WO2009129498A2 - Substance p and analogs thereof as a cancer immunogenic composition adjuvant - Google Patents

Substance p and analogs thereof as a cancer immunogenic composition adjuvant Download PDF

Info

Publication number
WO2009129498A2
WO2009129498A2 PCT/US2009/041022 US2009041022W WO2009129498A2 WO 2009129498 A2 WO2009129498 A2 WO 2009129498A2 US 2009041022 W US2009041022 W US 2009041022W WO 2009129498 A2 WO2009129498 A2 WO 2009129498A2
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
substance
cancer
tumor
composition
Prior art date
Application number
PCT/US2009/041022
Other languages
French (fr)
Other versions
WO2009129498A3 (en
Inventor
Michael Klaus Wilhelm
Hal N. Siegel
Adriana T. Larregina
Adrian Eduardo Morelli
Original Assignee
Immuneregen Biosciences, Inc.
University Of Pittsburgh Of The Commonwealth System Of Higher Education
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 Immuneregen Biosciences, Inc., University Of Pittsburgh Of The Commonwealth System Of Higher Education filed Critical Immuneregen Biosciences, Inc.
Publication of WO2009129498A2 publication Critical patent/WO2009129498A2/en
Publication of WO2009129498A3 publication Critical patent/WO2009129498A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/00117Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/046Tachykinins, e.g. eledoisins, substance P; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001148Regulators of development
    • A61K39/001149Cell cycle regulated proteins, e.g. cyclin, CDC, CDK or INK-CCR
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001176Heat shock proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001182Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00119Melanoma antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464454Enzymes
    • A61K39/464456Tyrosinase or tyrosinase related proteinases [TRP-1 or TRP-2]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/46449Melanoma antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/876Skin, melanoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule

Definitions

  • the present invention is based on the discovery that a neuropeptide can modulate the immunogenicity of an antigen including, but not limited to, a cancer- associated antigen such as a cancer tumor antigen.
  • a cancer-associated antigen such as a cancer tumor antigen.
  • the neuropeptide Substance P and certain analogs thereof provide a heretofore unrealized opportunity to formulate and use compositions comprising a select antigen and Substance P (or certain analogs thereof) as an effective cancer vaccine, including a dendritic cell-based vaccine, a solid tumor- based vaccine, and a non-solid tumor-based vaccine.
  • a method for treating cancer in a subject comprises administering an immunogenic composition comprising a cancer tumor antigen and an adjuvant effective amount of a Substance P or an analog thereof.
  • a method for preventing cancer in a subject at risk for developing cancer is provided.
  • a method for reducing the likelihood of developing cancer in a subject at risk for developing cancer is provided.
  • the cancer to be treated can be a solid tumor, such as a solid adenocarcinoma or a lympho-hematopoietic cancer.
  • the cancer to be treated can be pancreatic cancer, breast cancer, gastric cancer, renal cell carcinoma, colorectal cancer, melanoma, prostate cancer, ovarian cancer, lymphoma, non-Hodgkin's lymphoma, or leukemia.
  • the cancer to be prevented can be cervical, liver, breast, pancreas, colon, lung, ovarian or prostate cancer.
  • the antigen in the immunogenic composition can be from any source or derived by any methodology known in the art.
  • the antigen can be a shared tumor antigen, unique tumor antigen, inactivated whole tumor cells, or gene -modified cells.
  • the antigen can be a dendritic cell based antigen.
  • the antigen can be mucin 1 , HER2/NEU, melanoma-associated antigen, mammaglobin, carcinoembryonic antigen, or cyclin Bl.
  • the composition of the invention is useful for treatment, prevention, or amelioration of cancer in a subject; or, for reducing the likelihood of developing cancer in a subject at risk for developing cancer.
  • the composition comprises an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog.
  • the Substance P analog is of Formula (I): Zi-Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ -Xaa 1 l -Z 2 (I)
  • Xaa 1 is Arg, Lys, 6-N methyllysine or (6-N, 6-N) dimethyllysine;
  • Xaa 2 is Pro or Ala
  • Xaa 3 is Lys, Arg, 6-N-methyllysine or (6-N, 6-N) dimethyllysine;
  • Xaa 4 is Pro or Ala
  • Xaa 5 is GIn or Asn
  • Xaa 6 is GIn or Asn
  • Xaa 7 is Phe or Phe substituted with chlorine at position 2, 3 or 4;
  • Xaa 8 is Tyr, Phe, or Phe substituted with chlorine at position 2, 3 or 4;
  • Xaa 9 is GIy, Pro, Ala or N-methylglycine
  • Xaa 10 is Leu, VaI, He, Norleucine, Met, Met sulfoxide, Met sulfone, N-methylleucine, or N-methylvaline;
  • Xaa 11 is Met, Met sulfoxide, Met sulfone, or Norleucine;
  • Zi is R 2 N- or RC(O)NR-;
  • Z 2 is -C(O)NR 2 or -C(O)OR or a salt thereof; each R is independently R is H, (Ci -C 6 ) alkyl, (Ci -C 6 ) alkenyl, (Ci -C 6 ) alkynyl,
  • the Substance P analog can be of Formula (I) as described herein, wherein Xaa 1 is Arg; Xaa 2 is Pro; Xaa 3 is Lys; Xaa 4 is Pro; Xaa 5 is GIn; Xaa 6 is GIn; Xaa 7 is Phe or Phe substituted with chlorine at position 4; Xaa 8 is Tyr, Phe, or Phe substituted with chlorine at position 4; Xaa 9 is GIy, Pro or N- methylglycine; Xaa 10 is Leu; and Xaa 11 is Met, Met sulfoxide, Met sulfone or Norleucine.
  • the Substance P analog can be of Formula (I) as described herein wherein the "— " between residues Xaal through Xaal 1 designates -C(O)NH-; Zl is H2N-; and Z2 is -C(O)NH2.
  • the Substance P analog can be: RPKPQQFFGLM (SEQ ID NO.: 1);
  • RPKPQQFFGLNIe SEQ ID NO. : 2
  • RPKPQQFFPLM SEQ ID NO. : 3
  • RPKPQQFFMeGIyLM SEQ ID NO. : 4
  • RPKPQQFTGLM (SEQ ID NO. : 5);
  • RPKPQQF(4-C1)F(4-C1)GLM (SEQ ID NO. : 6);
  • RPKPQQFFGLM(O) (SEQ ID NO. : 7); RPKPQQFFMeGIyLM(O) (SEQ ID NO.: 8);
  • RPKPQQFFGLM(O 2 ) (SEQ ID NO. : 9); or
  • RPKPQQFFMeGIyLM(O 2 ) (SEQ ID NO.: 10).
  • the Substance P analog can be Zi-RPKPQQFFMeGlyLM(O 2 )-Z2; wherein Zi is NH 2 and Z 2 is C(O)NH 2 (SEQ ID NO:12).
  • the antigen can be a dendritic cell-based antigen selected from the group consisting of: tumor- dendritic cell hybrids; peptide-loaded dendritic cells; whole tumor cell-loaded dendritic cells; tumor mRNA-loaded dendritic cells; and genetically engineered or gene-loaded dendritic cells.
  • the antigen is a dendritic cell-based antigen resulting from a dendritic cell loaded with an antigen from a tumor; a dendritic cell exposed to a tumor lysate; a dendritic cell gene-loaded ex vivo; or a dendritic cell exposed to a tumor antigen ex vivo.
  • a particularly preferred embodiment is a composition which is a dendritic cell vaccine.
  • a composition comprises an antigen which is a cancer tumor antigen.
  • Such a cancer tumor antigen is selected from the group consisting of: a solid adenocarcinoma, a pancreatic cancer, a breast cancer, a gastric cancer, a renal cell cancer, a colorectal cancer, a melanoma, a lympho -hematopoietic cancer, a lymphoma, a Non-Hodgkin's lymphoma; a leukemia; mucin 1 ; HER2/NEU; melanoma-associated antigen; mammaglobin; carcinoembryonic antigen; and cyclin B 1.
  • cancer tumor antigen selected from the group consisting of: autologous or allogenic tumor cell antigen; tumor cell lysate antigen; shed antigens from autologous tumor cells; heat shock proteins from autologous tumor cells; tumor-associated antigens (TAA); shared tumor antigen, unique tumor antigen, inactivated whole tumor cell antigen; and gene -modified cell antigen.
  • a particularly preferred embodiment is a composition which is a melanoma vaccine.
  • the antigen is the subject's endogenous antigen.
  • the invention contemplates a composition in which the antigen and the Substance P or Substance P analog are in combination. In yet other embodiments, they are separate. Other embodiments contemplate a configuration which accommodates a situation in which the antigen and the Substance P or the Substance P analog are for contemporaneous administration. Still other configurations accommodate a situation in which the antigen and the Substance P or the Substance P analog are in separate, divided or undivided containers. Still other configurations accommodate a situation in which the antigen and the Substance P or the Substance P analog are for sequential administration.
  • the amelioration of cancer by the composition of the invention comprises reducing tumor load, slowing development of tumors or reducing severity when compared to the antigen alone.
  • any of the compositions of the invention can be a pharmaceutical composition.
  • the present invention is directed to a method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer, comprising providing to said subject an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog wherein the Substance P analog is of Formula (I) as set out elsewhere herein.
  • any method disclosed herein contemplates further defining Formula (I) as follows iXaa 1 is Arg; Xaa 2 is Pro; Xaa 3 is Lys; Xaa 4 is Pro; Xaa 5 is GIn; Xaa 6 is GIn; Xaa 7 is Phe or Phe substituted with chlorine at position 4; Xaa 8 is Tyr, Phe, or Phe substituted with chlorine at position 4; Xaa 9 is GIy, Pro or N- methylglycine; Xaa 10 is Leu; and Xaa 11 is Met, Met sulfoxide, Met sulfone or Norleucine.
  • any method of the present invention contemplates one or more Substance P analogs is selected from the group consisting of: RPKPQQFFGLM (SEQ ID NO.: 1);
  • RPKPQQFFGLNIe SEQ ID NO. : 2
  • RPKPQQFFPLM (SEQ ID NO. : 3);
  • RPKPQQFFMeGIyLM SEQ ID NO. : 4
  • RPKPQQFTGLM SEQ ID NO.: 5
  • RPKPQQF(4-C1)F(4-C1)GLM (SEQ ID NO. : 6);
  • RPKPQQFFMeGIyLM(O) (SEQ ID NO. : 8);
  • RPKPQQFFGLM(O 2 ) (SEQ ID NO. : 9); or RPKPQQFFMeGIyLM(O 2 ) (SEQ ID NO.: 10).
  • a preferred Substance P analog is: Zi-RPKPQQFFMeGlyLM(O 2 )-Z 2 , wherein Z x is NH 2 and Z 2 is C(O)NH 2 (SEQ ID NO: 12).
  • the method of the invention is suitable for a cancer which is a adenocarcinoma (for example, being selected from pancreatic, breast, gastric, renal cell and colorectal cancer), melanoma or a lympho-hematopoietic cancer (for example, being selected from lymphoma, Non-Hodgkin's lymphoma and leukemia).
  • adenocarcinoma for example, being selected from pancreatic, breast, gastric, renal cell and colorectal cancer
  • melanoma for example, being selected from lymphoma, Non-Hodgkin's lymphoma and leukemia.
  • the cancer to be treated or prevented is cervical, liver, breast, pancreas, colon, lung, ovarian, endometrial, fallopian tube or prostate and the increased risk is human papillomavirus infection, hepatitis B, PanIN, BRCAl, BRCA2, PSA, familial adenomatous polyposis syndrome (FAP) or hereditary non-polyposis colorectal cancer syndrome (HNPCC).
  • human papillomavirus infection hepatitis B, PanIN, BRCAl, BRCA2, PSA, familial adenomatous polyposis syndrome (FAP) or hereditary non-polyposis colorectal cancer syndrome (HNPCC).
  • FAP familial adenomatous polyposis syndrome
  • HNPCC hereditary non-polyposis colorectal cancer syndrome
  • the invention contemplates a broad scope of antigen including but not limited to a method wherein the antigen is selected from: a dendritic cell based antigen; a shared tumor antigen; a unique tumor antigen; inactivated whole tumor cells; gene-modified tumor cells or is comprised of mucin 1, HER2/NEU, melanoma- associated antigen, mammaglobin, carcinoembryonic antigen or cyclin Bl.
  • the invention further contemplates a broad scope of immunostimulatory capabilities exploited by the preferred methods, including methods wherein the immunogenic composition stimulates: (a) Natural killer tumor (NKT) cells, natural killer (NK) cells or B-lymphocytes; (b) antibody-dependent cellular cytotoxicity, or (c) THl cell activity (for example, stimulating T lymphocyte production or activity or cytotoxic T lymphocyte (CTL) activity (for example, the T lymphocyte cell being selected from CD4 + cells or CD4 CD25 + cells; for example, the T lymphocyte activity being selected from expression of CCR7, CD45, CCRlO or cutaneous leukocyte antigen; and for example, the CD45 being selected from the CCR7 CD45RO + isoform)).
  • NKT Natural killer tumor
  • NK natural killer
  • B-lymphocytes for example, antibody-dependent cellular cytotoxicity
  • THl cell activity for example, stimulating T lymphocyte production or activity or cytotoxic T lymphocyte (CTL) activity
  • CTL cytotoxic T
  • Another aspect of the present invention is directed to a composition
  • a composition comprising Substance P or an analog thereof; a melanoma-associated antigen; and a pharmaceutically acceptable carrier.
  • a related aspect of the present invention is a method of enhancing an immune response to a melanoma tumor in a subject having a melanoma tumor or at risk of having a melanoma tumor comprising the steps of providing Substance P or an analog thereof to the patient; and providing a melanoma-associated antigen. In some embodiments, either or both of the providing steps are repeated. In others, the Substance P or analog thereof and the antigen are provided separately but simultaneously. In yet others, it is contemplated that the Substance P or analog thereof is provided coincident with, prior to or subsequent to antigen. It is further contemplated that the Substance P or analog thereof is provided intradermally or topically.
  • an effective amount of antigen is provided to the patient biolistically; the effective amount of antigen can be provided as plasmid encoding the antigen.
  • the melanoma- associated antigen is the subject's endogenous antigen.
  • An additional aspect of the present invention is directed to a composition for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer, the composition comprising an immunogenic composition comprising a cell, an antigen and an adjuvant effective amount of Substance P or a Substance P analog.
  • the cell is selected from the group consisting of: an antigen processing cell; a cell engineered to express antigen; a cell engineered to express Substance P or a Substance P analog; and combinations of any one or more of the foregoing cells.
  • the cell and the immunogenic composition are the same.
  • the cell is endogenous to the subject.
  • the invention contemplates any method for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer comprising the step of providing the foregoing cell-containing composition to a subject in need thereof.
  • the present invention is directed to the use of an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog for the manufacture of a medicament for use in a method as defined in any one of the preceding claims.
  • the use of the immunogenic composition further comprises a cell.
  • FIG. 1 provides a comparison in bar graph format of the percentages of CD 11 C + CFSE + BMDCs (bone marrow derived dendritic cells) in DLNs ((skin) draining lymph nodes) following adoptive transference of TN-DCs, SarSP-TN-DCs, SarSP-DCs or untreated DCs as quantified by FACS analysis at different time points, i.e., at Day 5 and Day 7 (***p ⁇ 0.001, means ⁇ 1 SD of 3 independent experiments are shown).
  • the left most bar represents DCs
  • the bar second from the left represents SarSP-DCs
  • the bar second from the right represents TN-DCs
  • the rightmost bar represents SarSP-TN-DCs.
  • FIG. 2 shows that NKlR and CD40 signaling act synergistically to promote BMDC survival.
  • This bar graph provides a representation of the % of CDl Ic + BMDCs expressing Bc 1-2 after culture in medium alone or with SarSP +/- CD40mAb for different time points, i.e., at 24h, 28h, 72h, and 96h.
  • Agonistic CD40 mAb was added at 24h to cultures and the percentages of CDl IC + BMDCs expressing Bcl-2 was analyzed at each time point (*p ⁇ 0.05, **p ⁇ 0.01, and ***p ⁇ 0.001, means ⁇ 1 SD of 3 independent experiments are shown).
  • FIG. 3 is an illustration of a delay type hypersensitivity (DTH) assay showing cellular effector immune responses represented by ear thickness increases of mice sensitized with adoptively transferred TN-DCs, SarSP-TN-DCs, CD40-TN-DCs, or control (non-haptenized) SarSP-DCs or untreated DCs, as analyzed at different time points following delayed type hypersensitivity (DTH) elicitation (***p ⁇ 0.001, means ⁇ 1 SD of 3 independent experiments are shown).
  • DTH delay type hypersensitivity
  • FIG. 4 is a DTH assay illustrated by a bar graph showing the % of ear thickness increase in mice populations at 24 hours, 48 hours, and 72 hours after administration of one dose of either i) pCMV-luc (luciferase — control), ii) pCMV- TRP2, or iii) pCMV-TRP2 (melanoma self Ag) and the NKlRa agonist SarSP in the dorsal side of the ear.
  • the leftmost bar represents 24h; the middle bar represents 48h, and the rightmost bar represents 72h.
  • FIG. 5 are photos of mice sacrificed 22 days after intradermal injection with B16 melanoma cells. Mice were previously vaccinated with nothing (1), luciferase (irrelevant) plasmid by Gene Gun (GG) (2), TRP-2 melanoma Ag encoding plasmid by GG administered at a dose of 1-2 ⁇ g/dose (3), TRP-2 plasmid by GG and NKlR antagonist (4), TRP2 plasmid by GG and 1 dose of SarSP (5 nmol administered locally intradermally at the site of immunization) (5), and TRP-2 plasmid by 2 doses of SarSP (5 nmol injected locally at the immunization site and applied at the moment of the immunization or at day 3 after GG administration of TRP-2) (6).
  • GG Gene Gun
  • mice (1-4) are roughly the same as shown by the circumference of the circle highlighting the location of the tumor, whereas the size of the tumor on the mouse (5) is roughly 1/7 the size of the other tumors, and the size of the tumor on the mouse (6) is markedly smaller than that of the mouse (5) at least by half.
  • mice 7 is a Kaplan-Meier line graph showing the percent survival of mice from 0 to 100 days after administration of B 16/FO melanoma cells.
  • the mice that were i) na ⁇ ve (solid square), ii) administered only SarSP (diamond), iii) administered an irrelevant DNA plasmid via GG (upward pointing triangle), and iv) administered TRP -2 plasmid by GG (circle) were all deceased well before 50 days had passed.
  • TRP -2 via GG and SarSP 1 dose, downward pointing triangle
  • TRP -2 via GG and SarSP 2 doses, open square
  • FIG. 8 is a bar graph showing the % of ear thickness increase in mice populations at 24 hours, 48 hours, and 72 hours in a delayed type hypersensitivity (DTH) assay in mice surviving the melanoma tumors and challenged with GG delivered pCMV-TRP-2 in the dorsal side of the ears lOOd following the initial injection of melanoma cells and 12Od after the initial immunization.
  • the leftmost bar represents 24h; the middle bar represents 48h, and the rightmost bar represents 72h.
  • adjuvant refers to compounds that can increase, stimulate, induce, enhance or augment the immunogenicity of an antigen. In some embodiments, the adjuvant can assist or potentiate the antigenicity of an antigen. [0038] The term “adjuvant effective amount” refers to an amount effective to stimulate, potentiate, augment, enhance or increase the immunogenicity of an antigen.
  • alkyl refers to a saturated branched, straight chain or cyclic hydrocarbon radical.
  • Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, hexyl, and the like.
  • the alkyl groups are (Ci -C 6 ) alkyl.
  • alkenyl refers to an unsaturated branched, straight chain or cyclic hydrocarbon radical having at least one carbon-carbon double bond. The radical may be in either the cis or trans conformation about the double bond(s).
  • Typical alkenyl groups include, but are not limited to, ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, tert-butenyl, pentenyl, hexenyl and the like.
  • the alkenyl group is (Ci -C 6 ) alkenyl.
  • alkynyl refers to an unsaturated branched, straight chain or cyclic hydrocarbon radical having at least one carbon-carbon triple bond.
  • Typical alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, isobutynyl, pentynyl, hexynyl and the like.
  • the alkynyl group is (Ci - C 6 ) alkynyl.
  • aryl refers to an unsaturated cyclic hydrocarbon radical having a conjugated ⁇ electron system.
  • Typical aryl groups include, but are not limited to, penta-2,4-diene, phenyl, naphthyl, anthracyl, azulenyl, chrysenyl, coronenyl, fluoranthenyl, indacenyl, idenyl, ovalenyl, perylenyl, phenalenyl, phenanthrenyl, picenyl, pleiadenyl, pyrenyl, pyranthrenyl, rubicenyl, and the like.
  • the aryl group is (C5 -C 20 ) aryl, with (C5 -C 10) being particularly preferred.
  • alkaryl refers to a straight-chain alkyl, alkenyl or alkynyl group wherein one of the hydrogen atoms bonded to a terminal carbon is replaced with an aryl moiety.
  • Typical alkaryl groups include, but are not limited to, benzyl, benzylidene, benzylidyne, benzenobenzyl, naphthenobenzyl and the like.
  • the alkaryl group is (C 6 -C 26 ) alkaryl, i.e., the alkyl, alkenyl or alkynyl moiety of the alkaryl group is (Ci -C 6 ) and the aryl moiety is (C5 -C 20 ).
  • the alkaryl group is (C 6 -C 13 ) alkaryl, i.e., the alkyl, alkenyl or alkynyl moiety of the alkaryl group is (Ci -C 3 ) and the aryl moiety is (C 5 -C 10 ).
  • alkheteroaryl refers to a straight-chain alkyl, alkenyl or alkynyl group where one of the hydrogen atoms bonded to a terminal carbon atom is replaced with a heteroaryl moiety.
  • the alkheteroaryl group is 6-26 membered alkheteroaryl, i.e., the alkyl, alkenyl or alkynyl moiety of the alkheteroaryl is (Ci -C 6 ) and the heteroaryl is a 5-20-membered heteroaryl.
  • the alkheteroaryl is 6-13 membered alkheteroaryl, i.e., the alkyl, alkenyl or alkynyl moiety is a 5-10 membered heteroaryl.
  • heteroaryl refers to an aryl moiety wherein one or more carbon atoms is replaced with another atom, such as N, P, O, S, As, Se, Si, Te, etc.
  • Typical heteroaryl groups include, but are not limited to, acridarsine, acridine, arsanthridine, arsindole, arsindoline, carbazole, ⁇ -carboline, chromene, cinnoline, furan, imidazole, indazole, indole, indolizine, isoarsindole, isoarsinoline, isobenzofuran, isochromene, isoindole, isophosphoindole, isophosphinoline, isoquinoline, isothiazole, isoxazole, naphthyridine, perimidine, phenanthridine, phenanthroline, phenazine, phosphoin
  • the heteroaryl group is a 5-20 membered heteroaryl, with 5-10 membered aryl being particularly preferred.
  • substituted alkyl, alkenyl, alkynyl, aryl alkaryl, heteroaryl or alkheteroaryl refers to an alkyl, alkenyl, alkynyl, aryl, alkaryl, heteroaryl or alkheteroaryl group in which one or more hydrogen atoms is replaced with another substituent.
  • Preferred substituents include —OR, -SR, -NRR, -NO 2 , -CN, halogen, -C(O)R, -C(O)OR and -C(O)NR, where each R is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, alkaryl, heteroaryl or alkheteroaryl.
  • endogenous antigen refers to an antigen associated with a subject.
  • the occurrence, presence and/or production of an endogenous antigen in/by a subject can be normal for the subject (and its kind) or non-normal for the subject (and its kind).
  • an endogenous antigen can be removed from and returned to the subject in a modified, processed and/or derivatized form and still be an "endogenous antigen" for purposes of the present invention.
  • endogenous cell refers to a cell associated with a subject.
  • the occurrence, presence and/or production of an endogenous cell in/by a subject can be normal for the subject (and its kind) or non-normal for the subject (and its kind).
  • an endogenous cell can be removed from and returned to the subject in a modified, processed and/or derivatized form and still be an "endogenous cell” for purposes of the present invention.
  • Immunotherapy as an approach to treating and/or preventing cancer is gaining momentum.
  • the ability to increase anti-tumor T-cell precursors after administration of a cancer vaccine has been demonstrated in mice and humans. A number of clinical trials are underway. See, Lewis, 2004, Proc. Nat. Acad. Sci.
  • a method for treating cancer in a subject comprises administering an immunogenic composition comprising a cancer tumor antigen and an adjuvant effective amount of Substance P or an analog thereof.
  • a method for preventing cancer in a subject at risk for developing cancer is provided.
  • the invention provides a method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer.
  • a patient at risk for developing cancer may have, for example, a human papilloma virus infection, hepatitis B, PanIN, BRCAl, BRCA2, PSA, familial adenomatous polyposis syndrome (FAP), or hereditary non-polyposis colorectal cancer syndrome (HNPCC).
  • treatment or amelioration of cancer includes, for example, reducing tumor volume, reducing the number of tumors, reducing the spread of a tumor (for example, metastatic spread), or slowing the growth of a tumor.
  • treatment or amelioration of cancer includes, for example, reducing tumor volume, reducing the number of tumors, reducing the spread of a tumor (for example, metastatic spread), or slowing the growth of a tumor when compared to administration of antigen alone.
  • the method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer includes providing to the subject an immunogenic composition and a Substance P or a Substance P analog.
  • an immunogenic composition described in this application can be used according to this method, including, for example, but not limited to an antigen, such as a tumor cell antigen.
  • the antigen may be, for example, a dendritic cell-based antigen, a shared tumor antigen, a unique tumor antigen, inactivated whole tumor cells, gene -modified tumor cells, mucin 1, HER2/NEU, melanoma-associated antigen, MART-1/melan A, TRP-2, protein gplOO, mammaglobin, carcinoembryonic antigen, or cyclin Bl.
  • the antigen may be the subject's endogenous antigen.
  • the invention also contemplates use of analogs or fragments of antigens capable of inducing an immunologic response.
  • the method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer is administered to the subject prior to administration of Substance P or the Substance P analog.
  • Substance P or a Substance P analog is administered to the subject prior to administration of the immunogenic composition.
  • Substance P or a Substance P analog is administered to the subject simultaneously with the immunogenic composition, for example, together as a single composition, or separately as two compositions administered simultaneously.
  • a booster administration of Substance P or a Substance P analog and/or the immunogenic composition is provided to the subject at a time point after the initial administration of Substance P or a Substance P analog and immunogenic composition.
  • the booster administration provides the Substance P or Substance P analog prior to providing the immunogenic composition, while in another embodiment, the booster administration provides the immunogenic composition prior to providing the Substance P or Substance P analog.
  • a booster administration provides only Substance P or a substance P analog.
  • the immunogenic compositions disclosed herein and administered according to the method of invention stimulate natural killer T cells, natural killer cells, B lymphocytes, antibody-dependent cellular cytotoxicity, or ThI cell activity.
  • ThI cell activity includes stimulating type 1 IFN-gamma producing CD4 + T lymphocytes or activity, and cytotoxic TcI lymphocyte (CTL) activity such as the T lymphocyte cell being biased from CD4 + cells or a CD8 + cell lineage or expression of CCR7, CD45, CCRlO or cutaneous leukocyte antigen, and the CD45 expression being expression of CCR7 + CD45RO + isoform.
  • CTL cytotoxic TcI lymphocyte
  • the method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer may be used to treat solid adenocarcinomas such as pancreatic, breast, gastric, renal cell, and colorectal cancers, to treat melanoma, or to treat a lympho-hematopoietic cancer such as lymphoma, non-Hodgkin's lymphoma and leukemia.
  • the method may be used to treat cancers such as cervical, breast, bladder, colon, rectal, kidney, liver, pancreas, colon, lung, ovarian, endometrial, skin (non-melanoma), thyroid, fallopian tube or prostate cancer.
  • the invention contemplates an individualized, subject-by-subject course of treatment which provides to a subject an immunogenic composition comprising the subject's endogenous antigen, for example a cancer tumor antigen, in combination with Substance P or a Substance P analog. It is expected that such a vaccine composition can provide the subject with targeted immunoprotective capabilities, thereby permitting the subject optimal immune conditions for treatment and recovery.
  • endogenous antigen could be first presented to antigen-processing cells ex vivo and then returned to the subject as processed antigen per se; or returned to the subject in the form of antigen- loaded cells together with the aforementioned vaccine composition.
  • Yet another scenario contemplated herein involves formation endogenous Iy of the immunogenic composition of the present invention whereby a subject is provided with Substance P or a Substance P analog which then combines in vivo with the endogenous antigen to form the immunogenic composition; in certain circumstances, cells can accompany the Substance P or Substance P analog and combine in vivo with the endogenous antigen to form an immunogenic composition.
  • the invention provides a method of enhancing an immune response to a melanoma tumor in a subject or of providing protective immunity against melanoma in a subject having a risk of developing a melanoma tumor.
  • the method includes the steps of providing Substance P or a Substance P analog to a subject and providing a melanoma-associated antigen.
  • the melanoma associated antigen can be MART-1/melan A, TRP-I (tyrosinase related protein- 1 or gp75), TRP-2 (tyrosinase related protein-2), Restin, CEA, mucin 1, MAGE, melanoma-associated antigen tyrosinase(l-9), melanoma-melanocyte antigen gp 100(280-288), protein gplOO, melanoma associated antigen melan-A(27-35), gpl00:Pmell7, the subject's own endogenous melanoma associated antigen, or a combination of two or more melanoma antigens obtained by tumor cell lysis.
  • the melanoma associated antigen can be provided according to any of the methods described herein for providing an antigen or immunogenic composition.
  • an effective amount of an MAA is provided to the epidermis biolistically, for example, as a cDNA plasmid via Gene Gun transfection, while in another embodiment, cells, such as dendritic cells loaded with MAA or genetically engineered to express MAA are injected into the patient, for example, intradermally injected or provided according to any other mode of administration contemplated herein.
  • Substance P or a Substance P analog is also administered to patient in an effective amount according to the method.
  • Substance P or the Substance P analog may be administered via injection, for example intradermally, or according to any other mode of administration contemplated herein, such as topically.
  • the melanoma associated antigen is administered to the subject prior to administration of Substance P or the Substance P analog.
  • Substance P or a Substance P analog is administered to the subject prior to administration of the melanoma associated antigen.
  • Substance P or a Substance P analog is administered to the subject simultaneously with the melanoma associated antigen, for example, together as a single composition, or separately as two compositions administered simultaneously.
  • a booster administration of Substance P or a Substance P analog and/or the melanoma associated antigen is provided to the subject at a time point after the initial administration of Substance P or a Substance P analog and melanoma associated antigen.
  • the booster administration provides the Substance P or Substance P analog prior to providing the melanoma associated antigen, while in another embodiment, the booster administration provides the melanoma associated antigen prior to providing the Substance P or Substance P analog.
  • a booster administration provides only Substance P or a substance P analog.
  • a booster administration according to the invention is repeated, for example 1, 2, 3 or more times after the initial administration of the melanoma associated antigen and the Substance P or analog thereof.
  • the booster administration in one embodiment, is provided weekly after the first administration to provide a booster effect on the subject, for example, for three weeks.
  • the cancer to be treated can be a solid tumor, such as a solid adenocarcinoma or a lympho-hematopoietic cancer.
  • the cancer to be treated can be pancreatic cancer, breast cancer, gastric cancer, renal cell carcinoma, colorectal cancer, melanoma, prostate cancer, ovarian cancer, lymphoma, non-Hodgkin's lymphoma or leukemia.
  • the cancer to be prevented can be cervical, liver, breast, pancreas, colon, lung, ovarian or prostate cancer.
  • the antigen in the immunogenic composition can be from any source or derived by any methodology known in the art.
  • Several approaches to creating cancer immunogenic compositions have been attempted including polyvalent immunogenic compositions, wherein the antigen can be from autologous or allogenic tumor cells, tumor cell lysate, shed antigens and/or heat shock proteins prepared from autologous tumor cells.
  • Antigen-defined immunogenic compositions have been described comprised of tumor associated antigens (TAA), peptides, rDNA, recombinant virus or anti-idiotypic antibody.
  • TAA tumor associated antigens
  • DC Dendritic cell
  • the antigen can be a shared tumor antigen, unique tumor antigen, inactivated whole tumor cells or gene-modified cells.
  • the antigen can be a dendritic cell based antigen.
  • the antigen can be mucin 1 , HER2/NEU, melanoma-associated antigen, mammaglobin, carcinoembryonic antigen or cyclin Bl. See, Finn 2003, Nature 3: 630-641, Lü et al, 2005, Current Cancer Drug Targets 5: 611-627, Tabi and Man 2006, Adv. Drug Delivery Rev. 58: 902-915.
  • Dendritic cells induce, direct and control adaptive immune responses, those antigen-specific responses triggered by non-antigen specific innate immune responses. Dendritic cells capture and process antigens in peripheral tissues, then migrate to the lymphoid organs where they present antigens to na ⁇ ve T cells. The presentation of antigens induces an immune response involving both CD4 + T helper 1 cells and cytolytic CD8 + T cells. See, Sun and Bevan, 2003, Science, 300: 337-339, Shedlock and Shen, 2003, Science, 300: 337-339, Janssen et al.,2003, Nature, 421 :852-856.
  • dendritic cells play an important role in inducing a humoral immune response via their capacity to activate B cells and increase antigen-specific antibody production.
  • Dendritic cells can also activate natural killer (NK) cells and natural killer T (NKT) cells.
  • NK natural killer
  • NKT natural killer T
  • methods and compositions for preventing cancer in a subject at risk for developing cancer is provided.
  • a number of cancers have been linked to viral infections, genetic markers or other indicators.
  • presence of a genetic mutation, viral infection or the like is an indicator that the subject is at risk for developing cancer.
  • human papillomavirus (HPV) is found in over 50% of all cervical cancer tumors. Finn 2003, Nature 3:630-640 and Welters, et al, 2003, Cancer Res. 63: 636-641.
  • GardasilTM (Merck) is an available vaccine against HPV types 6, 11, 16 and 18. It has been projected that if a worldwide HPV vaccination program were started in 2010, cervical cancer would be eliminated by 2050. Plummer and Franceschi, 2002, Virus Res. 89: 285-293.
  • HBV hepatitis B virus
  • pancreatic intraepithelial neoplasia PanIN
  • PanIN-3 refers to a severe atypia that is likely to progress to invasive carcinoma.
  • the p 21 WAF1/CIP1 gene has been shown to be overexpressed early in the development of PanIN. Biankin et al., 2001, Cancer Res . 61 : 8830-8837. [0070] This overexpression appears to be cumulative, progressing from 9% in normal ducts to 85% in invasive carcinoma. Its exact role, however, remains unknown. Loss of pl6 INK4A may be the next hit, occurring slightly later than K-ras. Allelic loss has been detected in PanIN-1, and inactivation has been found to increase up to 100% in invasive cancers. Wilentz et al, 1998, Cancer Res. 58: 4740-4744, Moskaluk et al., 1997, Cancer Res. 84: 2140-2143.
  • Prostate specific antigen has been linked with an increased risk of developing prostate cancer.
  • Blood levels generally range from about 0 to about 20 ng/ml or more with about 0-about 2.5 ng/ml considered low to normal, about 2.6 to about 10 ng/ml considered slightly to moderately elevated, about 10 to about 19.9 ng.ml considered moderately elevated and over about 20 ng/ml considered significantly elevated.
  • individual blood levels are to be considered in a clinical context taking into consideration the overall health of the individual, including for example, the presence of benign prostate enlargement, inflammation (prostatitis), infection, age and race. National Comprehensive Cancer Network clinical Practice Guidelines in Oncology, Prostate Cancer, version 1, 2008.
  • Familial adenomatous polyposis is an autosomal dominant disorder characterized by early onset adenomatous polyps throughout the colon. Subjects with this syndrome, if left untreated, generally develop colon cancer by the age of about 35 to 40. The genetic defect is caused by a mutation in the adenomatous polyposis coli (APC) gene. Syndromes once thought to be distinct from FAP are now recognized to be, in reality, part of the phenotypic spectrum of FAP. Syndromes with a germline mutation in the APC gene include FAP, Gardner syndrome, some families with Turcot syndrome, and attenuated adenomatous polyposis coli (AAPC).
  • APC adenomatous polyposis coli
  • Gardner syndrome is characterized by colonic polyposis typical of FAP, along with osteomas (bony growth most commonly on the skull and the mandible), dental abnormalities, and soft tissue tumors.
  • Turcot syndrome is characterized by colonic polyposis typical of FAP, along with central nervous system tumors (medulloblastoma).
  • AAPC is characterized by fewer colonic polyps (average number of polyps, 30-35) as compared to classic FAP. The polyps also tend to develop at a later age (average age of 36 years), and they tend to involve the proximal colonic area. See, Itoh, et al., 1993, Int. J. Colorectal Disease 8(2): 87-94, Desai and Barket, 208, Gastroenterol. Clin. North Am. 37(1): 47-72.
  • HNPCC Hereditary nonpolyposis colorectal cancer
  • Lynch syndrome is an inherited cancer of the gastrointestinal tract. Mutations or variations of the MLHl, MSH2, MSH6, PMS and PMS2 genes increase the risk of developing HNPCC. Papadopoulos et al., 1994, Science 263(5153): 1625-1929, Fishel et al., 1993, Cell 75(5): 1027-1038. Subjects with HNPCC are at increased risk of developing cancer of the stomach, small intestine, liver, gallbladder ducts, hepatobiliary tract, upper urinary tract, brain, skin and prostate. Female subjects with HNPCC are at increased risk of endometrial and ovarian cancer.
  • Substance P refers to peptide: Arg Pro Lys Pro GIn GIn Phe Phe GIy Leu Met, or the single letter representation RPKPQQFFGLM (SEQ ID NO: 1).
  • a Substance P analog as used in the methods and compositions described herein refers to a Substance P analog that comprises one or more amino acids substitutions relative to SEQ ID NO: 1 and can either compete with Substance P for binding to its receptor (NK-I) or agonize the NK-I (neurokinin) receptor according to an assay conventional to the art, e.g., as described in Shue, et al.,Bioorgan Med Chem Letters 2006, 16(4): 1065-1069.
  • the amino acid substitutions can be conservative or non-conservative substitutions. Further, the amino acid substitutions can include substitutions of non- standard amino acids ⁇ e.g. , amino acids other than the 20 amino acids normally encoded by the genetic code).
  • the Substance P analog can comprise norleucine (NIe). In yet another example, the Substance P analog can comprise sarcosine (Sar) or N-methylglycine (MeGIy). In yet another example, the Substance P analog can comprise phenylalanine that is substituted with between 1 and 4 chlorines, more preferably 1 chlorine.
  • the methionine residue side chain sulfur (S) can be oxidated.
  • the methionine is methionine sulfoxide (-NH-CH(CO)- CH 2 -CH 2 -S(O)CH 3 ).
  • the methionine is methionine sulfone or methionine S, S, dioxide , (-NH-CH(CO)-CH 2 -CH 2 -S(O 2 )CH 3 ) , also referred to herein as Met(O) 2 .
  • the amino (designated herein as Zi) or carboxy terminus (designated herein as Z 2 ) of the Substance P analogs can be modified.
  • "Blocked" forms of the Substance P analogs i.e., forms of the Substance P analogs in which the N- and/or C-terminus is blocked with a moiety capable of reacting with the N-terminal -NH 2 or C-terminal -C(O)OH are also specifically contemplated.
  • the N- and/or C-terminal charges of the Substance P analogs can be an N-acylated peptide amide, ester, hydrazide, alcohol and substitutions thereof.
  • N-terminal blocking groups include RC(O)-, where R is -H, (Ci -C 6 ) alkyl, (Ci -C 6 ) alkenyl, (Ci -C 6 ) alkynyl, (C 5 -C 20 ) aryl, (C 6 -C 26 ) alkaryl, 5-20 membered heteroaryl or 6-26 membered alkheteroaryl.
  • Preferred N-terminal blocking groups include acetyl, formyl and dansyl.
  • Typical C-terminal blocking groups include -C(O)NRR and -C(O)OR, where each R is independently defined as above.
  • Preferred C-terminal blocking groups include those wherein each R is independently methyl. In another preferred embodiment the C-terminal group is amidated.
  • Substituted amides generally include, but are not limited to, groups of the formula -C(O)NR-, wherein R is (Ci -C 6 ) alkyl, substituted (Ci -C 6 ) alkyl, (Ci -C 6 ) alkenyl, substituted (Ci -C 6 ) alkenyl, (Ci -C 6 ) alkynyl, substituted (Ci -C 6 ) alkynyl, (C5 -C20) aryl, substituted (C5 -C20) aryl, (C 6 -C 26 ) alkaryl, substituted (C 6 -C 26 ) alkaryl, 5-20 membered heteroaryl, substituted 5-20 membered heteroaryl, 6-26 membered alkheteroaryl and substituted 6-26 membered alkheteroaryl.
  • R is (Ci -C 6 ) alkyl, substituted (Ci -C 6 )
  • Compounds having such non-amide linkages and methods for preparing such compounds are well-known in the art (see, e.g., Spatola, March 1983, Vega Data Vol. 1, Issue 3; Spatola, 1983, "Peptide Backbone Modifications" In: Chemistry and Biochemistry of Amino Acids Peptides and Proteins, Weinstein, ed., Marcel Dekker, New York, p. 267 (general review); Morley, 1980, Trends Pharm.
  • one or more amide linkages can be replaced with peptidomimetic or amide mimetic moieties which do not significantly interfere with the structure or activity of the peptides.
  • Suitable amide mimetic moieties are described, for example, in Olson et al., 1993, J. Med. Chem. 36:3039-3049.
  • compositions for administration of a Substance P analog as an adjuvant in conjunction with a immunogenic composition for combating cancer comprises an adjuvant effective amount of a Substance P analog according to Formula (I) as described herein.
  • the Substance P analogs can be administered by any suitable route that ensures bioavailability in the circulation. This can be achieved, for example, by parenteral routes of administration, including intravenous (IV), intramuscular (IM), intradermal (ID), subcutaneous (SC), cutaneous, and intraperitoneal (IP) administration.
  • parenteral routes of administration including intravenous (IV), intramuscular (IM), intradermal (ID), subcutaneous (SC), cutaneous, and intraperitoneal (IP) administration.
  • IV intravenous
  • IM intramuscular
  • ID intradermal
  • SC subcutaneous
  • IP intraperitoneal
  • other routes of administration can be used.
  • absorption through the gastrointestinal tract can be accomplished by oral routes of administration (including but not limited to ingestion, buccal and sublingual routes) provided appropriate formulations (e.g., enteric coatings) are used to avoid or minimize degradation of the active ingredient, e.g., in the harsh environments of the oral mucosa, stomach and/or small intestine.
  • transmucosal administration via mucosal tissue such as nasal, vaginal and rectal modes of administration may be utilized to avoid or minimize degradation in the gastrointestinal tract.
  • the compositions can be administered transcutaneously (e.g., transdermally), or by inhalation. It will be appreciated that the preferred route may vary with the condition and age of the recipient.
  • immunogenic compositions of the invention as described herein can be administered by any suitable route that ensures bioavailability in the circulation. This can be achieved, for example, by parenteral routes of administration, including intravenous (IV), intramuscular (IM), intradermal (ID), subcutaneous (SC), cutaneously, and intraperitoneal (IP) administration. However, other routes of administration can be used.
  • IV intravenous
  • IM intramuscular
  • ID intradermal
  • SC subcutaneous
  • IP intraperitoneal
  • other routes of administration can be used.
  • absorption through the gastrointestinal tract can be accomplished by oral routes of administration (including but not limited to ingestion, buccal and sublingual routes) provided appropriate formulations (e.g., enteric coatings) are used to avoid or minimize degradation of the active ingredient, e.g., in the harsh environments of the oral mucosa, stomach and/or small intestine.
  • appropriate formulations e.g., enteric coatings
  • transmucosal administration via mucosal tissue such as nasal, vaginal and rectal modes of administration may be utilized to avoid or minimize degradation in the gastrointestinal tract.
  • the compositions can be administered transcutaneously (e.g., transdermally), or by inhalation. It will be appreciated that the preferred route may vary with the condition and age of the recipient.
  • the compositions can be pharmaceutical compositions.
  • the pharmaceutical compositions can be administered by injection or inhalation.
  • the composition can be administered intramuscularly, intradermally, subcutaneously, transmucosally or by inhalation.
  • the actual dose of the Substance P analogs used will vary with the route of administration, and should be adjusted to achieve circulating plasma concentrations of 100 mg/L to 2 g/L, or circulating plasma concentrations as low as about 1 mg/L to as high as 2 g/L, or circulating plasma concentrations as low as about 10 "12 g/L to as high as 2 g/L.
  • Data obtained in animal model systems described herein show that the Substance P analogs can be administered by injection at a dose between 0.01 mg to 200 mg per dose, or at a dose of about 0.01 ug to about 200 mg per dose.
  • the Substance P analog can be administered at a dose of about 0.1 mg to about 100 mg per immunogenic composition dose.
  • the dose can be about 0.5 mg to about 50 mg. In another embodiment, the dose of the Substance P analog can be about 0.5 mg to about 15 mg per immunogenic composition dose. See, FDA Guidance Document, Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, Center for Biologies Evaluation and Research, United States Department of Health and Human Services. Concentrations of Substance P and Substance P analogs as administered, in one embodiment, range preferably from 10 "15 M to 10 "5 M, and more preferably, in another embodiment, from 10 "9 M to 10 "6 M.
  • Substance P or Substance P analogs are administered at a concentration of about 0.02 mg/kg to about 20 mg/kg, with such concentrations being useful, for example, for systemic administration. In another embodiment, Substance P or Substance P analogs are administered to provide about 0.01 ng to about 100 mg to the subject, with such amount of administration being useful when provided, for example, locally, for example, intradermally.
  • the composition can be given via the respiratory system (i.e. by inhalation) via the nose or mouth.
  • the inhalation composition is an aerosol. Aerosols comprised of solubilized agent in a liquid medium or dry powder compositions are known in the art. Aerosol compositions can be administered via nebulizer, metered dose inhalers, (MDIs) or dry powder inhalers (DPIs). MDIs and DPIs are also referred to as puffers.
  • compositions that can be administered in a frequency and duration for prevention or treatment of cancer in a human.
  • the compositions can be administered one time (e.g. single dose).
  • the compositions can be administered multiple times, for example a dose can be given days, weeks or even years after an initial immunogenic antigen plus adjuvant dose is given.
  • the compositions can be administered intermittently, for example, every 30 days, every 60 days, every 90 days, every 180 days, every 360 days, every 5 years, every 10 years and the like.
  • a composition of the invention is administered weekly.
  • the Substance P analogs can be used in combination with an antigen to increase the innate, humoral and/or cellular immune response to the antigen, e.g. act as an adjuvant.
  • the antigen and the Substance P analogs can be administered contemporaneously, for example, on the same day.
  • the antigen and the Substance P analogs can be administered at intervals, for example, the antigen given on Day 1 and the Substance P analogs can be administered on Day- 3, Day -1, Day 2, Day 3, or Day 7, and the like.
  • the Substance P analogs can be given on Day 1 and the antigen can be administered later on Day 1 , or Day 3 or Day 7 and the like.
  • compositions of Substance P and/or its analogs according to Formula (I) set forth herein in combination with an immunogenic composition are useful for the treatment, prevention, or amelioration of cancer in a patient or for reducing the likelihood of developing a cancer in the patient.
  • Amelioration of cancer includes but is not limited to reducing tumor load, slowing the growth or spread of a tumor, or reducing the severity of the tumor.
  • Substance P and/or Substance P analogs according to Formula (I) are provided in an amount sufficient enough for Substance P and/or Substance P analogs to act as adjuvant to the immune response triggered by the immunogenic composition.
  • Immunogenic compositions according to the invention may include antigens, cDNA plasmids encoding an antigen, an antigen presenting cell loaded with antigens, and a cell engineered to express an antigen.
  • Substance P or Substance P analogs are provided as peptides in one embodiment of the invention, while in another embodiment, cells engineered to express Substance P or a Substance P analog are provided in the composition.
  • the antigen of the immunogenic composition includes a dendritic cell-based antigen.
  • the dendritic cell- based antigen may be a tumor-dendritic cell hybrid, a peptide-loaded dendritic cell, whole tumor cell-loaded dendritic cell, a tumor mRNA-loaded dendritic cell, a genetically engineered or gene-loaded dendritic cell engineered to express an antigen.
  • the dendritic cell-based antigen may result from a dendritic cell loaded with a tumor antigen, a dendritic cell exposed to a tumor lysate, a dendritic cell gene-loaded ex vivo or in vivo, or a dendritic cell exposed to a tumor antigen ex vivo or in vivo.
  • the antigen of the immunogenic composition is provided bound to an artificially derived antigen- presenting structure or is provided in a lipid-membrane bound complex containing at least a tumor-derived antigen, and in a further embodiment, additionally containing MHC molecules, and co-stimulatory/immune activation molecules.
  • the antigen is a tumor cell antigen from a tumor cell such as a solid adenocarcinoma, a pancreatic cancer, a breast cancer, a gastric cancer, a renal cell cancer, a colorectal cancer, a melanoma, a lympho-hematopoietic cancer, a lymphoma, a non-Hodgkin's lymphoma, or a leukemia.
  • the tumor cell antigen may be mucin 1 , HER2/NEU, melanoma-associated antigen, MART-1/melan A, mammaglobin, carcinoembryonic antigen, or cyclin Bl.
  • the tumor cell antigen may also be a tumor cell antigen autologous or allogenic to the subject, a heat shock protein from autologous tumor cells, tumor-associated antigens (TAA), shared tumor antigen, unique tumor antigen, inactivated whole tumor cell antigen, and gene-modified cell antigen.
  • TAA tumor-associated antigens
  • a composition including a Substance P or Substance P analog and an immunogenic composition where the immunogenic composition includes a dendritic cell can be used as a dendritic cell vaccine.
  • a dendritic cell can be used to provide an antigen according to any number of mechanisms previously described for providing an antigen to a dendritic cell and the dendritic cell can be included in a composition with a Substance P analog or Substance P.
  • the composition when administered to a patient, acts as a vaccine to treat, prevent or ameliorate cancer in a subject, or to reduce the likelihood of the subject of developing cancer.
  • a composition of the invention includes an antigen and Substance P or a Substance P analog in combination with one another.
  • Substance P or a Substance P analog is provided together with an antigen in two separate containers, while in another embodiment, Substance P or a Substance P analog is provided together with an antigen in the same container.
  • the composition may be provided for contemporaneous administration of the antigen and Substance P or the Substance P analog, or the composition may be provided for sequential administration.
  • the antigen portion of the composition is administered first, followed by administration of the Substance P portion, whereas in another embodiment, the Substance P portion of the composition is administered first, followed by the antigen portion.
  • the cancer antigen and the Substance P analog adjuvant can be in separate, or divided or undivided containers.
  • the two agents can be in liquid, dried, lyophilized, or frozen form, as is convenient for the end user and good for shelf life.
  • the treatments can be administered at one time or sequentially, over a period of, for example, one day, one week, one month, six months or twelve months.
  • Suitable devices for administering the aerosol include atomizers and droppers as well as nebulizers and hand-held aerosol metered dose and dry powder inhaler ("puffer") devices.
  • Suitable regimens for administration include daily or multiple daily administrations by aerosol. Other modes of administration include continual intradermal infusion, transdermal infusion, intravenous injection, intramuscular, sublingual, subcutaneous injection and oral.
  • Suitable compositions of Substance P analogs for administration are any which are pharmaceutically acceptable and in which Substance P analogs retain biological activity. Generally, such compositions are Substance P analogs dissolved in sterile normal saline, buffered normal saline or sterile water.
  • compositions for changing stability, absorption and half-life characteristics can be used, including liposomal compositions, carrier molecules that physically or chemically retard the release of the adjuvant into physiological spaces and slow-release (depot) compositions.
  • Devices for effecting transdermal administration include, for example, electroporation, hollow microneedles, solid microneedles, patches, abrasive devices, and aerosol injectors.
  • Immunogenic compositions can include "pharmaceutically acceptable carriers," which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition.
  • these pharmaceutically acceptable carriers are sterile and pyrogen free.
  • Exemplary carriers are large, slowly metabolized macromolecules such as polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplet emulsions or liposomes) and inactive tumor particles.
  • Such carriers are well known to those of ordinary skill in the art.
  • these carriers can function as immune-stimulating agents.
  • the methods and compositions provide a kit for prophylactic or therapeutic immunotherapy for cancer.
  • the kit further comprises a chemotherapeutic agent.
  • the chemotherapeutic drug can be 5-azacitidine, 5-fluorouracil, 6-mercaptopurine, 6- thioguanine, actinomycin-D (dactinomycin), alemtuzumab, altretamine, aminoglutethimide, anagrelide, arsenic trioxide, asparaginase, bevacizumab, bexarotene, bleomycin, bortezomib, busulfan, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, dasatinib, daunomycin, daunorubicin, decitabine, docetaxel, doxorubicin, epirubicin, estramustine, etoposide, flox
  • the invention provides a kit including Substance P and/or one or more Substance P analogs in combination with an immunogenic composition as described herein.
  • the immunogenic composition may include any of an antigen, an antigen presenting cell, a cell containing an antigen or a cell engineered to express an antigen.
  • a composition is contemplated that is useful for treatment, prevention or ameliorating of cancer in a subject or for reducing the likelihood of developing cancer in a subject at risk for developing a cancer.
  • the composition includes an immunogenic composition including a cell, an antigen, and an adjuvant effective amount of Substance P or a Substance P analog.
  • the cell may be an antigen presenting cell, a cell engineered to express antigen, a cell engineered to express Substance P or a Substance P analog, or a cell endogenous to the subject and combinations of any of these cells.
  • Methods of the invention include administering to a subject the composition including a cell, an antigen, and an adjuvant effective amount of Substance P or Substance P analog.
  • Example 1 Assessment of adjuvant effects of Substance P analogs
  • This example provides exemplary methods to assess the adjuvant effects of Substance P analogs to potentiate the immune response against cancer in an immunotherapy.
  • a number of transgenic animal models are used in the development of cancer immunotherapy. See generally, McLaughlin 2003, Crit. Rev. One. Hemat. 40(1): 53-76, Ostrand-Rosenberg, 2004, Curr. Opin. Immunol. 16(2): 143-150, Xia, 2006, Cell Res. 16(3): 241-259.
  • the SV40 T-antigen-transgenic mouse is a model for human prostate cancer. Tumors spontaneously develop orthotopically with a disease progression that closely resembles the progress of human prostate cancer. Granziero, 1999, Eur. J. Immunol. 29(4): 1127-1138.
  • mice have been created that develop the full spectrum of pancreatic ductal adenocarcinoma from preinvasive neoplasias (PanINSs) to invasive and metastatic disease (infiltrating ductal adenocarcinoma of the pancreas).
  • PanINSs preinvasive neoplasias
  • metastatic disease infiltrating ductal adenocarcinoma of the pancreas.
  • KRASG 12D oncogenic KRAS with a glycine to aspartate substitution at codon 12
  • MistlKrasG12D/ + mutant mice also developed hepatocellular carcinoma. Tuveson et al, 2006, Can. Res. 66: 242-247. See also, Izeradjene et al, 2003, Cancer Cell 11(3): 229-243.
  • Isolated dendritic cells (DC) loaded with tumor antigen ex vivo and administered as a cellular vaccine have been found to induce protective and therapeutic anti-tumor immunity in experimental animals. Timmerman and Levy 1999, Ann. Rev. Medicine 50: 507-529.
  • DC based antigens can be given contemporaneously with the Substance P analogs described herein.
  • the adjuvant effect of the Substance P analogs is expected to ameliorate cancer by, for example, reducing tumor load, slowing development of tumors or reducing severity when compared to SV40 animals given DC vaccine alone.
  • any one or more of the above models are used to assess the effects of Substance P or an analog thereof on immunotherapy to treat, prevent, reduce the severity, reduce the likelihood of developing, and/or ameliorate a symptom of cancer.
  • the immunotherapy is administered to a statistically significant number of animals in the presence and absence of Substance P or an analog thereof.
  • Therapeutic benefit is assessed using an endpoint described in connection with the models discussed above, e.g., survival, immune response, and the like.
  • DCs myeloid dendritic cells
  • SarSP neurokininl receptor agonist
  • ex vzVo-generated DCs have been used in immunization approaches for prevention and treatment of cancer and infectious diseases. Schuler et al., 2003, Curr. Opin. Immunol. 15: 138-147.
  • BMDCs bone marrow derived dendritic cells
  • BMDCs were cultured in complete medium with or without [SaI 3 MBt(O 2 ) 1 ⁇ -Substance P (10 ⁇ 9 M). After 24h, BMDCs were left untreated (DCs, control) or haptenized with trinitrobenzene sulfonic acid (TNBS, 1 mM, 25 min at 37°C) (Sigma) (TN-DCs). DCs and TN-DCs were then labeled with carboxyfluorescein succinimidyl ester (CFSE, 1 ⁇ M, 15 min at 37°C) (Invitrogen).
  • CFSE carboxyfluorescein succinimidyl ester
  • mice were treated (or not) with two doses of anti-CD 154 blocking MRl mAb (Bio Express, West Riverside, NH) injected simultaneously and 2d after BMDC transfer (250 ⁇ g per dose, flank, i.p.). At indicated time points, mice were euthanized and local (popliteal) and distant (cervical and axillary) DLNs were dissected.
  • the number of viable CFSE-labeled CDl Ic + BMDCs in DLNs was analyzed by FACS, as previously described. Mathers et al., 2007, J. Immunol. 178: 7006-7017. An equal number of CDl Ic + DCs (10 6 ) per experimental situation was acquired by FACS and the percentage of CFSE-labeled CDl Ic + BMDCs was calculated with the following formula: [(number of CFSE + CDl Ic + BMDCs) / (total number of CDl Ic + DCs) xlOO].
  • CFSE-labeled BMDCs preincubated or not with Substance P (SarSP-DCs and DCs, respectively) were injected (footpad, s.c.) into B6 mice, and the number of viable CFSE-labeled CDl Ic + BMDCs was quantified in local DLNs for a period of 7d by FACS. Two days after adoptive transference, similar numbers of SarSP-DCs and control DCs were detected in DLNs, indicating that both DC subsets displayed equivalent DLN-homing capacities. Five days after injection, 95% of SarSP-DCs remained in DLNs compared to 63% of control DCs and by day seven 42% of SarSPDCs were present vs.
  • a composition of the present invention can be effective as an immunogenic composition as defined herein.
  • a composition including an immunogenic composition such as an antigen or an immunogenic or other cell, including a dendritic cell, prepared according to the disclosure herein to provide an antigen, and an adjuvant such as Substance P or analog of Substance P can be effective as an immunogenic composition.
  • Example 7.2.2 Synergistic NKlR and CD40 signaling prolong DC survival in vivo
  • SarSP-TN-DCs exhibit prolonged survival once they home in DLNs could be ascribed to sustained expression of anti-apoptotic molecules induced by [SaZMe ⁇ O 2 ) 1 ⁇ -Substance P or a combined effect of NKlR and CD40 signaling, the later provided by DC-activated CD4 + T cells in DLNs. Josien et al, 2000 J. Exp. Med.
  • mice that were non-treated with CD 154 blocking mAb had a significantly higher number of SarSP-TNDCs remaining in DLNs compared to TN-DCs.
  • these results suggest that the prolonged viability of BMDCs homed in DLNs was caused by a synergistic effect of NKlR and CD40 signaling.
  • Elicitation of DTH was performed 6d after sensitization by applying trinitrochlorobenzene (TNCB, 1% in acetone:olive oil, 4:1) (Sigma) on the dorsal surface of the ears.
  • TTCB trinitrochlorobenzene
  • the severity of the DTH responses was assessed by comparing the thickness of ears measured prior and after elicitation for up to 6d, using an electronic caliper (Mitutoyo, Aurora, IL).
  • the DTH responses were expressed as the percent in ear thickness increase using the formula: [(thickness of challenged ear - thickness of control ear) / (thickness of control ear) x 100].
  • mice were euthanized 6d after elicitation and samples from ear skin were fixed in 4% formaldehyde, embedded in paraffin, stained with H&E, and analyzed by microscopy or embedded in Tissue-Tek OCT (Miles Laboratories, Elkhart, IN) and snapfrozen in methyl-butane (Sigma).
  • Tissue-Tek OCT Transion-Tek OCT
  • the composition of the cellular infiltrate 6d after elicitation was assessed by immunofluorescence microscopy of cryostat sections of the ears, as previously described. Mathers et al., 2007, J. Immunol. 178: 7006-7017.
  • Cryostat sections (8 ⁇ m) were mounted on slides pre -treated with Vectabond (Vector Laboratories, Burlingame, CA), fixed in cold 96% ethanol, and incubated with Alexa Fluor 488-CD4 mAb (Invitrogen) and biotin-CD8 (eBioscience, San Diego, CA) or biotin-F4/80 mAbs (Invitrogen), followed by Cy3-streptavidin (Jackson). Cell nuclei were stained with DAPI.
  • mice were sensitized by administration of TN-DCs or SarSP-TN-DCs (footpad, s. c.) and DTH was elicited 5 days after by topical TNCB application to the dorsal skin of the ears. Mice were injected with haptenized CD40-TN-DCs or non- haptenized (untreated or SarSP-signaled) DCs to be included as positive and negative controls, respectively.
  • mice sensitized with TN-DCs, SarSP-TN-DCs, and CD40-TN-DCs exhibited a significant ear thickness increase vs. control mice lday post-elicitation (p ⁇ 0.001).
  • mice sensitized with TN-DCs the ear thickness diminished significantly 2 days after elicitation, and showed minimal differences vs. control mice 3 days post-elicitation.
  • mice sensitized with SarSP-TN-DCs and CD40-TN-DCs exhibited a further increase 2 days after elicitation, and showed a significant prolonged increase in ear thickness in comparison to TN-DCs and control mice for up to 6 days.
  • ThI -polarizing adjuvants in the skin induces the local secretion of pro- inflammatory mediators required to fully activate skin dendritic cells (DCs), polarizing them to induce ThI- and TcI -biased immunity, and promote development of CTLs, generating an effective and long-lasting anti-tumor immune response.
  • DCs skin dendritic cells
  • Th-I driving adjuvant such as SarSP
  • Skin-derived DCs generated under these conditions bias the differentiation of na ⁇ ve tumor- specific T cells into Th-I cells and promote development of cytotoxic T lymphocytes (CTLs), generating an effective and long-lasting anti-tumor immunity.
  • CTLs cytotoxic T lymphocytes
  • Transgenic TRP-2 was delivered by gene gun trans fection (GG) in the dorsal side of mouse ears (one dose). Measurements of % of ear thickness increase resulting from inflammation from an immune response were taken at 24h, 48h, and 72h. As shown in FIG. 4, mice immunized with TRP-2 in the presence of SarSP elicit a significant higher delayed- type hypersensitivity (DTH) response compared to mice immunized only with the GG, as shown by the increased thickness of mouse ears at 72h for those receiving SarSP as compared to the other groups.
  • DTH delayed- type hypersensitivity
  • mice were divided into 6 groups of 6 mice per group.
  • Group 1 received no vaccine.
  • Group 2 was administered the irrelevant transgenic antigen luciferase in plasmid form via GG in two shots of approximately 2 ⁇ g of plasmid/shot.
  • Group 3 received via GG plasmids encoding the melanoma self Ag TRP -2 in two shots of approximately 2 ⁇ g of plasmid/shot.
  • Group 4 received via GG the TRP-2 antigen in two shots of approximately 2 ⁇ g of plasmid/shot as well as an NKlR antagonist L733060 administered intradermally at a dose of 5 nmol.
  • Group 5 received via GG the TRP-2 antigen in two shots of approximately 2 ⁇ g of plasmid/shot with the NKlR agonist [Sar 9 Met (O 2 ) 11 J-SP (SarSP) being administered intradermally at a dose of 5 nmol.
  • Group 6 received three administrations of TRP-2 antigen via GG (approximately 2 ug/shot), the first administration simultaneously with the NKlR agonist [Sar 9 Met (O 2 ) 11 J-SP (SarSP) administered intradermally at a dose of 5 nmol, and in between the remaining two TRP-2 antigen GG doses, the mice were dosed intradermally again with SarSP at days 3, 9, and 16 at 5 nmol per dose.
  • mice were challenged at day 21 after the initial immunization with one dose of the highly aggressive syngeneic B 16/FO malignant melanoma cells injected intradermally in the lower flank (10 5 cells/injection) and tumor growth and mouse survival were analyzed.
  • the data from this experiment are shown in FIGS. 5-7, which demonstrate that a significant delay in tumor growth and mouse survival was observed in those mice receiving both 1 and 2 doses of SarSP and TRP-2.
  • both GG TRP-2 and GG TRP-2 plus NKlR agonist treatments inhibit tumor growth significantly as compared to GG encoding irrelevant Ag and non-immunized mice injected with B 16/FO cells.
  • mice receiving both GG pCMV-TRP-2 had final tumor areas approximately 16 times smaller than those of na ⁇ ve mice, and still even approximately 5 times smaller than mice receiving only TRP-2 based on visual inspection.
  • mice receiving one or 2 doses of the TRP- 2/SarSP vaccination had tumors significantly reduced in size compared to all other groups tested.
  • FIG. 7 an indefinite survival of 45% and of 50% is seen for mice injected either with one dose or with two doses of SarSP respectively, while other mice groups were completely deceased well before 40 days from the initial immunization.
  • GG-TRP-2 plus one or two doses of the NKlR agonist prolonged significantly the survival of mice compared to GG-TRP- 2 alone or with GG encoding an irrelevant Ag and non-immunized mice injected with B 16/FO cells.
  • mice surviving from melanoma tumors were subject to a DTH assay. Results are shown in FIG. 8. Mice were challenged with GG delivered pCMV-TRP-2 in the dorsal side of the ears. Percent of ear thickness increase was measured at 24, 48, and 72 hours after GG administration. As shown in FIG. 8, mice that were initially administered SarSP NKlR agonist and TRP-2 exhibited the greatest percent increase in ear thickness. This indicates that the vaccine was still effective in surviving mice at 4 months (12Od) post immunization and suggests the development of long lasting effector cellular immunity resulting from vaccination with TRP-2 and SarSP.
  • compositions and methods of the invention as described in the above examples and contemplated elsewhere herein are useful for treating, preventing, or ameliorating cancer in a patient, including such methods as administering to a patient Substance P or a Substance P analog as well as a tumor specific antigen according to any of the variety of methods contemplated herein.
  • these data also suggest that an immune response to a melanoma tumor can be enhanced by administering a melanoma antigen and a Substance P analog or Substance P.
  • Example 7.3.2 Development of local and systemic protective immunity by genetic immunization in the skin in the presence of Thl-driving adjuvants.
  • Example 7.3.2 A Induction of TRP-2 specific CTL responses by skin GG- immunization in the presence of Thl-driving adjuvants.
  • Cytotoxic activity of CD8 + T cells against TRP-2 Ag induced under experimental conditions is assessed by CTL assays. Briefly, B6 mice are GG- immunized with pCMV-TRP-2 in the absence (control) or the presence of Imiquimod or SarSP-NKla.
  • mice each are treated as follows: Group 1 receives no GG transfection and receives PBS as a control in lieu of an adjuvant; Group 2 receives pCMV-TRP-2 via GG and receives no adjuvant; Group 3 receives pCMB-TRP-2 via GG and 5nM SarSP-NKla adjuvant; Group 4 receives pCMV- TRP -2 by GG and 5% v/v Imiquimod adjuvant; Group 5 receives no transfection and receives 5 nM SP-NKIa adjuvant; and Group 6 receives no transfection and receives 5% v/v Imiquimod adjuvant.
  • mice are boosted on days 7 and 14. On day 21, mice are euthanatized and suspensions of spleen and lymph nodes cells are prepared. CTL assays are performed. Briefly, splenocytes and lymph node cells ( ⁇ 3xlO 7 ) are restimulated with TRP-2 protein and TRP-2 180"188 peptide (20ng/ml) in medium plus rhIL-2 (30 U/ml) and used as effector cells for CTL assays.
  • effector cells are cocultured with 51 Cr-labeled EL4 cells (syngeneic targets) ⁇ pulsed with TRP-2 " peptide (100ng/ml) at different effector/target cell ratios.
  • TRP-2 " peptide (100ng/ml) at different effector/target cell ratios.
  • CD8 T cells harvested from mice administered a composition of the present invention comprising an adjuvant (SarSP) and the exemplary TRP-2 melanoma self antigen exhibit the greatest cytotoxic activity as compared to other groups tested, thereby confirming effectiveness as a vaccine composition.
  • CD8 T cells harvested from mice administered TRP-2 with the SarSP adjuvant have at least the same activity as CD8 T cells harvested from mice administered TRP-2 with the Imiquimod adjuvant.
  • Example 7.3.2B Induction of local protective immunity
  • mice are GG-immunized in the skin with pCMV-TRP-2a in the absence (control) or the presence of Imiquimod or SP-NKIa adjuvant.
  • mice each are as follows: Group 1 — na ⁇ ve control mice with no GG transfection, no adjuvant (PBS control); Group 2 — no GG transfection, 5 nM Sp- NKIa adjuvant; Group 3 — no GG transfection, 5% v/v Imiquimod; Group 4 pCMV- mTRP-2 GG transfection with no adjuvant; Group 5 pCMV-mTRP-2 with 5 nM SP- NKIa adjuvant; and Group 6 — pCMV-mTRP-2 GG transfection with 5% v/v Imiquimod adjuvant. All groups are challenged with 2x10 5 B16 cells.
  • mice are challenged locally by intradermal injection in the flanks with syngeneic B16 melanoma cells expressing the self- Ag TRP-2.
  • the dose of cells is 2-10 times the LD50 (lethal dose to 50%).
  • the tumor size is monitored 3 times per week along with animal survival. Tumor size is assessed as mean of the tumor area (mm 2 ) ⁇ ISD. Survival is assessed as the percentage of surviving animals.
  • mice receiving the Substance P or Imiquimod adjuvant in addition to the GG immunization of pCMV-mTRP-2 exhibit statistically significant prolonged survival rates over the other groups of mice. It is also observed that the mean tumor area of mice receiving the Substance P or Imiquimod adjuvant in addition to the GG immunization of pCMV-mTRP-2 have average tumor sizes that are statistically significantly smaller than those of the other groups tested.
  • SarSP an adjuvant
  • TRP-2 melanoma self antigen results in modulation of tumor progression, thereby confirming effectiveness as a vaccine composition.
  • Example 7.3.2C Induction of systemic protective immunity against melanoma using senetic immunization in the presence of ThI -adjuvants:
  • This experiment demonstrates that GG-immunization in combination with administration of ThI -driving adjuvants induces generalized immune protection and prevents development of melanoma metastasis.
  • This experiment uses the same six (6) experimental groups as Example 7.3.2B with five (5) mice per group. Mice are immunized at day 1 with boosters administered at day 7 and day 14. At day 21, B 16 melanoma cells are injected intravenously (2xlO 5 cells/ 200 ⁇ l PBS/mouse) to permit homing of tumor cells in the lungs and liver of mice. Animals are euthanized 14 and 21 days after injection of tumor cells and the presence of melanoma metastatic nodules are quantified (number and size) by macroscopic examination and confirmed by microscopy.
  • mice receiving the Substance P or Imiquimod adjuvant in addition to the GG immunization of pCMV-mTRP-2 have statistically fewer and smaller metastatic nodules present in the lungs and as compared to mice in the other groups tested, indicating that the mice have a level of systemic protective immunity against melanoma resulting from the vaccine of the present invention.
  • Example 7.3.2D Induction of melanoma immunotherapy by genetic immunization in the presence ofThl-driving adjuvants
  • mice are injected intradermally in the flanks with 2 x 10 5 syngeneic B16 melanoma cells expressing the self- Ag TRP-2.
  • mice are GG-immunized in the skin with pCMV- TRP -2a alone (control), after local pre-treatment with Imiquimod, SP-NKIa or its vehicles (control); or left untreated (control).
  • Tumor size and animal survival are evaluated as described in Example 7.3.2B. Five mice per group are used. Three independent experiments are performed. The protocol is repeated again, except that the genetic immunization protocol is initiated 7 days after tumor injection, instead of at 3 days to observe the efficacy of immunotherapy in more advanced tumors.
  • mice receiving the adjuvant SarSP or Imiquimod and pCMV-TRP-2a have tumor volumes that are statistically significantly smaller than those in the other groups tested both when the immunization protocol is initiated at either 3 days and at 7 days.
  • Example 7.3.2E Induction of long lasting immunity to melanoma by genetic immunization in the presence of Th-I driving adjuvants
  • mice treated according to any of the methods in Examples 7.3.2A-D are monitored for long lasting effector cell memory. Surviving animals are monitored for the presence of specific Ab production to TRP-2 melanoma antigen 6 and 9 months after immunization and CTL assays are performed 9 months after immunization.
  • mice treated with the SarSP or Imiquimod adjuvant coupled with TRP-2 antigen have antibodies to melanoma antigen present at both 6 and 9 months after immunization, while no surviving mice from other groups has any significant antigen quantity present.
  • CTL assays performed reveal that mice treated with the SarSP or Imiquimod adjuvant coupled with TRP-2 antigen also have CTL memory cells such as CD4 + and CD8 + memory T cells against TRP-2 antigen, while no surviving mice from the other groups has any significant quantity of memory cells present.
  • the data suggest that the vaccination approach provides long lasting cellular immunity to TRP-2 antigen.
  • Example 7.3.2F Prophetic study investigating use of an immunogenic composition of the present invention for treatment, prevention or amelioration of cancer in a human subject, including use for reducing the risk of developing cancer.
  • Any and all of the following studies, when conducted, will be done so in compliance with all applicable FDA and IRB (Institutional Review Board) requirements. Human subjects having a cancer or at risk for developing a cancer will be treated with an immunogenic composition as defined herein.
  • An exemplary composition will comprise Substance P or a Substance P analog.
  • a particularly preferred composition will comprise SarSP. It is expected that existing cancers in human subjects treated in accordance with the methods described elsewhere herein will be ameliorated to a statistically significant extent; and the risk of developing a cancer will be reduced to a statistically significant extent following treatment in those deemed at risk.
  • compositions and methods of the invention as described in the above examples and contemplated elsewhere herein are useful for treating, preventing, or ameliorating cancer in a patient, including such methods as administering to a patient Substance P or a Substance P analog as well as a tumor specific antigen according to any of the variety of methods contemplated herein.
  • these data confirm that an immune response to a cancer tumor antigen, such as but not limited to a melanoma tumor antigen can be enhanced by administering a melanoma antigen and a Substance P analog or Substance P.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Oncology (AREA)
  • Cell Biology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Developmental Biology & Embryology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Reproductive Health (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Disclosed herein are methods and compositions effective for treating, preventing, and ameliorating cancer that utilize Substance P or Substance P analogs and antigens. Also disclosed are methods for vaccinating a patient against cancer that utilize Substance P or Substance P analogs and antigens.

Description

Substance P and Analogs Thereof as a Cancer Immunogenic
Composition Adjuvant
1. Cross-Reference to Related Applications
[0001] This application claims priority to and the benefit of U.S. provisional patent application number 61/045,856 filed April 17, 2008, the contents of which are incorporated by reference herein.
2. Statement of Government Interest
[0002] This invention was made with government support under R-Ol -CAl 00893-05 awarded by the National Institutes of Health. The government has certain rights in the invention.
3. Background of the Invention
[0003] Traditional cancer therapies have largely relied on small molecules to interrupt the division and proliferation of rapidly dividing cancer cells. More recently, monoclonal antibody therapies have been introduced into the clinician's armamentarium. However, even with these advances, cancer survival and cure rates are not acceptable. Improvements are needed.
4. Summary of the Invention
[0004] The present invention is based on the discovery that a neuropeptide can modulate the immunogenicity of an antigen including, but not limited to, a cancer- associated antigen such as a cancer tumor antigen. In its newly discovered capacity as an immune response modifier or adjuvant, the neuropeptide Substance P and certain analogs thereof provide a heretofore unrealized opportunity to formulate and use compositions comprising a select antigen and Substance P (or certain analogs thereof) as an effective cancer vaccine, including a dendritic cell-based vaccine, a solid tumor- based vaccine, and a non-solid tumor-based vaccine. The discussion and studies set forth below underscore the significance of this discovery and highlight the preventative as well as prophylactic aspects of the invention disclosed herein as well as preferred embodiments which can result from clinical exploitation of this discovery. [0005] Provided herein are methods and compositions for use of Substance P or an analog thereof as adjuvants in immunological compositions for use in the treatment, prevention, or amelioration of cancer. In one embodiment, a method for treating cancer in a subject comprises administering an immunogenic composition comprising a cancer tumor antigen and an adjuvant effective amount of a Substance P or an analog thereof. In one embodiment, a method for preventing cancer in a subject at risk for developing cancer is provided. In one embodiment, a method for reducing the likelihood of developing cancer in a subject at risk for developing cancer is provided. [0006] The cancer to be treated can be a solid tumor, such as a solid adenocarcinoma or a lympho-hematopoietic cancer. In a preferred embodiment, the cancer to be treated can be pancreatic cancer, breast cancer, gastric cancer, renal cell carcinoma, colorectal cancer, melanoma, prostate cancer, ovarian cancer, lymphoma, non-Hodgkin's lymphoma, or leukemia. In one embodiment, the cancer to be prevented can be cervical, liver, breast, pancreas, colon, lung, ovarian or prostate cancer.
[0007] The antigen in the immunogenic composition can be from any source or derived by any methodology known in the art. In a preferred embodiment, the antigen can be a shared tumor antigen, unique tumor antigen, inactivated whole tumor cells, or gene -modified cells. In a preferred embodiment, the antigen can be a dendritic cell based antigen. In one embodiment the antigen can be mucin 1 , HER2/NEU, melanoma-associated antigen, mammaglobin, carcinoembryonic antigen, or cyclin Bl.
[0008] In a currently preferred aspect, the composition of the invention is useful for treatment, prevention, or amelioration of cancer in a subject; or, for reducing the likelihood of developing cancer in a subject at risk for developing cancer. In certain embodiments, the composition comprises an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog.
[0009] In certain embodiments, the Substance P analog is of Formula (I): Zi-Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^-Xaa1 l-Z2 (I)
(SEQ ID NO: 11) or a pharmaceutically acceptable salt thereof, wherein: Xaa1 is Arg, Lys, 6-N methyllysine or (6-N, 6-N) dimethyllysine;
Xaa2 is Pro or Ala;
Xaa3 is Lys, Arg, 6-N-methyllysine or (6-N, 6-N) dimethyllysine;
Xaa4 is Pro or Ala; Xaa5 is GIn or Asn;
Xaa6 is GIn or Asn;
Xaa7 is Phe or Phe substituted with chlorine at position 2, 3 or 4;
Xaa8 is Tyr, Phe, or Phe substituted with chlorine at position 2, 3 or 4;
Xaa9 is GIy, Pro, Ala or N-methylglycine; Xaa10 is Leu, VaI, He, Norleucine, Met, Met sulfoxide, Met sulfone, N-methylleucine, or N-methylvaline;
Xaa11 is Met, Met sulfoxide, Met sulfone, or Norleucine;
Zi is R2N- or RC(O)NR-;
Z2 is -C(O)NR2 or -C(O)OR or a salt thereof; each R is independently R is H, (Ci -C6) alkyl, (Ci -C6) alkenyl, (Ci -C6) alkynyl,
(C5 -C2o) aryl, (C6 -C26) alkaryl, 5-20 membered heteroaryl or 6-26 membered alkheteroaryl; and each "— " between residues Xaa1 through Xaa11 independently designates an amide linkage, a substitute amide linkage or an isostere of an amide. [0010] In one embodiment, the Substance P analog can be of Formula (I) as described herein, wherein Xaa1 is Arg; Xaa2 is Pro; Xaa3 is Lys; Xaa4 is Pro; Xaa5 is GIn; Xaa6 is GIn; Xaa7 is Phe or Phe substituted with chlorine at position 4; Xaa8 is Tyr, Phe, or Phe substituted with chlorine at position 4; Xaa9 is GIy, Pro or N- methylglycine; Xaa10 is Leu; and Xaa11 is Met, Met sulfoxide, Met sulfone or Norleucine.
[0011] In a preferred embodiment, the Substance P analog can be of Formula (I) as described herein wherein the "— " between residues Xaal through Xaal 1 designates -C(O)NH-; Zl is H2N-; and Z2 is -C(O)NH2.
[0012] In another preferred embodiment, the Substance P analog can be: RPKPQQFFGLM (SEQ ID NO.: 1);
RPKPQQFFGLNIe (SEQ ID NO. : 2);
RPKPQQFFPLM (SEQ ID NO. : 3); RPKPQQFFMeGIyLM (SEQ ID NO. : 4);
RPKPQQFTGLM (SEQ ID NO. : 5);
RPKPQQF(4-C1)F(4-C1)GLM (SEQ ID NO. : 6);
RPKPQQFFGLM(O) (SEQ ID NO. : 7); RPKPQQFFMeGIyLM(O) (SEQ ID NO.: 8);
RPKPQQFFGLM(O2) (SEQ ID NO. : 9); or
RPKPQQFFMeGIyLM(O2) (SEQ ID NO.: 10).
[0013] In an even more preferred embodiment, the Substance P analog can be Zi-RPKPQQFFMeGlyLM(O2)-Z2; wherein Zi is NH2 and Z2 is C(O)NH2 (SEQ ID NO:12).
[0014] Regarding the antigen of the composition contemplated herein, the antigen can be a dendritic cell-based antigen selected from the group consisting of: tumor- dendritic cell hybrids; peptide-loaded dendritic cells; whole tumor cell-loaded dendritic cells; tumor mRNA-loaded dendritic cells; and genetically engineered or gene-loaded dendritic cells. In certain other embodiments, the antigen is a dendritic cell-based antigen resulting from a dendritic cell loaded with an antigen from a tumor; a dendritic cell exposed to a tumor lysate; a dendritic cell gene-loaded ex vivo; or a dendritic cell exposed to a tumor antigen ex vivo. A particularly preferred embodiment is a composition which is a dendritic cell vaccine. [0015] In another particularly preferred embodiment, a composition comprises an antigen which is a cancer tumor antigen. Such a cancer tumor antigen is selected from the group consisting of: a solid adenocarcinoma, a pancreatic cancer, a breast cancer, a gastric cancer, a renal cell cancer, a colorectal cancer, a melanoma, a lympho -hematopoietic cancer, a lymphoma, a Non-Hodgkin's lymphoma; a leukemia; mucin 1 ; HER2/NEU; melanoma-associated antigen; mammaglobin; carcinoembryonic antigen; and cyclin B 1. Other currently preferred embodiments comprise a cancer tumor antigen selected from the group consisting of: autologous or allogenic tumor cell antigen; tumor cell lysate antigen; shed antigens from autologous tumor cells; heat shock proteins from autologous tumor cells; tumor-associated antigens (TAA); shared tumor antigen, unique tumor antigen, inactivated whole tumor cell antigen; and gene -modified cell antigen. A particularly preferred embodiment is a composition which is a melanoma vaccine. In a particularly preferred embodiment, the antigen is the subject's endogenous antigen.
[0016] Regarding other embodiments of the composition, the invention contemplates a composition in which the antigen and the Substance P or Substance P analog are in combination. In yet other embodiments, they are separate. Other embodiments contemplate a configuration which accommodates a situation in which the antigen and the Substance P or the Substance P analog are for contemporaneous administration. Still other configurations accommodate a situation in which the antigen and the Substance P or the Substance P analog are in separate, divided or undivided containers. Still other configurations accommodate a situation in which the antigen and the Substance P or the Substance P analog are for sequential administration.
[0017] As contemplated herein, the amelioration of cancer by the composition of the invention comprises reducing tumor load, slowing development of tumors or reducing severity when compared to the antigen alone. Moreover, as further contemplated herein, any of the compositions of the invention can be a pharmaceutical composition.
[0018] In a related aspect, the present invention is directed to a method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer, comprising providing to said subject an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog wherein the Substance P analog is of Formula (I) as set out elsewhere herein. Similarly, any method disclosed herein contemplates further defining Formula (I) as follows iXaa1 is Arg; Xaa2 is Pro; Xaa3 is Lys; Xaa4 is Pro; Xaa5 is GIn; Xaa6 is GIn; Xaa7 is Phe or Phe substituted with chlorine at position 4; Xaa8 is Tyr, Phe, or Phe substituted with chlorine at position 4; Xaa9 is GIy, Pro or N- methylglycine; Xaa10 is Leu; and Xaa11 is Met, Met sulfoxide, Met sulfone or Norleucine. Moreover, any method contemplates use of a Substance P analog further defined as: the "— " between residues Xaa1 through Xaa11 designates -C(O)NH-; Zi is H2N-; and Z2 is -C(O)NH2.
[0019] Additionally, any method of the present invention contemplates one or more Substance P analogs is selected from the group consisting of: RPKPQQFFGLM (SEQ ID NO.: 1);
RPKPQQFFGLNIe (SEQ ID NO. : 2);
RPKPQQFFPLM (SEQ ID NO. : 3);
RPKPQQFFMeGIyLM (SEQ ID NO. : 4); RPKPQQFTGLM (SEQ ID NO.: 5);
RPKPQQF(4-C1)F(4-C1)GLM (SEQ ID NO. : 6);
RPKPQQFFGLM(O) (SEQ ID NO. : 7);
RPKPQQFFMeGIyLM(O) (SEQ ID NO. : 8);
RPKPQQFFGLM(O2) (SEQ ID NO. : 9); or RPKPQQFFMeGIyLM(O2) (SEQ ID NO.: 10).
[0020] A preferred Substance P analog is: Zi-RPKPQQFFMeGlyLM(O2)-Z2, wherein Zx is NH2 and Z2 is C(O)NH2 (SEQ ID NO: 12).
[0021] In certain preferred embodiments, the method of the invention is suitable for a cancer which is a adenocarcinoma (for example, being selected from pancreatic, breast, gastric, renal cell and colorectal cancer), melanoma or a lympho-hematopoietic cancer (for example, being selected from lymphoma, Non-Hodgkin's lymphoma and leukemia). In other preferred embodiments, the cancer to be treated or prevented is cervical, liver, breast, pancreas, colon, lung, ovarian, endometrial, fallopian tube or prostate and the increased risk is human papillomavirus infection, hepatitis B, PanIN, BRCAl, BRCA2, PSA, familial adenomatous polyposis syndrome (FAP) or hereditary non-polyposis colorectal cancer syndrome (HNPCC).
[0022] The invention contemplates a broad scope of antigen including but not limited to a method wherein the antigen is selected from: a dendritic cell based antigen; a shared tumor antigen; a unique tumor antigen; inactivated whole tumor cells; gene-modified tumor cells or is comprised of mucin 1, HER2/NEU, melanoma- associated antigen, mammaglobin, carcinoembryonic antigen or cyclin Bl.
[0023] The invention further contemplates a broad scope of immunostimulatory capabilities exploited by the preferred methods, including methods wherein the immunogenic composition stimulates: (a) Natural killer tumor (NKT) cells, natural killer (NK) cells or B-lymphocytes; (b) antibody-dependent cellular cytotoxicity, or (c) THl cell activity (for example, stimulating T lymphocyte production or activity or cytotoxic T lymphocyte (CTL) activity (for example, the T lymphocyte cell being selected from CD4+ cells or CD4 CD25+ cells; for example, the T lymphocyte activity being selected from expression of CCR7, CD45, CCRlO or cutaneous leukocyte antigen; and for example, the CD45 being selected from the CCR7 CD45RO+ isoform)).
[0024] Any of the methods disclosed herein contemplates that, when used in accordance with the teachings of the invention, the numbers of cancer cells in a subject are decreased.
[0025] Another aspect of the present invention is directed to a composition comprising Substance P or an analog thereof; a melanoma-associated antigen; and a pharmaceutically acceptable carrier.
[0026] And, a related aspect of the present invention is a method of enhancing an immune response to a melanoma tumor in a subject having a melanoma tumor or at risk of having a melanoma tumor comprising the steps of providing Substance P or an analog thereof to the patient; and providing a melanoma-associated antigen. In some embodiments, either or both of the providing steps are repeated. In others, the Substance P or analog thereof and the antigen are provided separately but simultaneously. In yet others, it is contemplated that the Substance P or analog thereof is provided coincident with, prior to or subsequent to antigen. It is further contemplated that the Substance P or analog thereof is provided intradermally or topically. In certain embodiments, an effective amount of antigen is provided to the patient biolistically; the effective amount of antigen can be provided as plasmid encoding the antigen. In a particularly preferred embodiment, the melanoma- associated antigen is the subject's endogenous antigen. [0027] An additional aspect of the present invention is directed to a composition for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer, the composition comprising an immunogenic composition comprising a cell, an antigen and an adjuvant effective amount of Substance P or a Substance P analog. In currently preferred embodiments, the cell is selected from the group consisting of: an antigen processing cell; a cell engineered to express antigen; a cell engineered to express Substance P or a Substance P analog; and combinations of any one or more of the foregoing cells. In one preferred embodiment, the cell and the immunogenic composition are the same. In another preferred embodiment, the cell is endogenous to the subject. In a related aspect, the invention contemplates any method for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer comprising the step of providing the foregoing cell-containing composition to a subject in need thereof.
[0028] Finally, the present invention is directed to the use of an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog for the manufacture of a medicament for use in a method as defined in any one of the preceding claims. The use of the immunogenic composition further comprises a cell.
5. Brief Description of the Drawings
[0029] FIG. 1 provides a comparison in bar graph format of the percentages of CD 11 C+CFSE+ BMDCs (bone marrow derived dendritic cells) in DLNs ((skin) draining lymph nodes) following adoptive transference of TN-DCs, SarSP-TN-DCs, SarSP-DCs or untreated DCs as quantified by FACS analysis at different time points, i.e., at Day 5 and Day 7 (***p<0.001, means ± 1 SD of 3 independent experiments are shown). The left most bar represents DCs, the bar second from the left represents SarSP-DCs, the bar second from the right represents TN-DCs and the rightmost bar represents SarSP-TN-DCs.
[0030] FIG. 2 shows that NKlR and CD40 signaling act synergistically to promote BMDC survival. This bar graph provides a representation of the % of CDl Ic+ BMDCs expressing Bc 1-2 after culture in medium alone or with SarSP +/- CD40mAb for different time points, i.e., at 24h, 28h, 72h, and 96h. Agonistic CD40 mAb was added at 24h to cultures and the percentages of CDl IC+ BMDCs expressing Bcl-2 was analyzed at each time point (*p<0.05, **p<0.01, and ***p<0.001, means ± 1 SD of 3 independent experiments are shown). For each time point represented, the leftmost bar represents the control, the bar second from the left represents SarSP, the bar second from the right represents CD40 mAb, and the rightmost bar represents SarSP + CD40 mAb. [0031] FIG. 3 is an illustration of a delay type hypersensitivity (DTH) assay showing cellular effector immune responses represented by ear thickness increases of mice sensitized with adoptively transferred TN-DCs, SarSP-TN-DCs, CD40-TN-DCs, or control (non-haptenized) SarSP-DCs or untreated DCs, as analyzed at different time points following delayed type hypersensitivity (DTH) elicitation (***p<0.001, means ± 1 SD of 3 independent experiments are shown). At day 6, the highest % is represented by SarSP-TN-DCs (square), followed by CD40-TN-DCs (triangle), followed by TN-DCs (circle), followed by DCs (X), followed by SarSP-DCs (diamond) with the lowest %. [0032] FIG. 4 is a DTH assay illustrated by a bar graph showing the % of ear thickness increase in mice populations at 24 hours, 48 hours, and 72 hours after administration of one dose of either i) pCMV-luc (luciferase — control), ii) pCMV- TRP2, or iii) pCMV-TRP2 (melanoma self Ag) and the NKlRa agonist SarSP in the dorsal side of the ear. The leftmost bar represents 24h; the middle bar represents 48h, and the rightmost bar represents 72h.
[0033] FIG. 5 are photos of mice sacrificed 22 days after intradermal injection with B16 melanoma cells. Mice were previously vaccinated with nothing (1), luciferase (irrelevant) plasmid by Gene Gun (GG) (2), TRP-2 melanoma Ag encoding plasmid by GG administered at a dose of 1-2 μg/dose (3), TRP-2 plasmid by GG and NKlR antagonist (4), TRP2 plasmid by GG and 1 dose of SarSP (5 nmol administered locally intradermally at the site of immunization) (5), and TRP-2 plasmid by 2 doses of SarSP (5 nmol injected locally at the immunization site and applied at the moment of the immunization or at day 3 after GG administration of TRP-2) (6). The tumor size for mice (1-4) are roughly the same as shown by the circumference of the circle highlighting the location of the tumor, whereas the size of the tumor on the mouse (5) is roughly 1/7 the size of the other tumors, and the size of the tumor on the mouse (6) is markedly smaller than that of the mouse (5) at least by half.
[0034] FIG. 6 is a line graph showing the tumor area in mm2 of mice from t=10 days from administration of B 16/FO melanoma cells to mice that are i) naϊve
(diamond shape; highest volume at day 21), ii) immunized with an irrelevant plasmid via GG (square; second highest volume at day 21), iii) immunized via GG with TRP-2 plasmid (triangle; second lowest volume at day 21), and iv) immunized via GG with TRP -2 and SarSP administered at the time of GG immunization and after initial dose (2 doses of SarSP) (circle; lowest volume at day 21). The data are the mean ± 1 SD of tumor area (mm2) of 6 mice treated per experimental group. [0035] FIG. 7 is a Kaplan-Meier line graph showing the percent survival of mice from 0 to 100 days after administration of B 16/FO melanoma cells. The mice that were i) naϊve (solid square), ii) administered only SarSP (diamond), iii) administered an irrelevant DNA plasmid via GG (upward pointing triangle), and iv) administered TRP -2 plasmid by GG (circle) were all deceased well before 50 days had passed. However, there was an indefinite survival of 45% and 50% for mice injected with TRP -2 via GG and SarSP (1 dose, downward pointing triangle) and with TRP -2 via GG and SarSP (2 doses, open square), respectively. The sample population for each group was 6 mice.
[0036] FIG. 8 is a bar graph showing the % of ear thickness increase in mice populations at 24 hours, 48 hours, and 72 hours in a delayed type hypersensitivity (DTH) assay in mice surviving the melanoma tumors and challenged with GG delivered pCMV-TRP-2 in the dorsal side of the ears lOOd following the initial injection of melanoma cells and 12Od after the initial immunization. The leftmost bar represents 24h; the middle bar represents 48h, and the rightmost bar represents 72h.
6. Detailed Description of the Invention 6.1 Definitions
[0037] The term "adjuvant" refers to compounds that can increase, stimulate, induce, enhance or augment the immunogenicity of an antigen. In some embodiments, the adjuvant can assist or potentiate the antigenicity of an antigen. [0038] The term "adjuvant effective amount" refers to an amount effective to stimulate, potentiate, augment, enhance or increase the immunogenicity of an antigen.
[0039] The term "alkyl" refers to a saturated branched, straight chain or cyclic hydrocarbon radical. Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, hexyl, and the like. In preferred embodiments, the alkyl groups are (Ci -C6) alkyl. [0040] The term "alkenyl" refers to an unsaturated branched, straight chain or cyclic hydrocarbon radical having at least one carbon-carbon double bond. The radical may be in either the cis or trans conformation about the double bond(s). Typical alkenyl groups include, but are not limited to, ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, tert-butenyl, pentenyl, hexenyl and the like. In preferred embodiments, the alkenyl group is (Ci -C6) alkenyl.
[0041] The term "alkynyl" refers to an unsaturated branched, straight chain or cyclic hydrocarbon radical having at least one carbon-carbon triple bond. Typical alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, isobutynyl, pentynyl, hexynyl and the like. In preferred embodiments, the alkynyl group is (Ci - C6) alkynyl.
[0042] The term "aryl" refers to an unsaturated cyclic hydrocarbon radical having a conjugated π electron system. Typical aryl groups include, but are not limited to, penta-2,4-diene, phenyl, naphthyl, anthracyl, azulenyl, chrysenyl, coronenyl, fluoranthenyl, indacenyl, idenyl, ovalenyl, perylenyl, phenalenyl, phenanthrenyl, picenyl, pleiadenyl, pyrenyl, pyranthrenyl, rubicenyl, and the like. In preferred embodiments, the aryl group is (C5 -C20) aryl, with (C5 -C 10) being particularly preferred.
[0043] The term "alkaryl" refers to a straight-chain alkyl, alkenyl or alkynyl group wherein one of the hydrogen atoms bonded to a terminal carbon is replaced with an aryl moiety. Typical alkaryl groups include, but are not limited to, benzyl, benzylidene, benzylidyne, benzenobenzyl, naphthenobenzyl and the like. In preferred embodiments, the alkaryl group is (C6 -C26) alkaryl, i.e., the alkyl, alkenyl or alkynyl moiety of the alkaryl group is (Ci -C6) and the aryl moiety is (C5 -C20). In particularly preferred embodiments, the alkaryl group is (C6 -C13) alkaryl, i.e., the alkyl, alkenyl or alkynyl moiety of the alkaryl group is (Ci -C3) and the aryl moiety is (C5-C10).
[0044] The term alkheteroaryl refers to a straight-chain alkyl, alkenyl or alkynyl group where one of the hydrogen atoms bonded to a terminal carbon atom is replaced with a heteroaryl moiety. In preferred embodiments, the alkheteroaryl group is 6-26 membered alkheteroaryl, i.e., the alkyl, alkenyl or alkynyl moiety of the alkheteroaryl is (Ci -C6) and the heteroaryl is a 5-20-membered heteroaryl. In particularly preferred embodiments the alkheteroaryl is 6-13 membered alkheteroaryl, i.e., the alkyl, alkenyl or alkynyl moiety is a 5-10 membered heteroaryl.
[0045] The term "heteroaryl" refers to an aryl moiety wherein one or more carbon atoms is replaced with another atom, such as N, P, O, S, As, Se, Si, Te, etc. Typical heteroaryl groups include, but are not limited to, acridarsine, acridine, arsanthridine, arsindole, arsindoline, carbazole, β-carboline, chromene, cinnoline, furan, imidazole, indazole, indole, indolizine, isoarsindole, isoarsinoline, isobenzofuran, isochromene, isoindole, isophosphoindole, isophosphinoline, isoquinoline, isothiazole, isoxazole, naphthyridine, perimidine, phenanthridine, phenanthroline, phenazine, phosphoindole, phosphinoline, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, quinazoline, quinoline, quinolizine, quinoxaline, selenophene, tellurophene, thiophene and xanthene. In preferred embodiments, the heteroaryl group is a 5-20 membered heteroaryl, with 5-10 membered aryl being particularly preferred. [0046] The term "substituted alkyl, alkenyl, alkynyl, aryl alkaryl, heteroaryl or alkheteroaryl" refers to an alkyl, alkenyl, alkynyl, aryl, alkaryl, heteroaryl or alkheteroaryl group in which one or more hydrogen atoms is replaced with another substituent. Preferred substituents include —OR, -SR, -NRR, -NO2, -CN, halogen, -C(O)R, -C(O)OR and -C(O)NR, where each R is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, alkaryl, heteroaryl or alkheteroaryl.
[0047] The term "endogenous antigen" refers to an antigen associated with a subject. The occurrence, presence and/or production of an endogenous antigen in/by a subject can be normal for the subject (and its kind) or non-normal for the subject (and its kind). As contemplated herein, an endogenous antigen can be removed from and returned to the subject in a modified, processed and/or derivatized form and still be an "endogenous antigen" for purposes of the present invention.
[0048] The term "endogenous cell" refers to a cell associated with a subject. The occurrence, presence and/or production of an endogenous cell in/by a subject can be normal for the subject (and its kind) or non-normal for the subject (and its kind). As contemplated herein, an endogenous cell can be removed from and returned to the subject in a modified, processed and/or derivatized form and still be an "endogenous cell" for purposes of the present invention. 6.2 Methods
[0049] Immunotherapy as an approach to treating and/or preventing cancer is gaining momentum. The ability to increase anti-tumor T-cell precursors after administration of a cancer vaccine has been demonstrated in mice and humans. A number of clinical trials are underway. See, Lewis, 2004, Proc. Nat. Acad. Sci.
101(supp 2): 14653-14656, Lage et al, 2005, Curr. Cancer Drug Targets 5: 611-627 and Tabi and Man, 2006, Adv. Drug Delivery Reviews 58: 902-915.
[0050] Provided herein are methods and compositions for use of Substance P or an analog thereof as adjuvants in immunological compositions to combat cancer. In one embodiment, a method for treating cancer in a subject comprises administering an immunogenic composition comprising a cancer tumor antigen and an adjuvant effective amount of Substance P or an analog thereof. In one embodiment, a method for preventing cancer in a subject at risk for developing cancer is provided.
[0051] In one aspect, the invention provides a method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer. A patient at risk for developing cancer may have, for example, a human papilloma virus infection, hepatitis B, PanIN, BRCAl, BRCA2, PSA, familial adenomatous polyposis syndrome (FAP), or hereditary non-polyposis colorectal cancer syndrome (HNPCC). According to one embodiment of the invention, treatment or amelioration of cancer includes, for example, reducing tumor volume, reducing the number of tumors, reducing the spread of a tumor (for example, metastatic spread), or slowing the growth of a tumor. According to one embodiment of the invention, treatment or amelioration of cancer includes, for example, reducing tumor volume, reducing the number of tumors, reducing the spread of a tumor (for example, metastatic spread), or slowing the growth of a tumor when compared to administration of antigen alone.
[0052] According to one embodiment, the method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer includes providing to the subject an immunogenic composition and a Substance P or a Substance P analog. Any of the immunogenic compositions described in this application can be used according to this method, including, for example, but not limited to an antigen, such as a tumor cell antigen. As explained herein, the antigen may be, for example, a dendritic cell-based antigen, a shared tumor antigen, a unique tumor antigen, inactivated whole tumor cells, gene -modified tumor cells, mucin 1, HER2/NEU, melanoma-associated antigen, MART-1/melan A, TRP-2, protein gplOO, mammaglobin, carcinoembryonic antigen, or cyclin Bl. In one embodiment, the antigen may be the subject's endogenous antigen. The invention also contemplates use of analogs or fragments of antigens capable of inducing an immunologic response.
[0053] According to one embodiment, the method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer, the immunogenic composition is administered to the subject prior to administration of Substance P or the Substance P analog. In another embodiment, Substance P or a Substance P analog is administered to the subject prior to administration of the immunogenic composition. In yet another embodiment, Substance P or a Substance P analog is administered to the subject simultaneously with the immunogenic composition, for example, together as a single composition, or separately as two compositions administered simultaneously.
[0054] In a further embodiment, a booster administration of Substance P or a Substance P analog and/or the immunogenic composition is provided to the subject at a time point after the initial administration of Substance P or a Substance P analog and immunogenic composition. In one embodiment, the booster administration provides the Substance P or Substance P analog prior to providing the immunogenic composition, while in another embodiment, the booster administration provides the immunogenic composition prior to providing the Substance P or Substance P analog. In yet another embodiment, a booster administration provides only Substance P or a substance P analog. [0055] The immunogenic compositions disclosed herein and administered according to the method of invention stimulate natural killer T cells, natural killer cells, B lymphocytes, antibody-dependent cellular cytotoxicity, or ThI cell activity. ThI cell activity includes stimulating type 1 IFN-gamma producing CD4+ T lymphocytes or activity, and cytotoxic TcI lymphocyte (CTL) activity such as the T lymphocyte cell being biased from CD4+ cells or a CD8 + cell lineage or expression of CCR7, CD45, CCRlO or cutaneous leukocyte antigen, and the CD45 expression being expression of CCR7+CD45RO+ isoform. [0056] According to one embodiment of the invention, the method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer may be used to treat solid adenocarcinomas such as pancreatic, breast, gastric, renal cell, and colorectal cancers, to treat melanoma, or to treat a lympho-hematopoietic cancer such as lymphoma, non-Hodgkin's lymphoma and leukemia. According to another embodiment of the invention, the method may be used to treat cancers such as cervical, breast, bladder, colon, rectal, kidney, liver, pancreas, colon, lung, ovarian, endometrial, skin (non-melanoma), thyroid, fallopian tube or prostate cancer. [0057] With respect to those embodiments using an endogenous antigen, the invention contemplates an individualized, subject-by-subject course of treatment which provides to a subject an immunogenic composition comprising the subject's endogenous antigen, for example a cancer tumor antigen, in combination with Substance P or a Substance P analog. It is expected that such a vaccine composition can provide the subject with targeted immunoprotective capabilities, thereby permitting the subject optimal immune conditions for treatment and recovery. In this regard, it is further contemplated that endogenous antigen could be first presented to antigen-processing cells ex vivo and then returned to the subject as processed antigen per se; or returned to the subject in the form of antigen- loaded cells together with the aforementioned vaccine composition. Yet another scenario contemplated herein involves formation endogenous Iy of the immunogenic composition of the present invention whereby a subject is provided with Substance P or a Substance P analog which then combines in vivo with the endogenous antigen to form the immunogenic composition; in certain circumstances, cells can accompany the Substance P or Substance P analog and combine in vivo with the endogenous antigen to form an immunogenic composition.
[0058] According to another aspect, the invention provides a method of enhancing an immune response to a melanoma tumor in a subject or of providing protective immunity against melanoma in a subject having a risk of developing a melanoma tumor. The method includes the steps of providing Substance P or a Substance P analog to a subject and providing a melanoma-associated antigen. For example, the melanoma associated antigen can be MART-1/melan A, TRP-I (tyrosinase related protein- 1 or gp75), TRP-2 (tyrosinase related protein-2), Restin, CEA, mucin 1, MAGE, melanoma-associated antigen tyrosinase(l-9), melanoma-melanocyte antigen gp 100(280-288), protein gplOO, melanoma associated antigen melan-A(27-35), gpl00:Pmell7, the subject's own endogenous melanoma associated antigen, or a combination of two or more melanoma antigens obtained by tumor cell lysis. The melanoma associated antigen (MAA) can be provided according to any of the methods described herein for providing an antigen or immunogenic composition. For example, according to one embodiment, an effective amount of an MAA is provided to the epidermis biolistically, for example, as a cDNA plasmid via Gene Gun transfection, while in another embodiment, cells, such as dendritic cells loaded with MAA or genetically engineered to express MAA are injected into the patient, for example, intradermally injected or provided according to any other mode of administration contemplated herein. Substance P or a Substance P analog is also administered to patient in an effective amount according to the method. Substance P or the Substance P analog may be administered via injection, for example intradermally, or according to any other mode of administration contemplated herein, such as topically.
[0059] According to one embodiment of the invention, the melanoma associated antigen is administered to the subject prior to administration of Substance P or the Substance P analog. In another embodiment, Substance P or a Substance P analog is administered to the subject prior to administration of the melanoma associated antigen. In yet another embodiment, Substance P or a Substance P analog is administered to the subject simultaneously with the melanoma associated antigen, for example, together as a single composition, or separately as two compositions administered simultaneously.
[0060] In a further embodiment, a booster administration of Substance P or a Substance P analog and/or the melanoma associated antigen is provided to the subject at a time point after the initial administration of Substance P or a Substance P analog and melanoma associated antigen. In one embodiment, the booster administration provides the Substance P or Substance P analog prior to providing the melanoma associated antigen, while in another embodiment, the booster administration provides the melanoma associated antigen prior to providing the Substance P or Substance P analog. In yet another embodiment, a booster administration provides only Substance P or a substance P analog. According to a further embodiment, a booster administration according to the invention is repeated, for example 1, 2, 3 or more times after the initial administration of the melanoma associated antigen and the Substance P or analog thereof. For example, the booster administration, in one embodiment, is provided weekly after the first administration to provide a booster effect on the subject, for example, for three weeks.
6.2.1 Antigens and Cancers
[0061] The cancer to be treated can be a solid tumor, such as a solid adenocarcinoma or a lympho-hematopoietic cancer. In a preferred embodiment, the cancer to be treated can be pancreatic cancer, breast cancer, gastric cancer, renal cell carcinoma, colorectal cancer, melanoma, prostate cancer, ovarian cancer, lymphoma, non-Hodgkin's lymphoma or leukemia. In one embodiment, the cancer to be prevented can be cervical, liver, breast, pancreas, colon, lung, ovarian or prostate cancer.
[0062] The antigen in the immunogenic composition can be from any source or derived by any methodology known in the art. Several approaches to creating cancer immunogenic compositions have been attempted including polyvalent immunogenic compositions, wherein the antigen can be from autologous or allogenic tumor cells, tumor cell lysate, shed antigens and/or heat shock proteins prepared from autologous tumor cells. Antigen-defined immunogenic compositions have been described comprised of tumor associated antigens (TAA), peptides, rDNA, recombinant virus or anti-idiotypic antibody. Dendritic cell (DC)-based immunogenic compositions have used as the antigen, tumor-DC hybrids, peptide loaded DC, whole tumor cell loaded DC, tumor mRNA loaded DC and genetically engineered DC. Mocellin et al., 2004, Exp. Cell Res. 299: 267-278, Janelsins, et al, 2008, Blood, published online Nov. 5, 2008, blood-2008-06-163121, Mathers et al, 2007, J. Immunol. 178: 7006-7017.
[0063] In a preferred embodiment, the antigen can be a shared tumor antigen, unique tumor antigen, inactivated whole tumor cells or gene-modified cells. In a preferred embodiment, the antigen can be a dendritic cell based antigen. In one embodiment the antigen can be mucin 1 , HER2/NEU, melanoma-associated antigen, mammaglobin, carcinoembryonic antigen or cyclin Bl. See, Finn 2003, Nature 3: 630-641, Lage et al, 2005, Current Cancer Drug Targets 5: 611-627, Tabi and Man 2006, Adv. Drug Delivery Rev. 58: 902-915.
[0064] Dendritic cells induce, direct and control adaptive immune responses, those antigen-specific responses triggered by non-antigen specific innate immune responses. Dendritic cells capture and process antigens in peripheral tissues, then migrate to the lymphoid organs where they present antigens to naϊve T cells. The presentation of antigens induces an immune response involving both CD4+ T helper 1 cells and cytolytic CD8+ T cells. See, Sun and Bevan, 2003, Science, 300: 337-339, Shedlock and Shen, 2003, Science, 300: 337-339, Janssen et al.,2003, Nature, 421 :852-856. In addition, dendritic cells play an important role in inducing a humoral immune response via their capacity to activate B cells and increase antigen-specific antibody production. Dendritic cells can also activate natural killer (NK) cells and natural killer T (NKT) cells. Fernandez et al., 1999, Nature Med 5:405-411 and Kadowaki et al., 2001, J. Exp. Med. 193: 1221-1226. [0065] Studies have shown that dendritic cells loaded with antigens from tumors can induce a protective and therapeutic antitumor immune response. Banchereau, et al., 2005, Nature Immunol. Rev. 5: 296-306. Methods of producing dendritic cell- based cancer vaccines have been evaluated in animal proof-of-concept studies, including gene-loading dendritic cells ex vivo and exposing dendritic cells to tumor lysate or tumor antigens ex vivo. See, Inaba et al., 1990, J. Exp. Med 172: 631-640, Somasse et al, 1992, J. Exp. Med 175: 15-21, Zitvogel, et al., 1996, J. Exp. Med 183, 87-97, Celluzzi et al, 1996, J. Exp. Med 183: 283-287, Nair et al, 1997, Int. J. Cancer 70: 706-715, Fu et al, 1996, Transplantation 62: 659-665 Ma et al, 2003, Eur. J. Immunol 33: 2123-2132, Asavaroengchai et al, 2002, Proc. Natl. Acad. Sci. USA 99: 931-936. Methods based on gene loading DCs and tumor lysate or antigen DCs have been found to be effective at promoting antigen-specific T cell immunity. Inaba et al, 1990, J. Exp. Med 111: 631-640. Gene loaded DCs have also been identified as efficacious at inducing humoral immunity. Somasse et al, 1992, J. Exp. Med 175: 15-21. [0066] In one embodiment, methods and compositions for preventing cancer in a subject at risk for developing cancer is provided. A number of cancers have been linked to viral infections, genetic markers or other indicators. In some instances, presence of a genetic mutation, viral infection or the like is an indicator that the subject is at risk for developing cancer. For example human papillomavirus (HPV) is found in over 50% of all cervical cancer tumors. Finn 2003, Nature 3:630-640 and Welters, et al, 2003, Cancer Res. 63: 636-641. Gardasil™ (Merck) is an available vaccine against HPV types 6, 11, 16 and 18. It has been projected that if a worldwide HPV vaccination program were started in 2010, cervical cancer would be eliminated by 2050. Plummer and Franceschi, 2002, Virus Res. 89: 285-293.
[0067] Chronically infected individuals with hepatitis B virus (HBV) are known to be at increased risk for liver cancer. In Taiwan, there has been a marked reduction in the incidence of childhood liver cancer after a national vaccination program against HBV was started in 1986. Huang and Lin, 2000, Vaccine, 18: S35-S38.
[0068] The link between pancreatic cancer and pancreatic precursor lesions has been recognized for many decades. Sommers et al., 1954, Gastroenterology 27: 629- 640. A standard classification system and progression model has been put forward by the National Cancer Institute Think Tank. Kern et al., 2001, Cancer Res. 61 : 4923- 4932. The term pancreatic intraepithelial neoplasia (PanIN) now describes the various changes seen in the pancreatic duct system. There is a grading, from 1 to 3, according to the degree of structural dysplasia and cytological atypia. For example, PanIN-3 refers to a severe atypia that is likely to progress to invasive carcinoma. Hruban et al., 2000, Am J. Pathol. 156: 1821-1825. Less than about 50% of PanIN-3 lesions are associated with loss of SMAD4 protein expression and is predominately seen in invasive cancers and generally is associated with a poorer prognosis.
[0069] The genetic mutations that take place in these precursor lesions appear to occur in a temporal, ordered sequence rather than a random fashion, confirming the theory that some act as gatekeepers in the initiation of neoplastic growth whilst others allow tumor growth and malignant progression. The first to appear is K-ras, and it is thus thought to represent a "gatekeeper" function. K-ras mutations are already present in normal pancreatic ductal cells, with the mutation rate increasing up to 100% for PanIN-3 lesions. Lϋttges et al. 1999 Cancer 85: 1703-1710 and Tada et al., 1996, Gastroenterology 110: 227-231. The p21WAF1/CIP1 gene has been shown to be overexpressed early in the development of PanIN. Biankin et al., 2001, Cancer Res . 61 : 8830-8837. [0070] This overexpression appears to be cumulative, progressing from 9% in normal ducts to 85% in invasive carcinoma. Its exact role, however, remains unknown. Loss of pl6INK4A may be the next hit, occurring slightly later than K-ras. Allelic loss has been detected in PanIN-1, and inactivation has been found to increase up to 100% in invasive cancers. Wilentz et al, 1998, Cancer Res. 58: 4740-4744, Moskaluk et al., 1997, Cancer Res. 84: 2140-2143. Overexpression of p53 is a later event still, usually occurring in PanIN-2 and -3. Wilentz et al., 1998, Cancer Res. 58: 4740-4744, DiGiuseppe, et al, 1994, Am. J. Clin. Pathol. 101 : 684-688, Apple, et al, 1999, Hum. Pathol. 30: 123-129, Heinmoller et al, 2000 Am. J. Pathol. 157: 83-92. Loss of SMAD4 protein expression, occurring in less than 50% of PanIN-3 lesions, is predominately seen in invasive cancers and may signify a poorer prognosis. Schwarte-Waldhoff, et al, 2000 Proc. Natl. Acad. Sci USA 97: 9624-9629, Lϋttges et al, 2001, Am. J. Pathol 157: 83-92, Tascilar et al, 2001, Clin. Cancer Res. 7: 4115- 4121. [0071] Mutations in the BRCAl and BRC A2 gene have been identified with breast and ovarian cancer susceptibility. Gayther et al., 1999, Am. J. Hum. Genet. 65: 1021-1029, Ganguly, 1997, Genet. Test 1 : 85-90, Lallas et al., 1999, MoI. Genet. Metab. 67: 357-363, Arver 1999, Genet. Test 3: 223-226, Bochar et al., 2000, Cell 102(2): 257-265, Irminger-Finger et al., 1999, Biol. Chem. 380(2): 117-28, Zhang et al., 1998, Oncogene 16(13): 1713-1721, Malone et al., 200, Cancer, 88(6): 1393- 1402, Ganguly et al., 1997, Genet. Test 1 : 85-90, Ramus et al., 1999, Genes Chromosomes Cancer 25(2): 91-96, Crook et al., 1998, Oncogene 17(13): 1681-1689, Abeliovich et al., 1997, Am. J. Hum. Genet. 60: 505-514. BRCAl mutations have also been linked to risk of colon cancer and fallopian tube carcinoma. Lin et al., 1999, Dis. Colon Rectum 42: 1041-1045, Garcia-Patino et al., 1998, Cancer Genet. Cytogenet. 104: 119-123, Zweemer et al., 2000, Gynecol. Oncol. 76(1): 45-50.
[0072] Prostate specific antigen (PSA) has been linked with an increased risk of developing prostate cancer. Blood levels generally range from about 0 to about 20 ng/ml or more with about 0-about 2.5 ng/ml considered low to normal, about 2.6 to about 10 ng/ml considered slightly to moderately elevated, about 10 to about 19.9 ng.ml considered moderately elevated and over about 20 ng/ml considered significantly elevated. It will be appreciated that individual blood levels are to be considered in a clinical context taking into consideration the overall health of the individual, including for example, the presence of benign prostate enlargement, inflammation (prostatitis), infection, age and race. National Comprehensive Cancer Network clinical Practice Guidelines in Oncology, Prostate Cancer, version 1, 2008. [0073] Familial adenomatous polyposis (FAP) is an autosomal dominant disorder characterized by early onset adenomatous polyps throughout the colon. Subjects with this syndrome, if left untreated, generally develop colon cancer by the age of about 35 to 40. The genetic defect is caused by a mutation in the adenomatous polyposis coli (APC) gene. Syndromes once thought to be distinct from FAP are now recognized to be, in reality, part of the phenotypic spectrum of FAP. Syndromes with a germline mutation in the APC gene include FAP, Gardner syndrome, some families with Turcot syndrome, and attenuated adenomatous polyposis coli (AAPC). Gardner syndrome is characterized by colonic polyposis typical of FAP, along with osteomas (bony growth most commonly on the skull and the mandible), dental abnormalities, and soft tissue tumors. Turcot syndrome is characterized by colonic polyposis typical of FAP, along with central nervous system tumors (medulloblastoma). AAPC is characterized by fewer colonic polyps (average number of polyps, 30-35) as compared to classic FAP. The polyps also tend to develop at a later age (average age of 36 years), and they tend to involve the proximal colonic area. See, Itoh, et al., 1993, Int. J. Colorectal Disease 8(2): 87-94, Desai and Barket, 208, Gastroenterol. Clin. North Am. 37(1): 47-72.
[0074] Hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome is an inherited cancer of the gastrointestinal tract. Mutations or variations of the MLHl, MSH2, MSH6, PMS and PMS2 genes increase the risk of developing HNPCC. Papadopoulos et al., 1994, Science 263(5153): 1625-1929, Fishel et al., 1993, Cell 75(5): 1027-1038. Subjects with HNPCC are at increased risk of developing cancer of the stomach, small intestine, liver, gallbladder ducts, hepatobiliary tract, upper urinary tract, brain, skin and prostate. Female subjects with HNPCC are at increased risk of endometrial and ovarian cancer.
6.2.2 Substance P and analogs thereof [0075] As will be understood by those of skill in the art, Substance P (SEQ ID NO 1) refers to peptide: Arg Pro Lys Pro GIn GIn Phe Phe GIy Leu Met, or the single letter representation RPKPQQFFGLM (SEQ ID NO: 1). As such, a Substance P analog as used in the methods and compositions described herein refers to a Substance P analog that comprises one or more amino acids substitutions relative to SEQ ID NO: 1 and can either compete with Substance P for binding to its receptor (NK-I) or agonize the NK-I (neurokinin) receptor according to an assay conventional to the art, e.g., as described in Shue, et al.,Bioorgan Med Chem Letters 2006, 16(4): 1065-1069. The amino acid substitutions can be conservative or non-conservative substitutions. Further, the amino acid substitutions can include substitutions of non- standard amino acids {e.g. , amino acids other than the 20 amino acids normally encoded by the genetic code). In one example, the Substance P analog can comprise norleucine (NIe). In yet another example, the Substance P analog can comprise sarcosine (Sar) or N-methylglycine (MeGIy). In yet another example, the Substance P analog can comprise phenylalanine that is substituted with between 1 and 4 chlorines, more preferably 1 chlorine.
[0076] In one embodiment the methionine residue side chain sulfur (S) can be oxidated. In one embodiment the methionine is methionine sulfoxide (-NH-CH(CO)- CH2-CH2-S(O)CH3). In one embodiment the methionine is methionine sulfone or methionine S, S, dioxide , (-NH-CH(CO)-CH2-CH2-S(O2)CH3) , also referred to herein as Met(O)2.
[0077] It will be apparent to one skilled in the art that the amino (designated herein as Zi) or carboxy terminus (designated herein as Z2) of the Substance P analogs can be modified. "Blocked" forms of the Substance P analogs, i.e., forms of the Substance P analogs in which the N- and/or C-terminus is blocked with a moiety capable of reacting with the N-terminal -NH2 or C-terminal -C(O)OH are also specifically contemplated. In some embodiments the N- and/or C-terminal charges of the Substance P analogs can be an N-acylated peptide amide, ester, hydrazide, alcohol and substitutions thereof. In a preferred embodiment, either the N- and/or C-terminus (preferably both termini) of the Substance P analogs are blocked. Typical N-terminal blocking groups include RC(O)-, where R is -H, (Ci -C6) alkyl, (Ci -C6) alkenyl, (Ci -C6) alkynyl, (C5 -C20) aryl, (C6 -C26) alkaryl, 5-20 membered heteroaryl or 6-26 membered alkheteroaryl. Preferred N-terminal blocking groups include acetyl, formyl and dansyl. Typical C-terminal blocking groups include -C(O)NRR and -C(O)OR, where each R is independently defined as above. Preferred C-terminal blocking groups include those wherein each R is independently methyl. In another preferred embodiment the C-terminal group is amidated.
[0078] Substituted amides generally include, but are not limited to, groups of the formula -C(O)NR-, wherein R is (Ci -C6) alkyl, substituted (Ci -C6) alkyl, (Ci -C6) alkenyl, substituted (Ci -C6) alkenyl, (Ci -C6) alkynyl, substituted (Ci -C6) alkynyl, (C5 -C20) aryl, substituted (C5 -C20) aryl, (C6 -C26) alkaryl, substituted (C6 -C26) alkaryl, 5-20 membered heteroaryl, substituted 5-20 membered heteroaryl, 6-26 membered alkheteroaryl and substituted 6-26 membered alkheteroaryl.
[0079] Amide isosteres generally include, but are not limited to, -CH2 NH-, -CH2 S-, -CH2CH2 -, -CH=CH- (cis and trans), -C(O)CH2 -, -CH(OH)CH2 - and -CH2 SO-. Compounds having such non-amide linkages and methods for preparing such compounds are well-known in the art (see, e.g., Spatola, March 1983, Vega Data Vol. 1, Issue 3; Spatola, 1983, "Peptide Backbone Modifications" In: Chemistry and Biochemistry of Amino Acids Peptides and Proteins, Weinstein, ed., Marcel Dekker, New York, p. 267 (general review); Morley, 1980, Trends Pharm.
Sci. 1 :463-468; Hudson et al, 1979, Int. J. Prot. Res. 14:177-185 (-CH2 NH-, -CH2 CH2 -); Spatola et al., 1986, Life Sci. 38:1243-1249 (-CH2 -S); Hann, 1982, J. Chem. Soc. Perkin Trans. I. 1 :307-314 (-CH=CH-, cis and trans); Almquist et al., 1980, J. Med. Chem. 23:1392-1398 (-COCH2 -); Jennings- White et al., Tetrahedron. Lett. 23 :2533 (-COCH2-); European Patent Application EP 45665 (1982) CA 97:39405 (-CH(OH)CH2 -); Holladay et al., 1983, Tetrahedron Lett. 24:4401-4404 (-C(OH)CH2 -); and Hruby, 1982, Life Sci. 31 :189-199 (-CH2 -S-).
[0080] Additionally, one or more amide linkages can be replaced with peptidomimetic or amide mimetic moieties which do not significantly interfere with the structure or activity of the peptides. Suitable amide mimetic moieties are described, for example, in Olson et al., 1993, J. Med. Chem. 36:3039-3049.
6.3 Compositions
[0081] In one embodiment, provided are compositions for administration of a Substance P analog as an adjuvant in conjunction with a immunogenic composition for combating cancer. In one embodiment, the composition comprises an adjuvant effective amount of a Substance P analog according to Formula (I) as described herein.
[0082] The Substance P analogs can be administered by any suitable route that ensures bioavailability in the circulation. This can be achieved, for example, by parenteral routes of administration, including intravenous (IV), intramuscular (IM), intradermal (ID), subcutaneous (SC), cutaneous, and intraperitoneal (IP) administration. However, other routes of administration can be used. For example, absorption through the gastrointestinal tract can be accomplished by oral routes of administration (including but not limited to ingestion, buccal and sublingual routes) provided appropriate formulations (e.g., enteric coatings) are used to avoid or minimize degradation of the active ingredient, e.g., in the harsh environments of the oral mucosa, stomach and/or small intestine. Alternatively, transmucosal administration via mucosal tissue such as nasal, vaginal and rectal modes of administration may be utilized to avoid or minimize degradation in the gastrointestinal tract. In yet another alternative, the compositions can be administered transcutaneously (e.g., transdermally), or by inhalation. It will be appreciated that the preferred route may vary with the condition and age of the recipient.
[0083] The immunogenic compositions of the invention as described herein, whether protein, DNA, or cell based, can be administered by any suitable route that ensures bioavailability in the circulation. This can be achieved, for example, by parenteral routes of administration, including intravenous (IV), intramuscular (IM), intradermal (ID), subcutaneous (SC), cutaneously, and intraperitoneal (IP) administration. However, other routes of administration can be used. For example, absorption through the gastrointestinal tract can be accomplished by oral routes of administration (including but not limited to ingestion, buccal and sublingual routes) provided appropriate formulations (e.g., enteric coatings) are used to avoid or minimize degradation of the active ingredient, e.g., in the harsh environments of the oral mucosa, stomach and/or small intestine. Alternatively, transmucosal administration via mucosal tissue such as nasal, vaginal and rectal modes of administration may be utilized to avoid or minimize degradation in the gastrointestinal tract. In yet another alternative, the compositions can be administered transcutaneously (e.g., transdermally), or by inhalation. It will be appreciated that the preferred route may vary with the condition and age of the recipient.
[0084] In a preferred embodiment, the compositions can be pharmaceutical compositions. In a more preferred embodiment, the pharmaceutical compositions can be administered by injection or inhalation. In an even more preferred embodiment, the composition can be administered intramuscularly, intradermally, subcutaneously, transmucosally or by inhalation.
[0085] The actual dose of the Substance P analogs used will vary with the route of administration, and should be adjusted to achieve circulating plasma concentrations of 100 mg/L to 2 g/L, or circulating plasma concentrations as low as about 1 mg/L to as high as 2 g/L, or circulating plasma concentrations as low as about 10"12 g/L to as high as 2 g/L. Data obtained in animal model systems described herein show that the Substance P analogs can be administered by injection at a dose between 0.01 mg to 200 mg per dose, or at a dose of about 0.01 ug to about 200 mg per dose. In another embodiment, the Substance P analog can be administered at a dose of about 0.1 mg to about 100 mg per immunogenic composition dose. In another embodiment, the dose can be about 0.5 mg to about 50 mg. In another embodiment, the dose of the Substance P analog can be about 0.5 mg to about 15 mg per immunogenic composition dose. See, FDA Guidance Document, Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, Center for Biologies Evaluation and Research, United States Department of Health and Human Services. Concentrations of Substance P and Substance P analogs as administered, in one embodiment, range preferably from 10"15M to 10"5M, and more preferably, in another embodiment, from 10"9M to 10"6M. In one embodiment, Substance P or Substance P analogs are administered at a concentration of about 0.02 mg/kg to about 20 mg/kg, with such concentrations being useful, for example, for systemic administration. In another embodiment, Substance P or Substance P analogs are administered to provide about 0.01 ng to about 100 mg to the subject, with such amount of administration being useful when provided, for example, locally, for example, intradermally. [0086] In one embodiment, the composition can be given via the respiratory system (i.e. by inhalation) via the nose or mouth. In one embodiment the inhalation composition is an aerosol. Aerosols comprised of solubilized agent in a liquid medium or dry powder compositions are known in the art. Aerosol compositions can be administered via nebulizer, metered dose inhalers, (MDIs) or dry powder inhalers (DPIs). MDIs and DPIs are also referred to as puffers.
[0087] Provided are methods and compositions that can be administered in a frequency and duration for prevention or treatment of cancer in a human. In one embodiment, the compositions can be administered one time (e.g. single dose). In one embodiment, the compositions can be administered multiple times, for example a dose can be given days, weeks or even years after an initial immunogenic antigen plus adjuvant dose is given. In one embodiment, the compositions can be administered intermittently, for example, every 30 days, every 60 days, every 90 days, every 180 days, every 360 days, every 5 years, every 10 years and the like. In another embodiment, a composition of the invention is administered weekly.
[0088] The Substance P analogs can be used in combination with an antigen to increase the innate, humoral and/or cellular immune response to the antigen, e.g. act as an adjuvant. In one embodiment, the antigen and the Substance P analogs can be administered contemporaneously, for example, on the same day. In one embodiment, the antigen and the Substance P analogs can be administered at intervals, for example, the antigen given on Day 1 and the Substance P analogs can be administered on Day- 3, Day -1, Day 2, Day 3, or Day 7, and the like. In one embodiment, the Substance P analogs can be given on Day 1 and the antigen can be administered later on Day 1 , or Day 3 or Day 7 and the like.
[0089] The invention contemplates compositions of Substance P and/or its analogs according to Formula (I) set forth herein in combination with an immunogenic composition. Such compositions are useful for the treatment, prevention, or amelioration of cancer in a patient or for reducing the likelihood of developing a cancer in the patient. Amelioration of cancer includes but is not limited to reducing tumor load, slowing the growth or spread of a tumor, or reducing the severity of the tumor.
[0090] According to the invention, Substance P and/or Substance P analogs according to Formula (I) are provided in an amount sufficient enough for Substance P and/or Substance P analogs to act as adjuvant to the immune response triggered by the immunogenic composition. Immunogenic compositions according to the invention may include antigens, cDNA plasmids encoding an antigen, an antigen presenting cell loaded with antigens, and a cell engineered to express an antigen. Substance P or Substance P analogs are provided as peptides in one embodiment of the invention, while in another embodiment, cells engineered to express Substance P or a Substance P analog are provided in the composition.
[0091] According to one embodiment of the invention, the antigen of the immunogenic composition includes a dendritic cell-based antigen. The dendritic cell- based antigen may be a tumor-dendritic cell hybrid, a peptide-loaded dendritic cell, whole tumor cell-loaded dendritic cell, a tumor mRNA-loaded dendritic cell, a genetically engineered or gene-loaded dendritic cell engineered to express an antigen. The dendritic cell-based antigen may result from a dendritic cell loaded with a tumor antigen, a dendritic cell exposed to a tumor lysate, a dendritic cell gene-loaded ex vivo or in vivo, or a dendritic cell exposed to a tumor antigen ex vivo or in vivo.
[0092] According to another embodiment of the invention, the antigen of the immunogenic composition is provided bound to an artificially derived antigen- presenting structure or is provided in a lipid-membrane bound complex containing at least a tumor-derived antigen, and in a further embodiment, additionally containing MHC molecules, and co-stimulatory/immune activation molecules.
[0093] According to a further embodiment of the invention, the antigen is a tumor cell antigen from a tumor cell such as a solid adenocarcinoma, a pancreatic cancer, a breast cancer, a gastric cancer, a renal cell cancer, a colorectal cancer, a melanoma, a lympho-hematopoietic cancer, a lymphoma, a non-Hodgkin's lymphoma, or a leukemia. The tumor cell antigen may be mucin 1 , HER2/NEU, melanoma-associated antigen, MART-1/melan A, mammaglobin, carcinoembryonic antigen, or cyclin Bl. The tumor cell antigen may also be a tumor cell antigen autologous or allogenic to the subject, a heat shock protein from autologous tumor cells, tumor-associated antigens (TAA), shared tumor antigen, unique tumor antigen, inactivated whole tumor cell antigen, and gene-modified cell antigen.
[0094] According to another embodiment of the invention, a composition including a Substance P or Substance P analog and an immunogenic composition where the immunogenic composition includes a dendritic cell can be used as a dendritic cell vaccine. For example, a dendritic cell can be used to provide an antigen according to any number of mechanisms previously described for providing an antigen to a dendritic cell and the dendritic cell can be included in a composition with a Substance P analog or Substance P. The composition, when administered to a patient, acts as a vaccine to treat, prevent or ameliorate cancer in a subject, or to reduce the likelihood of the subject of developing cancer.
[0095] According to another embodiment, a composition of the invention includes an antigen and Substance P or a Substance P analog in combination with one another. In another embodiment, Substance P or a Substance P analog is provided together with an antigen in two separate containers, while in another embodiment, Substance P or a Substance P analog is provided together with an antigen in the same container. The composition may be provided for contemporaneous administration of the antigen and Substance P or the Substance P analog, or the composition may be provided for sequential administration. For example, in one embodiment, the antigen portion of the composition is administered first, followed by administration of the Substance P portion, whereas in another embodiment, the Substance P portion of the composition is administered first, followed by the antigen portion.
[0096] The cancer antigen and the Substance P analog adjuvant can be in separate, or divided or undivided containers. The two agents can be in liquid, dried, lyophilized, or frozen form, as is convenient for the end user and good for shelf life. The treatments can be administered at one time or sequentially, over a period of, for example, one day, one week, one month, six months or twelve months.
[0097] Suitable devices for administering the aerosol are known in the art and include atomizers and droppers as well as nebulizers and hand-held aerosol metered dose and dry powder inhaler ("puffer") devices. Suitable regimens for administration include daily or multiple daily administrations by aerosol. Other modes of administration include continual intradermal infusion, transdermal infusion, intravenous injection, intramuscular, sublingual, subcutaneous injection and oral. Suitable compositions of Substance P analogs for administration are any which are pharmaceutically acceptable and in which Substance P analogs retain biological activity. Generally, such compositions are Substance P analogs dissolved in sterile normal saline, buffered normal saline or sterile water. Other compositions for changing stability, absorption and half-life characteristics can be used, including liposomal compositions, carrier molecules that physically or chemically retard the release of the adjuvant into physiological spaces and slow-release (depot) compositions. Devices for effecting transdermal administration include, for example, electroporation, hollow microneedles, solid microneedles, patches, abrasive devices, and aerosol injectors.
[0098] Immunogenic compositions can include "pharmaceutically acceptable carriers," which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Typically these pharmaceutically acceptable carriers are sterile and pyrogen free. Exemplary carriers are large, slowly metabolized macromolecules such as polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplet emulsions or liposomes) and inactive tumor particles. Such carriers are well known to those of ordinary skill in the art. In some embodiments, these carriers can function as immune-stimulating agents. [0099] In yet other embodiments, the methods and compositions provide a kit for prophylactic or therapeutic immunotherapy for cancer. In one embodiment, the kit further comprises a chemotherapeutic agent. In one embodiment the chemotherapeutic drug can be 5-azacitidine, 5-fluorouracil, 6-mercaptopurine, 6- thioguanine, actinomycin-D (dactinomycin), alemtuzumab, altretamine, aminoglutethimide, anagrelide, arsenic trioxide, asparaginase, bevacizumab, bexarotene, bleomycin, bortezomib, busulfan, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, dasatinib, daunomycin, daunorubicin, decitabine, docetaxel, doxorubicin, epirubicin, estramustine, etoposide, floxuridine, fludarabine, gefϊtinib, gemcitabine, gemtuzumab (gemtuzumab ozogamicin), goserelin, hydroxyurea, ibritumomab, idarubicin, ifosfamide, irinotecan, imatinib, lapatinib, lenalidomide, leuprolide, lomustine, mechlorethamine, mercaptopurine, methotrexate, melphalan, mitoxantrone, mitomycin, nelarabine, oxaliplatin, paclitaxel, pemetrexed, pentostatin, procarbazine, sorafenib, streptozocin, sunitinib, tretinoin, tositumomab (with or without 1131), temozolomide, temsirolimus, teniposide, thalidomide, thioguanine, thiotepa, topotecan, toremifene, vinblastine, vincristine, vinorelbine or vorinostat. [00100] In another embodiment, the invention provides a kit including Substance P and/or one or more Substance P analogs in combination with an immunogenic composition as described herein. For example, the immunogenic composition may include any of an antigen, an antigen presenting cell, a cell containing an antigen or a cell engineered to express an antigen.
[00101] According to yet another embodiment of the invention, a composition is contemplated that is useful for treatment, prevention or ameliorating of cancer in a subject or for reducing the likelihood of developing cancer in a subject at risk for developing a cancer. The composition includes an immunogenic composition including a cell, an antigen, and an adjuvant effective amount of Substance P or a Substance P analog. The cell may be an antigen presenting cell, a cell engineered to express antigen, a cell engineered to express Substance P or a Substance P analog, or a cell endogenous to the subject and combinations of any of these cells. Methods of the invention include administering to a subject the composition including a cell, an antigen, and an adjuvant effective amount of Substance P or Substance P analog.
7. Examples
7.1. Example 1: Assessment of adjuvant effects of Substance P analogs [00102] This example provides exemplary methods to assess the adjuvant effects of Substance P analogs to potentiate the immune response against cancer in an immunotherapy.
[00103] A number of transgenic animal models are used in the development of cancer immunotherapy. See generally, McLaughlin 2003, Crit. Rev. One. Hemat. 40(1): 53-76, Ostrand-Rosenberg, 2004, Curr. Opin. Immunol. 16(2): 143-150, Xia, 2006, Cell Res. 16(3): 241-259. The SV40 T-antigen-transgenic mouse is a model for human prostate cancer. Tumors spontaneously develop orthotopically with a disease progression that closely resembles the progress of human prostate cancer. Granziero, 1999, Eur. J. Immunol. 29(4): 1127-1138. Transgenic mice have been created that develop the full spectrum of pancreatic ductal adenocarcinoma from preinvasive neoplasias (PanINSs) to invasive and metastatic disease (infiltrating ductal adenocarcinoma of the pancreas). These mice express physiological levels of oncogenic KRAS with a glycine to aspartate substitution at codon 12 (KRASG 12D) in the progenitor cells of mouse pancreas. More recently it was noted that many MistlKrasG12D/+ mutant mice also developed hepatocellular carcinoma. Tuveson et al, 2006, Can. Res. 66: 242-247. See also, Izeradjene et al, 2003, Cancer Cell 11(3): 229-243.
[00104] Isolated dendritic cells (DC) loaded with tumor antigen ex vivo and administered as a cellular vaccine have been found to induce protective and therapeutic anti-tumor immunity in experimental animals. Timmerman and Levy 1999, Ann. Rev. Medicine 50: 507-529. Using an animal model, such as the SV40 T- antigen mouse, DC based antigens can be given contemporaneously with the Substance P analogs described herein. The adjuvant effect of the Substance P analogs is expected to ameliorate cancer by, for example, reducing tumor load, slowing development of tumors or reducing severity when compared to SV40 animals given DC vaccine alone.
[00105] Any one or more of the above models are used to assess the effects of Substance P or an analog thereof on immunotherapy to treat, prevent, reduce the severity, reduce the likelihood of developing, and/or ameliorate a symptom of cancer. Typically, the immunotherapy is administered to a statistically significant number of animals in the presence and absence of Substance P or an analog thereof. Therapeutic benefit is assessed using an endpoint described in connection with the models discussed above, e.g., survival, immune response, and the like.
7.2. Example 2: Effect of the neurokininl receptor agonist fSar9 Met(θ2)111-Substance P (SarSP) on dendritic cell survival [00106] As stated elsewhere herein, myeloid dendritic cells (DCs) are preferred target antigen-presenting cells for positive cellular vaccination protocols. Steinman and Banchereau 2007, Nature, 449: 419-426. For example, during the past decade, ex vzVo-generated DCs have been used in immunization approaches for prevention and treatment of cancer and infectious diseases. Schuler et al., 2003, Curr. Opin. Immunol. 15: 138-147. Additionally, other approaches to DC-based vaccines have focused on the adjuvant effect of pro-inflammatory mediators conferring DCs the capability to initiate and bias T cell immune responses. Mailliard et al., 2004, Cancer Res. 64: 5934-5937, Vieira et al., 2000, J. Immunol. 164: 4507-4512 and Hokey et al., 2005, Cancer Res. 65: 10059-10067. Heretofore, the advances have been limited given that DC-based vaccines described to date have not always elicited potent T cell immunity. Ridgway 2003, Cancer Invest. 21 : 873-886.
[00107] Generation of efficient T cell immunity using ex vzVo-generated DCs requires a critical number of adoptively transferred DCs capable of surviving apoptosis. Additionally, interaction of DCs with cells in early apoptosis downregulates the T cell-stimulatory ability of DCs and induces immunological tolerance.
[00108] The survival of DCs transporting Ag from peripheral tissues to DLNs is crucial for initiation of cellular immunity, and administration of DCs unable to survive in the periphery or DLNs represents a major drawback for current DC-based immune-stimulatory therapies. Park et al. 2006, Nat. Biotechnol. 24: 1581-1590, Hemans et al., 2000, J. Immunol. 164: 3095-3101, Nopora and Brocker 2002, J. Immunol. 169: 3006-3014.
[00109] Recently, it has been demonstrated that the nervous and immune systems interact to modulate the immune response via secretion of neuropeptides. The balance of proinflammatory and anti-inflammatory neuropeptides in the CNS and peripheral tissues is crucial in maintaining self-tolerance under the steady-state.
Indeed, a predominant secretion of the pro-inflammatory SP vs. anti-inflammatory neuropeptides favors chronic inflammatory and autoimmune disorders in the CNS and peripheral tissues. O'Connor et al., 2004, J. Cell Physiol. 201 : 167-180, Reinke et al., 2006, J. Neuroimmunol. 180: 117-125, Weinstock et al., 2003, J. Immunol. 171 :
3762-3767.
[00110] Recent publications have demonstrated that SP recruits immature DCs to inflammation sites, and that signaling skin DCs via NKlR participates in ThI polarization and induction of CTL/Tcl bias. Lambrecht 2001 Respir. Res. 2: 133- 138, Mathers et al., 2007, J. Immunol. 178: 7006-7017. However, the mechanisms involved in the relevant DC-stimulatory activities of pro-inflammatory tachykinins have hitherto been unknown. Moreover, the ability of proinflammatory tachykinins to promote the survival of ex vzVo-generated DCs relevant for DC based immunotherapies has not been previously investigated. [00111] Using a model of ex vzvo-generated BMDCs (bone marrow derived dendritic cells), we have confirmed that regardless of their maturation stage, BMDCs expressed constitutively the functional variant of NKlR, that SarSP induces apoptotic cell rescue mediated through NKlR signaling, and that SarSP anti-apoptotic effect is mediated by PI3K-Akt pathway.
[00112] Our data demonstrate a synergistic effect of signaling BMDCs via NKlR and CD40 confirmed in vivo by those results showing a significant increase of viable SarSP-TN DCs in the absence of the CD40L inhibition, compared to the number of TN-DCs that received CD40 signaling in the absence of exogenous NKlR agonist. The fact that inhibition of CD40-CD40L interaction diminished greatly the anti- apoptotic effects exerted by both DC survival signals demonstrates the relevance of DC-CD4+ Th cell interaction in DLNs for prolonging survival of NKlR-signaled BMDCs.
[00113] Importantly, enhanced longevity of Ag-loaded BMDCs signaled via NKlR and CD40 resulted in elicitation of a potent and sustained cellular immunity mediated mainly by CD8+ T cells and macrophages. Moreover, the observation that elicitation of DTH responses promoted by BMDCs signaled via NKlR persisted up to one week implies that the effector cells overcame the mechanisms of resolution of inflammation and remained active for the period that the Ag was present in the skin. Saint-Mezard, et al, 2003, J. Invest. Dermatol. 120: 641-647.
[00114] In summary, the data presented herein demonstrate that signaling BMDCs via NKlR with proinflammatory tachykinins enhances DC survival in vitro and in vivo, resulting in induction of potent immune-stimulatory DCs that promote robust cellular immunity and therefore provide guidance for making and using effective immune-stimulatory DCs for cell-based vaccination.
Example 7.2.1 NKlR-signaled BMDCs show enhanced survival in vivo
[00115] We analyzed in vivo the effect of signaling via NKlR on the survival of adoptively transferred BMDCs. BMDCs were cultured in complete medium with or without [SaI3MBt(O2)1 ^-Substance P (10~9 M). After 24h, BMDCs were left untreated (DCs, control) or haptenized with trinitrobenzene sulfonic acid (TNBS, 1 mM, 25 min at 37°C) (Sigma) (TN-DCs). DCs and TN-DCs were then labeled with carboxyfluorescein succinimidyl ester (CFSE, 1 μM, 15 min at 37°C) (Invitrogen). After thorough rinsing, CFSE-labeled DCs and TN-DCs were adoptively transferred (2xlO6 cells /50μl PBS, footpad, s.c.) into B6 mice (n=3 animals per group). For in vivo studies analyzing the survival of DCs in the presence or absence of CD40 ligation, mice were treated (or not) with two doses of anti-CD 154 blocking MRl mAb (Bio Express, West Lebanon, NH) injected simultaneously and 2d after BMDC transfer (250 μg per dose, flank, i.p.). At indicated time points, mice were euthanized and local (popliteal) and distant (cervical and axillary) DLNs were dissected. The number of viable CFSE-labeled CDl Ic+ BMDCs in DLNs was analyzed by FACS, as previously described. Mathers et al., 2007, J. Immunol. 178: 7006-7017. An equal number of CDl Ic+ DCs (106) per experimental situation was acquired by FACS and the percentage of CFSE-labeled CDl Ic+BMDCs was calculated with the following formula: [(number of CFSE+CDl Ic+ BMDCs) / (total number of CDl Ic+ DCs) xlOO].
[00116] CFSE-labeled BMDCs, preincubated or not with
Figure imgf000035_0001
Substance P (SarSP-DCs and DCs, respectively) were injected (footpad, s.c.) into B6 mice, and the number of viable CFSE-labeled CDl Ic+ BMDCs was quantified in local DLNs for a period of 7d by FACS. Two days after adoptive transference, similar numbers of SarSP-DCs and control DCs were detected in DLNs, indicating that both DC subsets displayed equivalent DLN-homing capacities. Five days after injection, 95% of SarSP-DCs remained in DLNs compared to 63% of control DCs and by day seven 42% of SarSPDCs were present vs. 24% of control DCs. No CFSE dilution was detected in CDl Ic+ BMDCs homed to local DLNs, indicating that the higher percentage of SarSP-DCs observed in the LNs was due to prolonged DC survival and not DC proliferation. Lyons 2000, J. Immunol. Methods 243: 147-154. Conversely, CFSE+ cells were not detected in non-draining homolateral or collateral LNs. These findings confirm that signaling via NKlR enhances survival of BMDCs in vivo.
[00117] Next, we addressed whether SarSP prolongs survival of Ag-loaded BMDCs in DLNs. CFSE labeled SarSP-DCs or control DCs were left untreated or haptenized with TNBS (SarSP-TNDCs and TN-DCs, respectively), injected (footpad, s.c.) into B6 mice, and quantified by FACS-analysis of DLNs at days 2, 5 and 7 following transfer. Regardless of the treatment, BMDCs showed similar DLN- homing capacities. However, the percentage of TN-DCs detected in DLNs 5 and 7 days after DC administration was significantly lower than the number of non- haptenized DCs (p<0.001) (FIG. 1). Importantly, the percentage of SarSP-TN-DCs homed in DLNs was significantly higher than that of TN-DCs, indicating that signaling via NKlR prolongs the survival of Ag-loaded BMDCs in DLNs. Thus, as contemplated elsewhere herein, a composition of the present invention can be effective as an immunogenic composition as defined herein. For example, as these data suggest, a composition including an immunogenic composition such as an antigen or an immunogenic or other cell, including a dendritic cell, prepared according to the disclosure herein to provide an antigen, and an adjuvant such as Substance P or analog of Substance P can be effective as an immunogenic composition.
Example 7.2.2 Synergistic NKlR and CD40 signaling prolong DC survival in vivo [00118] The finding that SarSP-TN-DCs exhibit prolonged survival once they home in DLNs could be ascribed to sustained expression of anti-apoptotic molecules induced by [SaZMe^O2)1 ^-Substance P or a combined effect of NKlR and CD40 signaling, the later provided by DC-activated CD4+ T cells in DLNs. Josien et al, 2000 J. Exp. Med. 191 : 495-502; Ridge et al., 1998, Nature 393: 474-478; Bennett et al., 1998, Nature 393: 478-480; Schoenberger et al., 1998, Nature 393 480-483; DeSmedt, et al., 1998, J. Immunol. 161 : 4476-4479; Miga et al., 2001, Eur. J. Immunol. 31 : 959-965. Thus, we analyzed the individual and collective contribution of NKlR and CD40 signaling in the expression of Bcl-2 and on the viability of BMDCs in vitro and in vivo, respectively. [00119] For in vitro experiments, BMDCs were pre-treated (or not) with
[Sa^MCt(O2)1 ^-Substance P during the first 24 hours of culture, and signaled with agonistic CD40 mAb added at 24 hours of culture. Expression of Bcl-2 by CDl Ic+ BMDCs was quantified up to 96 hours by FACS. As expected, NKlR signaling significantly increased the percentage of BMDCs expressing Bcl-2 after 24 hours of culture (p<0.001) (FIG. 2), and signaling via CD40 further enhanced Bcl-2 expression in both SarSP-DCs and control DCs 48 hours after culture (p<0.001). By 96 hours, the percentage of BMDCs expressing Bcl-2 diminished regardless of the treatment employed, however, CD40 ligation of SarSP-DCs significantly enhanced the number of cells expressing Bcl-2 vs. DCs (p<0.001), SarSP-DCs (p<0.01) or DCs signaled only via CD40 (p<0.05) (FIG. 2).
[00120] Next, we compared in vivo the survival of adoptively transferred BMDCs homed in DLNs in the presence or absence of CD40-CD40L (CD 154) interaction blockade. CFSE labeled TN-DCs or SarSP-TN-DCs were injected (footpad, s.c.) into B6 mice treated or not with CD 154 (CD40L) blocking MRl mAb (250 μg, i.p.) simultaneously and 2 days after DC administration. In vivo blockade of the CD40- CD 154 interaction decreased the percentage of transferred TN-DCs and SarSP-TN- DCs in DLNs 5 days after injection. Nevertheless, mice that were non-treated with CD 154 blocking mAb had a significantly higher number of SarSP-TNDCs remaining in DLNs compared to TN-DCs. Together, these results suggest that the prolonged viability of BMDCs homed in DLNs was caused by a synergistic effect of NKlR and CD40 signaling.
Example 7.2.3 BMDCs signaled via NKlR promote enhanced cellular immunity
[00121] The previous results indicate that the combination of NKlR and CD40 signaling enhances the interaction of BMDCs with T cells in DLNs, which might result in robust immune responses. We then compared the capability of TN-DCs and SarSP-TN-DCs to induce DTH responses. [00122] B6 mice (n=6 animals per group) were sensitized with one dose of BMDCs (2xlO6 cells / 50μl PBS, footpad, s.c.) pre-treated as follows: i) TNBS haptenized (TN-DCs); ii) TN-DCs cultured with [SaI3MBt(O2)1 ^-Substance P (10~9 M) (SarSP-TN-DCs); iii) TN-DCs cultured with CD40 mAb (CD40-TN-DCs); iv) non-haptenized DCs (DCs); or v) non-haptenized DCs cultured with [Sar9Met(θ2)π]- Substance P (SarSP-DCs). Elicitation of DTH was performed 6d after sensitization by applying trinitrochlorobenzene (TNCB, 1% in acetone:olive oil, 4:1) (Sigma) on the dorsal surface of the ears. The severity of the DTH responses was assessed by comparing the thickness of ears measured prior and after elicitation for up to 6d, using an electronic caliper (Mitutoyo, Aurora, IL). The DTH responses were expressed as the percent in ear thickness increase using the formula: [(thickness of challenged ear - thickness of control ear) / (thickness of control ear) x 100]. For histological analysis, mice were euthanized 6d after elicitation and samples from ear skin were fixed in 4% formaldehyde, embedded in paraffin, stained with H&E, and analyzed by microscopy or embedded in Tissue-Tek OCT (Miles Laboratories, Elkhart, IN) and snapfrozen in methyl-butane (Sigma). The composition of the cellular infiltrate 6d after elicitation was assessed by immunofluorescence microscopy of cryostat sections of the ears, as previously described. Mathers et al., 2007, J. Immunol. 178: 7006-7017. Cryostat sections (8 μm) were mounted on slides pre -treated with Vectabond (Vector Laboratories, Burlingame, CA), fixed in cold 96% ethanol, and incubated with Alexa Fluor 488-CD4 mAb (Invitrogen) and biotin-CD8 (eBioscience, San Diego, CA) or biotin-F4/80 mAbs (Invitrogen), followed by Cy3-streptavidin (Jackson). Cell nuclei were stained with DAPI.
[00123] B6 mice were sensitized by administration of TN-DCs or SarSP-TN-DCs (footpad, s. c.) and DTH was elicited 5 days after by topical TNCB application to the dorsal skin of the ears. Mice were injected with haptenized CD40-TN-DCs or non- haptenized (untreated or SarSP-signaled) DCs to be included as positive and negative controls, respectively.
[00124] The effector cellular immune response was analyzed by measuring the increase of ear thickness up to 6 days post-elicitation (FIG. 3). As expected, mice sensitized with TN-DCs, SarSP-TN-DCs, and CD40-TN-DCs exhibited a significant ear thickness increase vs. control mice lday post-elicitation (p<0.001). In mice sensitized with TN-DCs, the ear thickness diminished significantly 2 days after elicitation, and showed minimal differences vs. control mice 3 days post-elicitation. Conversely, mice sensitized with SarSP-TN-DCs and CD40-TN-DCs exhibited a further increase 2 days after elicitation, and showed a significant prolonged increase in ear thickness in comparison to TN-DCs and control mice for up to 6 days. [00125] We also characterized the cellular infiltrate in the ears of mice obtained 6 days after elicitation by microscopic analysis. Mice sensitized with TN-DCs presented scarce cellular infiltrate, whereas mice sensitized with SarSP-TN-DCs exhibited severe inflammatory infiltrate composed of mononuclear cells. Immuno fluorescence- microscopy analysis revealed that the enhanced cellular inflammatory infiltrate consisted of epidermal CD8+ T cells and dermal CD4+ T cells and F4/80+ macrophages, demonstrating that the immune response generated was a type-1 delayed type hypersensitivity (DTH) response. 7.3 Example 3. Effects of 'fSar9Met(O^nl-Substance P on melanoma [00126] Efficient anti-tumor immune responses are characterized by generation of specific anti-tumor CD8+ (cytotoxic T lymphocytes) CTLs (TcI) and CD4+ ThI cells. Use of ThI -polarizing adjuvants in the skin induces the local secretion of pro- inflammatory mediators required to fully activate skin dendritic cells (DCs), polarizing them to induce ThI- and TcI -biased immunity, and promote development of CTLs, generating an effective and long-lasting anti-tumor immune response. As shown through the examples below, the introduction of transgenic melanoma antigens into the skin in the presence of a Th-I driving adjuvant such as SarSP generates fully activated skin DCs with the ability to stimulate efficiently tumor antigen specific T cells. Skin-derived DCs generated under these conditions bias the differentiation of naϊve tumor- specific T cells into Th-I cells and promote development of cytotoxic T lymphocytes (CTLs), generating an effective and long-lasting anti-tumor immunity.
Example 7.3.1. Mouse model assessment of SarSP vaccine approach demonstrates achievement of long lasting cellular immunity
[00127] In order to assess the viability of a vaccine against the development of primary melanoma tumors utilizing the adjuvant effects of an NKlR agonist, the development of cellular effector immunity against the self melanoma Ag TRP -2 was analyzed. As shown by the following experiments, SarSP increases the effector cellular immune responses and reduces and delays the growth of melanoma in mice when administered with TRP -2.
[00128] To analyze development of cellular immune responses against the TRP-2 melanoma self antigen, the immune response in mouse ears was assessed. Transgenic TRP-2 was delivered by gene gun trans fection (GG) in the dorsal side of mouse ears (one dose). Measurements of % of ear thickness increase resulting from inflammation from an immune response were taken at 24h, 48h, and 72h. As shown in FIG. 4, mice immunized with TRP-2 in the presence of SarSP elicit a significant higher delayed- type hypersensitivity (DTH) response compared to mice immunized only with the GG, as shown by the increased thickness of mouse ears at 72h for those receiving SarSP as compared to the other groups.
[00129] To determine the feasibility of preventing primary melanoma by a vaccine approach, C57BL6 mice were divided into 6 groups of 6 mice per group. Group 1 received no vaccine. Group 2 was administered the irrelevant transgenic antigen luciferase in plasmid form via GG in two shots of approximately 2 μg of plasmid/shot. Group 3 received via GG plasmids encoding the melanoma self Ag TRP -2 in two shots of approximately 2 μg of plasmid/shot. Group 4 received via GG the TRP-2 antigen in two shots of approximately 2 μg of plasmid/shot as well as an NKlR antagonist L733060 administered intradermally at a dose of 5 nmol. Group 5 received via GG the TRP-2 antigen in two shots of approximately 2 μg of plasmid/shot with the NKlR agonist [Sar9Met (O2)11J-SP (SarSP) being administered intradermally at a dose of 5 nmol. Group 6 received three administrations of TRP-2 antigen via GG (approximately 2 ug/shot), the first administration simultaneously with the NKlR agonist [Sar9Met (O2)11J-SP (SarSP) administered intradermally at a dose of 5 nmol, and in between the remaining two TRP-2 antigen GG doses, the mice were dosed intradermally again with SarSP at days 3, 9, and 16 at 5 nmol per dose. Mice were challenged at day 21 after the initial immunization with one dose of the highly aggressive syngeneic B 16/FO malignant melanoma cells injected intradermally in the lower flank (105cells/injection) and tumor growth and mouse survival were analyzed. The data from this experiment are shown in FIGS. 5-7, which demonstrate that a significant delay in tumor growth and mouse survival was observed in those mice receiving both 1 and 2 doses of SarSP and TRP-2. [00130] For example, as shown in FIG. 6, both GG TRP-2 and GG TRP-2 plus NKlR agonist treatments (SarSP) inhibit tumor growth significantly as compared to GG encoding irrelevant Ag and non-immunized mice injected with B 16/FO cells. There was a significant inhibition of tumor growth in mice treated with GG pCMV- TRP -2 plus NKlR agonist compared to GG TRP2 alone. For example, mice receiving both GG pCMV-TRP-2 had final tumor areas approximately 16 times smaller than those of naϊve mice, and still even approximately 5 times smaller than mice receiving only TRP-2 based on visual inspection.
[00131] Further, as shown in FIG. 5, visual inspection of melanoma tumors on the abdomens of sacrificed mice reveals that mice receiving one or 2 doses of the TRP- 2/SarSP vaccination had tumors significantly reduced in size compared to all other groups tested. [00132] In addition, as shown in the survival curve in FIG. 7, an indefinite survival of 45% and of 50% is seen for mice injected either with one dose or with two doses of SarSP respectively, while other mice groups were completely deceased well before 40 days from the initial immunization. Accordingly, GG-TRP-2 plus one or two doses of the NKlR agonist prolonged significantly the survival of mice compared to GG-TRP- 2 alone or with GG encoding an irrelevant Ag and non-immunized mice injected with B 16/FO cells.
[00133] 100 days following the administration of tumor cells and 120 days following the initial immunizations, mice surviving from melanoma tumors were subject to a DTH assay. Results are shown in FIG. 8. Mice were challenged with GG delivered pCMV-TRP-2 in the dorsal side of the ears. Percent of ear thickness increase was measured at 24, 48, and 72 hours after GG administration. As shown in FIG. 8, mice that were initially administered SarSP NKlR agonist and TRP-2 exhibited the greatest percent increase in ear thickness. This indicates that the vaccine was still effective in surviving mice at 4 months (12Od) post immunization and suggests the development of long lasting effector cellular immunity resulting from vaccination with TRP-2 and SarSP.
[00134] Surviving mice subject to the DTH assay remained tumor free and were sacrificed 120 days after immunizations. [00135] Accordingly, as these data suggest, the compositions and methods of the invention as described in the above examples and contemplated elsewhere herein are useful for treating, preventing, or ameliorating cancer in a patient, including such methods as administering to a patient Substance P or a Substance P analog as well as a tumor specific antigen according to any of the variety of methods contemplated herein. In particular, these data also suggest that an immune response to a melanoma tumor can be enhanced by administering a melanoma antigen and a Substance P analog or Substance P.
Example 7.3.2. Development of local and systemic protective immunity by genetic immunization in the skin in the presence of Thl-driving adjuvants. Example 7.3.2 A. Induction of TRP-2 specific CTL responses by skin GG- immunization in the presence of Thl-driving adjuvants [00136] Cytotoxic activity of CD8+ T cells against TRP-2 Ag induced under experimental conditions is assessed by CTL assays. Briefly, B6 mice are GG- immunized with pCMV-TRP-2 in the absence (control) or the presence of Imiquimod or SarSP-NKla. Experimental groups of 3 mice each are treated as follows: Group 1 receives no GG transfection and receives PBS as a control in lieu of an adjuvant; Group 2 receives pCMV-TRP-2 via GG and receives no adjuvant; Group 3 receives pCMB-TRP-2 via GG and 5nM SarSP-NKla adjuvant; Group 4 receives pCMV- TRP -2 by GG and 5% v/v Imiquimod adjuvant; Group 5 receives no transfection and receives 5 nM SP-NKIa adjuvant; and Group 6 receives no transfection and receives 5% v/v Imiquimod adjuvant.
[00137] After GG-immunization on day 1, mice are boosted on days 7 and 14. On day 21, mice are euthanatized and suspensions of spleen and lymph nodes cells are prepared. CTL assays are performed. Briefly, splenocytes and lymph node cells (~3xlO7) are restimulated with TRP-2 protein and TRP-2180"188 peptide (20ng/ml) in medium plus rhIL-2 (30 U/ml) and used as effector cells for CTL assays. After 5 days, effector cells are cocultured with 51Cr-labeled EL4 cells (syngeneic targets) ± pulsed with TRP-2 " peptide (100ng/ml) at different effector/target cell ratios. After 5 hours, the amount Of 51Cr release into the 100 μl of culture supernatant is quantified. Percent of specific lysis is calculated with the formula: % SL= [(E)-(Sm)/ (M)-(Sm)] x 100, where SL: specific lysis, E: mean of release from 3 wells, Sm: mean Of 51Cr spontaneous minimum release, M: 51Cr maximum release.
[00138] To determine the role of different effector T cell populations, the same CTL experiment is performed after depletion of CD4+ T or CD8+ T cells by incubation with anti-Thy-1 mAb (M5/49), or anti-CD8 (ADH4) or anti-CD4 (GK-I) mAbs followed by lysis by complement. Four independent experiments are performed for statistical significance. Means of percent of cell lysis and SD are compared by ANOVA with appropriate data transformation. A value of p<0.05 is considered significant.
[00139] The data show that CD8 T cells harvested from mice administered a composition of the present invention comprising an adjuvant (SarSP) and the exemplary TRP-2 melanoma self antigen exhibit the greatest cytotoxic activity as compared to other groups tested, thereby confirming effectiveness as a vaccine composition. CD8 T cells harvested from mice administered TRP-2 with the SarSP adjuvant have at least the same activity as CD8 T cells harvested from mice administered TRP-2 with the Imiquimod adjuvant.
Example 7.3.2B. Induction of local protective immunity [00140] This experiment tests whether GG-immunization in combination with ThI- driving adjuvants induces local protective immunity against melanoma. To address this question, mice are GG-immunized in the skin with pCMV-TRP-2a in the absence (control) or the presence of Imiquimod or SP-NKIa adjuvant. Experimental and control groups of 5 mice each are as follows: Group 1 — naϊve control mice with no GG transfection, no adjuvant (PBS control); Group 2 — no GG transfection, 5 nM Sp- NKIa adjuvant; Group 3 — no GG transfection, 5% v/v Imiquimod; Group 4 pCMV- mTRP-2 GG transfection with no adjuvant; Group 5 pCMV-mTRP-2 with 5 nM SP- NKIa adjuvant; and Group 6 — pCMV-mTRP-2 GG transfection with 5% v/v Imiquimod adjuvant. All groups are challenged with 2x105 B16 cells. Priming immunizations on day 1 are followed by two boostings (days 7 and 14). At day 21, mice are challenged locally by intradermal injection in the flanks with syngeneic B16 melanoma cells expressing the self- Ag TRP-2. The dose of cells is 2-10 times the LD50 (lethal dose to 50%). The tumor size is monitored 3 times per week along with animal survival. Tumor size is assessed as mean of the tumor area (mm2) ± ISD. Survival is assessed as the percentage of surviving animals.
[00141] The data indicate that mice receiving the Substance P or Imiquimod adjuvant in addition to the GG immunization of pCMV-mTRP-2 exhibit statistically significant prolonged survival rates over the other groups of mice. It is also observed that the mean tumor area of mice receiving the Substance P or Imiquimod adjuvant in addition to the GG immunization of pCMV-mTRP-2 have average tumor sizes that are statistically significantly smaller than those of the other groups tested. Again, these data confirm that, when used in accordance with the teachings provided herein, treatment with a composition of the present invention comprising an adjuvant (SarSP) and the exemplary TRP-2 melanoma self antigen results in modulation of tumor progression, thereby confirming effectiveness as a vaccine composition. Example 7.3.2C. Induction of systemic protective immunity against melanoma using senetic immunization in the presence of ThI -adjuvants:
[00142] This experiment demonstrates that GG-immunization in combination with administration of ThI -driving adjuvants induces generalized immune protection and prevents development of melanoma metastasis. This experiment uses the same six (6) experimental groups as Example 7.3.2B with five (5) mice per group. Mice are immunized at day 1 with boosters administered at day 7 and day 14. At day 21, B 16 melanoma cells are injected intravenously (2xlO5 cells/ 200μl PBS/mouse) to permit homing of tumor cells in the lungs and liver of mice. Animals are euthanized 14 and 21 days after injection of tumor cells and the presence of melanoma metastatic nodules are quantified (number and size) by macroscopic examination and confirmed by microscopy.
[00143] The data show that mice receiving the Substance P or Imiquimod adjuvant in addition to the GG immunization of pCMV-mTRP-2 have statistically fewer and smaller metastatic nodules present in the lungs and as compared to mice in the other groups tested, indicating that the mice have a level of systemic protective immunity against melanoma resulting from the vaccine of the present invention. Once again, these studies confirm that, when used in accordance with the teachings provided herein, treatment with a composition of the present invention comprising an adjuvant (SarSP) and an exemplary cancer antigen results in modulation of tumor progression, thereby confirming effectiveness as a cancer vaccine composition.
Example 7.3.2D. Induction of melanoma immunotherapy by genetic immunization in the presence ofThl-driving adjuvants
[00144] This experiment shows that GG-immunization in combination with ThI- driving adjuvants induces tumor immunotherapy against melanoma. Groups of B6 mice, according to the groupings as in Example 7.3.2 A, are injected intradermally in the flanks with 2 x 105 syngeneic B16 melanoma cells expressing the self- Ag TRP-2. Three days after B 16 cell injections, mice are GG-immunized in the skin with pCMV- TRP -2a alone (control), after local pre-treatment with Imiquimod, SP-NKIa or its vehicles (control); or left untreated (control). Tumor size and animal survival are evaluated as described in Example 7.3.2B. Five mice per group are used. Three independent experiments are performed. The protocol is repeated again, except that the genetic immunization protocol is initiated 7 days after tumor injection, instead of at 3 days to observe the efficacy of immunotherapy in more advanced tumors.
[00145] The data show that mice receiving the adjuvant SarSP or Imiquimod and pCMV-TRP-2a have tumor volumes that are statistically significantly smaller than those in the other groups tested both when the immunization protocol is initiated at either 3 days and at 7 days. Once again, these studies confirm that, when used in accordance with the teachings provided herein, treatment with a composition of the present invention comprising an adjuvant (SarSP) and an exemplary cancer antigen results in modulation of tumor progression, thereby confirming effectiveness as a cancer vaccine composition.
Example 7.3.2E Induction of long lasting immunity to melanoma by genetic immunization in the presence of Th-I driving adjuvants
[00146] Mice treated according to any of the methods in Examples 7.3.2A-D are monitored for long lasting effector cell memory. Surviving animals are monitored for the presence of specific Ab production to TRP-2 melanoma antigen 6 and 9 months after immunization and CTL assays are performed 9 months after immunization.
[00147] The results indicate that mice treated with the SarSP or Imiquimod adjuvant coupled with TRP-2 antigen have antibodies to melanoma antigen present at both 6 and 9 months after immunization, while no surviving mice from other groups has any significant antigen quantity present. CTL assays performed reveal that mice treated with the SarSP or Imiquimod adjuvant coupled with TRP-2 antigen also have CTL memory cells such as CD4+ and CD8+ memory T cells against TRP-2 antigen, while no surviving mice from the other groups has any significant quantity of memory cells present. The data suggest that the vaccination approach provides long lasting cellular immunity to TRP-2 antigen. Once again, these studies confirm that, when used in accordance with the teachings provided herein, treatment with a composition of the present invention comprising an adjuvant (SarSP) and an exemplary cancer antigen results in modulation of tumor progression, thereby confirming effectiveness as a vaccine composition. Example 7.3.2F. Prophetic study investigating use of an immunogenic composition of the present invention for treatment, prevention or amelioration of cancer in a human subject, including use for reducing the risk of developing cancer. [00148] Any and all of the following studies, when conducted, will be done so in compliance with all applicable FDA and IRB (Institutional Review Board) requirements. Human subjects having a cancer or at risk for developing a cancer will be treated with an immunogenic composition as defined herein. Certain such subjects will have, or will be at risk of developing, melanoma. An exemplary composition will comprise Substance P or a Substance P analog. A particularly preferred composition will comprise SarSP. It is expected that existing cancers in human subjects treated in accordance with the methods described elsewhere herein will be ameliorated to a statistically significant extent; and the risk of developing a cancer will be reduced to a statistically significant extent following treatment in those deemed at risk.
[00149] Accordingly, as these data suggest, the compositions and methods of the invention as described in the above examples and contemplated elsewhere herein are useful for treating, preventing, or ameliorating cancer in a patient, including such methods as administering to a patient Substance P or a Substance P analog as well as a tumor specific antigen according to any of the variety of methods contemplated herein. In particular, these data confirm that an immune response to a cancer tumor antigen, such as but not limited to a melanoma tumor antigen can be enhanced by administering a melanoma antigen and a Substance P analog or Substance P. Once again, these studies confirm that, when used in accordance with the teachings provided herein, treatment with a composition of the present invention comprising an adjuvant (SarSP) and an exemplary antigen results in modulation of tumor progression, thereby confirming effectiveness as a vaccine composition.
[00150] Various embodiments have been described. The descriptions and examples are intended to be illustrative of the invention and not limiting. Indeed, it will be apparent to those of skill in the art that modifications may be made to the various embodiments described without departing from the spirit of the invention or scope of the appended claims set forth below. All references cited herein are incorporated herein by reference in their entireties for all purposes.

Claims

INTERNATIONAL PATENT APPLICATION Attorney Docket No.: IRB-013PCWhat is claimed is:
1. A composition for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer, the composition comprising an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog, wherein the Substance P analog is of Formula (I):
Z 1 -Xaal -Xaa2-Xaa3-Xaa4-Xaa5-Xaa6-Xaa7-Xaa8-Xaa9-Xaal O-Xaal 1 -Z2
(I) (SEQ ID NO: 11) or a pharmaceutically acceptable salt thereof, wherein:
Xaal is Arg, Lys, 6-N methyllysine or (6-N, 6-N) dimethyllysine;
Xaa2 is Pro or Ala;
Xaa3 is Lys, Arg, 6-N-methyllysine or (6-N, 6-N) dimethyllysine;
Xaa4 is Pro or Ala;
Xaa5 is GIn or Asn;
Xaa6 is GIn or Asn;
Xaa7 is Phe or Phe substituted with chlorine at position 2, 3 or 4;
Xaa8 is Tyr, Phe, or Phe substituted with chlorine at position 2, 3 or 4;
Xaa9 is GIy, Pro, Ala or N-methylglycine;
XaalO is Leu, VaI, He, Norleucine, Met, Met sulfoxide, Met sulfone, N- methylleucine, or N-methylvaline;
Xaal 1 is Met, Met sulfoxide, Met sulfone, or Norleucine;
Zl is R2N- or RC(O)NR-;
Z2 is -C(O)NR2 or -C(O)OR or a salt thereof; each R is independently R is H, (Cl -C6) alkyl, (Cl -C6) alkenyl, (Cl -C6) alkynyl, (C5 -C20) aryl, (C6 -C26) alkaryl, 5-20 membered heteroaryl or 6-26 membered alkheteroaryl; and each "— " between residues Xaal through Xaal 1 independently designates an amide linkage, a substitute amide linkage or an isostere of an amide.
2. The composition of claim 1, wherein the antigen is a dendritic cell-based antigen selected from the group consisting of: tumor-dendritic cell hybrids; peptide- loaded dendritic cells; whole tumor cell-loaded dendritic cells; tumor mRNA-loaded dendritic cells; and genetically engineered or gene-loaded dendritic cells.
3. The composition of claim 1, wherein the antigen is a dendritic cell-based antigen resulting from a dendritic cell loaded with an antigen from a tumor; a dendritic cell exposed to a tumor lysate; a dendritic cell gene-loaded ex vivo; or a dendritic cell exposed to a tumor antigen ex vivo.
4. The composition of claim 1, which is a dendritic cell vaccine.
5. The composition of claim 1, wherein the antigen is a cancer tumor antigen.
6. The composition of claim 5, wherein the cancer tumor antigen is an antigen selected from the group consisting of: a solid adenocarcinoma, a pancreatic cancer, a breast cancer, a gastric cancer, a renal cell cancer, a colorectal cancer, a melanoma, a lympho-hematopoietic cancer, a lymphoma, a Non-Hodgkin's lymphoma, a leukemia, mucin 1 , HER2/NEU, melanoma-associated antigen, mammaglobin, carcinoembryonic antigen, and cyclin Bl.
7. The composition of claim 5, wherein the cancer tumor antigen is selected from the group consisting of: autologous or allogenic tumor cell antigen; tumor cell lysate antigen; shed antigens from autologous tumor cells; heat shock proteins from autologous tumor cells; tumor-associated antigens (TAA); shared tumor antigen, unique tumor antigen, inactivated whole tumor cell antigen; and gene -modified cell antigen.
8. The composition of claim 1, is a melanoma vaccine.
9. The composition of claim 1, wherein the antigen and the Substance P or Substance P analog are in combination.
10. The composition of any one of the preceding claims wherein the antigen and the Substance P or the Substance P analog are for contemporaneous administration.
11. The composition of claim 1 , wherein the antigen and the Substance P or the Substance P analog are in separate, divided or undivided containers.
12. The composition of claim 11 , wherein the antigen and the Substance P or the Substance P analog are for sequential administration.
13. The composition of any one of the preceding claims wherein the amelioration of cancer comprises reducing tumor load, slowing development of tumors or reducing severity when compared to the antigen alone.
14. The composition of any one of the preceding claims which is a pharmaceutical composition.
15. A method of treating cancer in a subject having cancer or preventing cancer in a subject at risk for developing cancer, comprising providing to said subject an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog wherein the Substance P analog is of Formula
(I):
Zi-Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^Xaa^-Xaa11 -Z2 (I)
(SEQ ID NO: 11) or a pharmaceutically acceptable salt thereof, wherein:
Xaal is Arg, Lys, 6-N methyllysine or (6-N, 6-N) dimethyllysine;
Xaa2 is Pro or Ala;
Xaa3 is Lys, Arg, 6-N-methyllysine or (6-N, 6-N) dimethyllysine;
Xaa4 is Pro or Ala;
Xaa5 is GIn or Asn;
Xaa6 is GIn or Asn;
Xaa7 is Phe or Phe substituted with chlorine at position 2, 3 or 4;
Xaa8 is Tyr, Phe, or Phe substituted with chlorine at position 2, 3 or 4;
Xaa9 is GIy, Pro, Ala or N-methylglycine;
XaalO is Leu, VaI, He, Norleucine, Met, Met sulfoxide, Met sulfone, N- methylleucine, or N-methylvaline;
Xaal 1 is Met, Met sulfoxide, Met sulfone, or Norleucine;
Zl is R2N- or RC(O)NR-;
Z2 is -C(0)NR2 or -C(O)OR or a salt thereof; each R is independently R is H, (Cl -C6) alkyl, (Cl -C6) alkenyl, (Cl -C6) alkynyl, (C5 -C20) aryl, (C6 -C26) alkaryl, 5-20 membered heteroaryl or 6-26 membered alkheteroaryl; and each "— " between residues Xaal through Xaal 1 independently designates an amide linkage, a substitute amide linkage or an isostere of an amide.
16. The method of claim 15, wherein the Substance P analog is further defined as:
Xaa1 is Arg;
Xaa2 is Pro;
Xaa3 is Ly s;
Xaa4 is Pro;
Xaa5 is GIn;
Xaa6 is GIn;
Xaa7 is Phe or Phe substituted with chlorine at position 4;
Xaa8 is Tyr, Phe, or Phe substituted with chlorine at position 4;
Xaa9 is GIy, Pro or N-methylglycine;
XaalO is Leu; and
Xaal 1 is Met, Met sulfoxide, Met sulfone or Norleucine.
17. The method of claim 15, wherein the Substance P analog is further defined as: the "— " between residues Xaa1 through Xaa11 designates
-C(O)NH-; Zi is H2N-; and Z2 is -C(O)NH2.
18. The method of claim 15, wherein the Substance P analog is selected from the group consisting of:
RPKPQQFFGLM (SEQ ID NO.: 1);
RPKPQQFFGLNIe (SEQ ID NO. : 2);
RPKPQQFFPLM (SEQ ID NO. : 3);
RPKPQQFFMeGIyLM (SEQ ID NO. : 4);
RPKPQQFTGLM (SEQ ID NO. : 5);
RPKPQQF(4-C1)F(4-C1)GLM (SEQ ID NO. : 6);
RPKPQQFFGLM(O) (SEQ ID NO. : 7);
RPKPQQFFMeGIyLM(O) (SEQ ID NO. : 8); RPKPQQFFGLM(O2) (SEQ ID NO. : 9); or
RPKPQQFFMeGIyLM(O2) (SEQ ID NO.: 10).
19. The method of claim 15, wherein the Substance P analog is: Zi-RPKPQQFFMeGlyLM(O2)-Z2, wherein Zx is NH2 and Z2 is C(O)NH2.
20. The method of any one of claims 15-19, wherein the cancer is a solid adenocarcinoma (for example, being selected from pancreatic, breast, gastric, renal cell and colorectal cancer), melanoma or a lympho-hematopoietic cancer (for example, being selected from lymphoma, Non-Hodgkin's lymphoma and leukemia).
21. The method of any one of claim 15-19, wherein the cancer to be treated or prevented is cervical, liver, breast, pancreas, colon, lung, ovarian, endometrial, fallopian tube or prostate and the increased risk is human papillomavirus infection, hepatitis B, PanIN, BRCAl, BRC A2, PSA, familial adenomatous polyposis syndrome (FAP) or hereditary non-polyposis colorectal cancer syndrome (HNPCC).
22. The method of any one of claim 15-19, wherein the antigen is selected from: a dendritic cell based antigen; a shared tumor antigen; a unique tumor antigen; inactivated whole tumor cells; gene-modified tumor cells or is comprised of mucin 1, HER2/NEU, melanoma-associated antigen, mammaglobin, carcinoembryonic antigen or cyclin Bl.
23. The method of any one of claim 15-22, wherein the immunogenic composition stimulates: (a) Natural killer tumor (NKT) cells, natural killer (NK) cells or B- lymphocytes; (b) antibody-dependent cellular cytotoxicity, or (c) THl cell activity (for example, stimulating T lymphocyte production or activity or cytotoxic T lymphocyte (CTL) activity (for example, the T lymphocyte cell being selected from CD4+ cells or CD4+CD25+ cells; for example, the T lymphocyte activity being selected from expression of CCR7, CD45, CCRlO or cutaneous leukocyte antigen; and for example, the CD45 being selected from the CCR7+CD45RO+ isoform)).
24. The method of any one of claim 15-23, wherein the numbers of cancer cells in the subject are decreased.
25. The method of any one of claims 15-24, wherein the antigen is the subject's endogenous antigen.
26. A composition comprising:
Substance P or an analog thereof; a melanoma-associated antigen; and a pharmaceutically acceptable carrier.
27. A method of enhancing an immune response to a melanoma tumor in a subject having a melanoma tumor or at risk of having a melanoma tumor comprising: providing Substance P or an analog thereof to the patient; and providing a melanoma-associated antigen.
28. The method of claim 27, wherein the Substance P or analog thereof is provided coincident with, prior to or subsequent to antigen.
29. The method of claim 27, wherein the Substance P or analog thereof is provided intradermally or topically.
30. The method of claim 29, wherein an effective amount of antigen is provided to the patient biolistically.
31. The method of claim 30, wherein an effective amount of antigen is provided as plasmid encoding the antigen.
32. The method of claim 27, wherein either or both of the providing steps are repeated.
33. The method of claim 27, wherein the Substance P or analog thereof and the antigen are provided separately but simultaneously.
34. The method of claim 27, wherein the antigen is the subject's endogenous antigen.
35. A composition for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer, the composition comprising an immunogenic composition comprising a cell, an antigen and an adjuvant effective amount of Substance P or a Substance P analog.
36. The composition of claim 35, wherein the cell is selected from the group consisting of: an antigen processing cell; a cell engineered to express antigen; a cell engineered to express Substance P or a Substance P analog; and combinations of any one or more of the foregoing cells.
37. The composition of claim 36, wherein the cell is the immunogenic composition.
38. The composition of claim 35, wherein the cell is endogenous to the subject.
39. A method for use in the treatment, prevention or amelioration of cancer in a subject, or for reducing the likelihood of developing cancer in a subject at risk for developing cancer, the method comprising the step of providing the composition of claim any one of claims 35-38 to a subject in need thereof.
40. Use of an immunogenic composition comprising an antigen and an adjuvant effective amount of Substance P or a Substance P analog for the manufacture of a medicament for use in a method as defined in any one of the preceding claims.
41. The use of the immunogenic composition of claim 40, wherein the composition further comprises a cell.
PCT/US2009/041022 2008-04-17 2009-04-17 Substance p and analogs thereof as a cancer immunogenic composition adjuvant WO2009129498A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4585608P 2008-04-17 2008-04-17
US61/045,856 2008-04-17

Publications (2)

Publication Number Publication Date
WO2009129498A2 true WO2009129498A2 (en) 2009-10-22
WO2009129498A3 WO2009129498A3 (en) 2009-12-23

Family

ID=41100681

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/041022 WO2009129498A2 (en) 2008-04-17 2009-04-17 Substance p and analogs thereof as a cancer immunogenic composition adjuvant

Country Status (1)

Country Link
WO (1) WO2009129498A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110280897A1 (en) * 2008-07-25 2011-11-17 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Polypeptides and uses thereof
WO2020241972A1 (en) * 2019-05-28 2020-12-03 주식회사 바이오솔루션 Cosmetic composition comprising substance p for skin whitening

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001024822A2 (en) * 1999-10-01 2001-04-12 Cistem Biotechnologies Gmbh Pharmaceutical composition comprising an antigen
WO2002010194A1 (en) * 2000-07-31 2002-02-07 Dabur Research Foundation Substance p analogs for the treatment of cancer
WO2003102023A1 (en) * 2002-05-29 2003-12-11 Immatics Biotechnologies Gmbh Tumour-associated peptides that bond to mhc molecules

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001024822A2 (en) * 1999-10-01 2001-04-12 Cistem Biotechnologies Gmbh Pharmaceutical composition comprising an antigen
WO2002010194A1 (en) * 2000-07-31 2002-02-07 Dabur Research Foundation Substance p analogs for the treatment of cancer
WO2003102023A1 (en) * 2002-05-29 2003-12-11 Immatics Biotechnologies Gmbh Tumour-associated peptides that bond to mhc molecules

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LAMBRECHT B N ET AL: "Endogenously produced substance P contributes to lymphocyte proliferation induced by dendritic cells and direct TCR ligation." EUROPEAN JOURNAL OF IMMUNOLOGY DEC 1999, vol. 29, no. 12, December 1999 (1999-12), pages 3815-3825, XP002548220 ISSN: 0014-2980 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110280897A1 (en) * 2008-07-25 2011-11-17 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Polypeptides and uses thereof
US8748170B2 (en) * 2008-07-25 2014-06-10 University of Pittsburgh—of the Commonwealth System of Higher Education Polypeptides derived from cyclin B1 and uses thereof
WO2020241972A1 (en) * 2019-05-28 2020-12-03 주식회사 바이오솔루션 Cosmetic composition comprising substance p for skin whitening
KR20200136641A (en) * 2019-05-28 2020-12-08 주식회사 바이오솔루션 Cosmetic composition comprising substance P for whitening skin
KR102225547B1 (en) 2019-05-28 2021-03-10 주식회사 바이오솔루션 Cosmetic composition comprising substance P for whitening skin

Also Published As

Publication number Publication date
WO2009129498A3 (en) 2009-12-23

Similar Documents

Publication Publication Date Title
Heine et al. Clinical and immunological effects of mRNA vaccines in malignant diseases
Wong et al. Advances in therapeutic cancer vaccines
JP6134763B2 (en) Dendritic cells that have been produced using GM-CSF and interferon α and that have taken up cancer cells that have been heat-treated and killed
Sheng et al. Cancer immunotherapy and nanomedicine
JP5239041B2 (en) Cancer treatment
JP2006523688A (en) Nucleotide vaccine composition, nucleotide and cell vaccine composition production method, vaccine composition, vaccine composition use, immune response production method, disease treatment or prevention method, kit comprising antigen presenting cells
WO2000006602A1 (en) Cancer antigens based on tumor suppressor gene wt1 product
EP3347098B1 (en) Targeting mda-5 activation for cancer immunotherapy
Morisaki et al. Dendritic cell‐based combined immunotherapy with autologous tumor‐pulsed dendritic cell vaccine and activated T cells for cancer patients: rationale, current progress, and perspectives
EP1292321B1 (en) Method to increase class i presentation of exogenous antigens by human dendritic cells
JP2012097109A (en) Lactoferrin as adjuvant in cancer vaccine
JP6698541B2 (en) Medicament for use in a method of inducing or prolonging a cellular cytotoxic immune response
JP2022553192A (en) cancer vaccine
Bellone et al. Rejection of a nonimmunogenic melanoma by vaccination with natural melanoma peptides on engineered antigen-presenting cells.
WO2009129498A2 (en) Substance p and analogs thereof as a cancer immunogenic composition adjuvant
Murphy Frontiers in cancer immunotherapy
Boliukh et al. Immunological aspects of heat shock protein functions and their significance in the development of cancer vaccines
KR20190066028A (en) Immunogenic compounds for cancer treatment
Dunn et al. Dendritic cells and HNSCC: a potential treatment option?
Cebon et al. Immunotherapy of advanced or metastatic melanoma
den Brok et al. Adapted from: Expert Reviews In Vaccines, 2005 Oct; 4 (5): 699-710
Dranoff Advances in Therapeutic Cancer Vaccines
Shubina et al. Looking into the future of immunotherapy or how to find Cinderella?
Verbekea et al. Messenger RNA vaccination with α-galactosylceramide induces antitumor immunity by boosting cytotoxic and natural killer T cells
Jackson et al. Immunotherapy of cancer

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: 09733491

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09733491

Country of ref document: EP

Kind code of ref document: A2