. Cancer 33: 241-259 (1976).
The family of turn" antigen presenting cell lines are immunogenic variants obtained by
mutagenesis of mouse tumor cells or cell lines, as described by Boon et al., J. Exp. Med. 152: 1184-1193 (1980), the disclosure of which is incorporated by reference. To elaborate, turn
antigens are obtained by mutating tumor cells which do not generate an immune response in
syngeneic mice and will form tumors (i.e., "tum+" cells). When these turn" cells are
mutagenized, they are rejected by syngeneic mice, and fail to form tumors (thus "turn""). See
Boon et al., Proc. Natl. Acad. Sci. USA 74: 272 (1977), the disclosure of which is incorporated
by reference. Many tumor types have been shown to exhibit this phenomenon. See, e.g., Frost
et al, Cancer Res. 43: 125 (1983).
It appears that turn" variants fail to form progressive tumors because they initiate an
immune rejection process. The evidence in favor of this hypothesis includes the ability of "turn "
variants of tumors, i.e., those which do not normally form tumors, to do so in mice with immune
systems suppressed by sublethal irradiation, Van Pel et al., Proc. Natl. Acad. Sci. USA 76: 5282-
5285 (1979); and the observation that intraperitoneally injected turn" cells of mastocytoma P815
multiply exponentially for 12-15 days, and then are eliminated in only a few days in the midst
of an influx of lymphocytes and macrophages (Uyttenhove et al., J. Exp. Med. 152: 1175-1183
(1980)). Further evidence includes the observation that mice acquire an immune memory which
permits them to resist subsequent challenge to the same turn" variant, even when
immunosuppressive amounts of radiation are administered with the following challenge of cells
(Boon et al, Proc. Natl, Acad. Sci. USA 74: 272-275 (1977); Van Pel et al., supra: Uyttenhove
et al., supra). Later research found that when spontaneous tumors were subjected to
mutagenesis, immunogenic variants were produced which did generate a response. Indeed, these
variants were able to elicit an immune protective response against the original tumor. See Van
Pel et al., J. Exp. Med. 157: 1992-2001 (1983). Thus, it has been shown that it is possible to
elicit presentation of a so-called "tumor rejection antigen" in a tumor which is a target for a syngeneic rejection response. Similar results have been obtained when foreign genes have been
transfected into spontaneous tumors. SeeFearonetal., Cancer Res.48: 2975-1980(1988) in this
regard.
A class of antigens has been recognized which are presented on the surface of tumor cells
and are recognized by cytolytic T cells, leading to lysis. This class of antigens will be referred
to as "tumor rejection antigens" or "TRAs" hereafter. TRAs may or may not elicit antibody
responses. The extent to which these antigens have been studied, has been via cytolytic T cell
characterization studies, in vitro i.e., the study of the identification of the antigen by a particular
cytolytic T cell ("CTL" hereafter) subset. The subset proliferates upon recognition of the
presented tumor rej ection antigen, and the cells presenting the tumor rej ection antigens are lysed.
Characterization studies have identified CTL clones which specifically lyse cells expressing the
tumor rejection antigens. Examples of this work may be found in Levy et al., Adv. Cancer Res.
24: 1-59 (1977); Boon et al, J. Exp. Med. 152: 1184-1193 (1980); Brunner et al, J. Immunol.
124: 1627-1634 (1980); Maryanski et al., Eur. J. Immunol. 124: 1627-1634 (1980); Maryanski
et al., Eur. J. Immunol. 12: 406-412 (1982); Palladino et al., Cane. Res. 47: 5074-5079 (1987).
This type of analysis is required for other types of antigens recognized by CTLs, including minor
histocompatibility antigens, the male specific H-Y antigens, and the class of antigens referred
to as "turn " antigens, and discussed herein.
A tumor exemplary of the subject matter described supra is known as P815. See DePlaen
et al., Proc. Natl. Acad. Sci. USA 85: 2274-2278 (1988); Szikora et al., EMBO J 9: 1041-1050
(1990), and Sibille et al, J. Exp. Med. 172: 35-45 (1990), the disclosures of which are
incorporated by reference. The P815 tumor is a mastocytoma, induced in a DBA/2 mouse with
methylcholanthrene and cultured as both an in vitro tumor and a cell line. The P815 line has generated many turn" variants following mutagenesis, including variants referred to as P91A
(DePlaen, supra). 35B (Szikora, supra), and PI 98 (Sibille, supra). In contrast to tumor rejection
antigens - and this is a key distinction - the turn" antigens are only present after the tumor cells
are mutagenized. Tumor rejection antigens are present on cells of a given tumor without
mutagenesis. Hence, with reference to the literature, a cell line can be tuπT, such as the line
referred to as "PI", and can be provoked to produce turn" variants. Since the turn" phenotype
differs from that of the parent cell line, one expects a difference in the DNA of turn" cell lines
as compared to their turn" parental lines, and this difference can be exploited to locate the gene
of interest in turn" cells. As a result, it was found that genes of turn" variants such as P91 A, 35B
and PI 98 differ from their normal alleles by point mutations in the coding regions of the gene.
See Szikora and Sibille. supra, and Lurquin et al., Cell 58: 293-303 (1989). This has proved not
to be the case with the TRAs of this invention. These papers also demonstrated that peptides
derived from the turn" antigen are presented by H-2d Class I molecules for recognition by CTLs.
P91 A is presented by Ld, P35 by Dd and PI 98 by Kd.
PCT application PCT/US92/04354, filed on May 22, 1992 assigned to the same assignee
as the subject application, teaches a family of human tumor rejection antigen precursor coding
genes, referred to as the MAGE family. Several of these genes are also discussed in van der
Bruggen et al, Science 254: 1643 (1991). It is now clear that the various genes of the MAGE
family are expressed in tumor cells, and can serve as markers for the diagnosis of such tumors,
as well as for other purposes discussed therein. See also Traversari et al., Immunogenetics 35 :
145 (1992); van der Bruggen et al., Science 254: 1643 (1991) and De Plaen, et al.,
Immunogenetics 40: 360 (1994). The mechanism by which a protein is processed and presented
on a cell surface has now been fairly well documented. A cursory review of the development of the field may be found in Barinaga, "Getting Some 'Backbone': How MHC Binds Peptides",
Science 257: 880 (1992); also, see Fremont et al, Science 257: 919 (1992); Matsumura et al.,
Science 257: 927 (1992); Engelhard, Ann. Rev. Immunol 12:181-207 (1994); Madden, et al.,
Cell 75:693-708 (1993); Ramensee, etal, Ann. Rev. Immunol 11:213-244(1993); German, Cell
76: 287-299 ( 1994). These papers generally point to a requirement that the peptide which binds
to an MHC/HLA molecule be nine amino acids long (a "nonapeptide"), and to the importance
of the second and ninth residues of the nonapeptide. For H-2K\ the anchor residues are
positions 5 and 8 of an octamer, for H-2Db, they are positions 5 and 9 of a nonapeptide while the
anchor residues for HLA-A1 are positions 3 and 9 of a nonamer. Generally, for HLA molecules,
positions 2 and 9 are anchors.
Studies on the MAGE family of genes and many other TRAPs have now revealed that
these are processed to peptides which complex with HLA molecules, leading to lysis of the cells
presenting these by cytolytic T cells ("CTLs").
Research presented in, e.g., U.S. Patent No. 5,405,940 filed August 31 , 1992, and in U.S.
Patent No. 5,571,711, as well as many other issued U.S. patents found that when comparing
homologous regions of various MAGE genes to the region of the MAGE-1 gene coding for the
relevant nonapeptide, there is a great deal of homology, and identification of a family of
nonapeptides, all of which have the same N-terminal and C-terminal amino acids. These
nonapeptides are useful for various purposes including their use as immunogens, either alone or
coupled to carrier peptides. Nonapeptides are of sufficient size to constitute an antigenic epitope,
and the antibodies generated thereto were described as being useful for identifying the
nonapeptide, either as it exists alone, or as part of a larger polypeptide. The preceding survey of the relevant literature shows that various peptides, usually eight,
nine, or ten amino acids in length, complex with MHC molecules and present targets for
recognition by cytolytic T cells. A great deal of study has been carried out on melanoma, and
melanoma antigens which are recognized by cytolytic T cells are now divided into three broad
categories. The first, which includes many of the antigens discussed, supra, (e.g., MAGE), are
expressed in some melanomas, as well as other tumor types, and normal testis and placenta. The
antigens are the expression product of normal genes which are usually silent in normal tissues.
A second family of melanoma antigens includes antigens which are derived from mutant
forms of normal proteins. Examples of this family are MUM-1 (Coulie, et al., Proc. Natl. Acad.
Sci. USA92:7976-7980(1955)); CDK4(Wόlfel, etal., Science269:1281-1284(1955));Bcatenin
(Robbins, et al., J. Exp. Med. 183: 1185-1192 (1996)); and HLA- A2 (Brandel, et al., J. Exp. Med.
183:2501-2508 (1996)). A third category, also discussed, supra, includes the differentiation
antigens which are expressed by both melanoma and melanocytes. Exemplary are tyrosinase
gplOO, gp75, and Melan A Mart-1. See U.S. Patent No. 5,620,886 incorporated by reference,
with respect to Melan-A. See Wδlfel, et al., Eur. J. Immunol. 24: 759 (1994) and Brichard, et
al., Eur. J. Immunol. 26: 224 (1996) for tyrosinase; Kang, et al, J. Immunol. 155: 1343 (1995);
Cox, etal, Science 264: 716 (1994); Kawakami, etal., J. Immunol. 154: 3961 (1995) for gp 100;
Wang, et al, J. Exp. Med. 183: 1131 (1996) for gp 75.
Cytolytic T cells ("CTLs" hereafter) have been identified in peripheral blood
lymphocytes, and tumor infiltrating lymphocytes, of melanoma patients such as those who are
HLA-A*0201 positive. See Kawakami, et al, Proc. Natl. Acad. Sci. USA 91 :3515 (1994);
Coulie, et al, J. Exp. Med. 180:35 (1994). When ten HLA-A'0201 restricted Melan-A specific
CTLs were derived from different patients were tested, nine of them were found to recognize and react with the peptide Ala Ala Gly He Gly He Leu Thr Val, (SEQ ID NO:l), which consists of
amino acids 27-35 of Melan-A. (Kawakami, et al, J. Exp. Med 180:347-352 (1994)). Rivoltini,
et al, J. Immunol 154:2257 (1995), showed that Melan-A specific CTLs could be induced by
stimulating PBLs from HLA- A* 0201 positive normal donors, and melanoma patients, using SEQ
ID NO: 1. The strength of this response has led to SEQ ID NO: 1 being proposed as a target for
vaccine development. Further research has shown that a decapeptide, i.e.,
Glu Ala Ala Gly He Gly He Leu Thr Val
(SEQ ID NO:2), is actually a better target than SEQ ID NO: 1. See U.S. Patent Application
Serial No. 08/880,963, filed June 23 1997 and incorporated by references.
One difficulty in the area of cancer immunology is a lack of reliable protocols which can
be used to identify and to quantify in vivo cytolytic T lymphocyte responses. As a result, it is
difficult to characterize immune response, and to monitor vaccine trials. The invention described
hereafter was developed to address these, and other issues in the field. It has been found that
analysis of cytolytic T cells is greatly facilitated by the use of complexes containing a plurality
of T cell targets. More specifically, the invention relies on the known avidity of two binding
partners, such as avidin or streptavidin and biotin for each other. It is well known that every
molecule of avidin/streptavidin can bind to four biotin molecules. The invention involves
constructs where the avidin/streptavidin-biotin system is used to form complexes containing
multiple targets for cytolytic T cells, i.e., a plurality of immune complexes which comprise an
MHC molecule, such as an HLA molecule, a B2 microglobulin, and a peptide which binds to the
HLA molecule. The complex is labelled, and can be used to isolate, or to determine, cytolytic
T cells of interest in a sample. How these and other features of the invention are achieved will be seen in the disclosure
which follows.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows a correlation between frequency of cells positive for particular peptide containing
tetramers and cytotoxicity.
Figure 2 shows that CD8+ cells, isolated from bulk cultures and stimulated and expanded without
antigen had high tumoricidal activity.
Figure 3 presents data on determination of relative activity of peptides.
Figure 4 sets forth data showing that complexes of HLA-A2 and SEQ ID NO: 4 are more stable
than complexes of HLA-A2 and SEQ ID NO: 1.
Figure 5 presents data contributing to analysis of both polyclonal and monoclonal T cell
populations specific for tetramers of HLA- A* 0201 and SEQ ID NO: 4.
Figure 6 presents phenotype data for CD8+ cells specific for particular peptide containing
tetramers.
Figure 7 A-7C, inclusive, depict the results of experiments designed to compare fine specificities
of unsorted TILN populations.
Figure 8 summarizes data on stimulation of cytokine production by polyclonal and monoclonal
cell populations.
Figure 9 summarizes results of experiments designed to test PBL activity in the presence of NK
receptor specific monoclonal antibodies. Example 1
In order to make the desired tetramers, it was first necessary to prepare constructs which
would encode modified HLA-A*0201 molecules. To do this, total RNA was extracted from
