WO2017142988A1 - Methods and compositions for treating melanoma - Google Patents

Methods and compositions for treating melanoma Download PDF

Info

Publication number
WO2017142988A1
WO2017142988A1 PCT/US2017/018051 US2017018051W WO2017142988A1 WO 2017142988 A1 WO2017142988 A1 WO 2017142988A1 US 2017018051 W US2017018051 W US 2017018051W WO 2017142988 A1 WO2017142988 A1 WO 2017142988A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
ser
leu
gly
seq
Prior art date
Application number
PCT/US2017/018051
Other languages
French (fr)
Inventor
Rajasekharan Somasundaram
Meenhard Herlyn
Dorothee Herlyn
Original Assignee
The Wistar Institute Of Anatomy And Biology
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 The Wistar Institute Of Anatomy And Biology filed Critical The Wistar Institute Of Anatomy And Biology
Priority to EP17753785.9A priority Critical patent/EP3416668A4/en
Priority to CN201780024075.5A priority patent/CN109069574A/en
Priority to US15/998,913 priority patent/US11208468B2/en
Publication of WO2017142988A1 publication Critical patent/WO2017142988A1/en
Priority to US17/533,095 priority patent/US20220177548A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • BRAF is an intracellular signaling protein expressing frequently in melanomas for which alleles were identified as somatic mutations in 70% of melanomas, the majority of all types of nevi, and a minority of other cancers including lung, colon and ovary carcinomas, but not in normal cells.
  • the BRAF mutations were located in exons 11 or 15, with BRAF V 600E representing nearly all (92%) the BRAF alleles in melanoma.
  • BRAF V 6OOE has oncogenic activity through activation of the MAP kinase pathway.
  • BRAF V 6OOE based peptides that induce MHC Class I, HLA-A2-dependent cytotoxic T cell responses were identified in US Patent No. 7,811,993, which is incorporated herein by reference. These BRAFV6OOE based peptides were designed based on the amino acids 597 to 606 of the BRAF V 6OOE sequence (SEQ ID NO: 1) for use in prophylactic and therapeutic treatments for melanoma. More specifically, the
  • compositions described therein were useful in inducing responses that were not patient- specific, but that were specific for a mutation that occurs in about 70% of all melanoma patients.
  • a composition comprises a BRAF V 6OOE based peptide that induces MHC Class I, HLA-A2-dependent cytotoxic T cell responses peptide or a
  • this composition comprises peptides of the Formula II, III or IV as described herein.
  • a composition comprises a BRAF V 6OOE based peptide, including a peptide of Formula I, II, III or IV, described below that induces MHC Class I, HLA-A2- dependent cytotoxic T cell responses and a T helper peptide.
  • compositions recited herein further comprise an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • a composition comprises a nucleic acid sequence encoding a BRAF V 600E based peptide of Formula II, III or IV as described below.
  • composition comprises a nucleic acid sequence encoding a BRAF V 600E based peptide of Formula I, II, III or IV below that induces MHC Class I,
  • HLA-A2-dependent cytotoxic T cell responses and a nucleic acid sequence encoding a T helper sequence.
  • nucleic acid sequence-containing compositions recited herein further comprise a nucleic acid sequence encoding a checkpoint inhibitor or an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • a composition comprises a recombinant dendritic cell that is pretreated ex vivo with a composition containing a BRAF V 6OOE based peptide, an optional T helper peptide and/or an optional antibody that binds a checkpoint inhibitor or an optional molecule that mimics the function of a checkpoint inhibitor as described herein.
  • a recombinant dendritic cell pretreated ex vivo with a composition containing a nucleic acid sequence encoding a BRAF V 6OOE based peptide, optionally including a nucleic acid sequence encoding a T helper peptide and/or an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor as described herein.
  • a composition comprises the pretreated dendritic cell described herein with an antibody that binds a checkpoint inhibitor in a formulation suitable for simultaneous administration.
  • composition as described herein for use in the treatment or prophylaxis of melanoma in a mammalian subject is provided.
  • a method of treating or retarding or preventing the development of cancer, particularly melanoma, in a mammalian subject comprises administering to said subject one or more of the compositions as described herein.
  • FIG. 1 shows three graphs of results of proliferative assays for three different melanoma peripheral blood mononuclear cell (PBMC) samples labeled WM35, WM3457 and 3451 with peptides derived from the BRAF V6OOE sequence.
  • the peptides were screened for T-helper sequences (Th):
  • Peptide P8 is amino acids residues 608-622 of BRAF, i.e., SHQFEQLSGSILWHA SEQ ID NO: 15.
  • Peptide P9 is amino acids residues 546-600 of BRAF, i.e., EDLTVKIGDFGLATV SEQ ID NO: 16.
  • Peptide P10 is amino acids residues 547-601 of BRAF, i.e., DLTVKIGDFGLATVK SEQ ID NO: 17. These peptides bind to HLA-DR alleles using computer algorithms, as disclosed in Example 1 below. Proliferative responses were determined by standard HTdR incorporation assay. Data are expressed as mean cpm (triplicate determinations) plus SD (bar) of HTdR incorporation. * Values are significantly (p ⁇ 0.01) different from the value obtained with the control peptide. This figure demonstrates that T helper peptides can induce proliferative response in melanoma patients PBMCs.
  • FIG. 2 is a graph showing the induction of enriched CD 8 cytotoxic T cells (CTLs) in the presence of both a T helper and a BRAF V 6OO E - based CTL peptide.
  • the BRAF V 6OO E - based CTL peptide is that of the sequence LMTEKSRWSG SEQ ID NO: 25.
  • the T helper sequence is that of P9 SEQ ID NO: 16.
  • adherent monocytes were pulsed with the T helper synthetic peptides (25 ⁇ ) either alone or in combination with the affinity modified BRAF V6OOE - based CTL peptide and cell cultured.
  • T-cells were harvested and stained with anti-CD4 or anti-CD8 antibodies and analyzed for T-cell binding by standard FACS assay.
  • FIG. 3 is a graph showing the results of a standard 51 Cr-release assay conducted on melanoma cells exposed to lymphocytes that had been stimulated with Th peptide P9 SEQ ID NO: 16 identified above in FIG. 1, the BRAF V6 ooE-based CTL peptide SEQ ID NO: 25, or both peptides and with natural human IL-2, as described in Example 3.
  • the lymphocytes were tested for cytolytic activity against HLA-A2+ autologous WM35 (V600E+) or HLA-A2+ matched allogeneic WM3456 (V600E-) melanoma cells.
  • T-cells Indicated effector (T-cells) to tumor target ratios were used in the assay.
  • FIG. 4 is a graph showing the effect of a BRAF V 6OOE -based CTL peptide SEQ ID NO: 1
  • a checkpoint inhibitor which is an anti-PDl antibody
  • Tumor growth measurements are shown as Mean tumor volume and SEM and the results are compared with mice that received control HIV peptide and anti-PDl antibody. Combination of peptide immunization and anti-PDl antibody therapy showed significant inhibition of tumor growth (p ⁇ 0.05). Anti-PDl antibody alone was not effective in tumor inhibition when compared to isotype control antibody treatment.
  • compositions and methods are described herein that provide alternative and enhanced methods of treatment of melanoma and other cancers by the use of certain BRAF V 6ooE based peptides, compositions containing the peptides and nucleic acid compositions encoding these peptides..
  • BRAF V 6OOE refers to the human amino acid sequence of SEQ ID NO: 1 in which the amino acid residue at 600 is changed from the Val residue in the native sequence to a Glu.
  • the nucleic acid sequence encoding BRAF V 6OOE appears in SEQ ID NO: 21.
  • the nucleic acid sequence encoding amino acids 595 to 620 of SEQ ID NO: 1 was most often employed to make the changes from the native sequence to produce the peptides or nucleic acid compositions described herein.
  • Native human BRAF amino acid and encoding nucleic acid sequences are shown in the publically available NCBI database under Accession No. NM_004333.4. Other published sequences are available from NCBI or GenBank databases
  • BRAF V 6OOE based peptides or "BRAF V 6OOE based CTL peptides” refers to peptides based on amino acids 597-606 of BRAF V 6OOE sequence, namely Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly SEQ ID NO: 2 that induce MHC
  • such peptides included in this definition are the peptides identified in US Patent No. 7,811,993, incorporated by reference herein.
  • Such peptides are defined by the Formula I: Rl-Leu- AA2-AA3-Glu-AA5-AA6-AA7-Trp-AA9-AA10-R2 (SEQ ID NO: 3), or a
  • AA2 is an a substituted or unsubstituted Ala, Leu, Met, Val, Pro and Gly;
  • AA3 is a substituted or unsubstituted Thr and Ser;
  • AA5 is a substituted or unsubstituted Lys, Arg and His;
  • AA6 is a substituted or unsubstituted Ser and Thr;
  • AA7 is a substituted or unsubstituted Arg, Lys, and His;
  • AA9 is a substituted or unsubstituted Thr, Val, Leu and Ser;
  • AA10 is absent or is a substituted or unsubstituted Gly, Pro or Leu.
  • BRAF V 6OOE based peptides are the peptides defined by Formulae II, III and IV.
  • Formula II is Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp- Ser-AA10 -R2 (SEQ ID NO:4), wherein AA2 is a substituted or unsubstituted He, and AAIO is a hydrophobic residue.
  • Formula III is Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp- Ser-AA10 -R2 (SEQ ID NO: 5), wherein AA2 is a hydrophobic residue and AA10 is a substituted or unsubstituted Gly, Val or Leu.
  • Formula IV is Rl-Leu-AA2-Thr-Glu-Lys- Ser-Arg-Trp-Ser-AA10 -R2 (SEQ ID NO: 6), wherein AA2 is a substituted or
  • AA10 is a substituted or unsubstituted Gly, He, Val or Leu.
  • Rl and R2 are each independently absent or provide a spacer for coupling of a peptide of each formula to a second peptide or protein at the N- or C-termini of the peptide.
  • spacers may be amino acid sequences or chemical compounds ordinarily used as spacers.
  • Rl is a Cys; in another embodiment, Rl is a Gly-Ser. In another embodiment R2 is a Cys. In still another embodiment, R2 is a Gly-
  • Rl and R2 are identical. In another embodiment, Rl and R2 are different. In another embodiment of a peptide of Formula II, III or IV, Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein.
  • hydrophobic residue is meant a substituted or unsubstituted amino acid which imparts hydrophobicity to the resulting BRAF V 600E-based peptide.
  • Unsubstituted amino acids having hydrophobic side chains are glycine (Gly), alanine (Ala), valine (Val), leucine (Leu), isoleucine (He), proline (Pro), phenylalanine (Phe), methionine (Met), and tryptophan (Trp).
  • glycine Gly
  • alanine Al
  • valine val
  • Leu leucine
  • isoleucine He
  • Pro proline
  • Pro proline
  • Phe phenylalanine
  • methionine Metal
  • Trp tryptophan
  • Examples of specific BRAF V 6OOE based peptides of Formula I through IV include, among other s equences :
  • T helper peptide or "T helper sequence” or “Th peptide” as used herein is meant certain T helper sequences, or modified variants thereof of BRAF V 6OOE SEQ ID NO:
  • a suitable Th peptide is Ser-His-Gln-Phe-Glu-Gln-Leu-Ser-Gly-Ser-Ile-Leu- Trp-His-Ala SEQ ID NO: 15, which spans aa 607 to 622 of BRAF V6 OOE.
  • a suitable Th peptide is Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly- Leu-Ala-Thr-Val SEQ ID NO: 16, which spans AA 588 to 600 of BRAF V6 OOE.
  • a suitable Th peptide is Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu- Ala-Thr-Val-Lys SEQ ID NO: 17, which spans AA589 to 601 of BRAF V6 OOE.
  • This definition of suitable Th sequences also includes modified versions of the peptide sequences and pharmaceutically acceptable salts thereof. Peptides spanning both the T helper and BRAF V6 OOE based peptides, such as spanning AA597 to 622 SEQ ID NO: 32, or
  • BRAF V 6OOE based peptides and T helper peptides also include modified versions of the peptide sequences and pharmaceutically acceptable salts thereof.
  • modified peptides of the above Formulae I-IV is meant homologous or analogous modified sequences, wherein non-variable amino acids may be conservatively replaced individually by amino acid residues having similar characteristics.
  • the non-variable amino acid residues may be replaced by other amino acid residues bearing the same charge and/or similar side chain lengths.
  • the non- variable naturally-occurring amino acids may be replaced by non-naturally occurring amino acid residues.
  • an amino acid residue may be a naturally-occurring amino acids, meaning one of the twenty amino acids that occur in nature in L form, which include alanine, cysteine, aspartate, glutamate, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan, and tyrosine, or any derivative thereof produced through a naturally-occurring biological process or pathway.
  • non-naturally-occurring amino acids are also encompassed by the term "modifications" of the Formulae I - IV above.
  • This latter term is used herein to refer to an amino acid other than a naturally-occurring amino acid as defined above, which can be synthesized or "man-made", and including a derivative thereof, whether produced synthetically or via a biological process or pathway.
  • Non-naturally occurring amino acids include, without limitation, D amino acids, amino acids containing unnaturally substituted side chains, e.g., an N-methylated amino acid, cyclic amino acids, diamino acids, B-amino acids, homo amino acids.
  • non-naturally occurring amino acids used in the above formulae are only those that do not strain the binding formation by adding extra atoms to the peptide backbone, because backbone hydrogen bonding contact with the MHC is desirable for these peptides.
  • Non-naturally-occurring or unnatural amino acids may be characterized by novel backbone and side chain structures and are widely available from commercial reagent suppliers, such as Sigma- Aldrich (on the world-wide web at sigmaaldrich.com), on the world-wide web at Netchem.com, and other sites.
  • non-naturally occurring amino acid(s) when employed in the peptides and compositions herein are anticipated to make the compounds more resistant to degradation by mammalian enzymes in serum, saliva, stomach and intestines, and thus compounds that are composed of one or more such amino acids may confer upon the compound enhanced stability and bioavailability in vivo.
  • a variety of methods for producing non-natural amino acids are known and may be selected by one of skill in the art.
  • a class of non-naturally occurring amino acids includes L amino acids that effect stereochemistry.
  • one or more of the amino acids in the peptide may be in L form, while others may be in D form.
  • Another non-naturally occurring amino acid is an amino acid which is modified to contain a substitution on the alpha-carbon in the amino acid structure.
  • the alpha- carbon may be substituted by a suitable hydrocarbon moiety, such as aminoisobutyrate.
  • Still another class of non-naturally occurring amino acids is amino acids which are modified or mutated to extend their carbon chain length.
  • an amino acid with a single alpha-carbon chain may be extended with at least one additional carbon, i.e., a beta-carbon, and so on.
  • An additional modification to an amino acid is the insertion of a substituent on the nitrogen of the amino group.
  • An example of this type of modification is an N-methyl amino acid.
  • the addition of substituents on the alpha carbon or additional carbons or on the nitrogen of the amino acid molecule may occur in any of the amino acids of the formulae above.
  • substituents for creating the non-naturally occurring amino acids are straight chain, branched, cyclic or heterocyclic C 1-12 alkyl groups, and straight chain, branched, cyclic, or heterocyclic C 1-12 alkanoyl groups.
  • the amino acid may be also modified by the insertion of modifying sugars, imide groups and the like.
  • Other amino acids are substituted in the ortho or meta position by a substituent such as H, OH, CH 3 , halogen, OCH 3 , NH 2 , CH or N0 2 .
  • a non-exclusive list of modified or non-naturally occurring amino acids for inclusion in compounds fitting the formulae above include amino acids modified by N- terminal acetylation, C-terminal amidation, formylation of the N-terminal methionine, gamma-carboxy glutamic acid hydroxylation of Asp, Asn, Pro or Lys residues in the compound, methylation of Lys or Arg, preferably; phosphorylation of Ser, Thr, Tyr, Asp or His in the compound, use of a pyrrolidone carboxylic acid, which is an N-terminal glutamate which has formed an internal cyclic lactam, sulphonation of Tyr, generally.
  • Non-naturally occurring amino acids include use of or substitution with the following moieties: a 2-aminoadipic acid group, a 3-aminoadipic acid group, beta-Ala or beta-aminopropionic acid group, 2-aminobutryic acid, 4-aminobutyric acid, piperidinic acid, 6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutryic acid, 3- aminoisobutyric acid, 2-aminopimelic acid, 2, 4 diaminobutyric acid, desmosine, 2,2'- diaminopimelic acid, 2,3-diaminopropionic acid, N-ethylglycine, N-ethylglycine, N-ethyl asparagine, hydroxylysine, allo-hydroxylysine, 3 -hydroxy proline, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarco
  • peptides of Formulae I-IV include fusions into polypeptides or other multimeric constructs or compositions, when either Rl or R2 are absent or optional amino acids (e.g., Cys, -Gly-Ser-) or other amino acid or chemical compound spacers may be included at the N- or C-termini of the peptide for the purpose of linking two or more of the same or different BRAF V 600E-based peptides together or linking a peptide to a second peptide, such as one of the Th peptides or to a carrier.
  • optional amino acids e.g., Cys, -Gly-Ser-
  • other amino acid or chemical compound spacers may be included at the N- or C-termini of the peptide for the purpose of linking two or more of the same or different BRAF V 600E-based peptides together or linking a peptide to a second peptide, such as one of the Th peptides or to a carrier.
  • the spacer of Rl or R2 is a proteolytically sensitive spacer to permit cleavage of the epitope before it enters the cell compartment where it associates with MHC.
  • multiple copies of the same BRAF V 6ooE-based peptides are linked sequentially and expressed as a recombinantly or synthetically produced polypeptide.
  • multiple different BRAF V 600E-based peptides are linked sequentially, with and without spacer amino acids therebetween, to form a larger recombinant or synthetic fused polypeptide.
  • at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 repeating units of the same BRAF V 600E-based peptide forms a polypeptide.
  • BRAF V 600E-based peptide-T helper peptide fusion 4, 5, 6, 7, 8, 9, or 10 repeating units of the same fusion peptide, i.e., a BRAF V 600E-based peptide-T helper peptide fusion.
  • multiple BRAF V 600E-based peptides of fusion peptides as described are coupled to a carrier.
  • Such peptides and multimeric compositions may be produced synthetically or recombinantly by conventional methods.
  • the peptides are prepared conventionally by known chemical synthesis techniques.
  • carrier is meant a protein, peptide or other substrate that may enhance stability or delivery, improve the production, or change the activity spectrum of the peptide.
  • such carriers may be human albumin, keyhole limpet hemocyanin, polyethylene glycol, other biopolymers or other naturally or non- naturally occurring polymers.
  • the moiety is desirably a protein or other molecule which can enhance the stability of the peptide or enhance its penetration into the targeted cell.
  • Still other proteins or peptides to which the BRAF V 6ooE-based peptides described herein may be linked via the spacer include keyhole limpet hemocyanin or additional MHC molecules.
  • Still other carriers include a live antigen-presenting cell, such as a dendritic cell, which presents the peptides described herein.
  • Still alternative carriers or peptide-carrier constructs utilize lipopeptides.
  • nucleic acid sequence that encodes a BRAF V 6ooE-based peptide is meant an RNA or DNA sequence encoding the amino acid sequence of a BRAFv 6 ooE-based peptide as described herein, such as a peptide of any of Formula I through IV.
  • SEQ ID NO: 21 provides the DNA sequence of BRAF V 6OOE from which other modified nucleic acid sequences can be obtained, e.g., encoding the modified peptides or fusion peptides described herein.
  • nucleic acid sequence that encodes a T helper sequence is meant an RNA or DNA sequence encoding the amino acid sequence of T helper as described herein, such as those of SEQ ID Nos: 15, 16 and 17, among others. Nucleic acid sequences may also encode polypeptide or fusions of the referenced peptides. The nucleic acid sequences may be generated and/or modified by conventional techniques and useful in prophylactic, diagnostic, and therapeutic compositions and methods designed for delivery of the nucleic acid in vivo and expression of the peptide in vivo.
  • Nucleic acid sequences encoding the peptides described herein may be prepared by known recombinant DNA techniques and used to clone and express the peptides within a host microorganism or cell. Such nucleic acid sequences may also encode different or preferred codons for the peptide described.
  • nucleic acid sequences encoding these peptides may be designed to incorporate other nucleic acid sequences necessary for delivery to a subject, e.g., as naked DNA or other DNA vaccine forms.
  • a suitable plasmid may be constructed containing a nucleic acid sequence encoding the selected BRAF V 6ooE-based peptide or T helper peptide, or both, under the control of regulatory sequences directing expression thereof in a mammalian or vertebrate cell.
  • the components of the plasmid itself are conventional.
  • Still other nucleic acid constructs and vectors of viral and bacterial origin may be employed. See, e.g., US Patent No. 7,811,993, incorporated by reference herein.
  • checkpoint inhibitor refers to a composition or composition in the form of an antibody or a small molecule that binds or inhibits various checkpoint proteins.
  • checkpoint proteins include, without limitation, PD-1 , PD-L1, CTLA-4, BTLA and CD 160.
  • known checkpoint inhibitors include the antibodies ipilimumab (Yervoy®), pembrolizumab (Keytruda®), and nivolumab
  • checkpoint inhibitors developed as small molecules or other checkpoint binding antibodies or antibody fragments are included in this definition.
  • the term "antibody” refers to all types of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including antibody fragments.
  • the antibody can be monoclonal or polyclonal and can be of any species of origin, including (for example) mouse, rat, rabbit, horse, goat, sheep, camel, or human, or can be a chimeric antibody. See, e.g., Walker et al, Molec. Immunol. 26:403 (1989).
  • the antibodies can be recombinant monoclonal antibodies produced according to known methods, see, e.g., U.S. Patent Nos. 4,474,893 or 4,816,567, which are incorporated herein by reference.
  • the antibodies can also be chemically constructed according to known methods, e.g., US Patent No.
  • An antibody includes, without limitation, a synthetic antibody, a recombinant antibody, a chimeric antibody, a humanized antibody, a human antibody, a CDR-grafted antibody, a multispecific binding construct that can bind two or more targets, a dual specific antibody, a bi-specific antibody or a multi-specific antibody, or an affinity matured antibody.
  • Antibody fragments include without limitation antigen binding fragments such as a single antibody chain or an scFv fragment, a diabody, a single chain comprising complementary scFvs (tandem scFvs) or bispecific tandem scFvs, an Fv construct, a disulfide-linked Fv, a Fab construct, a Fab' construct, a F(ab')2 construct, an Fc construct, a monovalent or bivalent construct from which domains nonessential to monoclonal antibody function have been removed, a single-chain molecule containing one VL, one VH antigen-binding domain, and one or two constant "effector" domains optionally connected by linker domains, a univalent antibody lacking a hinge region, a single domain antibody, a dual variable domain immunoglobulin (DVD-Ig) binding protein, a nanobody, a domain antibody, a vaccibody, a linear antibody; a heavy chain or light chain complementarity determining region
  • antibody mimetics such as affibodies, i.e., a class of engineered affinity proteins, generally small (-6.5 kDa) single domain proteins that can be isolated for high affinity and specificity to any given protein target.
  • affibodies i.e., a class of engineered affinity proteins, generally small (-6.5 kDa) single domain proteins that can be isolated for high affinity and specificity to any given protein target.
  • antigen-binding fragments can be produced by known techniques.
  • patient or "subject” as used herein means a mammalian animal, including a human, a veterinary or farm animal, a domestic animal or pet, and animals normally used for clinical research.
  • the subject treated with the methods and composition is a human.
  • the subject treated with the methods and composition has a cancer.
  • the subject of these methods has melanoma.
  • cancer refers to or describes the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer means any cancer characterized by the presence of a solid tumor.
  • suitable cancers for treatment by the methods described herein include, cancers having the presence of the BRAF V 6OOE mutation.
  • suitable cancers include, without limitation, melanoma, breast cancer, lung cancer and ovarian cancer with such mutations.
  • suitable cancers include, without limitation, other forms of melanoma, brain cancer, colon/rectal cancer, lung cancer, ovarian cancer, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, bone cancer, endometrial cancer, esophageal cancer, eye cancer, kidney cancer, laryngeal cancer, liver cancer, head and neck cancer, nasopharyngeal cancer, osteosarcoma, oral cancer, pancreatic cancer, prostate cancer, rhabdosarcoma, salivary gland cancer, stomach cancer, testicular cancer, thyroid cancer, vaginal cancer, lung cancer, and neuroendocrine cancer
  • tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • the tumor targeted by the methods is characterized by hypoxia, significant infiltration with T lymphocytes, and low glucose in the tumor microenvironment.
  • therapeutic reagent or “regimen” is meant any type of treatment employed in the treatment of cancers with or without solid tumors, including, without limitation, chemotherapeutic pharmaceuticals, biological response modifiers, radiation, diet, vitamin therapy, hormone therapies, gene therapy, surgical resection, etc.
  • an immunotherapeutic composition or “immunotherapeutic vaccine” is meant any composition including the BRAF V 600E-based peptides or nucleic acid sequences encoding same that stimulate the subject's immune system. Such immunotherapeutic compositions are designed to elicit a humoral (e.g., antibody) or cellular (e.g., a cytotoxic T cell or T helper) response, to the BRAF target gene product delivered by the
  • immunogenic composition to a subject.
  • the immunotherapeutic compositions or vaccines, as described herein, are created using known recombinant and synthetic techniques.
  • immunotherapeutic compositions useful in these methods involve presentation of the BRAF V 600E-based peptides to the subject's immune system.
  • the immunotherapeutic composition used in the methods described herein is a DNA or RNA composition including a nucleic acid encoding the BRAF V 6OOE- based peptides.
  • the BRAF V 6ooE-based peptides or encoding nucleic acid sequences are delivered via virus vectors, e.g., adenovirus, adeno-associated virus, lentivirus, retrovirus, poxvirus or others, or via virus-like particles (VLP).
  • the immunotherapeutic composition used in the methods described herein is a monoclonal antibody or antigen-binding fragment(s) that specifically bind cancer antigens.
  • the immunotherapeutic composition is a dendritic cell.
  • Dendritic cell therapy involves the harvesting of autologous blood cells (monocytes) from a patient and pretreating the cells with, a BRAF V 6ooE-based peptide (optionally with a Th peptide) ex vivo.
  • pre-treatment with is meant that the autologous dendritic cell is cultured and expanded in the presence of the selected BRAFv6ooE-based peptide at between about 1 to about 500 ⁇ , to condition the dendritic cell to activate the immune system upon reinfusion into the subject.
  • the time of pretreatment in one embodiment means the entire time of in vitro culture, which can span several hours to at least several days.
  • the time of pretreatment is minimally 24 hours of in vitro culture.
  • Other time periods for pre-treatment with the peptides or nucleic acid components of the compositions described herein may be at least 1, 5, 10, 15, or 20 or more hours, or any intervening times between any specified number of hours stated herein.
  • the pretreated dendritic cells are reintroduced, e.g., by i.v. injection, back to a patient in order to allow massive dendritic cell participation in optimally activating the immune system.
  • vector is meant an entity that delivers a heterologous molecule to cells, either for therapeutic or vaccine purposes.
  • a vector may include any genetic element including, without limitation, naked DNA, a phage, transposon, cosmid, episome, plasmid, or a virus or bacterium.
  • Vectors are generated using the techniques and sequences provided herein, described in the examples and in conjunction with techniques known to those of skill in the art. Such techniques include conventional cloning techniques of cDNA such as those described in texts such as Green and Sambrook, Molecular Cloning: A Laboratory Manual. 4 th Edit, Cold Spring Harbor Laboratory Press, 2012, use of overlapping oligonucleotide sequences of the Salmonella genomes, polymerase chain reaction, and any suitable method which provides the desired nucleotide sequence.
  • compositions may further include a detergent to make the peptide more bioavailable, e.g., octylglucoside.
  • adjuvant is meant a substance that enhances the immune response when administered together with an immunogen or antigen.
  • a suitable adjuvant is CPG.
  • a suitable adjuvant is encapsulated PLG.
  • Still other useful adjuvants are known, and include those listed and disclosed in US Patent No. 7,811,993, and its cited references, which are all incorporated herein by reference.
  • administering or “route of administration” is meant delivery of the
  • administration method can occur with or without a pharmaceutical carrier or excipient, or with or without another chemotherapeutic agent into the subject.
  • Conventional and pharmaceutically acceptable routes of administration include, but are not limited to, systemic routes, such as intraperitoneal, intravenous, intradermal, subcutaneous, and other parenteral routes of administration or intratumoral or intranodal administration.
  • the route of administration is intradermal or subcutaneous for the peptide(s) and polypeptides.
  • the route of administration is intraperitoneal or intravascular for the checkpoint inhibitor antibodies.
  • the route of administration is intravascular or intravenous for delivery of the dendritic cell-based vaccine. Routes of administration may be combined, if desired. In some embodiments, the administration is repeated periodically, as discussed in detail below.
  • a refers to one or more, for example, “an miRNA,” is understood to represent one or more miRNAs.
  • the terms “a” (or “an”), “one or more,” and “at least one” are used interchangeably herein.
  • compositions for Use as Immunotherapeutic Vaccines are provided.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide having the Formula II: Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10 -R2 (SEQ ID NO:4).
  • Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein, and AA2 is a substituted or unsubstituted He and AA10 is a substituted or unsubstituted hydrophobic residue.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide having the Formula III: Rl -Leu- AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser- AA10 -R2 (SEQ ID NO:5).
  • Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein
  • AA2 is a hydrophobic residue
  • AAIO is a substituted or unsubstituted Gly, Val or Leu.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1 LAG3, or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide having the Formula IV: Rl-Leu-AA2- ⁇ ⁇ nr-Glu-Lys-Ser-Arg-T -Ser-AA10-R2
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 SEQ ID NO: 7 or a pharmaceutically acceptable salt thereof.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 SEQ ID NO: 1
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 SEQ ID NO: 9 or a pharmaceutically acceptable salt thereof.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Ile-R2 SEQ ID NO: 1
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Met-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 SEQ ID NO: 11 or a pharmaceutically acceptable salt thereof.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 SEQ ID NO: 1
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 SEQ ID NO: 13 or a pharmaceutically acceptable salt thereof.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Leu-R2 SEQ ID NO: 14 or a pharmaceutically acceptable salt thereof.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier.
  • a composition such as described herein includes a liposome.
  • a composition such as described herein includes an adjuvant.
  • a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide of Formula I, II, III or IV and a T helper peptide.
  • the T cell peptide is coupled or fused at either the N- or C- termini of the peptide of the BRAF V 6OOE- based peptide or polypeptide.
  • the T helper peptide is Ser-His-Gln- Phe-Glu-Gln-Leu- Ser-Gly-Ser-Ile-Leu-Trp-His-Ala SEQ ID NO: 15.
  • the T helper peptide is Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val SEQ ID NO: 16. In one embodiment, the T helper peptide is Asp-Leu-Thr-Val-Lys-Ile- Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val-Lys SEQ ID NO: 17.
  • composition containing a fused BRAF V 600E-based peptide or polypeptide and a T helper sequence is the sequence spanning the Th and CTL sequences found in BRAF V 6OOE- In one embodiment, this sequence spans about AA586 to AA 606 and has the formula, e.g., Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu- AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10 SEQ ID NO: 18 with AA2 and AA10 as described in any of Formula II, III or IV above.
  • this sequence spans about AA587 to AA 606 and has the formula, e.g., Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-AA2-Thr-Glu-Lys-Ser- Arg-Trp-Ser-AA10 SEQ ID NO: 19, with AA2 and AA10 as described in any of Formula II, III or IV above.
  • this sequence spans about AA597 to AA621 and has the formula, e.g., Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-Ser-His- Gln-Phe-Glu-Gln-Leu-Ser-Gly-Ser-Ile-Leu-Trp-Met-Ala SEQ ID NO: 20 with AA2 and
  • AAIO as described in any of Formula II, III or IV above. Still other embodiments of these sequences can contain modified amino acids, or replacement amino acids in the positions specified in Formula I, or conservative replacements, possible spacer amino acids on either end of the peptide, or multiple Th sequences flanking the amino- or carboxy amino acid of the whole peptide, as described above. Nucleic acid sequences encoding these sequences are also useful in these compositions.
  • a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • composition useful in the treatment of melanoma or other cancers comprises multiple copies of the same BRAF V 6OOE -based peptide or polypeptide fused or coupled together.
  • a composition useful in the treatment of melanoma or other cancers comprises multiple copies of a fused peptide, which comprises same BRAF V 6OOE -based peptide or polypeptide fused or coupled to a T helper peptide.
  • such compositions comprise a BRAF V 6OOE -based polypeptide or fusion formed from 5 to 10 of the same fused in frame or via a spacer.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR,
  • a composition useful in the treatment of melanoma or other cancers comprises one or multiple of the BRAF V 6OOE -based peptide or polypeptides or fusions with T helper sequences as described herein. Any number of single peptides or multimeric constructs may be mixed together to form a single composition.
  • the peptides may be formulated with a pharmaceutically acceptable carrier, adjuvant, diluent, other optional components, or some combination thereof.
  • the selected peptide(s) may be produced preferably synthetically, but also recombinantly, as disclosed above.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PDl, PDLl, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition useful in the treatment of melanoma or other cancers comprises one or multiple of the BRAFV6OOE -based peptides described herein in the form of a salt with an acid.
  • the compounds have at least one amino/amine groups which can form salts. Where two or more amino groups are present in the compound, a formulation of mixed salts can be prepared.
  • Acids which can be used preferably include compatible inorganic acids such as hydrochloric and organic acids (or salts thereof) more preferably those occurring in living organisms, including but not limited to oxalic acid, glucuronic acid, pyruvic acid, lactic acid, citric acid, isocitric acid- ketoglutaric acid, succinic acid, malic acid, and oxaloacetic acid.
  • the desired anion can be added either as the free acid, or a salt, preferably one which is highly soluble in water, for example the sodium or potassium salts, but also the lithium, magnesium, calcium or ammonium salts.
  • these salts can be used either in anhydrous or hydrated forms.
  • citric acid can be used as the anhydrous free acid, the monohydrate free acid, the anhydrous trisodium salt, or the dihydrate trisodium salt.
  • a composition useful in the treatment of melanoma or other cancers comprises a nucleic acid sequence, such as a DNA vaccine, encoding any of the components of the compositions containing BRAF V 600E-based peptides of Formula I, II, III or IV, or polypeptides comprising two or more copies of the BRAF V 600E-based peptides or fusions with T helper sequences described above.
  • a nucleic acid sequence for use as DNA vaccines can take the form of a recombinant vector carrying the above-described peptide-encoding nucleic acid sequence.
  • the nucleic acid sequences may be carried, and the BRAF V 6OOE peptides are expressed by, plasmid based systems, of which many are commercially available or in replicating or non-replicating recombinant viral vectors.
  • the nucleic acid sequences discussed herein may be expressed and produced using such vectors in vitro in desired host cells or in vivo in a mammalian subject.
  • the vector is a nonpathogenic virus.
  • the vector is a non-replicating virus.
  • a desirable viral vector may be a retroviral vector, such as a lentiviral vector.
  • a desirable vector is an adenoviral vector.
  • a suitable vector is an adeno-associated viral vector.
  • Adeno, adeno- associated and lentiviruses are generally preferred because they infect actively dividing as well as resting and differentiated cells such as the stem cells, macrophages and neurons.
  • a variety of adenovirus, lentivirus and AAV strains are available from the American Type Culture Collection, Manassas, Virginia, or available by request from a variety of commercial and institutional sources. Further, the sequences of many such strains are available from a variety of databases including, e.g., PubMed and GenBank.
  • a lentiviral vector is used.
  • useful vectors are the equine infectious anemia virus and feline as well as bovine immunodeficiency virus, and HIV-based vectors.
  • the vector used herein is an adenovirus vector.
  • adenovirus vector can be constructed using adenovirus DNA of one or more of any of the known adenovirus serotypes. See, e.g., T. Shenk et al, Adenoviridae: The Viruses and their
  • the vector used herein is an adeno-associated virus (AAV) vector.
  • AAV adeno-associated virus
  • the vector used herein is a bacterial vector.
  • the bacterial vector is Listeria monocytogenes. See, e.g., Lauer et al, Infect. Immunity, 76(8):3742-53 (Aug. 2008).
  • the bacterial vector is live-attenuated or photochemically inactivated.
  • the BRAF V 6OOE and/or T helper peptide or fusion peptides as described herein can be expressed recombinantly by the bacteria, e.g., via a plasmid introduced into the bacteria, or integrated into the bacterial genome, i.e., via homologous recombination.
  • These vectors also include conventional control elements that permit transcription, translation and/or expression of the nucleic acid sequences in a cell transfected with the plasmid vector or infected with the viral vector.
  • a great number of expression control sequences including promoters which are native, constitutive, inducible and/or tissue- specific, are known in the art and may be utilized.
  • the promoter is selected based on the chosen vector.
  • the promoter is U6, HI, CMV IE gene, EF-la, ubiquitin C, or phosphoglycero-kinase (PGK) promoter.
  • the promoter is an AAV, the promoter is an RSV, U6, or CMV promoter.
  • the promoter when the vector is an adenovirus, the promoter is RSV, U6, CMV, or HI promoters. In another embodiment, when the vector is Listeria monocytogenes, the promoter is a hly or actA promoter. Still other conventional expression control sequences include selectable markers or reporter genes, which may include sequences encoding geneticin, hygromicin, ampicillin or purimycin resistance, among others. Other components of the vector may include an origin of replication. Selection of these and other promoters and vector elements are conventional and many such sequences are available (see, e.g., the references cited herein).
  • nucleic acid sequences encoding the BRAF V 6OOE and/or T helper sequences for use in DNA vaccines are generated using the techniques and sequences provided herein, in conjunction with techniques known to those of skill in the art. Such techniques include conventional cloning techniques of cDNA such as those described in texts (Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring
  • one of skill in the art can construct a viral vector (or plasmid) that expresses the desired nucleic acid sequence encoding the BRAF V 6OOE and/or T helper sequences.
  • the BRAF V 6OOE and/or T helper polypeptides or proteins or fusion polypeptides or proteins encoded by these nucleic acid constructs may be expressed in vitro, or ex vivo in host cells or expressed in vivo by administration to a mammalian subject.
  • the BRAF V 6OOE and/or T helper polypeptides or proteins or fusion polypeptides may be generated synthetically by known chemical synthesis methodologies.
  • BRAF V 6OOE and/or T helper polypeptides or proteins or fusion polypeptides depending upon the components, the efficiency of the methodologies and the intended use, e.g., whether for administration as proteins, nucleic acids or in adoptive T cells, or otherwise to accomplish the desired therapeutic result.
  • nucleic acid compositions may include formulations with suitable vehicles for direct DNA, plasmid nucleic acid, or recombinant vector administration.
  • suitable vehicles include, without limitation, saline, sucrose, protamine, polybrene, polylysine, poly cations, proteins, or spermidine, etc. See e.g, International Patent Publication No. WO94/01139.
  • Other pharmaceutically acceptable vehicles, excipients and typical components of DNA compositions are optionally included in these compositions.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a composition as described above is provided containing one or more of the BRAF V 600E-based peptides, polypeptides or fusions or nucleic acid sequences encoding same, including DNA vaccines, in a pharmaceutically acceptable carrier, adjuvant or diluent or a combination thereof.
  • This composition can contain other pharmaceutically acceptable components for enhancing the penetration of the compound into a cell and/or for extending its bioavailability and increasing its resistance to enzymatic degradation in vivo.
  • Such a composition in one embodiment, is a pharmaceutical composition.
  • Such a composition in another embodiment is immunogenic.
  • compositions both peptide or nucleic acid compositions, may also be formulated to suit a selected route of administration, and may contain ingredients specific to the route of administration as known to one of skill in the art of pharmaceutical formulation.
  • auxiliary agents are lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like that do not deleteriously react with the active compounds.
  • the preparation of these pharmaceutically acceptable compositions, from the above-described components, having appropriate pH isotonicity, stability and other conventional characteristics is within the skill of the art.
  • a composition useful in the treatment of melanoma or other cancers comprises a recombinant dendritic cell pretreated ex vivo with any of the BRAFv600E-based peptides of Formula I, II, III or IV, or polypeptides comprising two or more copies of the BRAF V 6ooE-based peptides or fusions with T helper sequences described above.
  • the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
  • checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
  • a modified T cell is a T cell that has been transduced or transfected with one of the above-described vectors carrying the nucleic acid constructs encoding the BRAF V 6OOE- based peptides, polypeptides or fusions or nucleic acid sequences encoding same.
  • the T cell is a primary T cell, a CD8 (cytotoxic) T cell, or an NK T cell or other T cell obtained from the same mammalian subject into whom the modified T cell is administered or from another member of the mammalian species.
  • the T cell is an autologous human T cell or natural killer (NK) T cell obtained from the subject or from a bone marrow transplant match for the subject.
  • NK natural killer
  • Other suitable T cells include T cells obtained from resected tumors, a polyclonal or monoclonal tumor-reactive T cell.
  • the T cell is generally obtained by apheresis, and transfected or transduced with the selected nucleic acid construct to express the BRAF V 6OOE peptide or fusion with a T helper protein in vivo.
  • a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprises administering to a subject in need thereof, i.e., a subject with melanoma, a composition of any described above.
  • the method is useful in the treatment of melanomas to induce an MHC class I, HLA-A2 restricted CTL response by the patient against the cancer.
  • These therapeutic compositions and components of the methods may be administered to a patient, preferably suspended in a biologically compatible solution or pharmaceutically acceptable delivery vehicle.
  • the various components of the methods are prepared for administration by being suspended or dissolved in a pharmaceutically or physiologically acceptable carrier such as isotonic saline; isotonic salts solution or other formulations that will be apparent to those skilled in such administration.
  • aqueous and non-aqueous isotonic sterile injection solutions and aqueous and non-aqueous sterile suspensions known to be pharmaceutically acceptable carriers and well known to those of skill in the art may be employed for this purpose.
  • a pharmaceutical composition as described above may be administered by any appropriate route, such as subcutaneous injection for the peptide compositions or intravenous injection for the checkpoint inhibitor antibody and infusion of the dendritic cells. Still other routes of administration as described above may be selected depending upon the dosage, patient condition, cancer type and stage, and other clinical factors.
  • a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprises administering to said subject a BRAF V 6ooE-based modified peptide or a pharmaceutically acceptable salt thereof and at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor.
  • a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprising administering to said subject a nucleic acid sequence encoding a BRAF V 6ooE-based modified peptide and at least one of (a) a nucleic acid sequence encoding a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor.
  • a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprising administering to said subject a dendritic cell pretreated with a BRAF V 6ooE-based modified peptide or nucleic acid sequence encoding said peptide with at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor.
  • the dendritic cell is pretreated with the T helper peptide, a fusion of the BRAF V 6ooE-based modified peptide with the T helper peptide, or a peptide spanning the T helper and CTL peptides of BRAF V 6OOE as described above.
  • the dendritic cell is pretreated or transfected with nucleic acid molecules encoding these peptides.
  • a method of treating or preventing the development of a melanoma involves administering to a mammalian subject, preferably a human, an effective amount of a pharmaceutical composition described herein.
  • the amount of the protein, peptide or nucleic acid sequences present in each effective dose is selected with regard to consideration to the half-life of the compound, the identity and/or stage of the melanoma, the patient's age, weight, sex, general physical condition and the like.
  • the amount of active component required to induce an effective CTL response against the melanoma cells without significant adverse side effects varies depending upon the pharmaceutical composition employed and the optional presence of other components.
  • each dose will comprise between about 5 ⁇ g peptide/kg patient body weight to about 10 mg/kg.
  • a useful therapeutic dosage is between 1 to 5 mg peptide/kg body weight.
  • Another embodiment of a useful dosage may be about 500 ⁇ g/kg of peptide.
  • the composition is administered at a concentration of from and including 25 to 250 micromoles of the peptide or nucleic acid sequence encoding the peptide.
  • a suitable concentration of the dose of the BRAF V 6ooE-based peptide, polypeptide, fusion with T helper or nucleic acid sequences encoding same is administered in at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, to at least about 500 ⁇ .
  • the dosages may range between a unit dosage of between 0.01 mg to 100 mg of protein (which is equivalent to about 12.5 ⁇ g/kg body weight). If any of the compositions are administered as naked DNA, the dosages may range from about 50 ⁇ g to about 1 mg of DNA per mL of a sterile solution. If the compositions is administered as a DNA vaccine in a virus, a therapeutically effective adult human or veterinary dosage of a viral vector is generally in the range of from about 100 to about 100 mL of a carrier containing concentrations of from about 1 x 10 6 to about 1 x 10 15 particles, about 1 x 10 11 to 1 x 10 13 particles, or about 1 x 10 9 to lx
  • the dosage of the checkpoint inhibitor may be adjusted based on known toxicities of the particular antibody or small molecule used.
  • the selected composition is administered in a single dose.
  • an initial dose of a composition may be optionally followed by repeated administration for a duration selected by the attending physician.
  • one to three booster doses are administered.
  • Dosage frequency may also depend upon the factors identified above, and may range from 1 to 6 doses per day for a duration of about 3 days to a maximum of no more than about 1 week.
  • the compositions may also be administered as a continuous infusion for about 3-5 days, the specific dosage of the infusion depending upon the half-life of the compound.
  • the compounds may also be incorporated into chemotherapy protocols, involving repetitive cycles of dosing. Selection of the appropriate dosing method would be made by the attending physician.
  • the peptide or nucleic acid compositions and checkpoint inhibitors are independently administered systemically by intramuscular, intraperitoneal, intravenous, intratumoral, oral or intranodal administration.
  • the subject is treated with other anti-cancer therapies before, during or after treatment with said composition.
  • the subject can be treated with chemotherapy before administering said composition.
  • this method involving co-administering the
  • immunotherapeutic compositions with the pretreated dendritic cell can also include administering a checkpoint inhibitor in the form of an antibody or a small molecule.
  • the immunotherapeutic BRAF V 6OOE -based peptide, polypeptide or fusion compositions and T helper peptide or nucleic acid compositions encoding them, or dendritic cell pretreated with them are administered substantially simultaneously.
  • these immunotherapeutic compositions and T helper sequences, optionally with the checkpoint inhibitor are administered sequentially by the same or different routes of administration.
  • the routes of administration selected depend upon the nature of the compositions. For example, if the checkpoint inhibitor is a small chemical molecule, such molecules may be administered orally in doses known and accepted for other pharmaceutical uses of these drugs.
  • the other compositions or components of the method are administered in different routes as mentioned above.
  • the subject may be treated with other anticancer therapies before, during or after treatment with the compositions or individual components of the compositions and methods above-described. Such treatment may be concurrent or simultaneous with the BRAF V 6OOE -based peptide or nucleic acid compositions or dendritic cell compositions or overlap treatment with any of these components.
  • the methods involve treating the subject with chemotherapy before administering the immunotherapeutic compositions, T helper sequences and/or checkpoint inhibitor antibodies or small molecule mimics.
  • the method further comprises depleting the subject of lymphocytes and optionally surgically resecting the tumor prior to administration of the components of the above-described methods.
  • a number of such peptides were assayed as follows: Adherent monocytes (5 x 10 4 /well) were pulsed for 8 hours with synthetic peptides (25 ⁇ ): Peptide 8 SEQ ID NO: 15, Peptide 9 SEQ ID NO: 16, and Peptide 10 SEQ ID NO: 17. At the end of incubation, excess peptides were removed and the monocytes were cultured with one of three melanoma patient PBMC samples labeled WM35, WM3457 and 3451 (1 x 10 5 ) for 5 days.
  • Proliferative responses were determined by standard HTdR incorporation assay and the results shown in FIG. 1. Data are expressed as mean cpm (triplicate
  • Adherent monocytes (5 x 10 4 /well) were pulsed for 8 h with T-helper synthetic peptides (25 ⁇ ) either alone or in combination with an affinity modified BRAF V 6OOE- based peptide, i.e., a CTL peptide of the sequence LMTEKSRWSG; SEQ ID NO: 25.
  • Cell cultures were initiated as described in Example 1 and at the end of 7 days T-cells were harvested and stained with fluorescence conjugated anti-CD4 or anti-CD8 antibodies and analyzed for T-cell binding by standard FACS assay.
  • the Th peptides induced enriched CD8 CTLs in the presence of both the Th peptide and the BRAF V 6ooE-based CTL peptide.
  • T-cell cultures were initiated as described in Examples 1 and 2. After 7 days, growing lymphocyte cultures were harvested and restimulated with Th peptide 9 of sequence EDLTVKIGDFGLATV SEQ ID NO: 16, the BRAF V6 ooE-based peptide of SEQ ID NO: 25, or both, and 20 U/ml of natural human IL-2. This process was repeated every
  • a combination of the T helper and BRAF V 600E-based CTL peptide induces more potent CTLs than either the Th or CTL peptides alone.
  • mice received s.c. injection of mouse melanoma cell line (YUMM 1.7; lxlO 5 ). After 7 days the tumor was established. Mice received BRAF V 600E-based peptide SEQ ID NO: 25 immunization (50 ⁇ g, intradermally, every 7 days) in presence of CpG (30 ⁇ g) as adjuvant. Mice also received anti-PDl antibody (250 ⁇ g via intraperitoneal injection, every 5-6 days) either alone or in combination.
  • FIG. 4 shows tumor growth measurements as mean tumor volume and SEM. The results are compared with mice that received control HIV peptide and anti-PDl antibody. Combination of peptide immunization and anti-PDl antibody therapy showed significant inhibition of tumor growth (p ⁇ 0.05). Anti-PDl antibody alone was not effective in tumor inhibition when compared to isotype control antibody treatment.
  • FIG. 4 demonstrates that a combination of immunization with a BRAF V 600E-based CTL peptide and treatment with a checkpoint inhibitor, i.e., anti-PDl antibody, has significant effect on growth of mouse melanoma (YUMM 1.7) cells in an established tumor model and induces regression of established tumor.
  • Xaa is absent, Ser, Gly or Cys
  • Xaa is a substituted or unsubstituted Ala, Leu, Met, Val,
  • Xaa is a substituted or unsubstituted Thr or Ser
  • Xaa is a substituted or unsubstituted Lys, Arg or His
  • Xaa is a substituted or unsubstituted Ser or Thr
  • Xaa is a substituted or unsubstituted Arg, Lys, or His
  • Xaa is substituted or unsubstituted Thr, Val, Leu or Ser
  • Xaa is absent or is a substituted or unsubstituted Gly, Pro or Leu
  • Xaa is absent or is a Cys, Ser or Gly
  • Xaa is Ser, Gly, Cys or absent
  • Xaa is a substituted or unsubstituted He
  • Xaa is a substituted or unsubstituted Gly, Ala, Val, Leu,
  • Xaa is Ser, Gly, Cys or absent
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is a substituted or unsubstituted Gly, Ala, Val, Leu,
  • Xaa is a substituted or unsubstituted Gly, Val or Leu
  • Xaa is absent or a Cys, Gly or Ser
  • Xaa is absent or Gly, Ser or Cys
  • Xaa is a substituted or unsubstituted Ala, He, Met, or
  • Xaa is a substituted or unsubstituted Gly, He, Val or Leu.
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is absent, Gly, Ser or Cys
  • Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
  • Xaa is a substituted or unsubstituted Ala, He, Met, Leu, Gly, Val, Pro, Phe, or Trp
  • Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
  • Xaa is a substituted or unsubstituted Ala, He, Met, Leu, Gly, Val, Pro, Phe, or Trp SEQ ID NO: Free text under ⁇ 223>
  • Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
  • Xaa is a substituted or unsubstituted Ala, He, Met, Leu,

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Mycology (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Dispersion Chemistry (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compositions and methods for treating melanoma are provided. Compositions include BRAFV6ooE-based peptides, alone or admixed with T helper peptides. Other compositions include nucleic acid sequences encoding the BRAFV6ooE-based peptides, alone or admixed with nucleic acid sequences T helper peptides. Dendritic cells pretreated with the BRAFv6ooE-based peptides, alone or admixed with T helper peptides, are also provided. These compositions are useful to treat melanoma, optionally co-administered with antibodies to checkpoint inhibitors or molecules that mimic the action of such antibodies.

Description

METHODS AND COMPOSITIONS FOR TREATING MELANOMA
STATEMENT OF GOVERNMENT INTEREST
This invention was made with government support under Grant Nos. P01
CA114046 and P30 CA010815 awarded by the National Institutes of Health. The government has certain rights in the invention.
INCORPORATION-BY-REFERENCE OF MATERIAL SUBMITTED IN
ELECTRONIC FORM
Applicant hereby incorporates by reference the Sequence Listing material filed in electronic form herewith. This file is labeled "WST163PCT_ST25.txt " and dated February 15, 2017 with a size of 22KB.
BACKGROUND OF THE INVENTION
BRAF is an intracellular signaling protein expressing frequently in melanomas for which alleles were identified as somatic mutations in 70% of melanomas, the majority of all types of nevi, and a minority of other cancers including lung, colon and ovary carcinomas, but not in normal cells. The BRAF mutations were located in exons 11 or 15, with BRAFV600E representing nearly all (92%) the BRAF alleles in melanoma. BRAFV6OOE has oncogenic activity through activation of the MAP kinase pathway.
Certain BRAFV6OOE based peptides that induce MHC Class I, HLA-A2-dependent cytotoxic T cell responses were identified in US Patent No. 7,811,993, which is incorporated herein by reference. These BRAFV6OOE based peptides were designed based on the amino acids 597 to 606 of the BRAFV6OOE sequence (SEQ ID NO: 1) for use in prophylactic and therapeutic treatments for melanoma. More specifically, the
compositions described therein were useful in inducing responses that were not patient- specific, but that were specific for a mutation that occurs in about 70% of all melanoma patients.
There nevertheless remains a need in the art for additional pharmaceutical compositions and methods useful for treatment, prevention and diagnosis of melanoma in a large majority of patients. SUMMARY OF THE INVENTION
In one aspect, a composition comprises a BRAFV6OOE based peptide that induces MHC Class I, HLA-A2-dependent cytotoxic T cell responses peptide or a
pharmaceutically acceptable salt thereof. In one embodiment, this composition comprises peptides of the Formula II, III or IV as described herein.
In another aspect, a composition comprises a BRAFV6OOE based peptide, including a peptide of Formula I, II, III or IV, described below that induces MHC Class I, HLA-A2- dependent cytotoxic T cell responses and a T helper peptide.
In another aspect, the compositions recited herein further comprise an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
In one aspect, a composition comprises a nucleic acid sequence encoding a BRAFV600E based peptide of Formula II, III or IV as described below.
In another aspect, a composition comprises a nucleic acid sequence encoding a BRAFV600E based peptide of Formula I, II, III or IV below that induces MHC Class I,
HLA-A2-dependent cytotoxic T cell responses and a nucleic acid sequence encoding a T helper sequence.
In another aspect, the nucleic acid sequence-containing compositions recited herein further comprise a nucleic acid sequence encoding a checkpoint inhibitor or an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
In another aspect, a composition comprises a recombinant dendritic cell that is pretreated ex vivo with a composition containing a BRAFV6OOE based peptide, an optional T helper peptide and/or an optional antibody that binds a checkpoint inhibitor or an optional molecule that mimics the function of a checkpoint inhibitor as described herein.
In another aspect, a recombinant dendritic cell pretreated ex vivo with a composition containing a nucleic acid sequence encoding a BRAFV6OOE based peptide, optionally including a nucleic acid sequence encoding a T helper peptide and/or an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor as described herein. In another aspect, a composition comprises the pretreated dendritic cell described herein with an antibody that binds a checkpoint inhibitor in a formulation suitable for simultaneous administration.
In a further aspect, a composition as described herein for use in the treatment or prophylaxis of melanoma in a mammalian subject is provided.
In another aspect, a method of treating or retarding or preventing the development of cancer, particularly melanoma, in a mammalian subject comprises administering to said subject one or more of the compositions as described herein.
Other aspects and advantages of these compositions and methods are described further in the following detailed description of the preferred embodiments thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows three graphs of results of proliferative assays for three different melanoma peripheral blood mononuclear cell (PBMC) samples labeled WM35, WM3457 and 3451 with peptides derived from the BRAFV6OOE sequence. The peptides were screened for T-helper sequences (Th): Peptide P8 is amino acids residues 608-622 of BRAF, i.e., SHQFEQLSGSILWHA SEQ ID NO: 15. Peptide P9 is amino acids residues 546-600 of BRAF, i.e., EDLTVKIGDFGLATV SEQ ID NO: 16. Peptide P10 is amino acids residues 547-601 of BRAF, i.e., DLTVKIGDFGLATVK SEQ ID NO: 17. These peptides bind to HLA-DR alleles using computer algorithms, as disclosed in Example 1 below. Proliferative responses were determined by standard HTdR incorporation assay. Data are expressed as mean cpm (triplicate determinations) plus SD (bar) of HTdR incorporation. * Values are significantly (p<0.01) different from the value obtained with the control peptide. This figure demonstrates that T helper peptides can induce proliferative response in melanoma patients PBMCs.
FIG. 2 is a graph showing the induction of enriched CD 8 cytotoxic T cells (CTLs) in the presence of both a T helper and a BRAFV6OOE - based CTL peptide. The BRAFV6OOE - based CTL peptide is that of the sequence LMTEKSRWSG SEQ ID NO: 25. The T helper sequence is that of P9 SEQ ID NO: 16. According to the process of Example 2, adherent monocytes were pulsed with the T helper synthetic peptides (25 μΜ) either alone or in combination with the affinity modified BRAFV6OOE - based CTL peptide and cell cultured. T-cells were harvested and stained with anti-CD4 or anti-CD8 antibodies and analyzed for T-cell binding by standard FACS assay.
FIG. 3 is a graph showing the results of a standard 51Cr-release assay conducted on melanoma cells exposed to lymphocytes that had been stimulated with Th peptide P9 SEQ ID NO: 16 identified above in FIG. 1, the BRAFV6ooE-based CTL peptide SEQ ID NO: 25, or both peptides and with natural human IL-2, as described in Example 3. The lymphocytes were tested for cytolytic activity against HLA-A2+ autologous WM35 (V600E+) or HLA-A2+ matched allogeneic WM3456 (V600E-) melanoma cells.
Indicated effector (T-cells) to tumor target ratios were used in the assay.
FIG. 4 is a graph showing the effect of a BRAFV6OOE -based CTL peptide SEQ ID
NO: 25, and a checkpoint inhibitor, which is an anti-PDl antibody, in an established tumor model described in Example 4. Tumor growth measurements are shown as Mean tumor volume and SEM and the results are compared with mice that received control HIV peptide and anti-PDl antibody. Combination of peptide immunization and anti-PDl antibody therapy showed significant inhibition of tumor growth (p<0.05). Anti-PDl antibody alone was not effective in tumor inhibition when compared to isotype control antibody treatment.
DETAILED DESCRIPTION
Compositions and methods are described herein that provide alternative and enhanced methods of treatment of melanoma and other cancers by the use of certain BRAFV6ooE based peptides, compositions containing the peptides and nucleic acid compositions encoding these peptides..
Certain components and definitions used in the description of these compositions and methods are defined below.
As used herein the term BRAFV6OOE refers to the human amino acid sequence of SEQ ID NO: 1 in which the amino acid residue at 600 is changed from the Val residue in the native sequence to a Glu. The nucleic acid sequence encoding BRAFV6OOE appears in SEQ ID NO: 21. In one embodiment, the nucleic acid sequence encoding amino acids 595 to 620 of SEQ ID NO: 1 was most often employed to make the changes from the native sequence to produce the peptides or nucleic acid compositions described herein. Native human BRAF amino acid and encoding nucleic acid sequences are shown in the publically available NCBI database under Accession No. NM_004333.4. Other published sequences are available from NCBI or GenBank databases
As used herein, the term "BRAFV6OOE based peptides" or "BRAFV6OOE based CTL peptides" refers to peptides based on amino acids 597-606 of BRAFV6OOE sequence, namely Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly SEQ ID NO: 2 that induce MHC
Class I, HLA-A2-dependent cytotoxic T cell responses. In one embodiment, such peptides included in this definition are the peptides identified in US Patent No. 7,811,993, incorporated by reference herein. Such peptides are defined by the Formula I: Rl-Leu- AA2-AA3-Glu-AA5-AA6-AA7-Trp-AA9-AA10-R2 (SEQ ID NO: 3), or a
pharmaceutically acceptable salt thereof, wherein AA2 is an a substituted or unsubstituted Ala, Leu, Met, Val, Pro and Gly; AA3 is a substituted or unsubstituted Thr and Ser; AA5 is a substituted or unsubstituted Lys, Arg and His; AA6 is a substituted or unsubstituted Ser and Thr; AA7 is a substituted or unsubstituted Arg, Lys, and His; AA9 is a substituted or unsubstituted Thr, Val, Leu and Ser; AA10 is absent or is a substituted or unsubstituted Gly, Pro or Leu.
Also included in this definition of BRAFV6OOE based peptides are the peptides defined by Formulae II, III and IV. Formula II is Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp- Ser-AA10 -R2 (SEQ ID NO:4), wherein AA2 is a substituted or unsubstituted He, and AAIO is a hydrophobic residue. Formula III is Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp- Ser-AA10 -R2 (SEQ ID NO: 5), wherein AA2 is a hydrophobic residue and AA10 is a substituted or unsubstituted Gly, Val or Leu. Formula IV is Rl-Leu-AA2-Thr-Glu-Lys- Ser-Arg-Trp-Ser-AA10 -R2 (SEQ ID NO: 6), wherein AA2 is a substituted or
unsubstituted Ala, He, Met, or Leu, and wherein AA10 is a substituted or unsubstituted Gly, He, Val or Leu.
Also, according to Formula I, II, III or IV, in some embodiments of peptides of each formula, Rl and R2 are each independently absent or provide a spacer for coupling of a peptide of each formula to a second peptide or protein at the N- or C-termini of the peptide. Such spacers may be amino acid sequences or chemical compounds ordinarily used as spacers. For example, in one embodiment, Rl is a Cys; in another embodiment, Rl is a Gly-Ser. In another embodiment R2 is a Cys. In still another embodiment, R2 is a Gly-
Ser. In still further embodiments, Rl and R2 are identical. In another embodiment, Rl and R2 are different. In another embodiment of a peptide of Formula II, III or IV, Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein.
By "hydrophobic residue" is meant a substituted or unsubstituted amino acid which imparts hydrophobicity to the resulting BRAFV600E-based peptide. Unsubstituted amino acids having hydrophobic side chains are glycine (Gly), alanine (Ala), valine (Val), leucine (Leu), isoleucine (He), proline (Pro), phenylalanine (Phe), methionine (Met), and tryptophan (Trp). Various substitutions or modifications to the amino acids may make then more or less hydrophobic.
Examples of specific BRAFV6OOE based peptides of Formula I through IV include, among other s equences :
Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 7);
Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 8);
Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 9);
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Ile-R2 (SEQ ID NO: 10);
Rl-Leu-Met-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 11),
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 12);
Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 13); and
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Leu-R2 (SEQ ID NO: 14).
By "T helper peptide" or "T helper sequence" or "Th peptide" as used herein is meant certain T helper sequences, or modified variants thereof of BRAFV6OOE SEQ ID NO:
1, namely, amino acids 607-621, 588 to 600 and 589 to 601 of SEQ ID NO: 1. In one embodiment, a suitable Th peptide is Ser-His-Gln-Phe-Glu-Gln-Leu-Ser-Gly-Ser-Ile-Leu- Trp-His-Ala SEQ ID NO: 15, which spans aa 607 to 622 of BRAFV6OOE. In another embodiment, a suitable Th peptide is Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly- Leu-Ala-Thr-Val SEQ ID NO: 16, which spans AA 588 to 600 of BRAFV6OOE. In another embodiment, a suitable Th peptide is Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu- Ala-Thr-Val-Lys SEQ ID NO: 17, which spans AA589 to 601 of BRAFV6OOE. This definition of suitable Th sequences also includes modified versions of the peptide sequences and pharmaceutically acceptable salts thereof. Peptides spanning both the T helper and BRAFV6OOE based peptides, such as spanning AA597 to 622 SEQ ID NO: 32, or
AA588 to 607 SEQ ID NO: 33 or AA589 to 607 SEQ ID NO: 34, or other sequences derived from the BRAFV6OOE sequence are also possible. The definitions of BRAFV6OOE based peptides and T helper peptides also include modified versions of the peptide sequences and pharmaceutically acceptable salts thereof.
By "modified" peptides of the above Formulae I-IV is meant homologous or analogous modified sequences, wherein non-variable amino acids may be conservatively replaced individually by amino acid residues having similar characteristics. In one embodiment, the non-variable amino acid residues may be replaced by other amino acid residues bearing the same charge and/or similar side chain lengths. Similarly the non- variable naturally-occurring amino acids may be replaced by non-naturally occurring amino acid residues. For peptides of the formulae above, an amino acid residue may be a naturally-occurring amino acids, meaning one of the twenty amino acids that occur in nature in L form, which include alanine, cysteine, aspartate, glutamate, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan, and tyrosine, or any derivative thereof produced through a naturally-occurring biological process or pathway.
Also encompassed by the term "modifications" of the Formulae I - IV above are non-naturally-occurring amino acids. This latter term is used herein to refer to an amino acid other than a naturally-occurring amino acid as defined above, which can be synthesized or "man-made", and including a derivative thereof, whether produced synthetically or via a biological process or pathway. Non-naturally occurring amino acids include, without limitation, D amino acids, amino acids containing unnaturally substituted side chains, e.g., an N-methylated amino acid, cyclic amino acids, diamino acids, B-amino acids, homo amino acids. In some embodiments, the non-naturally occurring amino acids used in the above formulae are only those that do not strain the binding formation by adding extra atoms to the peptide backbone, because backbone hydrogen bonding contact with the MHC is desirable for these peptides. Non-naturally-occurring or unnatural amino acids may be characterized by novel backbone and side chain structures and are widely available from commercial reagent suppliers, such as Sigma- Aldrich (on the world-wide web at sigmaaldrich.com), on the world-wide web at Netchem.com, and other sites. Such non-naturally occurring amino acid(s) when employed in the peptides and compositions herein are anticipated to make the compounds more resistant to degradation by mammalian enzymes in serum, saliva, stomach and intestines, and thus compounds that are composed of one or more such amino acids may confer upon the compound enhanced stability and bioavailability in vivo. A variety of methods for producing non-natural amino acids are known and may be selected by one of skill in the art.
In one embodiment, a class of non-naturally occurring amino acids includes L amino acids that effect stereochemistry. Thus, in one embodiment of compounds, one or more of the amino acids in the peptide may be in L form, while others may be in D form.
Another non-naturally occurring amino acid is an amino acid which is modified to contain a substitution on the alpha-carbon in the amino acid structure. For example the alpha- carbon may be substituted by a suitable hydrocarbon moiety, such as aminoisobutyrate. Still another class of non-naturally occurring amino acids is amino acids which are modified or mutated to extend their carbon chain length. For example, an amino acid with a single alpha-carbon chain, may be extended with at least one additional carbon, i.e., a beta-carbon, and so on. An additional modification to an amino acid is the insertion of a substituent on the nitrogen of the amino group. An example of this type of modification is an N-methyl amino acid. The addition of substituents on the alpha carbon or additional carbons or on the nitrogen of the amino acid molecule may occur in any of the amino acids of the formulae above.
Among useful substituents for creating the non-naturally occurring amino acids are straight chain, branched, cyclic or heterocyclic C1-12 alkyl groups, and straight chain, branched, cyclic, or heterocyclic C1-12 alkanoyl groups. The amino acid may be also modified by the insertion of modifying sugars, imide groups and the like. Other amino acids are substituted in the ortho or meta position by a substituent such as H, OH, CH3, halogen, OCH3, NH2, CH or N02.
A non-exclusive list of modified or non-naturally occurring amino acids for inclusion in compounds fitting the formulae above include amino acids modified by N- terminal acetylation, C-terminal amidation, formylation of the N-terminal methionine, gamma-carboxy glutamic acid hydroxylation of Asp, Asn, Pro or Lys residues in the compound, methylation of Lys or Arg, preferably; phosphorylation of Ser, Thr, Tyr, Asp or His in the compound, use of a pyrrolidone carboxylic acid, which is an N-terminal glutamate which has formed an internal cyclic lactam, sulphonation of Tyr, generally. Still other modifications of non-naturally occurring amino acids include use of or substitution with the following moieties: a 2-aminoadipic acid group, a 3-aminoadipic acid group, beta-Ala or beta-aminopropionic acid group, 2-aminobutryic acid, 4-aminobutyric acid, piperidinic acid, 6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutryic acid, 3- aminoisobutyric acid, 2-aminopimelic acid, 2, 4 diaminobutyric acid, desmosine, 2,2'- diaminopimelic acid, 2,3-diaminopropionic acid, N-ethylglycine, N-ethylglycine, N-ethyl asparagine, hydroxylysine, allo-hydroxylysine, 3 -hydroxy proline, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N-methylisoleucine, 6-N- methyllysine, N-methylvaline, 6-N-methyllysine, norvaline, norleucine, and ornithine.
Still other modifications of peptides of Formulae I-IV include fusions into polypeptides or other multimeric constructs or compositions, when either Rl or R2 are absent or optional amino acids (e.g., Cys, -Gly-Ser-) or other amino acid or chemical compound spacers may be included at the N- or C-termini of the peptide for the purpose of linking two or more of the same or different BRAFV600E-based peptides together or linking a peptide to a second peptide, such as one of the Th peptides or to a carrier. Preferably, the spacer of Rl or R2 is a proteolytically sensitive spacer to permit cleavage of the epitope before it enters the cell compartment where it associates with MHC. In one embodiment multiple copies of the same BRAFV6ooE-based peptides are linked sequentially and expressed as a recombinantly or synthetically produced polypeptide. In one embodiment, multiple different BRAFV600E-based peptides are linked sequentially, with and without spacer amino acids therebetween, to form a larger recombinant or synthetic fused polypeptide. In one embodiment, at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 repeating units of the same BRAFV600E-based peptide forms a polypeptide. In another embodiment, at least 2, 3,
4, 5, 6, 7, 8, 9, or 10 repeating units of the same fusion peptide, i.e., a BRAFV600E-based peptide-T helper peptide fusion. In still a further embodiment, multiple BRAFV600E-based peptides of fusion peptides as described are coupled to a carrier. Such peptides and multimeric compositions may be produced synthetically or recombinantly by conventional methods. In one embodiment, the peptides are prepared conventionally by known chemical synthesis techniques.
By "carrier" is meant a protein, peptide or other substrate that may enhance stability or delivery, improve the production, or change the activity spectrum of the peptide. As a few well-known examples, such carriers may be human albumin, keyhole limpet hemocyanin, polyethylene glycol, other biopolymers or other naturally or non- naturally occurring polymers. In one embodiment, the moiety is desirably a protein or other molecule which can enhance the stability of the peptide or enhance its penetration into the targeted cell. Still other proteins or peptides to which the BRAFV6ooE-based peptides described herein may be linked via the spacer include keyhole limpet hemocyanin or additional MHC molecules. Still other carriers include a live antigen-presenting cell, such as a dendritic cell, which presents the peptides described herein. Still alternative carriers or peptide-carrier constructs utilize lipopeptides.
By the term "nucleic acid sequence that encodes a BRAFV6ooE-based peptide" as used herein is meant an RNA or DNA sequence encoding the amino acid sequence of a BRAFv6ooE-based peptide as described herein, such as a peptide of any of Formula I through IV. SEQ ID NO: 21 provides the DNA sequence of BRAFV6OOE from which other modified nucleic acid sequences can be obtained, e.g., encoding the modified peptides or fusion peptides described herein. By "nucleic acid sequence that encodes a T helper sequence" as used herein is meant an RNA or DNA sequence encoding the amino acid sequence of T helper as described herein, such as those of SEQ ID Nos: 15, 16 and 17, among others. Nucleic acid sequences may also encode polypeptide or fusions of the referenced peptides. The nucleic acid sequences may be generated and/or modified by conventional techniques and useful in prophylactic, diagnostic, and therapeutic compositions and methods designed for delivery of the nucleic acid in vivo and expression of the peptide in vivo. Nucleic acid sequences encoding the peptides described herein may be prepared by known recombinant DNA techniques and used to clone and express the peptides within a host microorganism or cell. Such nucleic acid sequences may also encode different or preferred codons for the peptide described.
Alternatively, such nucleic acid sequences encoding these peptides may be designed to incorporate other nucleic acid sequences necessary for delivery to a subject, e.g., as naked DNA or other DNA vaccine forms. For example, a suitable plasmid may be constructed containing a nucleic acid sequence encoding the selected BRAFV6ooE-based peptide or T helper peptide, or both, under the control of regulatory sequences directing expression thereof in a mammalian or vertebrate cell. The components of the plasmid itself are conventional. Still other nucleic acid constructs and vectors of viral and bacterial origin, may be employed. See, e.g., US Patent No. 7,811,993, incorporated by reference herein.
As used herein, the term "checkpoint inhibitor" refers to a composition or composition in the form of an antibody or a small molecule that binds or inhibits various checkpoint proteins. Such checkpoint proteins, include, without limitation, PD-1 , PD-L1, CTLA-4, BTLA and CD 160. As examples, known checkpoint inhibitors include the antibodies ipilimumab (Yervoy®), pembrolizumab (Keytruda®), and nivolumab
(Opdivo®), among others. Other checkpoint inhibitors developed as small molecules or other checkpoint binding antibodies or antibody fragments are included in this definition.
As used herein, the term "antibody" refers to all types of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including antibody fragments. The antibody can be monoclonal or polyclonal and can be of any species of origin, including (for example) mouse, rat, rabbit, horse, goat, sheep, camel, or human, or can be a chimeric antibody. See, e.g., Walker et al, Molec. Immunol. 26:403 (1989). The antibodies can be recombinant monoclonal antibodies produced according to known methods, see, e.g., U.S. Patent Nos. 4,474,893 or 4,816,567, which are incorporated herein by reference. The antibodies can also be chemically constructed according to known methods, e.g., US Patent No.
4,676,980 which is incorporated herein by reference. See also, US Patent No. 8,613,922, which is incorporated herein by reference. An antibody includes, without limitation, a synthetic antibody, a recombinant antibody, a chimeric antibody, a humanized antibody, a human antibody, a CDR-grafted antibody, a multispecific binding construct that can bind two or more targets, a dual specific antibody, a bi-specific antibody or a multi-specific antibody, or an affinity matured antibody. Antibody fragments include without limitation antigen binding fragments such as a single antibody chain or an scFv fragment, a diabody, a single chain comprising complementary scFvs (tandem scFvs) or bispecific tandem scFvs, an Fv construct, a disulfide-linked Fv, a Fab construct, a Fab' construct, a F(ab')2 construct, an Fc construct, a monovalent or bivalent construct from which domains nonessential to monoclonal antibody function have been removed, a single-chain molecule containing one VL, one VH antigen-binding domain, and one or two constant "effector" domains optionally connected by linker domains, a univalent antibody lacking a hinge region, a single domain antibody, a dual variable domain immunoglobulin (DVD-Ig) binding protein, a nanobody, a domain antibody, a vaccibody, a linear antibody; a heavy chain or light chain complementarity determining region, and multispecific antibodies formed from antibody fragments. Also included in this definition are antibody mimetics such as affibodies, i.e., a class of engineered affinity proteins, generally small (-6.5 kDa) single domain proteins that can be isolated for high affinity and specificity to any given protein target. Such antigen-binding fragments can be produced by known techniques.
The term "patient" or "subject" as used herein means a mammalian animal, including a human, a veterinary or farm animal, a domestic animal or pet, and animals normally used for clinical research. In one embodiment, the subject treated with the methods and composition is a human. In another embodiment, the subject treated with the methods and composition has a cancer. In another embodiment, the subject of these methods has melanoma.
As used herein the term "cancer" refers to or describes the physiological condition in mammals that is typically characterized by unregulated cell growth. In one embodiment, the term "cancer" means any cancer characterized by the presence of a solid tumor. In one embodiment, suitable cancers for treatment by the methods described herein, include, cancers having the presence of the BRAFV6OOE mutation. In another embodiment, suitable cancers include, without limitation, melanoma, breast cancer, lung cancer and ovarian cancer with such mutations. In another embodiment, suitable cancers include, without limitation, other forms of melanoma, brain cancer, colon/rectal cancer, lung cancer, ovarian cancer, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, bone cancer, endometrial cancer, esophageal cancer, eye cancer, kidney cancer, laryngeal cancer, liver cancer, head and neck cancer, nasopharyngeal cancer, osteosarcoma, oral cancer, pancreatic cancer, prostate cancer, rhabdosarcoma, salivary gland cancer, stomach cancer, testicular cancer, thyroid cancer, vaginal cancer, lung cancer, and neuroendocrine cancer
The term "tumor," as used herein, refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. In one embodiment, the tumor targeted by the methods is characterized by hypoxia, significant infiltration with T lymphocytes, and low glucose in the tumor microenvironment.
By "therapeutic reagent" or "regimen" is meant any type of treatment employed in the treatment of cancers with or without solid tumors, including, without limitation, chemotherapeutic pharmaceuticals, biological response modifiers, radiation, diet, vitamin therapy, hormone therapies, gene therapy, surgical resection, etc. By "an immunotherapeutic composition" or "immunotherapeutic vaccine" is meant any composition including the BRAFV600E-based peptides or nucleic acid sequences encoding same that stimulate the subject's immune system. Such immunotherapeutic compositions are designed to elicit a humoral (e.g., antibody) or cellular (e.g., a cytotoxic T cell or T helper) response, to the BRAF target gene product delivered by the
immunogenic composition to a subject. The immunotherapeutic compositions or vaccines, as described herein, are created using known recombinant and synthetic techniques.
In one embodiment immunotherapeutic compositions useful in these methods involve presentation of the BRAFV600E-based peptides to the subject's immune system. In another embodiment the immunotherapeutic composition used in the methods described herein is a DNA or RNA composition including a nucleic acid encoding the BRAFV6OOE- based peptides. In another embodiment the BRAFV6ooE-based peptides or encoding nucleic acid sequences are delivered via virus vectors, e.g., adenovirus, adeno-associated virus, lentivirus, retrovirus, poxvirus or others, or via virus-like particles (VLP). In another embodiment the immunotherapeutic composition used in the methods described herein is a monoclonal antibody or antigen-binding fragment(s) that specifically bind cancer antigens.
In another embodiment, the immunotherapeutic composition is a dendritic cell. Dendritic cell therapy involves the harvesting of autologous blood cells (monocytes) from a patient and pretreating the cells with, a BRAFV6ooE-based peptide (optionally with a Th peptide) ex vivo. By "pre-treatment with" is meant that the autologous dendritic cell is cultured and expanded in the presence of the selected BRAFv6ooE-based peptide at between about 1 to about 500 μΜ, to condition the dendritic cell to activate the immune system upon reinfusion into the subject. The time of pretreatment in one embodiment means the entire time of in vitro culture, which can span several hours to at least several days. In another embodiment, the time of pretreatment is minimally 24 hours of in vitro culture. Other time periods for pre-treatment with the peptides or nucleic acid components of the compositions described herein may be at least 1, 5, 10, 15, or 20 or more hours, or any intervening times between any specified number of hours stated herein. The pretreated dendritic cells are reintroduced, e.g., by i.v. injection, back to a patient in order to allow massive dendritic cell participation in optimally activating the immune system. By "vector" is meant an entity that delivers a heterologous molecule to cells, either for therapeutic or vaccine purposes. As used herein, a vector may include any genetic element including, without limitation, naked DNA, a phage, transposon, cosmid, episome, plasmid, or a virus or bacterium. Vectors are generated using the techniques and sequences provided herein, described in the examples and in conjunction with techniques known to those of skill in the art. Such techniques include conventional cloning techniques of cDNA such as those described in texts such as Green and Sambrook, Molecular Cloning: A Laboratory Manual. 4th Edit, Cold Spring Harbor Laboratory Press, 2012, use of overlapping oligonucleotide sequences of the Salmonella genomes, polymerase chain reaction, and any suitable method which provides the desired nucleotide sequence.
By the term "pharmaceutically acceptable carrier or vehicle" is meant a solution or suspension that is safe for human administration. Optionally such carriers enhance stability and/or immunogenicity. Such carriers include, for example, water, saline, buffered saline, alcohols, gum Arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates, such as lactose, amylase or starch, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, hydroxymethylcellulose, polyvinylpyrrolidone, liposomes, oil in water emulsions and others. The compositions may further include a detergent to make the peptide more bioavailable, e.g., octylglucoside.
By "adjuvant" is meant a substance that enhances the immune response when administered together with an immunogen or antigen. In one embodiment, a suitable adjuvant is CPG. In another embodiment a suitable adjuvant is encapsulated PLG. Still other useful adjuvants are known, and include those listed and disclosed in US Patent No. 7,811,993, and its cited references, which are all incorporated herein by reference.
By "administering" or "route of administration" is meant delivery of the
BRAFV6ooE-based peptides, polypeptides, nucleic acid constructs, T helper peptides or nucleic acid constructs encoding them, or the checkpoint inhibitor or the pre-treated dendritic cells used in the methods herein, to the subject. As discussed in detail below, these methods can be independent for each component of the method. Each
administration method can occur with or without a pharmaceutical carrier or excipient, or with or without another chemotherapeutic agent into the subject. Conventional and pharmaceutically acceptable routes of administration include, but are not limited to, systemic routes, such as intraperitoneal, intravenous, intradermal, subcutaneous, and other parenteral routes of administration or intratumoral or intranodal administration. In one embodiment, the route of administration is intradermal or subcutaneous for the peptide(s) and polypeptides. In another embodiment, the route of administration is intraperitoneal or intravascular for the checkpoint inhibitor antibodies. In another embodiment, the route of administration is intravascular or intravenous for delivery of the dendritic cell-based vaccine. Routes of administration may be combined, if desired. In some embodiments, the administration is repeated periodically, as discussed in detail below.
In the context of the compositions and methods described herein, reference to "one or more," "at least five," etc. of the compositions, peptides, antibodies or other components listed means any one or any and all combinations of them.
The words "comprise", "comprises", and "comprising" are to be interpreted inclusively rather than exclusively, i.e., to include other unspecified components or process steps.
The words "consist", "consisting", and its variants, are to be interpreted exclusively, rather than inclusively, i.e., to exclude components or steps not specifically recited.
As used herein, the phrase "consisting essentially of limits the scope of a described composition or method to the specified materials or steps and those that do not materially affect the basic and novel characteristics of the described or claimed method or composition. Whereever in this specification, a method or composition is described as "comprising" certain steps or features, it is also meant to encompass embodiments of the same method or composition consisting essentially of those steps or features and/or consisting of those steps or features.
As used herein, the term "about" means a variability of 10 % from the reference given, unless otherwise specified.
It is to be noted that the term "a" or "an", refers to one or more, for example, "an miRNA," is understood to represent one or more miRNAs. As such, the terms "a" (or "an"), "one or more," and "at least one" are used interchangeably herein.
Unless defined otherwise in this specification, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs and by reference to published texts, which provide one skilled in the art with a general guide to many of the terms used in the present application.
Compositions For Use as Immunotherapeutic Vaccines
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide having the Formula II: Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10 -R2 (SEQ ID NO:4). According to this formula, Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein, and AA2 is a substituted or unsubstituted He and AA10 is a substituted or unsubstituted hydrophobic residue. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG.
In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334
In another embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide having the Formula III: Rl -Leu- AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser- AA10 -R2 (SEQ ID NO:5). According to this formula, Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein, and AA2 is a hydrophobic residue and AAIO is a substituted or unsubstituted Gly, Val or Leu. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1 LAG3, or TIM334.
In another embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide having the Formula IV: Rl-Leu-AA2-ΓΓnr-Glu-Lys-Ser-Arg-T -Ser-AA10-R2
(SEQ ID NO:6). According to this formula, Rl and R2 are independently absent or a Cys for coupling to an additional peptide or protein, and AA2 is a substituted or unsubstituted Ala, He, Met, or Leu, and wherein AA10 is a substituted or unsubstituted Gly, He, Val or Leu. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 SEQ ID NO: 7 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 SEQ ID
NO: 8 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 SEQ ID NO: 9 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Ile-R2 SEQ ID
NO: 10 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Met-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 SEQ ID NO: 11 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 SEQ ID
NO: 12 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 SEQ ID NO: 13 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In one embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Leu-R2 SEQ ID NO: 14 or a pharmaceutically acceptable salt thereof. In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In another embodiment, a composition useful in the treatment of melanoma or other cancers comprises a peptide or a pharmaceutically acceptable salt thereof, the peptide of Formula I, II, III or IV and a T helper peptide. In one embodiment, the T cell peptide is coupled or fused at either the N- or C- termini of the peptide of the BRAFV6OOE- based peptide or polypeptide. In one embodiment, the T helper peptide is Ser-His-Gln- Phe-Glu-Gln-Leu- Ser-Gly-Ser-Ile-Leu-Trp-His-Ala SEQ ID NO: 15. In one embodiment, the T helper peptide is Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val SEQ ID NO: 16. In one embodiment, the T helper peptide is Asp-Leu-Thr-Val-Lys-Ile- Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val-Lys SEQ ID NO: 17.
In yet another embodiment of a composition containing a fused BRAFV600E-based peptide or polypeptide and a T helper sequence is the sequence spanning the Th and CTL sequences found in BRAFV6OOE- In one embodiment, this sequence spans about AA586 to AA 606 and has the formula, e.g., Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu- AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10 SEQ ID NO: 18 with AA2 and AA10 as described in any of Formula II, III or IV above.
In another embodiment, this sequence spans about AA587 to AA 606 and has the formula, e.g., Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-AA2-Thr-Glu-Lys-Ser- Arg-Trp-Ser-AA10 SEQ ID NO: 19, with AA2 and AA10 as described in any of Formula II, III or IV above. In still another embodiment, this sequence spans about AA597 to AA621 and has the formula, e.g., Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-Ser-His- Gln-Phe-Glu-Gln-Leu-Ser-Gly-Ser-Ile-Leu-Trp-Met-Ala SEQ ID NO: 20 with AA2 and
AAIO as described in any of Formula II, III or IV above. Still other embodiments of these sequences can contain modified amino acids, or replacement amino acids in the positions specified in Formula I, or conservative replacements, possible spacer amino acids on either end of the peptide, or multiple Th sequences flanking the amino- or carboxy amino acid of the whole peptide, as described above. Nucleic acid sequences encoding these sequences are also useful in these compositions.
In certain embodiments, a composition such as described herein includes a pharmaceutically acceptable vehicle or carrier. In certain embodiments, a composition such as described herein includes a liposome. In certain embodiments, a composition such as described herein includes an adjuvant. In certain embodiments, a composition such as described herein includes the adjuvant CPG or encapsulated PLG. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In another embodiment, a composition useful in the treatment of melanoma or other cancers comprises multiple copies of the same BRAFV6OOE -based peptide or polypeptide fused or coupled together. In another embodiment, a composition useful in the treatment of melanoma or other cancers comprises multiple copies of a fused peptide, which comprises same BRAFV6OOE -based peptide or polypeptide fused or coupled to a T helper peptide. In another embodiment, such compositions comprise a BRAFV6OOE -based polypeptide or fusion formed from 5 to 10 of the same fused in frame or via a spacer. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR,
OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In still further embodiments, a composition useful in the treatment of melanoma or other cancers comprises one or multiple of the BRAFV6OOE -based peptide or polypeptides or fusions with T helper sequences as described herein. Any number of single peptides or multimeric constructs may be mixed together to form a single composition. The peptides may be formulated with a pharmaceutically acceptable carrier, adjuvant, diluent, other optional components, or some combination thereof. For use in such compositions, the selected peptide(s) may be produced preferably synthetically, but also recombinantly, as disclosed above. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PDl, PDLl, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In still further embodiments, a composition useful in the treatment of melanoma or other cancers comprises one or multiple of the BRAFV6OOE -based peptides described herein in the form of a salt with an acid. The compounds have at least one amino/amine groups which can form salts. Where two or more amino groups are present in the compound, a formulation of mixed salts can be prepared. Acids which can be used preferably include compatible inorganic acids such as hydrochloric and organic acids (or salts thereof) more preferably those occurring in living organisms, including but not limited to oxalic acid, glucuronic acid, pyruvic acid, lactic acid, citric acid, isocitric acid- ketoglutaric acid, succinic acid, malic acid, and oxaloacetic acid. In the preferred case of an aqueous solution, the desired anion can be added either as the free acid, or a salt, preferably one which is highly soluble in water, for example the sodium or potassium salts, but also the lithium, magnesium, calcium or ammonium salts. Moreover, these salts can be used either in anhydrous or hydrated forms. For example citric acid can be used as the anhydrous free acid, the monohydrate free acid, the anhydrous trisodium salt, or the dihydrate trisodium salt. These salts can be prepared by the methods described in
International Patent Publication No. WO 96/02269, incorporated by reference herein.
In still another embodiment, a composition useful in the treatment of melanoma or other cancers comprises a nucleic acid sequence, such as a DNA vaccine, encoding any of the components of the compositions containing BRAFV600E-based peptides of Formula I, II, III or IV, or polypeptides comprising two or more copies of the BRAFV600E-based peptides or fusions with T helper sequences described above. In one embodiment, a nucleic acid sequence for use as DNA vaccines can take the form of a recombinant vector carrying the above-described peptide-encoding nucleic acid sequence. In another embodiment, the nucleic acid sequences may be carried, and the BRAFV6OOE peptides are expressed by, plasmid based systems, of which many are commercially available or in replicating or non-replicating recombinant viral vectors. The nucleic acid sequences discussed herein may be expressed and produced using such vectors in vitro in desired host cells or in vivo in a mammalian subject. In one embodiment, the vector is a nonpathogenic virus. In another embodiment, the vector is a non-replicating virus. In one embodiment, a desirable viral vector may be a retroviral vector, such as a lentiviral vector. In another embodiment, a desirable vector is an adenoviral vector. In still another embodiment, a suitable vector is an adeno-associated viral vector. Adeno, adeno- associated and lentiviruses are generally preferred because they infect actively dividing as well as resting and differentiated cells such as the stem cells, macrophages and neurons. A variety of adenovirus, lentivirus and AAV strains are available from the American Type Culture Collection, Manassas, Virginia, or available by request from a variety of commercial and institutional sources. Further, the sequences of many such strains are available from a variety of databases including, e.g., PubMed and GenBank.
In one embodiment, a lentiviral vector is used. Among useful vectors are the equine infectious anemia virus and feline as well as bovine immunodeficiency virus, and HIV-based vectors. A variety of useful lentivirus vectors, as well as the methods and manipulations for generating such vectors for use in transducing cells and expressing heterologous genes, e.g., N Manjunath et al, 2009 Adv Drug Deliv Rev., 61(9): 732-745; Porter et al, N Engl J Med. 2011 Aug 25;365(8):725-33), among others.
In another embodiment, the vector used herein is an adenovirus vector. Such vectors can be constructed using adenovirus DNA of one or more of any of the known adenovirus serotypes. See, e.g., T. Shenk et al, Adenoviridae: The Viruses and their
Replication", Ch. 67, in FIELD'S VIROLOGY, 6th Ed., edited by B.N Fields et al, (Lippincott Raven Publishers, Philadelphia, 1996), p. 111-2112; 6,083,716, which describes the genome of two chimpanzee adenoviruses; US Patent No. 7,247,472; WO 2005/1071093, etc. One of skill in the art can readily construct a suitable adenovirus vector to carry and express a nucleotide sequence encoding a desired BRAFV6OOE and/or T helper peptide or fusion peptides as described herein. In another embodiment, the vector used herein is an adeno-associated virus (AAV) vector. Such vectors can be constructed using AAV DNA of one or more of the known AAV serotypes. See, e.g., US Patent No. 7,803,611 ; US Patent No. 7,696,179, among others.
In yet another embodiment, the vector used herein is a bacterial vector. In one embodiment, the bacterial vector is Listeria monocytogenes. See, e.g., Lauer et al, Infect. Immunity, 76(8):3742-53 (Aug. 2008). Thus, in one embodiment, the bacterial vector is live-attenuated or photochemically inactivated. The BRAFV6OOE and/or T helper peptide or fusion peptides as described herein can be expressed recombinantly by the bacteria, e.g., via a plasmid introduced into the bacteria, or integrated into the bacterial genome, i.e., via homologous recombination.
These vectors also include conventional control elements that permit transcription, translation and/or expression of the nucleic acid sequences in a cell transfected with the plasmid vector or infected with the viral vector. A great number of expression control sequences, including promoters which are native, constitutive, inducible and/or tissue- specific, are known in the art and may be utilized. In one embodiment, the promoter is selected based on the chosen vector. In another embodiment, when the vector is lentivirus, the promoter is U6, HI, CMV IE gene, EF-la, ubiquitin C, or phosphoglycero-kinase (PGK) promoter. In another embodiment, when the vector is an AAV, the promoter is an RSV, U6, or CMV promoter. In another embodiment, when the vector is an adenovirus, the promoter is RSV, U6, CMV, or HI promoters. In another embodiment, when the vector is Listeria monocytogenes, the promoter is a hly or actA promoter. Still other conventional expression control sequences include selectable markers or reporter genes, which may include sequences encoding geneticin, hygromicin, ampicillin or purimycin resistance, among others. Other components of the vector may include an origin of replication. Selection of these and other promoters and vector elements are conventional and many such sequences are available (see, e.g., the references cited herein).
The nucleic acid sequences encoding the BRAFV6OOE and/or T helper sequences for use in DNA vaccines are generated using the techniques and sequences provided herein, in conjunction with techniques known to those of skill in the art. Such techniques include conventional cloning techniques of cDNA such as those described in texts (Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring
Harbor, NY), use of overlapping oligonucleotide sequences, polymerase chain reaction, the various known Crispr-Cas methodologies, and any suitable method which provides the desired nucleotide sequence.
Thus, in one embodiment, using the information taught herein and publically available and known vector construction components and techniques, one of skill in the art can construct a viral vector (or plasmid) that expresses the desired nucleic acid sequence encoding the BRAFV6OOE and/or T helper sequences. The BRAFV6OOE and/or T helper polypeptides or proteins or fusion polypeptides or proteins encoded by these nucleic acid constructs may be expressed in vitro, or ex vivo in host cells or expressed in vivo by administration to a mammalian subject. Alternatively the BRAFV6OOE and/or T helper polypeptides or proteins or fusion polypeptides may be generated synthetically by known chemical synthesis methodologies. One of skill in the art can select the appropriate method to produce these BRAFV6OOE and/or T helper polypeptides or proteins or fusion polypeptides depending upon the components, the efficiency of the methodologies and the intended use, e.g., whether for administration as proteins, nucleic acids or in adoptive T cells, or otherwise to accomplish the desired therapeutic result.
Such nucleic acid compositions may include formulations with suitable vehicles for direct DNA, plasmid nucleic acid, or recombinant vector administration. Such vehicles include, without limitation, saline, sucrose, protamine, polybrene, polylysine, poly cations, proteins, or spermidine, etc. See e.g, International Patent Publication No. WO94/01139. Other pharmaceutically acceptable vehicles, excipients and typical components of DNA compositions are optionally included in these compositions.
In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In still another embodiment, a composition as described above is provided containing one or more of the BRAFV600E-based peptides, polypeptides or fusions or nucleic acid sequences encoding same, including DNA vaccines, in a pharmaceutically acceptable carrier, adjuvant or diluent or a combination thereof. This composition can contain other pharmaceutically acceptable components for enhancing the penetration of the compound into a cell and/or for extending its bioavailability and increasing its resistance to enzymatic degradation in vivo. Such a composition, in one embodiment, is a pharmaceutical composition. Such a composition in another embodiment is immunogenic.
The pharmaceutical compositions, both peptide or nucleic acid compositions, may also be formulated to suit a selected route of administration, and may contain ingredients specific to the route of administration as known to one of skill in the art of pharmaceutical formulation. A non-exclusive list of auxiliary agents are lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like that do not deleteriously react with the active compounds. The preparation of these pharmaceutically acceptable compositions, from the above-described components, having appropriate pH isotonicity, stability and other conventional characteristics is within the skill of the art.
In still another embodiment, a composition useful in the treatment of melanoma or other cancers comprises a recombinant dendritic cell pretreated ex vivo with any of the BRAFv600E-based peptides of Formula I, II, III or IV, or polypeptides comprising two or more copies of the BRAFV6ooE-based peptides or fusions with T helper sequences described above. In still another embodiment, the composition further contains an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor. Among such checkpoint inhibitors are CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
In still another embodiment, useful in the treatment of melanoma or other cancers includes a modified T cell is a T cell that has been transduced or transfected with one of the above-described vectors carrying the nucleic acid constructs encoding the BRAFV6OOE- based peptides, polypeptides or fusions or nucleic acid sequences encoding same.
Desirably, the T cell is a primary T cell, a CD8 (cytotoxic) T cell, or an NK T cell or other T cell obtained from the same mammalian subject into whom the modified T cell is administered or from another member of the mammalian species. In one embodiment, the T cell is an autologous human T cell or natural killer (NK) T cell obtained from the subject or from a bone marrow transplant match for the subject. Other suitable T cells include T cells obtained from resected tumors, a polyclonal or monoclonal tumor-reactive T cell. The T cell is generally obtained by apheresis, and transfected or transduced with the selected nucleic acid construct to express the BRAFV6OOE peptide or fusion with a T helper protein in vivo.
Methods of Treatment
A method of treating or retarding or preventing the development of melanoma in a mammalian subject comprises administering to a subject in need thereof, i.e., a subject with melanoma, a composition of any described above. The method is useful in the treatment of melanomas to induce an MHC class I, HLA-A2 restricted CTL response by the patient against the cancer. These therapeutic compositions and components of the methods may be administered to a patient, preferably suspended in a biologically compatible solution or pharmaceutically acceptable delivery vehicle. The various components of the methods are prepared for administration by being suspended or dissolved in a pharmaceutically or physiologically acceptable carrier such as isotonic saline; isotonic salts solution or other formulations that will be apparent to those skilled in such administration. The appropriate carrier will be evident to those skilled in the art and will depend in large part upon the route of administration. Other aqueous and non-aqueous isotonic sterile injection solutions and aqueous and non-aqueous sterile suspensions known to be pharmaceutically acceptable carriers and well known to those of skill in the art may be employed for this purpose.
A pharmaceutical composition as described above may be administered by any appropriate route, such as subcutaneous injection for the peptide compositions or intravenous injection for the checkpoint inhibitor antibody and infusion of the dendritic cells. Still other routes of administration as described above may be selected depending upon the dosage, patient condition, cancer type and stage, and other clinical factors.
In another embodiment, a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprises administering to said subject a BRAFV6ooE-based modified peptide or a pharmaceutically acceptable salt thereof and at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor.
In another embodiment, a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprising administering to said subject a nucleic acid sequence encoding a BRAFV6ooE-based modified peptide and at least one of (a) a nucleic acid sequence encoding a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor.
In still a further embodiment, a method of treating or retarding or preventing the development of melanoma in a mammalian subject comprising administering to said subject a dendritic cell pretreated with a BRAFV6ooE-based modified peptide or nucleic acid sequence encoding said peptide with at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor. In another embodiment, the dendritic cell is pretreated with the T helper peptide, a fusion of the BRAFV6ooE-based modified peptide with the T helper peptide, or a peptide spanning the T helper and CTL peptides of BRAFV6OOE as described above. In yet another embodiment, the dendritic cell is pretreated or transfected with nucleic acid molecules encoding these peptides.
A method of treating or preventing the development of a melanoma involves administering to a mammalian subject, preferably a human, an effective amount of a pharmaceutical composition described herein. The amount of the protein, peptide or nucleic acid sequences present in each effective dose is selected with regard to consideration to the half-life of the compound, the identity and/or stage of the melanoma, the patient's age, weight, sex, general physical condition and the like. The amount of active component required to induce an effective CTL response against the melanoma cells without significant adverse side effects varies depending upon the pharmaceutical composition employed and the optional presence of other components. Generally, for the compositions containing protein/peptide, or fusion protein, each dose will comprise between about 5 μg peptide/kg patient body weight to about 10 mg/kg. Generally, a useful therapeutic dosage is between 1 to 5 mg peptide/kg body weight. Another embodiment of a useful dosage may be about 500μg/kg of peptide. In one embodiment, the composition is administered at a concentration of from and including 25 to 250 micromoles of the peptide or nucleic acid sequence encoding the peptide.
In one embodiment a suitable concentration of the dose of the BRAFV6ooE-based peptide, polypeptide, fusion with T helper or nucleic acid sequences encoding same is administered in at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, to at least about 500 μΜ. Similarly intervening concentration between any two numbers listed is encompassed in the term "suitable concentration. Other dosage ranges may also be contemplated by one of skill in the art. For example, dosages of the peptides may be similar to the dosages discussed for other peptide cancer therapeutics.
If the composition is administered as an antibody or other protein, the dosages may range between a unit dosage of between 0.01 mg to 100 mg of protein (which is equivalent to about 12.5 μg/kg body weight). If any of the compositions are administered as naked DNA, the dosages may range from about 50 μg to about 1 mg of DNA per mL of a sterile solution. If the compositions is administered as a DNA vaccine in a virus, a therapeutically effective adult human or veterinary dosage of a viral vector is generally in the range of from about 100 to about 100 mL of a carrier containing concentrations of from about 1 x 106 to about 1 x 1015 particles, about 1 x 1011 to 1 x 1013 particles, or about 1 x 109 to lx
1012 particles virus. The dosage of the checkpoint inhibitor may be adjusted based on known toxicities of the particular antibody or small molecule used.
In one embodiment, the selected composition is administered in a single dose. In another embodiment an initial dose of a composition may be optionally followed by repeated administration for a duration selected by the attending physician. In one embodiment, one to three booster doses are administered. Dosage frequency may also depend upon the factors identified above, and may range from 1 to 6 doses per day for a duration of about 3 days to a maximum of no more than about 1 week. The compositions may also be administered as a continuous infusion for about 3-5 days, the specific dosage of the infusion depending upon the half-life of the compound. The compounds may also be incorporated into chemotherapy protocols, involving repetitive cycles of dosing. Selection of the appropriate dosing method would be made by the attending physician.
In another embodiment, the peptide or nucleic acid compositions and checkpoint inhibitors are independently administered systemically by intramuscular, intraperitoneal, intravenous, intratumoral, oral or intranodal administration. In still further embodiments, the subject is treated with other anti-cancer therapies before, during or after treatment with said composition. For example, the subject can be treated with chemotherapy before administering said composition.
In another embodiment, this method involving co-administering the
immunotherapeutic compositions with the pretreated dendritic cell can also include administering a checkpoint inhibitor in the form of an antibody or a small molecule.
In one embodiment described herein, the immunotherapeutic BRAFV6OOE -based peptide, polypeptide or fusion compositions and T helper peptide or nucleic acid compositions encoding them, or dendritic cell pretreated with them are administered substantially simultaneously. In another embodiment, these immunotherapeutic compositions and T helper sequences, optionally with the checkpoint inhibitor, are administered sequentially by the same or different routes of administration. The routes of administration selected depend upon the nature of the compositions. For example, if the checkpoint inhibitor is a small chemical molecule, such molecules may be administered orally in doses known and accepted for other pharmaceutical uses of these drugs. The other compositions or components of the method are administered in different routes as mentioned above.
In still further aspects of these methods, the subject may be treated with other anticancer therapies before, during or after treatment with the compositions or individual components of the compositions and methods above-described. Such treatment may be concurrent or simultaneous with the BRAFV6OOE -based peptide or nucleic acid compositions or dendritic cell compositions or overlap treatment with any of these components. In one embodiment, the methods involve treating the subject with chemotherapy before administering the immunotherapeutic compositions, T helper sequences and/or checkpoint inhibitor antibodies or small molecule mimics. In still another embodiment, the method further comprises depleting the subject of lymphocytes and optionally surgically resecting the tumor prior to administration of the components of the above-described methods.
Other dosages are taught in the references recited herein and can be readily adjusted by one of skill in the art depending upon the treatment regimen, physical condition of the patient, type and stage and location of the tumor being treated, and taking into consideration other ancillary chemotherapies being used to treat the patient.
The following examples are provided for the purpose of illustration only and the invention should in no way be construed as being limited to these examples but rather should be construed to encompass any and all variations that become evident as a result of the teaching provided herein.
EXAMPLE 1 : T Helper Sequences
Amino acid sequences upstream and downstream of BRAFV6OOE mutation were screened for T-helper sequences (Th) that bind to HLA-DR alleles using computer algorithms. Table I illustrates a number of sequences tested. Table 1 : Human Modified BRAFV6OOE Peptides Predicted to Bind to HLA-A2
Figure imgf000032_0001
XFI- (MFI peptide MFI w/o eptide)" !
A number of such peptides were assayed as follows: Adherent monocytes (5 x 104/well) were pulsed for 8 hours with synthetic peptides (25 μΜ): Peptide 8 SEQ ID NO: 15, Peptide 9 SEQ ID NO: 16, and Peptide 10 SEQ ID NO: 17. At the end of incubation, excess peptides were removed and the monocytes were cultured with one of three melanoma patient PBMC samples labeled WM35, WM3457 and 3451 (1 x 105) for 5 days.
Proliferative responses were determined by standard HTdR incorporation assay and the results shown in FIG. 1. Data are expressed as mean cpm (triplicate
determinations) plus SD (bar) of HTdR incorporation. Significantly (p<0.01) different values compared to control were obtained in this assay with either Peptide 8 or Peptide 9 in each sample assayed. Thus, T helper peptides were shown to induce proliferative response in melanoma patients PBMCs. EXAMPLE 2: Induction of Cytotoxic T Cells
Adherent monocytes (5 x 104/well) were pulsed for 8 h with T-helper synthetic peptides (25 μΜ) either alone or in combination with an affinity modified BRAFV6OOE- based peptide, i.e., a CTL peptide of the sequence LMTEKSRWSG; SEQ ID NO: 25. Cell cultures were initiated as described in Example 1 and at the end of 7 days T-cells were harvested and stained with fluorescence conjugated anti-CD4 or anti-CD8 antibodies and analyzed for T-cell binding by standard FACS assay.
As shown in FIG. 2, the Th peptides induced enriched CD8 CTLs in the presence of both the Th peptide and the BRAFV6ooE-based CTL peptide.
EXAMPLE 3: Induction of More Potent CTLs
T-cell cultures were initiated as described in Examples 1 and 2. After 7 days, growing lymphocyte cultures were harvested and restimulated with Th peptide 9 of sequence EDLTVKIGDFGLATV SEQ ID NO: 16, the BRAFV6ooE-based peptide of SEQ ID NO: 25, or both, and 20 U/ml of natural human IL-2. This process was repeated every
7 days until day 56 when lymphocytes were harvested and tested for cytolytic activity against HLA-A2+ autologous WM35 (V600E+) or HLA-A2+ matched allogeneic WM3456 (V600E-) melanoma cells in standard 51Cr-release assay. Varied effector (T- cells) to tumor target ratios of 12.5: 1 or 25: 1 or 50: 1 were used in the assay.
As shown in FIG. 3, a combination of the T helper and BRAFV600E-based CTL peptide induces more potent CTLs than either the Th or CTL peptides alone.
EXAMPLE 4: Regression Of Established Tumor
In an established tumor model, C57B1/6 (4-6 weeks old; n=5; male) mice received s.c. injection of mouse melanoma cell line (YUMM 1.7; lxlO5). After 7 days the tumor was established. Mice received BRAFV600E-based peptide SEQ ID NO: 25 immunization (50 μg, intradermally, every 7 days) in presence of CpG (30 μg) as adjuvant. Mice also received anti-PDl antibody (250 μg via intraperitoneal injection, every 5-6 days) either alone or in combination.
FIG. 4 shows tumor growth measurements as mean tumor volume and SEM. The results are compared with mice that received control HIV peptide and anti-PDl antibody. Combination of peptide immunization and anti-PDl antibody therapy showed significant inhibition of tumor growth (p<0.05). Anti-PDl antibody alone was not effective in tumor inhibition when compared to isotype control antibody treatment. FIG. 4 demonstrates that a combination of immunization with a BRAFV600E-based CTL peptide and treatment with a checkpoint inhibitor, i.e., anti-PDl antibody, has significant effect on growth of mouse melanoma (YUMM 1.7) cells in an established tumor model and induces regression of established tumor.
TABLE 2 (Sequence Listing Free Text)
The following information is provided for sequences containing free text under numeric identifier <223>.
SEQ ID NO: Free text under <223>
(containing free
text)
2 <210> 2
<211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
3 <210> 3
<211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Ser, Gly or Cys
<220>
<221> MISC FEATURE
<222> (4)..(4) SEQ ID NO: Free text under <223>
(containing free
text)
<223> Xaa is a substituted or unsubstituted Ala, Leu, Met, Val,
Pro or Gly
<220>
<221> MISC FEATURE
<222> (5).. (5)
<223> Xaa is a substituted or unsubstituted Thr or Ser
<220>
<221> MISC FEATURE
<222> (7).. (7)
<223> Xaa is a substituted or unsubstituted Lys, Arg or His
<220>
<221> MISC FEATURE
<222> (8).. (8)
<223> Xaa is a substituted or unsubstituted Ser or Thr
<220>
<221> MISC FEATURE
<222> (9).. (9)
<223> Xaa is a substituted or unsubstituted Arg, Lys, or His
<220>
<221> MISC FEATURE
<222> (11).. (11)
<223> Xaa is substituted or unsubstituted Thr, Val, Leu or Ser
<220>
<221> MISC FEATURE
<222> (12).. (12)
<223> Xaa is absent or is a substituted or unsubstituted Gly, Pro or Leu
<220>
<221> MISC FEATURE SEQ ID NO: Free text under <223>
(containing free
text)
<222> (13).. (14)
<223> Xaa is absent or is a Cys, Ser or Gly
4 <210> 4
<211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is Ser, Gly, Cys or absent
<220>
<221> MISC FEATURE
<222> (4).. (4)
<223> Xaa is a substituted or unsubstituted He
<220>
<221> MISC FEATURE
<222> (12).. (12)
<223> Xaa is a substituted or unsubstituted Gly, Ala, Val, Leu,
He, Pro, Phe, Met or Trp
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is Ser, Gly, Cys or absent
5 <211> 14
<212> PRT
<213> Artificial Sequence
<220> SEQ ID NO: Free text under <223>
(containing free
text)
<223> Synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (4).. (4)
<223> Xaa is a substituted or unsubstituted Gly, Ala, Val, Leu,
He, Pro, Phe, Met or Trp
<220>
<221> MISC FEATURE
<222> (12).. (12)
<223> Xaa is a substituted or unsubstituted Gly, Val or Leu
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent or a Cys, Gly or Ser
6 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent or Gly, Ser or Cys
<220>
<221> MISC FEATURE SEQ ID NO: Free text under <223>
(containing free
text)
<222> (4).. (4)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, or
Leu,
<220>
<221> MISC FEATURE
<222> (12).. (12)
<223> Xaa is a substituted or unsubstituted Gly, He, Val or Leu.
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
7 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
8 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide SEQ ID NO: Free text under <223>
(containing free
text)
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
9 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
10 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2) SEQ ID NO: Free text under <223>
(containing free
text)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
11 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
12 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE SEQ ID NO: Free text under <223>
(containing free
text)
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
13 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
14 <211> 14
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (1)..(2)
<223> Xaa is absent, Gly, Ser or Cys
<220>
<221> MISC FEATURE
<222> (13).. (14)
<223> Xaa is absent, Gly, Ser or Cys
18 <211> 21 SEQ ID NO: Free text under <223>
(containing free
text)
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (13).. (13)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
Gly, Val, Pro, Phe, or Trp
<220>
<221> MISC FEATURE
<222> (21).. (21)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, Leu, Gly, Val, Pro, Phe, or Trp
19 <211> 20
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (12).. (12)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
Gly, Val, Pro, Phe, or Trp
<220>
<221> MISC FEATURE
<222> (20).. (20)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, Leu, Gly, Val, Pro, Phe, or Trp SEQ ID NO: Free text under <223>
(containing free
text)
20 211> 26
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
<220>
<221> MISC FEATURE
<222> (2).. (2)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
Gly, Val, Pro, Phe, or Trp
<220>
<221> MISC FEATURE
<222> (10).. (10)
<223> Xaa is a substituted or unsubstituted Ala, He, Met, Leu,
Gly, Val, Pro, Phe, or Trp
22 <211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
23 <211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
24 <211> 10
<212> PRT
<213> Artificial Sequence
<220> SEQ ID NO: Free text under <223> (containing free
text)
<223> synthetic peptide
25 <211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
26 <211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
27 <211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
28 <211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
29 <211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
30 <211> 11
<212> PRT
<213> Artificial Sequence SEQ ID NO: Free text under <223>
(containing free
text)
<220>
<223> synthetic peptide
31 211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
32 <211> 26
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
33 <211> 20
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
34 <211> 19
<212> PRT
<213> Artificial Sequence
<220>
<223> synthetic peptide
Each and every patent, patent application, including US Provisional Patent Application No. 62/296,705, and publication, including NCBI sequences, the sequence listing and websites cited throughout the disclosure and listed below, is expressly incorporated herein by reference in its entirety. All publicly available documents and public databases and publicly available DNA and nucleic acid sequences cited within this specification are incorporated herein by reference. The claims and the sequence listing are incorporated herein by reference. While this invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention are devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims include such embodiments and equivalent variations.

Claims

CLAIMS:
1. A composition comprising a peptide or a pharmaceutically acceptable salt thereof, the peptide having the formula
(a) Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-R2 SEQ ID NO: 4, wherein Rl and R2 are independently absent, Ser, Gly, or Cys for coupling to an additional peptide or protein, and wherein AA2 is a substituted or unsubstituted He and AAIO is a hydrophobic residue; or
(b) Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-R2 SEQ ID NO: 5, wherein Rl and R2 are independently absent, Ser, Gly, or Cys for coupling to an additional peptide or protein, and wherein AA2 is a hydrophobic residue and AAIO is a substituted or unsubstituted Gly, Val or Leu; or
(c) Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-R2 SEQ ID NO: 6, wherein Rl and R2 are independently absent, Ser, Gly, or Cys for coupling to an additional peptide or protein, and wherein AA2 is a substituted or unsubstituted He, Ala, Met or Leu, and wherein AAIO is a substituted or unsubstituted Gly, He, Val or Leu.
2. The composition according to claim 1, further comprising a pharmaceutically acceptable vehicle or carrier.
3. The composition according to claim 1(a), wherein AAIO is a substituted or unsubstituted Gly, Val or Leu.
4. The composition according to claim 1(b), wherein AA2 is a substituted or unsubstituted He, Ala, Met or Leu.
5. The composition according to claim 1, wherein said peptide is
Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 7);
Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO:33);
Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 9);
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Ile-R2 (SEQ ID NO: 10);
Rl-Leu-Met-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 11) or
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 12).
6. The composition according to claim 1, wherein one or more of said amino acids in said formula are selected from the group consisting of a D- amino acid, an amino acid containing a substitution for the alpha-carbon in the amino acid structure, an amino acid containing a beta-carbon in its amino acid structure, an amide substituted amino acid, an amino acid substituted with a straight chain, branched, cyclic or heterocyclic Cl-12 alkyl group, a straight chain, branched, cyclic, or heterocyclic Cl-12 alkanoyl group.
7. The composition according to claim 1, where said Rl or R2 is Cys.
8. The composition according to claim 1, further comprising a T helper peptide.
9. The composition according to claim 8, wherein the T cell peptide is coupled or fused at either the N- or C- termini of the peptide of (a), (b) or (c).
10. The composition according to claim 8, wherein said T helper peptide is
(a) Ser-His-Gln-Phe-Glu-Gln-Leu-Ser-Gly-Ser-Ile-Leu-Trp-His-Ala SEQ ID NO: 15;
(b) Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val SEQ ID NO: 16; or
(c) Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val-Lys SEQ ID NO: 17.
11. The composition according to claim 1, further comprising multiple copies of the same peptide (a), (b) or (c) fused or coupled together.
12. The composition according to claim 9, further comprising multiple copies of a fused peptide, which comprises the peptide (a), (b) or (c) fused or coupled to a T helper peptide.
The composition according to claim 11, comprising a polypeptide formed from 5 10 copies of the same peptide (a), (b) or (c) fused in frame or via a spacer.
14. The composition according to claim 12, comprising a polypeptide formed from 5 to 10 copies of the same fused peptide, each fused peptide in frame or fused via a spacer.
15. The composition according to claim 1, further comprising a liposome.
16. The composition according to claim 1, further comprising an adjuvant.
17. The composition according to claim 16, further wherein said adjuvant is CPG or encapsulated PLG.
18. A composition comprising a nucleic acid sequence encoding:
(a) Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-R2 SEQ ID NO: 4, wherein Rl and R2 are independently absent, Ser, Gly, or Cys for coupling to an additional peptide or protein, and wherein AA2 is a substituted or unsubstituted He and AAIO is a hydrophobic residue; or
(b) Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-R2 SEQ ID NO: 5, wherein Rl and R2 are independently absent, Ser, Gly, or Cys for coupling to an additional peptide or protein, and wherein AA2 is a hydrophobic residue and AAIO is a substituted or unsubstituted Gly, Val or Leu;
(c) Rl-Leu-AA2-Thr-Glu-Lys-Ser-Arg-Trp-Ser-AA10-R2 SEQ ID NO: 6, wherein Rl and R2 are independently absent, Ser, Gly, or Cys for coupling to an additional peptide or protein, and wherein AA2 is a substituted or unsubstituted He, Ala, Met or Leu, and wherein AAIO is a substituted or unsubstituted Gly, He, Val or Leu.
(d) a peptide or polypeptide encoding a fusion peptide comprising the peptide of (a), (b) or (c) fused to a T cell peptide at either the N- or C- termini; or
(e) a peptide or polypeptide encoding a fusion peptide comprising at least two copies of the same peptide of (a), (b), (c) or (d).
19. The composition according to claim 18, further comprising a pharmaceutically acceptable vehicle or carrier.
20. The composition according to claim 18(a), wherein AA10 is a substituted or unsubstituted Gly, Val or Leu.
21. The composition according to claim 18(b), wherein AA2 is a substituted or unsubstituted He, Ala, Met or Leu.
22. The composition according to claim 18, wherein said encoded peptide (a) or (b) is Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 7);
Rl-Leu-Ile-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 8);
Rl-Leu-Leu-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 9);
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Ile-R2 (SEQ ID NO: 10);
Rl-Leu-Met-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Gly-R2 (SEQ ID NO: 11) or
Rl-Leu-Ala-Thr-Glu-Lys-Ser-Arg-Trp-Ser-Val-R2 (SEQ ID NO: 12).
23. The composition according to claim 18, where said Rl or R2 is Cys.
24. The composition according to claim 18, further comprising a nucleic acid sequence encoding a T helper peptide.
25. The composition according to claim 18 or 24, wherein said T helper peptide is
(a) Ser-His-Gln-Phe-Glu-Gln-Leu- Ser-Gly-Ser-Ile-Leu-Trp-His-Ala SEQ ID NO: 15;
(b) Glu-Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val SEQ ID NO: 16; or
(c) Asp-Leu-Thr-Val-Lys-Ile-Gly-Asp-Phe-Gly-Leu-Ala-Thr-Val-Lys SEQ ID NO: 17.
26. The composition according to claim 18(e), wherein said nucleic acid sequence encodes a polypeptide formed from 5 to 10 of the same peptide or fused peptide via a spacer.
27. The composition according to claim 1, further comprising an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor.
28. The composition according to claim 18, further comprising an antibody that binds a checkpoint inhibitor or a molecule that mimics the function of a checkpoint inhibitor or a nucleic acid sequence encoding the inhibitor or molecule.
29. The composition according to any of claim 27 to 28, wherein the checkpoint inhibitor is CTLA4, PD1, PDL1, LAG3, ICOS, BTLA, KIR, OX40, CD27, CD40L, CD40, TIM3 or TIM334.
30. A composition comprising any one of
(a) a BRAFV6ooE-based modified peptide or a pharmaceutically acceptable salt thereof and at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor;
(b) a nucleic acid sequence encoding a BRAFV6ooE-based modified peptide and at least one of (a) a nucleic acid sequence encoding a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor;
(c) a dendritic cell pretreated with a BRAFV6ooE-based modified peptide or nucleic acid sequence encoding said peptide with at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor; or
(d) a composition of any of claims 1 to 29
for treating or retarding or preventing the development of melanoma in a mammalian subject.
31. A method of treating or retarding or preventing the development of melanoma in a mammalian subject comprising any one of:
(a) administering to said subject a BRAFV6ooE-based modified peptide or a pharmaceutically acceptable salt thereof and at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor;
(b) administering to said subject a nucleic acid sequence encoding a
BRAFV6ooE-based modified peptide and at least one of (a) a nucleic acid sequence encoding a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor;
(c) administering to said subject a dendritic cell pretreated with a BRAFV6OOE- based modified peptide or nucleic acid sequence encoding said peptide with at least one of (a) a T helper peptide and (b) an antibody that binds a checkpoint inhibitor or a synthetic small molecule that mimics the function of a checkpoint inhibitor; or
(d) administering to said subject a composition of any of claims 1 to 29;
32. The method according to claim 31, wherein the composition is administered in a single dose.
33. The method according to claim 31, wherein the composition is administered at a concentration of from and including 25 to 250 micromoles of the peptide or nucleic acid sequence encoding a peptide of claim 1.
34. The method according to claim 33, wherein said composition and checkpoint inhibitors are independently administered systemically by intramuscular, intraperitoneal, intravenous, intratumoral, oral or intranodal administration.
35. The method according to claim 33, further comprising treating the subject with other anti-cancer therapies before, during or after treatment with said composition or treating the subject with chemotherapy before administering said composition.
36. A composition comprising a recombinant dendritic cell pretreated ex vivo with a composition of any of claims 1 through 29.
PCT/US2017/018051 2016-02-18 2017-02-16 Methods and compositions for treating melanoma WO2017142988A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP17753785.9A EP3416668A4 (en) 2016-02-18 2017-02-16 Methods and compositions for treating melanoma
CN201780024075.5A CN109069574A (en) 2016-02-18 2017-02-16 For treating the method and composition of melanoma
US15/998,913 US11208468B2 (en) 2016-02-18 2017-02-16 Methods and compositions for treating melanoma
US17/533,095 US20220177548A1 (en) 2016-02-18 2021-11-22 Methods and Compositions for Treating Melanoma

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662296705P 2016-02-18 2016-02-18
US62/296,705 2016-02-18

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/998,913 A-371-Of-International US11208468B2 (en) 2016-02-18 2017-02-16 Methods and compositions for treating melanoma
US17/533,095 Division US20220177548A1 (en) 2016-02-18 2021-11-22 Methods and Compositions for Treating Melanoma

Publications (1)

Publication Number Publication Date
WO2017142988A1 true WO2017142988A1 (en) 2017-08-24

Family

ID=59626214

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/018051 WO2017142988A1 (en) 2016-02-18 2017-02-16 Methods and compositions for treating melanoma

Country Status (4)

Country Link
US (2) US11208468B2 (en)
EP (1) EP3416668A4 (en)
CN (1) CN109069574A (en)
WO (1) WO2017142988A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007002811A2 (en) * 2005-06-29 2007-01-04 The Wistar Institute Of Anatomy And Biology Methods and compositions for treating melanoma
WO2012044999A2 (en) * 2010-10-01 2012-04-05 Ludwig Institute For Cancer Research Ltd. Reversible protein multimers, methods for their production and use

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU684050B2 (en) 1992-07-13 1997-12-04 Baylor College Of Medicine Targeting somatic gene therapy to joints
AU2992295A (en) 1994-07-13 1996-02-16 Auckland Uniservices Limited Therapeutic method and compounds of use therein
US6083716A (en) 1996-09-06 2000-07-04 The Trustees Of The University Of Pennsylvania Chimpanzee adenovirus vectors
CA2466431C (en) 2001-11-21 2014-08-05 The Trustees Of The University Of Pennsylvania Simian adenovirus nucleic acid and amino acid sequences, vectors containing same, and methods of use
EP2000160A3 (en) 2002-10-30 2009-03-11 Gambro Lundia AB Method and apparatuses for determining the efficiency of dialysis
CA2553541C (en) 2004-01-23 2015-04-21 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Chimpanzee adenovirus vaccine carriers
US7803611B2 (en) 2005-02-03 2010-09-28 Benitec, Inc. RNAi expression constructs
CA2882292C (en) 2012-08-13 2023-10-17 The Rockefeller University Treatment and diagnosis of melanoma
KR102303166B1 (en) 2012-09-14 2021-09-17 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 T cell receptors recognizing mhc class ii-restricted mage-a3
FR3009959B1 (en) 2013-08-29 2018-05-18 L'oreal COSMETIC COMPOSITION COMPRISING AT LEAST ONE ACRYLIC AND / OR METHACRYLIC THICKENING POLYMER, AT LEAST ONE FIXING POLYMER AND AT LEAST ONE ANTI-SEBORRHEIC AGENT

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007002811A2 (en) * 2005-06-29 2007-01-04 The Wistar Institute Of Anatomy And Biology Methods and compositions for treating melanoma
WO2012044999A2 (en) * 2010-10-01 2012-04-05 Ludwig Institute For Cancer Research Ltd. Reversible protein multimers, methods for their production and use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3416668A4 *

Also Published As

Publication number Publication date
CN109069574A (en) 2018-12-21
EP3416668A1 (en) 2018-12-26
US20220177548A1 (en) 2022-06-09
US11208468B2 (en) 2021-12-28
US20190276517A1 (en) 2019-09-12
EP3416668A4 (en) 2020-02-19

Similar Documents

Publication Publication Date Title
US20210179735A1 (en) Trispecific binding proteins and methods of use
JP7055794B2 (en) Claudin-expressing bispecific trivalent antibody that binds to Claudin 6 or Claudin 18.2 and CD3 for the treatment of cancer diseases
US20200002387A1 (en) Cd20-binding immunotoxins for inducing cellular internalization and methods using same
US20210052727A1 (en) Cytokine fusion proteins
CN106414500B (en) anti-WT 1/HLA bispecific antibodies
ES2676630T3 (en) Immunogenic control of tumors and tumor cells
US20200155702A1 (en) Engineered Antibody Compounds and Conjuates Thereof
EP3565898A1 (en) Vegfr-2 car immune cells to treat cancers
WO2020113403A1 (en) Cytokine fusion proteins
WO2018009528A1 (en) Combination cancer immunotherapies with arginine depletion agents
JP6218175B2 (en) Modification of helper T cell inducible polypeptide
US20190336600A1 (en) Methods for detecting and reversing immune therapy resistance
US20220177548A1 (en) Methods and Compositions for Treating Melanoma
CN115304680A (en) Preparation and application of bispecific cell adaptor molecule constructed based on Pep42
CN115702002A (en) Improved peptide vaccines
KR20230147078A (en) T cells for use in therapy
CN116813796A (en) IL8 antibody-based signaling receptors
CN116836301A (en) TGF-beta antibody-based trap receptor
CN117126294A (en) Signal transduction receptor based on TGFbeta antibody
KR20220038742A (en) Antigenic Polypeptides and Methods of Using the Same
EA045360B1 (en) METHODS AND COMPOSITIONS THAT INCREASE THE EFFECTIVENESS OF SUPERANTIGEN-MEDIATED IMMUNOTHERAPY OF MALIGNANT TUMORS

Legal Events

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

Ref document number: 17753785

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2017753785

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017753785

Country of ref document: EP

Effective date: 20180918