Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Cancer ; 3(1): 60-74, 2022 01.
Article in English | MEDLINE | ID: mdl-35121988

ABSTRACT

Despite increased overall survival rates, curative options for metastatic breast cancer remain limited. We have previously shown that metadherin (MTDH) is frequently overexpressed in poor prognosis breast cancer, where it promotes metastasis and therapy resistance through its interaction with staphylococcal nuclease domain-containing 1 (SND1). Through genetic and pharmacological targeting of the MTDH-SND1 interaction, we reveal a key role for this complex in suppressing antitumor T cell responses in breast cancer. The MTDH-SND1 complex reduces tumor antigen presentation and inhibits T cell infiltration and activation by binding to and destabilizing Tap1/2 messenger RNAs, which encode key components of the antigen-presentation machinery. Following small-molecule compound C26-A6 treatment to disrupt the MTDH-SND1 complex, we showed enhanced immune surveillance and sensitivity to anti-programmed cell death protein 1 therapy in preclinical models of metastatic breast cancer, in support of this combination therapy as a viable approach to increase immune-checkpoint blockade therapy responses in metastatic breast cancer.


Subject(s)
Breast Neoplasms , Antigen Presentation , Breast Neoplasms/drug therapy , Endonucleases/metabolism , Female , Humans , Membrane Proteins/metabolism , Micrococcal Nuclease/metabolism , Nuclear Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
2.
Nat Cancer ; 3(1): 43-59, 2022 01.
Article in English | MEDLINE | ID: mdl-35121987

ABSTRACT

Metastatic breast cancer is a leading health burden worldwide. Previous studies have shown that metadherin (MTDH) promotes breast cancer initiation, metastasis and therapy resistance; however, the therapeutic potential of targeting MTDH remains largely unexplored. Here, we used genetically modified mice and demonstrate that genetic ablation of Mtdh inhibits breast cancer development through disrupting the interaction with staphylococcal nuclease domain-containing 1 (SND1), which is required to sustain breast cancer progression in established tumors. We performed a small-molecule compound screening to identify a class of specific inhibitors that disrupts the protein-protein interaction (PPI) between MTDH and SND1 and show that our lead candidate compounds C26-A2 and C26-A6 suppressed tumor growth and metastasis and enhanced chemotherapy sensitivity in preclinical models of triple-negative breast cancer (TNBC). Our results demonstrate a significant therapeutic potential in targeting the MTDH-SND1 complex and identify a new class of therapeutic agents for metastatic breast cancer.


Subject(s)
Endonucleases/metabolism , Membrane Proteins/metabolism , Micrococcal Nuclease , RNA-Binding Proteins/metabolism , Triple Negative Breast Neoplasms , Animals , Cell Adhesion Molecules/genetics , Humans , Membrane Proteins/genetics , Mice , RNA-Binding Proteins/genetics , Transcription Factors
3.
Nat Cell Biol ; 19(6): 711-723, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28530657

ABSTRACT

Tumour-initiating cells, or cancer stem cells (CSCs), possess stem-cell-like properties observed in normal adult tissue stem cells. Normal and cancerous stem cells may therefore share regulatory mechanisms for maintaining self-renewing capacity and resisting differentiation elicited by cell-intrinsic or microenvironmental cues. Here, we show that miR-199a promotes stem cell properties in mammary stem cells and breast CSCs by directly repressing nuclear receptor corepressor LCOR, which primes interferon (IFN) responses. Elevated miR-199a expression in stem-cell-enriched populations protects normal and malignant stem-like cells from differentiation and senescence induced by IFNs that are produced by epithelial and immune cells in the mammary gland. Importantly, the miR-199a-LCOR-IFN axis is activated in poorly differentiated ER- breast tumours, functionally promotes tumour initiation and metastasis, and is associated with poor clinical outcome. Our study therefore reveals a common mechanism shared by normal and malignant stem cells to protect them from suppressive immune cytokine signalling.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Interferons/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation , Cell Movement , Cell Self Renewal , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Phenotype , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transfection , Tumor Microenvironment
4.
Cancer Immunol Res ; 3(8): 946-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041735