HLA-A*0201 positive cells, and HLA-A*0201 was then cloned, using specific primers for the
molecule, and reverse transcription polymerase chain reaction (RT-PCR). Altman et al, Science
274: 94-96 (October 4, 1996) but with a new 3' primer, i.e. 5'-
GCAGGATCCCGGCTCCCATCCTCA GGGTGAGGGGC-3' (SEQ ID NO: 48) incorporated
by reference, was followed. Simultaneously with the RT-PCR, the amino terminal nucleotide
sequence was altered to optimize protein expression in the vector used. See Garboczi et al,
Proc. Natl. Acad. Sci. USA 89: 3429 (1992) incorporated by reference. Once this was done, the
extracellular coding portion of the molecule was amplified, again using specific primers. The
resulting construct was recloned into a vector which would produce a BirA biotinylation
recognition site in frame at the 3 '-end of the HLA-A'0201 heavy chain. The modified HLA-
A*0201 and B2 microglobulin were overexpressed in separate E. coli cultures. The resulting
inclusion bodies were purified and the HLA and B2 microglobulin recombinant proteins were
solubilized into urea, and then refolded, in a refolding solution, at 4 °C to form complexes. (The
refolding solution contained 100 mM Tris, at pH 8.0, L-arginine, 400 mM, EDTA, 2 mM,
reduced glutathione, 5 mM, oxidized glutathione, 0.5 mM, PMSF, 0.1 mM, HLA heavy chain,
and β2 microglobulin 1 μM, and 10 μM of the peptide of interest). The refolding solution was
concentrated to 7.5 ml, using standard techniques. Then, refolding buffer was exchanged with
BirA reaction buffer (Tris 100 mM, pH 7.5, NaCl 200 mM, Mg Cl2 5mM, PMSF 100 μM,
leupeptin 1 μM, and pepstatin 1 μM), the last three being added immediately before use. The complexes were then biotinylated with biotin holoenzyme synthase (the BirA
enzyme) by combining the refold mix containing the HLA-A2 complex with 50 μM enzyme,
100 mM biotin in 200 mM Tris, and 100 mM adenosine triphosphate. The mixture was
incubated overnight at room temperature. The biotinylated complexes were then purified, and
combined with phycoerythrin-labelled streptavidin, to produce tetrameric structures. These were
isolated, and reconstituted in small volumes, at a concentration of 1 mg/ml.
Example 2
Following isolation and reconstitution of the tetramers, these were tested to determine
if they could identify CTLs which express the cognate T cell receptor. To test this, monoclonal
CTL populations specific for SEQ ID NOS: 2, 4 and 5 (see infra), were used. In these
experiments, tetramers were prepared, using SEQ ID NOS: 3, 4 and 5 (see infra, but not SEQ
ID NO: 2) in a two color fluorescence assay with anti-CD8 antibodies, labelled with FITC. The
tetramers uniformly stained the CTLs, providing a signal two orders above background. The
specificity of CTL clone staining correlated well with the specificity of lytic activity displayed
by the same clones against 51Cr labelled T2 cells sensitized with SEQ ID NO: 2, 3, or 4, infra-
Example 3
The peptides, which were used in examples 1 and 2, i.e.:
Glu Ala Ala Gly He Gly He Leu Thr Val (SEQ ID NO: 2)
Glu Leu Ala Gly He Gly He Leu Thr Val (SEQ ID NO: 3)
Tyr Met Asp Gly Thr Met Ser Gin Val (SEQ ID NO: 4) and
Gly He Leu Gly Phe Val Phe Thr Leu (SEQ ID NO: 5) are all known to bind to HLA-A2 molecules, and stimulate CTLs. SEQ ID NO: 2 is derived
from Melan-A; SEQ ID NO: 3 is an analogue of SEQ ID NO: 2; SEQ ID NO: 4 is derived from
tyrosinase 2 (Skipper et al., J. Exp. Med. 183: 527 (1996)); SEQ ID NO: 5 is a control peptide
from influenza matrix protein (Gotch et al., 1987, Nature 326; 881). Hence, they were used in
the formation of the tetramers discussed supra. The tetramers were then used to detect specific
T lymphocytes in tumor infiltrated lymph nodes ("TILNS" hereafter).
TILNS were surgically removed from ten melanoma patients, who had been typed as
HLA- A* 0201 positive. The TILNS were treated in the same manner as the TILNS in Romero
etal.,J. Immunol. 159: 2366-2374 (1997), incorporated by reference. Control lymph nodes from
normal lymph nodes ("NLNs" hereafter) were also prepared. The TILNs were treated to form
single cell suspensions, and were either not cultured, or cultured for anywhere from 1 -22 days
for expansion of specific T lymphocytes in medium supplemented with rIL-2 and rIL-7, and then
samples containing from 0.5-l.OxlO6 were stained with fluorescent, anti-CD3, anti-CD8
antibodies, as well as the tetramers described supra.
The anti-CD3 antibody was labelled with PerCP (peridinin chlorophyll protein). The
anti-CD8 antibody was labelled with FITC. A total of 1-2 ug of the tetramer being tested, in 20
ul volumes of PBS, 0.02 mM, 0.2% bovine serum albumin, 0.2% sodium azide, was used. It was
combined with the cells, for 30 minutes, at 4-8 °C. The cells were washed once, with 3 ml of the
same buffer, and were then analyzed immediately via flow cytometry.
A surprising high number of lymphocytes (0.12 to 21% of CD3+ and CD8+ lymph node
cells) specific for complexes of SEQ ID NO: 2 or 3 were found, in 11 of the 12 TILN samples
tested. The binding lymphocytes were CD3+ CD8+ lymph node cells. There were also high numbers of TILN cells which bound to the tetramers containing SEQ ID NO: 4, ranging from
0.14 to 0.63%.
Example 4
The samples discussed supra were analyzed further, emphasis being placed on TILN
samples which had been incubated overnight (5 samples) or stained immediately (2 samples).
Of these, the levels ofSEQ ID NO: 3 tetramer binding lymphocytes ranged from 0.12 to 1.29%.
There were no SEQ ID NO: 4/HLA-A2 positive TILNs found in these samples which had been
cultured overnight. This suggests that highest levels of positive lymphocytes are achieved after
short term in vitro culture.
Example 5
In order to confirm the suggestion outlined supra, cell suspensions were prepared from
a heavily tumor infiltrated lymph node and a normal lymph node ("NLN") from the same patient.
The suspensions were cultured, as described supra, overnight, for 15 days, or for 21 days and
analyzed for tetramer staining as described supra. Tetramer positive cells rose from 0.12 to
12.2% in the TILNs, but from 0.14 to 0.81 % in the NLNs when the 21 day cultured lymph node
cells were used. What was more drastic, however, was the absolute number of tetramer positive
cells. Specifically, the apparent number of SEQ ID NO: 3/HLA-A2 tetramer positive CD3"
CD8+ cells rose from 210 to about 1.8x105 after 15 days, and 5.38x 105 after 21 days, representing
861 fold and 2562 fold expansions. Minor but significant expansion was seen in the NLNs, but
at two orders of magnitude less. Example 6
Studies were carried out to determine relationships between those TILN lymphocytes
which bind the tetramers, and CTL effectors capable of killing tumor cells expressing Melan A
antigen and to obtain highly homogenous populations of such CTL effectors.
To do so, TILN cells from a patient were cultured, for 21 days, as described supra, using
recombinant IL-2 and IL-7, and were stained with anti-CD3, labelled withperidinin chlorophyll
protein, and anti-CD8, labelled with FITC. These were then sorted into Melan A HLA-A2
tetramer positive, and negative cells, as described supra. Chromium release assays (Romero et
al. 1997, J. Immunol. ]_59: 2366) were carried out which showed that only the Melan- A/HLA-A2
tetramer positive T lymphocytes were able to kill T2 target cells sensitized with the peptide of
SEQ ID NO: 3.
Example 7
To further characterize the functional antigen specificity, TILNs which were sorted as
in example 5 were expanded for two weeks via non-specific stimulation with mitogen PHA, and
allogeneic, irradiated feeder cells. The sorted, expanded TILNs in a 51Cr release assay as
described supra against non-melanoma HLA-A2 expressing T2 cells, which had been sensitized
with one of seven peptides known to bind to HLA-A2 molecules, and to generate CTLs. The
assay was carried out under the conditions in example 6, supra.
It was found that lymphocytes which were specified to the tetramers containing the
decapeptide of SEQ ID NO: 3 only recognized this peptide. In contrast, tetramer negative cells
did not recognize any of the peptides. It was also observed that the Melan-A specific cells did in fact kill the autologous tumor
cell line efficiently.
Example 8
Apart from the importance of being able to visualize antigen specific CTLs, as is shown
supra, it is important to determine if these CTLs have encountered the antigen in vivo. Prior
work has established that expression of several antigens, including CD25, CD69, and CD45RO
are associated with T cell activation in CD8+ lymphocytes. These first two markers are
expressed transiently, while CD45RO is expressed in a more stable manner. Hence, the staining
method described supra was carried out, using anti-CD8 antibodies labelled with peridin
chlorophyll protein, anti-CD45RO, labelled with FITC, and the tetramers described supra. Ex
vivo cells suspensions were prepared from two patients, and then studied, with a tumor
infiltrated lymph node being tested with an unfiltrated node used for comparison.
It was found that the tetramer positive lymphocytes were predominantly, if not
exclusively in the CD45RO+, CD8+ lymphocytes. This indicated that such lymphocytes had
encountered antigen m vivo and were therefore being activated at the tumor site.
Example 9
In view of the experiments described supra, additional work was carried out to determine
if the tetramers could be used to isolated complex specific CTLs from circulating lymphocytes.
To do this, cryopreserved peripheral blood lymphocytes were thawed (37°C, 3 minutes), then
washed twice with standard culture medium. Magnetic cell sorting was carried out to select
CD8T cells, following standard techniques. The cells were then analyzed, using CD45RA" specific antibodies labelled with cytochrome, and CD28 labelled with FITC, following the
protocols of example 8. A total of 0.11% of the CD8+ PBLs were tetramer positive and, most
notably, most were CD28+. Approximately 44% of the CD8", tetramer specific cells, were
CD45RA\ This shows that uncultured, unstimulated blood lymphocytes can be assayed for
appropriate CTLs specific for the complex of interest.
Example 10
Further analysis were carried out with the tetramers of SEQ ID NO: 4. These stained
CTLs known to be specific for complexes of SEQ ID NO: 4 and HLA-A2, but not others. The
tetramer was then used to study PBMCs of individuals who were not known to be suffering from
influenza infection, to determine if the tetramers could be used to assay for low frequency CTLs,
like memory cells, ex vivo. The assay was carried out, as described in examples 8 and 9, supra.
The percentage of complex specific CD8+ cells was considerably higher than the
percentage estimated previously using limiting dilution analysis, but are similar to the percentage
observed, using ELISPOT analysis, as per Lalvani et al., J. Exp. Med. 186: 859-865 (1997),
which is incorporated by reference.
Example 11
Studies were then carried out to phenotype the complex specific, CD8+ cells. A large
number of PBMCs were obtained from an individual and, using the assay of example 10, supra,
the frequency of CD8+ cells specific for the complexes was consistently found to be about
1/3700 of the PBMCs, and 1/2500 of small lymphocytes which are activated, or "memory" CD8"
cells. When tested for cell surface markers, these were found to be CD45RO7CD45RA , consistent with such cells having previously encountered antigen. At least 74% of the positive
CTLs expressed the VB17 chain, as determined by cell surface staining, as compared to 4% of
the negatives, which confirms results shown by Lehner et al., J. Exp. Med. 181: 79-91 (1995).
Only 36%o of the positive cells were CD28+, compared to 75% of the negative CTLs. Again, this
is consistent with the majority of the cells having reduced proliferative potential due to longer
replicative history. This shows that, using the triple staining methodology, a very pure cell
population can be obtained, by applying, e.g., FACS on peripheral blood lymphocytes, thereby
identifying CTLs.
Example 12
Specificity and activation of the CD8+ PBMCs discussed supra was studied using the
ELISpot technique, referred to supra. This technique yields spots of color whenever a CTL
releases gamma interferon. In control experiments, a CTL clone specific for SEQ ID NO:
4 HLA-A2 complexes was stained with the tetramer and anti-CD8. Then, 100 double positive
cells were sorted directly into duplicate wells of ELISPOT plates containing T2 cells, which had
been prepared with either SEQ ID NO: 5, or as a control, SEQ ID NO: 2. No gamma interferon
was released from controls, indicating that tetramer staining did not per se activate the cells. The
SEQ ID NO: 5 containing matrices, however, provoked gamma interferon release corresponding
to about one spot per CTL per day, indicating all cells had been activated by their cognate
peptide.
Following this, the PBMCs were stained, sorted into sets of 15 cells and duplicate
ELISPOT wells for 1 or 2 days of incubation with peptide pulsed targets. Interferon release was
observed only when the peptide of SEQ ID NO: 5 was used. Pulsing with other peptides did not result in gamma interferon release, nor did tetramer negative CD8+ cells release IFN-gamma on
exposure to any peptide.
Since the number of spots formed was close to the number of PBMCs in the wells, this
suggests that the majority of the tetramer positive, CD8+ cells were capable of responding within
one day, thus supporting the concept that most circulating memory CD8+ cells are capable of
rapid effector function when triggered by exposure to cognate peptide in the absence of cytokine,
even when the presenting cells are not professional antigen presenting cells.
Example 13
Studies were then carried out to confirm the specificity of tetramer specific, CD8"
specific PBMCs, and to study proliferative potential. To do this, single PBMCs were sorted
directly into cloning wells. After two weeks, 15 of 60 wells seeded with these cells contained
proliferating blasts. The four most proliferative clones were tested in duplicate ELISPOT assays
for specificity, along with one proliferating CD 8', tetramer negative clone. What was found was
that all four of the CD8+, tetramer positive clones synthesized gamma interferon when exposed
to targets pulsed with SEQ ID NO: 4, but not when pulsed with a control peptide. The tetramer
negative, CD8+ cells failed to respond at all. Further, one of the clones was tested in a chromium
release assay, where it specifically lysed target cells pulsed with SEQ ID NO: 5.
Example 14
The potential of tetramer based, FACS sorting in analyzing tumor specific CTLs
responses was studied using tetramers containing SEQ ID NO: 2, as described supra. The
tetramer was then used to stain and sort a polyclonal CTL line, which had been generated from an HLA-A2", melanoma infiltrated lymph node. Only a small percentage of cells (6%), could
be double stained with the specific tetramers and antibody to CD8. The polyclonal line did kill
A2 matched targets pulsed with SEQ ID NO: 2, although it was poor at killing A2 matched
melanoma lines expressing Melan-A, and demonstrated small background killing of target cells
pulsed with a negative control.
An aliquot of the polyclonal line was then enriched for tetramer specific, CD8 cells by
FACS sorting. This line killed melanoma lines and target pulsed with SEQ ID NO: 2 much more
efficiently than the original polyclonal line, and showed no background killing. The specific
cytotoxicity correlated entirely with the percentage which stained tetramers, indicating that
tetramer staining, followed by FACS sorting, can be used to generate highly effective,
tumoricidal, CTL lines.
Example 15
These experiments describe in vitro expansion of Melan A specific precursors from
PBMCs of melanoma patients.
Samples of PBMCs were taken from seven melanoma patients, using standard
techniques, and were enriched for CD8+ lymphocytes via magnetic cell sorting, to give
populations of cells which were over 99% CD37CD87
These cells were then cultured, overnight, under standard conditions, and were then
stained with tetramers of HLA-A* 0201 and SEQ ID NO: 3, which were labelled with
phycoerythrin, and anti-CD8 monoclonal antibodies (Staining was accomplished by combining
the cells with 20μl of PBS with 2% FCS, and incubating for 15 minutes at 4°C, after which 20μl
of anti-CD8 monoclonal antibodies labelled with FITC were added, and incubated for 30 minutes at 4°C). Cells were then washed, once, with the buffer described supra, and analyzed
by standard flow cytometry.
The frequency of tetramer positive cells ranged from 0.03% to 0.09% of the total CD8+
population. These frequencies were close to the detection limit of the flow cytometer, which led
to the next experiments.
Example 16
In view of the low levels of positive cells found, PBMCs were taken from 3 of the 7
melanoma patients described supra, and were stimulated three times, at weekly intervals, with
either SEQ ID NO: 2 or SEQ ID NO: 3 pulsed, autologous PBMC cells. To elaborate, the
samples were stimulated, initially, by adding 1 μM of peptide directly into the medium. Cultures
were then stimulated weekly by adding autologous PBMCs (3x106 cells/well), which had been
pulsed for 2 hours at 37°C, in serum free medium, with lμM of the peptide of SEQ ID NO:2 or
SEQ ID NO: 3 and 3μg/ml of human β2 microglobulin. These peptide pulsed cells were washed
extensively, irradiated (3000 rad), and adjusted to an appropriate volume before being added to
the responder cell population. Both IL-2 ( 1 OU/ml), and IL-7 ( 1 Ong/ml), were added during the
first two stimulation cycles, and IL-2 alone was added thereafter, at 100 U/ml.
The assay described in example 15 was carried out on the samples 7 days after each
stimulation cycle.
During the first week of stimulation, the total number of tetramer positive cells declined
consistently, increased moderately after the second stimulation, and significantly after the third
one. Stimulation with the peptide of SEQ ID NO: 3 resulted in more vigorous expansion than
did stimulation with SEQ ID NO: 2. The difference in stimulatory capacity was apparent after the first cycle and became very marked after the third one. The results are summarized in the
following table.
Figure imgf000023_0001
Example 17
Cultures were tested seven days after the third stimulation cycle for their capacity to lyse
T2 cells, in the absence or presence of each of the two peptides. (T2 cells are known as cells
which are HLA- A* 0201 positive, are defective in antigen processing ability, but present
exogenously supplied peptides effectively). The assays were carried out by labelling the T2 cells
with 51Cr for 1 hour at 37°C, followed by two washes. Labelled target cells (1000 cells in 5 Oμl)
were then added to varying amounts of effector cells (in 50μl) in V-bottom microwells, either
in the presence of 1 μg/ml of the peptide of SEQ ID NO: 2, 1 μg/ml of the peptide of SEQ ID NO:
3, or no peptide.
The effector cells had been preincubated for at least 20 minutes at 37 °C in the presence
of unlabelled K562 cells (50,000/well) to eliminate non-specific lysis due to NK-like effectors
in the stimulated T cell populations. Chromium release was measured after 4 hours. The results are presented in figure 1. Frequency of HLA-A*0201/SEQ ID NO: 3
tetramer positive cells detected in the cultures correlated directly with peptide specific
cytotoxicity. Also, CTLs which were generated following stimulation with SEQ ID NO: 3 cross
recognized SEQ ID NO: 2.
Example 18
Additional experiments were carried out to document the specificity of CD8+, HLA- A*
0201 /peptide tetramer positive lymphocytes which had been generated supra.
In these experiments, PBMCs from one of the three patients described supra were
stimulated with autologous PBMCs that had been pulsed with SEQ ID NO: 1, SEQ ID NO: 2,
SEQ ID NO: 3, the peptide Ala Ala Ala Gly He Gly He Leu Thr Val (SEQ ID NO: 37), Ala Leu
Ala Gly He Gly He Leu Thr Val (SEQ ID NO: 38), or an immunodominant HLA-A2 restricted
epitope from influenza matrix protein, presented as SEQ ID NO: 5, supra. (SEQ ID NOS: 37
and 38 are Melan A analogues, and hence are related to SEQ ID NOS: 1,2 and 3). The
stimulation protocol was identical to that given in example 17, supra.
A small number of HLA- A* 0201/ peptide tetramer positive cells were detected at the
end of the first stimulation cycle in the cultures stimulated using the influenza peptide, and was
equivalent to the number of such cells detected in uncultured cells. The number declined after
a second round of stimulation.
Expansion of the tetramer positive lymphocytes occurred with both SEQ ID NOS : 1 and
2, directly demonstrating the ability of the peptides to stimulate in vitro expansion of Melan-A
specific lymphocytes in peripheral blood. Their stimulatory capacity appears to be similar. Notwithstanding this point, the increase in proportion of tetramer positive lymphocytes
wasmuchmorepronouncedwhenanalogues ofSEQIDNO: 1 were used. The largest expansion
occurred with SEQ ID NO: 38. The following table summarizes these results. The values show
the percentage of cells which were positive both for the tetramer discussed supra, and the FITC
labelled, anti CD8 antibody.
Peptide
Figure imgf000025_0001
Example 19
Following the work in example 18, the viable cells recovered after the third stimulation
were titrated as effectors in a 51Cr release assay of the type described supra.
Frequency of tetramer positive cells correlated directly with the level of specific
cytotoxicity, and CTLs resulting from the analogue driven expansion effectively lysed target
cells sensitized with SEQ ID NO: 2.
Example 20
Examples 15-19 show that it is possible to directly visualize antigen specific T
lymphocytes using flourescent tetramers, and to separate them from bulk cultures. They suggest the possibility of early stage, in vitro stimulation with peptide, and continued expansion in
antigen independent fashion. This was tested in the experiments which follow.
Samples of CD8+, tetramer positive cells were isolated from each of the cultures
described in example 18, seven days after the second cycle of stimulation. These sorted cells
were then expanded in vitro by stimulation with phytohemagglutinin, and the resulting cells were
tested for their lytic activity on T2 cells, in the same manner described supra, in the absence or
presence of the peptide used in the particular culture from which the cells were taken originally,
or SEQ ID NO: 2. Concurrently, the ability of these CD8+ cells to lyse tumor cells was tested,
using Melan-A positive tumor cell line Me 290 as the target.
The results, presented in figure 2, show that all the cell populations exhibited a high
degree of specific lysis against target cells which had been pulsed with the corresponding
stimulatory peptide and with cells pulsed with SEQ ID NO: 2. They also exhibited high
tumoricidal activity. Me 290 is illustrative of additional Melan-A positive tumor cell lines which
were lysed. Melan-A negative tumor cell lines were not lysed. Effectoπtarget ratios necessary
for achieving half maximal lysis ranged from 3:1 to 15:1.
Example 21
The relative avidity of peptide antigen recognition of the different cell populations which
had been obtained was assessed in CTL assays, following standard protocols. Specifically, 1000
target cells (50μls) were incubated with varying concentrations of peptides (50μl) for 15 minutes
at room temperature before effector cells were added. The concentration of each peptide
required to achieve 50% maximal lysis of target was determined, using standard methods. To
make comparison easier, relative activity of peptide was calculated as the 50% concentration of SEQ ID NO: 1, divided by the 50% concentration of the tested peptide. Figure 3 shows these
results. These results are also presented in the table which follows. The values are the peptide
concentrations required for 50% maximal activity.
The relative avidity of the different lines for SEQ ID NO: 1 and SEQ ID NO: 2 were
remarkably similar, except for the line obtained using SEQ ID NO: 39, which recognized SEQ
ID NO: 1 about 3 fold less efficiently, and SEQ ID NO: 2 5-7 fold less efficiently as compared
to other lines.
The results are also presented in terms of relative antigenic activity. Note that, regardless
of the peptide used for in vitro expansion, all lines recognized SEQ ID NO: 2 more efficiently
than SEQ ID NO: 1. Analogues were recognized more efficiently than both parental sequences
by all lines, notwithstanding some differences in relative antigenicity.
Preference of a line for a certain analogue did not correlate with the analogue used to
generate the line: in all cases, SEQ ID NO: 38 was the peptide most efficiently recognized.
Culture stimulated with:
Melan-A Melan-A E26A A27L E26A/A27
27-35 26-35 L
(SEQ ID (SEQ ID (SEQ ID (SEQ ID (SEQ ID NO: 1) NO: 2) NO: 37) NO: 3) NO: 38)
Peptide [nM] 50%:
Melan-A 27-35 (SEQ ID NO: 1) 27 22 18 37 65
Melan-A 26-35 (SEQ ID NO: 2) 3 3 5 5 20
E26A (SEQ ID NO: 37) 0.09 0.14 0.12 0.19 1 Melan-A Melan-A E26A A27L E26A/A27
27-35 26-35 L
A27L (SEQ ID NO: 3) 0.15 0.2 0.2 0.16 0.4
E26A/A27L (SEQ ID NO: 38) 0.02 0.01 0.008 0.005 0.5
Relative antigenic activity:
Melan-A 27-35 (SEQ ID NO: 1) 1 1 1 1 1 Melan-A 26-35 (SEQ ID NO: 2) 9 7 4 7 3
E26A (SEQ ID NO: 37) 300 157 183 194 65 A27L (SEQ ID NO: 3) 180 110 110 231 162 E26A/A27L (SEQ ID NO: 38) 1350 2200 2750 7400 130
Example 22
The peptide presented supra as SEQ ID NO: 4 is derived from the tyrosinase molecule,
which is known to function as a tumor rejection antigen precursor, with the peptide of SEQ ID
NO: 4 functioning as a tumor rejection antigen. See, e.g., Brichard, et al., J. Exp. Med. 178:489
(1993); Wδlfel, et al., Eur. J. Immunol. 24:759 (1994). Work by others has focused on study of
the binding properties of an analog of this peptide, i.e., one where asparagine at position 3 of the
peptide, has been replaced by aspartic acid. (See SEQ ID NO: 22, supra). This modification
occurs naturally, via post transiational processes. Skipper, et al., J. Exp. Med. 183:527 (1996)
have shown that this difference significantly affects T cell recognition. The experiments which
follow are directed to a determination of what role single amino acid side chains contribute to
the interaction between SEQ ID NO: 4 and HLA-A* 0201 molecules.
First, single alanine substituted derivatives of SEQ ID NO: 4 were prepared, i.e.: Ala Met Asp Gly Thr Met Ser Gin Val (SEQ ID NO: 39)
Tyr Ala Asp Gly Thr Met Ser Gin Val (SEQ ID NO: 40)
Tyr Met Ala Gly Thr Met Ser Gin Val (SEQ ID NO: 41)
Tyr Met Asp Ala Thr Met Ser Gin Val (SEQ ID NO: 42)
Tyr Met Asp Gly Ala Met Ser Gin Val (SEQ ID NO: 43)
Tyr Met Asp Gly Thr Ala Ser Gin Val (SEQ ID NO: 44)
Tyr Met Asp Gly Thr Met Ala Gin Val (SEQ ID NO: 45)
Tyr Met Asp Gly Thr Met Ser Ala Val (SEQ ID NO: 46)
Tyr Met Asp Gly Thr Met Ser Gin Ala (SEQ ID NO: 47)
These peptides were synthesized using standard methodologies, and were then tested in
a functional competition assay. To elaborate, the assumption was that these peptides would bind
to HLA-A*0201 molecules. It is known that SEQ ID NO: 1 binds to HLA-A*0201. Hence, T2
