Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Oncotarget ; 12(20): 2051-2067, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34611479

ABSTRACT

Multiple Myeloma (MM) patients suffer disease relapse due to the development of therapeutic resistance. Increasing evidence suggests that immunotherapeutic strategies can provide durable responses. Here we evaluate the possibility of adoptive cell transfer (ACT) by generating ex vivo T cells from peripheral blood mononuclear cells (PBMCs) isolated from MM patients by employing our previously devised protocols. We designed peptides from antigens (Ags) including cancer testis antigens (CTAs) that are over expressed in MM. We exposed PBMCs from different healthy donors (HDs) to single peptides. We observed reproducible Ag-specific cluster of differentiation 4+ (CD4+) and CD8+ T cell responses on exposure of PBMCs to different single peptide sequences. These peptide sequences were used to compile four different peptide cocktails. Naïve T cells from PBMCs from MM patients or HDs recognized the cognate Ag in all four peptide cocktails, leading to generation of multiclonal Ag-specific CD4+ and CD8+ effector and central memory T (TEM and TCM, respectively) cells which produced interferon-gamma (IFN-γ), granzyme B and perforin on secondary restimulation. Furthermore, this study demonstrated that immune cells from MM patients are capable of switching metabolic programs to induce effector and memory responses. Multiple peptides and cocktails were identified that induce IFN-γ+, T1-type, metabolically active T cells, thereby paving the way for feasibility testing of ACT in phase I clinical trials.

2.
Oncotarget ; 8(7): 10785-10808, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-27974697

ABSTRACT

Effective adoptive immunotherapy has proved elusive for many types of human cancer, often due to difficulties achieving robust expansion of natural tumor-specific T-cells from peripheral blood. We hypothesized that antigen-driven T-cell expansion might best be triggered in vitro by acute activation of innate immunity to mimic a life-threatening infection. Unfractionated peripheral blood mononuclear cells (PBMC) were subjected to a two-step culture, first synchronizing their exposure to exogenous antigens with aggressive surrogate activation of innate immunity, followed by γ-chain cytokine-modulated T-cell hyperexpansion. Step 1 exposure to GM-CSF plus paired Toll-like receptor agonists (resiquimod and LPS), stimulated abundant IL-12 and IL-23 secretion, as well as upregulated co-stimulatory molecules and CD11c expression within the myeloid (CD33+) subpopulation. Added synthetic long peptides (>20aa) derived from widely expressed oncoproteins (MUC1, HER2/neu and CMVpp65), were reliably presented to CD4+ T-cells and cross-presented to CD8+ T-cells. Both presentation and cross-presentation demonstrated proteasomal and Sec61 dependence that could bypass the endoplasmic reticulum. Step 2 exposure to exogenous IL-7 or IL-7+IL-2 produced selective and sustained expansion of both CD4+ and CD8+ peptide-specific T-cells with a predominant interferon-γ-producing T1-type, as well as the antigen-specific ability to lyse tumor targets. Other γ-chain cytokines and/or combinations were initially proliferogenic, but followed by a contractile phase not observed with IL-7 or IL-7+IL-2. Regulatory T-cells were minimally propagated under these culture conditions. This mechanistically rational culture sequence, effective even for unvaccinated donors, enables rapid preparation of T-cells recognizing tumor-associated antigens expressed by the majority of human cancers, including pancreatic cancers, breast cancers and glioblastomas.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-7/pharmacology , Mucin-1/immunology , Receptor, ErbB-2/immunology , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cross-Priming/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Imidazoles/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-7/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mucin-1/metabolism , Neoplasms/immunology , Neoplasms/therapy , Peptides/immunology , Peptides/pharmacology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
3.
Biomolecules ; 6(3)2016 06 29.
Article in English | MEDLINE | ID: mdl-27367740

ABSTRACT

Cancer vaccines have often failed to live up to their promise, although recent results with checkpoint inhibitors are reviving hopes that they will soon fulfill their promise. Although mutation-specific vaccines are under development, there is still high interest in an off-the-shelf vaccine to a ubiquitous antigen, such as MUC1, which is aberrantly expressed on most solid and many hematological tumors, including more than 90% of breast carcinomas. Clinical trials for MUC1 have shown variable success, likely because of immunological tolerance to a self-antigen and to poor immunogenicity of tandem repeat peptides. We hypothesized that MUC1 peptides could be optimized, relying on heteroclitic optimizations of potential anchor amino acids with and without tumor-specific glycosylation of the peptides. We have identified novel MUC1 class I peptides that bind to HLA-A*0201 molecules with significantly higher affinity and function than the native MUC1 peptides. These peptides elicited CTLs from normal donors, as well as breast cancer patients, which were highly effective in killing MUC1-expressing MCF-7 breast cancer cells. Each peptide elicited lytic responses in greater than 6/8 of normal individuals and 3/3 breast cancer patients. The CTLs generated against the glycosylated-anchor modified peptides cross reacted with the native MUC1 peptide, STAPPVHNV, suggesting these analog peptides may offer substantial improvement in the design of epitope-based vaccines.


Subject(s)
Cancer Vaccines , HLA-A2 Antigen , Mucin-1 , Peptides , T-Lymphocytes, Cytotoxic/immunology , Aged , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Female , Glycosylation , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immune Tolerance , Interferon-gamma , MCF-7 Cells , Middle Aged , Mucin-1/immunology , Mucin-1/metabolism , Peptides/immunology , Peptides/metabolism
4.
PLoS One ; 11(1): e0145920, 2016.
Article in English | MEDLINE | ID: mdl-26788922

ABSTRACT

It remains challenging to produce decisive vaccines against MUC1, a tumor-associated antigen widely expressed by pancreas, breast and other tumors. Employing clinically relevant mouse models, we ruled out such causes as irreversible T-cell tolerance, inadequate avidity, and failure of T-cells to recognize aberrantly glycosylated tumor MUC1. Instead, every tested MUC1 preparation, even non-glycosylated synthetic 9mer peptides, induced interferon gamma-producing CD4(+) and CD8(+) T-cells that recognized glycosylated variants including tumor-associated MUC1. Vaccination with synthetic peptides conferred protection as long as vaccination was repeated post tumor challenge. Failure to revaccinate post challenge was associated with down-regulated tumor MUC1 and MHC molecules. Surprisingly, direct admixture of MUC1-expressing tumor with MUC1-hyperimmune T-cells could not prevent tumor outgrowth or MUC1 immunoediting, whereas ex vivo activation of the hyperimmune T-cells prior to tumor admixture rendered them curative. Therefore, surrogate T-cell preactivation outside the tumor bed, either in culture or by repetitive vaccination, can overcome tumor escape.


Subject(s)
Cancer Vaccines/therapeutic use , Mucin-1/genetics , Mucin-1/immunology , Neoplasms, Experimental/prevention & control , Peptides/therapeutic use , Animals , Antigens/chemistry , Antigens/immunology , Antigens/therapeutic use , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Glycosylation , Humans , Mice , Mice, Transgenic , Mucin-1/metabolism , Neoplasms, Experimental/immunology , Peptides/chemistry , Peptides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape
5.
Immunol Invest ; 41(6-7): 680-710, 2012.
Article in English | MEDLINE | ID: mdl-23017141

ABSTRACT

The receptor tyrosine kinase inhibitor, sunitinib, is astonishingly effective in its capacity to reduce MDSCs in peripheral tissues such as blood (human) and spleen (mouse), restoring responsiveness of bystander T lymphocytes to TcR stimulation. Sunitinib blocks proliferation of undifferentiated MDSCs and decreases survival of more differentiated neutrophilic MDSC (n-MDSC) progeny. Ironically, sunitinib's profound effects are observed even in a total absence of detectable anti-tumor therapeutic response. This is best explained by the presence of disparate MDSC-conditioning stimuli within individual body compartments, allowing sensitivity and resistance to sunitinib to coexist within the same mouse or patient. The presence or absence of GM-CSF is likely the major determinant in each compartment, given that GM-CSF's capacity to preempt STAT3-dependent with dominant STAT5-dependent hematopoietic programming confers sunitinib resistance and redirects differentiation from the n-MDSC lineage to the more versatile monocytoid (m-MDSC) lineage. The clinical sunitinib experience underscores that strategies for MDSC and Treg depletions must be mindful of disparities among body compartments to avoid sanctuary effects. Ironically, m-MDSCs manifesting resistance to sunitinib also have the greatest potential to differentiate into tumoricidal accessory cells, by virtue of their capacity to respond to T cell-secreted IFN-γ or to TLR agonists with nitric oxide and peroxynitrate production.


Subject(s)
Indoles/therapeutic use , Myeloid Progenitor Cells/drug effects , Neoplasms/pathology , Neoplasms/therapy , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , T-Lymphocytes/drug effects , Tumor Escape , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immune Tolerance , Indoles/pharmacology , Mice , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Neoplasms/immunology , Neoplasms/metabolism , Organ Specificity , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , Sunitinib , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
Proc Natl Acad Sci U S A ; 109(1): 261-6, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22171012

ABSTRACT

The mucin MUC1 is typically aberrantly glycosylated by epithelial cancer cells manifested by truncated O-linked saccharides. The resultant glycopeptide epitopes can bind cell surface major histocompatibility complex (MHC) molecules and are susceptible to recognition by cytotoxic T lymphocytes (CTLs), whereas aberrantly glycosylated MUC1 protein on the tumor cell surface can be bound by antibodies to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). Efforts to elicit CTLs and IgG antibodies against cancer-expressed MUC1 have not been successful when nonglycosylated MUC1 sequences were used for vaccination, probably due to conformational dissimilarities. Immunizations with densely glycosylated MUC1 peptides have also been ineffective due to impaired susceptibility to antigen processing. Given the challenges to immuno-target tumor-associated MUC1, we have identified the minimum requirements to consistently induce CTLs and ADCC-mediating antibodies specific for the tumor form of MUC1 resulting in a therapeutic response in a mouse model of mammary cancer. The vaccine is composed of the immunoadjuvant Pam(3)CysSK(4), a peptide T(helper) epitope and an aberrantly glycosylated MUC1 peptide. Covalent linkage of the three components was essential for maximum efficacy. The vaccine produced CTLs, which recognized both glycosylated and nonglycosylated peptides, whereas a similar nonglycosylated vaccine gave CTLs which recognized only nonglycosylated peptide. Antibodies elicited by the glycosylated tripartite vaccine were significantly more lytic compared with the unglycosylated control. As a result, immunization with the glycosylated tripartite vaccine was superior in tumor prevention. Besides its own aptness as a clinical target, these studies of MUC1 are likely predictive of a covalent linking strategy applicable to many additional tumor-associated antigens.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Mucin-1/immunology , Neoplasms/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/chemistry , Cancer Vaccines/chemistry , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Glycosylation , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Mammary Tumor Virus, Mouse/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/immunology , Vaccines, Synthetic/chemistry
7.
BMC Cancer ; 11: 365, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21859454

ABSTRACT

BACKGROUND: Several studies have demonstrated that sites of chronic inflammation are often associated with the establishment and growth of various malignancies. A common inflammatory condition in humans is autoimmune arthritis (AA). Although AA and cancer are different diseases, many of the underlying processes that contribute to the disorders of the joints and connective tissue that characterize AA also affect cancer progression and metastasis. Systemically, AA can lead to cellular infiltration and inflammation of the lungs. Several studies have reported statistically significant risk ratios between AA and breast cancer. Despite this knowledge being available, there has been minimal research linking breast cancer, arthritis, and metastasis associated with breast cancer. Notably both diseases are extremely prevalent in older post-menopausal women. METHODS: To establish the novel link between arthritis induced inflammation and secondary metastasis associated with breast cancer, PyV MT mice that spontaneously develop mammary gland carcinoma were injected with Type II collagen (CII) to induce arthritis at 9 and 18 weeks of age for pre-metastatic and metastatic condition. The sites of secondary metastasis and the associated inflammatory microenvironment were evaluated. RESULTS: A significant increase in breast cancer-associated secondary metastasis to the lungs and bones was observed in the arthritic versus the non-arthritic PyV MT mice along with an increase in primary tumor burden. We report significant increases in the levels of interstitial cellular infiltrates and pro-inflammatory cytokines such as interleukin-17 (IL-17), interleukin-6 (IL-6), Pro- Matrix metallopeptidase 9 (Pro-MMP9), insulin like growth factor-II (GF-II) and macrophage colony stimulating factor (M-CSF) in the arthritic lung and bone milieu as well as in the circulation. These pro-inflammatory cytokines along with the inflammatory microenvironment may be the underlying factors facilitating tumor progression and metastasis in arthritic PyV MT mice. This was further substantiated by treatment with celecoxib, an anti-inflammatory drug + αIL-17 antibody that significantly reduced the secondary metastasis to lung and bone. CONCLUSIONS: The data generated not only reveal the underlying mechanism of high susceptibility to bone and lung metastasis in an arthritic condition but our combination therapies may lead to treatment modalities that will be capable of reducing tumor burden, and preventing relapse and metastasis in arthritic patients with breast cancer.


Subject(s)
Arthritis, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Celecoxib , Collagen Type II , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Enzyme Precursors/metabolism , Female , Histocytochemistry , Inflammation/metabolism , Inflammation/pathology , Insulin-Like Growth Factor II/metabolism , Interleukin-17/antagonists & inhibitors , Interleukin-17/metabolism , Interleukin-6/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophage Colony-Stimulating Factor/metabolism , Mammary Neoplasms, Experimental/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Neoplasm Metastasis , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism
8.
Cancer Immunol Immunother ; 59(12): 1801-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20824430

ABSTRACT

Since epithelial mucin 1 (MUC1) is associated with several adenocarcinomas at the mucosal sites, it is pertinent to test the efficacy of a mucosally targeted vaccine formulation. The B subunit of the Vibrio cholerae cholera toxin (CTB) has great potential to act as a mucosal carrier for subunit vaccines. In the present study we evaluated whether a MUC1 tandem repeat (TR) peptide chemically linked to CTB would break self-antigen tolerance in the transgenic MUC1-tolerant mouse model (MUC1.Tg) through oral or parenteral immunizations. We report that oral immunization with the CTB-MUC1 conjugate along with mucosal adjuvant, unmethylated CpG oligodeoxynucleotide (ODN) and interleukin-12 (IL-12) did not break self-antigen tolerance in MUC1.Tg mice, but induced a strong humoral response in wild-type C57BL/6 mice. However, self-antigen tolerance in the MUC1.Tg mouse model was broken after parenteral immunizations with different doses of the CTB-MUC1 conjugate protein and with the adjuvant CpG ODN co-delivered with CTB-MUC1. Importantly, mice immunized systemically with CpG ODN alone and with CTB-MUC1 exhibited decreased tumor burden when challenged with a mammary gland tumor cell line that expresses human MUC1.


Subject(s)
Cholera Toxin/immunology , Mucin-1/immunology , Neoplasms, Experimental/therapy , Animals , Autoantigens/immunology , Cell Line, Tumor , Humans , Immune Tolerance , Immunization , Injections, Intraperitoneal , Interleukin-12/pharmacology , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology
9.
Breast Cancer Res ; 11(4): R56, 2009.
Article in English | MEDLINE | ID: mdl-19643025

ABSTRACT

INTRODUCTION: Sites of chronic inflammation are often associated with the establishment and growth of various malignancies including breast cancer. A common inflammatory condition in humans is autoimmune arthritis (AA) that causes inflammation and deformity of the joints. Other systemic effects associated with arthritis include increased cellular infiltration and inflammation of the lungs. Several studies have reported statistically significant risk ratios between AA and breast cancer. Despite this knowledge, available for a decade, it has never been questioned if the site of chronic inflammation linked to AA creates a milieu that attracts tumor cells to home and grow in the inflamed bones and lungs which are frequent sites of breast cancer metastasis. METHODS: To determine if chronic inflammation induced by autoimmune arthritis contributes to increased breast cancer-associated metastasis, we generated mammary gland tumors in SKG mice that were genetically prone to develop AA. Two breast cancer cell lines, one highly metastatic (4T1) and the other non-metastatic (TUBO) were used to generate the tumors in the mammary fat pad. Lung and bone metastasis and the associated inflammatory milieu were evaluated in the arthritic versus the non-arthritic mice. RESULTS: We report a three-fold increase in lung metastasis and a significant increase in the incidence of bone metastasis in the pro-arthritic and arthritic mice compared to non-arthritic control mice. We also report that the metastatic breast cancer cells augment the severity of arthritis resulting in a vicious cycle that increases both bone destruction and metastasis. Enhanced neutrophilic and granulocytic infiltration in lungs and bone of the pro-arthritic and arthritic mice and subsequent increase in circulating levels of proinflammatory cytokines, such as macrophage colony stimulating factor (M-CSF), interleukin-17 (IL-17), interleukin-6 (IL-6), vascular endothelial growth factor (VEGF), and tumor necrosis factor-alpha (TNF-alpha) may contribute to the increased metastasis. Treatment with anti-IL17 + celecoxib, an anti-inflammatory drug completely abrogated the development of metastasis and significantly reduced the primary tumor burden. CONCLUSIONS: The data clearly has important clinical implications for patients diagnosed with metastatic breast cancer, especially with regards to the prognosis and treatment options.


Subject(s)
Arthritis/complications , Autoimmune Diseases/complications , Bone Neoplasms/secondary , Cyclooxygenase 2/physiology , Interleukin-17/physiology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/therapeutic use , Arthritis/chemically induced , Arthritis/drug therapy , Arthritis/immunology , Arthritis/physiopathology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Bone Neoplasms/physiopathology , Bronchoalveolar Lavage Fluid/immunology , Celecoxib , Cell Line, Tumor/transplantation , Cyclooxygenase 2 Inhibitors/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Female , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Lung Neoplasms/physiopathology , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Organ Specificity , Osteoclasts/physiology , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , ZAP-70 Protein-Tyrosine Kinase/deficiency , ZAP-70 Protein-Tyrosine Kinase/genetics
10.
Breast Cancer Res ; 7(4): R422-35, 2005.
Article in English | MEDLINE | ID: mdl-15987447

ABSTRACT

INTRODUCTION: Inhibitors of cyclo-oxygenase (COX)-2 are being extensively studied as anticancer agents. In the present study we evaluated the mechanisms by which a highly selective COX-2 inhibitor, celecoxib, affects tumor growth of two differentially invasive human breast cancer cell lines. METHODS: MDA-MB-231 (highly invasive) and MDA-MB-468 (moderately invasive) cell lines were treated with varying concentrations of celecoxib in vitro, and the effects of this agent on cell growth and angiogenesis were monitored by evaluating cell proliferation, apoptosis, cell cycle arrest, and vasculogenic mimicry. The in vitro results of MDA-MB-231 cell line were further confirmed in vivo in a mouse xenograft model. RESULTS: The highly invasive MDA-MB-231 cells express higher levels of COX-2 than do the less invasive MDA-MB-468 cells. Celecoxib treatment inhibited COX-2 activity, indicated by prostaglandin E2 secretion, and caused significant growth arrest in both breast cancer cell lines. In the highly invasive MDA-MB-231 cells, the mechanism of celecoxib-induced growth arrest was by induction of apoptosis, associated with reduced activation of protein kinase B/Akt, and subsequent activation of caspases 3 and 7. In the less invasive MDA-MB-468 cells, growth arrest was a consequence of cell cycle arrest at the G0/G1 checkpoint. Celecoxib-induced growth inhibition was reversed by addition of exogenous prostaglandin E2 in MDA-MB-468 cells but not in MDA-MB-231 cells. Furthermore, MDA-MB-468 cells formed significantly fewer extracellular matrix associated microvascular channels in vitro than did the high COX-2 expressing MDA-MB-231 cells. Celecoxib treatment not only inhibited cell growth and vascular channel formation but also reduced vascular endothelial growth factor levels. The in vitro findings corroborated in vivo data from a mouse xenograft model in which daily administration of celecoxib significantly reduced tumor growth of MDA-MB-231 cells, which was associated with reduced vascularization and increased necrosis in the tumor mass. CONCLUSION: The disparate molecular mechanisms of celecoxib-induced growth inhibition in human breast cancer cells depends upon the level of COX-2 expression and the invasive potential of the cell lines examined. Data suggest a role for COX-2 not only in the growth of cancer cells but also in activating the angiogenic pathway through regulating levels of vascular endothelial growth factor.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Celecoxib , Cell Cycle/drug effects , Cyclooxygenase 2/biosynthesis , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/biosynthesis
11.
Mol Cancer Res ; 2(11): 632-42, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15561779

ABSTRACT

Cyclooxygenase-2 (COX-2) inhibitors are rapidly emerging as a new generation of therapeutic drug in combination with chemotherapy or radiation therapy for the treatment of cancer. The mechanisms underlying its antitumor effects are not fully understood and more thorough preclinical trials are needed to determine if COX-2 inhibition represents a useful approach for prevention and/or treatment of breast cancer. The purpose of this study was to evaluate the growth inhibitory mechanism of a highly selective COX-2 inhibitor, celecoxib, in an in vivo oncogenic mouse model of spontaneous breast cancer that resembles human disease. The oncogenic mice carry the polyoma middle T antigen driven by the mouse mammary tumor virus promoter and develop primary adenocarcinomas of the breast. Results show that oral administration of celecoxib caused significant reduction in mammary tumor burden associated with increased tumor cell apoptosis and decreased proliferation in vivo. In vivo apoptosis correlated with significant decrease in activation of protein kinase B/Akt, a cell survival signaling kinase, with increased expression of the proapoptotic protein Bax and decreased expression of the antiapoptotic protein Bcl-2. In addition, celecoxib treatment reduced levels of proangiogenic factor (vascular endothelial growth factor), suggesting a role of celecoxib in suppression of angiogenesis in this model. Results from these preclinical studies will form the basis for assessing the feasibility of celecoxib therapy alone or in combination with conventional therapies for treatment and/or prevention of breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/secondary , Cyclooxygenase Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Breast Neoplasms/metabolism , Celecoxib , Cell Division/drug effects , Dinoprostone/blood , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein
12.
Ann Surg Oncol ; 11(3): 328-39, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14993030

ABSTRACT

BACKGROUND: In several neoplastic diseases, including breast cancer, immunosuppression correlates with disease stage, progression, and outcome. Thus, thorough analysis of immune parameters in breast cancer patients may be beneficial in designing effective anticancer immune-based therapies. METHODS: We investigated dendritic cell and T-cell function in breast cancer patients at various stages of the disease and in age-matched controls. We also evaluated cyclooxygenase-2 (COX-2) expression and prostaglandin E2 (PGE2) levels within the tumor milieu and in the circulation. RESULTS: T cells from cancer patients showed decreased proliferation in response to CD3 antibody stimulation. Analysis of T-cell helper type 1 and 2 cytokines revealed reduced levels of interferon-gamma, tumor necrosis factor-alpha, interleukin (IL)-12, and IL-2 and increased levels of IL-10 and IL-4. Dendritic cells from these patients showed significantly reduced expression of co-stimulatory molecules (B7 and CD40) and demonstrated reduced phagocytic ability, reduced antigen presentation to T cells, and reduced ability to mature in response to lipopolysaccharide. Data revealed increased synthesis of PGE2, an immune suppressor, along with increased expression of COX-2, a key regulator of PGE2 synthesis. CONCLUSIONS: COX-2-induced PGE2 may contribute to immunosuppression and may directly block antitumor immunity while promoting tumor growth, providing us with the rationale for using COX-2 inhibition combined with immunotherapy.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Dendritic Cells/physiology , Dinoprostone/metabolism , Dinoprostone/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Immunosuppression Therapy , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , T-Lymphocytes/physiology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Case-Control Studies , Cell Division , Cyclooxygenase 2 , Disease Progression , Female , Humans , Isoenzymes/pharmacology , Membrane Proteins , Middle Aged , Phenotype , Prognosis , Prostaglandin-Endoperoxide Synthases/pharmacology , Tumor Cells, Cultured
13.
Breast Dis ; 20: 53-63, 2004.
Article in English | MEDLINE | ID: mdl-15687707

ABSTRACT

To study immunology in breast tumors, we have utilized a mammary gland adenocarcinoma model in which mice develop spontaneous tumors of the mammary gland which are initiated at puberty and express a human tumor antigen, MUC1. MUC1 (CD227) is over-expressed in 90% of human breast cancers and its glycosylation status and pattern of expression in cancer cells is altered. Humoral and cellular responses to MUC1 have been reported in breast cancer patients and therefore, MUC1 is being evaluated as a target for immune intervention. This mouse model of spontaneous breast cancer allows the evaluation of anti-MUC1 immune responses at all stages of the disease. In this report, we review the model as it pertains to a) the development of the tumor, b) MUC1 expression, and the native immune responses against MUC1 as tumors progress, and c) the immune suppressive microenvironment within the developing tumor. Finally, we report our latest findings describing the therapeutic efficacy of adoptively transferred MUC1-specific cytotoxic T lymphocytes (MUC1-CTL) in these mice and discuss ways to increase their effectiveness by agonistic monoclonal antibody against CD137 T cell costimulatory molecule.


Subject(s)
Breast Neoplasms/therapy , Mammary Neoplasms, Animal/therapy , Mucin-1/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD , Breast Neoplasms/immunology , Disease Models, Animal , Humans , Immunotherapy , Mammary Neoplasms, Animal/immunology , Mice , Tumor Necrosis Factor Receptor Superfamily, Member 9
14.
J Surg Res ; 107(2): 196-202, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12429175

ABSTRACT

BACKGROUND: Surgical resection of pancreatic tumors removes gross disease but not metastases. Adjuvant therapy such as chemotherapy and radiation treatment is of little value in metastatic pancreatic cancer. The hypothesis of this investigation is that specific and effective immunotherapeutic vaccine (dendritic/tumor cell fusion) will activate cytotoxic T lymphocytes (CTLs), leading to the eradication of spontaneous pancreatic cancer. METHODS: We have developed a double transgenic mouse model (MET) that forms spontaneous pancreatic tumors and expresses the human MUC1 antigen. Seven-week-old MET mice (n = 8) were treated every 3 weeks with the vaccine. In addition, these mice received 50 microg of superantigen staphylococcal enterotoxin B (SEB), a known T cell stimulant, prior to the first vaccination. A second treatment group received SEB alone (n = 8) and controls received no treatment (n = 9). MUC1-specific CTLs were measured by chromium release assay. At 10 weeks of age and at necropsy, MUC1 serum levels were measured using a MUC1-specific ELISA. RESULTS: Mice were known to harbor microscopic foci of cancer at birth. Survival was enhanced in vaccine as well as SEB-treated mice (75% CI +/- 0.42) compared to controls (11% CI +/- 0.28) and both groups of treated mice exhibited mature CTLs without in vitro stimulation. MUC1 serum levels of the vaccine group were 50% less than that of control (P < 0.04) at 10 weeks. MUC1 serum levels directly correlated with tumor weight at necropsy (r = 0.86). CONCLUSIONS: This is the first evidence that MUC1-specific CTLs can be stimulated to enhance survival in a spontaneous tumor model.


Subject(s)
Cancer Vaccines/therapeutic use , Cell Fusion , Dendritic Cells/immunology , Enterotoxins/pharmacology , Pancreatic Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Disease Models, Animal , Immunization , Immunotherapy , Mice , Mice, Inbred C57BL , Mucin-1/blood , Mucin-1/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...