ABSTRACT

DNA vaccines have demonstrated antitumor efficacy in multiple preclinical models, but low immunogenicity has been observed in several human clinical trials. This has led to many approaches seeking to improve the immunogenicity of DNA vaccines. We previously reported that a DNA vaccine encoding the cancer-testis antigen SSX2, modified to encode altered epitopes with increased MHC class I affinity, elicited a greater frequency of cytolytic, multifunctional CD8(+) T cells in non-tumor-bearing mice. We sought to test whether this optimized vaccine resulted in increased antitumor activity in mice bearing an HLA-A2-expressing tumor engineered to express SSX2. We found that immunization of tumor-bearing mice with the optimized vaccine elicited a surprisingly inferior antitumor effect relative to the native vaccine. Both native and optimized vaccines led to increased expression of PD-L1 on tumor cells, but antigen-specific CD8(+) T cells from mice immunized with the optimized construct expressed higher PD-1. Splenocytes from immunized animals induced PD-L1 expression on tumor cells in vitro. Antitumor activity of the optimized vaccine could be increased when combined with antibodies blocking PD-1 or PD-L1, or by targeting a tumor line not expressing PD-L1. These findings suggest that vaccines aimed at eliciting effector CD8(+) T cells, and DNA vaccines in particular, might best be combined with PD-1 pathway inhibitors in clinical trials. This strategy may be particularly advantageous for vaccines targeting prostate cancer, a disease for which antitumor vaccines have demonstrated clinical benefit and yet PD-1 pathway inhibitors alone have shown little efficacy to date.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Cancer Vaccines/immunology , Epitopes/immunology , Neoplasms/immunology , Peptide Fragments/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vaccines, DNA/immunology , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Epitopes/genetics , Gene Expression , Humans , Immunization , Mice , Mice, Knockout , Neoplasms/metabolism , Neoplasms/therapy , Peptide Fragments/genetics , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Vaccines, DNA/administration & dosage
5.
Cancer Cell ; 26(1): 92-105, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-24981741

ABSTRACT

The Metadherin gene (MTDH) is prevalently amplified in breast cancer and associated with poor prognosis; however, its functional contribution to tumorigenesis is poorly understood. Using mouse models representing different subtypes of breast cancer, we demonstrated that MTDH plays a critical role in mammary tumorigenesis by regulating oncogene-induced expansion and activities of tumor-initiating cells (TICs), whereas it is largely dispensable for normal development. Mechanistically, MTDH supports the survival of mammary epithelial cells under oncogenic/stress conditions by interacting with and stabilizing Staphylococcal nuclease domain-containing 1 (SND1). Silencing MTDH or SND1 individually or disrupting their interaction compromises tumorigenenic potential of TICs in vivo. This functional significance of MTDH-SND1 interaction is further supported by clinical analysis of human breast cancer samples.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Mammary Glands, Animal/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Breast Neoplasms/chemically induced , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/virology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Viral , Endonucleases , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mammary Glands, Animal/pathology , Mammary Glands, Animal/virology , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/pathogenicity , Medroxyprogesterone Acetate , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Phenotype , Protein Binding , RNA Interference , RNA-Binding Proteins , Time Factors , Transfection , Tumor Cells, Cultured
6.
Vaccine ; 32(15): 1707-15, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24492013

ABSTRACT

Plasmid DNA serves as a simple and easily modifiable form of antigen delivery for vaccines. The USDA approval of DNA vaccines for several non-human diseases underscores the potential of this type of antigen delivery method as a cost-effective approach for the treatment or prevention of human diseases, including cancer. However, while DNA vaccines have demonstrated safety and immunological effect in early phase clinical trials, they have not consistently elicited robust anti-tumor responses. Hence many recent efforts have sought to increase the immunological efficacy of DNA vaccines, and we have specifically evaluated several target antigens encoded by DNA vaccine as treatments for human prostate cancer. In particular, we have focused on SSX2 as one potential target antigen, given its frequent expression in metastatic prostate cancer. We have previously identified two peptides, p41-49 and p103-111, as HLA-A2-restricted SSX2-specific epitopes. In the present study we sought to determine whether the efficacy of a DNA vaccine could be enhanced by an altered peptide ligand (APL) strategy wherein modifications were made to anchor residues of these epitopes to enhance or ablate their binding to HLA-A2. A DNA vaccine encoding APL modified to increase epitope binding elicited robust peptide-specific CD8+ T cells producing Th1 cytokines specific for each epitope. Ablation of one epitope in a DNA vaccine did not enhance immune responses to the other epitope. These results demonstrate that APL encoded by a DNA vaccine can be used to elicit increased numbers of antigen-specific T cells specific for multiple epitopes simultaneously, and suggest this could be a general approach to improve the immunogenicity of DNA vaccines encoding tumor antigens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunodominant Epitopes/immunology , Neoplasm Proteins/immunology , Repressor Proteins/immunology , Vaccines, DNA/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/immunology , Prostatic Neoplasms/therapy
7.
Clin Cancer Res ; 19(17): 4547-9, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23922303

ABSTRACT

Tumor-derived factors can induce a premetastatic niche, yet little is known about how metastatic microenvironments are influenced by external insults, such as acute infections commonly seen in patients with cancer. New findings reveal increased metastasis to the lung after acute bacterial infection via the CXCR4/ubiquitin axis, suggesting new targets for antimetastasis therapeutics.


Subject(s)
Acute Lung Injury/genetics , Lung Neoplasms/genetics , Receptors, CXCR4/genetics , Ubiquitin/genetics , Animals , Humans
8.
J Mol Med (Berl) ; 91(4): 411-29, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23515621

ABSTRACT

Tumor metastasis is driven not only by the accumulation of intrinsic alterations in malignant cells, but also by the interactions of cancer cells with various stromal cell components of the tumor microenvironment. In particular, inflammation and infiltration of the tumor tissue by host immune cells, such as tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, have been shown to support tumor growth in addition to invasion and metastasis. Each step of tumor development, from initiation through metastatic spread, is promoted by communication between tumor and immune cells via the secretion of cytokines, growth factors, and proteases that remodel the tumor microenvironment. Invasion and metastasis require neovascularization, breakdown of the basement membrane, and remodeling of the extracellular matrix for tumor cell invasion and extravasation into the blood and lymphatic vessels. The subsequent dissemination of tumor cells to distant organ sites necessitates a treacherous journey through the vasculature, which is fostered by close association with platelets and macrophages. Additionally, the establishment of the pre-metastatic niche and specific metastasis organ tropism is fostered by neutrophils and bone marrow-derived hematopoietic immune progenitor cells and other inflammatory cytokines derived from tumor and immune cells, which alter the local environment of the tissue to promote adhesion of circulating tumor cells. This review focuses on the interactions between tumor cells and immune cells recruited to the tumor microenvironment and examines the factors allowing these cells to promote each stage of metastasis.


Subject(s)
Neoplasm Metastasis/immunology , Animals , Cytokines , Disease Progression , Humans , Inflammation/immunology , Neoplasm Invasiveness/immunology , Neoplasm Metastasis/therapy , Neoplastic Cells, Circulating , Neovascularization, Pathologic/immunology , Stromal Cells/immunology , Tumor Microenvironment/immunology
9.
Cancer Immunol Immunother ; 61(7): 1137-47, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22210552

ABSTRACT

CTLA-4 blockade has demonstrated antitumor efficacy in human clinical trials. The antitumor mechanism is presumably mediated in part by the expansion of tumor-specific T cells. Androgen deprivation, the cornerstone of treatment for patients with metastatic prostate cancer, has been shown to elicit prostate tissue apoptosis and lymphocytic inflammation. We hypothesized that treatment with androgen deprivation, followed by an anti-CTLA-4 antibody, could augment a tumor-specific immune response elicited by androgen deprivation. We report here the results of a phase I trial evaluating a humanized monoclonal antibody targeting CTLA-4, CP-675,206 (tremelimumab), in combination with androgen deprivation using an antiandrogen. Eligible patients were those with PSA-recurrent prostate cancer after primary surgery and/or radiation therapy, not previously treated with androgen deprivation, and without radiographic evidence of metastatic disease. Subjects were treated in two cycles, 3 months apart, in which they received bicalutamide 150 mg daily days 1-28 and tremelimumab on day 29. The primary endpoint of the trial was safety. Secondary endpoints included measures of PSA kinetics and identification of a maximum tolerated dose. Eleven patients were enrolled and completed at least 1 year of follow-up. Dose-limiting toxicities included grade 3 diarrhea and skin rash. No favorable changes in PSA doubling time were observed in a period shortly after completing treatment; however, three patients experienced a prolongation in PSA doubling time detectable several months after completing treatment. The identification of delayed, prolonged favorable changes in serum PSA suggests that future studies could explore this combination in studies evaluating time to disease progression.


Subject(s)
Androgen Antagonists/administration & dosage , Anilides/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Nitriles/administration & dosage , Prostate-Specific Antigen/blood , Prostatic Neoplasms/drug therapy , Tosyl Compounds/administration & dosage , Aged , Anilides/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease Progression , Dose-Response Relationship, Drug , Humans , Male , Middle Aged , Nitriles/adverse effects , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Tosyl Compounds/adverse effects , Treatment Outcome
10.
Prostate ; 72(5): 487-98, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21748755

ABSTRACT

BACKGROUND: We retrospectively explored changes in immunological parameters in men with biochemically recurrent prostate cancer treated with either 5 or 25 mg of lenalidomide in a randomized phase 2 trial, and determined whether those changes correlated with disease progression. METHODS: Cytokine levels were compared for each patient at baseline and after 6 months of treatment with lenalidomide. Regression models for correlated data were used to assess associations of cytokine levels with lenalidomide treatment effect. Estimates were obtained using generalized estimating equations. Changes in circulating anti-prostate antibodies were evaluated using a high-throughput immunoblot technique. RESULTS: Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Disease progression in treated patients was associated with an increase in circulating IL-8 levels, while IL-8 levels decreased significantly in non-progressors. CONCLUSIONS: Lenalidomide demonstrates immunomodulatory properties in patients with biochemically recurrent prostate cancer. The induction of novel anti-prostate antibodies is a potential mechanism for lenalidomide response. Changes in serum IL-8 levels may serve as a potential biomarker in treated patients. These hypotheses require formal testing in future prospective trials.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/therapeutic use , Interleukin-8/metabolism , Neoplasm Recurrence, Local/drug therapy , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/therapy , Thalidomide/analogs & derivatives , Adenocarcinoma/diagnosis , Adenocarcinoma/immunology , Aged , Aged, 80 and over , Antigens, Surface/immunology , Biomarkers, Tumor/immunology , Disease Progression , Humans , Immunomodulation/drug effects , Lenalidomide , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/immunology , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/immunology , Radiotherapy, Adjuvant , Retrospective Studies , Thalidomide/therapeutic use
11.
Cancer Res ; 71(21): 6785-95, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21880588

ABSTRACT

Recent U.S. Food and Drug Administration approval of the first immunotherapy for prostate cancer encourages efforts to improve immune targeting of this disease. The synovial sarcoma X chromosome breakpoint (SSX) proteins comprise a set of cancer-testis antigens that are upregulated in MHC class I-deficient germline cells and in various types of advanced cancers with a poor prognosis. Humoral and cell-mediated immune responses to the SSX family member SSX2 can arise spontaneously in prostate cancer patients. Thus, SSX2 and other proteins of the SSX family may offer useful targets for tumor immunotherapy. In this study, we evaluated the expression of SSX family members in prostate cancer cell lines and tumor biopsies to identify which members might be most appropriate for immune targeting. We found that SSX2 was expressed most frequently in prostate cell lines, but that SSX1 and SSX5 were also expressed after treatment with the DNA demethylating agent 5-aza-2'-deoxycytidine. Immunohistochemical analysis of microarrayed tissue biopsies confirmed a differential level of SSX protein expression in human prostate cancers. Notably, SSX expression in patient tumor samples was restricted to metastatic lesions (5/22; 23%) and no expression was detected in primary prostate tumors examined (0/73; P < 0.001). We determined that cross-reactive immune responses to a dominant HLA-A2-specific SSX epitope (p103-111) could be elicited by immunization of A2/DR1 transgenic mice with SSX vaccines. Our findings suggest that multiple SSX family members are expressed in metastatic prostate cancers which are amenable to simultaneous targeting.


Subject(s)
Adenocarcinoma/metabolism , Antigens, Neoplasm/physiology , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Repressor Proteins/physiology , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cancer Vaccines/immunology , Cell Line, Tumor/metabolism , Decitabine , Epitopes/immunology , Female , Gene Expression Regulation, Neoplastic/drug effects , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-DR1 Antigen/genetics , HLA-DR1 Antigen/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Multigene Family , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , T-Cell Antigen Receptor Specificity
12.
J Immunother ; 34(8): 569-80, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21904219

ABSTRACT

The cancer-testis antigen synovial sarcoma X breakpoint-2 (SSX-2) is a potentially attractive target for tumor immunotherapy based upon its tissue-restricted expression to germline cells and its frequent expression in malignancies. The goal of this study was to evaluate genetic vaccine encoding SSX-2 to prioritize human leukocyte antigen (HLA)-A2-specific epitopes and determine if a DNA vaccine can elicit SSX-2-specific cytotoxic T lymphocytes (CTLs) capable of lysing prostate cancer cells. HLA-A2-restricted epitopes were identified based on their in vitro binding affinity for HLA-A2 and by the ability of a genetic vaccine to elicit peptide-specific CTL in A2/DR1 (HLA-A2.1+/HLA-DR1+/H-2 class I-/class II-knockout) transgenic mice. We found that SSX-2 peptides p41-49 (KASEKIFYV) and p103-111 (RLQGISPKI) had high affinity for HLA-A2 and were immunogenic in vivo; however, peptide p103-111 was immunodominant with robust peptide-specific immune responses elicited in mice vaccinated with a plasmid DNA vaccine encoding SSX-2. Furthermore, p103-111-specific CTLs were able to lyse an HLA-A2+ prostate cancer cell line. The immunodominance of this epitope was found not to be due to a putative HLA-DR1 epitope (p98-112) flanking p103-111. Finally, we demonstrated that SSX-2 epitope-specific CTLs could be detected and cultured from the peripheral blood of HLA-A2+ prostate cancer patients, notably patients with advanced prostate cancer. Overall, we conclude that SSX-2 peptide p103-111 is an immunodominant HLA-A2-restricted epitope, and epitope-specific CD8 T cells can be detected in patients with prostate cancer, suggesting that tolerance to SSX-2 can be circumvented in vivo. Together, these findings suggest that SSX-2 may be a relevant target antigen for prostate cancer vaccine approaches.


Subject(s)
Cancer Vaccines/immunology , Epitopes/immunology , HLA-A2 Antigen/immunology , Neoplasm Proteins/immunology , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cells, Cultured , HLA-DR1 Antigen/immunology , Humans , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptides/immunology , Prostatic Neoplasms/immunology , Vaccines, DNA/immunology
13.
J Biomed Biotechnol ; 2011: 454861, 2011.
Article in English | MEDLINE | ID: mdl-21197272

ABSTRACT

We previously demonstrated that IgG responses to a panel of 126 prostate tissue-associated antigens are common in patients with prostate cancer. In the current report we questioned whether changes in IgG responses to this panel might be used as a measure of immune response, and potentially antigen spread, following prostate cancer-directed immune-active therapies. Sera were obtained from prostate cancer patients prior to and three months following treatment with androgen deprivation therapy (n = 34), a poxviral vaccine (n = 31), and a DNA vaccine (n = 21). Changes in IgG responses to individual antigens were identified by phage immunoblot. Patterns of IgG recognition following three months of treatment were evaluated using a machine-learned Bayesian Belief Network (ML-BBN). We found that different antigens were recognized following androgen deprivation compared with vaccine therapies. While the number of clinical responders was low in the vaccine-treated populations, we demonstrate that ML-BBN can be used to develop potentially predictive models.


Subject(s)
Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Immunoglobulin G/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Algorithms , Artificial Intelligence , Bayes Theorem , Biomarkers, Tumor/blood , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Computational Biology , Humans , Immunoblotting , Immunoglobulin G/blood , Male , Prostatic Neoplasms/metabolism , Signal Transduction , Treatment Outcome , Vaccines, DNA/immunology
14.
Clin Dev Immunol ; 2010: 150591, 2010.
Article in English | MEDLINE | ID: mdl-20981248

ABSTRACT

Cancer-testis antigens (CTAs) represent an expanding class of tumor-associated proteins defined on the basis of their tissue-restricted expression to testis or ovary germline cells and frequent ectopic expression in tumor tissue. The expression of CTA in MHC class I-deficient germline cells makes these proteins particularly attractive as immunotherapeutic targets because they serve as essentially tumor-specific antigens for MHC class I-restricted CD8+ T cells. Moreover, because CTAs are expressed in many types of cancer, any therapeutic developed to target these antigens might have efficacy for multiple cancer types. Of particular interest among CTAs is the synovial sarcoma X chromosome breakpoint (SSX) family of proteins, which includes ten highly homologous family members. Expression of SSX proteins in tumor tissues has been associated with advanced stages of disease and worse patient prognosis. Additionally, both humoral and cell-mediated immune responses to SSX proteins have been demonstrated in patients with tumors of varying histological origin, which indicates that natural immune responses can be spontaneously generated to these antigens in cancer patients. The current review will describe the history and identification of this family of proteins, as well as what is known of their function, expression in normal and malignant tissues, and immunogenicity.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasm Proteins/immunology , Neoplasms/therapy , Repressor Proteins/immunology , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cytotoxicity, Immunologic , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , Male , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/immunology , Prognosis , Repressor Proteins/metabolism
15.
J Hered ; 99(4): 407-16, 2008.
Article in English | MEDLINE | ID: mdl-18310068

ABSTRACT

The K homology (KH) domain is a conserved sequence present in a wide variety of RNA-binding proteins. The rough sheath2-interacting KH domain (RIK) protein of maize has been implicated in the maintenance of the repressed chromatin state of knox genes during leaf primordia initiation. The amino acid sequences of the publicly available plant RIK proteins contain a splicing factor 1 (SF1)-like KH domain core sequence motif that distinguishes them from all other SF1-like KH domain-containing proteins. We demonstrate that the maize RIK gene exhibits surprisingly little nucleotide sequence diversity among Zea species and subspecies. Microarray hybridization experiments demonstrate that RIK has a higher level of expression in the shoot apical meristem as compared with 14-day seedling. Reverse transcriptase-polymerase chain reaction analysis of RIK indicates that the gene is expressed in many tissues, albeit at lower levels in older leaf samples. Taken together, these data suggest that the RIK protein may be involved in the maintenance of an inactive chromatin state of knox and possibly other genes in nonmeristematic tissues.


Subject(s)
Genes, Plant , Plant Proteins/genetics , Polymorphism, Single Nucleotide , Zea mays/genetics , Amino Acid Sequence , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Plant , Genome, Plant , Meristem/genetics , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phylogeny , Plant Proteins/metabolism , Repressor Proteins/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL