Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Ann Oncol ; 23 Suppl 8: viii6-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22918931

ABSTRACT

The immune system has the greatest potential for the specific destruction of tumours with no toxicity to normal tissue and for long-term memory that can prevent cancer recurrence. The last 30 years of immuno-oncology research have provided solid evidence that tumours are recognised by the immune system and their development can be stopped or controlled long term through a process known as immunosurveillance. Tumour specificity of the immune response resides in the recognition of tumour antigens. Viral proteins in tumours caused by viruses and mutated proteins from oncogenes or other genes, as well as nonmutated but abnormally expressed self proteins found on all tumours, have been shown to be good antigens and good targets for immunosurveillance. In many cancers, however, malignant progression is accompanied by profound immune suppression that interferes with an effective antitumour response and tumour elimination. Initially, most of the escape from immunosurveillance was ascribed to changes in the tumour cells themselves (loss of tumour antigens, loss of human leukocyte antigen molecules, loss of sensitivity to complement, or T cell or natural killer (NK) cell lysis), making them a poor target of an immune attack. However, it has become clear that the suppression comes from the ability of tumours to subvert normal immune regulation to their advantage. The tumour microenvironment can prevent the expansion of tumour antigen-specific helper and cytotoxic T cells and instead promote the production of proinflammatory cytokines and other factors, leading to the accumulation of suppressive cell populations that inhibit instead of promote immunity. The best described are regulatory T cells and myeloid-derived suppressor cells. Great conceptual and technical advances in the field of immuno-oncology over the past 30 years have provided us with the knowledge and techniques to develop novel immunotherapeutic approaches for the treatment of cancer. These include methods that enhance tumour immunity by blocking inhibitory pathways and inhibitory cells in the tumour microenvironment (e.g. antibodies against cytotoxic T-lymphocyte-associated antigen-4, programmed death 1 or its ligand programmed death ligand 1, or low-dose chemotherapy). Of equal importance, they include methods that can enhance the specificity of antitumour immunity by inducing the expansion of T cells and antibodies directed to well-defined tumour antigens (e.g. cancer vaccines, potent adjuvants, immunostimulatory cytokines). Even as monotherapies, these approaches are having a substantial impact on the treatment of some patients with advanced, previously untreatable, malignancies. Most exciting of all, these successes provide a rationale to expect that used in various combinations or earlier in disease, current and future immunotherapies may transform cancer treatment, improving a prognosis for many patients.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Cytokines/biosynthesis , Humans , Immunosuppression Therapy , Monitoring, Immunologic , T-Lymphocytes, Cytotoxic/immunology
2.
Cell Death Differ ; 15(1): 70-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18007660

ABSTRACT

Cancer patients mount adaptive immune responses against their tumors. However, tumor develops many mechanisms to evade effective immunosurveillance. T-cell death caused by tumor plays a critical role in establishing tumor immunotolerance. Chronic stimulation of T cells by tumors leads to activation-induced cell death. Abortive stimulation of T cells by tolerogenic antigen-presenting cells loaded with tumor antigens leads to autonomous death of tumor-specific T cells. Therapeutic approaches that prevent T-cell death in the tumor microenvironment and tumor draining lymph nodes, therefore, should boost adaptive immune responses against cancer.


Subject(s)
Apoptosis , Immune Tolerance , Immunologic Surveillance , Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Tumor Escape , Antigens, Neoplasm/immunology , Autophagy , Dendritic Cells/immunology , Humans , Immunity, Cellular , Immunotherapy , Neoplasms/pathology , Neoplasms/physiopathology , Neoplasms/therapy , Suppressor Factors, Immunologic/physiology , T-Lymphocyte Subsets/physiology
3.
Novartis Found Symp ; 279: 206-12; discussion 213-5, 216-9, 2006.
Article in English | MEDLINE | ID: mdl-17278396

ABSTRACT

The immune system is alerted to the presence of a pathogen through the activation of the innate immune system. The message is transmitted to the cells of the adaptive immunity through activated antigen-presenting cells. The development of specific immunity capable of eliminating the pathogen is orchestrated by cytokines and chemokines produced by the innate system. When everything functions optimally, the pathogen is eradicated and specific memory response is established. This finely tuned system can be subverted by pathogens, leading to disease. Immunity to cancer is orchestrated in the same way and it is now recognized that the early stages of tumour development are recognized by the cells of innate immunity that transmit this message to the cells of adaptive immunity. The molecules that alert the immune system and are also its targets are tumour antigens. Two important antigens for lung tumour-specific immunity are MUC1 and cyclin B1. We discuss how each molecule interacts with the innate and the adaptive immunity and the types of the immune responses that result for these interactions. We also discuss the state of immunosuppression of adaptive immunity in cancer patients due to chronic activation of the innate immune system.


Subject(s)
Immunity, Innate/immunology , Lung Neoplasms/immunology , Animals , Humans
4.
J Exp Med ; 194(9): 1313-23, 2001 Nov 05.
Article in English | MEDLINE | ID: mdl-11696596

ABSTRACT

We eluted peptides from class I molecules of HLA-A2.1(+) breast adenocarcinoma and loaded reverse phase high-performance liquid chromatography (HPLC) fractions onto dendritic cells to prime naive CD8(+) T cells. Fractions that supported growth of tumor-specific cytotoxic T lymphocytes were analyzed by nano-HPLC micro-ESI tandem mass spectrometry. Six HLA-A2.1-binding peptides, four 9-mers (P1-P4) differing in the COOH-terminal residue, and two 10-mers (P5 and P6) with an additional COOH-terminal alanine, were identified in one fraction. Peptide sequences were homologous to cyclin B1. We primed CD8(+) T cells from another HLA-A2.1(+) healthy donor with synthetic peptides and generated P4-specific responses. We also detected memory T cells specific for one or more of these peptides in patients with breast cancer and squamous cell carcinomas of the head and neck (SCCHN). T cells from one patient, restimulated once in vitro, could kill the tumor cell line from which the peptides were derived. Immunohistochemical analysis of tumor lines and tissue sections showed cyclin B1 overexpression and aberrant localization in the cytoplasm instead of the nucleus. Sequencing genomic DNA and cDNA corresponding to P1-P6 region showed that differences in COOH-terminal residues were not due to either DNA mutations or errors in transcription, suggesting a high error rate in translation of cyclin B1 protein in tumors.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Cyclin B/immunology , Head and Neck Neoplasms/immunology , Antigens, Neoplasm/genetics , Base Sequence , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/cytology , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cyclin B/biosynthesis , Cyclin B/genetics , Cyclin B1 , DNA , DNA, Complementary , Female , Gene Expression , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Head and Neck Neoplasms/pathology , Health Status , Humans , Immunologic Memory , Molecular Sequence Data , Mutagenesis , Peptides/genetics , Peptides/immunology , RNA , Tissue Donors , Tumor Cells, Cultured
5.
Protein Expr Purif ; 22(1): 92-100, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11388805

ABSTRACT

MUC1, a member of the mucin family of molecules, is a transmembrane glycoprotein abundantly expressed on human ductal epithelial cells and tumors originating from those cells. MUC1 expressed by malignant cells is aberrantly O-glycosylated. Differences in O-glycosylation of the tandem repeat region of MUC1 make tumor and normal forms of this antigen immunologically distinct. The tumor-specific glycoform is, therefore, expected to be a good target for immunotherapy and a good immunogen for generation of antitumor immune responses. We have generated a renewable source of this glycoform by expressing MUC1 cDNA in Sf-9 insect cells using a baculovirus vector. This form of MUC1 (BV-MUC1) is O-glycosylated at a very low level, approximately 0.3% (w/w), and this is not due to the lack of appropriate glycosylotransferases in insect cells. Peptidyl GalNAc-transferases isolated from Sf-9 cells were able to glycosylate in vitro a synthetic MUC1 peptide as efficiently as the transferases isolated from human milk. Neither preparation of peptidyl GalNAc-transferases, however, was able to glycosylate BV-MUC1. This underglycosylated recombinant MUC1 mimics underglycosylated MUC1 on human tumor cells and could serve as an immunogen to stimulate responses that would recognize MUC1 on tumor cells. To test this we immunized mice with Sf-9 cells expressing BV-MUC1. Sera from immunized mice recognized MUC1 on human tumor cells. We also generated MUC1-specific T cells that proliferated in response to synthetic MUC1 peptide.


Subject(s)
Mucin-1/chemistry , Mucin-1/immunology , Spodoptera , Amino Acid Sequence , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Flow Cytometry , Glycosylation , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Molecular Sequence Data , Mucin-1/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spodoptera/cytology , Spodoptera/metabolism , T-Lymphocytes/immunology , Tandem Repeat Sequences
6.
Cancer Res ; 61(12): 4756-60, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406548

ABSTRACT

Impaired T-cell function in patients with advanced cancer has been a widely acknowledged finding, but mechanisms reported thus far are those primarily operating in the tumor microenvironment. Very few mechanisms have been put forth to explain several well-described defects in peripheral blood T cells, such as reduction in expression of signaling molecules, decreased production of cytokines, or increased apoptosis. We have closely examined the peripheral blood mononuclear cell (PBMC) samples derived from patients and healthy individuals, and we have observed an important difference that may underlie the majority of reported defects. We observed that in samples from patients only, an unusually large number of granulocytes copurify with low density PBMCs on a density gradient rather than sediment, as expected, to the bottom of the gradient. We also show that activating granulocytes from a healthy donor with N-formyl-L-methionyl-L-leucyl-L-phenylalanine could also cause them to sediment aberrantly and copurify with PBMCs, suggesting that density change is a marker of their activation. To confirm this, we looked for other evidence of in vivo granulocyte activation and found it in drastically elevated plasma levels of 8-isoprostane, a product of lipid peroxidation and a marker of oxidative stress. Reduced T-cell receptor zeta chain expression and decreased cytokine production by patients' T cells correlated with the presence of activated granulocytes in their PBMCs. We showed that freshly obtained granulocytes from healthy donors, if activated, can also inhibit cytokine production by T cells. This action is abrogated by the addition of the hydrogen peroxide (H(2)O(2)) scavenger, catalase, implicating H(2)O(2) as the effector molecule. Indeed, when added alone, H(2)O(2) could suppress cytokine production of normal T cells. These findings indicate that granulocytes are activated in advanced cancer patients and that granulocyte-derived H(2)O(2) is the major cause of severe systemic T-cell suppression.


Subject(s)
Granulocytes/immunology , Hydrogen Peroxide/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Adenocarcinoma/blood , Adenocarcinoma/immunology , Breast Neoplasms/blood , Breast Neoplasms/immunology , Colonic Neoplasms/blood , Colonic Neoplasms/immunology , Granulocytes/metabolism , Humans , Hydrogen Peroxide/blood , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Interleukin-2/biosynthesis , Interleukin-2/blood , Interleukin-4/biosynthesis , Interleukin-4/blood , Membrane Proteins/biosynthesis , Membrane Proteins/blood , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neoplasms/blood , Oxidative Stress/immunology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/blood , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Immunol ; 166(11): 6555-63, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11359807

ABSTRACT

Low-frequency CTL and low-titer IgM responses against tumor-associated Ag MUC1 are present in cancer patients but do not prevent cancer growth. Boosting MUC1-specific immunity with vaccines, especially effector mechanisms responsible for tumor rejection, is an important goal. We studied immunogenicity, tumor rejection potential, and safety of three vaccines: 1) MUC1 peptide admixed with murine GM-CSF as an adjuvant; 2) MUC1 peptide admixed with adjuvant SB-AS2; and 3) MUC1 peptide-pulsed dendritic cells (DC). We examined the qualitative and quantitative differences in humoral and T cell-mediated MUC1-specific immunity elicited in human MUC1-transgenic (Tg) mice compared with wild-type (WT) mice. Adjuvant-based vaccines induced MUC1-specific Abs but failed to stimulate MUC1-specific T cells. MUC1 peptide with GM-CSF induced IgG1 and IgG2b in WT mice but only IgM in MUC1-Tg mice. MUC1 peptide with SB-AS2 induced high-titer IgG1, IgG2b, and IgG3 Abs in both WT and MUC1-Tg mice. Induction of IgG responses was T cell independent and did not have any effect on tumor growth. MUC1 peptide-loaded DC induced only T cell immunity. If injected together with soluble peptide, the DC vaccine also triggered Ab production. Importantly, the DC vaccine elicited tumor rejection responses in both WT and MUC1-Tg mice. These responses correlated with the induction of MUC1-specific CD4+ and CD8+ T cells in WT mice, but only CD8(+) T cells in MUC1-Tg mice. Even though MUC1-specific CD4+ T cell tolerance was not broken, the capacity of MUC1-Tg mice to reject tumor was not compromised.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Graft Rejection/immunology , Lymphoma, T-Cell/immunology , Mucin-1/immunology , Peptide Fragments/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Dendritic Cells/immunology , Dendritic Cells/transplantation , Disease-Free Survival , Graft Rejection/genetics , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin Isotypes/biosynthesis , Interferon-gamma/physiology , Lymphocyte Activation/genetics , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mucin-1/administration & dosage , Mucin-1/genetics , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Repetitive Sequences, Amino Acid , Tumor Cells, Cultured
9.
Clin Cancer Res ; 7(3 Suppl): 773s-780s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300472

ABSTRACT

We describe a method for discovery of new tumor antigens that uses dendritic cells (DCs) as antigen-presenting cells to prime autologous naive CD4+ and CD8+ T cells from healthy donors against tumor proteins and peptides. For the identification of HLA class I-restricted tumor antigens, peptides were extracted from tumor HLA class I molecules, fractionated by reverse phase-high performance liquid chromatography, and loaded onto in vitro-generated DCs to prime naïve CD8+ T cells. Our results show that we were able to prime naive CD8+ T cells in vitro to several peptide fractions and generate specificity for the tumor. Electrospray ionization mass spectrometry was used to confirm that these fractions contained peptides derived from MHC class I molecules, and the primed CD8+ T cells were used to further analyze the immunostimulatory peptide fractions. For the identification of HLA class II-restricted tumor antigens, we fractionated tumor protein extracts using reverse phase-high performance liquid chromatography and loaded individual fractions onto DCs to prime naive CD4+ T cells. Our results show that we were also able to prime naive CD4+ T cells to several protein fractions and generate specificity for the tumor. These results illustrate the potential of this method to identify new immunostimulatory MHC class I- and class II-restricted tumor antigens.


Subject(s)
Antigens, Neoplasm/metabolism , Dendritic Cells/metabolism , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Division , Cells, Cultured , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Genes, MHC Class I , HLA Antigens/metabolism , Humans , Macrophages/metabolism , Mass Spectrometry , Mice , Peptides/chemistry , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Subcellular Fractions/metabolism , T-Lymphocytes/metabolism , Tumor Cells, Cultured
10.
Clin Cancer Res ; 7(3 Suppl): 781s-787s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300473

ABSTRACT

Transmembrane glycoprotein tumor antigen MUC1 that is overexpressed on pancreatic and breast tumor cells can be found in large amounts in soluble form in serum and ascites fluid. MUC1 has been identified as a target of human antitumor antibody and CTL responses that are generated in the absence of helper T cells. The soluble form of MUC1 should support generation of helper T cells, but we have found recently that this form, although effectively endocytosed by dendritic cells, remains trapped in early endosomes and is not trafficked to antigen-processing compartments. The exact biochemical structure of this form of MUC1 has not been elucidated to date, and it is thus not clear what structural characteristics may be responsible for its retention in early endosomes. We have purified soluble MUC1 from ascites fluid of breast/pancreatic cancer patients (ASC-MUC1) and quantitated O-linked carbohydrates. We have altered ASC-MUC1 by enzymatic treatment: trypsin or clostripain digestion, desialylation, and further in vitro glycosylation. We have found that desialylated ASC-MUC1 was further glycosylated by peptidyl N-acetylgalactosamine transferases and was not when sialic acid was present. These alterations created new forms of ASC-MUC1 that might be handled more efficiently by antigen-presenting cells to generate better tumor-specific immunity and used to identify structures that are directly involved in retention of this antigen in early endosomes.


Subject(s)
Ascites/metabolism , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Mucin-1/biosynthesis , Mucin-1/chemistry , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Carbohydrates/chemistry , Cysteine Endopeptidases/pharmacology , Electrophoresis, Polyacrylamide Gel , Endosomes/metabolism , Glycosylation , Humans , Immunoblotting , Microscopy, Electron , Models, Genetic , Monosaccharides/metabolism , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Protein Structure, Tertiary , Trypsin/pharmacology
11.
Clin Cancer Res ; 7(3 Suppl): 933s-939s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300494

ABSTRACT

Suppression of various functions of T cells derived from cancer patients has been linked previously to changes in the T-cell receptor (TCR)-associated signal transduction molecules, in particular the zeta chain of the TCR complex. In this study, we have examined the TCRzeta chain expression and cytokine production in vivo and in vitro in T cells of patients with metastatic adenocarcinomas of the pancreas that participated in a Phase I clinical trial of the MUC1 peptide plus bacillus Calmette-Guerin cancer vaccine. A majority of the patients had reduced TCRzeta chain expression and interleukin 4 production by T cells, and all of the patients showed decreased production of IFN-gamma of their peripheral T cells when compared with healthy individuals. Peripheral blood T cells were activated with the phorbol ester phorbol myrisate acetate and ionomycin to show that although aberrant TCRzeta chain expression and decreased cytokine production were often correlated, the reduced cytokine production was not simply a consequence of an impaired TCRzeta chain expression. Rather, these are two separate but parallel defects in signal transduction in T cells, which are potentially modulated by the same mechanisms. Half of the patients showed an improvement for TCRzeta chain or IFN-gamma expression after vaccination.


Subject(s)
Cytokines/biosynthesis , Membrane Proteins/biosynthesis , Pancreatic Neoplasms/metabolism , Receptors, Antigen, T-Cell/biosynthesis , Adenocarcinoma/metabolism , Biopsy , Flow Cytometry , Humans , Immunohistochemistry , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Mucin-1/chemistry , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Neoplasms/pathology , T-Lymphocytes/metabolism , Time Factors
12.
J Immunol ; 165(7): 3730-41, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11034378

ABSTRACT

Immunity to tumor Ags in patients is typically weak and not therapeutic. We have identified a new mechanism by which potentially immunogenic glycoprotein tumor Ags, such as MUC1, fail to stimulate strong immune responses. MUC1 is a heavily glycosylated membrane protein that is also present in soluble form in sera and ascites of cancer patients. We show that this soluble protein is readily taken up by dendritic cells (DC), but is not transported to late endosomes or MHC class II compartments for processing and binding to class II MHC. MUC1 uptake is mediated by the mannose receptor, and the protein is then retained long term in early endosomes without degradation. Long-term retention of MUC1 does not interfere with the ability of DC to process and present other Ags. We also demonstrate inhibited processing of another important glycoprotein tumor Ag, HER-2/neu. This may, therefore, be a frequent obstacle to presentation of tumor Ags and an important consideration in the design of cancer vaccines. It should be possible to overcome this obstacle by providing DC with a form of tumor Ag that can be better processed. For MUC1 we show that a 140-aa-long synthetic peptide is very efficiently processed by DC.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance , Intracellular Fluid/immunology , Lectins, C-Type , Mannose-Binding Lectins , Mucin-1/immunology , Mucin-1/metabolism , Amino Acid Sequence , Antibodies, Blocking/pharmacology , Antigen Presentation/drug effects , Biological Transport/drug effects , Biological Transport/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Dendritic Cells/drug effects , Dendritic Cells/ultrastructure , Endosomes/immunology , Endosomes/metabolism , Glycosylation , Hemocyanins/immunology , Hemocyanins/metabolism , Humans , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Mannans/pharmacology , Mannose/pharmacology , Mannose Receptor , Molecular Sequence Data , Mucin-1/blood , Mucin-1/ultrastructure , Protein Binding/drug effects , Protein Binding/immunology , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/immunology , Receptors, Cell Surface/physiology , Time Factors
15.
Clin Cancer Res ; 5(7): 1918-24, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10430099

ABSTRACT

CTLs and antibody responses to the tumor-associated antigen MUC1 mucin can be detected in patients with adenocarcinomas of the breast, pancreas, colon, and ovary. However, neither response is generally effective at controlling disease. Methods to augment immunity to MUC1 are being designed, with the expectation that this will lead to an antitumor response. The key to eliciting potent immunity to tumor MUC1 may be in generating MUC1-specific T-helper cell responses, which, to date, have not been reported in cancer patients. We have recently demonstrated that a synthetic vaccine representing five copies of the MUC1 tandem repeat peptide can be used to prime MUC1-specific human CD4+ T cells in vitro. Here, we extend these studies to test the immunogenicity and safety of the tandem repeat peptide in the chimpanzee, which has the identical MUC1 tandem repeat sequence to the human. To promote induction of Th1-type responses, we used the novel adjuvant LeIF, a Leishmania-derived protein that is known to stimulate human peripheral blood mononuclear cells (PBMCs) and antigen-presenting cells, to produce a Th1-type cytokine profile. We found that MUC1 tandem repeat peptide administered with LeIF elicited positive, albeit transient, proliferative T-cell responses to MUC1 in the PBMCs from four of four chimpanzees. Immunization induced MUC1-specific IFN-gamma but not interleukin 4 expression in CD4+ T cells from PBMCs and draining lymph nodes. MUC1-specific CTLs were also generated that did not induce detectable autoimmune dysfunction during the 1 year of observation. We conclude that the MUC1 tandem repeat peptide can be used to elicit both T-helper and cytotoxic cell responses to MUC1 in the primate and holds promise as a safe and effective cancer vaccine.


Subject(s)
Mucin-1/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Immunization , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/immunology , Lymph Nodes/immunology , Lymphocyte Activation , Pan troglodytes , Peptides/immunology , Repetitive Sequences, Amino Acid
16.
Cell Immunol ; 194(2): 143-9, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383817

ABSTRACT

We previously reported that the glycosylated MUC1 tumor antigen circulating as soluble protein in patients' serum is not processed by dendritic cells and does not elicit MHC-Class II-restricted T helper responses in vitro. In contrast, a long synthetic peptide from the MUC1 tandem repeat region is presented by Class II molecules, resulting in the initiation of T helper cell responses. Here we addressed the ability of dendritic cells to present various glycosylated or not glycosylated forms of MUC1 by MHC Class I. We found that three different forms of MUC1, ranging from glycosylated and underglycosylated protein to unglycosylated synthetic peptide, were able to elicit MUC1-specific, Class-I-restricted CTL responses. The efficiency of processing and the resulting strength of CTL activity were inversely correlated with the degree of glycosylation of the antigen. Furthermore, the more efficiently processed 100mer peptide primed a broader repertoire of CTL than the glycosylated protein.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Mucin-1/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antigen-Presenting Cells/immunology , Cell Line , Dendritic Cells/cytology , Epitopes, T-Lymphocyte/immunology , Female , Glycosylation , Histocompatibility Antigens Class II/immunology , Humans , Mice , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/immunology
17.
Cancer Res ; 58(22): 5066-70, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9823312

ABSTRACT

Epithelial cell mucin MUC1 is expressed on adenocarcinomas in an underglycosylated form that serves as a tumor antigen in breast, pancreatic, ovarian, and other tumors. Two predominant MUC1-specific immune responses are found in patients: CD8+ CTLs, which recognize tandemly repeated epitopes on the MUC1 protein core, and IgM antibodies. There have been no reports to date of MUC1-specific CD4+ T-helper cells in cancer patients. We show here that MUC1-specific CD4+ T cells are neither deleted nor tolerized and that CD4+ T cell responses can be generated when an appropriate soluble form of MUC1 is used. Naive CD4+ T cells from healthy donors were primed in vitro to a synthetic MUC1 peptide of 100 amino acids, representing five unglycosylated tandem repeats, presented by dendritic cells. They produced IFN-gamma and had moderate cytolytic activity. We identified one core peptide sequence, PGSTAPPAHGVT, that elicits this response when it is presented by HLA-DR3.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , Histocompatibility Antigens Class II/immunology , Mucin-1/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Herpesvirus 4, Human/immunology , Humans , Molecular Sequence Data , Mucin-1/chemistry , Peptide Fragments/chemistry , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tandem Repeat Sequences , Tumor Cells, Cultured
18.
J Immunother ; 21(4): 247-56, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9672846

ABSTRACT

MUC-1 mucin is an epithelial cell antigen whose aberrant expression plays a role in autoimmunity and tumor immunity and is thus an attractive candidate for immunotherapy of gene therapy. Because the MUC-1 cDNA is composed almost entirely of 60-bp tandem repeats and is susceptible to homologous recombination, it presents a special challenge to cloning and expression in viral vectors. Nevertheless, we have been successful in constructing a retroviral vector (MFG-MUC-1) with a 22-tandem repeat MUC-1 cDNA. Both stable and transient packaging cell lines are capable of producing high-titer retroviruses that can transfer the expression of MUC-1 to murine 3T3 cells. Transduced cells express uniformly high levels of MUC-1 on their surface, and western blot analysis reveals that the molecule expressed is of full length and extensively glycosylated. We have used the MFG-MUC-1 vector to stably transduce an immortalized murine dendritic cell line and show that immunization of mice with transduced cells elicits specific immune responses to mucin. The ability of this vector to transfer expression of the MUC-1 tumor antigen to potent antigen-presenting cells is expected to be of use in the immunotherapy of epithelial cancers.


Subject(s)
Immunotherapy , Mucin-1/immunology , 3T3 Cells , Animals , DNA, Complementary/chemistry , Genetic Therapy , Humans , Mice , Mice, Inbred BALB C , Mucin-1/genetics , Recombinant Proteins/immunology , Repetitive Sequences, Nucleic Acid , Retroviridae/genetics , T-Lymphocytes/immunology
19.
J Immunother ; 21(3): 188-97, 1998 May.
Article in English | MEDLINE | ID: mdl-9610910

ABSTRACT

We have previously reported that tumor-specific cytotoxic T lymphocytes (CTLs) derived from pancreatic and breast cancer patients recognize specific epitopes on the mucin polypeptide core. These CTLs recognize breast and pancreatic tumor cells in a major histocompatibility complex (MHC)-unrestricted fashion, and the lytic activity of these T cells is mediated through the T cell receptor (TCR). To characterize the TCR-mediated MHC-unrestricted CTL function, we used semiquantitative polymerase chain reaction (PCR) and cytofluorometry to analyze the TCR repertoire in CTL lines established from cancer patients and specific for mucin-expressing tumors. We found three TCR Vbeta genes, Vbeta9, Vbeta13.1. and Vbeta17, predominantly expressed in these functional cell lines, established either from one patient by stimulation with various mucin-expressing targets or from different patients. Sequencing of these preferentially used TCR genes unveiled usage of distinct Jbeta and Cbeta but a potentially interesting conservation of certain amino acids in the CDR3 region.


Subject(s)
Breast Neoplasms/immunology , Mucins/immunology , Pancreatic Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Base Sequence , Humans , Lymph Nodes/immunology , Polymorphism, Single-Stranded Conformational , Receptors, Antigen, T-Cell, alpha-beta/immunology , Sequence Analysis, DNA , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...