cells, described supra, were labelled with 51 Cr in the presence of a well known monoclonal
antibody, i.e., W6/32, which binds MHC - Class I molecules.
Then, varying concentrations of test peptides SEQ ID NOS: 39-47 were added, in 50 μl
aliquots, to the labelled T2 cells (1000 cells/well), for 15 minutes at room temperature, after
which a suboptimal dose of SEQ ID NO: 1 (125nM in 50 μl), was added, together with 5000
cells/well of an HLA-A*0201/SEQ ID NO: 1 restricted CTL clone, as taught by Valmori, et al.,
J. Immunol 160: 1750 (1998). Chromium release was measured after 4 hours of incubation at
37 °C. The concentration of each competitor peptide required to achieve 50% inhibitions of target
cell lysis was determined. This is indicated in the Table which follows, as [nM] 50%. The
comparison is facilitated by presenting relative competitor activity as [nM] 50% of SEQ ID NO: 4. This peptide' s activity is assigned arbitrary competitor activity of 1. As a control, an
influenza matrix peptide was used.
It was found that SEQ ID NO: 4 displays intermediate competitor activity, i.e., to secure
50%) of maximal lysis, a concentration of lOOnM was needed, as compared to 6nM for SEQ ID
NO: 5, known to be a high affinity binder. It was surprising that changes at putative anchor
positions 2 and 9 had marginal impact. Changes at positions 7 and 8, for example, had a much
greater impact.
IΛJD 5545.1-JEL/NDH
Table
Binding of single A substituted Tyrosinase 368-376 peptide variants to HLA-A* 0201
Peptide Sequence Competitor Relative
activity competitor
[nM] 50% activity
G
Tyrosinase 368-376 YMDGTMSQV (SEQ ID NO: 4) 102
CZ5
1 H— H analogues AMDGTMSQV (SEQ ID NO: 39) 200 0.5
H YADGTMSQV (SEQ ID NO: 40) 300 0.3
H
W s YMAGTMSQV (SEQ ID NO: 41) 90 1.1
X YMDATMSQV (SEQ ID NO: 42) 100 1.0 w m YMDGAMSQV (SEQ ID NO: 43) 80 1.3
H YMDGTASQV (SEQ ID NO: 44) 200 0.6 d YMDGTMAQV (SEQ ID NO: 45) 50 r 2.0 w YMDGTMSAV (SEQ ID NO: 46) 30 3.3 t YMDGTMSQA (SEQ ID NO: 47) 80 1.3
Figure imgf000031_0001
Influenza A matrix 58-66 GILGFVFTL (SEQ ID NO: 5) 17
Example 23
It had been observed previously that complexes formed between HLA- A* 0201 molecules
and SEQ ID NO: 2 are unstable, dissociating completely in under one hour when incubated at
37°C. Stability studies were carried out on complexes of HLA-A*0201 and SEQ ID NO: 4. In
brief, T2 cells were loaded with saturating concentrations (lOμM) of one of SEQ ID NO: 4,
SEQ ID NO: 2, or control peptide SEQ ID NO: 5. Following overnight incubation with the
peptides and β2-micro-globulin (3μg/ml) in serum free medium, the T2 cells were treated with
ementine (104 M) to inhibit protein synthesis, and incubated at 37 °C. The cells were stained,
at different time periods, by incubating an aliquot with anti-HLA-A2 specific monoclonal
antibody BB7.2, to measure HLA-A2 antigen expression. The results were expressed as relative
complex stability, defined by the formula:
[Mean fluorescence test peptide - background mean fluorescence] [Mean fluorescence SEQ ID NO: 48 - background mean fluorescence]
"Background" refers to fluorescence values obtained on T2 cells treated similarly but for absence
of exogenous peptide.
Figure 4 presents these results. They confirm the instability of HLA-A2/SEQ ID NO:
1 complexes, and show that the complexes of HLA-A2 and SEQ ID NO: 4 are stable over a 6
hour period.
Example 24
It has been observed, by Romero, et al, J. Exp. Med. 188:1641 (1998) incorporated by
reference, and in the preceding examples, that significant numbers of HLA-A2/SEQ ID NO: 4 positive, CD8+ T lymphocytes can be detected in short term cultured TILNs from HLA-A*0201
positive melanoma patients. Studies were carried out to assess the antigenic and functional
specificity of such positive lymphocytes.
In brief, positive and negative fractions were separated via flow cytometry sorting, using
tetramers of HLA-A2/SEQ ID NO: 4, and CD8 specific monoclonal antibodies, as described
supra. Following separation, cells were expanded by stimulation with PHA and irradiated
allogeneic PBMCs, as described supra, over a two week period. The fractions were tested for
their ability to lyse 51Cr labelled T2 cells in the absence or presence of 4μM of SEQ ID NO: 4.
The CD8~, tetramer positive fraction lysed the T2 cells in the presence of SEQ ID NO: 4, but not
its absence. Negative fractions did not lyse the target.
Example 25
These experiments were designed to obtain monoclonal T cell populations specific for
SEQ ID NO: 4 containing complexes. To do this, limiting dilution cultures were set up
immediately after the cell sorting described in example 24. Limiting dilution was carried out
following Valmori, et al., J. Immunol. 160:1750 (1998), incorporated by reference, using
irradiated allogeneic PBMCs, EBV transformed B lymphocytes, PHA and recombinant IL-2.
Six specific CTL clones were secured, and a seventh clone was obtained from
unfractionated TILNs after in vitro stimulation in an independent cloning experiment. These
constitute monoclonal T cell populations.
Example 26 These experiments were designed to analyze both polyclonal, monospecific T cell
populations, and the monoclonal T cell populations described supra, for tumor antigen
recognition, avidity, and fine specificity. This example describes the antigen recognition
experiments.
Three cell lines were used. NA8-MEL is an HLA-A*0201 positive melanoma cell line
which does not express tyrosinase. NA8-MEL tyr" is a cell line derived by transfecting NA8
with cDNA encoding tyrosinase. Cell line Me 290 described supra as an HLA-A* 0201 positive,
Melan-A+ cell line is also tyrosinase+.
Samples of each of these lines were combined with one of a polyclonal, monospecific
CTL population obtained by using the tetramers of SEQ ID NO: 4 and HLA-A* 0201, supra, or
the monoclonal T cell populations described supra.
The melanoma cell lines and T cells were combined, at varying ratios, either with or
without the peptide, at lμM concentration. The tumor cells had been labelled with 51Cr prior to
combining them with the T cells. 51Cr release was measured after 4 hours of incubation, using
the same parameters as are described supra.
These results are set out in figure 5. The panel labelled Tyr. tetr+ shows data from the
polyclonal cell population. The others are the monoclonal lines described supra.
All of the clones and the polyclonal population lysed the NA8-MEL cell line, which is
tyrosinase negative, only when the peptide of SEQ ID NO: 4 was added. The other lines, i.e.,
Me 290 and NA8-MELtyr+ were lysed equally well, both in the presence of the peptide and
without it.
Example 27 Fine antigenic specificity of the T cell populations discussed in example 26 was analyzed,
by quantitating the relative antigenic activity of the variants set out in SEQ ID NOS: 39-47 in
standard CTL assays. These were carried out by using a 5ICr release assay, as described supra.
Relative avidity was determined by comparison to the concentration of the peptide of SEQ ID
NO: 4 required to obtain half maximal lysis at an effector: target ratio of 10:1. The results are
presented in the following Table.
LUD 5545.1-JEL/NDH
Figure imgf000036_0001
LUD 5545.1-JEL/NDH
45 YMDGTMAQV O.0004 <0.003 <0.0004 0.04 O.003 <0.02 <0.00 <0.015
2
46 YMDGTMSAV 0.05 <0.003 O.0004 1 0.05 <0.02 <0.00 100
2
47 YMDGTMSQA 0.06 O.003 0.05 1 0.5 0.5 0.2 0.03
GO c
GO
H
H Cl H m w
U1
GO
X w
H
Figure imgf000037_0001
Each clone displayed unique fine specificity. Replacement of residues at positions 2 and
9 did not drastically affect antigen recognition in the majority of cases, which is consistent with
lack of direct action between side chains located at these positions, and T cell receptors, as
suggested by Madden, et al., Cell 75:693 (1993). The results indicate that position 7 of the
peptide (a Ser residue), was important, while replacement at different positions gave highly
variable results. The net result is a showing of wide heterogeneity in the recognition of epitopes
by CTLs specific to SEQ ID NO: 4, and derived from a single, infiltrated lymph node.
Example 28
Example 15, supra, showed how tetramers of HLA-A* 0201 and a Melan-A derived
peptide could be used to measure the frequency of CD8+ T lymphocytes in a population. Similar
experiments were carried out using tetramers of HLA-A* 0201 and SEQ ID NO: 4.
The same protocol as was used in example 15, but for a different tetramer, was carried
out on highly enriched CD8+ T cells from PBMCs of 10 HLA-A* 0201 positive melanoma
patients. Seven samples showed frequencies of tetramer+ cells ranging from less than 0.01 % and
up to 0.03%). These ranges were not very much above the level of detection afforded by the flow
cytometry assay, so the enriched, CD8+ T lymphocytes for all 10 samples were stimulated with
SEQ ID NO: 4, using the protocol of example 16. supra, with minor modifications. Specifically,
cells were initially stimulated with 100 μM of peptide directly into the culture medium, and
weekly stimulations were carried out with additional lOOμM samples of peptide. Additionally,
IL-2 (100 U/ml) and IL-7 (10 ng/ml) were added during the first stimulation cycle, and IL-2
alone (100 U/ml) thereafter. Cultures were monitored seven days after stimulation for the
presence of tetramer positive cells. The results are summarized in the following Table. What is interesting is that positive
lymphocytes were detectable in six out of 10 samples after only short term culture. (The other
four samples were positive, but at the limits of detection for the assay).
Figure imgf000039_0001
The values refer to the percentage of A2/SEQ ID NO:4 tetramer positive T cells, relative to the
total number of CD8+ lymphocytes.
Example 29
In additional experiments, tetramers of HLA-A *0201 molecules and SEQ ID NOS: 2,
4 and now SEQ ID NO: 9, i.e., MAGE-3 derived peptide FLWGPRALV, described supra, were
made, as described in the preceding examples. These tetramers were then used to stain CTLs,
as described in the previous examples. To review, cell samples were stained with PE labelled
tetramer for 15 minutes at 37°C, after which tricolor labelled, anti-CD8" antibodies were added,
on ice, for 15 minutes. Following this, the materials were washed extensively, and then analyzed
by FACS. The tetramers stained CTLs which had previously been identified as being specific for
complexes of HLA-A *0201 and the peptide used, but did not stain with other, non-specific
peptides.
Example 30
These experiments describe staining of different types of samples from different patients.
Five patients were typed for expression of tumor antigens, following standard
methodologies. Different types of samples were taken from these patients, all of whom had
histologically confirmed, malignant melanoma. The characteristics of the patients and samples
involved are set forth in the following table:
Figure imgf000040_0001
Figure imgf000041_0001
Patient L02 was leukapheresed, and PBLs from this patient were cryopreserved
immediately. These PBLS were analyzed after thawing and overnight culture in Iscove's medium
with 5% human serum added. Two additional PBL samples were taken over aperiod of 3 weeks
for comparative analysis.
In the case of the patient referred to as DECH, PBLs were separated from a blood sample,
and cryopreserved. Following thawing, these PBLs were pulsed with lOμM of SEQ ID NO. 9 in "CTL medium" (Iscove's medium, 5% human serum, IL-2 at 100 U/ml), plus IL-7 (lOng/ml).
After this, they were cultured in CTL medium for 2 weeks until they were analyzed.
Patient MM 14 and MM 18 had melanoma - infiltrated lymph nodes removed from the left
or right axillae, respectively, disrupted, and cultured in the CTL medium described supra, plus
IL-7 (lOng/ml). Any proliferating blasts were expanded in the CTL medium for 14 days (for
MM14), or 23 days (for MM18), before analysis.
In the case of MM 15 , a skin metastasis was removed from the left shoulder, mechanically
disrupted, and cultured in the medium described supra for 13 weeks. There was a small
population of TILs, which was stimulated with PHA (5μg/ml), and expanded in medium for two
more weeks before analysis.
Tetramer staining was carried out as described supra, using appropriate tetramers. In
other words, tetramers containing SEQ ID NO. 9 were used in MAGE-3 positive samples from
DECH. Melan-A positive patient samples were screened with tetramers containing SEQ ID NO.
3, while tyrosinase positive patient staples were screened with tetramers containing SEQ ID NO.
4.
Tissue culture of tumor infiltrated lymph nodes of MM 14 revealed CD8^ cells specific
for both the tyrosinase and Melan-A peptides. The cultured LNs from MM18 were positive for
SEQ ID NO. 3 only. Patient L02 had readily detectable CD8+ cells specific to SEQ ID NO: 3
in peripheral blood at a frequency similar to the frequency of influenza specific CTLs in normal
patients, as described by Dunbar, et al., Curr. Biol 8:413 (1998). This frequency remained fairly
constant over the 3 week period of analysis. Patient DECH showed no CD8+ cells specific for
SEQ ID NO: 9 in PBLs on direct analysis; however, two weeks after a single pulse with the
peptide, a small population of cells were detected. Finally, patient MM15 showed very few CD8+ cells in a PHA stimulated culture from skin metastasis; however, a high portion of these
were stained by the tetramers containing SEQ ID NO. 3.
Example 31
The preceding example described how CD8+ cells were identified. In these experiments,
CTLs are cloned, directly, from these CD8+ populations.
To do this, single cells were sorted, directly, into U bottom, 96 well plates coated
previously with anti-CD3 and anti-CD28 antibodies (both at 100 ng/ml), in PBS, containing 105
