Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(5): e0285707, 2023.
Article in English | MEDLINE | ID: mdl-37192201

ABSTRACT

Next generation sequencing of human cancer mutations has identified novel therapeutic targets. Activating Ras oncogene mutations play a central role in oncogenesis, and Ras-driven tumorigenesis upregulates an array of genes and signaling cascades that can transform normal cells into tumor cells. In this study, we investigated the role of altered localization of epithelial cell adhesion molecule (EpCAM) in Ras-expressing cells. Analysis of microarray data demonstrated that Ras expression induced EpCAM expression in normal breast epithelial cells. Fluorescent and confocal microscopy showed that H-Ras mediated transformation also promoted epithelial-to-mesenchymal transition (EMT) together with EpCAM. To consistently localize EpCAM in the cytosol, we generated a cancer-associated EpCAM mutant (EpCAM-L240A) that is retained in the cytosol compartment. Normal MCF-10A cells were transduced with H-Ras together with EpCAM wild-type (WT) or EpCAM-L240A. WT-EpCAM marginally effected invasion, proliferation, and soft agar growth. EpCAM-L240A, however, markedly altered cells and transformed to mesenchymal phenotype. Ras-EpCAM-L240A expression also promoted expression of EMT factors FRA1, ZEB1 with inflammatory cytokines IL-6, IL-8, and IL1. This altered morphology was reversed using MEK-specific inhibitors and to some extent JNK inhibition. Furthermore, these transformed cells were sensitized to apoptosis using paclitaxel and quercetin, but not other therapies. For the first time, we have demonstrated that EpCAM mutations can cooperate with H-Ras and promote EMT. Collectively, our results highlight future therapeutic opportunities in EpCAM and Ras mutated cancers.


Subject(s)
Epithelial-Mesenchymal Transition , Signal Transduction , Humans , Cell Line, Tumor , Cytosol/metabolism , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Epithelial-Mesenchymal Transition/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
2.
Onco Targets Ther ; 14: 3813-3820, 2021.
Article in English | MEDLINE | ID: mdl-34188489

ABSTRACT

INTRODUCTION: Esophageal cancer (EC) is an aggressive cancer type that is increasing at a high rate in the US and worldwide. Extensive sequencing of EC specimens has shown that there are no consistent driver mutations that can impact treatment strategies. The goal of this study was to identify activated tyrosine kinase receptors (TKRs) in EC samples as potential targets in the treatment of EC. METHODS: Activated tyrosine kinase receptors were detected using a dot-blot array for human TK receptors. Human esophageal cancer cell lines were transplanted into immunocompromised mice, and tumor xenografts were subjected to tyrosine kinase inhibitors based on the dot-blot array data. RESULTS: Using the OE33 esophageal cancer cell line, we identified activated EGF receptor (EGFR), as well as ErbB2 and ErbB3. Treatment of this cell line with erlotinib, a specific inhibitor of EGFR, did not impact the growth of this tumor cell line. Treating the OE33 cell line with afatinib, a pan-EGFR family inhibitor resulted in the growth inhibition of OE33, indicating that the ErbB2 and ErbB3 receptors were contributing to tumor cell proliferation. Afatinib treatment of mice growing OE33 tumors inhibited growth of the OE33 tumor cells. DISCUSSION: Activated tyrosine kinase receptors were readily detected in both cancer cell lines and human esophageal cancer samples. By identifying the activated receptors and then using the appropriate tyrosine kinase inhibitors, we can block tumor growth in vitro and in animal xenografts. We propose that identifying and targeting activated TKRs can be used as a personalized EC tumor treatment strategy.

3.
BMC Cancer ; 21(1): 541, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33980181

ABSTRACT

BACKGROUND: EpCAM (Epithelial cell adhesion molecule) is often dysregulated in epithelial cancers. Prior studies implicate EpCAM in the regulation of oncogenic signaling pathways and epithelial-to-mesenchymal transition. It was recently demonstrated that EpCAM contains a thyroglobulin type-1 (TY-1) domain. Multiple proteins with TY-1 domains are known to inhibit cathepsin-L (CTSL), a cysteine protease that promotes tumor cell invasion and metastasis. Analysis of human cancer sequencing studies reveals that somatic EpCAM mutations are present in up to 5.1% of tested tumors. METHODS: The Catalogue of Somatic Mutations in Cancer (COSMIC) database was queried to tabulate the position and amino acid changes of cancer associated EpCAM mutations. To determine how EpCAM mutations affect cancer biology we studied C66Y, a damaging TY-1 domain mutation identified in liver cancer, as well as 13 other cancer-associated EpCAM mutations. In vitro and in vivo models were used to determine the effect of wild type (WT) and mutant EpCAM on CTSL activity and invasion. Immunoprecipitation and localization studies tested EpCAM and CTSL protein binding and determined compartmental expression patterns of EpCAM mutants. RESULTS: We demonstrate that WT EpCAM, but not C66Y EpCAM, inhibits CTSL activity in vitro, and the TY-1 domain of EpCAM is responsible for this inhibition. WT EpCAM, but not C66Y EpCAM, inhibits tumor cell invasion in vitro and lung metastases in vivo. In an extended panel of human cancer cell lines, EpCAM expression is inversely correlated with CTSL activity. Previous studies have demonstrated that EpCAM germline mutations can prevent EpCAM from being expressed at the cell surface. We demonstrate that C66Y and multiple other EpCAM cancer-associated mutations prevent surface expression of EpCAM. Cancer-associated mutations that prevent EpCAM cell surface expression abrogate the ability of EpCAM to inhibit CTSL activity and tumor cell invasion. CONCLUSIONS: These studies reveal a novel role for EpCAM as a CTSL inhibitor, confirm the functional relevance of multiple cancer-associated EpCAM mutations, and suggest a therapeutic vulnerability in cancers harboring EpCAM mutations.


Subject(s)
Cathepsin L/antagonists & inhibitors , Epithelial Cell Adhesion Molecule/genetics , Mutation , Neoplasms/genetics , Animals , Cathepsin L/physiology , Epithelial Cell Adhesion Molecule/physiology , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness
4.
Genome Med ; 13(1): 56, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33879241

ABSTRACT

BACKGROUND: Preclinical studies and early clinical trials have shown that targeting cancer neoantigens is a promising approach towards the development of personalized cancer immunotherapies. DNA vaccines can be rapidly and efficiently manufactured and can integrate multiple neoantigens simultaneously. We therefore sought to optimize the design of polyepitope DNA vaccines and test optimized polyepitope neoantigen DNA vaccines in preclinical models and in clinical translation. METHODS: We developed and optimized a DNA vaccine platform to target multiple neoantigens. The polyepitope DNA vaccine platform was first optimized using model antigens in vitro and in vivo. We then identified neoantigens in preclinical breast cancer models through genome sequencing and in silico neoantigen prediction pipelines. Optimized polyepitope neoantigen DNA vaccines specific for the murine breast tumor E0771 and 4T1 were designed and their immunogenicity was tested in vivo. We also tested an optimized polyepitope neoantigen DNA vaccine in a patient with metastatic pancreatic neuroendocrine tumor. RESULTS: Our data support an optimized polyepitope neoantigen DNA vaccine design encoding long (≥20-mer) epitopes with a mutant form of ubiquitin (Ubmut) fused to the N-terminus for antigen processing and presentation. Optimized polyepitope neoantigen DNA vaccines were immunogenic and generated robust neoantigen-specific immune responses in mice. The magnitude of immune responses generated by optimized polyepitope neoantigen DNA vaccines was similar to that of synthetic long peptide vaccines specific for the same neoantigens. When combined with immune checkpoint blockade therapy, optimized polyepitope neoantigen DNA vaccines were capable of inducing antitumor immunity in preclinical models. Immune monitoring data suggest that optimized polyepitope neoantigen DNA vaccines are capable of inducing neoantigen-specific T cell responses in a patient with metastatic pancreatic neuroendocrine tumor. CONCLUSIONS: We have developed and optimized a novel polyepitope neoantigen DNA vaccine platform that can target multiple neoantigens and induce antitumor immune responses in preclinical models and neoantigen-specific responses in clinical translation.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Immunity , Translational Research, Biomedical , Vaccines, DNA/immunology , Adult , Animals , Antigen Presentation/immunology , Cell Proliferation , Disease Models, Animal , Female , HeLa Cells , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Male , Mammary Neoplasms, Animal/pathology , Mice, Inbred C57BL , Neoplasm Metastasis , Neuroendocrine Tumors/immunology , Neuroendocrine Tumors/pathology , Peptides/immunology , T-Lymphocytes/immunology
5.
Cancer Gene Ther ; 25(1-2): 27-38, 2018 02.
Article in English | MEDLINE | ID: mdl-29242639

ABSTRACT

Adenoviral (Ad) vector vaccines represent one of the most promising modern vaccine platforms, and Ad vector vaccines are currently being investigated in human clinical trials for infectious disease and cancer. Our studies have shown that specific targeting of adenovirus to dendritic cells dramatically enhanced vaccine efficacy. However, this was achieved using a molecular adapter, thereby necessitating a two component vector approach. To address the mandates of clinical translation of our strategy, we here sought to accomplish the goal of DC targeting with a single-component adenovirus vector approach. To redirect the specificity of Ad vector vaccines, we replaced the Ad fiber knob with fiber-fibritin chimeras fused to DC1.8, a single-domain antibody (sdAb) specific for murine immature DC. We engineered a fiber-fibritin-sdAb chimeric molecule using the coding sequence for DC1.8, and then replaced the native Ad5 fiber knob sequence by homologous recombination. The resulting Ad5 virus, Ad5FF1.8, expresses the chimeric fiber-fibritin sdAb chimera. Infection with Ad5FF1.8 dramatically enhances transgene expression in DC2.4 dendritic cells compared with infection with native Ad5. Ad5FF1.8 infection of bone marrow-derived DC demonstrates that Ad5FF1.8 selectively infects immature DC consistent with the known specificity of DC1.8. Thus, sdAb can be used to selectively redirect the tropism of Ad5 vector vaccines, providing the opportunity to engineer Ad vector vaccines that are specifically targeted to DC, or specific DC subsets.


Subject(s)
Adenoviridae , Dendritic Cells/immunology , Genetic Vectors , Vaccines , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Genetic Vectors/genetics , Genetic Vectors/immunology , Mice , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Vaccines/genetics , Vaccines/immunology , Viral Proteins/genetics , Viral Proteins/immunology
6.
Cancer Immunol Res ; 5(7): 516-523, 2017 07.
Article in English | MEDLINE | ID: mdl-28619968

ABSTRACT

Next-generation sequencing technologies have provided insights into the biology and mutational landscape of cancer. Here, we evaluate the relevance of cancer neoantigens in human breast cancers. Using patient-derived xenografts from three patients with advanced breast cancer (xenografts were designated as WHIM30, WHIM35, and WHIM37), we sequenced exomes of tumor and patient-matched normal cells. We identified 2,091 (WHIM30), 354 (WHIM35), and 235 (WHIM37) nonsynonymous somatic mutations. A computational analysis identified and prioritized HLA class I-restricted candidate neoantigens expressed in the dominant tumor clone. Each candidate neoantigen was evaluated using peptide-binding assays, T-cell cultures that measure the ability of CD8+ T cells to recognize candidate neoantigens, and preclinical models in which we measured antitumor immunity. Our results demonstrate that breast cancer neoantigens can be recognized by the immune system, and that human CD8+ T cells enriched for prioritized breast cancer neoantigens were able to protect mice from tumor challenge with autologous patient-derived xenografts. We conclude that next-generation sequencing and epitope-prediction strategies can identify and prioritize candidate neoantigens for immune targeting in breast cancer. Cancer Immunol Res; 5(7); 516-23. ©2017 AACR.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Animals , Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Epitope Mapping , Epitopes/genetics , Exome/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Mice , Mutation/genetics , Mutation/immunology , T-Lymphocytes, Cytotoxic/immunology , Xenograft Model Antitumor Assays
8.
Fertil Steril ; 103(2): 469-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25497447

ABSTRACT

OBJECTIVE: To exclude minimal residual disease in remaining ovarian tissue after harvesting the ovarian cortex for cryopreservation, by means of a tailor-made approach. DESIGN: Retrospective case series. SETTING: Hospital laboratory. PATIENT(S): We evaluated the ovarian and tubal tissue from 47 cancer patients (breast cancer, [non-]Hodgkin lymphoma; osteo-, Ewing, myxoid lipo-, and oropharyngeal synovial sarcoma; cervical, rectal, and esophageal cancer), who had stored ovarian tissue for fertility preservation. INTERVENTION(S): Immunohistochemistry (IHC) with tumor-related antibodies and genetic mutation analysis were performed to detect micrometastases by multiple sectioning at three levels of the paraffin-embedded formalin-fixed material. Molecular assays were performed with the use of tissue between these three levels of sectioning. MAIN OUTCOME MEASURE(S): Detection of micrometastases in ovaries. RESULT(S): We analyzed 847 ovarian slides to detect isolated tumor cells (ITCs) or micrometastases by IHC. In only one case (1/47) were ITCs detected in the fallopian tube. That patient had an intra-abdominal metastatic esophageal carcinoma. Additional DNA analyses of breast and rectal cancer, Ewing sarcoma, and human papilloma virus in cervical patients did not show evidence of micrometastases in the ovarian tissue. CONCLUSION(S): The tailor-made approach consisted of patient-specific tumor markers which were used to search for ovarian micrometastases. We found evidence of metastatic disease within the fallopian tube of a patient with intraperitoneal metastatic esophageal adenocarcinoma.


Subject(s)
Cryopreservation/methods , Fertility Preservation/methods , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/secondary , Ovary/pathology , Adolescent , Adult , Biomarkers, Tumor/genetics , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/genetics , Esophageal Neoplasms/secondary , Fallopian Tube Neoplasms/diagnosis , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/secondary , Female , Humans , Ovarian Neoplasms/genetics , Retrospective Studies , Transplantation, Autologous/methods , Young Adult
9.
Mol Cancer Res ; 11(4): 418-26, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23378578

ABSTRACT

The epithelial cell adhesion molecule (EpCAM) is a 40-kD type I transmembrane protein that is overexpressed in human epithelial cancers and is currently the target of molecular therapy based on its overexpression at the cell surface. Recently, we and others have shown a role for EpCAM in cell signaling and carcinogenesis, and EpCAM expression seems to promote breast cancer invasion. Interleukin-8 (IL-8/CXCL-8) is an inflammatory cytokine that has recently been shown to modulate breast cancer invasion and angiogenesis. In preliminary experiments, we identified a correlation between EpCAM and IL-8 expression in primary human breast cancers. Specific ablation of EpCAM in breast cancer cell lines results in decreased IL-8 expression, and IL-8 contributes to EpCAM-dependent breast cancer invasion. Specific ablation of EpCAM is also associated with decreased NF-κB transcription factor activity, decreased phosphorylation of the NF-κB family member RELA, and increased IκBα protein expression. EpCAM modulates IL-8 expression at baseline, and following IL-1ß stimulation, which is known to be a potent inducer of NF-κB in breast cancer. In functional rescue experiments, specific ablation of RELA or forced expression of the NF-κB inhibitor protein IκBα prevented EpCAM-dependent rescue of IL-8 promoter activity. These studies show for the first time that EpCAM can modulate NF-κB transcription factor activity and IL-8 expression in breast cancer and confirm the role of EpCAM signaling in modulating breast cancer invasion. Further study is required to define the molecular mechanism(s) of EpCAM signaling in breast cancer and to direct the rational development of molecular therapies targeting EpCAM.


Subject(s)
Antigens, Neoplasm/biosynthesis , Breast Neoplasms/metabolism , Cell Adhesion Molecules/biosynthesis , Interleukin-8/biosynthesis , NF-kappa B/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Female , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , MCF-7 Cells , NF-kappa B/genetics , Neoplasm Invasiveness , Signal Transduction
10.
Breast Cancer Res Treat ; 138(1): 109-18, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22678162

ABSTRACT

Mammaglobin-A (Mam-A) is a 10 kDa secretory protein that is overexpressed in 80 % of primary and metastatic human breast cancers. Previous studies from our laboratory demonstrated that Mam-A cDNA vaccine can induce Mam-A-specific CD8 T cell responses and mediate regression of human breast cancer xenografts in NOD/SCID mice. In this article, we present our results on a phase I clinical trial of a Mam-A cDNA vaccination in breast cancer patients with stage-IV metastatic disease, including the impact of vaccination on the expression of the inducible co-stimulator molecule (ICOS) on CD4 T cells. Specimens from seven patients with stage-IV metastatic cancer were available for these analyses. Patients were vaccinated with a Mam-A cDNA vaccine on days 0, 28, and 56, and immune responses were assessed at serial time points following vaccination. At 6 months following the first vaccination, flow cytometric analysis demonstrated a significant increase in the frequency of CD4+ICOS(hi) T cells from 5 ± 2 % pre-vaccination to 23 ± 4 % (p < 0.001), with a concomitant decrease in the frequency of CD4+FoxP3+ T cells (regulatory T cells [Treg]) from 19 ± 6 to 10 ± 5 % (p < 0.05). ELISpot analysis of CD4+ICOS(hi) sorted T cells demonstrated that following vaccination the cytokines produced by Mam-A-specific T cells switched from IL-10 (78 ± 21 spm pre-vaccination to 32 ± 14 spm 5 months post-vaccine p < 0.001) to IFN-γ (12 ± 6 spm pre-vaccination to 124 ± 31 spm 5 months post-vaccine p < 0.001). The ratio of CD4+ICOS(hi) T cells to CD4+FoxP3+ T cells increased from 0.37 ± 0.12 before vaccination to 2.3 ± 0.72 (p = 0.021) following vaccination. Further, these activated CD4+ICOS(hi) T cells induced preferential lysis of human breast cancer cells expressing Mam-A protein. We conclude that Mam-A cDNA vaccination is associated with specific expansion and activation of CD4+ICOS(hi) T cells, with a concomitant decrease in Treg frequency. These encouraging results strongly suggest that Mam-A cDNA vaccination can induce antitumor immunity in breast cancer patients.


Subject(s)
Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Mammaglobin A/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology , Breast Neoplasms/therapy , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , DNA, Complementary , Female , Forkhead Transcription Factors/metabolism , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lymphocyte Count , Mammaglobin A/genetics , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
11.
Proc Natl Acad Sci U S A ; 109(31): 12716-21, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22802630

ABSTRACT

Activation of naïve cluster of differentiation (CD)8(+) cytotoxic T lymphocytes (CTLs) is a tightly regulated process, and specific dendritic cell (DC) subsets are typically required to activate naive CTLs. Potential pathways for antigen presentation leading to CD8(+) T-cell priming include direct presentation, cross-presentation, and cross-dressing. To distinguish between these pathways, we designed single-chain trimer (SCT) peptide-MHC class I complexes that can be recognized as intact molecules but cannot deliver antigen to MHC through conventional antigen processing. We demonstrate that cross-dressing is a robust pathway of antigen presentation following vaccination, capable of efficiently activating both naïve and memory CD8(+) T cells and requires CD8α(+)/CD103(+) DCs. Significantly, immune responses induced exclusively by cross-dressing were as strong as those induced exclusively through cross-presentation. Thus, cross-dressing is an important pathway of antigen presentation, with important implications for the study of CD8(+) T-cell responses to viral infection, tumors, and vaccines.


Subject(s)
Antigen Presentation , Antigens, CD/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Integrin alpha Chains/immunology , Vaccination , Animals , Antigens, CD/genetics , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , Dendritic Cells/cytology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Integrin alpha Chains/genetics , Mice , Mice, Knockout , Peptides/genetics , Peptides/immunology
12.
Hum Immunol ; 73(1): 11-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22074997

ABSTRACT

Human breast cancer-associated antigen, mammaglobin-A (Mam-A), potentially offers a novel therapeutic target as a breast cancer vaccine. In this study, we define the CD8(+) cytotoxic T lymphocyte (CTL) response to Mam-A-derived candidate epitopes presented in the context of HLA-A24 (A*2402). HLA-A24 has a frequency of 72% in Japanese, 27% in Asian Indian, and 18% in Caucasian populations. Using a human leukocyte antigen (HLA)-binding prediction algorithm we identified 7 HLA-A24-restricted Mam-A-derived candidate epitopes (MAA24.1-7). Membrane stabilization studies with TAP-deficient T2 cells transfected with HLA-A2402 (T2.A24) indicated that MAA24.2 (CYAGSGCPL) and MAA24.4 (ETLSNVEVF) have the highest HLA-A24 binding affinity. Further, 2 CD8(+) CTL cell lines generated in vitro against T2.A24 cells individually loaded with Mam-A-derived candidate epitopes demonstrated significant cytotoxic activity against MAA24.2 and MAA24.4. In addition, the same CD8(+) CTL lines lysed the HLA-A24(+)/Mam-A(+) stable transfected human breast cancer cell lines AU565 and MDA-MB-361. However, these CTLs had no cytotoxicity against HLA-A24(-)/Mam-A(+) and HLA-A24(+)/Mam-A(-) breast cancer cell lines. In summary, our results define HLA-A24-restricted, Mam-A-derived, CD8(+) CTL epitopes that can potentially be employed for Mam-A-based breast cancer vaccine therapy to breast cancer patients with HLA-A24 phenotype.


Subject(s)
Breast Neoplasms/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A24 Antigen/immunology , Mammaglobin A/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Binding, Competitive/immunology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Female , HLA-A24 Antigen/genetics , HLA-A24 Antigen/metabolism , Humans , Mammaglobin A/genetics , Protein Binding/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transfection
13.
Breast Cancer Res ; 13(6): R124, 2011.
Article in English | MEDLINE | ID: mdl-22132731

ABSTRACT

INTRODUCTION: EpCAM is a cell-surface glycoprotein that is overexpressed in the majority of epithelial carcinomas. However, the functional role of EpCAM in regulating cancer invasion remains controversial, and the mechanism(s) underlying EpCAM-mediated regulation of breast cancer invasion remain to be defined. METHODS: EpCAM expression was manipulated in breast cancer cell lines using RNA interference and cDNA expression constructs. Recombinant EpCAM was used to rescue EpCAM signaling following specific ablation of EpCAM. Protein and gene expression, invasion, transcription factor activity, and protein phosphorylation were measured using standard molecular biology techniques. RESULTS: In loss-of-function, and gain-of-function experiments we demonstrate that EpCAM expression is associated with increased breast cancer invasion in vitro and in vivo. We demonstrate further that specific ablation of EpCAM expression is associated with decreased activator protein-1 (AP-1) transcription factor activity. Phosphoprotein analyses confirm that specific ablation of EpCAM is associated with decreased phosphorylation of the AP-1 subunit c-Jun. Recombinant soluble extracellular EpCAM (rEpCAM) is able to rescue invasion, AP-1 transcription factor activity, and c-Jun phosphorylation in a dose-dependent fashion. Pharmacologic inhibitors, and constitutively active constructs of the c-Jun N-terminal kinase (JNK) signal transduction pathway, suggest that the impact of EpCAM expression on AP-1 transcription factor activity is mediated through the JNK pathway. In functional rescue experiments, forced expression of c-Jun rescues invasion in breast cancer cells following specific ablation of EpCAM. CONCLUSIONS: These data demonstrate for the first time that EpCAM expression can influence the JNK/AP-1 signal transduction pathway, and suggest that modulation of AP-1 transcription factor activity contributes to EpCAM-dependent breast cancer invasion. These data have important implications for the design and application of molecular therapies targeting EpCAM.


Subject(s)
Antigens, Neoplasm/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Transcription Factor AP-1/metabolism , Animals , Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Neoplasm Invasiveness , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Xenograft Model Antitumor Assays
14.
J Biomed Opt ; 16(8): 080503, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21895303

ABSTRACT

Photoacoustic tomography is a hybrid modality based on optical absorption excitation and ultrasonic detection. It is sensitive to melanin, one of the primary absorbers in skin. For cells that do not naturally contain melanin, melanin production can be induced by introducing the gene for tyrosinase, the primary enzyme responsible for expression of melanin in melanogenic cells. Optical resolution photoacoustic microscopy was used in the ex vivo study reported here, where the signal from transfected cells increased by more than 10 times over wild-type cells. A subsequent in vivo experiment was conducted to demonstrate the capability of photoacoustic microscopy to spectrally differentiate between tyrosinase-catalyzed melanin and various other absorbers in tissue.


Subject(s)
Genes, Reporter , Microscopy/methods , Molecular Imaging/methods , Monophenol Monooxygenase/metabolism , Signal Processing, Computer-Assisted , Tomography/methods , Equipment Design , HEK293 Cells , Hemoglobins , Humans , Melanins/chemistry , Melanins/metabolism , Microscopy/instrumentation , Oxyhemoglobins , Phantoms, Imaging , Tomography/instrumentation , Ultrasonography/methods
15.
Cancers (Basel) ; 3(4): 4191-211, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-24213133

ABSTRACT

New DNA sequencing platforms have revolutionized human genome sequencing. The dramatic advances in genome sequencing technologies predict that the $1,000 genome will become a reality within the next few years. Applied to cancer, the availability of cancer genome sequences permits real-time decision-making with the potential to affect diagnosis, prognosis, and treatment, and has opened the door towards personalized medicine. A promising strategy is the identification of mutated tumor antigens, and the design of personalized cancer vaccines. Supporting this notion are preliminary analyses of the epitope landscape in breast cancer suggesting that individual tumors express significant numbers of novel antigens to the immune system that can be specifically targeted through cancer vaccines.

16.
Breast Cancer Res Treat ; 127(1): 81-9, 2011 May.
Article in English | MEDLINE | ID: mdl-20544273

ABSTRACT

Mammaglobin-A (MGBA), a 10-kD protein, is over expressed in 80% of primary and metastatic human breast cancers. Breast cancer patients demonstrate high frequencies of CD8(+) cytotoxic T lymphocytes (CTL) specific to MGBA. Defining CD8(+) CTL responses to HLA class I-restricted MGBA-derived epitopes assumes significance in the context of our ongoing efforts to clinically translate vaccine strategies targeting MGBA for prevention and/or treatment of human breast cancers. In this study, we define the CD8(+) CTL response to MGBA-derived candidate epitopes presented in the context of HLA-B7, which has a frequency of 17.7% in Caucasian and 15.5% in African American populations. We identified seven MGBA-derived candidate epitopes with high predicted binding scores for HLA-B7 using a computer algorithm. Membrane stabilization studies with TAP-deficient T2 cells transfected with HLA-B7 indicated that MGBA B7.3 (VSKTEYKEL), B7.6 (KLLMVLMLA), B7.7 (NPQVSKTEY), and B7.1 (YAGSGCPLL) have the highest HLA-B7 binding affinities. Further, two CD8(+) CTL cell lines generated in vitro against T2.B7 cells individually loaded with MGBA-derived candidate epitopes showed significant cytotoxic activity against MGBA B7.1, B7.3, B7.6, and B7.7. In addition, the same CD8(+) CTL lines lysed the HLA-B7(+)/MGBA(+) human breast cancer cell line DU-4475 but had no significant cytotoxicity against HLA-B7(-) or MGBA(-) breast cancer cell lines. Cold-target inhibition studies strongly suggest that MGBA B7.3 is an immunodominant epitope. In summary, our results define HLA-B7-restriced, MGBA-derived, CD8(+) CTL epitopes with all of the necessary features for developing novel vaccine strategies against HLA-B7 expressing breast cancer patients.


Subject(s)
Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-B7 Antigen/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Uteroglobin/immunology , Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Antibody Specificity/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Mammaglobin A
17.
Vaccine ; 28(8): 1911-8, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20188246

ABSTRACT

It is commonly believed that delivery of antigen into the class I antigen presentation pathway is a limiting factor in the clinical translation of DNA vaccines. This is of particular concern in the context of cancer vaccine development as many immunodominant peptides derived from self tumor antigens are not processed and presented efficiently. To address this limitation, we have engineered completely assembled peptide/MHC class I complexes whereby all three components (class I heavy chain, beta(2)m, and peptide) are attached by flexible linkers and expressed as a single polypeptide (single chain trimers or SCT). In this study, we tested the efficacy of progressive generations of SCT DNA vaccines engineered to (1) enhance peptide binding, (2) enhance interaction with the CD8 coreceptor, and/or (3) activate CD4(+) helper T cells. Disulfide trap SCT (dtSCT) have been engineered to improve peptide binding, with mutations designed to create a disulfide bond between the class I heavy chain and the peptide linker. dtSCT DNA vaccines dramatically enhance the immune response to model low affinity antigens as measured by ELISPOT analysis and tumor challenge. SCT engineered to enhance interaction with the CD8 coreceptor have a higher affinity for the TCR/CD8 complex, and are associated with more robust CD8(+) T cell responses following vaccination. Finally, SCT constructs that coexpress a universal helper epitope PADRE, dramatically enhance CD8(+) T cell responses. Taken together, our data demonstrate that dtSCT DNA vaccines coexpressing a universal CD4 epitope are highly effective in generating immune responses to poorly processed and presented cancer antigens.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Genes, MHC Class I , Lymphocyte Activation , Vaccines, DNA/immunology , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , HLA-A2 Antigen/immunology , Humans , Mammaglobin A , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Neoplasm Proteins/immunology , Ovalbumin/immunology , Protein Binding , Protein Engineering , Uteroglobin/immunology , Vaccines, DNA/biosynthesis , Vaccines, DNA/genetics , beta 2-Microglobulin/immunology
18.
Breast Cancer Res Treat ; 123(3): 691-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19967557

ABSTRACT

Several gene expression profiles have been reported to predict breast cancer response to neoadjuvant chemotherapy. These studies often consider breast cancer as a homogeneous entity, although higher rates of pathologic complete response (pCR) are known to occur within the basal-like subclass. We postulated that profiles with higher predictive accuracy could be derived from a subset analysis of basal-like tumors in isolation. Using a previously described "intrinsic" signature to differentiate breast tumor subclasses, we identified 50 basal-like tumors from two independent clinical trials associated with gene expression profile data. 24 tumor data sets were derived from a 119-patient neoadjuvant trial at our institution and an additional 26 tumor data sets were identified from a published data set (Hess et al. J Clin Oncol 24:4236-4244, 2006). The combined 50 basal-like tumors were partitioned to form a 37 sample training set with 13 sequestered for validation. Clinical surveillance occurred for a mean of 26 months. We identified a 23-gene profile which predicted pCR in basal-like breast cancers with 92% predictive accuracy in the sequestered validation data set. Furthermore, distinct cluster of patients with high rates of cancer recurrence was observed based on cluster analysis with the 23-gene signature. Disease-free survival analysis of these three clusters revealed significantly reduced survival in the patients of this high recurrence cluster. We identified a 23-gene signature which predicts response of basal-like breast cancer to neoadjuvant chemotherapy as well as disease-free survival. This signature is independent of tissue collection method and chemotherapeutic regimen.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Testing , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/genetics , Adult , Aged , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Clinical Trials as Topic , Cluster Analysis , Disease-Free Survival , Female , Gene Expression Profiling/methods , Humans , Kaplan-Meier Estimate , Middle Aged , Neoadjuvant Therapy , Neoplasms, Basal Cell/mortality , Neoplasms, Basal Cell/pathology , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Reproducibility of Results , Time Factors , Treatment Outcome
19.
J Surg Res ; 156(1): 50-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19524940

ABSTRACT

RNA interference (RNAi) is a powerful new tool for the selective ablation of gene expression, facilitating loss-of-function studies. However, appropriate controls are considered essential to confirm the specificity of RNAi experiments. The most stringent control is rescue of the target gene in a form that is refractory to RNAi. To facilitate rescue of the target gene, we have created improved dual expression lentiviral vectors with the ability to simultaneously drive expression of a shRNA for RNA interference and a rescue transgene in a single vector system. In proof-of-principle experiments, we ablated more than 90% of target gene expression by targeting either the open reading frame, or the 3' UTR region. Target gene expression was successfully rescued with a cDNA containing silent third-codon point mutations in the targeted region or with native cDNA when the 3' UTR was targeted. Finally, expression of the rescue transgene can be manipulated by positional cloning and appropriate promoter selection. The dual expression lentiviral vectors described here represent a versatile strategy for confirming the integrity of RNAi experiments and may facilitate functional analyses even in the absence of an established gain-of-function model system.


Subject(s)
Genetic Vectors , Lentivirus , RNA Interference , Cell Line , Gene Expression , Humans , Transgenes
20.
Cancer Cell Int ; 9: 8, 2009 Mar 23.
Article in English | MEDLINE | ID: mdl-19309500

ABSTRACT

BACKGROUND: Mammaglobin (MAM) has been used as a specific molecular marker for breast cancer diagnosis. Recently, several groups of researchers proposed a number of therapeutic strategies targeting this molecule. Some of the strategies are based upon an essential but not demonstrated hypothesis - mammaglobin is associated with the surface of breast cancer cells, which strongly disputes the therapeutic strategies. RESULTS: We conducted a computer-based predictive analysis and identified a small fragment at the N-end of MAM as a potential transmembrane domain. We provided several evidences to demonstrate the presence of the membrane-associated MAM. We isolated the membrane protein components from known MAM positive breast cancer cells (MDA-MB361 and MDA-MB415). We showed that about 22-64% of MAM proteins, depending upon the types of the cancer cells, directly attached on the membrane of breast cancer cells, by Western blotting assays. To directly visualize the presence of the membrane-bound MAM protein, we incubated the MAM positive cancer cells with FITC labeled anti-MAM antibody, and observed clear fluorescent signals on the surface of the cells. In studying the MAM protein distribution in human breast cancer tissues, we first identified two immunostain patterns that are associated with the membrane-bound MAM: the membrane stain pattern and luminary surface stain pattern. To test whether the membrane-associated MAM can serve as a molecular target for drug delivery, we conjugated anti-MAM antibody to human low-density lipoprotein (LDL) and loaded doxorubicin (Dox) in the core of LDL. Specific binding and cytotoxicity of the MAM targeted and Dox loaded LDL was tested in the MAM positive breast cancer cells in vitro. CONCLUSION: We first showed that some of MAM protein directly associated with the surface of breast cancer cells. The membrane-associated MAM protein may be utilized as a useful molecular marker for breast cancer targeted drug delivery.

SELECTION OF CITATIONS
SEARCH DETAIL
...