Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Front Oncol ; 12: 1023510, 2022.
Article in English | MEDLINE | ID: mdl-36419901

ABSTRACT

NK cells have unique attributes to react towards cells undergoing malignant transformation or viral infection. This reactivity is regulated by activating or inhibitory germline encoded receptors. An impaired NK cell function may result from an aberrant expression of such receptors, a condition often seen in patients with hematological cancers. Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer worldwide and NK cells have emerged as crucial targets for developing immunotherapies. However, there are important gaps concerning the phenotype and behavior of NK cells during emergence of ALL. In this study we analyze the phenotype and function of NK cells from peripheral blood in pediatric patients with ALL at diagnosis. Our results showed that NK cells exhibited an altered phenotype highlighted by a significant reduction in the overall expression and percent representation of activating receptors compared to age-matched controls. No significant differences were found for the expression of inhibitory receptors. Moreover, NK cells with a concurrent reduced expression in various activating receptors, was the dominant phenotype among patients. An alteration in the relative frequencies of NK cells expressing NKG2A and CD57 within the mature NK cell pool was also observed. In addition, NK cells from patients displayed a significant reduction in the ability to sustain antibody-dependent cellular cytotoxicity (ADCC). Finally, an aberrant expression of activating receptors is associated with the phenomenon of leukemia during childhood.

2.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32518090

ABSTRACT

BACKGROUND: Natural killer and cytotoxic CD8+ T cells are major players during antitumor immunity. They express NKG2D, an activating receptor that promotes tumor elimination through recognition of the MHC class I chain-related proteins A and B (MICA and MICB). Both molecules are overexpressed on a great variety of tumors from different tissues, making them attractive targets for immunotherapy. However, tumors shed MICA and MICB, and the soluble forms of both (sMICA and sMICB) mediate tumor-immune escape. Some reports indicate that anti-MICA antibodies (Ab) can promote the restoration of antitumor immunity through the induction of direct antitumor effects (antibody-dependent cell-mediated cytotoxicity, ADCC) and scavenging of sMICA. Therefore, we reasoned that an active induction of anti-MICA Ab with an immunogenic protein might represent a novel therapeutic and prophylactic alternative to restore antitumor immunity. METHODS: We generated a highly immunogenic chimeric protein (BLS-MICA) consisting of human MICA fused to the lumazine synthase from Brucella spp (BLS) and used it to generate anti-MICA polyclonal Ab (pAb) and to investigate if these anti-MICA Ab can reinstate antitumor immunity in mice using two different mouse tumors engineered to express MICA. We also explored the underlying mechanisms of this expected therapeutic effect. RESULTS: Immunization with BLS-MICA and administration of anti-MICA pAb elicited by BLS-MICA significantly delayed the growth of MICA-expressing mouse tumors but not of control tumors. The therapeutic effect of immunization with BLS-MICA included scavenging of sMICA and the anti-MICA Ab-mediated ADCC, promoting heightened intratumoral M1/proinflammatory macrophage and antigen-experienced CD8+ T cell recruitment. CONCLUSIONS: Immunization with the chimeric protein BLS-MICA constitutes a useful way to actively induce therapeutic anti-MICA pAb that resulted in a reprogramming of the antitumor immune response towards an antitumoral/proinflammatory phenotype. Hence, the BLS-MICA chimeric protein constitutes a novel antitumor vaccine of potential application in patients with MICA-expressing tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Histocompatibility Antigens Class I/immunology , Lymphoma/immunology , Recombinant Fusion Proteins/immunology , Urinary Bladder Neoplasms/immunology , Animals , Brucella/enzymology , Female , Histocompatibility Antigens Class I/genetics , Lymphoma/pathology , Lymphoma/therapy , Male , Mice , Mice, Inbred C57BL , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy
3.
Nat Rev Drug Discov ; 17(7): 509-527, 2018 07.
Article in English | MEDLINE | ID: mdl-29904196

ABSTRACT

Immune cell functions are regulated by co-inhibitory and co-stimulatory receptors. The first two generations of cancer immunotherapy agents consist primarily of antagonist antibodies that block negative immune checkpoints, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte protein 4 (CTLA4). Looking ahead, there is substantial promise in targeting co-stimulatory receptors with agonist antibodies, and a growing number of these agents are making their way through various stages of development. This Review discusses the key considerations and potential pitfalls of immune agonist antibody design and development, their differentiating features from antagonist antibodies and the landscape of agonist antibodies in clinical development for cancer treatment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Animals , CTLA-4 Antigen/antagonists & inhibitors , Humans , Immunotherapy/methods , Molecular Targeted Therapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors
4.
Sci Transl Med ; 10(424)2018 01 17.
Article in English | MEDLINE | ID: mdl-29343622

ABSTRACT

Antibodies targeting immune checkpoints are emerging as potent and viable cancer therapies, but not all patients respond to these as single agents. Concurrently targeting additional immunosuppressive pathways is a promising approach to enhance immune checkpoint blockade, and bifunctional molecules designed to target two pathways simultaneously may provide a strategic advantage over the combination of two single agents. M7824 (MSB0011359C) is a bifunctional fusion protein composed of a monoclonal antibody against programmed death ligand 1 (PD-L1) fused to the extracellular domain of human transforming growth factor-ß (TGF-ß) receptor II, which functions as a "trap" for all three TGF-ß isoforms. We demonstrate that M7824 efficiently, specifically, and simultaneously binds PD-L1 and TGF-ß. In syngeneic mouse models, M7824 suppressed tumor growth and metastasis more effectively than treatment with either an anti-PD-L1 antibody or TGF-ß trap alone; furthermore, M7824 extended survival and conferred long-term protective antitumor immunity. Mechanistically, the dual anti-immunosuppressive function of M7824 resulted in activation of both the innate and adaptive immune systems, which contributed to M7824's antitumor activity. Finally, M7824 was an effective combination partner for radiotherapy or chemotherapy in mouse models. Collectively, our preclinical data demonstrate that simultaneous blockade of the PD-L1 and TGF-ß pathways by M7824 elicits potent and superior antitumor activity relative to monotherapies.


Subject(s)
Antibodies, Monoclonal/immunology , Programmed Cell Death 1 Receptor/immunology , Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/chemistry , Immunotherapy/methods , Mice , Programmed Cell Death 1 Receptor/chemistry , Transforming Growth Factor beta/chemistry
5.
J Immunol ; 194(3): 878-82, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548231

ABSTRACT

CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.


Subject(s)
Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , CpG Islands , DNA Methylation , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Transgenic , Neoplasms/metabolism , Transforming Growth Factor beta/metabolism
6.
Cancer Epidemiol Biomarkers Prev ; 21(2): 337-46, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22144502

ABSTRACT

BACKGROUND: Macrophage inhibitory cytokine-1 (MIC-1/GDF15) mediates nonsteroidal anti-inflammatory drug (NSAID) protection from colonic polyps in mice and is linked to the development of colorectal carcinoma in humans. Therefore, changes in serum MIC-1/GDF15 levels could predict the presence of premalignant colonic polyposis and assist in population screening strategies. METHODS: Serum MIC-1/GDF15 levels were measured in subjects in the Polyp Prevention Trial, in which NSAID use and colon cancer risk factors were defined. Subjects had an initial adenoma removed, a repeat colonoscopy removing previously unidentified polyps, and serum MIC-1/GDF15 estimation. Three years later recurrent adenomas were identified and serum MIC-1/GDF15 levels reestimated. The relationship between serum MIC-1/GDF15 levels and adenoma presence or recurrence was examined. RESULTS: Serum MIC-1/GDF15 levels differed by adenoma status and were significantly related to colon cancer risk factors. In addition, mean serum MIC-1/GDF15 levels rose with increasing numbers of adenomas present and high-risk adenoma recurrence. NSAID users had higher serum MIC-1/GDF15 concentrations, which were related to protection from adenoma recurrence. Furthermore, adjusted serum MIC-1/GDF15 levels at final follow-up were related to adenoma recurrence (highest quartile MIC-1/GDF15; OR = 14.7, 95% CI: 3.0-73). CONCLUSIONS: These data suggest that MIC-1/GDF15 mediates at least some of the protection afforded by NSAIDs against human colonic polyposis. Furthermore, serum MIC-1/GDF15 levels vary with the development of adnenomatous colonic polyps. IMPACT: Serum MIC-1/GDF15 determination may hold promise as the first serum screening test to assist the detection of premalignant adenomatous colonic polyposis.


Subject(s)
Adenoma/prevention & control , Colonic Neoplasms/prevention & control , Growth Differentiation Factor 15/blood , Adenoma/blood , Adenoma/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers, Tumor/blood , Colonic Neoplasms/blood , Colonic Neoplasms/drug therapy , Colonic Polyps/blood , Colonic Polyps/prevention & control , Female , Humans , Male , Mass Screening , Mice , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Risk Factors
7.
Cancer Immunol Immunother ; 60(2): 197-206, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20976449

ABSTRACT

We have previously shown that the suppressive function of regulatory T cells (Tregs) from peripheral blood mononuclear cells (PBMCs) is enhanced in patients with prostate cancer when compared with healthy individuals. Two phase II studies using the PSA-TRICOM vaccine in patients with metastatic castration-resistant prostate cancer (mCRPC) showed evidence of patient benefit in terms of enhanced survival. The Halabi nomogram has been used to predict survival (HPS) of patients with mCRPC treated with conventional chemotherapy or second-line hormonal therapy. Tregs from PBMCs of patients (n = 23) with mCRPC were obtained pre- and post-three monthly vaccinations, and analyzed for number, phenotype, and suppressive function. Changes post- versus pre-vaccination in these parameters were compared with 3-year survival and HPS. No differences in Treg numbers were observed post- versus pre-vaccination. Trends (P = 0.029) were observed between overall survival (OS) and a decrease in Treg suppressive function post- versus pre-vaccination. Trends were also observed in analyzing effector:Treg (CD4(+)CD25(+)CD127(-)FoxP3(+)CTLA4(+)) ratio post- versus pre-vaccination with OS versus HPS. These data provide preliminary evidence for a possible association between improved OS and a decrease in Treg function when PBMCs are analyzed after three monthly vaccinations. Patients with an OS > HPS were more likely to have decreased Treg function following vaccine. Larger studies to confirm and extend these findings are warranted.


Subject(s)
Cancer Vaccines/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cancer Vaccines/administration & dosage , Flow Cytometry , Humans , Immune Tolerance/immunology , Lymphocyte Count , Male , Neoplasm Metastasis/immunology , Prostatic Neoplasms/immunology , Survival Analysis , T-Lymphocytes, Regulatory/metabolism
8.
J Immunother ; 33(7): 697-705, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664357

ABSTRACT

IL-12 is a potent antitumor cytokine that exhibits significant clinical toxicities after systemic administration. We hypothesized that intratumoral (i.t.) administration of IL-12 coformulated with the biodegradable polysaccharide chitosan could enhance the antitumor activity of IL-12 while limiting its systemic toxicity. Noninvasive imaging studies monitored local retention of IL-12, with and without chitosan coformulation, after i.t. injection. Antitumor efficacy of IL-12 alone and IL-12 coformulated with chitosan (chitosan/IL-12) was assessed in mice bearing established colorectal (MC32a) and pancreatic (Panc02) tumors. Additional studies involving depletion of immune cell subsets, tumor rechallenge, and CTL activity were designed to elucidate mechanisms of regression and tumor-specific immunity. Coformulation with chitosan increased local IL-12 retention from 1 to 2 days to 5 to 6 days. Weekly i.t. injections of IL-12 alone eradicated ≤10% of established MC32a and Panc02 tumors, while i.t. chitosan/IL-12 immunotherapy caused complete tumor regression in 80% to 100% of mice. Depletion of CD4(+) or Gr-1(+) cells had no impact on chitosan/IL-12-mediated tumor regression. However, CD8(+) or NK cell depletion completely abrogated antitumor activity. I.t. chitosan/IL-12 immunotherapy generated systemic tumor-specific immunity, as >80% of mice cured with i.t. chitosan/IL-12 immunotherapy were at least partially protected from tumor rechallenge. Furthermore, CTLs from spleens of cured mice lysed MC32a and gp70 peptide-loaded targets. Chitosan/IL-12 immunotherapy increased local retention of IL-12 in the tumor microenvironment, eradicated established, aggressive murine tumors, and generated systemic tumor-specific protective immunity. Chitosan/IL-12 is a well-tolerated, effective immunotherapy with considerable potential for clinical translation.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Chitosan/administration & dosage , Colorectal Neoplasms/therapy , Immunotherapy , Pancreatic Neoplasms/therapy , Animals , Carcinoembryonic Antigen/genetics , Cells, Cultured , Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic/drug effects , Immunologic Memory , Interleukin-12/administration & dosage , Interleukin-12/adverse effects , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/immunology , Remission Induction , Tumor Burden/drug effects
9.
Cancer Epidemiol Biomarkers Prev ; 19(6): 1441-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501764

ABSTRACT

BACKGROUND: Serum adiponectin, leptin, C-peptide, and homocysteine are indicators for obesity, hyperinsulinemia, and chronic inflammation, which have all been associated with colorectal cancer. AIMS: To determine whether serum adiponectin, leptin, C-peptide, and homocysteine are associated with fat, fiber, fruit and vegetable, flavonol, or dry bean intake and colorectal adenoma recurrence. METHODS: Using logistic regression, we estimated odds ratios (OR) and 95% confidence intervals (95% CI) for adenoma recurrence in 627 participants from the control arm of the Polyp Prevention Trial, a 4-year trial that examined the effectiveness of a low-fat, high-fiber, high-fruit and vegetable diet on adenoma recurrence. RESULTS: Serum concentrations of C-peptide and homocysteine were inversely related to fiber, fruit and vegetable, and flavonol intake and positively related to percentage of calories from fat (all P(trend) < or = 0.01). High homocysteine concentrations were associated with any (4th versus 1st quartile: OR, 2.26; 95% CI, 1.30-3.94) and more than one adenoma recurrence (OR, 2.11; 95% CI, 1.01-4.40). Individuals in the highest, versus lowest, tertile of serum leptin concentration had a decreased risk of advanced adenoma recurrence (OR, 0.22; 95% CI, 0.06-0.79). CONCLUSION: Our results suggest that serum homocysteine may serve as an indicator of dietary exposure, including a low-fat and high-fiber, high-fruit and vegetable, and high-flavonol diet, as well as colorectal adenoma recurrence. IMPACT: Discovering biomarkers that are both modifiable and can predict cancer risk is critical. We identified serum homocysteine as a novel indicator that is modified by diet and predicts risk of adenoma recurrence.


Subject(s)
Adenoma/blood , Colorectal Neoplasms/blood , Neoplasm Recurrence, Local/blood , Adenoma/prevention & control , Adiponectin/blood , Adult , Aged , Aged, 80 and over , C-Peptide/blood , Colorectal Neoplasms/prevention & control , Diet , Female , Follow-Up Studies , Homocysteine/blood , Humans , Leptin/blood , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Recurrence , Risk Factors
10.
Cancer Res ; 69(15): 6192-9, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638573

ABSTRACT

Intravesical BCG has been used successfully to treat superficial bladder cancer for three decades. However, 20% to 30% of patients will fail initial BCG therapy and 30% to 50% of patients will develop recurrent tumors within 5 years. Alternative or complementary strategies for the management of superficial bladder cancer are needed. Interleukin-12 (IL-12) is a potent T(H)1 cytokine with robust antitumor activity and the ability to potentiate immunologic memory. Unfortunately, intravesical IL-12 did not show antitumor efficacy in a recent clinical study of patients with recurrent superficial bladder cancer. We hypothesized that coformulation of IL-12 with chitosan, a biocompatible, mucoadhesive polysaccharide, could improve intravesical IL-12 delivery and provide an effective and durable alternative for the treatment of superficial bladder cancer. In antitumor studies, 88% to 100% of mice bearing orthotopic bladder tumors were cured after four intravesical treatments with chitosan/IL-12. In contrast, only 38% to 60% of mice treated with IL-12 alone and 0% treated with BCG were cured. Antitumor responses following chitosan/IL-12 treatments were durable and provided complete protection from intravesical tumor rechallenge. Urinary cytokine analysis showed that chitosan/IL-12 induced multiple T(H)1 cytokines at levels significantly higher than either IL-12 alone or BCG. Immunohistochemistry revealed moderate to intense tumor infiltration by T cells and macrophages following chitosan/IL-12 treatments. Bladder submucosa from cured mice contained residual populations of immune cells that returned to baseline levels after several months. Intravesical chitosan/IL-12 is a well-tolerated, effective immunotherapy that deserves further consideration for testing in humans for the management of superficial bladder cancer.


Subject(s)
Carcinoma, Transitional Cell/therapy , Chitosan/administration & dosage , Interleukin-12/administration & dosage , Urinary Bladder Neoplasms/therapy , Administration, Intravesical , Animals , BCG Vaccine/administration & dosage , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/immunology , Cell Line, Tumor , Female , Immunohistochemistry , Interferon-gamma/blood , Interferon-gamma/urine , Interleukin-12/blood , Interleukin-12/urine , Luciferases/biosynthesis , Luciferases/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Transfection , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
11.
Clin Cancer Res ; 15(7): 2387-96, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276249

ABSTRACT

PURPOSE: IFN-alpha is a pleiotropic cytokine possessing immunomodulatory properties that may improve the efficacy of therapeutic cancer vaccines. The aim of this study was to evaluate the effectiveness and compatibility of combining recombinant IFN-alpha with poxvirus vaccines targeting the human carcinoembryonic antigen (CEA) in murine models of colorectal and pancreatic adenocarcinomas, where CEA is a self-antigen. EXPERIMENTAL DESIGN: The phenotypic and functional effects of IFN-alpha were evaluated in the draining inguinal lymph nodes of tumor-free mice. We studied the effect of the site of IFN-alpha administration (local versus distal) on antigen-specific immune responses to poxvirus vaccination. Mechanistic studies were conducted to assess the efficacy of IFN-alpha and CEA-directed poxvirus vaccines in tumor-bearing CEA transgenic mice. RESULTS: We identified a dose and schedule of IFN-alpha that induced a locoregional expansion of the draining inguinal lymph nodes and improved cellular cytotoxicity (natural killer and CD8(+)) and antigen presentation. Suppression of the vaccinia virus was avoided by administering IFN-alpha distal to the site of vaccination. The combination of IFN-alpha and vaccine inhibited tumor growth, improved survival, and elicited CEA-specific CTL responses in mice with CEA(+) adenocarcinomas. In mice with pancreatic tumors, IFN-alpha slowed tumor growth, induced CTL activity, and increased CD8(+) tumor-infiltrating lymphocytes. CONCLUSIONS: These data suggest that IFN-alpha can be used as a biological response modifier with antigen-directed poxvirus vaccines to yield significant therapeutic antitumor immune responses. This study provides the rationale and mechanistic insights to support a clinical trial of this immunotherapeutic strategy in patients with CEA-expressing carcinomas.


Subject(s)
Adenocarcinoma/drug therapy , Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Fowlpox virus/genetics , Interferon-alpha/therapeutic use , Vaccinia virus/genetics , Adenocarcinoma/immunology , Animals , Cancer Vaccines/genetics , Carcinoembryonic Antigen/genetics , Cell Line, Tumor , Combined Modality Therapy , DNA, Recombinant/analysis , Female , Histocompatibility Antigens Class I/metabolism , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
12.
J Cell Physiol ; 219(1): 132-42, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19097033

ABSTRACT

We examined the role of the extracellular signal regulated kinases (ERK) in 1,25-dihydroxyvitamin D (1,25(OH)(2)D(3))-induced gene expression in the differentiated Caco-2 cells. 1,25(OH)(2)D(3)-regulated expression of the 25-hydroxyvitamin D, 24-hydroxylase (CYP24) gene (both natural gene and promoter construct) was strongly modulated by altering ERK activity (i.e., reduced by MEK inhibitors and dominant negative (dn) ERK1 and ERK2, activated by epidermal growth factor) but ERK inhibition had no effect on 1,25(OH)(2)D(3)-regulated expression of the transient receptor potential cation channel, subfamily V, member 6 (TRPV6). ERK5-mediated phosphorylation of the transcription factor Ets-1 enhanced 1,25(OH)(2)D(3)-mediated CYP24 gene transcription in proliferating but not differentiated Caco-2 cells due to reduced levels of ERK5 and Ets-1 (total and phosphoprotein levels) in differentiated cells. MEK inhibition reduced 1,25(OH)(2)D(3)-induced 3X-VDRE promoter activity but had no impact on the association of vitamin D receptor (VDR) with chromatin suggesting a role for co-activator recruitment in ERK-modulation of vitamin D-regulated CYP24 gene activation. Chromatin immunoprecipitation assays revealed that the ERK1/2 target, mediator 1 (MED1), is recruited to the CYP24, but not the TRPV6, promoter following 1,25(OH)(2)D(3) treatment. MED1 phosphorylation was sensitive to activators and inhibitors of the ERK1/2 signaling and MED1 siRNA reduced 1,25(OH)(2)D(3)-regulated human CYP24 promoter activity. This suggests ERK1/2 signaling enhances 1,25(OH)(2)D(3) effects on the CYP24 promoter by MED1-mediated events. Our data show that there are both promoter-specific and cell stage-specific roles for the ERK signaling pathway on 1,25(OH)(2)D(3)-mediated gene induction in enterocyte-like Caco-2 cells.


Subject(s)
Calcitriol/metabolism , Gene Expression Regulation, Enzymologic , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Caco-2 Cells/physiology , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Enzyme Inhibitors/metabolism , Epidermal Growth Factor/metabolism , Humans , Imidazoles/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , RNA Interference , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Steroid Hydroxylases/antagonists & inhibitors , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Two-Hybrid System Techniques , Vitamin D3 24-Hydroxylase , Vitamins/metabolism
13.
Vaccine ; 26(42): 5407-15, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18706954

ABSTRACT

Regular moderate exercise has been proposed to enhance immune function, but its effects on immunity and their consequences have not been well studied. Mice without (AL) or with access (AL+EX) to voluntary running wheels were vaccinated with a model antigen (ovalbumin (OVA)) via intranasal or subcutaneous routes to target the mucosal and systemic immune compartments, respectively. EX enhanced OVA-specific CD4(+) T cell cytokine production and proliferation in all lymphoid organs examined without changes in cell distribution in any organ. These results suggest that coupling moderate exercise with vaccination may enhance vaccine efficacy for the prevention and/or therapy of numerous diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Motor Activity , Vaccination , Animals , Body Composition , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/biosynthesis , Cytokines/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Substrate Specificity
14.
Clin Cancer Res ; 14(13): 4316-25, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594015

ABSTRACT

PURPOSE: Saccharomyces cerevisiae, a nonpathogenic yeast, has been used previously as a vehicle to elicit immune responses to foreign antigens, and tumor-associated antigens, and has been shown to reduce tumor burden in mice. Studies were designed to determine if vaccination of human carcinoembryonic antigen (CEA)-transgenic (CEA-Tg) mice (where CEA is a self-antigen) with a recombinant S. cerevisiae construct expressing human CEA (yeast-CEA) elicits CEA-specific T-cell responses and antitumor activity. EXPERIMENTAL DESIGN: CEA-Tg mice were vaccinated with yeast-CEA, and CD4(+) and CD8(+) T-cell responses were assessed after one and multiple administrations or vaccinations at multiple sites per administration. Antitumor activity was determined by tumor growth and overall survival in both pulmonary metastasis and s.c. pancreatic tumor models. RESULTS: These studies demonstrate that recombinant yeast can break tolerance and that (a) yeast-CEA constructs elicit both CEA-specific CD4(+) and CD8(+) T-cell responses; (b) repeated yeast-CEA administration causes increased antigen-specific T-cell responses after each vaccination; (c) vaccination with yeast-CEA at multiple sites induces a greater T-cell response than the same dose given at a single site; and (d) tumor-bearing mice vaccinated with yeast-CEA show a reduction in tumor burden and increased overall survival compared to mock-treated or control yeast-vaccinated mice in both pulmonary metastasis and s.c. pancreatic tumor models. CONCLUSIONS: Vaccination with a heat-killed recombinant yeast expressing the tumor-associated antigen CEA induces CEA-specific immune responses, reduces tumor burden, and extends overall survival in CEA-Tg mice. These studies thus form the rationale for the incorporation of recombinant yeast-CEA and other recombinant yeast constructs in cancer immunotherapy protocols.


Subject(s)
Antigens, Neoplasm/chemistry , Antineoplastic Agents/pharmacology , Carcinoembryonic Antigen/chemistry , Gene Expression Regulation , Immunotherapy/methods , Saccharomyces cerevisiae/metabolism , Vaccines, DNA/chemistry , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Carcinoembryonic Antigen/metabolism , Cell Proliferation , Female , Humans , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Recombinant Proteins/chemistry , Vaccines, DNA/metabolism
15.
J Nutr ; 138(1): 115-22, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18156413

ABSTRACT

The prevalence of obesity, an established risk factor for several chronic diseases, including cancer, has risen dramatically over the past 4 decades. Dietary change and/or increased physical activity are the most commonly recommended lifestyle-based strategies for preventing or reversing obesity. One of several physiological systems that may be enhanced by dietary change and exercise is the immune system. In this study, we examined the effects of energy restriction (ER; 30% reduction relative to control energy intake) and/or exercise (EX; voluntary wheel running) on systemic and mucosal immune function. Female C57BL/6 mice were randomized into 4 treatment conditions: 1) controls consumed ad libitum (AL); 2) AL with access to running wheels (AL + EX); 3) 30% ER; and 4) 30% ER with access to running wheels (ER + EX). Both ER and EX reduced spleen weight and the number of splenic T and B lymphocytes (P < 0.05). ER enhanced natural killer (NK) cell function, but reduced concanavalin A (Con A)-induced T-cell proliferation (P < 0.05). In contrast, EX enhanced Con A-induced proliferation and cytokine production from Peyer's patch cells (P < 0.05). These data suggest that ER and EX enhance some, but not all, components of the immune system and are likely working via different biological mechanisms to regulate NK and T-cell function.


Subject(s)
Energy Intake/physiology , Immunity, Mucosal/immunology , Motor Activity/physiology , Animals , Body Composition , Female , Mice , Mice, Inbred C57BL , Organ Size , Peyer's Patches/cytology , Peyer's Patches/metabolism , Spleen/anatomy & histology , T-Lymphocytes
16.
Vaccine ; 25(52): 8673-86, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18037196

ABSTRACT

Sustained, local delivery of immunomodulatory cytokines is under investigation for its ability to enhance vaccine and anti-tumor responses both clinically and preclinically. This study evaluates the ability of chitosan, a biocompatible polysaccharide, to (1) control the dissemination of a cytokine, GM-CSF, and (2) enhance the immunoadjuvant properties of GM-CSF. While cytokines have previously been delivered in lipid-based adjuvants and other vehicles, these do not have the clinical safety profile or unique properties of chitosan. We found that chitosan solution maintained a measurable depot of recombinant GM-CSF (rGM-CSF) at a subcutaneous injection site for up to 9 days. In contrast, when delivered in a saline vehicle, rGM-CSF was undetectable in 12-24h. Furthermore, a single s.c. injection of 20 microg rGM-CSF in chitosan solution (chitosan/rGM-CSF(20 microg)) transiently expanded lymph nodes up to 4.6-fold and increased the number of MHC class II expressing cells and dendritic cells by 7.4-fold and 6.8-fold, respectively. These increases were significantly greater than those measured when rGM-CSF was administered in saline at the standard preclinical dose and schedule, i.e. 4 daily s.c. injections of 20 microg. Furthermore, lymph node cells from mice injected with chitosan/rGM-CSF(20 microg) induced greater allogeneic T cell proliferation, indicating enhanced antigen presenting capability, than lymph node cells from mice injected with rGM-CSF alone. Finally, in vaccination experiments, chitosan/rGM-CSF was superior to either chitosan or rGM-CSF alone in enhancing the induction of antigen-specific CD4(+) proliferation, peptide-specific CD8(+) pentamer staining and cytotoxic T cell lysis. Altogether, chitosan/rGM-CSF outperformed standard rGM-CSF administrations in dendritic cell recruitment, antigen presentation and vaccine enhancement. We conclude that chitosan solution is a promising delivery platform for the sustained, local delivery of rGM-CSF.


Subject(s)
Chitosan/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytotoxicity Tests, Immunologic , Delayed-Action Preparations/administration & dosage , Dendritic Cells/immunology , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Histocompatibility Antigens Class II/genetics , Injections, Subcutaneous , Lymph Nodes/immunology , Lymphocyte Subsets/immunology , Mice , T-Lymphocytes, Cytotoxic/immunology
17.
Front Biosci ; 12: 4997-5029, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17569626

ABSTRACT

A number of lifestyle factors that reduce cancer risk in the primary prevention setting may be potential new targets for use in combination with cancer vaccines. This review discusses the modulation of energy balance (physical activity, calorie restriction, and obesity prevention), and the supplementation with natural and synthetic analogs of vitamins A and E, as potential interventions for use in combination with cancer vaccines. Additionally, the pharmacologic manipulation of nutrient metabolism in the tumor microenvironment (e.g., arachidonic acid, arginine, tryptophan, and glucose metabolism) is discussed. This review includes a brief overview of the role of each agent in primary cancer prevention; outlines the effects of these agents on immune function, specifically adaptive and/or anti-tumor immune mechanisms, when known; and discusses the potential use of these interventions in combination with therapeutic cancer vaccines. Modulation of energy balance through exercise and strategies targeting nutrient metabolism in the tumor microenvironment represent the most promising interventions to partner with therapeutic cancer vaccines. Additionally, the use of vitamin E succinate and the retinoid X receptor-directed rexinoids in combination with cancer vaccines offer promise. In summary, a number of energy balance- and nutrition-related interventions are viable candidates for further study in combination with cancer vaccines.


Subject(s)
Cancer Vaccines , Diet , Exercise , Neoplasms/prevention & control , Combined Modality Therapy , Energy Intake/immunology , Humans , Immunity, Innate , Neoplasms/complications , Neoplasms/immunology , Obesity/complications , Obesity/immunology , Risk Factors
18.
Vaccine ; 25(11): 2085-94, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17258843

ABSTRACT

The development of safe, novel adjuvants is necessary to maximize the efficacy of new and/or available vaccines. Chitosan is a non-toxic, biocompatible, biodegradable, natural polysaccharide derived from the exoskeletons of crustaceans and insects. Chitosan's biodegradability, immunological activity and high viscosity make it an excellent candidate as a depot/adjuvant for parenteral vaccination. To this end, we explored chitosan solution as an adjuvant for subcutaneous vaccination of mice with a model protein antigen. We found that chitosan enhanced antigen-specific antibody titers over five-fold and antigen-specific splenic CD4+ proliferation over six-fold. Strong increases in antibody titers together with robust delayed-type hypersensitivity (DTH) responses revealed that chitosan induced both humoral and cell-mediated immune responses. When compared with traditional vaccine adjuvants, chitosan was equipotent to incomplete Freund's adjuvant (IFA) and superior to aluminum hydroxide. Mechanistic studies revealed that chitosan exhibited at least two characteristics that may allow it to function as an immune adjuvant. First, the viscous chitosan solution created an antigen depot. More specifically, less than 9% of a protein antigen, when delivered in saline, remained at the injection site after 8 h. However, more than 60% of a protein antigen delivered in chitosan remained at the injection site for 7 days. Second, chitosan induced a transient 67% cellular expansion in draining lymph nodes. The expansion peaked between 14 and 21 days after chitosan injection and diminished as the polysaccharide was degraded. These mechanistic studies, taken together with the enhancement of a vaccine response, demonstrate that chitosan is a promising and safe platform for parenteral vaccine delivery.


Subject(s)
Adjuvants, Immunologic , Antibodies/blood , Chitosan/immunology , Vaccination , Vaccines/immunology , Aluminum Hydroxide/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Chitosan/pharmacokinetics , Enzyme-Linked Immunosorbent Assay , Female , Freund's Adjuvant , Histocytochemistry , Hypersensitivity, Delayed , Injections, Subcutaneous , Lipids , Lymph Nodes/cytology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Models, Animal , Skin/pathology , Vaccines/administration & dosage , beta-Galactosidase/immunology
19.
J Cell Biochem ; 100(3): 617-28, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-16960876

ABSTRACT

We examined the effects of 1,25 dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) on the distribution and mobility of the vitamin D receptor (VDR) in the enterocyte-like Caco-2 cell. Confocal microscopy showed that a green fluorescent protein-vitamin D receptor (GFP-VDR) fusion protein is predominantly nuclear (58%) and it does not associate with the apical or basolateral membrane of proliferating or polarized, differentiated cells. In contrast to the previously studied cell types, neither endogenous VDR nor GFP-VDR levels accumulate in the nucleus following 1,25(OH)(2)D(3) treatment (100 nM, 30 min). However, in nuclear photobleaching experiments nuclear GFP-VDR import was significantly increased by 1,25(OH)(2)D(3) during both an early (0-5 min) and later (30-35 min) period (20% per 5 min). Compared to the natural ligand, nuclear import of GFP-VDR was 60% lower in cells treated with the 1,25(OH)(2)D(3) analog, 1-alpha-fluoro-16-ene-20-epi-23-ene-26,27-bishomo-25-hydroxyvitamin D(3) (Ro-26-9228, 5 min, 100 nM). Downstream events like ligand-induced association of VDR with chromatin at 1 h and the accumulation of CYP24 mRNA were significantly lower in Ro-26-9228 treated cells compared to 1,25(OH)(2)D(3) (60 and 95% lower, respectively). Collectively our data are consistent with a role for ligand-induced nuclear VDR import in receptor activation. In addition, ligand-dependent VDR nuclear import appears to be balanced by export, thus accounting for the lack of nuclear VDR accumulation even when VDR import is significantly elevated.


Subject(s)
Colon/metabolism , Receptors, Calcitriol/metabolism , Base Sequence , Caco-2 Cells , Calcitriol/pharmacology , Colon/cytology , Colon/drug effects , DNA Primers , Green Fluorescent Proteins/metabolism , Humans , Protein Transport , Transcription, Genetic
20.
J Natl Cancer Inst ; 97(13): 966-75, 2005 Jul 06.
Article in English | MEDLINE | ID: mdl-15998949

ABSTRACT

BACKGROUND: Inflammatory breast carcinoma (IBC) appears to be a clinicopathologic entity distinct from noninflammatory locally advanced breast cancer (LABC). We examined incidence and survival trends for IBC in Surveillance, Epidemiology, and End Results (SEER) Program data with a case definition designed to capture many of its unique clinical and pathologic characteristics. METHODS: We analyzed breast cancer cases diagnosed in the SEER 9 Registries (n = 180,224), between 1988 and 2000. Breast cancer cases were categorized using SEER's "Extent of Disease" codes in combination with International Classification of Diseases for Oncology morphology code 8530/3 and classified as IBC (n = 3648), LABC (n = 3636), and non-T4 breast cancer (n = 172,940). We compared changes in incidence rates over 3-year intervals by breast cancer subtype and race using SEER*Stat. Survival differences by breast cancer subtype and race were assessed using Kaplan-Meier curves and log-rank statistics. All statistical tests were two-sided. RESULTS: Between 1988 and 1990 and 1997 and 1999, IBC incidence rates (per 100,000 woman-years) increased from 2.0 to 2.5 (P < .001), whereas those for LABC declined (2.5 to 2.0, P = .0025), as did those for non-T4 breast cancer (108 to 101, P = .0084). IBC incidence rates were statistically significantly higher in black women (3.1) than in white women (2.2) during the study period (P < .001). Women diagnosed with IBC had statistically significantly poorer survival than women with either LABC or non-T4 breast cancer (log-rank test, P < .001). Median survival of women with IBC (2.9 years) was statistically significantly shorter than that of women with LABC (6.4 years; P < .0001) or non-T4 breast cancer (> 10 years, P < .0001). Black women with IBC or LABC had poorer survival than white women with IBC or LABC, respectively (log-rank test, P < .001). CONCLUSIONS: Throughout the 1990s, IBC incidence rose, and survival improved modestly. Substantial racial differences were noted in age at diagnosis, age-specific incidence rates, and survival outcomes.


Subject(s)
Black or African American/statistics & numerical data , Breast Neoplasms/epidemiology , Carcinoma/epidemiology , White People/statistics & numerical data , Adult , Age Distribution , Age Factors , Aged , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma/chemistry , Carcinoma/mortality , Carcinoma/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Incidence , Inflammation , Middle Aged , Receptors, Estrogen/analysis , Research Design , SEER Program , Survival Rate/trends , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...