CA2605749C - Compositions and methods for cancer immunotherapy - Google Patents

Compositions and methods for cancer immunotherapy Download PDF

Info

Publication number
CA2605749C
CA2605749C CA2605749A CA2605749A CA2605749C CA 2605749 C CA2605749 C CA 2605749C CA 2605749 A CA2605749 A CA 2605749A CA 2605749 A CA2605749 A CA 2605749A CA 2605749 C CA2605749 C CA 2605749C
Authority
CA
Canada
Prior art keywords
alkyl
cancer
group
alkoxy
optionally substituted
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CA2605749A
Other languages
French (fr)
Other versions
CA2605749A1 (en
Inventor
Daniel P. Rossignol
Sally T. Ishizaka
Lynn D. Hawkins
Scott Fields
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eisai R&D Management Co Ltd
Original Assignee
Eisai R&D Management Co Ltd
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 Eisai R&D Management Co Ltd filed Critical Eisai R&D Management Co Ltd
Publication of CA2605749A1 publication Critical patent/CA2605749A1/en
Application granted granted Critical
Publication of CA2605749C publication Critical patent/CA2605749C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/57Skin; melanoma

Abstract

The invention relates to immunotherapeutic compounds and to methods for stimulating an immune response in a subject individual at risk for developing cancer, diagnosed with a cancer, in treatment for cancer, or in post-therapy recovery from cancer or the compounds of the invention can be administered as a prophylactic to a subject individual to prevent or delay the development of cancer.

Description

Compositions and Methods for Cancer Immunotherapy Field of the Invention The invention provides compositions and methods for cancer immunotherapy.
1. Background of the Invention Cancer immunotherapy involves the use of compositions and methods to elicit and enhance an individual's own immune system against cancerous cells, or infections that predispose to cancer. Cancer vaccines function by triggering the immune system to mount a response to an antigen (e.g., typically a protein, peptide, or carbohydrate) that is introduced into the body in a non-carcinogenic form and triggers the body to confer immunity or obtain a long-lived "memory" immune response. See, e.g., Kast, Peptide-Based Cancer Vaccines, Landes Bioscience (2000); Stern et al, Cancer Vaccines and Inzinunotherapy, Cambridge University Press (2000). Once the immune system response is established, exposure of the immune system to this antigen (e.g., in the form of a cancerous tumor) results in a rapid and robust immune response.
It is often necessary to enhance the immune response to the antigens present in a vaccine in order to stimulate the immune system to a sufficient extent to make a vaccine effective, i.e., to confer immunity. Many protein, peptide and carbohydrate antigens, administered alone, do not generate a sufficient response to confer immunity. The reasons for this may be that the antigens recognized by cancer reactive immune responses originate from proteins that are expressed in normal tissue of the same histological type as the cancer, such that immunologic tolerance may prevent effective immune responses to the antigens. Such antigens need to be presented to the immune system in such a way that they will generate an immune response. To this end, adjuvants have been devised which immobilize antigens and enhance the immune response. The best known adjuvant, Freund's complete adjuvant, consists of a mixture of mycobacteria in an oil/water emulsion. Freund's adjuvant works (i) by enhancing cell and humoral-mediated immunity and (ii) by blocking rapid dispersal of the antigen challenge (the depot effect). Freund's adjuvant is used primarily with experimental therapies to help stimulate the immune system in animals, and in humans the mycobacterial preparation Bacille Calmette-Guerin (BCG), is an immunotherapy approved as a treatment for bladder cancer.
Another molecule that has been shown to have immunostimulatory or adjuvant activity is endotoxin, also known as lipopolysaccharide (LPS). LPS is also a model adjuvant that can overcome tolerance to self antigens. Waldner, et al., .1.
Gin. Invest., (2004); 113 990-997. While LPS is too toxic to be a viable adjuvant, molecules that are structurally related to endotoxin, such as monophosphoryl lipid A ("MPL"), are being tested as adjuvants in clinical trials. The only FDA-approved adjuvant for use in humans are aluminum salts, alum.
There is a need in the art for safe and effective compositions that can stimulate the immune system as a cancer immunotherapeutic. The invention is directed to this, as well as other, important ends.
2. Summary of the Invention The present invention relates to a composition having (i) at least one immunotherapeutic agent which may be one or more cancer antigens, one or more antigens derived from a virus associated with cancer, one or more anti-cancer antibodies;
and an anti-idiotypic antibody to an anti-cancer antibody; and, (ii) one or more compounds of formulae (1), (11), (III), (IV), and (V) and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
The compounds of the invention can be administered to a subject individual at risk for developing cancer, diagnosed with a cancer, in treatment for cancer, or in post-therapy recovery from cancer or the compounds of the invention can be administered as a prophylactic to a subject individual to prevent or delay the development of cancer.
The invention further relates to methods for stimulating an immune response in a subject individual by (a) administering to the individual at least one immunotherapeutic agent selected from one or more cancer antigens; one or more viral derived antigen associated with cancer; one or more anti-cancer antibodies; and one or more anti-idiotypic antibodies to an anti-cancer antibody; and (b) administering to the individual one or more compound selected from formulae (I), (II), (III), (IV) and (V) and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
The present invention more particularly relates to a composition, comprising:

, , at least one immunotherapeutic agent selected from one or more cancer antigens; one or more antigens from a virus associated with cancer; one or more anti-cancer antibodies;
and one or more anti-idiotypic antibodies to an anti-cancer antibody; and, one or more compounds of formula (I) .,`.... v (CH

., (CI12)a R kt_,rivb I I

I I
MO¨P=¨"T T=P ¨OM
I I

(CH2)d (C112)e (C).¨H2)e' 1r2/W2 X2¨K
i \
WI (C112)d' \ / \

\ /
(CH2)e (MA

/ \

formula (I) wherein:
R1 is:
(a) -C(0)-;
(b) -C(0)-C1_14 alkyl-C(0)- or-C(0)-C1-14 alkenyl-C(0)-;
wherein the -Ci_14 alkyl- or -C1_14 alkenyl- is optionally substituted with one or more substituents selected from the group consisting of hydroxy, 2a C1_6 alkyl, C1_6 alkoxy, C1.6 alkyldioxy, C1-5 alkylamino, carboxy, C1-6 alkoxycarbonyl, C1-6 carbamoyl, C1_6 acylamino, and (aryl)C1-6 alkyl; and wherein the aryl moiety of the (aryl)C1_6 alkyl is optionally substituted with one or more substituents selected from the group consisting of C1-6 alkyl, C1-alkoxy, C1.6 alkylamino, C1_6 alkoxyamino, C1_6 alkylamino-C1.6 alkoxy, -0-C1-6 alkylamino-C1_6 alkoxy, -0-C1-6 alkylamino-C(0)-C1-6 alkyl-C(0)0H, alkylamino-C(0)-C1-6 alkyl-C(0)-C1-6 alkyl, -0-C1.6 alkyl-NH-C -6 alkyl-0-C1-6 alkyl, -0-C1-6 alkyl-NH-C(0)C1-6 alkyl-C(0)0H, and ¨0-C1-6 alkyl-NH-C(0)C1_6 alkyl-C(0)-C1-6 alkyl;
(c) a C2 to C15 straight or branched chain alkyl group optionally substituted with one or more hydroxyl or alkoxy groups; or (d) -C(0)-C6_12 aryl-C(0)- wherein the aryl is optionally substituted with one or more group selected from the group consisting of hydroxy, halo, nitro, amino, C1-6 alkyl and C1_6 alkoxy groups;
a and b are each independently 0, 1, 2, 3 or 4;
d and e are each independently 1, 2, 3, 4, 5 or 6;
d' and e' are each independently 0, 1, 2, 3 or 4;
d" and e" are each independently 0, 1, 2, 3 or 4;
T is oxygen or sulfur;
Xi, X2, Y1 and Y2 are each independently null, oxygen, NH, -N(C(0)C1.4 alkyl)-, or -N(C1.4 alkyl)-;
WI and W2 are each independently selected from the group consisting of carbonyl, methylene, sulfone and sulfoxide;
R2, R3, R4, R5, R6 and R7 are each independently:
a) C2 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
b) C2 to C20 straight chain or branched chain alkenyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
2b C) C2 to C20 straight chain or branched chain alkoxy, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
d) ¨NH-C2_20 straight chain or branched chain alkyl, wherein the alkyl group is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
e)-C(0)-C2_20 straight chain or branched chain alkyl or alkenyl, wherein the alkyl or alkenyl is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;

M' Z is 0 or NH; and M' and N are each independently C2 to C20 straight chain or branched chain alkyl, alkenyl, alkoxy, acyloxy, alkylamino, or acylamino;
or RI
R8 is C1-6 straight or branched chain alkyl or C2-6 straight or branched chain alkenyl or alkynyl;
R9 and R19 are independently selected from the group consisting of (i) C1 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy; and 2c , , GO C2 to C20 straight chain or branched chain alkenyl or alkynyl which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy;
G1, G2, G3, G4 and G5 are each independently oxygen, methylene, -NH-, thiol, -N(C1_4 alkyl)-, -N[C(0)-C1_4 alkyl]-, -NH-C(0)-, -NH-S02-, -C(0)-0-, -C(0)-NH-, -0-C(0)-, -0-C(0)NH-, -0-C(0)0-, -NH-C(0)-NH-, -C(0)NH-, -C(0)N(C1_4 alkyl), aryl, or -S(0),-, wherein n is 0, I, or 2;
or G2R4or G4R7 may together be a hydrogen atom or hydroxyl; and M is independently selected from a hydrogen atom and a pharmaceutically acceptable cation;
or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
The present invention also relates to the use of at least one immunotherapeutic agent selected from one or more cancer antigens; one or more viral antigen associated with cancer;
one or more anti-cancer antibodies; and one or more anti-idiotypic antibodies to an anti-cancer antibody; and one or more compound of formula (I) 2d y I
=,(rT.T.
(0-12)a R

MO¨PT T =P ¨OM

(CH2)d (CH2)e (C)---H2):\272 W1 (CH2)(r GI
(CH2)e (C1-12)en G2 ¨( G4 formula (I) wherein:
RI is:
(a) -C(0)-;
(b) -C(0)-C1-14 alkyl-C(0)- or-C(0)-C1_14 alkenyl-C(0)-;
wherein the -Ci_14 alkyl- or -C1_14 alkenyl- is optionally substituted with one or more substituents selected from the group consisting of hydroxy, C1_6 alkyl, C1-6 alkoxy, CI-6 alkyldioxy, C1-5 alkylamino, carboxy, C1-6 alkoxycarbonyl, C1..6 carbamoyl, C1-6 acylamino, and (aryl)C1.6 alkyl; and wherein the aryl moiety of the (aryl)C1.6 alkyl is optionally substituted with one or more substituents selected from the group consisting of CI-6 alkyl, C1-alkoxy, C1..6 alkylamino, C1.6 alkoxyamino, C1.6 alkylamino-C1.6 alkoxy, -0-C1-6 alkylamino-Ci -6 alkoxy, -0-C1-6 alkylamino-C(0)-C1-6 alkyl-C(0)0H, -0-C1-6 alkylamino-C(0)-C1_6 alkyl-C(0)-C1.6 alkyl, -0-C1_6 alkyl-NH-C1-6 2e alkyl-O-C1 -6 alkyl, -0-C1.6 alkyl-NH-C(0)C1..6 alkyl-C(0)0H, and ¨0-C1-6 alkyl-NH-C(0)C1 -6 alkyl-C(0)-C 1-6 alkyl;
(c) a C2 to C15 straight or branched chain alkyl group optionally substituted with one or more hydroxyl or alkoxy groups; or (d) -C(0)-C6-12 aryl-C(0)- wherein the aryl is optionally substituted with one or more group selected from the group consisting of hydroxy, halo, nitro, amino, C1..6 alkyl and C1-6 alkoxy groups;
a and b are each independently 0, 1, 2, 3 or 4;
d and e are each independently 1, 2, 3, 4, 5 or 6;
d' and e' are each independently 0, 1, 2, 3 or 4;
d" and e" are each independently 0, 1, 2, 3 or 4;
T is oxygen or sulfur;
X1, X2, Y1 and Y2 are each independently null, oxygen, NH, -N(C(0)C1_4 alkyl)-, or -N(C 1-4 alkyl)-;
W1 and W2 are each independently selected from the group consisting of carbonyl, methylene, sulfone and sulfoxide;
R2, R3, R4, R5, R6 and R7 are each independently:
a) C2 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
b) C2 to C20 straight chain or branched chain alkenyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
c) C2 to Cal straight chain or branched chain alkoxy, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
d) ¨NH-C2_20 straight chain or branched chain alkyl, wherein the alkyl group is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
2f e) -C(0)-C2_20 straight chain or branched chain alkyl or alkenyl, wherein the alkyl or alkenyl is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;

M' Z is 0 or NH; and M' and N are each independently C2 to C20 straight chain or branched chain alkyl, alkenyl, alkoxy, acyloxy, alkylamino, or acylamino;
or g) Ri R8 is C1_6 straight or branched chain alkyl or C2_6 straight or branched chain alkenyl or alkynyl;
R9 and RI are independently selected from the group consisting of (i) C1 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy; and (ii) C2 to C20 straight chain or branched chain alkenyl or alkynyl which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy;
GI, G2, G3, G4 and G5 are each independently oxygen, methylene, -NH-, thiol, -N(C14 alkyl)-, -N[C(0)-C14 alkyl]-, -NH-C(0)-, -NH-S02-, -C(0)-0-, -C(0)-NH-, 2g -0-C(0)-, -0-C(0)NH-, -0-C(0)0-, -NH-C(0)-NH-, -C(0)NH-, -C(0)N(C1_4 alkyl), aryl, or -S(0)-, wherein n is 0, 1, or 2;
or G2R4 or G4R7 may together be a hydrogen atom or hydroxyl; and M is independently selected from a hydrogen atom and a pharmaceutically acceptable cation;
or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof, for stimulating an immune response in a subject individual.
These and other aspects of the invention are described in more detail herein.
3. Figures Figure 1 is a graph depicting the percentage of tumor-bearing mice surviving after treatment with subcutaneous B16 GM-CSF(R) cells, E6020, or B16 GM-CSF (r) cells and E6020, or with no treatment.
2h Figure 2 is a graph depicting the percentage of tumor-bearing mice surviving after intratumoral treatment with B16 GM-CSF (R) cells, or B16 GM-CSF(r) cells and E6020, or with no treatment.
Figure 3 is a graph depicting the percentage of animals without sign of disease after no treatment or treatment with vaccine and E6020.
Figure 4 is a graph depicting the percentage of animals alive after no treatment, treatment with vaccine, or treatment with E6020.
4. Detailed Description of the Invention The invention provides compositions comprising (i) at least one immunotherapeutic agent selected from one or more cancer antigens, one or more antigens derived from a virus associated with cancer, one or more an anti-cancer antibody, and one or more anti-idiotypic antibody to an anti-cancer antibody, and (ii) one or more compounds selected from formulae (I), (1), (III), (IV), and (V), and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof. The compounds are described in detail below.
The compositions may be therapeutic, that is, the compounds are administered to treat an existing cancer, or to prevent the recurrence of a cancer, or prophylactic, that is, the compounds are administered to prevent or delay the development of cancer. When the compositions are used therapeutically, they are administered to cancer patients and are designed to elicit an immune response to stabilize a tumor by preventing or slowing the growth of the existing cancer, to prevent the spread of a tumor or of metastases, to reduce the tumor size, to prevent the recurrence of treated cancer, or to eliminate cancer cells not killed by earlier treatments. A composition used as a prophylactic treatment is administered to individuals who do not have cancer, and are designed to elicit an immune response to target potential cancer cells or to target an antigen derived from a virus associated with cancer.
The compositions of this invention may include more than one immunotherapeutic agent with another immunotherapeutic agent, for example, a cancer antigen in combination with one or more antigens derived from a virus associated with cancer, one or more anti-cancer antibodies, and one or more anti-idiotypic antibodies to an anti-cancer antibody. Another embodiment of the compositions may include one or more cancer antigens and an anti-cancer antibody and/or an anti-idiotypic antibody to an anti-cancer antibody. Other embodiments of the compositions may include an anti-cancer antibody and an anti-idiotypic antibody to an anti-cancer antibody. Another embodiment may include one or more antigens derived from a virus associated with cancer and an anti-cancer antibody and/or an anti-idiotypic antibody to an anti-cancer antibody.
4.1 Cancer Antigens One of the immunotherapeutic agents of the pharmaceutical composition may be one or more cancer antigens. A cancer antigen is (a) a cell surface antigen that can be found on a malignant cell, (b) an antigen that can be found inside a malignant cell or (c) a mediator of tumor cell growth.
The term "cancer antigen" refers to (i) tumor-specific antigens, (ii) tumor-associated antigens, (iii) cells that express tumor-specific antigens, (iv) cells that express tumor-associated antigens, (v) embryonic antigens on tumors, (vi) autologous tumor cells, (vii) tumor-specific membrane antigens, (viii) tumor-associated membrane antigens, (ix) growth factor receptors, (x) growth factor ligands, and (xi) any other type of antigen or antigen-presenting cell or material that is associated with cancer.
The cancer antigen can be any type of cancer antigen known in the art. The cancer antigen may be an epithelial cancer antigen, (e.g., breast, gastrointestinal, lung), a prostate specific cancer antigen (PSA) or prostate specific membrane antigen (PSMA), a bladder cancer antigen, a lung (e.g., small cell lung) cancer antigen, a colon cancer antigen, an ovarian cancer antigen, a brain cancer antigen, a gastric cancer antigen, a renal cell carcinoma antigen, a pancreatic cancer antigen, a liver cancer antigen, an esophageal cancer antigen, a head and neck cancer antigen, or a colorectal cancer antigen.
In another embodiment, the cancer antigen is a lymphoma antigen (e.g., non-Hodgkin's lymphoma or Hodgkin's lymphoma), a B-cell lymphoma cancer antigen, a leukemia antigen, a myeloma (i.e., multiple myeloma or plasma cell myeloma) antigen, an acute lymphoblastic leukemia antigen, a chronic myeloid leukemia antigen, or an acute myelogenous leukemia antigen.
In another embodiment, the cancer antigen is a mucin-1 protein or peptide (MUC-1) that is found on all human adenocarcinomas: pancreas, colon, breast, ovarian, lung, prostate, head and neck, including multiple myelomas and some B
cell lymphomas. Patients with inflammatory bowel disease, either Crohn's disease or ulcerative colitis, are at an increased risk for developing colorectal carcinoma. MUC-1 is a type I transmembrane glycoprotein. The major extracellular portion of has a large number of tandem repeats consisting of 20 amino acids which comprise immunogenic epitopes. In some cancers it is exposed in an unglycosylated form that is recognized by the immune system. See Gendler, S.J., et al., J. Biol. Chem.
265:15286-15293 (1990).
In another embodiment, the cancer antigen is a mutated B-Raf antigen, which is associated with melanoma and colon cancer. The vast majority of these mutations represent a single nucleotide change of T-A at nucleotide 1796 resulting in a valine to glutarnic acid change at residue 599 within the activation segment of B-Raf.
Raf proteins are also indirectly associated with cancer as effectors of activated Ras proteins, oncogenic forms of which are present in approximately one-third of all human cancers. Normal non-mutated B-Raf is involved in cell signaling, relaying signals from the cell membrane to the nucleus. The protein is usually only active when needed to relay signals. In contrast, mutant B-Raf has been reported to be constantly active, disrupting the signaling relay. Mercer and Pritchard, Biochim Biophys Acta. (2003);1653(1):25-40; Sharkey, et al, Cancer Res.
(2004);64(5):1595-9.
In one embodiment, the cancer antigen is a human epidermal growth factor receptor-2 (HER-2/neu) antigen. Cancers that have cells that overexpress HER-2/neu are referred to as HER-2/neu+ cancers. Exemplary HER-2/neu+ cancers include prostate cancer, lung cancer, breast cancer, ovarian cancer, pancreatic cancer, skin cancer, liver cancer (e.g., hepatocellular adenocarcinoma), intestinal cancer, and bladder cancer.
HER-2/neu has an extracellular binding domain (ECD) of approximately 645 aa, with 40% homology to epidermal growth factor receptor (EGFR), a highly hydrophobic transmembrane anchor domain (TMD), and a carboxyterminal intracellular domain (ICD) of approximately 580 aa with 80% homology to EGFR.
The nucleotide sequence of HER-2/neu is available at GENBANK Accession Nos.
AH002823 (human HER-2 gene, promoter region and exon 1); M16792 (human HER-2 gene, exon 4): M16791 (human HER-2 gene, exon 3); M16790 (human HER-2 gene, exon 2); and M16789 (human HER-2 gene, promoter region and exon 1). The amino acid sequence for the HER-2/neu protein is available at GENBANK
Accession No. AAA58637. Based on these sequences, one skilled in the art could develop HER-2/neu antigens using known assays to find appropriate epitopes that generate an effective immune response. Exemplary HER-2/neu antigens include p369-377 (a HER-2/neu derived HLA-A2 peptide); dHER2 (Corixa Corporation); ii-Key MHC class II epitope hybrid (Generex Biotechnology Corporation); peptide (amino acids 378-398); peptide P7 (amino acids 610-623); mixture of peptides (amino acids 544-560) and P7; mixture of peptides P4, P6 and P7; HER2 [9754];
and the like.
In one embodiment, the cancer antigen is an epidermal growth factor receptor (EGFR) antigen. The EGFR antigen can be an EGFR variant 1 antigen, an EGFR
variant 2 antigen, an EGFR variant 3 antigen and/or an EGFR variant 4 antigen.

Cancers with cells that overexpress EGFR are referred to as EGFR + cancers.
Exemplary EGFR + cancers include lung cancer, head and neck cancer, colon cancer, colorectal cancer, breast cancer, prostate cancer, gastric cancer, ovarian cancer, brain cancer and bladder cancer.
The nucleotide sequence (mRNA) of EGFR variant 1 is available at GENBANK Accession No. NM_005228. The nucleotide sequence (mRNA) of EGFR variant 2 is available at GENBANK Accession No. NM_201282. The nucleotide sequence (mRNA) of EGFR variant 3 is available at GENBANK
Accession No. NM_201283. The nucleotide sequence (mRNA) of EGFR variant 4 is available at GENBANK Accession No. NM_201284. Exemplary EGFR antigens include GI-3001; peptide aa 1168-1181; and the like.
In one embodiment, the cancer antigen is a vascular endothelial growth factor receptor (VEGFR) antigen. The VEGFR antigen can be VEGFR variant 1 antigen or VEGFR variant 2 antigen. VEGFR variant 1 is synonymous with Flt-1. VEGFR
variant 2 is synonymous with Flk-1 and Kdr (i.e., kinase insert domain protein receptor). VEGFR is considered to be a regulator of cancer-induced angiogenesis.
Cancers with cells that overexpress VEGFR are called VEGFR + cancers.
Exemplary VEGFR + cancers include breast cancer, lung cancer, small cell lung cancer, colon cancer, colorectal cancer, renal cancer, leukemia, and lymphocytic leukemia.
The nucleotide sequence (DNA) for VEGFR variant 1 (Flt-1) is available at GENBANK Accession No. D64016 (human gene for vascular endothelial growth factor receptor, promoter and exon 1). The nucleotide sequence (mRNA) for VEGFR
variant 2 (Flk-1 or Kdr) is available at GENBANK Accession No. AF063658 (human vascular endothelial growth factor receptor 2).

In one embodiment the cancer antigen is prostate-specific antigen (PSA) and/or prostate-specific membrane antigen (PSMA) that are prevalently expressed in androgen-independent prostate cancers.
In another embodiment, the cancer antigen is Gp-100 Glycoprotein 100 (gp 100) is a tumor-specific antigen associated with melanoma.
In one embodiment, the cancer antigen is a carcinoembryonic (CEA) antigen.
Cancers with cells that overexpress CEA are referred to as CEA + cancers.
Exemplary CEA + cancers include colorectal cancer, gastric cancer and pancreatic cancer.
The nucleotide sequence (mRNA) for human carcinoembryonic antigen-like 1 is available at GENBANK Accession No. NM_020219. Exemplary CEA antigens include CAP-1 (i.e., CEA aa 571-579), CAP1-6D, CAP-2 (i.e., CEA aa 555-579), CAP-3 (i.e., CEA aa 87-89), CAP-4 (CEA aa 1-11), CAP-5 (i.e., CEA aa 345-354), CAP-6 (i.e., CEA aa 19-28) and CAP-7.
In one embodiment, the cancer antigen is carbohydrate antigen 10.9 (CA 19.9).
CA 19.9 is an oligosaccharide related to the Lewis A blood group substance and is associated with colorectal cancers.
In another embodiment, the cancer antigen is a melanoma cancer antigen.
Melanoma cancer antigens are useful for treating melanoma. Exemplary melanoma cancer antigens include MART-1 (e.g., MART-1 26-35 peptide, MART-1 27-35 peptide); MART-1/Melan A; pMe117; pMe117/gp100; gp100 (e.g., gp 100 peptide 280-288, gp 100 peptide 154-162, gp 100 peptide 457-467); TRP-1; TRP-2; NY-ESO-1; p16; beta-catenin; mum-1; and the like.
In one embodiment, the cancer antigen is a mutant or wild type ras peptide.
The mutant ras peptide can be a mutant K-ras peptide, a mutant N-ras peptide and/or a mutant H-ras peptide. Mutations in the ras protein typically occur at positions 12 (e.g., arginine or valine substituted for glycine), 13 (e.g., asparagine for glycine), 61 (e.g., glutatnine to leucine) and/or 59. Mutant ras peptides can be useful as lung cancer antigens, gastrointestinal cancer antigens, hepatoma antigens, myeloid cancer antigens (e.g., acute leukemia, myelodysplasia), skin cancer antigens (e.g., melanoma, basal cell, squamous cell), bladder cancer antigens, colon cancer antigens, colorectal cancer antigens, and renal cell cancer antigens.
In another embodiment of the invention, the cancer antigen is a mutant and/or wildtype p53 peptide. The p53 peptide can be used as colon cancer antigens, lung cancer antigens, breast cancer antigens, hepatocellular carcinoma cancer antigens, lymphoma cancer antigens, prostate cancer antigens, thyroid cancer antigens, bladder cancer antigens, pancreatic cancer antigens and ovarian cancer antigens.
The cancer antigen can be a cell, a protein, a peptide, a fusion protein, DNA
encoding a peptide or protein, RNA encoding a peptide or protein, a glycoprotein, a lipoprotein, a phosphoprotein, a carbohydrate, a lipopolysaccharide, a lipid, a chemically linked combination of two or more thereof, a fusion or two or more thereof, or a mixture of two or more thereof. In another embodiment, the cancer antigen is a peptide comprising about 6 to about 24 amino acids; from about 8 to about 20 amino acids; from about 8 to about 12 amino acids; from about 8 to about 10 amino acids; or from about 12 to about 20 amino acids. In one embodiment, the cancer antigen is a peptide having a MHC Class I binding motif or a MHC Class II
binding motif. In another embodiment, the cancer antigen comprises a peptide that corresponds to one or more cytotoxic T lymphocyte (CTL) epitopes.
In another embodiment, the cancer antigen is in the form of a foreign homologous cancer antigen. Foreign homologous cancer antigens and methods for making them are described in US Patent No. 6,942,862. Because many human cancer antigens are self proteins (i.e., proteins normally produced by an individual and not necessarily unique to cancer), immunologic tolerance may exist and represent a barrier to effective vaccination against the human cancer antigens. This aspect of the invention overcomes immunologic tolerance by immunizing a patient with a protein or peptide that is foreign (i.e., not identical to that in the individual) but nevertheless homologous to an individual's self cancer antigen or portion thereof.
"Foreign" cancer antigens can be generated from, for example, rabbits, rats, mice and pigs.
Generally, a foreign cancer antigen (e.g., protein or peptide) will possess at least about 75%
sequence homology to the cancer antigen targeted. Sequence homology means either identical amino acids at the same positions in the sequence (i.e., sequence identity), or conservative substitutions of amino acids at the same positions in the sequence.
Conservative substitutions are well known in the art. Examples are isoleucine for leucine, valine for alanine, glutamic acid for aspartic acid, threonine for serine, etc.
Typically, a foreign cancer antigen (e.g., proteins or peptides) will possess about 80%, 85%, 90%, 95% or 99% sequence homology. Preferred foreign cancer antigens (e.g., proteins or peptides) are those which are highly homologous, e.g., with from about but less than 100% sequence homology. Particularly preferred foreign cancer antigens (e.g., proteins or peptides) are those wherein the aforementioned sequence homology , , , , percents each represent percent sequence identity.
4.2 Viral Antigens Derived from Virus Associated with Cancer One of the immunotherapeutic agents of the pharmaceutical composition may be one or more antigens derived from a virus associated with cancer. Infection from certain viruses are known to lead to the development of different types of cancers, for example, human papilloma virus (HPV), hepatitis viral infections, Epstein-Barr virus (EBV), human herpes virus 8 (HHV-8), human T-cell leukemia virus-1 (HTLV-1) and human T-cell leukemia virus-2 (HTLV-2).
Patients who are infected or who are at risk of being infected with the human papilloma virus (HPV) are at a higher risk for developing cervical cancer than HPV
negative patients. The risk for cervical cancer is particularly high for patients who have HPV-16, HPV-18, HPV-31, HPV-33 and/or HPV-35 infections. The HPV antigen that can be used in the pharmaceutical compositions and the methods of the invention can be an HPV-16 antigen, an HPV-18 antigen, an HPV-31 antigen, an HPV-33 antigen and/or an HPV-35 antigen; and is preferably an HPV-16 antigen and/or HPV-18 antigen.
The genome of HPV-16 is described in Virology, 145:181-185 (1985) and DNA
sequences encoding HPV-18 are described in US Patent No. 5,840,306. HPV-16 antigens (e.g., seroreactive regions of the El and/or E2 proteins of HPV-16) are described in US Patent No. 6,531,127, and HPV-18 antigens (e.g., seroreactive regions of the Ll and/or L2 proteins of HPV-18) are described in US Patent No. 5,840,306. Based on the sequences and antigens for HPV-16 and HPV-18 described in these references, one skilled in the art could develop other HPV antigens using known assays to find appropriate epitopes that generate an effective immune response.
Patients who are infected or are at risk of being infected with hepatitis viral infections, such as hepatitis B (HBV) and/or hepatitis C (HCV) viral infections, are at a higher risk for developing liver cancer than patients who do not have hepatitis viral , infections. HBV antigens and HCV antigens can be used in the pharmaceutical compositions and the methods of the invention. The complete genome for HBV is available at GENBANK Accession No. NC _003977, the disclosure of which is incorporated herein.
The genome of HCV is described in European Patent Application No. 318 216.
PCT/US90/01348 discloses sequence information ____________________________________ 9a of clones of the HCV genome, amino acid sequences of HCV viral proteins and methods of making and using such compositions for HCV vaccines comprising HCV
proteins and peptides derived there from. Based on the sequences and antigens for HBV and HCV described in these references, one skilled in the art could develop other HBV and/or HCV antigens using known assays to find appropriate epitopes that generate an effective immune response.
Patients who are infected or are at risk for being infected with Epstein-Barr virus (EBV) are at a higher risk for developing Burkitt's lymphoma, nasopharyngeal carcinoma and Hodgkin's disease than EBV negative patients. An EBV antigen can be used in the pharmaceutical compositions and the methods of the invention.
The nucleotide sequence of EBV DNA is described, for example, in US Patent No.
4,707,358. Based on this sequences for EBV, one skilled in the art could develop EBV antigens using known assays to find appropriate epitopes that generate an effective immune response. The compounds of the invention, EBV antigens and immunostimulatory compounds can be administered separately or in the form of a composition. The composition can be in the form of a prophylactic vaccine (i.e., for patients who are EBV negative) or therapeutic vaccine (i.e., for patients who are EBV
positive).
Patients who are infected or are at risk of being infected with human herpes virus 8 (HHV-8) are at a higher risk for developing Kaposi's sarcoma than HHV-negative patients. The HHV-8 antigen can be used in the pharmaceutical compositions and the methods of the invention. The nucleotide sequence of HHV-8 is described, for example, by Russo et al, "Nucleotide sequence of the Kaposi sarcoma-associated herpes virus (HHV8)," Proc. Natl. Acad. Sci USA, 93:14862-14867 (1996).
Based on the known sequence for HHV-8, one skilled in the art could develop antigens using known assays to find appropriate epitopes that generate an effective immune response.
Patients who are infected or at risk of being infected with human T-cell leukemia virus-1 (HTLV-1) or human T-cell leukemia virus-2 (HTLV-2) are at a higher risk for developing T-cell leukemia than HTLV-1 or HTLV-2 negative patients. The sequences of HTLV-1 and HTLV-2 are well known in the art and are described in Wong-Staal F, Gallo RC. Human T-lymphotropic retroviruses. Nature 317:395-403, 1985.

4.3 Anti-Cancer Antibodies One of the immunotherapeutic agents of the pharmaceutical composition may be one or more anti-cancer antibodies, that is, an antibody that has been generated to one or more cancer antigens. Exemplary anti-cancer antibodies include the following:
trastuzumab (HERCEPTIN by Genentech) which is used to treat HER-2/neu positive breast cancer or metastatic breast cancer;
bevacizumab (AVASTIN by Genentech) which is used to treat colorectal cancer, metastatic colorectal cancer, breast cancer, metastatic breast cancer, non-small cell lung cancer, or renal cell carcinoma;
rituximab (RITUXAN by Genentech), which is used to treat non-Hodgkin's lymphoma or chronic lymphocytic leukemia;
pertuzumab (OMNITARG by Genentech) which is used to treat breast cancer, prostate cancer, non-small cell lung cancer, or ovarian cancer;
cetuximab (ERBITUX by ImClone Systems Incorporated) which can be used to treat colorectal cancer, metastatic colorectal cancer, lung cancer, head and neck cancer, colon cancer, breast cancer, prostate cancer, gastric cancer, ovarian cancer, brain cancer, pancreatic cancer, esophageal cancer, renal cell cancer, prostate cancer, cervical cancer, or bladder cancer;
IMC-1C11 (ImClone Systems Incorporated) which is used to treat colorectal cancer, head and neck cancer, as well as other potential cancer targets;
tositumomab and tositumomab and iodine 1131 (BEXXAR by Corixa Corporation) which is used to treat non-Hodgkin's lymphoma, which can be CD20 positive, follicular, non-Hodgkin's lymphoma, with and without transformation, whose disease is refractory to Rituximab and has relapsed following chemotherapy;
mm ibirtumomab tiuxetan; Y9 ibirtumomab tiuxetan; Inm ibirtumomab tiuxetan and Y9 ibirtumomab tiuxetan (ZEVALIN by Biogen Idec) which is used to treat lymphoma or non-Hodgkin's lymphoma, which can include relapsed follicular lymphoma; relapsed or refractory, low grade or follicular non-Hodgkin's lymphoma;
or transformed B-cell non-Hodgkin's lymphoma;
EMD 7200 (EMD Pharmaceuticals) which is used for treating for treating cancer is non-small cell lung cancer or cervical cancer;
SGN-30 (genetically engineered monoclonal antibody targeted to CD30 antigen by Seattle Genetics) (Hodgkin's lymphoma or non-Hodgkin's lymphoma);
SGN-15 (genetically engineered monoclonal antibody targeted to a LewisT-related antigen that is conjugated to doxorubicin by Seattle Genetics)( non-small cell lung cancer); SGN-40 (humanized monoclonal antibody targeted to CD40 antigen by Seattle Genetics)( multiple myeoloma or non-Hodgkin's lymphoma); SGN-35 (genetically engineered monoclonal antibody targeted to a CD30 antigen that is conjugated to Auristatin E by Seattle Genetics)(non-Hodgkin's lymphoma); SGN-17/19 (fusion protein containing antibody and enzyme conjugated to melphalan prodrug by Seattle Genetics)( melanoma or metastatic melanoma).
The anti-cancer antibody can be a fragment of an antibody; a complex comprising an antibody; or a conjugate comprising an antibody. The antibody can optionally be chimeric or humanized.
The mechanism(s) of action of many of these antibodies are not entirely clear, but generation of an immune response such as antibody-dependent cell-mediated cytotoxicity (ADCC) is often believed to be part of the therapeutic action.
Activation of ADCC by TLR4 ligation on Pc receptor-bearing cells may enhance the anti-tumor efficacy of the antibody. The use of the anti-cancer antibodies in combination with one or more compounds selected from formulae (I), (II), (ifi), (IV), and (V) will increase the immune response.
4.4 Anti-Idiotypic Antibodies Antibodies to antigens have a serologically unique structure at the antigen-binding site, called an idiotype. An antibody may be generated to the original antibody, resulting in the production of anti-idiotypic antibodies. The original antibody is designated Ab 1, and the anti-idiotypic antibody Ab2. The Ab2 antibodies recognize the antigen-binding site of Abl, and therefore share a motif or structural similarities with the original antigen. An antibody raised to the binding site on Ab2 may therefore react with the original antigen. If the original antigen is a cancer antigen, the anti-Ab2 antibody may have a therapeutic effect.
When administered alone, the anti-idiotypic antibody may not be able to generate sufficient immune response to a cancer antigen. However, when the anti-idiotypic antibody is used in combination with one or more compound selected from formulae (I), (II), (In), (IV), and (V) as a pharmaceutical composition or in a method of administration of this invention, an immune response is generated. The adjuvant compounds of formulae (I), (II), (III), (IV), and (V) can improve the immunogenicity of the anti-idiotypic antibody or provide the anti-idiotypic antibody with the ability to generate an immune response by breaking immunological tolerance. In addition, the pharmaceutical composition of the invention may also reduce the amount of anti-idiotypic antibodies needed to induce an immune response and/or reduce the number of administrations needed to induce the desired immune response.
Anti-idiotypic antibodies are known in the art. The anti-idiotypic antibody may be an antibody to an antibody that that is produced in response to a cancer antigen described above or to an anti-cancer antibody. Exemplary anti-idiotypic antibodies include (described in US Patent No. 6,042,827); BEC2 (ImClone Systems Incorporated);

(Igeneon, a subsidiary of Aphton Corporation); and the like. The production of anti-idiotypic antibodies are well known in the art and are described for example in US
Patent No.
6,926,893.
4.5 Modes of Administration of Pharmaceutical Compositions Administration of the pharmaceutical composition can be accomplished by several routes that would be suitable for delivery. Exemplary delivery modes include parenteral administration, e.g., subcutaneous injection, transcutaneous, intravenous, intra-tumoral, peri-tumoral, intra-nasal, ophthalmic, intramuscular, intradermal, intraperitoneal, pulmonary, and non-parenteral administration, e.g., transmucosal, transdermal, inhalation, intravaginal, rectal or oral administration.
4.6 Methods of Treatment The pharmaceutical composition provides a method for stimulating or eliciting or enhancing an immune response in a subject individual. The subject individual is preferably human, although the invention can be applied in veterinary applications to animal species, including mammals or avian species.
The subject individual may be at risk for developing cancer, diagnosed with a cancer, in treatment for cancer, or in post-therapy recovery from cancer.
The term "immune response" encompasses both cellular and humoral immune responses, including stimulating the production of cytokines, stimulating the proliferation of immune cells, stimulating the activation of immune cells, or stimulating the lytic activity of immune cells. Examples of immune responses stimulated by the methods of the invention are the secretion of cytokines, the activation of NK cells, the proliferation of B cells, T cells, macrophages, monocytes, and other immune cells, and other immune responses. To detect a cellular immune response, for example, T cell effector activity against cells expressing the antigen can _____________________________________________________________ 13a be detected using standard assays, e.g., target-cell killing, macrophage activation, B-cell activation or lymphokine production. Humoral responses can be measured by detecting the appearance of, or the increase in titer of, for example, antigen-specific antibodies, using routine methods such as ELISA. The progress of the antibody response can be determined by measuring class switching, such as the switch from an early IgM response to a later IgG response.
As used herein, the term "stimulate an immune response" includes stimulating, eliciting, increasing, enhancing, sustaining, and/or improving the stimulation of new immune response or of a preexisting immune response. Thus, "stimulating an immune response" as an impnunotherapy refers to enhancing the therapeutic efficacy, increasing survival time, slowing the progression of a cancerous tumor or shrinking the cancerous tumor size, preventing the spread of a tumor or of metastases, preventing or slowing the recurrence of treated cancer, eliminating cancer cells not killed by earlier treatments, targeting potential cancer cells or targeting antigens derived from a virus associated with cancer. In the methods of this invention, the immunotherapeutic agent and the compound of selected from formulae (I), (II), (DI), (IV) and (V) are administered in an amount effective to stimulate an immune response in the subject individual at a dose sufficient to generate an effective immune response without unacceptable toxicity. As will be understood by one of skill in the art, the magnitude of the immune response and the maintenance of that response may have varying degrees which will be recognized a having a potential therapeutic or prophylactic benefit.
To stimulate an immune response, the subject individual is administered (i) at least one immunotherapeutic agent selected from one or more cancer antigens, one or more antigens derived from a virus associated with cancer, an anti-cancer antibody, and an anti-idiotypic antibody to an anti-cancer antibody, and (ii) one or more compounds selected from formulae (I), (II), (III), (IV) and (V), and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof. Typically, the administration of the immunotherapeutic agent and the compound of selected from formulae (I), (II), (III), (IV) and (V) will be in the form of a vaccine or administered in a vaccine regimen.
The therapeutic agent and the compound of selected from formulae (I), (II), (W), (IV) and (V) can be administered at about the same time to the subject individual, or can be administered separately and/or sequentially. "At about the same time" includes administering one or more immunotherapeutic agents and one or more compounds selected from formulae (I), (II), (III), (IV), and (V) at the same time, at the same time, but through different modes of administration or at different sites on the body, at different times on the same day, or on different days, provided that they are administered as part of an overall dosing treatment regimen. When administered separately, or sequentially, the one or more immunotherapeutic agents and one or more compounds selected from formulae (I), (II), (111), (IV), and (V) are part of an overall treatment regimen, such as a therapeutic cocktail or a combination therapy.
The dosing schedule can be routinely determined by one of skill in the art and may be varied or modified according to the appropriate treatment for the subject individual.
For example, the immunotherapeutic agent and a compound of formulae (I), (II), (III), (IV) and (V) can be administered at about the same time as a single dose, or by a dose of the therapeutic agent and a dose of a compound of formulae (I), (11), (IV) and (V). The dosing schedule can be continued at regular intervals, such as at 1 to 4 week time periods, followed by dosing at regular intervals of 1 to 3 months, for example. In another embodiment, the dosing schedule can be based on a "prime" and "boost"
treatment, in which the immunotherapeutic agent is administered to prime or stimulate the production of CTLs and then another dose of immunotherapeutic agent in combination with one or more compounds selected from formulae (I), (II), (M), (IV), and (V) to boost the production of neutralizing antibodies and antibody dependent cellular cytotoxcity. The immune response in the subject individual can be assessed and monitored through known methods.
In some instance, these treatments can be used in combination with conventional cancer therapies or pharmaceutical formulations useful for treating cancer or infectious diseases. These treatments can include surgical procedures, radiation therapy and/or ablation therapy (e.g., laser therapy, infrared therapy and the like).
Cancer therapies including dendritic cell therapy, chemokines, cytokines, tumor necrosis factors (e.g., TNF-a), chemotherapeutic agents (e.g., adenosine analogs (e.g., cladribine, pentostatin), alkyl sulfanates (e.g., busulfan)), anti-tumoral antibiotics (e.g., bleomycin, dactinomycin, daunorabicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, mitomycin), aziridines (e.g., thiotepa), camptothecin analogs (e.g., irinotecan, topotecan), cryptophycins (e.g., cryptophycin 52, cryptophicin 1), dolastatins (e.g., dolastatin 10, dolastatin 15), enedyine anticancer drugs (e.g., esperamicin, calicheamicin, dynemicin, neocarzinostatin, neocarzinostatin chromophore, kedarcidin, kedarcidin chromophore, C-1027 chromophore, and the like), epipodophyllotoxins (e.g., etoposide, teniposide), folate analogs (e.g., methotrexate), maytansinoids (e.g., maytansinol and maytansinol analogues), microtubule agents (e.g., docetaxel, paclitaxel, vinblastine, vincristine, vinorelbine), nitrogen mustards (e.g., chlorambucil, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, melphalan), nitrosoureas (e.g., carmustine, lamustine, streptoxacin), nonclassic alkylators (e.g., altretamine, dacarbazine, procarbazine, temozolamide), platinum complexes (e.g., carboplatin, cisplatin), purine analogs (e.g., fludarabine, mercaptopurine, thioguanine), pyrimidine analogs (e.g., capecitabine, cytarabine, depocyt, floxuridine, fluorouracil, gemcitabine), substituted ureas (e.g., hydroxyurea)]; anti-angiogenic agents (e.g., canstatin, troponin I, ), biologic agents (e.g., ZD 1839, virulizin and interferon), antibodies and fragments thereof (e.g., anti EGFR, anti-HER-2/neu, anti-KDR, Th4C-C225), anti-emetics (e.g., lorazepam, metroclopramide, and domperidone), epithelial growth factor inhibitors (e.g., transforming growth factor beta 1), anti-mucositic agents (e.g., dyclonine, lignocaine, azelastine, glutamine, corticoid steroids and allopurinol), anti-osteoclastic agents (e.g., bisphosphonates {e.g., etidronate, pamidronate, ibandronate, and osteoprotegerin 1), hormone regulating agents (e.g., anti-androgens, LHRH agonists, anastrozole, tamoxifen), hematopoietic growth factors, anti-toxicity agents (e.g., amifostine), kinase inhibitors (gefitinib, imatinib), and mixtures of two or more thereof.
Antibodies that block immunosuppressive functions, for example, anti-CTLA4 antibodies that block a receptor on T cells that turns off activation may also be used in combination with the immunotherapeutic agent and a compound of formulae (I), (II), (111), (IV) and (V). Thus, administering the immunotherapeutic agent and a compound of formulae (I), (II), (III), (IV) and (V) with anti-CTLA4 antibodies will increase immune response in the subject individual.
With regard to both prophylactic and therapeutic methods of treatment, such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics. "Pharmacogenomics", as used herein, refers to the application of genomics technologies such as gene sequencing, statistical genetics, and gene expression analysis to drugs in clinical development and on the market.
More specifically, the term refers to the study of how a patient's genes determine his or her response to a drug (e.g., a patient's "drug response phenotype" or "drug response genotype"). Thus, another aspect of the invention provides methods for tailoring an individual's prophylactic or therapeutic treatment according to that individual's drug response genotype. Pharmacogenomics allows a clinician or physician to target prophylactic or therapeutic treatments to patients who will most benefit from the treatment and to avoid treatment of patients who will experience toxic drug-related side effects. The clinician or physician can thereby tailor the type of treatment that may be necessary to the specific patient.
4.7 Optional Immunostimulatory Compounds In one embodiment of the methods of immunotherapy, the immune response is further augmented by the administration of compounds that may act as an immunostimulatory compound. Exemplary immunostimulatory compounds include toll like receptor (TLR) agonists (e.g., TLR4, TLR7, TLR9), N-acetylmuramyl-L-alanine-D-isoglutamine (MDP), lipopolysaccharides (LPS), genetically modified and/or degraded LPS, alum, glucan, colony stimulating factors (e.g., EPO, GM-CSF, G-CSF, M-CSF, pegylated G-CSF, SCF, IL-3, IL6, PIXY 321), interferons (e.g., y-interferon, a-interferon), interleukins (e.g., IL-2, IL-7, IL-12, IL-15, IL-18), MHC
Class II binding peptides, saponins (e.g., QS21), unmethylated CpG sequences, methyl tryptophan, arginase inhibitors, cyclophosphamide, antibodies that block immunosuppressive functions (e.g., anti-CTLA4 antibodies), and mixtures of two or more thereof. Exemplary TLR4 agonists include lipopolysaccharides (LPS); E.
coil LPS; and P. gingivalis LPS. Exemplary TLR7 agonists include imidazoquinoline compounds (e.g., imiquimod, resiquimod and the like); and loxoribine.
4.8 Pharmaceutical Formulations The pharmaceutical composition is formulated to be compatible with its intended route of administration, and will typically include a pharmaceutically acceptable carrier. As used herein the term "pharmaceutically acceptable carrier"
includes solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions. The pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000 and Encyclopedia of Pharmaceutical Technology, (eds. J. Swarbrick and J. C.
Boylan), 1988-1999, Marcel Dekker, New York).

Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH
can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the selected particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride are included in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating compound selected from formulae (I), (II), (III), (IV), and (V) in the specified amount in an appropriate solvent with one or a combination of ingredients enumerated above, as needed, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and other ingredients selected from those enumerated above or others known in the art. In the case of sterile powders for the preparation of sterile injectable solutions, the methods of preparation can be vacuum drying and freeze-drying, which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
4.9 Covalent Bonding, Complexes and Conjugates In one embodiment, one or more immunotherapeutic agent and a compound selected from formulae (I), (II), (III), (IV), and (V) can be covalently bonded together through an alkyl, amino, carbonyl, ether, hydroxyl, phosphate, phosphonyl, sulfonyl, sulfate, thiol ether, or thiol moiety of compound selected from formulae (I), (II), (III), (IV), and (V). For example, one or more immunotherapeutic agent can be covalently bonded to the R1, X1 and/or Y1 substituents in the compounds selected from formulae (I), (II), (III), (IV), and (V). For example, the immunotherapeutic agent can be covalently bonded to a carbonyl moiety (e.g., the C1 carbonyl) of the -C(0)- group or to the -C(0)-Ci_i4alkyl-C(0)- group of the R1 substituent in one or more compounds of the invention. As another example, the immunotherapeutic agent can be covalently bonded through a nitrogen atom in the X1 and/or Y1 substituents in the compound selected from formulae (I), (II), (III), (IV), and (V).
One skilled in the art would be able to link one or more immunotherapeutic agent to a compound selected from formulae (I), (II), (III), (IV), and (V) following the methods described, for example, by Hoffman et al., Biol. Chem. Hoppe-Sayler, 370:575-582 (1989);
Wiesmuller et al., Vaccine, 7:29-33 (1989); Wiesmuller et al., Int. J Peptide Protein Res., 40:255-260 (1992); Defourt etal., Proc. Natl. Acad. Sci., 89:3879-3883 (1992);
Tohokuni et al., J. Ani. Chem. Soc., 116:395-396 (1994); Reichel, Chem. Commun., 2087-2088 (1997);
Kamitakahara, Agnew. Chem. Int. Ed. 37:1524-1528 (1998); Dullenkopf et al., Chem. Eur.
J, 5:2432-2438 (1999).

In one embodiment, the immunotherapeutic agent can be in the form of a complex.
The complex can comprise at least one cancer antigen (optionally bonded to the compound selected from formulae (I), (II), (III), (IV), and (V)) and one or more proteins, peptides, immunostimulatory compounds and/or cells. Exemplary proteins and peptides include heat shock proteins, heat shock peptides, MHC Class I proteins, MHC Class I
peptides, MHC
Class II proteins, MHC Class II peptides, and the like.
1 9a Exemplary cells include dendritic cells, autologous dendritic cells, dendritic cells pulsed with cancer antigens, autologous dendritic cells pulsed with cancer antigens, and the like. Exemplary immunostimulatory compounds include TLR agonists (e.g., TLR4, TLR7, TLR9), N-acetylmuramyl-L-alanine-D-isoglutamine (MDP), lipopolysaccharides (LPS), genetically modified and/or degraded LPS, alum, glucan, colony stimulating factors (e.g., EPO, GM-CSF, G-CSF, M-CSF, PEGylated G-CSF, SCF, 1L-3, IL6, PIXY 321), interferons (e.g., 7-interferon, a-interferon), interleukins (e.g., IL-2, 1L-7, IL-12, IL-15, IL-18), saponins (e.g., QS21), monophosphoryl lipid A, 3 De-O-acylated monophosphoryl lipid A (3D-MPL), unmethylated CpG sequences, 1-methyl tryptophan, arginase inhibitors, cyclophosphamide, antibodies that block immunosuppressive functions (e.g., anti-CTLA4 antibodies), and the like. The complex with at least one cancer antigen (optionally bonded to compound selected from formulae (I), (1), (III), (IV), and (V)) and the proteins, peptides, immunostimulatory compounds and/or cells can be non-covalent, ionic, covalent, van der Waals forces, hydrogen bonding, and the like.
In another embodiment, the cancer antigen (optionally bonded to compound selected from formulae (I), (II), (III), (IV), and (V)) is in the form of a complex comprising a heat shock protein or heat shock peptide (HSP), particularly gp96. In one embodiment, the complex is formed by in vitro peptide pulsing the heat shock proteins and/or heat shock peptides. The complex can be covalent or non-covalent. In another embodiment, the cancer antigen (optionally bonded to compound selected from formulae (I), (II), (ifi), (W), and (V)) is in the form of a complex comprising a peptide, at least one heat shock protein and/or heat shock peptide, and at least one MHC Class I and/or II protein or peptide.
In another embodiment, the cancer antigen (optionally bonded to compound selected from formulae (I), (II), (III), (IV), and (V)) is in the form of a complex comprising a peptide and dendritic cells. In one embodiment, the complex is formed by in vitro peptide pulsing the dendritic cells to form cancer antigen loaded dendritic cells. The dendritic cells can be pulsed with cancer antigens and/or organisms (e.g., recombinant viruses or bacteria) expressing cancer antigens. The dendritic cells can be autologous.
In another embodiment, the cancer antigen (optionally bonded to compound selected from formulae (I), (II), (111), (IV), and (V)) is in the form of a complex comprising a peptide and an MHC Class I protein or peptide; and/or comprising a peptide and a MHC Class II protein or peptide. In one embodiment, the MHC
Class I
protein or peptide is HLA (e.g., HLA-A*0201, HLA-A2, HLA-A3, HLA-A*1101, HLA-A*3101, HLA-A*3301, HLA-A*6801, HLA-A24). The complex can be covalent or non-covalent. In another embodiment, the cancer antigen (optionally bonded to compound selected from formulae (I), (II), (III), (IV), and (V)) is in the form of a complex comprising a peptide, a MHC Class I and/or II protein or peptide, and at least one heat shock protein and/or heat shock peptide.
In another embodiment, the cancer antigen is in the form of a conjugate. For example, one or more cancer antigens can be chemically linked to one or more proteins, peptides, carbohydrates, polymers, lipids and/or toxic moieties. The conjugate can be of the formula: A-L-X, wherein "A" is one or more cancer antigens as described herein, "L" is one or more linking groups, and "X" is one or more proteins, peptide, carbohydrates, polymers, lipids and/or toxic moieties.
Exemplary linking groups include chemical linking groups and peptide linking groups. The most common methods for linking "A" and "X" rely on the presence of free amino (a-amino or Lys), sufhydryl (Cys), or carboxylic acid groups (Asp, Glu, or a-carboxyl).
Linking methods should be used that link the peptides to the carrier proteins, peptides, carbohydrates, lipids and/or toxic moieties via the carboxy- or amino-terminal residue.
Other common linking methods include maleimide and carbodiimide coupling chemistry. The linking group can be selected so that enzymes act upon the linking group so that "A" and "X" separate from each other in vivo. Alternatively, a linking group can be selected so that "A" and "X" remain covalently bonded via the linking group in vivo. In still other embodiments, "L" can simply be a covalent bond between "A" and "X." "X" can be a peptide, protein (such as an MHC Class I protein or an MHC Class II protein); keyhole limpet hemocyanin; albumin; bovine serum albumin;
ovalbumin; rabbit serum albumin, antibody and the like. "X" can be a peptide.
The peptide can be cancer antigen that is the same as or different from "A." Such a conjugate may be referred to as a multiple antigen peptide (MAP), and the peptides can form a fusion protein with or without the presence of a linking group "L."
The peptide can be a Class I and/or Class II peptide that can assist in generating an immune response. One skilled in the art will appreciate that the term "fusion protein"
may be used when two peptides are linked together, in which case the "L" group may drop out of the structure. In other embodiments, "X" can be a toxic moiety.
Exemplary toxic moieties include viral toxins, bacterial toxins (e.g., diphtheria toxins, tetanus toxins, clostridia toxin, cholera toxin, anthrax toxin, botulinum toxin, pertussis toxin, tracheal toxin,
5. Compounds of Formulae (I), (II), (III), (IV) and (V) The pharmaceutical compositions include the immunotherapeutic agents described above and a compound selected from formulae (1), (II), (III), (IV), and (V) and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof. ). The compounds of Formula (I), (II), (IV) and (V) can function as adjuvants and/or as immunostimulatory compounds depending on the application in which they are used.
X1 yl (CH2)a R1 (CH2)b MO- P¨=1T T=P¨OM

(CH2)d (CH2)e X2 _________________ < _______________________________ y2 \w2 (CH2)d, (CH2)e.

(CH2)d..
(CH2)e"
G2 _____________________ <
R3 ) ____ G4 R4 \ 7 (1) X1 yl (cid2) Ri (C. \
IA ,2/b Z1¨(CH2)a.-0¨P¨=T 1=-- P ¨ 0¨ (CH2)iy¨Z2 (CF12)d (CH2)e / _______________________ < y2 ./> \
wl (C1-12)d. (CH2)e vv2 .
/
\R2 G1 \ /

\
/
(CH2)d"
(CH2)e"
G2 _______________________ <
/ R3 ) ______ G4 R4 \R7 (1) Xi yi (cF12) Ri (c.1_, 2) ,,b R , ,,._N+_Ri2 R i ....¨N+_ R12 (CH2)d (CF12)e X2 _________________ <
/ />
wl (CF12)cr (CH2)e' w2 \ /
R2 \ G/1 \ /
(CH2)d"
(CH2)e"
G2 _____________________ ( i R3 ) ____ G4 R4 \R7 (11) X1 yl (CH2)a Ri (CHAD

MO-P=T 1P¨OM
I I

(CH2)d (CH2)e f.r.n.
,..... A 2,..1 d, 1 (012)e, I

,, A
G R3 G-' Fr RI , R4 ' (IV) Xi yl (CH2)a R1 (01-12)b I I

I I
Z1¨(0H2)a.-0¨P=1- T=P-0¨(CH2)1Y¨Z2 (CH2)d (CF12)e 4r,G1 "
k.,.........2)di 1, (CHO,. I
Fr R5 ,, , G` Fr R6 RI ,9' 1 R,' (V) wherein:
R1 is: (a) -C(0)-;
(b) -C(0)-Ci_malkyl-C(0)- or -C(0)-C1_ma1keny1-C(0)-;

wherein the -C1_14alky1- or -Ci..14alkenyl- is optionally substituted with one or more substituents selected from hydroxy, Ci.6alkoxy, Ci_6alkyldioxy, C1_5 alkylamino, carboxy, C1_6 alkoxycarbonyl, C1_6 carbamoyl, C1-6 acylamino, and/or (aryl)C1_6alkyl; and wherein the aryl moiety of the (aryl)C1_6-alkyl is optionally substituted with one or more substituents selected from Ci_6alkyl, C1-6alkoxy, Ci_6alkylamino, C1_6alkoxyamino, C1_6alkylamino-Ci_6alkoxy, -0-C1..6alkylamino-C1_6alkoxy, -0-C1_6alkylamino-C(0)-C1_6alkyl-C(0)0H, -0-Ci_6alkylamino-C(0)-Ci_6alkyl-C(0)-Ci_6alkyl, -0-C1_6alkyl-NH-Ci_6alkyl-O-C1_6 alkyl, -0-C1_6alkyl-NH-C(0)-C1_6alkyl-C(0)0H, and/or -0.-Ci_6alkyl-NH-C(0)-Ci_6alkyl-C(0)-Ci_6alkyl;
(c) a C2 to C15 straight or branched chain alkyl group optionally substituted with one or more hydroxy and/or alkoxy groups; or (d) -C(0)-C6_12ary1-C(0)- wherein the aryl is optionally substituted with one or more hydroxy, halo (e.g., fluoro), nitro, amino, Ci_6alkyl and/or Ci_6alkoxy groups;
a and b are each independently 0, 1, 2, 3 or 4; (preferably 2);
a' and b' are independently 2, 3, 4, 5, 6, 7 or 8; (preferably 2);
d and e are each independently 1, 2, 3, 4, 5 or 6;
d' and e' are each independently 0, 1, 2, 3 or 4; (preferably 0, 1 or 2);
cr and e" are each independently 0, 1, 2, 3 or 4; (preferably 1, 2, 3 or 4);
T is oxygen or sulfur;
x2, -1 and Y2 are each independently null, oxygen, NH, -N(C(0)C1..4alkyl)-, or -N(Ci_4alkyl)-;
W1 and W2 are each independently carbonyl, methylene, sulfone or sulfoxide;
R2, R3, R4, R.5, K and R7 are each independently:
(a) C2 to, C20 straight chain or branched chain alkyl, which is optionally substituted with one or more oxo, halo (preferably fluoro), hydroxy and/or alkoxy groups;
(b) C2 to C20 straight chain or branched chain alkenyl, which is optionally substituted with one or more of oxo, halo (preferably fluoro), hydroxy and/or alkoxy groups;
(c) C2 to C20 straight chain or branched chain alkoxy, which is optionally substituted with one or more oxo, halo (e.g., fluoro), hydroxy and/or alkoxy groups;
(d) -NH-C2_20 straight chain or branched chain alkyl, wherein the alkyl group is optionally substituted with one or more oxo, halo (e.g., fluoro), hydroxy and/or alkoxy groups;
(e) -C(0)-C2_20 straight chain or branched chain alkyl or alkenyl, wherein the alkyl or alkenyl is optionally substituted with one or more oxo, halo (e.g., fluoro), hydroxy and/or alkoxy groups;
(0 Z M
\)N
Z is 0 or NH; and M and N are each independently C2 to C20 straight c haM or branched chain alkyl, alkenyl, alkoxy, acyloxy, alkylamino, or acylamino;
(g) RS
G5j R9 iiR10 R8 is C1_6 straight or branched chain alkyl or C2_6 straight or branched chain alkenyl or alkynyl;
R9 and RI are independently selected from the group consisting of (i) C1 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more halo, oxo, hydroxy and/or alkoxy; and (ii) C2 to C20 straight chain or branched chain alkenyl or alkynyl which is optionally substituted with one or more halo, oxo, hydroxy and/or alkoxy;
GI, G2, G3, G4 and G5 are each independently oxygen, methylene, ¨NH¨, thiol, 4alkyl)¨, ¨N[C(0)¨Ci_4alkyli¨, ¨NH¨C(0)¨, ¨NH¨S02-, -C(0)-0¨, ¨C(0)¨NH¨, -0-C(0)¨, ¨0¨C(0)¨NH¨, ¨0¨C(0)-0¨, ¨NH¨C(0)¨NH¨, -C(0)NH-, -C(0)N(C1-4alkyl), aryl, and -S(0)n¨, where n is 0, 1, or 2;

or GiR2, G2-4, K 03R5 and/or G4R7 may together be a hydrogen atom or hydroxyl;
Z1 and Z2 are each independently selected from -0P(0)(OH)2, -P(0)(011)2, -0P(0)(0R8)(OH) {where R8 is a Ci_4alkyl}, -0S(0)20H, -S(0)20H-, -CO2H, -0B(OH)2, -OH, -CH3, -NH2, and -N(R9)2 {where R9 is a Ci_4alkyl} ;
R12 is H or a C1_4 straight or branched alkyl; and M is independently selected from a hydrogen atom and a pharmaceutically acceptable cation {a monovalent cation will take the place of one M, while a divalent cation will take the place of two M variables}.
In one embodiment, R1 in the compounds of Formula (I)-(V) is -C(0)- or -C(0)-C1_malkyl-C(0)-. In another embodiment, R1 in the compounds of Formula (I)-(V) is -C(0)-.
In one embodiment of the invention, T is oxygen in the compounds of Formula (I)-(V).
In another embodiment, G1, G2, G3 and G4 in the compounds of Formula (I)-(V) are each independently oxygen, -NH-, -NH-C(0)-, -C(0)-0-, -C(0)-NH-, -0-C(0)-, -0-C(0)-NH-, -0-C(0)-0-, -NH-C(0)-NH-, or -C(0)NH-. In another embodiment, G1, G2, G3 and G4 in the compounds of Formula (I)-(V) are each independently oxygen, -C(0)-0- or -0-C(0)-. In another embodiment, G1 and G3 in the compounds of Formula (I)-(V) are -0-C(0)-.
In one embodiment for the compounds of Formula (I)-(V) {preferably compounds of Formula (I)-(11) 1, R2 and R5 are each independently substituents selected from (a), (b), (c), (d) and (f) in the definitions of R2 and R5 herein; R3 and R6 are each independently substituents selected from (a) and (b) in the definitions of R3 and R6 herein; and R4 and R7 are each independently substituents selected from (a), (b), (c) and (e) in the definitions of R4 and R7 herein.
In other embodiments for the compounds of Formula (I)-(V) {preferably compounds of Formula (IV) or (V)}, R2, R3, R4, R5, R6 and R7 are each independently substituents selected from (a), (b), (g) and (h) in the definitions of R2, R3, R4, R5, R6 and R7 herein.
In other embodiments for the compounds of Formulas (I)-(III), one or more of the following is present: each of a and b is 2; each of X1 and Y1 is NH; R1 is -C(0)- or -C(0)-C1_14a1kyl-C(0)-; each of d' and e' is 1; each of cr and e- is 1; x is 0 or NH, more preferably NH; and W is C(0); or each of d' and e' are 2.
In other embodiments for Formulas (1)-(111), R1 is a -C(0)C1_14 alkyl-C(0)-, wherein the C114a1ky1 is substituted, for example, with a C1_5alkoxy group.
In one embodiment, the compounds of Formulas (I)-(11) are "Type 1" wherein the values of a and b are the same; the values of d and e are the same; the values of d' and e' are the same; the values of d" and e" are the same; X1 and Y1 are the same; X2 and Y2 are the same; W1 and W2 are the same; R2 and R5 are the same; G1 and G3 are the same; R3 and R6 are the same; G2 and G4 are the same; and R4 and R7 are the same.
In another embodiment, the compounds of Formulas (I)-(111) are "Type 2"
wherein the values of a and b are different, the values of d and e are the same, the values of d' and e' are different; the values of d" and e" are the same; X1 and Y1 are different; X2 and Y2 are different; W1 and W2 are different; R2 and R5 are different; G1 and G3 are different; R3 and R6 are different; G2 and G4 are different; or R4 and R7 are different.
In another embodiment, the compounds of Formulas (I)-(111) are "Type 3"
wherein the values of a and b are different, the values of d and e are different, the values of d' and e' are different; the values of d" and e" are different; X1 and Y1 are different; X2 and Y2 are different; W1 and W2 are different; R2 and R5 are different; G1 and G3 are different; R3 and R6 are different; G2 and G4 are different; or R4 and R7 are different.
In other embodiments, the compounds of Formulas (I), (1) and/or (ifi) are preferably:
ER 803022; a pharmaceutically acceptable salt thereof other than the sodium -salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

o 0).L(CH2)10CH3 " \/NO(CH2)6CH3 0¨P-0 /
HN 0 Na HN(CH2)12CH3 (-)L/r.0 rsu HN
v kvi 12/10µ..1 13 so¨P¨e cH2,6 _ _yNo( cH
_ _3 0 Na HN(CH2)12CH3 ER 803058; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0 kw. .2110w. .3 it /O¨P-0 0(CH2)6CH3 HN/ 0 Na HN(CH2)12CH3 HN 0 k%./ I 12/10....n 13 O¨P-0 0(CH2)6CH3 0 Na HN(CH2)12CH3 ER 803732; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

o QA(CH)CH3 0¨P-CY (1)(CH2)6CH3 HN 0 Na HN(CH2)12CH3 r irsu A
HN %-1 k%-, I 12110% I 13 \ 0 II
(CH2) 6CH3 0 Na HN(CH2)12CH3 ER 804053; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

(NA
0 kµal 12/10vi 13 I I
O¨P-0 o(CH2)6CH3 HN/¨// I
0 Na (CH2)10CH3 HN 0 0)'L(CH2)10CH3 O¨P-0 07(CH2)6C H3 0 Na Hi\-1 (CH2)10CH3 0 0 =
ER 804058; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

scrr-s.1.4 .3 I I
/0¨P-OYNO(CH2)6CH3 HN/ 0 Na HN (CH2)10CH3 HN
.2/6.,.
/NIA
(l/r12)6µ.../n3 0 Na HN (CH2)10CH3 ER 804059; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0 0)(0H2)50H3 NO(CH2)6CH3 HN/ 0 Na (CH2)10CH3 HNr,u (l./1-12)3 \al 13 \--\

12/6(-saa ..4 o Na (CH2)10CH3 00 =
ER 804442; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0 0 (CH2)10CH3 /0P00(CH2)6CH3 HN/ 0 Na HN,NH-(C1-12)11C0H3 HN 0 0)L(CH2)10CH3 O¨P-0 0(CH2)6CH3 NaMH
HN
(La--12)1 LA--13 0 =

ER 804680; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0 0 0 (CH2)1 0CH3 O-P-0N CH2,)( 1 s6CH3 HN/--/0 NaYNH l,L'nr,"
HN-li2/12L413 HN 00 0).L(CH2)10CH3 rsu 0 Na HN1. (CH2)12CH3 ;or ER 804764; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0 0)L(CH2)10CH3 / 0(CH2)6CH3 HN(CH2)12CH3 )-L
\__\9 0 (CH2)1 oCH3 0 Na (C1- 12)6C3 HN(CH2)12CH3 In one embodiment, the preferred compound is 112066; a stereoisomer thereof;
a pharmaceutically acceptable salt thereof other than the sodium salt shown below;
and/or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0).L(CH2)10CH3 /O-P-0 10(CH2)6CH3 HN 0 Na HN (CH2)10CH3 II II

HN0)L(CF12)10CH3 \__\
O-P-0 o(CH2)6CF13 0 Na HN (CH2)10CH3 In one embodiment, the preferred compound is ER 804057; a pharmaceutically acceptable salt thereof other than the sodium salt shown below; or the one or more sodium cations shown below can be replaced with hydrogen atoms:

0 2/10`-'"3 I I
0¨P-OMO(CH
/ 2)6CH3 HN 0 Na HN (CH2)10CH3 HN (:))/f"NI.J \ r=u kk-ii 12/1 0µ-'"3 0¨P-0 401H
_ ( _ _2)6CH3 0 Na HN (CH2)10CH3 In some embodiments for Formulas (IV) and (V), one or more of the following limitations is present: each of a and b is 2; each of X1 and Y1 is NH; each of d and e is 1 or 2; and each of d' and e' is 0, 1, or 2. In certain preferred embodiments, each of d and e is 1 and each of d' and e' is 0. In certain other preferred embodiments, each of d and e is 1 and each of d' and e is 1 or 2.
In some embodiments for Formulas (IV) and (V), R1 is ¨C(0)¨ or -C(0)-Ci_14alkyl-C(0)¨, wherein the Ci_i4alkyl is optionally substituted with one or two substituents selected from the group consisting of hydroxy, Ci_6alkoxy, C1-6alkyldioxy, C1_6alkylamino, or (aryl)Ci_6alkyl, wherein the aryl moiety of the (aryl)Ci_6alkyl is optionally substituted with Ci_6alkyl, C1_6alkoxy, Ci_6alkylamino, (C1_6alkoxy)C1_6alkylamino, (C1_6alkylamino)C1_6alkoxy, -0-Ci_6alkyl-NH-C1_6alky1-0-Ci_6alkyl, -0-C1_6alkyl-NH-C(0)-Ci_6alkyl-C(0)0H, or -0-Ci_6alkyl-NH-C(0)-Ci_6alkyl-C(0)-Ci_6alkyl.
In some embodiments for Formulas (IV) and (V), G1, G2, G3, and G4 are each independently selected from the group consisting of -NH-C(0)- and -0-C(0)-.
In some embodiments for Formula (IV) and (V), at least two of R2, R3, R4, R5, R6, R7, R9 and R1 are C6-20 straight or branched chain alkyl, alkenyl, or alkynyl; any of which may optionally be substituted with one or more substituents selected from the group consisting of halo, oxo, hydroxy and/or alkoxy. In other embodiments, at least two of R2, R3, R4, R5, R6, R7, R9 and R1 are C8-15 straight or branched chain alkyl, alkenyl, or alkynyl; any of which may optionally be substituted with one or more substituents selected from the group consisting of halo, oxo, hydroxy and alkoxy.
In some embodiments for Formulas (IV) and (V), at least four of R2, R3, R4, R5, R6, R7, R9 and R1 are C6-20 straight or branched chain alkyl, alkenyl or alkynyl;
any of which may optionally be substituted with one or more substituents selected from the group consisting of halo, oxo, hydroxy and alkoxy. In certain preferred embodiments, at least four of R2, R3, R4, R5, R6, 1(-7, R9 and Rth are C8_15 straight or branched chain alkyl, alkenyl or alkynyl; any of which may optionally be substituted with one or more substituents selected from the group consisting of halo, oxo, hydroxy and alkoxy.
In some embodiments for Formulas (IV) and (V), at least six of R2, R3, R4, R5, R6, R7, R9 and R1 are C6-20 straight or branched chain alkyl, alkenyl, or alkynyl; any of which may optionally be substituted with one or more substituents selected from the group consisting of halo, oxo, hydroxy and alkoxy. In other embodiments, at least six of R2, R3, R4, R5, R6, R7, R9 and R1 are C8-15 straight or branched chain alkyl, alkenyl or alkynyl; any of which may optionally be substituted with one or more substituents selected from the group consisting of halo, oxo, hydroxy and alkoxy.
In other embodiments, the invention provides compounds of Formula (I), (II), (HI), (IV) or (V) wherein T is sulfur. In other embodiments, the invention provides compounds of Formula (I), (II), (H), (IV) or (V) wherein T is sulfur; provided that the compound is not Compound No. 804678.

In another embodiment of the invention, there is a proviso that the compounds of Formula (I), (IL) or (III) are not:

0 0 (cF12)14CH3 II

(CH
, - -2,14- 3 0" Na+
______________________ OH 0 0" Na+
I
1._, \-..2\/14n,-,..p II
0 0 (CH2)1 4C H3 or 0 0 ____ =-=õ,..___rrr- (0112)50H3 I I I

01- Na+ (CHACH3 OH

0- Na+

0-P-0 0 _ _ (CH2)5CH3 II
00..,...--......--...õ--iõ_...-=-L.,,, (CH2)5CH3 As used herein, the term "alkyl" includes substituted or unsubstituted, straight or branched chain monovalent or bivalent aliphatic hydrocarbon groups. One skilled in the art will appreciate the distinction between a monovalent alkyl group and a bivalent alkyl group in view of the context of the term "alkyl" in the definition for any particular substituent. When an alkyl is a terminal group, it will be monovalent, such as -CH3, -CH2CH3, -CH2CH2CH3, -CH2CH2CH2CH3, and the like. When an alkyl is between other moieties, such as "-C(0)-C114a1ky1-C(0)-" in the definition of R1, the alkyl group will be bivalent, such as -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH2CH2CH2CH2-, and the like.
As used herein, the term "alkenyl" includes substituted or unsubstituted, straight or branched chain unsaturated monovalent or bivalent aliphatic hydrocarbon groups. The "alkenyl" group can have any number of carbon-carbon double bonds, preferably one or two. One skilled in the art will appreciate the distinction between a monovalent alkenyl group and a bivalent alkenyl group in view of the context of the term "alkenyl" in the definition for any particular substituent. When an alkenyl is a terminal group, it will be monovalent, such as -CH=CH2, -CH=CHCH3, and the like.
When an alkenyl is between other moieties, such as "-C(0)-C144alkenyl-C(0)-"
in the definition of R1, the alkenyl group will be bivalent, such as -CH=CH-, -CH=CHCH2-, -CH2CH=CHCH2-, and the like.
As used herein, the term "aryl" includes substituted or unsubstituted, monovalent or bivalent aromatic hydrocarbon groups. One skilled in the art will appreciate the distinction between a monovalent aryl group and a bivalent aryl group in view of the context of the term "aryl" in the definition for any particular substituent.
When an aryl is a terminal group, it will be monovalent. When an aryl is between other moieties, such as "-C(0)-C6_12aryl-C(0)-" in the definition of R1, the aryl group will be bivalent.
Boc is t-butyloxycarbonyl.
Null with reference to a given substituent means that the substituent is absent, and the chemical groups between which the substituent is positioned are directly attached to each other by way of a covalent bond.
The compounds of Formulas (I), (II), (III), (IV) and (V) may have one or more asymmetric carbon atoms, depending upon the substituents, and can have stereoisomers, which are within the scope of the invention. The compounds of Formulas (I), (II), (LE), (IV) and/or (V) can be administered in the form of a pharmaceutically acceptable salt (e.g., where M in the compounds of Formulas (I), (1), (III), (IV) and/or (V) is a pharmaceutically acceptable cation). The compounds of Formulas (I), (II), (III), (IV) and/or (V) can be administered in the form of a pharmaceutically acceptable salt of a stereoisomer of the compounds.
"Pharmaceutically acceptable salt" refers to salts which retain their biological effectiveness. Pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases. Exemplary pharmaceutically acceptable salts derived from inorganic bases include sodium, potassium, lithium, ammonium, calcium and magnesium salts. Exemplary salts derived from organic bases include salts of primary, secondary and tertiary amines. Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Exemplary salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric , acid, phosphoric acid, and the like. Exemplary salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, furnaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
Methods for making the compounds of Formulas (I), (II), (III), (IV) and (V) are described in US Publication No. 2004/0006242, US Publication No. 2003/0153532, US
Publication No. 2002/0176861, US Publication No. 2002/0049314, U.S. Patent No.
6,551,600, U.S. Patent No. 6,521,776, U.S. Patent No. 6,290,973, and WO
03/099195.
Some compounds of Formulas (I), (II), (III), (IV) and (V) and methods for making them are also described by Hawkins et al, The Journal of Pharmacology and Experimental Therapeutics, 300(2):655-661 (2000); Lien et al, The Journal of Biological Chemistry, 276(3):1873-1880 (2001); Przetak eta!, Vaccine, 21:961-970 (2003); and Seydel et al, Eur. J Immunot, 33:1586-1592 (2003).
Exemplary compounds falling within the scope of the compounds of Formulas (I)-(V) are set forth below.
No. Structure _ _ 0(CH2)5CH3 ,o-p-eyNoccHo6cH.
Hi--- 6 Na HNIry(CH2)10CH3 .
( r HN O'IL(CH2)5CH3 so-i-soc.-(6H2),cH3 0 Na HNy--)I(CH2)10CH3 No. Structure Q(CH2)5CH3 HN/-- 0 Na HN (CH2)10CH3 HN 0)---(CH2)5CH3 0¨P¨eyNo(CH2)6CH3 _ _2,10 _ 6 Na HN(CH1CH3 0)*(CH2)5CF13 0¨P-OrNo(CH2)6CH3 HN / I
0 Na HN (CF12)10CH3 HN 0).(CH2)5CF13 0-1:1)--00(CH2)6CH3 0 Na HN (CH2)10CH3 (31).(CH2)5CF13 /0-11:L00(CH2)6C H3 HN 6 Na HN (CH2)10CH3 HN 0)(CH2)5CH3 0¨P-00(CH2)6CH3 0 Na HN (CH2)10CH3 No. Structure .215,..n÷ i-4 .3 o4-o-/y-N
/
HN/ 0 Na HN (CH2)10CH3 HN ../. .
k2/5.r,... u .3 \----\0-P-00(CF12)6C H3 0 Na HN (CF12)10CH3 OH
7,0-rg-0r-0(0F12)6C H3 HN Na HN (CF12)10CH3 0 Na HNyy(CH2)10CH3 OH

0-P--00(CH2)6C1-13 HN 0 Na HNIry(CH2)6CH3 0 0 (CHACH3 -\---\ 9 'PHACH3 ) - _2,16_ -CH3 (I Na HNICH
0 (CH2)5C H3 No. Structure OH

HN

/¨ 0 Na HN(CH2)10CH3 HN OH
0-1=1)-00(CH2)6CH3 0 Na HN (CH2)10CH3 OH

II

HN 6 Na HN (CH2)6CH3 112048 (CH2,) ,6CH3 ) HN OH
0 Na HNyy(CH 1 CH
_ 2,6 3 0) (CH2,6CH3 OH

"
0-1-eYNO(CH2)6CH3 /
HN/ 0 Na HNr(CH
s 2)6C

00 (CF12)5CH3 HN OH
\\
0 Na1 CH
__2,10 3 No. Structure O cyjr-4-11 I I
0-13-00(CF12)6CF13 HN Na HN (CH2)10CH3 HN OH
0¨P-0 _ _2,110 _ _CH3 0 Na HN,i(CH

0 0 (CH2)ioCH3 O¨P-0 O Na (CH2)10CH3 HN 0)L(CH2)10CH3 0 Na HN (CH2)10CH3 0 0-j(CF12)1oCH3 O Na HN (CH2)10CH3 HN 9A*(CF12)10CH3 \---\ 9 0 Na HN in1-1 (-1-1 \-2,10-3 No. Structure 0 (CH2)10CH3 p¨P-Oo(CH2)6CH3 HN7¨' Na HN (CH2)10CH3 HN 0 (CH2)10CH3 Na HNIry (CH 1 C
_ -2,10 - H3 O 0(CF12)14CH3 II
0-PI 0 (CH2)14CH3 0 a Na+
112071 _________________ OH 0 Na+
II
0 o(CH2)14CH3 ,r- (CH2)5CH3 (CH2)5CH3 (5- Na+

0" Na+
(CH2)5CH3 II

No. Structure oA,irsu \ nu k%.,11215...1 .3 9 _ 0-p-oio -----------"--(cH2)6cH, HN/ _2,6 _ 3 0 Na HN (CH 1 CH
(r 0 0 _ (CH2)5CH3 HN 0(CH2)5CH3 _ 0¨P-0-----No(CH2)6CH3 I
0 Na HN (CH2)6C_H_ 3 sCH2, 0 0.1._. ( )5CH3 o)(tr-,LI N rsu k....112/5..al 13 0 ¨
0¨P-070(CH2)6CH3 / I
HN /0 Na HN (CH ,__ 2)6 _CH 3 (0 0 0 _ (CH2)5CH3 oõJ'ir-q.4 (CH) HN HN k.....2/5, .3 0¨P¨eYNO----.-"-----)'''(CH2)6CH3 O Na HN (CH2)6CH3 sCH2,5CH3 No. Structure 0)--(CH2)5CH3 /0-17-00(CH2)6CH3 HN/ 0 Na HN (CH2)6CH3 112093 0 0 0 (CH2)5CH3 HN 0)¨(CH2)5CH3 0-1=1)-0YNo(CH2)6CH3 0 Na 1 HN (CH CH
2, 6 3 0 (CH2)6CH3 OH

0-P-0O(CH2)6C H3 /
0 Na HN (CH2)6CH3 HN
0 0(CH2)5CH3 HN OH
0--00(cH2)6CH3 0 Na HN (CH 1 CH
_ 2,6 _ 3 0 0.1(^,,,./\=,..-- (CHACH3 OH

/0-P-eYN0(CH2)6CH3 I
0 Na HN (CH CH

2, 6 _ 112099 0 Oy---(CH2)5CH3 HN OH
0-Fi'-eYNo(CH2)6CH3 0 Na HN (C H
Fi2, 6C 3 0 (CH2)6CH3 No. Structure OH

/0-P-00(CH2)6CH3 HN/-0 Na HN1y(CH2)6CH3 0 C5r. (CHACH3 HN O)W-(CH2)5CH3 0---Fi)-eyNo(cH2)6cH3 0 Na HNIry(CH2)10CH3 OH
fi?
/0-P-Oci(CH2)6CH3 HN/--0 Na HNIry(CH2)6CH3 I. 0 0 0(CH2)5CH3 HN OH
0-P-eYNo(CH2)6CH3 O Na HN (CH ) CH
=,,ry, 2 6 3 0 (CH2)5CH3 =

OH

/0-F1)--00(CH2)6CH3 HN/ 0 Na HN (CH2)6CH3 Oy (CH2)5C H3 = 0 HN OH
O-P-00(CH2)6C H3 Na HN (CH 1 CH
yy, 2,6 3 No. Structure OH

¨-00(CH2)6C H3 0 Na HN (CH2)6CH3 sCH2,6CH3 0 ) 112861 .

HN OH
0¨P-00(C H2)6C H3 0 Na HN(CH
_ _2)6 _ _CH3 sCH2, 5CH3 0 O ) y_.

OH

/
,_ 2,6 _ _ HN/ 0 Na HN (CH)CH3 oJ 0 (CH2)5CH3 HN OH
\--\
o¨p¨o=rN0(CH2)6CH3 O Na HN (CH2)6CH3 0 (11.-(CH2)5C H3 OH

o¨ig-orNo(cH2)6c H3 I
HN/
0 Na HN(CH
,_ _2)6 _CH_3 0 ,CH2, 5CH3 HN OH
sCH2, 3 6C
0 Na HN- () H
0 ) ,CH2,6CH3 No. Structure OH

HN/ 0 Na HN (CH 1 CH
,_ _2,6 _ _ 3 0O (CF12)5CH3 113643 0Nr_o 0 HN OH
0¨P¨OrNio(CH2)6C H3 O Na HN (CH2)6CH3 0 Oy(CH2)3CH3 OH

/ r.
0-i (CH2)6v1 13 I
HN /0 Na Hi\lry(CH ,_ _2)6 _ CH_3 0 (CH2)6CH3 ><0 O"'"\16 HN OH
H2)6C H3 _ 2,6 _ 0 Na HIA.ry(CH 1 CH3 (CH2)6CH3 0)(C H2)5C H3 HN/0 Na HN (CH ,_ _2)10 _CH 3 HN 0).(CH2)5C H3 ,_ ¨2,1 ,,CF13 0 Na HN.r(CH

No. Structure OH
/0-1=1)-00(CH2)6C H3 HN 0 Na HN (CH2)6CH3 Cr (CH2)5CH3 _ 2,6 _ 0 Na HN (C1CH3H
0 0 ( sCH2)5CH3 OH

HN/x_ _2,16_ 0 Na HN.-y(CH CH
0 sCH2.5CH3 HN OH
0 Na HN ,__ 2,6 _ _ CH_3 r (CH
, 0 0-(CH2)5CH3 r=L.1 /0¨F13-0 kvi 1216..,1 13 HN

/' 0 Na HN I(CF12)10CH3 HN
0).(CH2)5C H3 \
HO (C H2)6C H3 , ¨2,10 0 Na HN (CH 1 CH3 No. Structure (rsw. u .2)3w. .3 /0-P-eYNO(CH2)6CH3 HN/--0 Na HN (CH2)10CH3 HN 9 0)(CH2)6CH3 NHBoc _ 2,10 _ 3 0 Na HN (CH 1 CH

0¨.-(CH2)6CH3 zo-p-00(0H2)6C H3 Na HN(CF12)12CH3 HN 0).---(CH2)6CH3 0-1:1)-00(CH2)6CH3 0 Na HNy(CH2)12CH3 0)---(CH2)6CH3 /0-F1)-eYN0(CH2)6CH3 HN/ 0 Na HI\k.(CH2)10CH3 CO
HN 0).(CH2)6C H3 ID-0N0(CH2)6C H3 Na HN(CF12)10CH3 No. Structure 9 0(CH2)5CH3 ,0-1=1)-eYNo(CH2)6CH3 HN/ 0 Na HN(CH2)14CH3 HN C:1)(CH2)5CH3 0¨Fi'-00(CH2)6CH3 O Na HN(CF12)14CH3 O
OThCH2)5CH3 it /0-17-00(CH2)6CH3 HN 0 Na HN (CH2)10CH3 HN 0)(CH2)5CH3 \O¨P-00 O _1CH3 2,10 _ Na HN (CH

0)(CH2)5CH3 O
,0-11)-00(CH2)6CH3 HN/ 0 Na HN (CH2)5CH3 HN 0)(CH2)5CH3 O
0¨P-00(CH2)6CH3 Na FINI.(CH2)5CH3 No. Structure 0 , r.0)*L µ-' f kµa"2/10"3 o(CH2)6CH3 HN/¨ Na H1\17(CH2)12CH3 HN 0)L(CH2)10CH3 0-p-eYNo(CH2)6CH3 0 Na HI\J(CH2)12CH3 C1)(CH2)5CH3 II
O-P-0 Q(CH2)6CH3 / I
HN/ 0 Na HNk(CF12)12CH3 HN Ca).(CH2)5CH3 0¨¨eYNC(0H2)6CF13 0 Na EIN.r(CH2)12CH3 0)(CH2)5CH3 /0-11:LeYNo(CH2)6CH3 HN Na HNk(CH2)12CH3 HN
0).(CH2)5CH3 o 0-P-eYN0 NHBoc Na HN(CH2)12CH3 No. Structure cyji1r=LA\r-2/5%...1 II
10-P-0 (2)(CH2)6CH3 HN 6 Na EIN(CH2)12CH3 (-4_1 (-1_4 NHBoc 9 0¨P-00(CH2)6CH3 0 Na HN(CH2)12CH3 0 .3 /0¨P-00(CH2)6CF13 HN/--' 6 Na HN(CH2)12CH3 =
HN
0(CH2)5CH3 \
0 0P00(CH)CH
6 Na HN
NH2 (CH2)12CH3 I I

0).(CH2)5CH3 II
0¨P-0 c)(CH2)6CH3 /
HN/ 0 Na Hi\k(CH2)12CH3 0(CH2)5CH3 0-1-010(CH2)6CH3 0 Na HyCH2)12CH3 No. Structure o i0-P-eY(CH2)6C H3 O Na HN(CH2)12CH3 0)(CH2)5CH3 0-F,)-ey(CH2)6C H3 0 Na HN(CH2)12CH3 0 0)(C H2)6C H3 I I
/0-P-00(CH2)6C H3 I
HN/ 0 Na HN(CH2)12CH3 HN 0)(CH2)5CH3 0 0-p-00(c1-12)6cH3 \¨\ 0 HN-' Na HN(CH2)12CH3 ( 9 0)L(CH2)1 0CH3 / rs"
\-1 (CH2)6vi t3 HN/ 0 Na HN(CH2)12CH3 ) 9 t-N /f,IA2/10%..nL rsu HN kµa113 \--\
0-F1)-0 o(cHoecH3 0 Na HN(CH2)12CH3 No. Structure rsu kvi ovi 13 0(C H2)6 C H 3 Na C\(CH2)12CH3 HN 0A(CH2)10CH3 0(CH2)6C H3 Na 0(CH2)12CH3 frsu r=LA
kµal 12/1 ok,n3 zo--p-0-0(cH2)6C H3 HN Na 6,=,(cH2)12cH3 HN 0 (CH2)10CH3 ,9 o-p-clo(cH2)6C H3 o Na (5,(ci-12)12cF13 p¨P1 I N
(CH216.3 Na HN HN(CH2)12CH3 803596 ><

HN 0).(CH ) CH

0¨ .1)-00(C H2)6 C FI3 0 Na FIN(CH2)12CH3 No. Structure OH

/0¨P-00(CH2)6CH3 HN/--- 0 Na HfCkv(CH2)12CH3 803597 çO

HN OH
0¨P-00(CH2)6C
O Na 1-1F11.(CH2)12CH3 OH

oll õ r.
/\\iu kJ¨F ¨1/41 - ivevr-sui i3 HN/ /
0 Na FIF1(CH2)12CH3 \---\
0¨P-0 ¨(CH2)6C H3 0 Na FIF1(CH2)12CH3 OH
O
io-i2)-oo(cH2)6C H3 HN Na 1-1(CH2)12CH3 HN 0).(CH2)10CH3 O-F1)-oo(cH2)6cH3 O Na HNIr(CH2)12CH3 OH
O
0¨P-00(CH2)6C H3 / I
HN/ 0 Na 41(CH2)12CH3 rsLA
803613 HN rlAirsu 12110%./1 oi)--oNo(CH2)6CH3 0 Na HNy(CH2)12CH3 No. Structure 0 (CI-12)10CH3 \ rµu zu-r-k.) i 1 u kv2/6%-ii HN 0 Na HN(CH2)12CH3 HN 0(CF12)10CH3 o Na HN(CH2)12CH3 r)*LII-14 r11.4 kv. .2/10w. .3 z0¨P-00(CH2)6CH3 HN Na HN(CH2)12CH3 HN rAAA
kir-,sw.

\--\0¨A-00(CF12)6C1-13 0 Na HN(CH2)12CH3 No. Structure kvi 1215rs%.."1t-1 /0¨P-00(C112)6C H3 Na 4.1(CH2)12CH3 HN
HO ..20 0 HN
r.)./r,i_.1 rs1-1 kµ-li 12/5vi 0¨P-00(CH2)6CH3 0 Na HNy(CH2)12CH3 II ACH2)17CH3 /
0 Na HRI(CH2)12CH3 HN

HN
(CH2)17CH3 0 Na HiC(CEI2)12CH3 No. Structure /0-1g--Oci(CH2)17C11-13 /¨ 6 Na H1(01-12)12CH3 HN

HN
\O¨P-00'(CH2)17CH3 0 Na 1-11(CH2)12CH3 0).(CH2)10CH3 /0¨P-00(CH2)6CH3 HN 6 Na HN(CH2)12CH3 803789 . 0 HN 0).(CF12)100H3 0-11D-oNo(cH2)6CH3 0 Na 1-11-<,-(CH2)12CH3 = 0)(CH2)5CH3 /0¨P-00(CH2)6CH3 HN 6 Na (CF12)10CH3 HN(4_1 12/5,-,113 0¨-eYeN0(CH2)6CH3 0 Na HN (CF12)10CH3 No. Structure 9 0 (CH2)10CH3 p¨P-ONo(CH2)6CH3 HN Na HN (CH2)10CH3 yY 0 HN 0 (CH2)10CH3 0¨P-0 0- -(CH2)6C H3 0 Na (CH2)10CH3 (CH2)1 1CH3 / CH2) 00( 6CH3 I
HN 0 Na HNõ.. (CH2)12CH3 >-0 0 (CH2)11CH3 0 sCH2) 0 Na HN/. (CH2)12CH3 \

No. Structure o (CH2)11OH3 0¨ Or-NclC(CH2)6CH3 0 Na HN (CH2)12CH3 HN
I I

HN
(CH2)11 CH3 0 ¨ P ¨ (Y'Y'Ncy (CH2)6C H3 0 Na HN.(CF12)12CH3 0 (CH2)10CH3 (:)(cH2)6C H3 HN N

HN
yNj.L(CH2)10CH3 )o 0 "

HN rsi I
kµ-,1 [2/10k.,n3 \\
0--o NaMOPH2)60H3 Nj.(CF12)10CH3 No. Structure )*
0 (CH2)10CH3 II
/01-00(CH2)6CH3 HN/ 0 Na HN

\
HN 12ji coal 13 \___\
0-1-0C)(0H2)6CH3 0 Na HN

0 (CH2)10CH3 0-P-00(CH2)6CH3 / I
HN/ 0 Na Fin rst ..*'(lif12)13lif13 0 HN 0)L(Ch12)10CH3 O-P-0 - (CH2)6CH3 Na.
frsL_I rsu ."'kk-,n2)13kan3 HNA(CF12)100H3 .../0-Fi)-0(:)(0H2)6CH3 HN II 0 Na HN(CH2)12CF13 HN HN)L(CF12)10CH3 \--\0+-00C(CH2)6CH3 0 Na HN(CH2)12CF13 0).L(CH2)10CH3 p-ILOMNO
_ H3 HN/¨ Na HNo nu HN 0*)(CF12)10CH3 O-P-00(CH2)6CH3 0 Na HN
(CH2)1 3k-,n1 u 13 No. Structure o HN (CH2)10CH3 HN O Na HN
)rC-1(cH2) 11CH
? 3 HN HN(CH2)10CH3 \\ 9 (!) Na HN1-----(cH2)11CH3 II

o O'k(CH2)10CH3 /
HN/ 0 Na N, CH2 804339 ) HN 0)L(CF12)10CH3 0-.1 --00(CH2)6CH3 0 Na õ,_, (l112)13L1r13 0\
0' (CH2)11 CH3 HN""
HN (1) Na HN(CH2)12CH3 0.(CH2)11CH3 HN
0 Na HNIT(CH2)12CH3 No. Structure 0 (CH2)1 oCH3 I I

HN/ 0 Na ACH2)11CH3 HN 0 0 (CH2)10CH3 O-P-0 0(CH2)6CH3 01 NaT
HNy0(CH2)iiCH3 0)L(CH2)10CH3 /0-114-00(CH2)6CH3 HN/-0 Na HN,s,(CH2)11CH3 804558 \\

HN 0'k(CH2)10CH3 0-P-00(CH2)6CH3 o Na HN,,(CH2)iiCH3 (Atr..14 0-P-00(CH2)6CH3 Na HNI.(CH2)12CH3 k%-,112110rs%.,u1 i3 0-P-040(CH2)6CH3 Na HNIr(CH2)12CH3 No. Structure 9 0 ktan2/10µa"3 /0¨P-00'(CH2)6CH3 HN Na / I (µ-,r12)13%-,n3 0 HN 0)L(CH2)10CH3 0¨P-00(CH2)6CH3 O Na N
'PH2)13CH3 0 (CH2)10CH3 HN/ 0 Na HNk,ACH2)12CH3 HN 0-k(CH2)10CH3 0 Na HN(CH2)12CH3 o 0 (CH2)10CH3 /0ONN(CH2)6C H3 HN/ 0 Na HN(CH2)12CH3 HN 9)*L(CH2)10CH3 0¨P¨eYNN
O Na HN(CH2)12CH3 No. Structure 9 0 (CH2)10CH3 Na TN
HN H (CH2)12CH3 804732 HN1.r/ 0 HN 0 0)L(CH2)10CH3 0¨P¨OMN
Na H
Hr\kr. (CH2)12CH3 I I

0 0 (CH2)10CH3 /0-1"-eOJ y(CH2)7CH3 HN/
0 Na HN (CH2)12CH3 0 Il HN 0).(CH2)10CH3 \0_00--\\ (1? .õ---4,-(CH2)7CH3 Na HN (CH2)12CH3 No. Structure 0 0 (CH2)10C1-13 x-(CH2)12CH3 HN HN
)804947 0 0 0 HN
0 0 (CH2)10CH3 N'"¨(/ CH 1 CH
( 2,6 3 X- HN(CH2)12CH3 804638 9 HN)L(CH2)12CH3 01-0 (CH2)10CH3 /--/ 0 Na HN 0 (CH2)10CH3 HN
\--\9 HN(CH2)12C1-13 0-1:1)-0 (CH2)10CI-13 0 Na 0(CH2)10CH3 No. Structure 804666 9 HIV A*(CF12)12CH3 00¨(CH2)9CH3 /¨j 0 Na HN 0(CF12)10C1-13 o HN
\--\ 9 H1\12(CF12)12CH3 0-1-0----(CH2)9CH3 0 Na Oy (CH2)10CH3 804874 9 HN)L(CF12)12CH3 (CF12)10CH3 Na rsu rsu HN \-/v..,r-12)10v1-13 Oro HN
HN (CF12)12CH3 (CF12)10CH3 Na 11(CH2)10CH3 No. Structure 805028 0 HN'-'*(CH2)12CH3 (CH2)10CH3 I
0 Na HN 0 (CH2)10CH3 HN
\
0 HN (CH2)12CH3 0¨P-0 (CH2)10CH3 0 Na Oy. (CH2)1 oCH3 805520 0 HN**L(CF12)12CH3 I I
/0 P (CH2)12CH3 Isomer A 0 Na HN 0 (CH2)1 oCH3 HN
HN*(CH2)12CH3 0 ¨P (CH2)12CH3 0 Na O. (C H2)1 0CH3 )L
805270 0 HN (CH2)12CH3 O¨P-0 (CH2)12CH3 Isomer B HN 0 g./.(CH2)10CH3 HN
(1? HN(CH2)12CH3 (CH2)12CH3 01 Na (CH2)10CH3 No. Structure 805271u (liF12)9k-1113 0¨P-0 0 Na HN (cF12)11cH3 HN
\¨\rsi kvi J2/9vi 0(CH2)9CH3 0 Na (3.(CH2)11CH3 rsu 805274 0 12)9k-,113 O¨P-0 / I (CH)CH
0 Na HN HN (CH2)11CH
\õ/ 3 0 ?
HN
r/\ frj q r.0 12/9v113 II
0¨P-0(CH2)9CH3 01 Na HN (CF12)11CH3 0-4)-0 (CH2)8CH3 /
0 Na HN HN1r(CH2)12CH3 ) 0 0 0 HN
r.0 \ 0 kvi 12/12tri 13 (CH2)8CH3 0 Na HNIr(CH2)12CH3 No. Structure 805329 0 EINI)(CF12)12CH3 (CHACH3 0¨Fi)N-0 z__/ a HN HN (CF12)12CH3 HN
(R HNA(CH2)12CH3 0 Na HN (CF12)120H3 805517 0 HIA(CH2)12CH3 /0¨ 0 12/7saii3 0 Na (CH2)1 CH3 HN

HN
HN)L(CF12)12CH3 0-7-0 1.4 0 Na 0 (CH2)11 CH3 =

805518 0 HN)L'(CF12)12CH3 II
O¨P ¨ 0 (CH
/ I
2)8CH3 Isomer A
0 Na HN (CH2)1 oCH3 HN o HN(CH2)12CH3 \ II
0¨P-0 (CH2)8CH3 0 Na Olr (CH2)1 oCH3 No. Structure 805519 0 HN).L(CH2)12CI-13 O¨P-0 (CH2)8CH3 I
Isomer B / 0 Na HN 0 (CH2)10CH3 HN
.) \¨\ HNL (CH2)12CH3 0-7-0 - (cH2)8cH3 0 Na 0( (CH2)10CH3 Examples Example 1 Intraperitoneal Administration of a TLR Agonist Enhances Therapeutic Efficacy of a Vaccine To determine the effect of E6020 when administered intraperitoneally with a cancer vaccine, e.g., granulocyte-macrophage colony stimulating factor (GM-CSF) secreting tumor cells, a mouse model using melanoma cells was used. E6020 is a TLR-4 (Toll-like receptor-4) agonist. B6 mice (C57BL/6 mice) were engrafted subcutaneously with 1 x 106 syngeneic B 16F10 murine melanoma cells. Three days after tumor cell inoculation, the mice were either (1) vaccinated subcutaneously (s.c.) with 1 x 106 B16F10 tumor cells that were genetically modified to stably express and secrete murine GM-CSF (B16-GM-CSF cells); (2) vaccinated intraperitoneally (i.p.) with E6020; or (3) were treated with a combination of s.c. GM-CSF cell vaccination and i.p. E6020 (the GM-CSF cell vaccination and E6020 vaccination were administered at separate sites in the mice). In these experiments, the GM-CSF
cells were inactivated by gamma-irradiation prior to inoculation. Survival of the animals was monitored.
These experiments demonstrated that vaccination of the animals with the B16 GM-CSF cells and i.p. E6020 enhanced the therapeutic efficacy of the GM-CSF
cells (Figure 1).
Example 2 Local Administration of a TLR Agonist Enhances the Therapeutic Efficacy of a Cancer Vaccine The effect of E6020 on treatment of B6 mice that were engrafted subcutaneously with 1 x 106syngeneic B16F10 murine melanoma cells was examined.
When tumors became palpable, the mice were injected intratumorally with GM-CSF

cells alone, or in combination with E6020 (about 3-10 tg). Survival of the animals was monitored.
It was found that the population of animals treated intratumorally with a combination of GM-CSF cells and E6020 had increased survival compared to animals that were untreated or treated with GM-CSF cells alone (Figure 2).

Example 3 MUC-1/E6020 Vaccine Therapeutic Effects To test the effects of a MUC-1 vaccine with E6020 adjuvant for treating inflammatory bowel disease (IBD) and subsequent development of colon adenocarcinoma, an engineered mouse strain that lacks the IL-10 gene and expresses transgenic human MUC1 was used. Such mice spontaneously develop intestinal inflammation resembling IBD followed by colon adenocarcinoma. These data were presented and published in Beatty et al., AACR Annual Meeting 2006, Washington D.C., April 4, 2006.
In these experiments, mice were immunized intranasally with 30 mg/flare of Tn MUC100mer (HGVTSAPDTRPAPGSTAPPA) x 5, SEQ ID NO:1) and 3 mg of E6020. Animals were vaccinated at about 4.5 weeks and boosted at about 6.5 weeks and 9 weeks.
MUC1 IL-10 -/- mice treated with vaccine and E6020 had delayed onset of IBD or did not develop IBD during the period of the experiment (Figure 3).
Mice treated with vaccine and E6020 had improved survival and did not develop colon cancer (Figure 4 and Table 1).
Table 1 Treatment/no, of mice treated Age (weeks) Colon tumors Vaccine/6 mice 14 - 35.5 0/6 Adjuvant/3 mice 12.5 - 18 3/3 No treatment/4 mice 8 - 15.5 4/4 The data demonstrate that the addition of E6020 to the MUC-1 vaccine can slow the progression to rectal prolapse associated with IBD, and suppresses the appearance of histologically detected tumors.
Example 4 EGFRvIll Therapeutic Effects with Adjuvant To determine whether an adjuvant can enhance the effect of an oncology antigen (i.e., an antigen that can be used to vaccinate an animal against a cancer), C57/BL6J mice were immunized subcutaneously with a tumor-associated peptide, LEEKKGNYVVTDHC (SEQ ID NO: 2) (derived from a mutant form of EGFR, EGFRv111) conjugated to the protein carrier keyhole limpet hemocyanin (KLH), with or without E6020 or murine GM-CSF, a cytokine used in cancer vaccine trials to boost immune response. E6020 was dosed at 3 tg, GM-CSF at 5 pig and the peptide-KLH conjugate at 2511g per dose. Mice were immunized three times at intervals of three weeks. Serum was prepared from mice two weeks after each immunization and tested for EGFRvIll peptide-specific antibodies using ELISA on plates coated with EGFRvIll peptide conjugated to bovine serum albumin. The results in Table 2 are presented as titers from individual animals. The titer is defined as the last serum dilution at which a signal at 0.25 OD units above background was observed.
The data from these experiments demonstrate that E6020 enhanced the mean titer of antigen-specific IgG2a, which binds high affinity Fc receptors involved in antibody-dependent cell-mediated cytotoxicity (ADCC). IgG2a is the mouse correlate of the human IgG1 isotype that is used in currently marketed human anti-tumor monoclonal antibodies, because it is most efficacious in tumor killing.
The combination of E6020 and GM-CSF in the vaccination with EGFRvIII
peptide demonstrated a greater effect on IgG2a titers than either material alone. These data demonstrate the usefulness of combinations of E6020 with other immunoenhancers.
Table 2. Antibody titers to EGFRvIII peptide are enhanced by E6020 Antigen administered with: PBS E6020 GM-CSF

Antibody subclass: IgG1 IgG2a IgG1 IgG2a IgG1 IgG2a IgG1 IgG2a Geometric mean titer: 22008 The scope of the claims should not be limited by the preferred embodiments set forth in the examples, but should be given the broadest interpretation consistent with the description as a whole.

Claims (27)

1. A composition, comprising:
at least one immunotherapeutic agent selected from one or more cancer antigens; one or more antigens from a virus associated with cancer; one or more anti-cancer antibodies; and one or more anti-idiotypic antibodies to an anti-cancer antibody; and one or more compounds of formula (I) wherein:
R1 is:
(a) -C(O)-;
(b) -C(O)-C1-14 alkyl-C(O)- or-C(O)-C1-14 alkenyl-C(O)-;

wherein the -C1-14 alkyl- or -C1-14 alkenyl- is optionally substituted with one or more substituents selected from the group consisting of hydroxy, C1-6 alkyl, C1-6 alkoxy, C1-6 alkyldioxy, C1-5 alkylamino, carboxy, C1-6 alkoxycarbonyl, C1-6 carbamoyl, C1-6 acylamino, and (aryl)C1-6 alkyl; and wherein the aryl moiety of the (aryl)C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, C1-6 alkylamino, C1-6 alkoxyamino, C1-6 alkylamino-C1-6 alkoxy, -O-C1-6 alkylamino-C1-6 alkoxy, -O-C1-6 alkylamino-C(O)-C1-6 alkyl-C(O)OH, -O-C1-6 alkylamino-C(O)-C1-6 alkyl-C(O)-C1-6 alkyl, -O-C1-6 alkyl-NH-C1-6 alkyl-O-C1-6 alkyl, -O-C1-6 alkyl-NH-C(O)C1-6 alkyl-C(O)OH, and -O-C1-6 alkyl-NH-C(O)C1-6 alkyl-C(O)-C1-6 alkyl;
(c) a C2 to C15 straight or branched chain alkyl group optionally substituted with one or more hydroxyl or alkoxy groups; or (d) -C(O)-C6-12 aryl-C(O)- wherein the aryl is optionally substituted with one or more group selected from the group consisting of hydroxy, halo, nitro, amino, C1-6 alkyl and C1-6 alkoxy groups;
a and b are each independently 0, 1, 2, 3 or 4;
d and e are each independently 1, 2, 3, 4, 5 or 6;
d' and e' are each independently 0, 1, 2, 3 or 4;
d" and e" are each independently 0, 1, 2, 3 or 4;
T is oxygen or sulfur;
X1, X2, Y1 and Y2 are each independently null, oxygen, NH, -N(C(O)C1-4 alkyl)-, or -N(C1-4 alkyl)-;
W1 and W2 are each independently selected from the group consisting of carbonyl, methylene, sulfone and sulfoxide;
R2, R3, R4, R5, R6 and R7 are each independently:

a) C2 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
b) C2 to C20 straight chain or branched chain alkenyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
c) C2 to C20 straight chain or branched chain alkoxy, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
d) -NH-C2-20 straight chain or branched chain alkyl, wherein the alkyl group is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
e) -C(O)-C2-20 straight chain or branched chain alkyl or alkenyl, wherein the alkyl or alkenyl is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
Z is O or NH; and M' and N are each independently C2 to C20 straight chain or branched chain alkyl, alkenyl, alkoxy, acyloxy, alkylamino, or acylamino; or R8 is C1-6 straight or branched chain alkyl or C2-6 straight or branched chain alkenyl or alkynyl;
R9 and R19 are independently selected from the group consisting of (i) C1 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy; and (ii) C2 to C20 straight chain or branched chain alkenyl or alkynyl which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy;
G1, G2, G3, G4 and G5 are each independently oxygen, methylene, -NH-, thiol, -N(C1-4 alkyl)-, -N[C(O)-C1-4 alkyl]-, -NH-C(O)-, -NH-SO2-, -C(O)-O-, -C(O)-NH-, -O-C(O)-, -O-C(O)NH-, -O-C(O)O-, -NH-C(O)-NH-, -C(O)NH-, -C(O)N(C1-4 alkyl), aryl, or -S(O)n-, wherein n is 0, 1, or 2;
or G2R4 or G4R7 may together be a hydrogen atom or hydroxyl; and M is independently selected from a hydrogen atom and a pharmaceutically acceptable cation;
or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
2. The composition of claim 1, wherein the immunotherapeutic agent is one or more cancer antigens.
3. The composition of claim 2, wherein the cancer antigen is MUC-1-
4. The composition of claim 2, wherein the cancer antigen is selected from the group consisting of mutant B-raf, HER-2/neu, epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), prostate specific antigen (PSA), prostate specific membrane antigen (PSMA), GP-100, carcinoembryonic antigen (CEA), CA 19.9, MART-1, K-ras, N-ras, H-ras, and p53.
5. The composition of claim 4, wherein the cancer antigen is selected from EGFR variants 1-4.
6. The composition of claim 4, wherein the cancer antigen is selected from VEGFR variants 1-2.
7. The composition of claim 1, wherein the antigen from a virus associated with cancer is human papilloma virus (HPV), hepatitis B or C virus (HBV or HCV), Epstein-Barr virus (EBV), human herpes virus-8 (HHV-8), human T-Iymphotrophic virus (HTLV)-1, or HTLV-2.
8. The composition of claim 1, wherein the anti-cancer antibody is trastuzumab, bevacizumab, rituximab, pertuzumab, cetuximab, IMC-1C11, BEXXAR®, ZEVALIN®, EMD 7200, SGN-30, SGN-15, SGN-30, SGN-40, SGN-35, or SGN-17/19.
9. The composition of claim 1, wherein the anti-idiotypic antibody recognizes the antigen-binding site of an antibody to MUC-1, mutant B-raf, HER-2/neu, EGFR, VEGFR, PSMA, GP-100, CEA, CA 19.9, MART-1, K-ras, N-ras, H-ras, or p53.
10. The composition of claim 9, wherein the anti-idiotypic antibody recognizes the antigen-binding site of an antibody to EGFR variants 1-4.
11. The composition of claim 9, wherein anti-idiotypic antibody recognizes the antigen-binding site of an antibody to VEGFR variants 1-2.
12. The composition of claim 1, wherein the compound is selected from ER 803022, ER 803058, ER 803732, ER 804053, ER 804058, ER 804059, ER 804442, ER 804680, ER 804764, ER 112066, and ER 804057, or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
13. The composition of claim 12 wherein the compound is ER 804057 and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
14. The composition of claim 1, wherein the immunotherapeutic agent is one or more cancer antigens and the composition further comprises a second immunotherapeutic agent.
15. Use of at least one immunotherapeutic agent selected from one or more cancer antigens; one or more viral antigen associated with cancer; one or more anti-cancer antibodies; and one or more anti-idiotypic antibodies to an anti-cancer antibody; and one or more compound of formula (I) wherein:
R1 is:
(a) -C(O)-;
(b) -C(O)-C1-14 alkyl-C(O)- or-C(O)-C1-14 alkenyl-C(O)-;
wherein the -C1-14 alkyl- or -C1-14 alkenyl- is optionally substituted with one or more substituents selected from the group consisting of hydroxy, C1-6 alkyl, C1-6 alkoxy, C1-6 alkyldioxy, C1-5 alkylamino, carboxy, C1-6 alkoxycarbonyl, C1 -6 carbamoyl, C1-6 acylamino and (aryl)C1-6 alkyl; and wherein the aryl moiety of the (aryl)C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, C1-6 alkylamino, C1-6 alkoxyamino, C1-6 alkylamino-C1-6 alkoxy, -O-C1-6 alkylamino-C1-6 alkoxy, -O-C1-6 alkylamino-C(O)-C1-6 alkyl-C(O)OH, -O-C1-6 alkylamino-C(O)-C1-6 alkyl-C(O)-C1-6 alkyl, -O-C1-6 alkyl-NH-C1-6 alkyl-O-C1-6 alkyl, -O-C1-6 alkyl-NH-C(O)C1-6 alkyl-C(O)OH and alkyl-NH-C(O)C1-6 alkyl-C(O)-C1-6 alkyl;
(c) a C2 to C15 straight or branched chain alkyl group optionally substituted with one or more hydroxyl or alkoxy groups; or (d) -C(O)-C6-12 aryl-C(O)- wherein the aryl is optionally substituted with one or more group selected from the group consisting of hydroxy, halo, nitro, amino, C1-6 alkyl and C1-6 alkoxy groups;
a and b are each independently 0, 1, 2, 3 or 4;
d and e are each independently 1, 2, 3, 4, 5 or 6;
d' and e' are each independently 0, 1, 2, 3 or 4;
d" and e" are each independently 0, 1, 2, 3 or 4;
T is oxygen or sulfur;
X1, X2, Y1 and Y2 are each independently null, oxygen, NH, -N(C(O)C1-4 alkyl)-, or -N(C1-4 alkyl)-;
W1 and W2 are each independently selected from the group consisting of carbonyl, methylene, sulfone and sulfoxide;
R2, R3, R4, R5, R6 and R7 are each independently:
a) C2 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
b) C2 to C20 straight chain or branched chain alkenyl, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
c) C2 to C20 straight chain or branched chain alkoxy, which is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
d) -NH-C2-20 straight chain or branched chain alkyl, wherein the alkyl group is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;

e) -C(O)-C2-20 straight chain or branched chain alkyl or alkenyl, wherein the alkyl or alkenyl is optionally substituted with one or more groups selected from the group consisting of oxo, halo, hydroxy and alkoxy groups;
Z is O or NH; and M' and N are each independently C2 to C20 straight chain or branched chain alkyl, alkenyl, alkoxy, acyloxy, alkylamino, or acylamino; or R8 is C1-6 straight or branched chain alkyl or C2-6 straight or branched chain alkenyl or alkynyl;
R9 and R10 are independently selected from the group consisting of (i) C1 to C20 straight chain or branched chain alkyl, which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy; and (ii) C2 to C20 straight chain or branched chain alkenyl or alkynyl which is optionally substituted with one or more groups selected from the group consisting of halo, oxo, hydroxy and alkoxy;
G1, G2, G3, G4 and G5 are each independently oxygen, methylene, -NH-, thiol, -N(C1-4 alkyl)-, -N[C(O)-C1-4 alkyl]-, -NH-C(O)-, -NH-SO2-, -C(O)-O-, -C(O)-NH-, -O-C(O)-, -O-C(O)NH-, -O-C(O)O-, -NH-C(O)-NH-, -C(O)NH-, -C(O)N(C1-4 alkyl), aryl, or -S(O)n-, wherein n is 0, 1, or 2;
or G2R4 or G4R7 may together be a hydrogen atom or hydroxyl; and M is independently selected from a hydrogen atom and a pharmaceutically acceptable cation;
or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof, for stimulating an immune response in a subject individual.
16. The use of claim 15, wherein the immunotherapeutic agent and the compound of formula (I) are to be used at about the same time.
17. The use of claim 15, wherein the immunotherapeutic agent and the compound of formula (I) are to be used separately.
18. The use of claim 15, wherein the subject individual is at risk for developing cancer, diagnosed with a cancer, in treatment for cancer, or in post-therapy recovery from cancer.
19. The use of claim 18, in combination with a surgical procedure to remove or reduce the size of a cancer tumor, radiation therapy, chemotherapy, and/or ablation therapy.
20. The use of claim 18, to stabilize a tumor by preventing or slowing the growth of the existing cancer, to prevent the spread of a tumor or of metastases, to reduce the tumor size, to prevent the recurrence of treated cancer, or to eliminate cancer cells not killed by earlier treatments.
21. The use of claim 15, in combination with a cancer therapy agent.
22. The use of claim 21, wherein the cancer therapy agent is dendritic cells, chemokines, cytokines, tumor necrosis factors, chemotherapeutic agents, adenosine analogs, alkyl sulfanates, anti-tumoral antibiotics, aziridines, camptothecin analogs, cryptophycins, dolastatins, enedyine anticancer drugs, epipodophyllotoxins, folate analogs, maytansinoids, microtubule agents, nitrogen mustards, nitrosoureas, nonclassic alkylators, platinum complexes, purine analogs, pyrimidine analogs, substituted ureas, anti-angiogenic agents, biologic agents, antibodies and fragments thereof, anti-emetics, epithelial growth factor inhibitors, anti-mucositic agents, anti-osteoclastic agents, hormone regulating agents, hematopoietic growth factors, anti-toxicity agents or mixtures of two or more thereof.
23. The use of claim 22, wherein the cancer therapy agent is TNF-.alpha., cladribine, pentostatin, busulfan, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, mitomycin, thiotepa, irinotecan, topotecan, cryptophycin 52, cryptophicin 1, dolastatin 10, dolastatin 15, esperamicin, calicheamicin, dynemicin, neocarzinostatin, neocarzinostatin chromophore, kedarcidin, kedarcidin chromophore, C-1027 chromophore, etoposide, teniposide, methotrexate, maytansinol, maytansinol analogues, docetaxel, paclitaxel, vinblastine, vincristine, vinorelbine, chlorambucil, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, melphalan, carmustine, lamustine, streptoxacin, altretamine, dacarbazine, procarbazine, temozolamide, carboplatin, cisplatin, fludarabine, mercaptopurine, thioguanine, capecitabine, cytarabine, depocyt, floxuridine, fluorouracil, gemcitabine, hydroxyurea, canstatin, troponin I, ZD
1839, virulizin, interferon, anti EGFR, anti-HER-2/neu, anti-KDR, IMC-C225, lorazepam, metroclopramide, domperidone, transforming growth factor beta 1, dyclonine, lignocaine, azelastine, glutamine, corticoid steroids, allopurinol, bisphosphonates, etidronate, pamidronate, ibandronate, osteoprotegerin, anti-androgens, luteinizing hormone releasing hormone agonists, anastrozole, tamoxifen, amifostine or mixtures of two or more thereof.
24. The use of claim 15, to prevent or delay the development of cancer.
25. The use of claim 15, in combination with an immunostimulatory compound.
26. The use of claim 25, wherein said immunostimulatory compound is a toll like receptor (TLR) agonist, N-acetylmuramyl-L-alanine-D-isoglutamine (MDP), lipopolysaccharides (LPS), genetically modified and/or degraded LPS, alum, glucan, colony stimulating factors, interferons, interleukins, major histocompatability complex Class ll binding peptides, saponins, unmethylated CpG sequences, 1-methyl tryptophan, arginase inhibitors, cyclophosphamide, or antibodies that block immunosuppressive functions, or mixtures of two or more thereof.
27. The use of claim 26, wherein said immunostimulatory compound is TLR4, TLR7, TLR9, erythropoietin, granulocyte macrophage colony-stimulating factor, granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor, pegylated G-CSF, stem cell factor, interleukin (IL)-3, IL6, PIXY 321, y-interferon, a-interferon, IL-2, IL-7, IL-12, IL-15, IL-18, QS21, anti-CTLA4 antibodies, or mixtures of two or more thereof.
CA2605749A 2005-04-26 2006-04-26 Compositions and methods for cancer immunotherapy Active CA2605749C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US67468005P 2005-04-26 2005-04-26
US60/674,680 2005-04-26
PCT/US2006/015668 WO2006116423A2 (en) 2005-04-26 2006-04-26 Compositions and methods for cancer immunotherapy

Publications (2)

Publication Number Publication Date
CA2605749A1 CA2605749A1 (en) 2006-11-02
CA2605749C true CA2605749C (en) 2015-06-30

Family

ID=37215428

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2605749A Active CA2605749C (en) 2005-04-26 2006-04-26 Compositions and methods for cancer immunotherapy

Country Status (8)

Country Link
US (3) US7976852B2 (en)
EP (1) EP1874342B1 (en)
JP (2) JP5185813B2 (en)
KR (1) KR101205064B1 (en)
CN (2) CN101355928B (en)
AU (1) AU2006241206B2 (en)
CA (1) CA2605749C (en)
WO (1) WO2006116423A2 (en)

Families Citing this family (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6835721B2 (en) 1999-02-01 2004-12-28 Eisai Co., Ltd. Immunomodulatory compounds and methods of use thereof
US20040006242A1 (en) * 1999-02-01 2004-01-08 Hawkins Lynn D. Immunomodulatory compounds and method of use thereof
US7915238B2 (en) * 1999-02-01 2011-03-29 Eisai R & D Management Co., Ltd. Immunomodulatory compounds and methods of use thereof
US7906492B2 (en) * 2001-01-16 2011-03-15 Sloan-Kettering Institute For Cancer Research Therapy-enhancing glucan
US7507724B2 (en) * 2001-01-16 2009-03-24 Sloan-Kettering Institute For Cancer Research Therapy-enhancing glucan
US20070292418A1 (en) * 2005-04-26 2007-12-20 Eisai Co., Ltd. Compositions and methods for immunotherapy
CN101355928B (en) 2005-04-26 2013-05-22 卫材R&D管理株式会社 Compositions and methods for cancer immunotherapy
WO2007070660A2 (en) 2005-12-13 2007-06-21 President And Fellows Of Harvard College Scaffolds for cell transplantation
US20090053221A1 (en) * 2006-01-17 2009-02-26 Cheung Nai-Kong V Immune response enhancing glucan
US8323644B2 (en) * 2006-01-17 2012-12-04 Sloan-Kettering Institute For Cancer Research Therapy-enhancing glucan
US9770535B2 (en) * 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
WO2009003082A2 (en) * 2007-06-26 2008-12-31 Vanderbilt University Immunological compositions as cancer biomarkers and/or therapeutics
EP2215471B1 (en) 2007-10-29 2012-02-08 Eisai R&D Management Co., Ltd. Methods for prognosing the ability of a zearalenone analog compound to treat cancer
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
CN102006891B (en) 2008-02-13 2017-04-26 哈佛学院董事会 Continuous cell programming devices
WO2011063336A2 (en) * 2009-11-20 2011-05-26 President And Fellows Of Harvard College Secondary site of antigen stimulation for therapeutic vaccination
US9012399B2 (en) 2008-05-30 2015-04-21 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
EP2310006A4 (en) * 2008-07-03 2012-04-25 Mayo Foundation Treating cancer
HUE026194T2 (en) * 2009-03-24 2016-05-30 Transgene Sa Biomarker for monitoring patients
WO2010120749A2 (en) 2009-04-13 2010-10-21 President And Fellow Of Harvard College Harnessing cell dynamics to engineer materials
JP5926180B2 (en) 2009-07-31 2016-05-25 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Methods of cell programming for tolerogenic therapy
CN102859355B (en) * 2009-08-24 2015-10-07 基因泰克公司 The susceptibility of cell to B-RAF inhibitor for treating is measured by detection KRAS and RTK expression
EP2542230A4 (en) 2010-03-05 2013-08-28 Harvard College Enhancement of skeletal muscle stem cell engrafment by dual delivery of vegf and igf-1
BR112012027745A2 (en) * 2010-04-27 2017-01-10 Univ Johns Hopkins method and immunogenic composition for treatment of neoplasia
KR101477194B1 (en) * 2010-05-27 2014-12-29 심천 사루브리스 퍼머수티칼스 컴퍼니 리미티드 Chemical synthesis and anti-tumor and anti-metastatic effects of dual functional conjugate
EP2585053A4 (en) 2010-06-25 2014-02-26 Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
AU2011281192A1 (en) 2010-07-19 2013-02-28 Yeda Research And Development Co. Ltd. Peptides based on the transmembrane domain of a Toll-like receptor (TLR) for treatment of TLR-mediated diseases
CN103458902B (en) * 2010-10-01 2017-11-07 帆德制药股份有限公司 TLR activators and the treatment use of therapeutic alliance
PT2624873T (en) 2010-10-06 2020-03-04 Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
WO2012064697A2 (en) 2010-11-08 2012-05-18 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
WO2012105219A1 (en) * 2011-01-31 2012-08-09 オリンパス株式会社 Antibody therapy effect-enhancing drug
WO2012148684A1 (en) 2011-04-27 2012-11-01 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
ES2878089T3 (en) 2011-04-28 2021-11-18 Harvard College Injectable preformed macroscopic three-dimensional scaffolds for minimally invasive administration
US9427477B2 (en) 2011-05-09 2016-08-30 Mayo Foundation For Medical Education And Research Cancer treatments
MX371526B (en) * 2011-05-27 2020-01-31 Ambrx Inc Compositions containing, methods involving, and uses of non-natural amino acid linked dolastatin derivatives.
EP2714073B1 (en) 2011-06-03 2021-03-10 President and Fellows of Harvard College In situ antigen-generating cancer vaccine
WO2013019620A2 (en) * 2011-07-29 2013-02-07 Glaxosmithkline Llc Method of treating cancer using combination of braf inhibitor, mek inhibitor, and anti-ctla-4 antibody
CN108434440B (en) * 2011-09-06 2022-08-23 新加坡科技研究局 Polypeptide vaccine
EP2766384B1 (en) 2011-10-10 2016-11-23 Yeda Research and Development Co. Ltd. Toll-like receptor 4 (tlr-4) agonist peptides for modulating tlr-4 mediated immune response
ES2773895T3 (en) 2012-04-16 2020-07-15 Harvard College Mesoporous Silica Compositions to Modulate Immune Responses
SG11201408153YA (en) * 2012-06-07 2015-01-29 Ambrx Inc Prostate-specific membrane antigen antibody drug conjugates
CN112587658A (en) * 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
EP2903610B1 (en) 2012-10-01 2021-11-03 Mayo Foundation For Medical Education And Research Cancer treatments
CN103768596B (en) * 2012-10-17 2016-06-22 苏州丁孚靶点生物技术有限公司 For the combination product of oncotherapy, its purposes and correlation technique
AU2013355337B2 (en) * 2012-12-03 2018-07-12 Merrimack Pharmaceuticals, Inc. Combination therapy for treating HER2-positive cancers
US10548985B2 (en) 2014-01-10 2020-02-04 Birdie Biopharmaceuticals, Inc. Compounds and compositions for treating EGFR expressing tumors
EP3137105A4 (en) 2014-04-30 2017-12-27 President and Fellows of Harvard College Combination vaccine devices and methods of killing cancer cells
CN113134095A (en) 2014-06-16 2021-07-20 梅约医学教育与研究基金会 Treatment of myeloma
CN105233291A (en) * 2014-07-09 2016-01-13 博笛生物科技有限公司 Combined therapy composition and combined therapy method for treating cancers
DK3166976T3 (en) 2014-07-09 2022-04-11 Birdie Biopharmaceuticals Inc ANTI-PD-L1 COMBINATIONS FOR TREATMENT OF TUMORS
US11219672B2 (en) 2014-08-07 2022-01-11 Haruki Okamura Therapeutic agent for cancer which comprises combination of IL-18 and molecule-targeting antibody
CN112587672A (en) 2014-09-01 2021-04-02 博笛生物科技有限公司 anti-PD-L1 conjugates for the treatment of tumors
US9446148B2 (en) 2014-10-06 2016-09-20 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
GB201501302D0 (en) * 2015-01-27 2015-03-11 Ostara Biomedical Ltd Embryo implantation
EP3250250A4 (en) 2015-01-30 2019-05-22 President and Fellows of Harvard College Peritumoral and intratumoral materials for cancer therapy
CN107708756A (en) 2015-04-10 2018-02-16 哈佛学院院长等 Immunocyte acquisition equipment and its preparation and application
US10842763B2 (en) 2015-07-31 2020-11-24 The Johns Hopkins University Methods for cancer and immunotherapy using prodrugs of glutamine analogs
CN108135875B (en) 2015-07-31 2021-12-31 约翰霍普金斯大学 Methods and compositions for treating metabolic reprogramming disorders
SI3328827T1 (en) 2015-07-31 2023-11-30 The Johns Hopkins University Prodrugs of glutamine analogs
TW201707725A (en) 2015-08-18 2017-03-01 美國馬友醫藥教育研究基金會 Carrier-antibody compositions and methods of making and using the same
TW201713360A (en) 2015-10-06 2017-04-16 Mayo Foundation Methods of treating cancer using compositions of antibodies and carrier proteins
MA44334A (en) 2015-10-29 2018-09-05 Novartis Ag ANTIBODY CONJUGATES INCLUDING A TOLL-TYPE RECEPTOR AGONIST
CN106943597A (en) 2016-01-07 2017-07-14 博笛生物科技(北京)有限公司 Anti-EGFR for treating tumour is combined
CN115350279A (en) 2016-01-07 2022-11-18 博笛生物科技有限公司 anti-HER 2 combinations for the treatment of tumors
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
CN115554406A (en) 2016-01-07 2023-01-03 博笛生物科技有限公司 anti-CD 20 combinations for the treatment of tumors
EP3411475A4 (en) 2016-02-06 2019-09-11 President and Fellows of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11351254B2 (en) 2016-02-12 2022-06-07 Mayo Foundation For Medical Education And Research Hematologic cancer treatments
CN107126563B (en) * 2016-02-26 2021-09-10 博生吉医药科技(苏州)有限公司 Composition containing low-dose antibody for blocking VEGF signal path and application thereof
CA3018340A1 (en) 2016-03-21 2017-09-28 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug
WO2017165440A1 (en) 2016-03-21 2017-09-28 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US10618969B2 (en) 2016-04-06 2020-04-14 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
AU2017295704B2 (en) 2016-07-13 2023-07-13 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
EP4177271A1 (en) 2016-09-01 2023-05-10 Mayo Foundation for Medical Education and Research Carrier-pd-l1 binding agent compositions for treating cancers
US11160876B2 (en) 2016-09-01 2021-11-02 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
EP3509635A1 (en) 2016-09-06 2019-07-17 Vavotar Life Sciences LLC Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
CA3035653A1 (en) 2016-09-06 2018-03-15 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
EP3510048A1 (en) 2016-09-06 2019-07-17 Mayo Foundation for Medical Education and Research Methods of treating pd-l1 expressing cancer
WO2018160993A1 (en) * 2017-03-03 2018-09-07 Obsidian Therapeutics, Inc. Compositions and methods for immunotherapy
WO2018166298A1 (en) * 2017-03-13 2018-09-20 Fudan University Immunopotentiator, immunotherapeutic pharmaceutical composition and its preparation and use
CN108567977B (en) * 2017-03-13 2022-04-12 复旦大学 Immunopotentiator, immunotherapy pharmaceutical composition, preparation and application thereof
CN108794467A (en) 2017-04-27 2018-11-13 博笛生物科技有限公司 2- amino-quinoline derivatives
BR112019027025A2 (en) 2017-06-23 2020-06-30 Birdie Biopharmaceuticals, Inc. pharmaceutical compositions
WO2019102265A1 (en) 2017-11-23 2019-05-31 Theraphage Inc. Peptide displaying bacteriophage nanoparticles and related compositions and methods
US20190275134A1 (en) 2018-03-12 2019-09-12 Janssen Pharmaceuticals, Inc Vaccines against urinary tract infections
CN112334141A (en) 2018-06-14 2021-02-05 株式会社明治 Compositions for facilitating immune checkpoint inhibition therapy
WO2021050696A1 (en) * 2019-09-10 2021-03-18 The Research Foundation For The State University Of New York Compositions and methods for improving tumor penetration of tumor specific antibodies
AU2020358862A1 (en) 2019-10-02 2022-04-14 Janssen Vaccines & Prevention B.V. Staphylococcus peptides and methods of use
BR112022013720A2 (en) 2020-01-16 2022-10-11 Janssen Pharmaceuticals Inc MUTANT FIMH, COMPOSITIONS WITH IT AND ITS USE
EP4277921A1 (en) 2021-01-12 2023-11-22 Janssen Pharmaceuticals, Inc. Fimh mutants, compositions therewith and use thereof
BR112023019874A2 (en) 2021-04-01 2023-11-07 Janssen Pharmaceuticals Inc PRODUCTION OF E. COLI O18 BIOCONJUGATES
WO2023168342A2 (en) * 2022-03-02 2023-09-07 Abreos Biosciences, Inc. Methods and compositions for measuring serum analyte levels from biological matrices

Family Cites Families (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4707358A (en) 1984-01-30 1987-11-17 The University Of Chicago Vaccine against Epstein-Barr Virus
US5030621A (en) 1987-04-23 1991-07-09 Bystryn Jean Claude Shed melanoma antigen compositions
DE318216T1 (en) 1987-11-18 1990-06-13 Chiron Corp., Emeryville, Calif., Us NANBV DIAGNOSTICS AND VACCINE.
ATE130764T1 (en) 1988-10-17 1995-12-15 Cancer Res Campaign Tech ANTI-IDIOTYPE ANTIBODIES INDUCTION OF AN ANTITUMOR RESPONSE.
JPH06104789B2 (en) 1989-03-31 1994-12-21 日本ペイント株式会社 Metal pigment composition and aqueous coating composition containing the same
DE4123760C2 (en) 1991-07-18 2000-01-20 Dade Behring Marburg Gmbh Seroreactive areas on the HPV 16 proteins E1 and E2
WO1993004672A1 (en) 1991-09-11 1993-03-18 Pitman-Moore, Inc. Method for enhancing the immune system in a host employing liposome-encapsulated polypeptides
US5484911A (en) 1993-04-01 1996-01-16 Health Research, Inc. Nucleoside 5'-diphosphate conjugates of ether lipids
US5961970A (en) 1993-10-29 1999-10-05 Pharmos Corporation Submicron emulsions as vaccine adjuvants
AU5543294A (en) 1993-10-29 1995-05-22 Pharmos Corp. Submicron emulsions as vaccine adjuvants
DE4499552C1 (en) 1993-12-09 1997-07-17 Heinrich Dr Exner New incomplete and complete adjuvants for antigens
GB9326253D0 (en) 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
US5798100A (en) 1994-07-06 1998-08-25 Immunomedics, Inc. Multi-stage cascade boosting vaccine
US5840306A (en) 1995-03-22 1998-11-24 Merck & Co., Inc. DNA encoding human papillomavirus type 18
DE19511276C2 (en) 1995-03-27 1999-02-18 Immuno Ag Adjuvant based on colloidal iron compounds
US6180111B1 (en) 1995-05-18 2001-01-30 University Of Maryland Vaccine delivery system
US5681824A (en) 1995-06-05 1997-10-28 Eisai Co., Ltd. Substituted liposaccharides useful in the treatment and prevention of endotoxemia
EP0840744A4 (en) 1995-06-07 1999-03-10 Genta Inc Phosphonic acid-based cationic lipids
TW343975B (en) 1995-12-15 1998-11-01 Boehringer Mannheim Gmbh New phospholipid derivatives of phosphonocarboxylic acids, the production thereof as well as their use as antiviral pharmaceutical agents
US6942862B2 (en) 1996-04-01 2005-09-13 University Of Washington Methods and compositions to generate immunity in humans against self tumor antigens by immunization with homologous foreign proteins
US5834015A (en) 1996-09-11 1998-11-10 Albany Medical College Protein-lipid vesicles and autogenous vaccine comprising the same
US6355257B1 (en) 1997-05-08 2002-03-12 Corixa Corporation Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
GB9712347D0 (en) 1997-06-14 1997-08-13 Smithkline Beecham Biolog Vaccine
BR9910269A (en) 1998-05-07 2001-01-09 Corixa Corp Adjuvant composition and methods for its use
US6146659A (en) 1998-07-01 2000-11-14 Neopharm, Inc. Method of administering liposomal encapsulated taxane
US6306404B1 (en) 1998-07-14 2001-10-23 American Cyanamid Company Adjuvant and vaccine compositions containing monophosphoryl lipid A
NZ512722A (en) * 1999-01-13 2003-07-25 Igeneon Krebs Immuntherapie Use of Ep-CAM antibodies for anticancer vaccination
US6551600B2 (en) 1999-02-01 2003-04-22 Eisai Co., Ltd. Immunological adjuvant compounds compositions and methods of use thereof
US20040006242A1 (en) 1999-02-01 2004-01-08 Hawkins Lynn D. Immunomodulatory compounds and method of use thereof
US7915238B2 (en) 1999-02-01 2011-03-29 Eisai R & D Management Co., Ltd. Immunomodulatory compounds and methods of use thereof
US6835721B2 (en) 1999-02-01 2004-12-28 Eisai Co., Ltd. Immunomodulatory compounds and methods of use thereof
DE60011571T2 (en) 1999-02-01 2005-08-18 Eisai Co., Ltd. COMPOUNDS WITH IMMUNOLOGICAL ADJUVANT EFFECT
WO2000073263A1 (en) 1999-05-28 2000-12-07 Vical Incorporated Cytofectin dimers and methods of use thereof
US7189397B2 (en) * 1999-10-08 2007-03-13 Arius Research Inc. Cytotoxicity mediation of cells evidencing surface expression of CD44
WO2001046127A1 (en) 1999-12-22 2001-06-28 Om Pharma Acyl pseudopeptides bearing a functionalised auxiliary spacer
AU5345901A (en) * 2000-04-13 2001-10-30 Univ Rockefeller Enhancement of antibody-mediated immune responses
CN1606446A (en) 2000-05-19 2005-04-13 科里克萨有限公司 Method for preventing and treating communicable diseases and other diseases using monosaccharides and disaccharides
EP1307466B1 (en) 2000-07-31 2005-11-16 Eisai Co., Ltd. Immunological adjuvant compounds
JP2004507552A (en) 2000-08-31 2004-03-11 メルク エンド カムパニー インコーポレーテッド Phosphate derivatives as immunomodulators
US7507724B2 (en) * 2001-01-16 2009-03-24 Sloan-Kettering Institute For Cancer Research Therapy-enhancing glucan
JP2004534088A (en) * 2001-07-06 2004-11-11 スローン−ケッタリング・インスティテュート・フォア・キャンサー・リサーチ Multivalent conjugate vaccine for cancer
AU2003224989B2 (en) * 2002-04-19 2008-12-04 Endocyte, Inc. Adjuvant enhanced immunotherapy
US20040197312A1 (en) * 2003-04-02 2004-10-07 Marina Moskalenko Cytokine-expressing cellular vaccine combinations
CA2536654A1 (en) * 2003-08-26 2005-03-03 Board Of Regents, The University Of Texas System Anti-cancer vaccines
FR2863890B1 (en) * 2003-12-19 2006-03-24 Aventis Pasteur IMMUNOSTIMULATING COMPOSITION
US20070292418A1 (en) * 2005-04-26 2007-12-20 Eisai Co., Ltd. Compositions and methods for immunotherapy
CN101355928B (en) 2005-04-26 2013-05-22 卫材R&D管理株式会社 Compositions and methods for cancer immunotherapy
JP6136343B2 (en) 2012-06-12 2017-05-31 株式会社リコー Information processing system, information processing method, program, and recording medium

Also Published As

Publication number Publication date
EP1874342B1 (en) 2018-06-06
KR101205064B1 (en) 2012-11-27
JP5539460B2 (en) 2014-07-02
EP1874342A2 (en) 2008-01-09
JP2012211196A (en) 2012-11-01
US20070020232A1 (en) 2007-01-25
US20140065100A1 (en) 2014-03-06
AU2006241206B2 (en) 2011-06-09
US20110250171A1 (en) 2011-10-13
US8603482B2 (en) 2013-12-10
AU2006241206A1 (en) 2006-11-02
CN103285392A (en) 2013-09-11
WO2006116423A2 (en) 2006-11-02
WO2006116423A3 (en) 2008-10-09
JP2008539249A (en) 2008-11-13
CN101355928A (en) 2009-01-28
CA2605749A1 (en) 2006-11-02
US7976852B2 (en) 2011-07-12
CN101355928B (en) 2013-05-22
EP1874342A4 (en) 2012-08-08
KR20070122510A (en) 2007-12-31
JP5185813B2 (en) 2013-04-17

Similar Documents

Publication Publication Date Title
CA2605749C (en) Compositions and methods for cancer immunotherapy
US20110243935A1 (en) Compositions and Methods for Immunotherapy
JP6713160B2 (en) Combination of WT1 antigen peptide and immunomodulator
JP2018021047A (en) Cancer immunotherapy
CA2617009C (en) Defective ribosomal products in blebs (dribbles) and methods of use to stimulate an immune response
JP2017171678A (en) Novel four ctl epitope-joined peptide
AU2014227019B2 (en) Novel peptide having 5 linked CTL epitopes
KR102375057B1 (en) Anti-cancer vaccine composition comprising HSP90 antigenic peptide
AU2011211457B2 (en) Compositions and methods for cancer immunotherapy

Legal Events

Date Code Title Description
EEER Examination request