irradiated B cells in CTL medium, as described supra, plus IL-4 (20ng/ml). The anti-CD3
provides a solid phase mitogenic signal, and the anti-CD28 antibodies were provided in case the
cells were capable of costimulation through CD28.
The cloning plates were incubated at 37°C in 5% CO2 for 10-14 days, without any
manipulation. Then, they were restimulated using PHA (5 μg/ml), together with irradiated,
allogenic PBLs and B cells. All clones were expanded to at least 5xl06 cells, in order to permit
characterization and banking. Several of the clones were stimulated further, in order to test
proliferative potential, and the total potential yield was calculated from the number of CTLs
generated in culture, and the number banked.
The table within follows summarizes these results. Essentially, between 2 and 13% of
sorted clones (average: 6.5%) expanded sufficiently to cryopreserve. Additionally, about three
times as many proliferated to a lesser degree.
The expanded clones were then tested for antigen specificity using either tetramers as
described supra, with confirmance by a 51Cr release assay and ELISPOT analysis. With respect to the 51Cr release assay, this was carried out by T2 cells which had been
pulsed with relevant peptide (SEQ ID NOS: 3, 4 or 9), or a negative control, as well as cell lines
SK-Mel-29 and SK-Mel 23, both of which are HLA-A2.T, and express both Melan-A and
tyrosinase. The lysis percentage was calculated by the formula:
100 x (specific release - spontaneous release) (maximal release - spontaneous release)
Thirty three of thirty four of the clones were positive for their antigen, showing that the direct
cloning was extremely efficient, as compared to methods where the vast majority of cloned cells
do not have the desired specificity. Ten of these were tested by 51Cr release, and three of these
were tested via ELISPOT. All showed specificity for relevant peptide.
With respect to cloning efficiency, no clear differences were seen, although peptide
stimulated cultures required the greatest number of starting lymphocytes to generate clones:
Figure imgf000044_0001
Example 32 The expanded CTLs were then tested for surface phenotytpes via FACS, using FITC
labelled antibodies against CD62L (L-Selectin), CDl lb, CD44, CD45RO, CD45RA, TCRαβ,
and CD 49a-e., inclusive (integrin-Bl-6, VLA 1-6), followed by goat-antimouse Ig. Also, anti-
CLA (cutaneous lymphocyte antigen), and FITC labelled mouse-antirat IgM labelled with FITC,
were used.
All clones tested were CD8+, and TCRαβ^, which was to be expected. They were also
all CD45RO", CD45RA \ CD44+ and CD62L", which is consistent with previous antigenic
stimulation. The lack of CD62L on surfaces suggested that if these clones were infused during
immunotherapy, they would not home to lymph nodes via high endothelial venues, but would
traffic through peripheral circulation. The clones were CD1 lb and CD49d positive, which is
consistent with cells capable of migrating through activated vascular endothelium which express
VCAM-1 - a phenotype associated with metastatic melanoma (Rice, et al., Science 246:1303
(1989); Rice, et al., J. Exp. Med. 171 :1369 (1990)).
A minority of clones were CLA positive suggesting that clones could be selected which
would or would not home to skin following infusion. See Picker, et al., Nature 349:796 (1991);
Ogg, et al., J. Exp. Med. 188:1203 (1998). Hence, it may be possible to pre-select clones for
immunotherapy according to their homing markers, via triple staining for surface molecules prior
to cloning. Also, clones with the best proliferative potential might be pre-selected by only
sorting tetramer + cells which express markers, such as CD28.
Example 33
In this example, and the examples which follow, additional characterization studies were
carried out on both melanoma patients and healthy volunteers. Patients and donors were selected based upon HLA-A2 expression, determined by using
allele specific mAb BB7.2. PBMCs were isolated from samples, using standard methods. The
CD8+ cells in these samples were purified via two rounds of positive selection, using magnetic
beads, as described supra. These cells were greater than 98% CD3+CD8+.
These CD8+ cells were then stained with tetramers of HLA-A*0201 and SEQ ID NO: 3,
SEQ ID NO: 4 or control peptide SEQ ID NO: 5 and anti-CD8+ monoclonal antibodies labelled
with FITC and Cy-Chrome (a tandem flourescent conjugate system) (106 cells added to 50 μl of
PBS containing the antibodies and the tetramers, as well as 2% azide and 2% BSA. The
incubation period was 40 minutes, at a temperature of 4 °C). Following the incubation, the cells
were washed, once, in the buffer used for the binding step, and then analyzed immediately via
FACS.
It was found that CD8+ cells specific to tetramers containing either SEQ ID NO: 3 or
control peptide SEQ ID NO: 5 were present in both melanoma patients and healthy subjects.
CD8+ cells specific to tetramers containing SEQ ID NO: 4 were not found. Note,
however, that these samples had not been stimulated with antigen in vitro. As was shown, supra.
even a short period of in vitro stimulation with antigen was sufficient to cause expansion of CD8*
cells specific to SEQ ID NO: 4 to the point where they could be detected in 6 of 10 patients
tested.
Example 34
A series of experiments were then carried out to determine levels of non-specific, HLA-
A2 tetramer positive CD8+ cells in healthy, HLA-A2 negative patients. The same tetramers as
are described in example 33, supra, were used, and the same protocol for separation and analysis were used. These results are set out in the table which follows For compaπson. tests were also run on samples taken from HLA-A2 positive, healthy donors, HLA-A2 positive melanoma patients, and HLA-A2 positive patients who suffered from both melanoma and viti go.
Percentage of circulating HLA-A2 Melan-A26 3S A27L tetramer and Flu-MA<8 66 tetramer' in CD8~ T cells
Figure imgf000047_0001
The lower detection limit for CD8* cells which reacted with tetramers of SEQ ID NO 3 was about 0.04%, while it was under 0.02% for CD8~ cells positive for SEQ ID NO: 5 containing tetramers These limits of detection are lower than limits of detection for TILS, as shown m Romero, et al, J. Exp. Med. 188: 1641-1650 (1998), due to differences m the type of sample assayed. Of the 13 melanoma and melanoma/vitihgo patients tested, 10 showed significant numbers of HLA-A2/SEQ ID NO 3 positive CD8* cells, while six of ten healthy HLA-A2 positive donors showed significant numbers as well Note that these CDS" cells were almost all naive cells, as compared to the mix of naive and antigen experienced cells found in
melanoma patients.
Example 35
Prior work has established that the CD45RA and CD45RO isoforms can be used to
identify naive and memory T cells, respectively. See Young, et al., Eur. J. Immunol. 27: 2383-
2390 (1997), incorporated by reference. It has also been shown that circulating, CD28" CD8+ T
cells present direct, ex vivo cytolytic activity (Azuma, et al., J. Immunol. 150: 1147-1159 (1993),
and Hamann, et al., J. Exp. Med. 186: 1407-1418 (1997), have proposed that such cells
correspond to effector type CTLs. In view of these prior observations, individuals who presented
significant amounts of circulating cells positive for tetramers of SEQ ID NO: 3 or SEQ ID NO:
5 were phenotyped for CD28, CD45RA, and CD45RO.
To do this, CD8+ lymphocyte populations were purified, as described supra. The highly
purified samples were divided into two, and then contacted with one of the tetramers described
supra, and with antibodies against CD45RA labelled with Cy-Chrome, and either antibody
against CD45RO labelled with FITC, or antibody against CD28 labelled with FITC. The
procedures for carrying out the staining are described, supra, and are not repeated here.
Virtually all circulating CD8+ T cells from healthy donors were positive for CD28, and
CD45RA, and negative for CD45RO when the cells were positive for tetramers of SEQ ID NO:
3. In contrast, CD8+ T cells which were positive for SEQ ID NO: 5 tetramers were CD28 and
CD45RO positive, and CD45RA negative (i.e., CD45RAlow). This suggests that the CD8" T cells
specific for SEQ ID NO: 5 were antigen experienced memory cells, which is compatible with the idea that SEQ ID NO: 5 represents a recall antigen. The phenotype of the CD8+ T cells from
healthy patients, which are specific to SEQ ID NO: 3, in contrast, suggests a naive phenotype.
This analysis was extended to melanoma patients. The majority of circulating CD8* T
cells specific for SEQ ID NO: 3 containing tetramers were CD28 and CD45RA positive, but
CD45RO negative. The results are presented graphically in figure 6.
Of the ten melanoma patients tested, the circulating CD8+ T cells specific for SEQ ID
NO: 3 in seven presented a uniformly naive phenotype (i.e., CD28 and CD45RA positive,
CD45RO negative), a phenotype identical to that of healthy donors' CD8+ T cells. Three of
these melanoma patients either displayed greater than 35% memory cells (CD28 and CD45RO
positive, CD45RA negative), or more than 90% effector-like cells (CD28 negative, CD45RA
intermediate levels, CD45RO negative).
Phenotype and frequency of CD8+ T cells positive for Melan-A tetramers were not
correlated, in general. For example, the memory cells found in the two melanoma patients were
not found at increased frequencies, while the high frequencies of CD8+ T cells positive for the
Melan-A containing tetramers were not of memory phenotype.
When the CD8+ T cells which were positive for tetramers containing SEQ ID NO: 5 were
analyzed, the memory phenotype discussed supra was found in 11 of the 13 melanoma and
melanoma/vitiligo patients tested, as well as in all healthy donors. There was some heterogeneity
in two of the melanoma patients.
Example 36
In these experiments, the staining methodology, limiting dilution assays, and the
ELISPOT assay technique, all of which are described, supra, were compared. To start, sorted cells from two healthy donors (HD 329 and HD 604, in the table which
follows), and two melanoma patients (LAU 132 and LAU 203), were compared, using the
functional assays described supra.
In the two healthy patients and one of the melanoma patients, nearly all cells were
CD45RA'
(95, 95 and 94%), while only 54% of the second melanoma patient's cells were CD45RA+.
When samples were tested via limiting dilution, almost all of the Melan-A specific CTL
activity was found in the CD45RA" naive subsets (98, 99 and 90%), while 73% of the CTL
activity for patient LAU 132 was found in the CD45RA" (CD45RAlow) subset. Distribution of
CTL precursors among naive and memory subsets, as analyzed by LDA, paralleled the results
obtained using the flow cytometry assays; however, frequency of the CTL precursors was
underestimated by LDA, especially in the CD45RA+ population. The level of underestimation
was by a factor of 3.6 for CD45RA" (CD45RA,0W) cells, but 11.4 for CD45RA* cells.
ELISPOT assays were then carried out on unsorted PBLs, due to an insufficiency of pure
cell populations. The number of positives for Melan-A peptide antigen was extremely low,
indicating high underestimation using the technique.
In the table which follows, the values for CD45RA - (CD45RAlow) cells for HD 329, HD
604 and LAU 203 were normalized, because frequency values were below the lower limit of
detection. Melan-A specific CD8+ T-cell frequen cy [10-5]
Tetramers LDA ELISPOT
CD8+ subset: CD45RA+ CD45RA CD45RA+ CD45RA total
(CD45RA|0W) (CD45RAl0W)
HD329 98 ± 11 7 ± 2 13 ± 0.2 0.3 ± 0.0 0.9 ± 0.9 HD604 190 ± 6 12 ± 3 9.7 ± 1.9 0.1 ± 0.0 0.0 ± 0.0 LAU 132 36 33 3.3 ± 0.4 9.1 ± 0.1 5.5 ± 5.6 LAU 203 100 4 13 ± 2.9 1.3 ± 0.1 0.0 ± 0.0
Example 37
Additional experiments were carried out to provide a better comparison of the sensitivity
of the ELISPOT technique to tetramer staining. In these experiments, PBLs from the 10 healthy
donors, and twelve of the thirteen melanoma patients were analyzed, using SEQ ID NOS: 3 and
5, staining assays and the ELISPOT assay.
Significant levels of spots specific for SEQ ID NO: 5 were found in all cases (over three
times background), with deduced frequency correlating to the tetramer calculations. The
ELISPOT assay did generally underestimate the frequency of CTL precursors specific for SEQ
ID NO: 5.
On the other hand, SEQ ID NO: 3 specific spots were only detectable in one melanoma
patient. The apparent frequency of SEQ ID NO: 3 specific, ELISPOT positive cells was 93 times
lower than that obtained using tetramer staining. When some portion of the SEQ ID NO: 3
tetramer positive cells presented antigen experienced phenotype, frequency was less
underestimated (12, and 4 fold, in two assays). This appears to show that efficient detection by
ELISPOT is restricted to memory phenotype, antigen specific cells. Example 38
The fate of SEQ ID NO: 3 specific T cells in vivo over time was studied. In these
experiments, blood samples were taken from a melanoma patient over a period of two years. To
elaborate, the patient had presented primary skin melanoma of the lower limb. Inguinal lymph
node dissection revealed that 4 of 6 lymph nodes were infiltrated by melanoma cells. The patient
received isolated limb perfusion therapy (melphalan), and then adjuvant IFN alpha therapy, over
1.5 years. A second inguinal lymph node dissection showed that 15/16 lymph nodes were
infiltrated with melanoma. The patient was tumor free for nearly two years. Over that period,
he received immunizations of melanoma specific peptides (3-4 weekly in subcutaneous inj ections
of lOOμg of each of various peptides in PBS). The peptides corresponded to amino acids 26-35
of Melan-A (i.e., SEQ ID NO: 2), amino acids 1-9 of tyrosinase (i.e., MLLAVLYCLL, SEQ ID
NO: 49), amino acids 368-378 of tyrosinase (i.e., SEQ ID NO: 4), amino acids 280-288 of gp
100, (YLLEPGPVTA, SEQ ID NO: 50) and amino acids 457-466 of tyrosinase (i.e.,
LLDGTATLRL; SEQ ID NO: 51). Five cycles of immunization were given, four of which
included daily, subcutaneous administration of GM-CSF, starting 4 days before immunization,
and covering the entire period of immunization.
Prior to the first immunization cycle, SEQ ID NO: 3 tetramer positive CD8* T cells of
the patient (0.04% of total CD8+ T cells), presented a naive, CD45RA" phenotype (The assay
used is as described supra, using the same purification methods, anti-CD45RA Cy-Chrome
labelled mAbs, and anti-CD28 FITC labelled mAbs).
One month after the end of the first injection, and until the end of the second cycle of
immunization, half of the tetramer" cells presented antigen experienced, CD45RA~(CD45RAlow)
phenotype, accompanied by a small increase in frequency of SEQ ID NO: 3 positive cells (0.04% to 0.07%). During the second year, the CD45RA (CD45RAl0W) SEQ ID NO: 3 positive cells
gradually disappeared (dropping from 51% to 23%), while the frequency of total SEQ ID NO:
3 tetramer positive cells remained constant. The vast majority of these cells continually
displayed CD28 positive phenotypes.
Example 39
The experiments described herein were designed to obtain tumor antigen specific T cells
from tumor infiltrated lymph nodes ("TILN" hereafter). Single cell suspensions were prepared
from a TILN sample taken from a melanoma patient, in accordance with Romero, et al., J.
Immunol. 159: 2366-2374 (1997), incorporated by reference. The patient had been typed,
previously, as HLA-AT 0201 positive. The single cells were cultured in medium (2 ml of
Iscove's Dulbecco medium, supplemented with Asn, Arg and Gin, 10%> pooled, human A* serum
and recombinant IL-2 (100 U/ml) and recombinant IL-7 (lOng/ml). After 16 days of culture, the
cells were analyzed as described supra, using tetramers containing SEQ ID NO: 3, mAbs to CD3
labelled with peridinin chlorophyll protein (PerCP), and mAbs to CD8, labelled with FITC. The
TILN were found to contain more than 90% CD37CD8+ cells. Of these, 19.4% were positive
for the tetramers, which is a high percentage.
Example 40
These experiments detail the expansion of the positive population described supra. First,
cells were sterile sorted into positive cells, using standard methods. The positive and negative
cells were then cultured for 2 weeks in the presence of irradiated, allogeneic PBMCs and
phytohemagglutinin, as described supra. Following the culture, the cells were retested. using tetramers and the CD8 specific mAb described supra. The positive portion contained 94.4%
tetramer positive cells, and the negative portion, 1.1% positives.
Example 41
Following the experiments described supra, the expanded populations of positive and
negative cells, as well as TILN from LAU 203 were tested in a 51Cr release assay, as described
supra, together with the peptide of SEQ ID NO:2, which was added at lμM, or without it, using
T2 cells labelled with the 51Cr as a target.
No cytotoxicity was observed without the peptide. The tetramer positive population
showed enhanced antigen specific cytolytic activity as compared to unfractionated TILN, which
was proportional to the level of enrichment. A low level of cytolysis was detected in the negative
cells, possibly resulting from a small percentage of tetramer positive T cells in the culture.
Example 42
These experiments were designed to compare the fine specificities of the unsorted, TILN
population and the tetramer positive cells described supra. This was done by carrying out 51Cr
release assays, as described supra, using T2 cells which had been incubated with varying
concentrations of different peptides, in 50 μl samples. The peptides tested were SEQ ID NOS:
1, 2, 3, 37 and 38. The T2 cells were incubated with the peptides for 15 minutes at room
temperature before adding either unsorted TILN population cells or the sorted, tetramer positive
cells, at an effector/target ratio of 10/1. Chromium release was measured after 4 hours at 37°C.
The results are presented in figures 7 A and 7B , where figure 7 A shows the results obtained using
the unsorted TILN population and 7B the sorted population. The two populations showed similar lysis patterns. Parental decapeptide (SEQ ID NO: 1) was recognized about 5 fold more
efficiently than the parental nonapeptide (SEQ ID NO: 1), but decapeptide SEQ ID NO: 3 was
recognized much more efficiently, with half maximal lysis being obtained at low peptide
concentrations, i.e., 10"10 to 10"" M.
These experiments show nearly identical fine specificity in sorted and unsorted TILN,
showing that the tetramer positive, sorted cell population can be considered a polyclonal,
monospecific T cells population, representative of the total T cell population which is specific
for the antigen/MHC complex of interest.
Example 43
T cell receptor down regulation is one of the earliest T cell activation events induced upon
antigen recognition and subsequent TCR triggering. The tetramer sorted population discussed
supra was used to study this.
Specifically, T2 cells were pulsed for 1 hour at 37°C with varying concentrations of
peptides, and were then washed, twice, to avoid peptide autopresentation by T cells. Samples
(1 x 105) of the T cells were then stimulated by peptide pulsed T2 cells (2 x 105). After ό hours
of incubation at 37°C, the cells were stained for CD3 using a fluorescent labelled monoclonal
antibody, and were then analyzed, via standard methods. The parental Melan-A peptides (SEQ
ID NOS: 1 and 2) induced only weak TCR down regulation even at the high peptide doses that
were efficient in target CTL sensitization; however, maximal TCR down regulation was induced
by SEQ ID NO: 3. Example 44
These experiments describe studies of Ca2* responses. These responses occur a few
seconds after T cell receptors are engaged. Essentially, when T cells are activated, calcium enters
the cells, and can be measured. T cells were loaded with a commercially available fluorescent
Ca2* indicator, Indo-1 AM, described by Grynkiewicz, et al. J. Biol. Chem. 260(6): 3440-3450
(1985) following Valitutti, et al., J. Exp. Med. 183: 1917-1921 (1996), both of which are
incorporated by reference. As a positive control, a CTL clone was used which had been derived
from a normal HLA-A2 positive donor, PBMCs of whom had been stimulated, in vitro, with
SEQ ID NO: 2. Samples of this CTL were combined with T2 cells that had been pulsed, for 2
hours at 37°C, with serial dilutions of SEQ ID NO: 2 or SEQ ID NO: 3. Cells were centrifuged
for one minute at 1500 x g, incubated for 1 minute at 37°C, and then resuspended and analyzed
by flow cytometry.
SEQ ID NO : 3 triggered a full Ca2+ response, but the CTL clone referred to supra required
a concentration of about 1 ng/ml. In contrast, the polyclonal, tetramer positive cells discussed
supra were triggered for Ca2+ at a broader range of peptide analogue concentrations. Some cells
even reacted at the lowest peptide concentration, i.e., 10"3 ng/ml. Further, at the maximal peptide
concentrations tested, SEQ ID NO: 2 triggered Ca2+ influx in only 60% of the cells of the CTL
clone, and 90% of the polyclonal population. In contrast, SEQ ID NO: 3 mobilized Ca2* in
nearly 100% of the CTL sample.
The data also showed that almost 50% of TCR down-regulation was required to attain
a full Ca response. SEQ ID NO: 3 also induced a more rapid Ca2* response as compared to the
parental peptide. The observation that 90% of the tetramer positive population increased Ca2" in response
to the parental decapeptide confirmed that the tetramer positive population was highly specific
for the parental peptide.
Example 45
In addition to the early activation events described in examples 43 and 44, late activation
events were also studied, i.e., cytokine synthesis, and induction of cytolytic effector cell function.
The former was done via intracellular staining of cytokines, which is described by Jung, et al,
J. Immunol. Meth. 159:197-207 (1993), as a very sensitive method for determining cytokine
response at the single cell level. Labelled monoclonal antibodies against TNF-alpha, INF
gamma, GM-CSF, and IL-4 were used. All mAbs were labelled with phycoerythrin. In brief,
5 x 104 T cells were stained with different mAbs, at concentrations of 5mg/ml, for 30 minutes
at room temperature. Dilutions and washes were carried out using PBS 10.1 %, containing 0.1%
saponin. The saponinpermeabilizes the cell, allowing entry of the mAbs. The production of each
of these cytokines by the CTL line described supra, and the polyclonal, tetramer positive
population, in response to different peptide concentrations were measured. Peptides defined by
SEQ ID NOS: 1, 2 and 3 were tested. The results are presented in figure 8. "Clone 1.13" is the
CTL clone referred to supra.
None of the peptides stimulated the entire monoclonal or polyclonal population, which
is consistent with prior reports that a fraction of CTLs is refractory to cytokine production. SEQ
ID NOS: 1 and 2 only stimulated small fractions of the CTLs (e.g., only 30%o of the cells of the
CTL clone produced TNF-alpha at the highest concentrations of these peptides). Similarly, 45%
and 10% of the polyclonal population produced TNF-alpha in response to SEQ ID NO: 1 and SEQ ID NO: 2, respectively. In contrast, SEQ ID NO: 3 stimulated the totality of CTLs able to
produce cytokines, with a plateau being reached at 1 μM, for each population. It is interesting
to note that 45%o of the polyclonal population produced some TNF-alpha, while 90% of these
cells increased intracellular Ca2* concentration at the highest concentration of SEQ ID NOS: 1
and 2 used.
The percentage of the population which produced IL-2 and IL-4 was always less than the
percentage producing TNF-alpha, regardless of the peptide concentrations used.
The polyclonal population was distinguishable from the monoclonal population in that
it had a greater capacity to produce IFN-gamma, and was incapable of producing IL-4, which is
consistent with prior observations.
Double staining experiments were carried out on both populations, following stimulation
with 10 μM of SEQ ID NOS: 1, 2, and 3. The double stainings were IFN-gamma and TNF-
alpha, and IFN-gamma and IL-2. SEQ ID NOS: 1 and 2 stimulated low fractions of these cells
to produce the cytokines; however, when SEQ ID NO: 3 was used, both the percentages of cells
producing the cytokines, and the amount of cytokine produced per cell were increased.
Most cells produced TNF-alpha and IFN-gamma, while only a fraction of IFN-gamma
positive cells produced IL-2. More importantly, however, the fraction of IL-2 producing cells
and the amount of IL-2 produced on a per cell basis were highly increased after stimulation with
SEQ ID NO: 3.
Example 46
It is known that full CTL activation is associated with cell division and induction of
cytolytic effector function. Such functions require strongly agonistic peptides, and/or higher peptide concentrations, in order to maximize induction of expansion and cytolytic effector
function, as compared to target sensitization, or induction of cytokine synthesis. In the case of
the peptides being studied herein, the low potential of SEQ ID NOS: 1 and 2 to induce cytokine
production by specific CTLs may mean that these peptides are inefficient activators of cytolytic
effector function. These experiments were designed to test this.
Peripheral blood lymphocytes of patent LAU 203 were stimulated with syngeneic
stimulator cells (PBMCs) that had been pulsed for 2 hours at 37°C in serum free medium together
with lμM of one of SEQ ID NOS: 1, 2, 3, 38 or 39, and 3 ug/ml of human β2-microglobulin.
These peptide pulsed PBMCs were washed, twice, irradiated (3000 rads), and then adjusted to
an appropriate volume before adding to the responder cells. In addition to the stimulator cells,
IL-2 (10 U/ml), and IL-7 (10 ng/ml), were added during the first two stimulation cycles (week
1 and week 2), and then IL-2 alone (100 U/ml) was added during the third week.
After 3 weeks of incubation, the PBMCs were analyzed via flow cytometry. The results
showed that cultures that had been stimulated with SEQ ID NO: 1 or 2 had 1.7% and 3.5%
tetramer positive cells, while those populations stimulated with SEQ ID NOS: 3, 38 and 39 had
11.2% (SEQ ID NO: 3), 19.7% (SEQ ID NO: 38), and 37.0% (SEQ 10 NO: 39) percent
positives.
Cytotoxicity was also tested, in 51Cr release assays as described supra. The cytotoxicity
induced by SEQ ID NOS : 1 and 2 was weak, while analogue peptides induced strong cytotoxicity
against target cells labelled with parent peptides SEQ ID NOS: 1 and 2, and autologous tumor
cells. This strong cytotoxicity was caused by high frequency of antigen specific cells in the
culture, and strong activation of these cells. Example 47
Experiments were carried out to study inhibition of CTL function. More particularly,
notwithstanding the observations described in the previous examples, it is known that in vivo T
cell activity does not protect most cancer patients.
There has been some speculation that cytolytic function is inhibited through the activity
of natural killer ("NK" hereafter) receptors. Two families of these have been observed, i.e., type
I transmembrane proteins, which belong to the imrnunoglobulin (Ig) superfamily, such as p58.2
(Moretta, et al., J. Exp. Med. 182:875-884 (1995)) and ILT2 (Colonna, et al., J. Exp. Med.
186:1809- 1818 (1997)), and those which are type II transmembrane proteins which contain a C-
type lectin domain, such as the CD94/NKG2 heterodimer. (Maretta, etal., J. Exp. Med. 180:545-
555 (1994)) . The experiments in this example were designed to determine whether NK receptors
may interfere with in vivo tumor specific responses.
First, CD8* cells were examined for NK receptor phenotype. To do this, blood samples
and lymph nodes were obtained from patients with advanced stage malignant melanoma, using
standard techniques. (The patients had been typed as HLA-A*0201 positive via flow cytometry
of PBMCs, using mAb BB7.2, described supra.)
PBLs were separated from blood samples taken from patients who suffered from vitiligo
as well as melanoma via centrifugation over Ficoll-Paque, washed 3 times, and then
cryopreserved in RPMI 1640, 40% fetal calf serum, and 10% DMSO. Vials containing from 5-
lOxlO6 cells were stored in liquid nitrogen.
Lymph nodes were collected by surgical dissection, dissociated into single cell
suspensions in sterile RPMI 1640 supplemented with 10% fetal calf serum, washed and
cyropreserved, as described supra. Aliquots of cells were placed in 24 well tissue culture plates in 2ml of Iscove's Dulbecco medium, supplemented with 0.55 mM Arg, 0.24 mM Asn, 1.5 mM
Gin, 10% pooled human A^serum, recombinant human IL-2 ( 100 U/ml), and recombinant human
IL-7 (lOng/ml).
Both CD8* cells and the cells taken from the lymph nodes were contacted with
monoclonal antibodies against p58.2/CD158b, CD94, CD94/NKG2A, and ILT2, each of which
is an NK receptor, and tetramers of SEQ ID NO:3 labelled with phycoerythrin.
The TILNs contained high percentages of CD8* tetramer * cells (1.3-5.1%). In one of the
samples analyzed, these were largely negative for p58.2 and ILT2, but were highly positive for
CD94 and CD94/NKG2A. A sample of TILN from another patient expressed some ILT2, but
low levels of the others. Yet a third sample expressed ILT2, CD94, and CD94/NKG2A.
The PBLs were found to contain from 0.10 to 0.17% CD8* tetramer ~ cells. One sample
was negative for all of the receptors. The CD8* tetramer * cells of a second sample were almost
all positive for CD94 and CD94/NKG2A, and about half expressed ILT2. A third sample of
PBLs showed a low percentage of NK receptor positive cells.
Example 48
The relatively high frequency of tumor specific CTLs in the vitiligo patients permitted
investigation of the in vivo phenotype of these cells. Fluorochrome labelled anti-CD45RA mAbs
were used, in an assay as described supra. Two samples had high levels of CD45RA positive
cells, which are indicative of naive T cells, while reduced levels were found in a third.
Additional assays were carried out using fluorochrome labelled mAbs against CD28 and
adhesion molecule CD57, since downregulation of CD28 and upregulation of CD57 have been
recognized in activated effector CTLs. It was found that samples taken from the one vitiligo subject, described supra, with a high
percentage of NK receptor expressing CTLs were predominantly CD28" and CD57+, while the
other two subjects were CD28* and CD57".
Example 49
Given that the NK receptors had been typed, it was important to determine if they were
functional. Previously, it had been shown that antigen specific, T cell cytotoxicity can be
inhibited through the triggering of the receptor CD94/NKG2A (Noppen, et al., Eur. J. Immunol.
28:1134-1142 (1998)); or p58.2 (Phillips, et al., Science 268: 403-405 (1995); Ikeda, et al,
Immunity 6: 199-208 (1997), Bakker, et al., J. Immunol. 160:5239-5245 (1998)). Experiments
were developed, and are described herein, to investigate cytotoxicity of cells ex vivo, directly
after withdrawal from the patient. PBLs were sorted from two different samples, and sorted via
FACS, using methodologies described supra, using anti CD8 and either anti p58.2 or anti
CD94/NKG 2A antibodies. The sorted suspensions were cultured, overnight, in Iscove
Dulbecco's medium supplemented with 0.55 mM Arg, 0.24 mM Asn, 1.5mM Gin, 10% pooled
human A* serum, and recombinant human IL-2 (10 U/ml).
Cytolytic activity was then tested in anti-CD3 mAb redirected 51Cr release assays. In
these assays, Fcγ-receptor expressing P815 mastocytoma cells were radiolabelled with Na5 ' CrO4
for 1 hour at 37°C, at 5% CO2. After washing, 103 P815 target cells were coincubated with PBLs
which were either CD94/NKG2 A positive or CD94/NKG2 A negative, at varying effector: target
levels, together with varying concentrations of anti-CD 3 mAbs. The anti-CD3 mAbs cross link
CD3 molecules on T cells to the Fcγ-receptors, leading to the lysis. Samples were incubated for 4 hours at 37°C, after which supernatant were collected and radioactivity measured. To
determine the role of the receptor, anti CD94/NKG2A antibody was either added, or withheld.
As a control, anti-CD 19 antibodies were used.
The results are shown in figure 9. Squares represent tests carried out using anti
CD94/NKG2A antibody, and triangles, the control, anti-CD 19 antibody.
In the presence of the anti-CD94/NKG2A antibody, the cells positive for this receptor
showed reduced killing, as compared to killing in the presence of anti CD 19 antibody, indicating
that the antibody stimulated the NK receptor, i.e., the CD94/NKG2A receptor. In contrast,
cytolytic activity of those CD8^ cells negative for this receptor were not reduced in the presence
of the antibody. Further, the NK receptor positive population showed stronger cytolytic activity,
as compared to the NK receptor negative population.
Example 50
Phenotypic analysis of various samples revealed one which had an unusually high
frequency of αβTCR*, CD28 negative cells which expressed p58.2. These cells provided a
singular opportunity to test functional consequences of p58.2 binding to its natural ligand HLA-
Cw3. As in example 49, cells of this sample were sorted via FACS, into CD8* p58.2* and CD8*
p58.2" populations, and were tested as described supra, but without any antibodies. P815 cells
were used, as were P815 cells transfected to express HLA-Cw3, using known methods.
The P815 cells were killed efficiently by both p58.2 positive and negative cells at
effector: target ratios of 5.1 ; however, lysis of transfected cells was reduced markedly, indicating
inhibition of lysis via interaction of p58.2 with HLA-Cw3. In the absence of CD3 specific
antibodies, killing was below 2%. The foregoing examples describe aspects of the invention, which is a multicomponent
complex useful, e.g., in isolating cytolytic T cells specific for a particular target, from a sample.
The complex comprises a first binding partner and a second binding partner, wherein the first and
second binding partner are specific for each other. These can be, e.g., avidin or streptavidin and
biotin, an antibody or a binding portion of an antibody specific to biotin, and so forth. The key
feature is that the second binding partner must be bound to a plurality of complexes of an MHC
molecule, a B2 microglobulin molecule and a peptide which binds specifically to said MHC
molecule, and the multicomponent complex must be labelled. The MHC molecules are
preferably HLA molecules, such as HLA-A2 molecules. The examples all show HLA-A*0201,
however, it will be understood by the artisan of ordinary skill that any HLA molecule could be
used. With respect to the peptide of interest, many references, including review articles, U.S. and
non-U.S. patents, and so forth describe peptides beyond SEQ ID NOS: 2-5 and their binding
partner HLA molecule. All are encompassed by the invention. Exemplary peptides and their
HLA molecule partners are presented later in this application.
Preferably, the second binding partner is biotin, but it may also be, e.g., an antibody
which is specific for a component of the HLA B2 microglobulin peptide complex, such as an
HLA specific antibody, or a B2 microglobulin specific antibody. Similarly, the first binding
partner may be e.g., recombinant or naturally occurring protein L, recombinant or naturally
occurring protein A, or even a second antibody. The complex can be in soluble form, or bound,
e.g., to a removable solid phase, such as a magnetic bead.
The number of HLA/B2 microglobulin/peptide complexes in the large molecule of the
invention may vary. It comprises at least two complexes, and preferably at least four, but more
may be present as well. The complex of binding partners and HLA/B2 microglobulin/peptide may be labelled,
using any of the labels known to the art. Examples of fluorescent labels are given supra.
Enzymatic labels, such as alkaline phosphatase, metal particles, colored plastics made of
synthetic materials, radioactive labels, etc., may all be used.
A third binding partner may also be used, which binds, specifically, to the first binding
partner. For example, if the first binding partner is streptavidin, and the second binding partner
is biotin, then the third binding partner may be a streptavidin specific antibody. When three or
more binding partners are used, the label refeπed to supra may be attached to any of the binding
partners so long as engagement with the HLA/B2 microglobulin peptide complexes is not
impaired.
The complexes may be used, e.g., to identify or to isolate cytolytic T cells present in a
sample, where these cells are specific for the HLA/B2 microglobulin/peptide complex. As the
examples show, such cytolytic T cells bind to the immunocomplexes of the invention. In a
preferred embodiment, the sample being tested is treated with a reactant which specifically binds
to a cytolytic T cell, wherein said label provides a detectable signal. The sample, including
labelled CTLs, is then contacted to the complex, where it binds, and can be separated via any of
the standard, well known approaches to cell separation. Preferably, FACS is used, but other
separation methodologies will be known to the skilled artisan as well. The peptide used is left
to the needs of the skilled artisan, and will depend, e.g., on the nature of the specific MHC
system under consideration, a table of exemplary, but no means the only, peptides for which
CTLs are known, follows. These are also set forth as SEQ ID NOS: 6-36. Gene MHC Peptide SEQ ID
MAGE-1 HLA-A1 EADPTGHSY 6
HLA-Cwl6 SAYGEPRKL 7
MAGE-3 HLA-A1 EVDPIGHLY 8
HLA-A2 FLWGPRALV 9
HLA-B44 MEVDPIGHLY 10
BAGE HLA-Cwl6 AARAVFLAL 11
GAGE- 1,2 HLA-Cwl6 YRPRPRRY 12
RAGE HLA-B7 SPSSNRIRNT 13
GntV HLA-A2 VLPDVFIRC(V) 14
MUM-1 HLA-B44 EEKLIVVLF 15
EEKLSVVLF 16
CDK4 HLA-A2 ACDPHSGHFV 17
ARDPHSGHFV 18
B-catenin HLA-A24 SYLDSGIHF 19
SYLDSGIHS 20
Tyrosinase HLA-A2 MLLAVLYCL 21
HLA-A2 YMNGTMSQV 22
HLA-A24 AFLPWHRLF 23
HLA-B44 SEIWRDIDF 24
HLA-B44 YEIWRDIDG 25
HLA-DR4 QNILLSNAPLGPGFP 26
HLA-DR4 DYSYLQDSDPDSFQD 27
Melan-AMart-' HLA-A2 (E)AAGIGILTV 28
HLA-A2 ILTVILGVL 29 gpl00Pmel117 HLA-A2 KTWGQYWQV 30
HLA-A2 ITDQVPFSV 31
HLA-A2 YLEPGPVTA 32
HLA-A2 LLDGTATLRL 33 HLA-A2 VLYRYGSFSV 34
DAGE HLA-A24 LYVDSLFFL 35
MAGE-6 HLA-Cwl6 KISGGPRISYPL 36
Additional peptides may be found, e.g., in U.S. patent application Serial Nos. 08/672,351,
08/718,964, 08/530,569, and 08/880,963, all of which are incorporated by reference, as well as
U.S. Patent No. 5,821,122, also incorporated by reference.
Additionally, the method can be used to monitor the status of tumors, following
administration of a particular therapeutic agent, such as a vaccine. Further, since the
methodology can be used to identify cytolytic T cell precursors, as shown, supra, one can identify
candidates for potential therapies by determining if they possess the relevant T cell precursors.
Also apart of the invention is the use of tetramers as described, in conjunction with other
steps, to yield populations of T cells with desired features. These include specific phenotypes,
such as phenotypes associated with antigen experienced, or memory cells, or naive cells, and so
forth. Such populations can be cultured, in the absence of peptides, to yield cell populations
which can be used, e.g., diagnostically or therapeutically. This culturing can be carried out, e.g.,
with a mitogen, such as phytohemagluttinin, and without peptides.
The invention also involves methods for obtaining desired T cells via in vivo recruitment.
The examples show, e.g., that one can inject a peptide of interest into a subject, e.g.,
subcutaneously, leading to recruitment of T cells. In turn, the T cells can be separated, and
cultured, in the same manner described supra.
The invention also involves the ability to improve the lytic activity of a T cell population.
As has been shown, one can assay a population of T cells, to determine if these T cells express
NK receptors. Expression of said receptors allows one to gauge the lytic potential of the T cell population involved. An antibody assay is one way to do this, but hybridization assays, and other
approaches may also be used.
Once the NK receptor expression levels are determined, one can improve the lytic ability
of the population by adding NK receptor inhibitors, such as soluble, MHC molecules like soluble
HLA-A* 0201 or HLA-Cw*3 molecules, or agents which permit the T cell to function
notwithstanding expression of NK receptors. An example of such an agent, as was shown, is an
anti-CD3 antibody.
Other aspects of the invention will be clear to the skilled artisan, and need not be
elaborated further.
The terms and expressions which have been employed are used as terms of description
and not of limitation, and there is no intention in the use of such terms and expressions of
excluding any equivalents of the features shown and described or portions thereof, it being
recognized that various modifications are possible within the scope of the invention.

Claims

Claims
1. An isolated complex comprising a first binding partner and a second binding partner
which bind to each other, at least one of which is labelled to provide a detectable signal,
and a plurality of immune complexes comprising an MHC molecule, a B2 microglobulin
molecule, and a peptide which specifically binds with said MHC molecule.
2. The isolated complex of claim 1 , wherein said second binding partner is biotin.
3. The isolated complex of claim 1, wherein said first binding partner is avidin or
streptavidin.
4. The isolated complex of claim 1, comprising at least four of said immune complexes.
5. The isolated complex of claim 4, wherein said immune complexes are identical to each
other.
6. The isolated complex of claim 1, wherein said complex is labelled with a fluorescent or
enzymatic label.
7. The isolated complex of claim 1, wherein said immune complex is a complex found on
surfaces of cancer cells.
8. The isolated complex of claim 1, wherein said first binding partner is an antibody or a
binding fragment of an antibody.
9. The isolated complex of claim 1, wherein said second binding partner is an antibody.
10. The isolated complex of claim 1, further comprising a third binding partner which
specifically binds to said first binding partner.
11. The isolated complex of claim 10, wherein said third binding partner is an antibody.
12. The isolated complex of claim 1 , further comprising a solid phase to which said isolated
complex is bound.
13. The isolated complex of claim 1 , wherein said solid phase is a magnetic bead.
14. A method for determining presence of cytolytic T cells specific for an immune complex
of an HLA molecule, a B2 microglobulin molecule and a peptide, comprising contacting
a T cell containing sample with the isolated complex of claim 1 , and determining binding
of cytolytic T cells to said isolated complex as a determination of said cytolytic T cells
in said sample.
15. The method of claim 14, further comprising labelling said cytolytic T cells with a
detectable label which specifically binds to cytolytic T cells.
16. The method of claim 15 , wherein said label which specifically binds to cytolytic T cells
is an anti-CD8, anti-CD28, anti-CD45RO, or antiCD45RA antibody.
17. A method for isolating cytolytic T cells which specifically bind to a complex of an HLA
molecule, a B2 microglobulin molecule, and a peptide, comprising contacting a sample
containing said cytolytic T cells with the isolated complex of claim 1 to bind cytolytic
T cells thereto, separating said isolated complex from said sample, and removing any
cytolytic T cells bound thereto.
18. A method for monitoring status of a tumor, comprising contacting a T cell containing
sample from a subject with a tumor with the multimeric complex of claim 1 at a first
point in time, repeating contact at a second point in time, and determining variation in
T cell type or number at said second point in time as compared to T cell type or number
at said first point in time as a determination of tumor status.
19. The method of claim 18, further comprising administering a therapeutic agent to said
subject after said first point in time and before said second point in time.
20. A method for identifying a candidate for treatment of a condition characterized by a T
cell response to said condition, comprising contacting a T cell containing sample from
said candidate with the multimeric complex of claim 1, determining any cytolytic T cell
precursors which bind thereto, and determining whether any of said cytolytic T cell
precursors are of a type sufficient to alleviate said condition.
21. A method for preparing a population of cytolytic T cells specific for a complex of an
MHC molecule and a peptide, comprising:
(a) contacting a sample containing T cells specific for said complex with the
complex of claim 1, under conditions favoring binding of said T cells to said complex,
(b) removing said T cells, and
(c) culturing said T cells in a medium which contains no peptides.
22. The method of claim 21 , wherein said complex is a tetramer.
23. The method of claim 22, wherein said tetramer comprises four MHC molecules,
and four peptide molecules, each of which is complexed with one of said four
MHC molecules.
24. The method of claim 23, wherein said four peptide molecules are tumor rejection
antigens.
25. The method of claim 21 , further comprising adding an amount of said peptide to said
sample prior to contacting with said tetramer, for a time sufficient to stimulate T cells in
said sample.
26. The method of claim 21 , wherein said medium contains a mitogen.
27. The method of claim 26, wherein said mitogen is phytohemaglutinin.
28. An isolated peptide, the amino acid sequence of which consists of an amino acid
sequence set forth in one of SEQ ID NOS: 39-47.
29. The isolated peptide of claim 28, the amino acid sequence of which is set forth at SEQ
ID NO: 41, 43, 45 or 46.
30. The isolated complex of claim 4, comprising 4 molecules of SEQ ID NO: 1, 2 , 3 or 4,
each of which is complexed to an HLA-A* 0201 molecule.
31. The method of claim 25, wherein said peptide is the peptide of SEQ ID NO: 4.
32. The isolated complex of claim 4, comprising 4 molecules of SEQ ID NO: 9, each of
which is complexed to an HLA-A* 0201 molecule.
33. The method of claim 25, wherein said peptide is the peptide of SEQ ID NO: 9.
34. A method for obtaining a population of T cells, comprising, injecting skin of a subject
with an amount of a peptide sufficient to stimulate recruitment of cytolytic T cells to the
site of injection, taking a skin biopsy from said site of injection, separating any
lymphocytes from said biopsy, isolating a subset of lymphocytes specific for complexes
of said peptide and an MHC molecule from said lymphocytes, by contacting the
lymphocytes with the isolated complex of claim 4, and stimulating said subset of
lymphocytes.
35. The method of claim 34. wherein said subject is afflicted with cancer.
36. The method of claim 34, further comprising separating said subset of lymphocytes into
naive cells and memory cells.
37. The method of claim 34, comprising stimulating said subset of lymphocytes with an
amount of the peptide used for injection.
38. The method of claim 34, comprising stimulating said subset of lymphocytes in the
absence of any peptide and in the presence of a mitogen.
39. The method of claim 34, further comprising separating said subset of lymphocytes by
determining cell surface markers thereon.
40. A method for stimulating production of T cells of interest, comprising contacting a cell
sample believed to contain T cells with the isolated complex of claim 4, separating any
T cells which bind to said complex from said samples, phenotyping said T cells based
upon cell surface markers, separating a population of said T cells which possess cell
surface markers of interest, and stimulating proliferation of said population.
41. The method of claim 40, wherein said cell surface molecules of interest are markers
which indicate that said T cells are antigen experienced.
42. A method for assessing lytic ability of a T cell population, comprising assaying said T
cell population to determine NK receptor expression by said T cell population.
43. The method of claim 42, comprising assaying said T cell population with an antibody
which specifically binds to an NK receptor.
44. The method of claim 43 , wherein said NK receptor is p58.2 or CD94/NKG2 A.
45. A method for improving lytic activity of a T cell population, comprising adding an agent
to said T cell population which inhibits activity of any NK receptors expressed by said
T cell population.
46. The method of claim 45, wherein said agent is a soluble MHC molecule or a portion of
an MHC molecule which binds to said NK receptor.
47. The method of claim 46, wherein said agent is a soluble HLA-A2 or HLA-Cw*3
molecule.
48. A method for improving lytic activity of a T cell population, at least a portion of which
express NK receptors, comprising adding an agent to said T cell population which
facilitates binding of a T cell to its target.
49. The method of claim 48, wherein said agent is a cross-linking antibody.
0. The method of claim 49, wherein said cross-linking antibody is an anti-CD3 antibody.
PCT/US1999/006615 1998-03-27 1999-03-25 Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof WO1999050637A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU33654/99A AU3365499A (en) 1998-03-27 1999-03-25 Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4985098A 1998-03-27 1998-03-27
US09/049,850 1998-03-27

Publications (3)

Publication Number Publication Date
WO1999050637A2 true WO1999050637A2 (en) 1999-10-07
WO1999050637A8 WO1999050637A8 (en) 2000-01-06
WO1999050637A3 WO1999050637A3 (en) 2000-04-13

Family

ID=21962080

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/006615 WO1999050637A2 (en) 1998-03-27 1999-03-25 Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof

Country Status (2)

Country Link
AU (1) AU3365499A (en)
WO (1) WO1999050637A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6268411B1 (en) 1997-09-11 2001-07-31 The Johns Hopkins University Use of multivalent chimeric peptide-loaded, MHC/ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
WO2001070772A2 (en) * 2000-03-23 2001-09-27 Pierre Fabre Medicament Molecule of pharmaceutical interest comprising at its n-terminal a glutamic acid or a glutamine in the form of an addition salt to an acid
WO2001090198A1 (en) * 2000-05-24 2001-11-29 Ludwig Institute For Cancer Research Multicomponent conjugates which bind to target molecules and stimulate cell lysis
US6448071B1 (en) 1996-03-28 2002-09-10 The Johns Hopkins University Soluble divalent and multivalent heterodimeric analogs of proteins
US6458354B1 (en) 1996-03-28 2002-10-01 The Johns Hopkins University Molecular complexes which modify immune responses
US7094555B2 (en) 2001-04-05 2006-08-22 Benaroya Research Institute At Virginia Mason Methods of MHC class II epitope mapping, detection of autoimmune T cells and antigens, and autoimmune treatment
EP1766393A2 (en) * 2004-05-07 2007-03-28 Beckman Coulter, Inc. Mhc bridging system for detecting ctl-mediated lysis of antigen presenting cells
US7678379B2 (en) 2004-06-17 2010-03-16 Beckman Coulter, Inc. Mycobacterium tuberculosis epitopes and methods of use thereof
US7892559B2 (en) * 2002-01-30 2011-02-22 Survac Aps Survivin-derived peptides and use thereof
US8815528B2 (en) 2002-10-11 2014-08-26 Beckman Coulter, Inc. Methods and systems for detecting MHC class I binding peptides
US10030065B2 (en) 2007-07-03 2018-07-24 Dako Denmark A/S MHC multimers, methods for their generation, labeling and use
US10336808B2 (en) 2007-03-26 2019-07-02 Dako Denmark A/S MHC peptide complexes and uses thereof in infectious diseases
US10369204B2 (en) 2008-10-02 2019-08-06 Dako Denmark A/S Molecular vaccines for infectious disease
US10611818B2 (en) 2007-09-27 2020-04-07 Agilent Technologies, Inc. MHC multimers in tuberculosis diagnostics, vaccine and therapeutics
US10722562B2 (en) 2008-07-23 2020-07-28 Immudex Aps Combinatorial analysis and repair
US10968269B1 (en) 2008-02-28 2021-04-06 Agilent Technologies, Inc. MHC multimers in borrelia diagnostics and disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0817244D0 (en) 2008-09-20 2008-10-29 Univ Cardiff Use of a protein kinase inhibitor to detect immune cells, such as T cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DUNBAR R.: 'Characterization of Human Cytotoxic T-Lymphocyte (CTL) Responses to Tumor Antigens using fluorogenic MHC Class I/Peptide complexes' TUMOR IMMUNOLOGY vol. 92, December 1997, page 12, SEE ABSTRACT NO. 3.3, XP002923255 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6448071B1 (en) 1996-03-28 2002-09-10 The Johns Hopkins University Soluble divalent and multivalent heterodimeric analogs of proteins
US6458354B1 (en) 1996-03-28 2002-10-01 The Johns Hopkins University Molecular complexes which modify immune responses
US6734013B2 (en) 1997-03-28 2004-05-11 The Johns Hopkins University Use of multivalent chimeric peptide-loaded, MHC/Ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
US6268411B1 (en) 1997-09-11 2001-07-31 The Johns Hopkins University Use of multivalent chimeric peptide-loaded, MHC/ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
FR2806727A1 (en) * 2000-03-23 2001-09-28 Pf Medicament MOLECULE OF PHARMACEUTICAL INTEREST COMPRISING IN THE N-TERMINAL END A GLUTAMIC ACID OR GLUTAMINE IN THE FORM OF A PHYSIOLOGICALLY ACCEPTABLE ACID ADDITION SALT
WO2001070772A3 (en) * 2000-03-23 2003-02-13 Pf Medicament Molecule of pharmaceutical interest comprising at its n-terminal a glutamic acid or a glutamine in the form of an addition salt to an acid
WO2001070772A2 (en) * 2000-03-23 2001-09-27 Pierre Fabre Medicament Molecule of pharmaceutical interest comprising at its n-terminal a glutamic acid or a glutamine in the form of an addition salt to an acid
WO2001090198A1 (en) * 2000-05-24 2001-11-29 Ludwig Institute For Cancer Research Multicomponent conjugates which bind to target molecules and stimulate cell lysis
US7094555B2 (en) 2001-04-05 2006-08-22 Benaroya Research Institute At Virginia Mason Methods of MHC class II epitope mapping, detection of autoimmune T cells and antigens, and autoimmune treatment
US7892559B2 (en) * 2002-01-30 2011-02-22 Survac Aps Survivin-derived peptides and use thereof
USRE48522E1 (en) 2002-01-30 2021-04-20 Survac Aps Survivin-derived peptides and uses thereof
US9534030B2 (en) 2002-01-30 2017-01-03 Survac Aps Survivin-derived peptides and uses thereof
US8815528B2 (en) 2002-10-11 2014-08-26 Beckman Coulter, Inc. Methods and systems for detecting MHC class I binding peptides
US8318174B2 (en) 2003-01-30 2012-11-27 Survac Aps Survivin-derived peptides and use thereof
JP2007536522A (en) * 2004-05-07 2007-12-13 ベックマン コールター インコーポレーティッド MHC cross-linking system for detecting CTL-mediated lysis of antigen presenting cells
EP1766393A4 (en) * 2004-05-07 2008-06-18 Beckman Coulter Inc Mhc bridging system for detecting ctl-mediated lysis of antigen presenting cells
EP1766393A2 (en) * 2004-05-07 2007-03-28 Beckman Coulter, Inc. Mhc bridging system for detecting ctl-mediated lysis of antigen presenting cells
US7678379B2 (en) 2004-06-17 2010-03-16 Beckman Coulter, Inc. Mycobacterium tuberculosis epitopes and methods of use thereof
US10336808B2 (en) 2007-03-26 2019-07-02 Dako Denmark A/S MHC peptide complexes and uses thereof in infectious diseases
US10030065B2 (en) 2007-07-03 2018-07-24 Dako Denmark A/S MHC multimers, methods for their generation, labeling and use
US10611818B2 (en) 2007-09-27 2020-04-07 Agilent Technologies, Inc. MHC multimers in tuberculosis diagnostics, vaccine and therapeutics
US10968269B1 (en) 2008-02-28 2021-04-06 Agilent Technologies, Inc. MHC multimers in borrelia diagnostics and disease
US10722562B2 (en) 2008-07-23 2020-07-28 Immudex Aps Combinatorial analysis and repair
US10369204B2 (en) 2008-10-02 2019-08-06 Dako Denmark A/S Molecular vaccines for infectious disease

Also Published As

Publication number Publication date
WO1999050637A8 (en) 2000-01-06
AU3365499A (en) 1999-10-18
WO1999050637A3 (en) 2000-04-13

Similar Documents

Publication Publication Date Title
Jäger et al. Clonal expansion of Melan A‐specific cytotoxic T lymphocytes in a melanoma patient responding to continued immunization with melanoma‐associated peptides
Zaremba et al. Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen
Ikeda et al. Characterization of an antigen that is recognized on a melanoma showing partial HLA loss by CTL expressing an NK inhibitory receptor
JP5627180B2 (en) Tumor-associated peptide that binds to human leukocyte antibody (HLA) class II molecules indiscriminately
van Elsas et al. Peptide‐pulsed dendritic cells induce tumoricidal cytotoxic T lymphocytes from healthy donors against stably HLA‐A* 0201‐binding peptides from the Melan‐A/MART‐1 self antigen
WO1999050637A2 (en) Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof
US8519109B2 (en) Tumour rejection antigens
US6326200B1 (en) Isolated nona-and decapeptides which bind to HLA molecules, and the use thereof
US8075900B2 (en) Melanoma associated peptide analogues and vaccines against melanoma
Bettinotti et al. Stringent allele/epitope requirements for MART-1/Melan A immunodominance: implications for peptide-based immunotherapy
CA2278189A1 (en) Peptides and peptide-loaded antigen presenting cells for the activation of ctl
Spagnoli et al. Peptide‐specific ctl in tumor‐infiltrating lymphocytes from metastatic melanomas expressing mart‐1/melan‐a, gp100 and tyrosinase genes: A study in an unselected group of hla‐a2. 1‐positive patients
US6368857B1 (en) Method for provoking proliferation of cytolytic T cells via the use of decapeptides which complex with HLA-A2 molecules
AU764550B2 (en) Isolated peptides which bind to HLA-B35 molecules
Maccalli et al. TCR beta-chain variable region-driven selection and massive expansion of HLA-class I-restricted antitumor CTL lines from HLA-A* 0201+ melanoma patients.
CA2259944C (en) Melanoma associated peptide analogues and vaccines against melanoma
US6506875B1 (en) Isolated peptides which bind to HLA-C molecules and uses thereof
WO2001016320A1 (en) Isolated nona and decapeptides which bind to hla molecules, and the use thereof
US7041502B2 (en) Isolated peptides which bind to HLA-B18 molecules and uses thereof
JP2002502224A (en) Methods for identifying individuals with cellular abnormalities
EP0854727B1 (en) Tumor rejection antigens presented by hla-b44 molecules, and uses thereof
JP5736333B2 (en) Tumor-associated peptide that binds to human leukocyte antibody (HLA) class II molecules indiscriminately
JP5736334B2 (en) Tumor-associated peptide that binds to human leukocyte antibody (HLA) class II molecules indiscriminately
Dietrich et al. Vaccination with a Melan-A Peptide Selects

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: C1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: C1

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

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i
AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A3

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase