NZ701000B2 - Novel melanoma antigen peptide and uses thereof - Google Patents

Novel melanoma antigen peptide and uses thereof Download PDF

Info

Publication number
NZ701000B2
NZ701000B2 NZ701000A NZ70100012A NZ701000B2 NZ 701000 B2 NZ701000 B2 NZ 701000B2 NZ 701000 A NZ701000 A NZ 701000A NZ 70100012 A NZ70100012 A NZ 70100012A NZ 701000 B2 NZ701000 B2 NZ 701000B2
Authority
NZ
New Zealand
Prior art keywords
peptide
melanoma
seq
antigen peptide
meloe
Prior art date
Application number
NZ701000A
Other versions
NZ701000A (en
Inventor
Rogel Anne
Lang Francois
Nathalie Labarriere
Bobinet Mathilde
Original Assignee
Centre National De La Recherche Scientifique (Cnrs)
Inserm (Institut National De La Sante Et De La Recherche Medicale)
Universite D'angers
Universite De Nantes
Filing date
Publication date
Application filed by Centre National De La Recherche Scientifique (Cnrs), Inserm (Institut National De La Sante Et De La Recherche Medicale), Universite D'angers, Universite De Nantes filed Critical Centre National De La Recherche Scientifique (Cnrs)
Priority claimed from PCT/IB2012/001310 external-priority patent/WO2013175256A1/en
Publication of NZ701000A publication Critical patent/NZ701000A/en
Publication of NZ701000B2 publication Critical patent/NZ701000B2/en

Links

Abstract

The present invention relates to a melanoma antigen peptide comprising the amino acids sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 5 or a function-conservative variant thereof. Moreover the invention also relates to a melanoma antigen peptide according to the invention for use in the prevention or the treatment of melanoma in patient. the invention for use in the prevention or the treatment of melanoma in patient.

Description

NOVEL MELANOMA ANTIGEN E AND USES THEREOF FIELD OF THE INVENTION: The present invention relates to a melanoma antigen peptide comprising the amino acids sequence selected in the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID N023, SEQ ID N024 or SEQ ID NO:5 or a on-conservative variant thereof. Moreover the invention also relates to a melanoma antigen peptide according to the invention for use in the prevention or the treatment ofmelanoma in patient.
BACKGROUND OF THE INVENTION: In antitumor immune ses, CD8 cytotoxic T cytes (CTL) have been identified as the most powerful effector cells (Vesely MD et al., 2011). As a consequence, most previous anti-cancer vaccines use class I HLA—restricted peptides derived from tumor antigens in order to ate CTL responses. r, the clinical impact of peptide-based cancer vaccines remains still modest, even if a recent gplOO-derived peptide vaccination was shown to increase patient survival in melanoma berg SA et al., 2004 and tzentruber DJ et al., 2011). In addition to a variety of immune suppressive mechanisms originating from the tumor , suboptimal design of vaccines used so far may explain this failure. In particular, short epitopic peptides, could induce vanishing CTL responses or tolerance s targeted antigens (Bijker MS et al., 2007 and Toes RE et al., 1996). In the meanwhile, CD4 helper T cells have gained interest in anti-tumor immunity and therapy. Indeed, tumor-reactive CD4+ T helper 1 T cells (Th1) produce several cytokines (such as IFN-y, TNF-d and IL-2) essential for the ion of cell-mediated immunity against tumors. One widely accepted model demonstrates the ability of CD4+ T cells to ‘license’ dendritic cells (DCs) for efficient CD8+ T cell priming through the interaction of costimulatory receptors (Bennett SR et al., 1998 and Smith CM et al., 2004).
The cytokines secreted by CD4+ Th1 cells also exert direct antitumor and antiangiogenic effects. Furthermore, it has been demonstrated in a mouse model that only tumor-reactive CD4+ T cells have been found to ensure efficient effector CTLs recruitment at the tumor site.
In a al standpoint, a high density of tumor-infiltrating CD4+ Th1 cells has been recently shown as a good prognostic marker in colorectal cancer patients emphasizing the role of these cells in cancer immunosurveillance. In melanoma, tumor-reactive CD4 T cells have also been ated with a good clinical outcome (Robbins PF et al., 2002), and more recently the same group showed that tumor specific CD4 T cells were present in at least 20% of metastatic melanomas, and suggested that the infiision of TIL populations containing CD4 specific T cells could enhance the efficacy of adoptive cell y (Friedman KM et al., 2012). In the same line of thought, it has been demonstrated in a melanoma patient that the adoptive cell transfer of CD4 T cells specific for NYESO-l antigen induces durable clinical remission and led to endogenous responses against non-targeted tumor antigens, suggesting the stimulation of immune responses by transferred CD4 T cells (Hunder NN et al., 2008).
In the field of peptide vaccination, it has been documented twenty years ago, in a mouse model that the generation of a strong CD8 response against a LCMV-derived peptide ed on the ce of CD4 helper T cells (Fayolle C et al., 1991). These results have been more recently confirmed in a al setting by the use of synthetic long peptides (SLP) in ctal cancer, using P53 derived SLP (Speetjens FM et al., 2009), in vulvar intraepithelial neoplasia (Kenter GG et al., 2009) and cervical cancer patients (Welters MJ et al., 2008) using HPVlé-derived SLP. In the case of vulvar neoplasia, clinical responses ed to be ated with the induction of strong HPV16 specific immune responses.
Synthetic long peptides containing immunogenic CD8 and CD4 tumor epitopes are therefore attractive tools to implement therapeutic cancer e.
One of the main issues in the field of long e vaccination in solid tumors is to identify immunogenic long peptides derived from relevant tumor associated antigens. Target antigens should be widely expressed, and able to induce robust CD8 and CD4 anti-tumor T cell responses. In ma, the Melan—A antigen fillfills these requirements and the inventors recently reported the efficiency of a Melan—A modified SLP, to cross-prime human tumor-reactive T cells (Chauvin JM et al., 2012). Another attractive target for melanoma vaccination would be the MELOE—l antigen (46 amino , specifically overexpressed in melanoma. Indeed, the inventors previously reported that the infusion of tumor infiltrating lymphocytes (TIL) c for the MELOE-l antigen was associated with a prolonged relapse-free survival for HLA-A2 melanoma patients who ed TIL therapy (Godet Y et al., 2008). Furthermore, they nted the presence of a large and tumor reactive CD8 T cell repertoire in HLA—A2 melanoma patients (Godet Y et al., 2010) and the presence of two class II es in the vicinity of the class I epitope, located at the C-terminal end of the polypeptide (Rogel A et al., 2011).
Despite these results, the identification of onal melanoma ns with a documented immunogenic potential remains a major issue to address for melanoma immunotherapy.
SUMMARY OF THE INVENTION: In a first aspect, the present invention provides use of a melanoma antigen peptide of less than amino acids long comprising the amino acid sequence SEQ ID NO:5 in the manufacture of a ment for the prevention or the treatment of ma in a t.
In a second aspect, the present invention provides use of a fusion protein comprising a melanoma antigen peptide as defined in the first aspect and a melanoma antigen peptide comprising the amino acids motif: - TX2NDECWPX9 (SEQ ID NO: 23) wherein X2 is e, nine, valine, isoleucine or glutamine and X9 is alanine, valine or leucine in the manufacture of a medicament for the prevention or the treatment of melanoma in a patient.
In a third aspect, the present invention provides use of a melanoma antigen peptide according to the first aspect or a fusion protein according to the second aspect, wherein the patient is genotyped with HLA-DQβ1*0201 or HLA-DQβ1*0202 s.
In a fourth aspect, the present invention provides use of a T lymphocyte that izes ically a melanoma antigen peptide as defined in the first aspect in the manufacture of a medicament for the prevention or the treatment of melanoma in a patient.
In a fifth , the present invention provides a method for producing said T cytes as defined in the fourth aspect, said method comprising the steps of: (a) stimulating PBMCs or tumor infiltrating lymphocytes obtained from a patient with at least one melanoma n peptide according to the first aspect or a fusion protein according to the second aspect, (b) enriching the population of T lymphocytes specific for the melanoma antigen peptide(s) used in (a), (c) optionally cloning said population of T lymphocytes specific for the melanoma antigen peptide(s) used in (a).
In a sixth aspect, the t invention provides a melanoma antigen peptide comprising the amino acid sequence SEQ ID NO:5, wherein said peptide has an amino acid sequence of less than 15 amino acids long, or consists of the sequence SEQ ID NO:2 or SEQ ID NO:4.
In a seventh aspect, the present invention provides a fusion protein comprising a melanoma antigen peptide as defined in the sixth aspect and a melanoma antigen peptide comprising the amino acids motif: - TX2NDECWPX9 (SEQ ID NO: 23) wherein X2 is leucine, nine, valine, isoleucine or glutamine and X9 is alanine, valine or e.
AH26(12781302_1):EOR In an eighth aspect, the t ion provides a nucleic acid sequence encoding a melanoma antigen peptide according to the sixth aspect or a fusion protein according to the seventh aspect.
In a ninth aspect, the present invention provides an expression vector comprising a nucleic acid sequence according to the eighth aspect.
In a tenth aspect, the t invention provides a host cell comprising an expression vector according to the ninth aspect, n the host cell is not within a human body.
In an eleventh aspect, the present invention es antibody or fragment thereof that binds to a melanoma antigen peptide according to the sixth aspect.
In a twelfth aspect, the present ion provides an immunising composition comprising: (a) at least one ma antigen peptide according to the sixth aspect or (b) at least one fusion protein according to the seventh aspect, or (c) at least one nucleic acid sequence according to the eighth aspect or (d) at least one expression vector according to the ninth aspect, or (e) at least one host cell according to the tenth aspect, or (f) at least one antibody according to the eleventh aspect.
In a thirteenth aspect, the present invention provides use of an antibody or fragment thereof that binds to a melanoma antigen peptide as defined in the first aspect in the manufacture of a ment for the prevention or the treatment of ma in a patient.
In this study, the inventors find new epitopes located all along the MELOE-1 ce and characterized by their T helper profile of CD4 T cell response.
Thus, the invention relates to a melanoma antigen peptide comprising the amino acids sequence selected in the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:5 or a function-conservative variant thereof. Moreover the invention also relates to a ma antigen peptide according to the invention for use in the prevention or the ent of melanoma in patient.
DETAILED DESCRIPTION OF THE INVENTION: Definitions: Throughout the specification, several terms are employed and are defined in the following paragraphs.
As used herein, the term “peptide” refers to an amino acid sequence having less than 50 amino acids. As used herein, the term de” encompasses amino acid sequences having less than 50 amino acids, less than 40 amino acids, less than 30 amino acids, less than 25 amino acids, less than 20 amino acids, less than 15 amino acids or less than 10 amino acids.
AH26(12387646_1):EOR Melanoma n peptides of the invention are described in table A.
SEQ ID number Sequences Nomenclatures used in the patent application Peptide SEQ ID NO:1 SCVGYPDEATSREQFLPSEC MELOE21 or 2-21 Peptide SEQ ID NO:2 VGYPDEATSREQFLPS MELOE19 or 4-19 Peptide SEQ ID NO:3 VGYPDEATSREQFL 14-17 or 4-17 Peptide SEQ ID NO:4 GYPDEATSREQFLPS MELOE19 or 5-19 Peptide SEQ ID NO:5 PDEATSREQFLPS MELOE19 or 7-19 AH26(12387646_1):EOR SmrOm |-6 or 26-46 Peptide SEQ ID \0 7 m |.5 or 32-45 Peptide SEQ ID \0 8 SmrOm |_4 or 32-44 Peptide SEQ ID \'o:9 ViELOE-13144 or 31-44 Peptide SEQ ID \o:10 \/[EL0E-111_30 or 11-30 Peptide SEQ ID \'o:11 E—113_27 or 13-27 Peptide SEQ ID \‘0212 VIELOE—lpm or 14-27 Peptide SEQ ID 30:13 MELOE-115_27 or 15-27 Peptide SEQ ID 30:14 111_23 or 11-23 Peptide SEQ ID 50:15 MELOE-lim or 12-24 e SEQ ID 30:16 MELOE-lig_37 or 18-37 Peptide SEQ ID x017 MELOE-lzus or 22-46 Peptide SEQ ID 1\O:18 MELOE-124,37 or 24-37 Peptide SEQ ID \o:19 \/[ELOE-124_36 or 24-36 Peptide SEQ ID \0220 MSCVGYPDEATSREQFLPSEGAACPPW MELOE-11_46 or 1-46 HPSERISSTLNDECWPASL e SEQ ID \O:2l GHGHSYTTAEELAGIGILTVILGVL Sa$A16—40L Table A: melanoma antigen peptides ofthe invention As used herein, the term "antibody" refers to a protein capable of specifically binding an n, typically and preferably by g an epitope or antigenic determinant or said antigen. The term ody" also includes recombinant proteins comprising the binding domains, as well as variants and fragments of antibodies. Examples of fragments of antibodies e Fv, Fab, Fab', F(ab')2, dst, scFv, 2, diabodies and multispeciflc antibodies formed from antibody fragments.
"Function-conservative variants" as used herein refer to those in which a given amino acid residue in a protein or enzyme has been changed (inserted, deleted or substituted) without altering the overall conformation and function of the polypeptide. Such variants e protein having amino acid alterations such as deletions, insertions and/or substitutions. A “deletion” refers to the absence of one or more amino acids in the protein. An “insertion” refers to the addition of one or more of amino acids in the protein. A “substitution” refers to the replacement of one or more amino acids by another amino acid residue in the protein. Typically, a given amino acid is replaced by an amino acid with one having similar properties (such as, for example, polarity, hydrogen bonding potential, acidic, basic, hydrophobic, aromatic, and the like). Amino acids other than those indicated as ved may differ in a protein so that the t protein or amino acid sequence similarity between any two proteins of similar function may vary and may be, for example, from 70 % to 99 % as determined according to an alignment scheme such as by the Cluster Method, wherein similarity is based on the MEGALIGN algorithm. A "function—conservative varian " also includes a polypeptide which has at least 60 % amino acid identity as determined by BLAST or FASTA algorithms, preferably at least 75 %, more preferably at least 85%, still preferably at least 90 %, and even more preferably at least 95%, and which has the same or substantially r properties or fiinctions as the native or parent protein to which it is compared. Two amino acid sequences are "substantially homologous" or "substantially similar" when greater than 80 %, preferably greater than 85 %, preferably greater than 90 % of the amino acids are identical, or greater than about 90 %, preferably greater than 95 %, are similar (functionally identical) over the whole length of the shorter sequence. Preferably, the similar or gous ces are identified by alignment using, for example, the GCG (Genetics Computer Group, Program Manual for the GCG Package, Version 7, Madison, Wisconsin) pileup program, or any of sequence comparison algorithms such as BLAST, FASTA, etc.
The term “Major Histocompatibility x” (MHC) is a generic designation meant to encompass the histo-compatibility antigen systems described in different species including the human leucocyte ns (HLA).
The term “melanoma” as used herein includes, but is not limited to, all types of melanocytes cancers at all stages ofprogression like metastatic melanocyte cancer.
The term “treating” a disorder or a condition refers to reversing, alleviating or inhibiting the s of one or more symptoms of such disorder or condition. The term nting” a disorder or condition refers to preventing one or more ms of such disorder or ion.
As used herein, the term “patient” denotes a , such as a rodent, a feline, a canine, and a primate. Preferably a patient according to the invention is a human.
A “therapeutically effective amount” as used herein is ed for a minimal amount of active agent which is necessary to impart therapeutic benefit to a patient. For example, a “therapeutically effective amount of the active agent” to a patient is an amount of the active agent that induces, ameliorates or causes an improvement in the pathological symptoms, disease progression, or physical conditions ated with the disease ing the patient.
The term “adjuvant” as used herein refers to a compound or a mixture that may be non-immunogenic when administered in the host alone, but that augments the host’s immune response to an antigen when administered conjointly with that antigen.
Peptide, fusion protein and uses thereof A first object of the invention relates to a melanoma antigen peptide comprising the amino acids sequence selected in the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID N023, SEQ ID N024 or SEQ ID NO:5 or a fianction-conservative variant thereof.
In one embodiment, the ma n peptide has the sequence SEQ ID NO:5.
Melanoma antigen peptides of the invention, are generated from the peptide MELOE- l (SEQ ID NO:20) as described in the patent application WC 2010 026165 and in table A.
In one embodiment, the melanoma antigen peptide of the invention is not the peptide MELOE-l (SEQ ID NO:20).
In one embodiment of the invention, by “antigen peptide” is meant a peptide capable ofbinding to HLA le and causing a cellular or l response in a patient.
In a preferred embodiment of the invention, said antigen peptide may comprise a specific motif such that the polypeptide binds an HLA molecule and induces a CTL se.
In another preferred embodiment of the invention, said antigen e may se a specific motif such that the polypeptide binds an HLA molecule and induces a helper T cell response.
In one embodiment of the invention, said melanoma antigen peptides as described here above are HLA-DQB1*0201 restricted.
In one embodiment of the invention, said melanoma antigen peptides as described here above are B1*0202 restricted.
In one ment ofthe invention, said antigen peptide is an amino acid sequence of less than 50 amino acids long that comprises the amino acid motif SEQ ID NO: 1, 2, 3, 4 or 5 as defined here above.
In another embodiment of the invention, said antigen e is an amino acid sequence of less than 45 amino acids long that comprises the amino acid motif SEQ ID NO: 1, 2, 3, 4 or 5 as defined here above.
In another embodiment of the invention, said antigen peptide is an amino acid ce of less than 40 amino acids long that comprises the amino acid motif SEQ ID NO: 1, 2, 3, 4 or 5 as defined here above.
In another embodiment of the invention, said antigen peptide is an amino acid sequence of less than 30 amino acids long that comprises the amino acid motif SEQ ID NO: 1, 2, 3, 4 or 5 as defined here above.
In another ment of the invention, said antigen peptide is an amino acid ce of less than 25 amino acids long that comprises the amino acid motif SEQ ID NO: 1, 2, 3, 4 or 5 as defined here above.
In one embodiment, the antigen peptide according to the invention comprises at least 60% identity over said SEQ ID NO: 1, 2, 3, 4 or 5 even more preferably at least 700%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97% and is still able to bind to HLA molecule and causing a cellular or humoral se in a patient.
In another embodiment, the antigen peptide according to the invention consists in the amino acid sequence as set forth in SEQ ID NO: 1, 2, 3, 4 or 5 or a variant f comprising at least 60%, preferably at least 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% identity with SEQ ID N021 and is still able to bind to HLA molecule and causing a cellular or humoral response in a patient.
The invention also encompasses peptides that are function-conservative variants of antigen peptides comprising SEQ ID NO: 1, 2, 3, 4 or 5 as described here above.
Typically, the invention encompasses peptides substantially identical to antigen es comprising SEQ ID NO: 1, 2, 3, 4 or 5 in which one or more residues have been conservatively tuted with a functionally similar residue and which displays the functional aspects of the n peptides comprising SEQ ID NO: 1, 2, 3, 4 or 5 as described here above, i.e. being still able to bind to an HLA molecule in substantially the same way as a peptide consisting ofthe given amino acid sequence.
Examples of conservative substitutions include the substitution of one non-polar (hydrophobic) residue such as isoleucine, valine, leucine or methionine for r, the substitution of one polar (hydrophilic) residue for r such as between arginine and lysine, between glutamine and asparagine, between glycine and serine, the substitution of one basic e such as lysine, arginine or histidine for another, or the substitution of one acidic residue, such as aspartic acid or glutamic acid or r.
The term "conservative substitution" also includes the use of a chemically derivatized residue in place of a non-derivatized residue. "Chemical derivative" refers to a t peptide having one or more residues chemically derivatized by reaction of a functional side group.
Examples of such derivatized molecules include for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides. Free hydroxyl groups may be derivatized to form O-acyl or O-alkyl derivatives. The o 1e nitrogen of histidine may be derivatized to form N-im- benzylhistidine. Chemical derivatives also include peptides which contain one or more naturally-occurring amino acid derivatives of the twenty standard amino acids. For examples: oxyproline may be substituted for proline; 5-hydroxylysine may be substituted for ; ylhistidine may be substituted for histidine; homoserine may be substituted for serine; and ne may be tuted for lysine.
According to the invention, the n peptides of the invention can be obtained by synthesizing the peptides according to the method for peptide sis known in the art.
In r embodiment, the antigen peptides of the invention may be incorporated into polytopes or fiision proteins. Two or more peptides of the invention can be joined together directly, or via the use of flanking sequences. Thompson et al., Proc. Natl. Acad. Sci. USA 92 (13): 5845- 5849 (1995), teaches the direct linkage of relevant epitopic ces. The use of polytopes or fiision proteins as vaccines is well known. See, e. g. t et al., Nat.
Biotechnol. 15 (12): 1280—1284 (1997); Thomson et al. Thomson et al., J. Immunol. , supra; 157 (2): 822- 826 (1996); Tam et al., J. Exp. Med. 171 (1) : 299-306 (1990), all ofwhich are incorporated by reference. The Tam et a1. reference in particular shows that polytopes or fusion proteins, when used in a mouse model, are useful in generating both antibody and protective immunity.
Thus, the invention also relates to a filSiOI‘l protein comprising a melanoma antigen peptide according to the invention and a melanoma antigen peptide comprising the amino acids motif: — TX2NDECWPX9 (SEQ ID NO: 22) wherein X2 is leucine, methionine, valine, isoleucine or glutamine and X9 is alanine, valine or leucine.
In one embodiment of the invention, said second melanoma antigen peptide is selected in the group consisting ofpeptides having the sequence SEQ ID NO: 23 to SEQ ID NO: 37 as described below.
Peptide SEQ ID NO 23 Peptide SEQ ID NO 24 Peptide SEQ ID )0 25 Peptide SEQ ID NO 26 Peptide SEQ ID \0 27 e SEQ ID \0 28 e SEQ ID \0 29 Peptide SEQ ID \0 30 Peptide SEQ ID \0 31 Peptide SEQ ID \0 32 Peptide SEQ ID \0 33 Peptide SEQ ID \0 34 Peptide SEQ ID \'O 35 Peptide SEQ ID \'O 36 Peptide SEQ ID \'O 37 In another embodiment, the melanoma antigen peptide according to the invention or the fusion protein according to the invention may be use in the tion or the treatment of melanoma in t.
In one embodiment, said patient is genotyped with HLA-DQB1*0201 or HLA— DQB1*0202 alleles.
Nucleic acids, vectors, recombinant host cells and uses thereof Another object of the invention relates to a nucleic acid sequence ng a melanoma antigen peptide according to the invention or a fiision protein according to the ion.
Another object of the invention relates to an sion vector comprising a nucleic acid sequence encoding a melanoma antigen peptide according to the invention or a fitsion protein ing to the invention.
In one embodiment of the invention, said expression vector comprises the nucleic acid sequence corresponding to a melanoma antigen peptide having the sequence SEQ ID NO:1 to SEQ ID NO: 5.
According to the invention, expression vectors suitable for use in the invention may se at least one expression control element operationally linked to the c acid ce. The expression control elements are inserted in the vector to control and regulate the expression of the nucleic acid sequence. Examples of expression control elements include, but are not limited to, lac system, or and promoter regions of phage lambda, yeast promoters and promoters derived from polyoma, adenovirus, retrovirus, lentivirus or SV40.
Additional preferred or required operational elements include, but are not limited to, leader sequence, termination codons, polyadenylation signals and any other sequences necessary or red for the riate transcription and uent translation of the nucleic acid sequence in the host system. It will be tood by one skilled in the art that the correct combination of required or preferred expression control elements will depend on the host system chosen. It will further be understood that the expression vector should n additional elements necessary for the er and subsequent replication of the expression vector containing the nucleic acid sequence in the host system. Examples of such elements include, but are not d to, origins of replication and selectable markers. It will further be understood by one skilled in the art that such vectors are easily constructed using conventional methods or commercially ble.
Another object of the invention is a host cell comprising an expression vector as described here above.
According to the invention, examples of host cells that may be used are eukaryote cells, such as animal, plant, insect and yeast cells and prokaryotes cells, such as E. coli. The means by which the vector carrying the gene may be introduced into the cells include, but are not limited to, microinjection, electroporation, transduction, or transfection using DEAE- dextran, lipofection, m phosphate or other procedures known to one skilled in the art.
In a red embodiment, otic expression vectors that fiinction in eukaryotic cells are used. Examples of such vectors include, but are not limited to, Viral vectors such as retrovirus, adenovirus, adeno-associated virus, herpes virus, vaccinia virus, us, poliovirus; lentivirus, bacterial expression vectors, plasmids, such as pcDNA3 or the baculovirus er vectors. Preferred eukaryotic cell lines include, but are not limited to, COS cells, CHO cells, HeLa cells, NIH/3T3 cells, 293 cells (ATCC# CRL1573), T2 cells, dendritic cells, or monocytes.
In one embodiment, the nucleic acid sequence according to the invention or the expression vector according to the invention or the host cell ing to the invention may be use in the prevention or the treatment ofmelanoma in patient.
Antibodies and uses thereof r object of the invention relates to an dy or fragment thereof that binds to the melanoma antigen peptide according to invention.
In one embodiment of the invention, said dy or fragment thereof binds to melanoma antigen peptide having the sequence SEQ ID NO: 1 to SEQ ID NO: 5.
In one ment of the invention, said antibody is monoclonal. In another embodiment ofthe invention, said antibody is polyclonal.
Such antibodies may be easily prepared, for example, according to the method described in “Antibodies: A laboratory manual”, Lane H. D. et al. eds, Cold Spring Harbor Laboratory Press, New York, 1989 or Antibody Engineering: Methods and Protocols, 2003, Benny K. Lo.
MHC/peptide multimer Another object of the invention relates to a MHC/peptide multimer comprising a melanoma antigen peptide as bed here above. According to the invention, said ptide multimer include, but are not limited to, a MHC/peptide dimer, , tetramer or pentamer.
In one embodiment of the invention, said MHC/peptide multimer is a HLA-class II/peptide multimer.
In another embodiment of the invention, said MHC/peptide multimer is a HLA- DQB l *0201 II/melanorna n peptide multimer or a HLA—DQB1*0202/melanoma antigen peptide multimer.
Methods for obtaining MHC/peptide ers are described in WO96/26962 and W001/18053, which are orated by reference.
In one embodiment of the invention, said MHC/peptide er can be used to visualise T cell populations that are specific for the complex HLA—DQB1*0201/ melanoma antigen peptide or HLA-DQBl *0202/melanoma antigen peptide multimer as bed here above.
In another embodiment of the invention, said MHC/peptide multimer can be used for the detection and/or isolation by screening (in flow cytometry or by immunomagnetic screening) of T cell population that are specific for a complex HLA/ melanoma antigen peptide as described here above.
In another embodiment of the invention, said HLA-DQBl*0201/ melanoma antigen peptide or Bl*0202/melanoma antigen peptide er can be used for the detection and/or isolation by screening (in flow cytometry or by magnetic screening) of T cell population that are specific for a complex HLA-DQBl*0201/ ma antigen peptide or HLA—DQBl *0202/melanoma antigen peptide multimer as described here above.
Another object of the invention is beads coated with MHC/peptide multimers as described here above.
Immunizing composition and uses thereof Another object of the invention relates to an immunising composition comprising: (a) at least one melanoma antigen peptide as described here above or (b) at least one fusion protein as bed here above, or (c) at least one nucleic acid sequence as described here above, or (d) at least one expression vector as described here above, or (e) at least one host cell as described here above, or (t) at least one antibody as bed here above.
In one ment, said immunising composition comprises a melanoma antigen peptide having a ce SEQ ID NO: 1 to SEQ ID NO: 5.
The lactic administration of the immunizing composition of the invention should serve to t or attenuate melanoma in a mammal. In a preferred embodiment mammals, preferably human, at high risk for melanoma are prophylactically treated with the immunising composition of the invention. Examples of such mammals e, but are not limited to, humans with a family history ofmelanoma.
When provided eutically, the immunising composition of the invention is provided to enhance the patient's own immune se to the melanoma antigen present on the melanoma or metastatic melanoma.
In one embodiment of the invention, the peptides of the invention may be conjugated with lipoprotein or administered in liposomal form or with adjuvant.
In one embodiment, said immunising composition is a pharmaceutical composition.
In such embodiment, said immunising composition, for human use, comprises at least one n peptide as described here above or at least one antibody as described here above, together with one or more pharmaceutically acceptable carriers and, optionally, other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. The immunising compositions may conveniently be presented in unit dosage form and may be prepared by any method well-known in the ceutical art.
Immunising compositions suitable for intravenous, intradermal, intramuscular, subcutaneous, or intraperitoneal administration conveniently comprise sterile aqueous ons of the active agent with solutions which are preferably isotonic with the blood of the recipient. Such itions may be conveniently prepared by dissolving solid active ient in water containing physiologically compatible substances such as sodium chloride (eg. 0.1-2.0M), glycine, and the like, and having a buffered pH compatible with physiological conditions to produce an aqueous solution, and rendering said solution sterile. These may be present in unit or multi—dose containers, for example, sealed ampoules or Vials.
The immunising compositions of the invention may incorporate a stabilizer. rative stabilizers are polyethylene glycol, proteins, saccharides, amino acids, inorganic acids, and organic acids which may be used either on their own or as ures. These stabilizers are preferably incorporated in an amount of 0.11-10,000 parts by weight per part by weight of active agent. If two or more stabilizers are to be used, their total amount is preferably within the range specified above. These stabilizers are used in aqueous solutions at the appropriate concentration and pH. The specific osmotic pressure of such aqueous ons is generally in the range of 0.1-3.0 osmoles, preferably in the range of 0.8-1.2. The pH ofthe aqueous solution is adjusted to be within the range of 5.0—9.0, preferably within the range of 6-8.
Additional pharmaceutical methods may be employed to control the duration of action.
Controlled release preparations may be achieved through the use of polymer to complex or absorb the peptides of the invention. The controlled ry may be exercised by selecting appropriate macromolecules (for example polyester, polyamirio acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) and the concentration of macromolecules as well as the methods of incorporation in order to control release. Another possible method to control the duration of action by controlled-release preparations is to incorporate the antigen peptides of the invention into particles of a polymeric material such as polyesters, ino acids, hydrogels, poly(lactic acid) or ethylene ceiate copolymers. Alternatively, instead of incorporating these agents into polymeric particles, it is possible to entrap these als in microcapsules ed, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxy— methylcellulose or gelatin-microcapsules and poly(methylmethacylate) microcapsules, respectively, or in colloidal drug delivery systems, for example, mes, n pheres, microemulsions, nanoparticles, and nanocapsules or in macroemulsions.
When oral preparations are desired, the itions may be combined with typical carriers, such as lactose, sucrose, starch, talc magnesium stearate, crystalline cellulose, methyl ose, ymethyl cellulose, glycerin, sodium alginate or gum arabic among others.
Immunisation of a t with the immunising composition of the invention can be conducted by conventional s, for example, in the presence of tional adjuvants.
Examples of conventional adjuvant include, but are not limited to, metal salts, oil in water emulsions, Toll like receptors agonists, ns, lipid A, alkyl glucosaminide phosphate, Freund's adjuvant, keyhole limpet haemocyanin (KLH), mannan, BCG, alum, cytokines such as IL—1, IL—2, macrophage colony stimulating factor, and tumor necrosis ; and other substances that act as stimulating agents such as muramyl peptides or bacterial cell wall components, toxins, toxoids and TLR ligands.
The immunising composition can be administered by any route appropriate for antibody production and/or T cell activation such as intravenous, intraperitoneal, intramuscular, aneous, and the like. The immunising composition may be stered once or at periodic intervals until a significant titre of anti-Nectin4 immune cells or anti- 4 antibody is produced. The presence of anti-Nectin4 immune cells may be assessed by measuring the frequency of precursor CTL ic T-lymphocytes) against the antigen peptides of the invention prior to and after immunization by specific tetramer labelling or by a CTL precursor analysis assay. The antibody may be detected in the serum using an immunoassay.
Antibodies ed to the antigens of the invention can also be used directly as anti- melanoma agents. To prepare antibodies, a host animal may be zed using melanoma antigen e as described here above. The host serum or plasma is collected following an riate time to provide a composition comprising antibodies reactive to said n peptides. The gamma globulin fraction or the IgG antibodies can be ed, for example, by use of saturated ammonium sulfate or DEAE Sephadex, or other techniques known to those skilled in the art. The antibodies are substantially free of many of the adverse side effects which may be associated with other anti-cancer agents such as chemotherapy.
The immunising composition ofthe invention comprising antibodies as described here above can be made even more compatible with the host system by minimizing potential adverse immune system responses. This is accomplished by removing all or a portion of the Fc portion of a foreign species antibody or using an dy of the same species as the host patient, for example, the use of antibodies from human/human hybridomas. Humanized antibodies (i.e., nonimmunogenic in a human) may be produced, for e, by replacing an immunogenic portion of an antibody with a ponding, but nonimmunogenic portion (i.e., chimeric antibodies). Such chimeric antibodies may contain the reactive or antigen binding portion of an antibody from one species and the Fc portion of an antibody munogenic) from a ent species. Examples of chimeric antibodies, include but are not limited to, an mammal-human chimeras, rodent—human chimeras, murine—human and rat-human chimeras.
Methods for obtaining said antibodies, chimeric antibodies and humanized chimeric antibodies are well—known in the art.
The immunising composition comprising the antibodies of the invention can also be used as a means of enhancing the immune response. The antibodies can be administered in amounts similar to those used for other therapeutic administrations of antibody. For example, pooled gamma globulin is administered at a range of about 1mg to about 100mg per patient.
Thus, antibodies reactive with the antigen peptides of the invention can be passively administered alone or in conjunction with other anti-cancer therapies to a mammal afflicted with cancer. Examples of anti-cancer therapies include, but are not limited to, chemotherapy, radiation therapy, adoptive immunotherapy therapy with TIL.
The antibodies or chimeric antibodies described herein may also be coupled to toxin molecules, sotopes and drugs by conventional methods. Examples oftoxins to which the antibodies may be coupled to included, but are not limited to, ricin or diphtheria toxin. es of drugs or chemotherapeutic agents include, but are not limited to, cyclophosphamide or doxorubicin. Examples of radioisotopes, include, but are not limited to, 1311. Antibodies covalently conjugated to the aforementioned agents can be used in cancer immunotherapy for treating melanoma.
If the patient to be immunized is already afflicted with cancer or metastatic cancer, the immunising composition of the invention can be administered in ction with other therapeutic treatments. Examples of other therapeutic treatments includes, but are not limited to, adoptive T cell therapy, coadministration of cytokines or other therapeutic drugs for cancer.
The dose of antigen peptide of the invention to be administered to a patient may be adjusted as appropriate depending on, for example, the disease to be treated, the age and the body weight of said patient. Ranges of n peptides of the invention that may be administered are about 0.001 to about 100 mg per t, preferred doses are about 0.01 to about 10mg per patient.
The immunising composition of the invention may be ted first in animal models, lly rodents, and in an primates and finally in humans. The safety of the immunization procedures is determined by looking for the effect of immunization on the general health of the immunized animal (weight change, fever, appetite behavior etc.) and looking for pathological changes on autopsies. After initial testing in animals, cancer patients can be tested. Conventional s would be used to te the immune se of the patient to ine the efficiency of the immunising composition.
Another object of the invention s to an immunising composition as described above for use in the prevention or treatment of melanoma in a patient in need thereof.
In another embodiment, said patient is genotyped with HLA-DQB1*0201 or HLA- DQ[31*0202 alleles.
Antigen presenting cell Another object of the invention is an antigen presenting cell sing a complex HLA n and a melanoma antigen peptide ofthe invention.
In one embodiment of the invention, said x HLA antigen is a HLA- DQBI*0201 or HLA-DQBI *0202 antigen.
In one embodiment of the invention, said antigen presenting cell is derived from the patient to be d.
The term "antigen presenting cell" (APCs) refers to any cell that expresses an HLA antigen capable of presenting the n peptide of the invention on its surface. Dendritic cells, which are reported to have an especially high antigen-presenting ability, are preferred.
In another embodiment, artificial APCs may also be used such ian cells (fibroblast, endothelial cells, keratinocytes), or cell lines.
In order to prepare such APCs of the invention, cells having an antigen-presenting ability are isolated from the patient to be treated, and pulsed ex vivo with at least one antigen e of the ion to form a complex with the HLA-DQB1*0201 or HLA-DQB1*0202 antigen.
In case dendritic cells are used, the APC of the invention can be prepared as follows.
Lymphocytes are isolated from peripheral blood of the patient to be treated by Ficoll method; adherent cells are separated from non-adherent cells; the adherent cells are then cultured in the presence of GM-CSF and IL-4 to induce dendritic cells; and the dendritic cells are pulsed by culturing with at least one antigen peptide of the invention to obtain the APCs of the invention. The dendritic cells should be exposed to the antigen peptide for sufficient time to allow the antigens to be alized and presented on the dendritic cells surface. The resulting dendritic cells can then be re-administrated to the patient to be treated. Such s are described in WO93/208185 and EP0563485, which are incorporated by reference.
Another object of the invention is a composition for active immunotherapy comprising n presenting cells comprising a complex HLA n and a melanoma antigen peptide ofthe invention.
In one embodiment of the ion, said antigen presenting cells comprise a complex HLA-DQBI *0201 or HLA—DQB1*0202 antigen and a melanoma antigen peptide of the invention.
Said APCs may be preferably ned in physiological saline, phosphate buffered saline (PBS), culture medium, or the like. Administration may be achieved, for example, intravenously, hypodermically, or intradermally.
Lymphocytes T and uses thereof Another object of the invention relates to a T lymphocyte that recognizes specifically the melanoma antigen e ofthe invention or a fusion protein of the invention.
In one embodiment of the invention, said T lymphocyte is a T lymphocyte helper.
In another ment of the invention, said T lymphocyte is HLA-DQB1*0201 or HLA-DQBl *0202 restricted.
In another embodiment of the invention, said T lymphocyte is a T cell Clone.
In another embodiment, said T lymphocyte is a genetically modified T cyte that expresses a TCR that recognizes specifically the melanoma antigen peptide of the invention. r object of the invention is a composition for adoptive therapy sing said T lymphocytes as described here above that recognizes specifically the melanoma antigen peptide of the invention or a filSlOIl protein of the invention.
In the case of melanoma, it has been observed that an adoptive immunotherapy wherein intratumoral T cell infiltrate taken from the patient to be treated are cultured ex vivo in large quantities, and then ed into the patient achieves a therapeutic gain.
It is preferred that the T cells are contained in physiological saline, phosphate ed saline (PBS), culture medium, or the like in order to their stable maintain. Administration may be achieved, for example, intravenously or tumoraly. By returning the T cells that recognizes specifically the antigen peptide of the invention into the patient’s body, the toxicity of said T cells, or the stimulation of CD8 xic T cells by said cells towards tumor cells is enhanced in the patient who is positive for the melanoma n peptide of the invention. The tumor cells are yed and thereby the treatment oftumor is achieved.
Examples of where T-lymphocytes can be ed, include but are not limited to, peripheral blood cells lymphocytes (PBL), lymph nodes, or tumor infiltrating lymphocytes (TIL).
Such lymphocytes can be isolated from tumor or peripheral blood of the individual to be d by methods known in the art and cultured in Vitro. Lymphocytes are cultured in media such as RPMI or RPMI 1640 for 2—5 weeks, preferably for 2—3 weeks. Viability is assessed by trypan blue dye exclusion assay. The cytes are exposed to the antigen peptide of the invention for all of the culture duration.
In a red embodiment the lymphocytes are exposed to the melanoma antigen peptide of the ion at a concentration of about 1 to about 10 micrograms(ug)/ml for all the on of lymphocyte culture. Cytokines may be added to the lymphocyte culture such as IL-2.
The melanoma antigen peptide of the invention may be added to the culture in presence of antigen presenting cells such as dendritic cells or allogeneic ated cancer cell line cells.
After being sensitized to the peptide, the T—lyrnphocytes are administered to the patient in need of such treatment.
Examples of how these sensitized T-cells can be administered to the mammal include but are not limited to, intravenously, intraperitoneally or intralesionally. Parameters that may be assessed to ine the efficacy of these ized T-lymphocytes include, but are not limited to, production of immune cells in the patient being treated or tumor regression.
Conventional methods are used to assess these parameters. Such treatment can be given in conjunction with cytokines or gene modified cells (Rosenberg, S.A. et a1. (1992) Human Gene Therapy, 3: 75-90; Rosenberg, SA. et a1. (1992) Human Gene Therapy, 3: 57-73).
Another object of the invention is a method for producing T cytes that recognize specifically a melanoma n peptide of the invention, said method comprising the steps of: (a) stimulating peripheral blood mononuclear cells (PBMCs) or tumor infiltrating lymphocytes (TIL) obtained from a patient with at least one melanoma antigen peptide of the invention or a fusion protein of the invention, (b) ing the population of T lymphocytes specific for the melanoma antigen peptide(s) used in (a), (c) optionally cloning said population of T lymphocytes specific for the melanoma antigen peptide(s) used in (a).
Enrichment and/or cloning may be carried out by using an MHC/peptide multimer as described here above. g may also be carried out by conventional s.
In one embodiment of the invention, the T lymphocytes that recognize specifically a melanoma antigen peptide of the invention are HLA—DQB1*0201 or HLA-DQBI*0202 restricted. In such embodiment, enrichment and/or cloning may be carried out by using an HLA-DQ[31*0201 or HLA-DQ[31*0202 /peptide multimer as described here above.
Stimulation ofPBMCs may be carried out with at least one melanoma antigen peptide of the invention alone, or presented by an antigen presenting cell such as dendritic cells or allogeneic irradiated cancer cell line cells. Typically, cytokines such as IL-2 may also be added to the culture.
Another object of the invention is a composition for adoptive y that comprises lymphocytes that recognizes specifically the melanoma antigen peptide of the invention for preventing or treating melanoma in a patient in need f, wherein said T lymphocytes are to be re-administrated to the patient.
In one embodiment, said lymphocytes that recognizes specifically the antigen peptide ofthe invention are HLA-DQB1*0201 or HLA-DQ[31*0202 restricted.
The invention also relates to a method for preventing or treating ma in a patient in need thereof, comprising administering a therapeutically effective amount of (a) at least one melanoma antigen peptide as described here above or (b) at least one fusion n as described here above, or (c) at least one nucleic acid sequence as described here above, or (d) at least one expression vector as bed here above, or (e) at least one host cell as described here above, or (f) at least one antibody as described here above.
The invention also relates to a method for preventing or treating melanoma in a patient in need f, comprising administering a therapeutically ive amount of T lymphocytes that recognizes specifically the ma antigen peptide of the invention. In one ment, said T lymphocytes are Bl *0201 or HLA-DQB1*0202 restricted.
The invention also relates to a method for preventing or treating melanoma in a patient in need f, comprising administering a therapeutically effective amount of antigen presenting cells sing a complex HLA antigen and a ma antigen peptide of the invention. In one embodiment, said complex HLA/peptide is a complex HLA-DQB1*0201 or HLA-DQB1*0202 / antigen peptide of the invention.
The invention will be further rated by the following figures and examples.
However, these examples and figures should not be interpreted in any way as ng the scope of the present invention.
FIGURES: Figure 1: (A) Percentages of TNF-Ot producing CD4 T cells among positive ultures. Fourteen days after PBMC stimulation with MELOE-l whole polypeptide (2- 46), microcultures were re—stimulated with each indicated peptide during 5 hours. TNF-Ot production was then assessed by a double labeling TNF-Ot -CD4. Results were analyzed with a non—parametric test (Kruskal—Wallis) followed by a Dunns post—test. (B) Frequency of microcultures containing CD4 T cells specific for MELOE-l peptides (assessed by TNF-(l intracellular staining after re-stimulation with the 4 indicated peptides). 624 microcultures (from 7 healthy donors) were analyzed by a contingency table ed by a Fisher exact test.
Figure 2: (A) tages of cytokine producing CD4 T cells among positive microcultures. Fourteen days after PBMC stimulation with MELOE-l whole polypeptide (2- 46), microcultures were re-stimulated with each indicated peptide during 5 hours. IFN—y (left panel) and IL4 (right panel) production was then assessed by a triple ng IFN—y -IL4- CD4. Results were analyzed with a non-parametric test (Kruskal-Wallis) followed by a Dunns post-test. (B) Frequency of microcultures ning CD4 T cells specific for l peptides (assessed by IFN—y or 1L4 intracellular staining after re-stimulation with the 3 indicated peptides). 576 microcultures (from 10 melanoma patients) were analyzed by a contingency table followed by a Fisher exact test.
Figure 3: HLA-restricting element of MELOE-l c T cell clones and reactivity against HLA-matched melanoma cell lines. The HLA restriction of MELOE—l specific T cell clones was first assessed using anti-HLA blocking antibodies (upper panel). T cell clones were stimulated for 5 hours in the presence of din A (10 ug/mL) either with peptide alone (10 uM) in an autopresentation assay and in presence or not of blocking antibodies at a concentration of 12.5 ug/rnL. HLA restriction was confirmed with HLA-matched B—EBV cell lines pulsed 2 hrs with the cognate peptide, at a ratio 1: 2 (middle panel). Reactivity of each T cell clone against HLA-class II expressing melanoma cells was assessed, in presence or not of exogeneous peptide (lower panel). After 5 hours of stimulation, cells were then stained with njugated anti-CD4 mAb, fixed with 4% paraformaldehyde, labeled with PE- conjugated anti—TNF—or mAb and analyzed by flow cytometry.
Figure 4: Class II epitopes are naturally processed from MELOE-l whole antigen. gous DC, were loaded (before or after fixation) with MELOE-12_46 (1 uM) or, as a ve control, with AlMOL peptide (1 uM), and matured. T cell clones were then stimulated with DC at a ratio 1: 1, during 5h in presence of Brefeldin A, then stained with APC-conjugated anti-CD3 mAb, fixed with 4% paraformaldehyde, labeled with PE- conjugated anti-TNF-or mAb and analyzed by flow cytometry. Histograms illustrated the % of TNF-OL producing cells among CD3 positive T cells.
Figure 5: l peptides recognized by MELOE—l specific CD4 T cell clones.
MELOE-l specific CD4 T cell clones were incubated with various concentrations of the indicated es during 5 hours in presence of Brefeldin A. TNF-OL production was ed by intracellular labeling with an anti-TNF-(x specific antibody. The core peptide sequence is indicated in bold on each figure panel, and black circles rate the best fitting peptide.
MELOE-l MSCVGYPDEATSREQFLPSEGAACPPWHPSERISSTLNDECWPASL MELOE-12.21 SCVGYPDEATSREQFLPSEG l MELOE-111_30 TSREQFLPSEGAACPPWHPS 10 118_37 PSEGAACPPWHPSERISSTL 16 MELOE'126-46 PWHPSERISSTLNDECWPASL 6 MELOE-122.45 AACPPWHPSERISSTLNDECWPA SL 17 Table I: MELOE—l and l derived peptide sequences.
All the peptides were sed from Millegen y (France), with a purity > 85%. In bold are indicated the DR—ll (SEQ ID N018) and the DQ—6 (SEQ ID NO:8) overlapping epitopes already described, and in italics is indicated the HLA-A2 restricted class I epitope (TLNDECWPA, SEQ ID NO:23)). ultures containing MELOE-l specific CD4+ T cells TNF-(x roduction MELOE-111_ MELOE-118_ 126_ MELOE-IMI Class II HLA 37 46 DPB 1 *0902/1501 9/96 13/96 6/96 5/96 DQBl*0301 (1.4%::O.6) (2.4%115) (1.4%i0.6) (1.5%:05) DRBl*1104/1201 DPBl*0401/0301 6/96 26/96 7/96 10/96 HD” DQBl*0301/0603 (3.2%:19) (8.2%i6.5) (30/0107) (5.2%:41) DRBl*1101/1301 ngglfioligéém* 1/96 2/96 2/96 HD22 0/96 (1%) (3.9%:t3.3) (1%i0-1) DRBl*0701 DPBl*0401/0101 HD24 0/96 0/96 DQBI*0501/0602 (1 33:60 8). 0 . 0:619?”0 ~ DRBI*0101/1501 mm 0/96 (0 63/9160. nggBolglz/gioz o . 1) (1 23/9160 7). o . (1 ' 11/5260 9) ° - 0301 DPBl NA /96 HD27 0/96 0/96 0/96 DQBI*501/0201 (50/ i 3 4)o - DRBl*0101/0301 DPBI NA /48 14/48 0/48 Hms 0/96 DQNA Table 11: Assessment of MELOE—l CD4 T cell responses in PBMC from y donors.
PBMC from healthy donors were stimulated with 10 uM of MELOE-l. Afier 14 days, the ce of CD4 T cells specific for the different regions of l was assessed by restimulating cells with 12_21, MELOE-111_30, MELOE—118_37 and 125_46 peptides, followed by CD4 / TNF—(x double staining and flow cytometry analysis. Between brackets is indicated the mean % of TNF—oc producing CD4 T cells, in positive microcultures.
NA : not available.
Microcultures containin_ MELOE-l s l ecific CD4+ T cells Th2 responses (1L4 positive m1crocultures) mlcrocultures) -MELOE- MELOE- MELOE—ln MELOE- MELOE- MELOE- 12-21 111-30 46 12-21 111-30 122-46 1/48 10/48 11/48 1/48 4/48 0/48 (0.7%) (1.4%407) (4.8%i72) (1%) (0.8%::0.3) 4/96 16/96 2/96 ”6 0/96 0/96 .2) (1.6%i1.2) (0.6%::0.02) Pt¢3 DPBl*0201/ 2001 5/96 4/96 DQ1315001303/* (16.24133.0 0/96 0/96 - - (1.9%i1.3) DRBl*0101/ 9/48 1/48 (”48 0/48 0/48 (0.9%405) (07%) 1/48 6/48 2/48 1/48 (0.6%) 1.1)-(0.7%40.1> “8 (0.8%) /48 0/48 0/48 0/48 0/48 0/48 (1.5%2‘21-9) 28/48 7/48 (2.9%43.5)(1.5%4=0.7> 2/48 1/48 1/48 9/48 3/48 l/48 Ptyé9 0/48 0/48 0/48 0 (0.6%i0.09 (0.7%:t0,15 ) ) Table III: Assessment of MELOE-l CD4 T cell responses in PBMC from melanoma patients.
PBMC from melanoma patients were stimulated with 10 MM of MELOE—l. After 14 days, the presence of CD4 T cells specific for the different regions of MELOE-l was assessed by restimulating cells with MELOE-12_21, MELOE-111_30 iVI-E-LQE—l-w and MELOE-122_46 peptides, followed by CD4/IFN—y double staining for the detection of Th1 responses, and by CD4/1L4 double staining for Th2 responses. n brackets is indicated the mean % of cytokine producing CD4 T cells, in ve microcultures.
MELOE21 Specific CD4 T cell clone (9C12- DQBI*0202) TNF 1g GM-CSF 1g 1L4 'g MELOE-111_30 s.ecific CD4 T cell clone I51 *0101 V beta chain a% "g CDR3 beta IFN ‘ J beta chain 1L2 °W GM-CSF 03 1L4 03 l—tl—t hr5‘”negH(IO ILlO neg MELOE-125_46 specific CD4 T cell clone (4E2- DQB 1 *0201) TNF "g GM-CSF 1g D—l|—( rr‘m4}- _%oa(to IL13 ILlO neg Table IV: TCR characterization and cytokine profile of MELOE-l specific CD4 T cell clones CD4 T cell clones were stimulated for 5 hours in the presence of brefeldin A (10 ug/mL) either with the cognate peptide (10 uM) in an autopresentation assay. After 5 hours of stimulation, cells were stained with AFC-conjugated D4 mAb, fixed with 4% paraformaldehyde, labeled with PE- conjugated anti-cytokine mAb and analyzed by flow cytometry.
Material & Methods Cells Blood samples from healthy subjects and melanoma patients were respectively obtained from ssement is du Sang, Nantes, France and from the department of onco-dermatology, Nantes Hospital, France. Melanoma and B-EBV cell-lines were maintained in RPMI 1640 ) containing 10% fetal calf serum (FCS). cytes were grown in RPMI 1640 8% human serum (HS) with 50 or 75 IU/ml of recombinant interleukin-2 (IL-2, Chiron, France) and 2nM of L—Glutamin. For experiments using dendritic cells (DC), RPMI supplemented with 20mg/mL of human albumin (LFB BIOMEDICAMENTS, France) was used to avoid peptide degradation by serum proteases.
Reagents Antibodies were purchased fiom BD Biosciences-France or from Miltenyi Biotec, France. Purified cytokines were purchased from CellGeniX, Germany. The different peptides (Millegen, France, purity >85%) used in this study are described in Table I. HLA- A*0201/MELOE-136,44 rs were generated by the recombinant protein facility of our institute (SFR 26).
Dendritic cells tion and loading Monocytes were purified from PBMC of healthy donors by a CD14-enrichment kit, according the recommendations of the supplier (Stem Cell, France). Immature dendritic cells (iDC) were generated by culturing monocytes in RPMI supplemented with 20mg/mL of human albumin, 1000 IU/mL of GM-CSF and 200 IU/mL of 1L-4 for 5 days. Then, iDC were pulsed with the whole MELOE-l (luM) n or the ed Melan—A16_40 A27L as ve control (1 uM) and matured with 20 ng/mL ofTNF-0t and 50 ug/mL of PolyI:C for 4 hours at 37°C. Finally they were fixed for 1 minute with PBS/0.015% glutaraldehyde. atively, iDC were first matured, fixed and then pulsed with antigens at the same concentration.
Stimulation ofMELOE-l specific T cells PBMC from healthy donors or melanoma patients (2.105 cells/well) were cultured for 14 days with 10 uM of MELOE-l whole antigen (46 amino acids) in RPMI medium supplemented with 8% HS, 50 IU/ml of rIL-2 (Chiron, France) and L-Glutamin, in 96—well multiplates. Microcultures were then restimulated individually with each pping peptide (MELOE-12_21, MELO E—111_3o, MELOE-l 1837, MELOE-126_46 or MELOE-122_45 for melanoma patients) in the presence of 10 ug/mL brefeldin A for 5 hours and the percentage of CD4+ specific T cells was assessed by TNF-0t, IFN—y or 1L-4 intracellular staining. A negative control without e was included in all experiments.
Alternatively, MELOE-l specific CD4+ T cell clones were stimulated by autologous MELOE-l loaded and matured DC at a 1:1 ratio.
T cell cloning and TCR characterization Polyclonal cultures containing specific CD4+ T cells were cloned by limiting dilution as previously bed (Gervois N. et al., 2000). After 2 weeks, each clone was checked for peptide specificity by TNF production assay. For TCR sequencing, RNA from 5.106 T cell clones was extracted with RNable reagent (Eurobio, ) according to the supplier's ctions. Reverse transcriptions, PCR cations and cing were performed as described (Davodeau F. et a1, 2001). We used the TCR nomenclature established by Arden et al. (Arden et al., 1995); TNF production assay CD4+ T cell clones were cultured for 5 hours at 37°C in the presence of the recognized -mer peptide. e supernatants were harvested and TNF was measured in a biological assay using cytotoxicity against WEHI 164 clone 13 (Espevik T. et al., 1986).
Cfiokine intracellular staining Lymphocytes were stimulated for 5 hours in the presence of brefeldin A (10 ug/mL) either with peptide alone (10 uM) in an autopresentation assay or with B—EBV or HLA-class II expressing melanoma cells pulsed 2 hours with the cognate peptide, at a ratio 1: 2. In some ments, blocking mAb against HLA-DP (clone B7.21 from Dr n, UMR940, Paris), HLA—DQ (clone SPVL3, Beckman Coulter) or HLA—DR (clone L243, BD Biosciences) were added at a concentration of 12,5 ng/ml. Cells were then stained with APC- conjugated anti-CD4 mAb, fixed with 4% paraformaldehyde, labeled with PE-conjugated anti-cytokine mAb and analyzed by flow cytometry.
Statistical analyses Statistical analyses were done with GraphPad Prism® software. Bar graphs were used to compare ncies of T cells specific for MELOE-l—derived es, in all donors and patients and were analyzed by a contingency table followed by 21 Fisher exact test. Scatter-dot graphs were made to compare the percentage of TNFOL positive cells among positive microcultures and were analyzed with a non-parametric test (Kruskal—Wallis followed by a Dunns post-test).
Frequency and distribution of MELOE-l specific CD4 responses in healthy donor’s PBMC stimulated with l n Our purpose was to look for the nce of class II helper epitopes all along MELOE-l sequence (SEQ ID NO: 20), in order to document the immunogenicity of the different regions of this melanoma antigen. We stimulated 2.107 PBMC from seven healthy donors with MELOE-l whole n and tested, after a 14-day culture period the presence of CD4 T cells specific for each region of the protein. Microcultures were screened for TNFOL production by CD4+ T cells, after restimulation with four MELOE-l derived overlapping peptides (Table I), in an autopresentation assay. As shown in table II, all donors exhibited CD4 responses against at least 1 out of 4 overlapping peptides. ses against the N- terminal region of MELOE-l (2-21) were detected in 6/7 donors, with rather low frequencies (from 1 to 9% of positive microcultures containing between 0.6 to 5.6% of TNFOL producing CD4 T cells), unless in HD28 healthy donor, who exhibited 62% of positive microcultures.
The region 11-30 appears ally immunogenic, with CD4 specific responses detected in each tested donor (from 2 to 48% of positive microcultures containing between 0.7 to 24 % of TNFO. producing CD4 T cells), and with very high frequencies in three donors (HDl7, HD24 and HD28). On the contrary, the central region 18—37, containing an already described DRl l- restricted epitope (24-37) located just at the end of this 20-mer peptide (Rogel et al., 2011), induced specific responses in microcultures deriving from only 2/7 donors (HD9 and HD17, both expressing the DRII element). In these two donors, we detected 6 and 7% of positive microcultures, containing between 0.6 to 3.7 % of TNFoc producing CD4 T cells. Finally, the C-terminal region (26-46), containing an already described DQ6-restricted epitope (Rogel et al., 2011), was recognized by ated microcultures from 4 out of 7 donors (all do not expressing the DQ6 element), with frequencies ranging from 2 to 16% of microcultures containing between 0.5 and 16% of TNFOt producing CD4 T cells. Overall, the frequency of MELOE-111_3o ve microcultures was significantly higher than frequency of ultures specific for the three other regions of MELOE-l (Figure 1B) (p<0.0001). The two terminal regions (2-21 and 26-46) were equivalent in terms of frequencies of positive microcultures, and these two regions induced significantly more responses than the central region 18-37 (Figure 1A). Nonetheless, the mean fractions of CD4 reactive T cells induced in positive microcultures were not significantly ent from a region to another (Figure 1B).
Frequency, distribution and Th profile of MELOE-l specific CD4 responses in melanoma patient’s PBMC stimulated with MELOE-l antigen In order to confirm the immunogenicity of each MELOE-l s in melanoma patients, we stimulated melanoma ts PBMC with the MELOE-l whole protein, and tested the reactivity of ated lymphocytes towards the three most immunogenic regions: 2-21, 11-30, and 22-46. For this study, instead of challenging microcultures with the 18-37 e, that appeared poorly immunogenic, we extended the C—terminus region from 26-46 to 22—46, in order to also detect ses to our previously described HLA—DRll—restricted epitope (24-37). Indeed, the location of this epitope just at the end of the 18-37 e could be deleterious for the detection of c responses in onal DR contexts, and we previously showed that CD4 T cells c for MELOE-lz4_36 epitope were efficiently induced by 22-46 peptide stimulation (Rogel et al., 2011). We tested the ion of CD4 specific responses from MELOE-l stimulated PBMC of 10 melanoma patients. We documented CD4 responses c for the central region of MELOE-l (11-30) for 7/9 patients, whereas responses specific for MELOE-12_21 and MELOE-122_46 were respectively detected in 4/9 and 5/9 patients (Table 111). These responses were mainly Thl responses (IFN— g production) while less frequent Th2 responses c for the three regions of MELOE-l were detected in 3/9 patients for 12_21, 2/9 patients for MELOE-111_30 and 5/9 patients for 122_46 (Table III). Considering the various regions of MELOE-l, Thl responses specific for the N—term region of MELOE-l (2-21) were significantly less frequent than those specific for the central region (p<0.0001) and the C-term region 001), with respectively 3.3%, 10.8% and 9.6% of 576 tested microculutures (Figure 2A). Concerning Th2 responses, much less frequent, the C-term region appeared to induce more frequently the growth of lL-4 producing CD4 T cells than the two other regions (Figure 2A). As observed for healthy donors, even if the frequencies were different, the mean fiaction of reactive T cells (Thl and Th2) induced in positive microcultures were not significantly ent from a region to another (Figure 2B). In summary, stimulation of patient’s PBMC with MELOE-l induced Thl responses specific for diverse epitopes located all along the protein sequence, and among the different regions, the central region (1 1—3 0) and the C-term region (22-46) appeared to be especially genic in term of frequency of responses.
Production and characterization of CD4 T cell clones specific for the different regions In order to ly characterize the recognized epitopes, we derived CD4 T cell clones specific for each region of MELOE—l by ng dilution, from microcultures of healthy donors or melanoma patients, ning at least 0.5% of specific CD4 T cells. We succeeded to derive CD4 specific T cell clones from HD17, HD22, HD25 and Pt¢3 microcultures, which were reactive against MELOE-12_21 (HD22), MELOE-111_30 (HDl7 and Pt¢3) and MELOE-lg6_46 (HD25). From each g experiment, we obtained between one and ten reactive CD4 T cell clones, that turned out to be the same clonotype after CDR3B sequencing (Table IV). A single CD4 T cell clone for each specificity was used for further experiments. The HLA-restriction was determined for each T cell clone, first by using HLA- class II blocking monoclonal antibodies (Figure 3, upper panel), and further by testing the recognition of HLA—matched B—EBV cell lines loaded with each recognized long e (Figure 3 middle panel). The two T cell clones named 9Cl2 and 4E2, derived from HD22 and HD25 and specific for MELOE-12_21 and MELOE—12646 were restricted by the HLA- DQBl*0202 and the 201 molecules respectively. As, these two donors were homozygous for the HLA—DQ locus, a single HLA-DQ matched B—EBV cell line was tested to confirm the HLA restriction of these two CD4 T cell clones. As shown on figure 3 (upper , the two other T cell clones 1A5 and 5F9 recognized the 11-30 region of MELOE-l, in a HLA-DR context. The use of HLA-matched B-EBV cell lines allowed to precise that 1A5 T cell clone was restricted by the B] *1 101 molecule and the 5F9 T cell clone by the HLA—DR131*0101 molecule.
We further tested the reactivity of these CD4 T cell clones against HLA—matched melanoma cell lines positive for meloe expression, by qPCR is. All the ma cell lines tested expressed HLA-DQ and DR at the cell surface. All the T cell clones were reactive against HLA-matched melanoma cell lines when loaded with the cognate e (Figure3, lower panel, black bars). The two DQ2-restricted T cell clones were reactive against peptide- loaded DQBl *0201 and 0202 melanoma cell lines. In absence of peptide, only the 4E2 DQ 131*0201-restricted T cell clone was able to recognize unloaded M77 melanoma cell line (also DQBl *0201), but not the DQ131*0202 ma cell line, M88. Similarly, the DRl-restricted T cell clone 5F9 also recognized one ofthe DR131*0101 ma cell line, in the e of exogenous peptide (M101).
We also documented the T helper profile of each T cell clone, by stimulating the CD4 T cell clones with the cognate peptide, and analyzing cytokine production. All the clones sed Th1 cytokines (TNFor, IFNy, IL2 and GM-CSF). On the contrary, these clones differ in their expression of Th2 cytokines. Indeed, the two DQ2 restricted T cell clones (9C 12 and 4E2) and the DRl restricted one (5F9) also strongly express two Th2 cytokines (IL4, and IL13), whereas the DRl l-restricted T cell clone only weakly expressed IL4 (Table IV). None ofthe CD4 T cell expresses ILlO or 1L5 at a significant level.
Processing of the recognized epitopes from autologous DC loaded with MELOE-l antigen Initial PBMC stimulation was carried out with MELOE—l whole antigen, and thus we assume that CD4 T cell responses were generated against peptides naturally processed by monocytes. Nonetheless, we could not formally exclude that the 14-day culture period artificially generated shorter class II epitopes that elicited CD4 T cell responses. Thus, it remained l to assess that all these new epitopes were naturally processed by autologous dendritic cells loaded with MELOE-l whole protein, in serum-free medium. To this end, we loaded gous iDC with 1 uM of MELOE—l antigen in serum-free medium, in presence of maturating agents, and fixed these DC before ation of the CD4 specific T cell . In these conditions, the four T cell clones were reactive against MELOE-l loaded autologous DC (Figure 4, left panel), whereas we could not detect any reactivity of CD4 T cell clones to DC loaded with an irrelevant long peptide, synthesized in the same conditions (Melan—A16_ 40L).
As an additional control, we loaded autologous DC with MELOE-l after DC fixation, and we could observe only a weak recognition by specific CD4 T cell , indicating that only a small fraction of the protein had been externally degraded into shorter peptides (Figure 4, right panel). Thus, the four new epitopes identified by PBMC stimulation, are naturally processed from MELOE-l n.
Characterization of the minimal recognized epitopes Our T cell clones were reactive against 20-mer peptides that are probably not the exact peptides naturally processed. In order to formally identify the minimal recognized epitopes, we tested shorter peptides derived from each of the MELOE-l recognized regions, chosen on the basis of the core peptide ce ed to be recognized by the T cell clones (indicated in bold on figure 5).
Three shorter peptides were better recognized by the DQ2-restricted 9C12 T cell clone, the st one being MELOE-17_19 (13-mer), recognized with an ECSO of 100 nM.
The deletion of the two amino acids in C-term strongly reduces T cell clone ition. The other DQ2-restricted T cell clone (4E2) better recognized a l4-mer peptide (31-44) also with an ECSO of 100 nM, and also recognized to a lower extent the 32-44 epitope (Figure 5) previously described in the HLA-DQBI*0603 context (Rogel et al., 2011). Concerning the DR-restricted T cell clones, optimal shorter es were 13-mer peptides, MELOE-l 15,27 for the DR131*1101 restricted T cell clone 1A5 (EC50=100 nM), and MELOE-111_23 or MELOE- 112_24 for the DRBl*0101 restricted one e 5). Nonetheless all of these clones recognized a series of ned peptides, thus we cannot formally assess that the shortest ones will be the exact es naturally presented on class II molecules.
REFERENCES: hout this application, various references describe the state of the art to which this invention pertains. The disclosures of these nces are hereby incorporated by reference into the present disclosure.
Arden B, Clark SP, Kabelitz D, and Mak TW Human T-cell receptor variable gene segment families. Immunogenetics 1995; 42: 455—500.
Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF, and Heath WR Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature 1998; 393: 478-80.
Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, Offringa R, and van der Burg SH CD8+ CTL priming by exact peptide epitopes in incomplete Freund's adjuvant induces a vanishing CTL response, whereas long peptides induce ned CTL reactivity. J Immunol 2007; 179: 5033-40.
Chauvin JM, Larrieu P, Sarrabayrouse G, t-Blondel A, Lengagne R, Desfrancois J, et al. HLA Anchor Optimization of the Melan-A-HLA—AZ Epitope within a Long Peptide Is ed for Efficient Cross—Priming of Human Reactive T Cells. J Immunol2012; 188: 0.
Davodeau F, Difilippantonio M, Roldan E, en M, Casanova JL, Couedel C, et al. The tight interallelic positional coincidence that distinguishes T-cell receptor Jalpha usage does not result from homologous chromosomal pairing during ValphaJalpha rearrangement.
EMBO J 2001; 20: 4717-29.
Espevik T and Nissen—Meyer J A highly sensitive cell line, WEHI 164 clone 13, for measuring cytotoxic factor/tumor necrosis factor from human monocytes. J Immunol Methods 1986; 95: 99-105.
Gervois N, Labarriere N, Le Guiner S, Pandolfino MC, Fonteneau JF, Guilloux Y, et al. High y melanoma-reactive cytotoxic T lymphocytes are efficiently induced from peripheral blood lymphocytes on ation by peptide-pulsed melanoma cells. Clin Cancer Res 2000; 6.‘ 1459-67.
Godet Y, Desfrancois J, Vignard V, Schadendorf D, Khammari A, Dreno B, et al.
Frequent occurrence of high affinity T cells against MELOE-l makes this antigen an tive target for melanoma immunotherapy. Eur J Immunol 2010; 40: 1786-94.
Godet Y, Moreau-Aubry A, Guilloux Y, Vignard V, Khammari A, Dreno B, et a1.
MELOE-l is a new antigen overexpressed in melanomas and involved in ve T cell transfer efficiency. J Exp Med 2008; 205: 2673-82.
Hunder NN, Wallen H, Cao J, cks DW, Reilly JZ, Rodmyre R, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against -l. N Engl J Med 2008; 358: 2698-703.
Fayolle C, Deriaud E, and Leclerc C In vivo induction of cytotoxic T cell response by a free synthetic peptide requires CD4+ T cell help. J l 1991; 147: 4069-73.
Friedman KM, Prieto PA, Devillier LE, Gross CA, Yang JC, Wunderlich JR, et al. specific CD4+ ma Tumor—infiltrating Lymphocytes. J Immunother 2012.
Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, et al. Vaccination against HPV-l6 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009; 361: 1838-47.
Robbins PF, El-Gamil M, Li YF, Zeng G, Dudley M, and erg SA le HLA class II-restricted melanocyte differentiation antigens are recognized by tumor- infiltrating lymphocytes from a patient with melanoma. J Immun012002; 169: 7.
Rogel A, Vignard V, Bobinet M, Labarriere N, and Lang F A long peptide from MELOE-l contains multiple HLA class II T cell epitopes in addition to the HLA-A*0201 epitope: an attractive candidate for melanoma vaccination. Cancer Immunol ther 2011; 60: 327-37.
Rosenberg SA, Yang JC, and Restifo NP Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004; 10: 909-15.
Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, et al. gplOO peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med 2011; 364: 2119-27.
Smith CM, Wilson NS, Waithman J, Villadangos JA, Carbone FR, Heath WR, et al.
Cognate CD4(+) T cell licensing of dendritic cells in CD8(+) T cell immunity. Nat Immunol 2004; 5: 1143-8. ens FM, Kuppen PJ, Welters MJ, Essahsah F, Voet van den Brink AM, Lantrua MG, et al. Induction of p53-specific immunity by a p53 tic long peptide vaccine in patients treated for metastatic colorectal . Clin Cancer Res 2009; 15: 5.
Toes RE, ga R, Blom RJ, Melief CJ, and Kast WM Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc Natl Acad Sci U S A 1996; 93: 7855-60.
Vesely MD, Kershaw MH, Schreiber RD, and Smyth MJ Natural innate and adaptive immunity to cancer. Annu Rev l 2011; 29: 235-71.
Welters MJ, Kenter GG, Piersma SJ, Vloon AP, Lowik MJ, Berends-van der Meer DM, et a1. Induction of tumor—specific CD4+ and CD8+ T—cell immunity in cervical cancer patients by a human papillomavirus type 16 E6 and E7 long peptides vaccine. Clin Cancer Res 2008; 14: 178-87.
I

Claims (16)

WE CLAIM:
1. Use of a melanoma antigen peptide of less than 20 amino acids long comprising the amino acid sequence SEQ ID NO:5 in the manufacture of a medicament for the prevention or the treatment of melanoma in a patient.
2. Use of a melanoma n peptide according to claim 1, wherein said peptide is an amino acid ce of less than 15 amino acids long, or wherein said peptide consists of the sequence SEQ ID NO:5
3. Use of a fusion n comprising a melanoma antigen peptide as defined in any one of claims 1 or 2 and a melanoma antigen peptide sing the amino acids motif: - TX2NDECWPX9 (SEQ ID NO: 23) n X2 is leucine, methionine, valine, isoleucine or glutamine and X9 is alanine, valine or leucine in the manufacture of a medicament for the prevention or the ent of melanoma in a patient.
4. Use of a melanoma antigen peptide according to any one of claims 1 or 2 or a fusion protein according to claim 3, n the patient is genotyped with HLA-DQβ1*0201 or HLA- DQβ1*0202 alleles.
5. Use of a T lymphocyte that recognizes specifically a melanoma n peptide as d in any one of claims 1 or 2 in the manufacture of a medicament for the prevention or the treatment of melanoma in a patient.
6. Use of a composition comprising T lymphocytes according to claim 5, wherein said T lymphocytes that recognize specifically a melanoma antigen e or a fusion protein are to be re-administrated to the patient.
7. A method for producing said T lymphocytes as defined in claim 5, said method sing the steps of: (a) stimulating PBMCs or tumor infiltrating cytes obtained from a patient with at least one melanoma antigen peptide according to any one of claims 1 or 2 or a fusion protein according to claim 3, (b) enriching the population of T lymphocytes specific for the melanoma antigen peptide(s) used in (a), (c) optionally cloning said population of T lymphocytes specific for the melanoma antigen peptide(s) used in (a).
8. A ma antigen peptide comprising the amino acid sequence SEQ ID NO:5, wherein said peptide has an amino acid sequence of less than 15 amino acids long, or consists of the sequence SEQ ID NO:2 or SEQ ID NO:4.
9. A melanoma antigen peptide according to claim 8, wherein said peptide consists of any one of the sequences SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:5.
10. A fusion protein comprising a melanoma n peptide as defined in any one of claims 8 or 9 and a melanoma antigen peptide comprising the amino acids motif: AH26(12781302_1):EOR - TX2NDECWPX9 (SEQ ID NO: 23) wherein X2 is leucine, methionine, valine, isoleucine or glutamine and X9 is alanine, valine or leucine.
11. A nucleic acid sequence ng a melanoma antigen e according to any one of claims 8 or 9 or a fusion protein according to claim 10.
12. An expression vector comprising a nucleic acid sequence according to claim 11.
13. A host cell comprising an expression vector according to claim 12, n the host cell is not within a human body.
14. An antibody or fragment thereof that binds to a melanoma antigen peptide according to any one of claims 8 or 9.
15. An immunising composition comprising: (a) at least one ma antigen peptide according to any one of claims 8 or 9 or (b) at least one fusion protein according to claim 10, or (c) at least one nucleic acid sequence according to claim 11 or (d) at least one sion vector ing to claim 12, or (e) at least one host cell according to claim 13, or (f) at least one antibody according to claim 14.
16. Use of an antibody or fragment thereof that binds to a melanoma antigen peptide as defined in any one of claims 1 or 2 in the manufacture of a medicament for the prevention or the treatment of ma in a patient.
NZ701000A 2012-05-22 Novel melanoma antigen peptide and uses thereof NZ701000B2 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2012/001310 WO2013175256A1 (en) 2012-05-22 2012-05-22 Novel melanoma antigen peptide and uses thereof

Publications (2)

Publication Number Publication Date
NZ701000A NZ701000A (en) 2017-03-31
NZ701000B2 true NZ701000B2 (en) 2017-07-04

Family

ID=

Similar Documents

Publication Publication Date Title
US20230414735A1 (en) Neoantigens and methods of their use
TWI730306B (en) Novel peptides and combination of peptides for use in immunotherapy against cll and other cancers
EP3140319B1 (en) Novel immunotherapy against several tumors of the blood, such as acute myeloid leukemia (aml)
US6602510B1 (en) HLA class I A2 tumor associated antigen peptides and vaccine compositions
TWI670282B (en) Novel immunotherapy against several tumors of the blood, in particular chronic lymphoid leukemia (cll)
JP5502823B2 (en) Proteins belonging to the BCL-2 family and fragments thereof, and their use in cancer patients
US20160022791A1 (en) Cytotoxic T Lymphocyte Inducing Immunogens For Prevention Treatment and Diagnosis of Cancer
US20080279924A1 (en) HLA class I A2 tumor associated antigen peptides and vaccine compositions
JP2021527426A (en) Neoantigen and its use
CA2661651A1 (en) Cytotoxic t-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
JP6523685B2 (en) Native and agonist CTL epitopes of MUC1 tumor antigen
JP2021527655A (en) Neoantigens and their use
CA2393730A1 (en) Hla class i a2 tumor associated antigen peptides and vaccine compositions
US9475841B2 (en) Melanoma antigen peptide and uses thereof
US20030224036A1 (en) Hla class I a2 tumor associated antigen peptides and vaccine compositions
US9573975B2 (en) Melanoma antigen peptide and uses thereof
JP2020014460A (en) Novel immunotherapy for several tumors of blood, in particular chronic lymphatic leukemia (cll)
US10898562B1 (en) Immunotherapy against several tumors of the blood, such as acute myeloid leukemia (AML)
US20160331818A1 (en) Novel antigen peptide and uses thereof
NZ701000B2 (en) Novel melanoma antigen peptide and uses thereof
US20140147490A1 (en) Hla class i a2 tumor associated antigen peptides and vaccine compositions