Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Clin Cancer Res ; 23(19): 5892-5911, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28679769

ABSTRACT

Purpose: We investigated the role of the human endogenous retrovirus type K (HERV-K) envelope (env) gene in pancreatic cancer.Experimental Design: shRNA was employed to knockdown (KD) the expression of HERV-K in pancreatic cancer cells.Results: HERV-K env expression was detected in seven pancreatic cancer cell lines and in 80% of pancreatic cancer patient biopsies, but not in two normal pancreatic cell lines or uninvolved normal tissues. A new HERV-K splice variant was discovered in several pancreatic cancer cell lines. Reverse transcriptase activity and virus-like particles were observed in culture media supernatant obtained from Panc-1 and Panc-2 cells. HERV-K viral RNA levels and anti-HERV-K antibody titers were significantly higher in pancreatic cancer patient sera (N = 106) than in normal donor sera (N = 40). Importantly, the in vitro and in vivo growth rates of three pancreatic cancer cell lines were significantly reduced after HERV-K KD by shRNA targeting HERV-K env, and there was reduced metastasis to lung after treatment. RNA-Seq results revealed changes in gene expression after HERV-K env KD, including RAS and TP53. Furthermore, downregulation of HERV-K Env protein expression by shRNA also resulted in decreased expression of RAS, p-ERK, p-RSK, and p-AKT in several pancreatic cancer cells or tumors.Conclusions: These results demonstrate that HERV-K influences signal transduction via the RAS-ERK-RSK pathway in pancreatic cancer. Our data highlight the potentially important role of HERV-K in tumorigenesis and progression of pancreatic cancer, and indicate that HERV-K viral proteins may be attractive biomarkers and/or tumor-associated antigens, as well as potentially useful targets for detection, diagnosis, and immunotherapy of pancreatic cancer. Clin Cancer Res; 23(19); 5892-911. ©2017 AACR.


Subject(s)
Carcinogenesis/genetics , Endogenous Retroviruses/genetics , Pancreatic Neoplasms/genetics , Viral Envelope Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Endogenous Retroviruses/pathogenicity , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Gene Knockdown Techniques , Host-Pathogen Interactions/genetics , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Signal Transduction/genetics
3.
Br J Haematol ; 178(3): 413-426, 2017 08.
Article in English | MEDLINE | ID: mdl-28508448

ABSTRACT

The identification of novel tumour-associated antigens is urgently needed to improve the efficacy of immunotherapy for multiple myeloma (MM). In this study, we identified a membrane protein MMSA-1 (multiple myeloma special antigen-1) that was specifically expressed in MM and exhibited significantly positive correlation with MM. We then identified HLA-A*0201-restricted MMSA-1 epitopes and tested their cytotoxic T lymphocyte (CTL) response. The MMSA-1 epitope SLSLLTIYV vaccine was shown to induce an obvious CTL response in vitro. To improve the immunotherapy, we constructed a multi-epitope peptide vaccine by combining epitopes derived from MMSA-1 and Dickkopf-1 (DKK1). The effector T cells induced by multi-epitope peptide vaccine-loaded dendritic cells lysed U266 cells more effectively than MMSA-1/DKK1 single-epitope vaccine. In myeloma-bearing severe combined immunodeficient mice, the multi-epitope vaccine improved the survival rate significantly compared with single-epitope vaccine. Consistently, multi-epitope vaccine decreased the tumour volume greatly and alleviated bone destruction. The frequencies of CD4+ and CD8+ T cells was significantly increased in mouse blood induced by the multi-epitope vaccine, indicating that it inhibits myeloma growth by changing T cell subsets and alleviating immune paralysis. This study identified a novel peptide from MMSA-1 and the multi-epitope vaccine will be used to establish appropriate individualized therapy for MM.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Intercellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Multiple Myeloma/therapy , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Apoptosis/physiology , Bone Diseases/etiology , Bone Diseases/immunology , Cancer Vaccines/immunology , Cell Cycle Checkpoints/physiology , Cell Movement/physiology , Cell Proliferation , Cytokines/blood , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Female , Gene Silencing , Humans , Immunotherapy/methods , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice, SCID , Multiple Myeloma/complications , Multiple Myeloma/immunology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Sci Rep ; 7: 41960, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165048

ABSTRACT

Human endogenous retroviruses (HERVs), which make up approximately 8% of the human genome, are overexpressed in some breast cancer cells and tissues but without regard to cancer subtype. We, therefore, analyzed TCGA RNA-Seq data to evaluate differences in expression of the HERV-K family in breast cancers of the various subtypes. Four HERV-K loci on different chromosomes were analyzed in basal, Her2E, LumA, and LumB breast cancer subtypes of 512 breast cancer patients with invasive ductal carcinoma (IDC). The results for all four loci showed higher HERV-K expression in the basal subtype, suggesting similar mechanisms of regulation regardless of locus. Expression of the HERV-K envelope gene (env) was highly significantly increased in basal tumors in comparison with the also-upregulated expression of other HERV-K genes. Analysis of reverse-phase protein array data indicated that increased expression of HERV-K is associated with decreased mutation of H-Ras (wild-type). Our results show elevation of HERV-K expression exclusively in the basal subtype of IDC breast cancer (as opposed to the other subtypes) and suggest HERV-K as a possible target for cancer vaccines or immunotherapy against this highly aggressive form of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Basal Cell/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Endogenous Retroviruses/genetics , Viral Envelope Proteins/genetics , Breast Neoplasms/genetics , Carcinoma, Basal Cell/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/genetics , Databases, Factual , Female , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Genome, Human , Humans
5.
Oncol Rep ; 36(5): 2435-2443, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666407

ABSTRACT

Depression is a common symptom among gastric cancer (GC) patients and serves as a potential indication of poor prognosis and advanced cancer clinical stage. However, the molecular mechanism of depression­associated poor prognoses of GC patients remains unclear. Recent studies have revealed that GC patients with depression are under high levels of oxidative stress (OS) status that is accompanied by the dysfunction of numerous proto­oncogenes, including the ABL proto­oncogene 1 (ABL1), which is a non­receptor tyrosine kinase. Recent evidence indicates that ABL1 was dysregulated in both major depressive disorder (MDD) and cancer patients with depression, and high levels of reactive oxygen species (ROS) can lead to the activation of ABL1 in response to OS and that activated ABL1 subsequently contributes to development of GC via interactions with the downstream targets and corresponding signaling pathways. In this review, we examine the evidence to illuminate the molecular mechanism of ABL1 in the progression of GC patients with depression and identify out new and effective methods for the initial and long­term treatment of GC.


Subject(s)
Depressive Disorder/genetics , Oncogene Proteins v-abl/biosynthesis , Stomach Neoplasms/genetics , Depressive Disorder/drug therapy , Depressive Disorder/etiology , Depressive Disorder/pathology , Gene Expression Regulation, Neoplastic , Humans , Oncogene Proteins v-abl/genetics , Oxidative Stress/genetics , Prognosis , Reactive Oxygen Species , Stomach Neoplasms/complications , Stomach Neoplasms/drug therapy , Stomach Neoplasms/psychology
6.
Int J Oncol ; 49(5): 1791-1800, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27633118

ABSTRACT

Gastric cancer (GC) remains one of the most prevalent tumors worldwide and affects human health due to its high morbidity and mortality. Mechanisms underlying occurrence and development of GC have been widely studied. Studies have revealed reactive oxygen species (ROS) generated by cells under oxidative stress (OS) are involved in gastric tumorigenesis, and modulate expression of microRNAs (miRs). As such, miRs have been shown to be associated with OS-related GC. Given the association of OS and miRs in development of GC, this review aims to summarize the relationship between miRs and OS and their role in GC development. Serving as a link between OS and GC, miRs may offer new approaches for gaining a more in-depth understanding of mechanisms of GC and may lead to the identification of new therapeutic approaches against GC.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Oxidative Stress/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Humans
7.
Oncotarget ; 7(51): 84093-84117, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27557521

ABSTRACT

Human endogenous retrovirus type K (HERV-K) Env protein was previously demonstrated to be overexpressed in human breast cancer (BC) cells and tissues. However, the molecular pathways driving the specific alterations are unknown. We now show that knockdown of its expression with an shRNA (shRNAenv) blocked BC cell proliferation, migration, and invasion. shRNAenv transduction also attenuated the ability of BC cells to form tumors, and notably prevented metastasis. Mechanistically, downregulation of HERV-K blocked expression of tumor-associated genes that included Ras, p-RSK, and p-ERK. The major upstream regulators influenced by HERV-K knockdown were p53, TGF- ß1, and MYC. Of interest, when the HERV-K env gene was overexpressed in shRNAenv-transduced BC cells using an HERV-K env expression vector, Ras/Raf/MEK/ERK pathway signaling was restored. CDK5, which alters p53 phosphorylation in some cancers, was upregulated and p53 was downregulated when HERV-K was overexpressed. CDK5 is also a mediator of TGF-ß1-induced epithelial-mesenchymal transition and migration in cancer cells, and is involved in tumor formation. Importantly, reductions in migration, invasion, and transformation of BC cells stably transduced with shRNAenv was reversed after adding back a vector with a synonymous mutation of HERV-K env. Taken together, these results indicate that HERV-K Env protein plays an important role in tumorigenesis and metastasis of BC.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Gene Products, env/genetics , Xenograft Model Antitumor Assays , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interleukin Receptor Common gamma Subunit/genetics , MCF-7 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , RNA Interference , RNAi Therapeutics , Signal Transduction/genetics
8.
APMIS ; 124(1-2): 105-15, 2016.
Article in English | MEDLINE | ID: mdl-26818265

ABSTRACT

Endogenous retroviral sequences are spread throughout the genome of all humans, and make up about 8% of the genome. Despite their prevalence, the function of human endogenous retroviruses (HERVs) in humans is largely unknown. In this review we focus on the brain, and evaluate studies in animal models that address mechanisms of endogenous retrovirus activation in the brain and central nervous system (CNS). One such study in mice found that TRIM28, a protein critical for mouse early development, regulates transcription and silencing of endogenous retroviruses in neural progenitor cells. Another intriguing finding in human brain cells and mouse models was that endogenous retrovirus HERV-K appears to be protective against neurotoxins. We also report on studies that associate HERVs with human diseases of the brain and CNS. There is little doubt of an association between HERVs and a number of CNS diseases. However, a cause and effect relationship between HERVs and these diseases has not yet been established.


Subject(s)
Brain Diseases/virology , Brain/physiology , Brain/virology , Endogenous Retroviruses/physiology , Animals , Brain/growth & development , Brain/physiopathology , Brain Diseases/etiology , Brain Diseases/therapy , Central Nervous System/virology , Disease Models, Animal , Endogenous Retroviruses/genetics , Humans , Mice , Neurotoxins/toxicity , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Tripartite Motif-Containing Protein 28 , Virus Activation/genetics
9.
Oncoimmunology ; 4(11): e1047582, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26451325

ABSTRACT

We have previously reported that human endogenous retrovirus-K (HERV-K) envelope (env) protein is a tumor-associated antigen (TAA) for cancer vaccines, and that its antibodies (mAbs) possess antitumor activity against cancer. In this study, a chimeric antigen receptor (CAR) specific for HERV-K env protein (K-CAR) was generated using anti-HERV-K mAb. K-CAR T cells from peripheral blood mononuclear cells (PBMCs) of 9 breast cancer (BC) patients and 12 normal donors were able to inhibit growth of, and to exhibit significant cytotoxicity toward, BC cells but not MCF-10A normal breast cells. The antitumor effects in cancer cells were significantly reduced when control T cells were used, or the expression of HERV-K was knocked down by an shRNA. Secretion of multiple cytokines, including IFNγ, TNF-α, and IL-2, was significantly enhanced in culture media of BC cells treated with K-CARs. Significantly reduced tumor growth and tumor weight was observed in xenograft models bearing MDA-MB-231 or MDA-MB-435.eB1 BC cells. Importantly, the K-CAR prevented tumor metastasis to other organs. Furthermore, downregulation of HERV-K expression in tumors of mice treated with K-CAR correlated with upregulation of p53 and downregulation of MDM2 and p-ERK. Importantly, the expression of HERV-K env protein in metastatic tumor tissues treated with K-CAR T cells correlated with the expression of Ras. Our results indicate that HERV-K env protein is an oncoprotein and may play an important role in tumorigenesis related to p53 and Ras signaling pathways. Anti-HERV-K treatment, including K-CAR treatment, shows potential for immunotherapy of BC.

10.
Clin Cancer Res ; 21(14): 3241-51, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25829402

ABSTRACT

PURPOSE: The human endogenous retrovirus (HERV-K) envelope (env) protein is a tumor-associated antigen (TAA) expressed on melanoma but not normal cells. This study was designed to engineer a chimeric antigen receptor (CAR) on T-cell surface, such that they target tumors in advanced stages of melanoma. EXPERIMENTAL DESIGN: Expression of HERV-K protein was analyzed in 220 melanoma samples (with various stages of disease) and 139 normal organ donor tissues using immunohistochemical (IHC) analysis. HERV-K env-specific CAR derived from mouse monoclonal antibody was introduced into T cells using the transposon-based Sleeping Beauty (SB) system. HERV-K env-specific CAR(+) T cells were expanded ex vivo on activating and propagating cells (AaPC) and characterized for CAR expression and specificity. This includes evaluating the HERV-K-specific CAR(+) T cells for their ability to kill A375-SM metastasized tumors in a mouse xenograft model. RESULTS: We detected HERV-K env protein on melanoma but not in normal tissues. After electroporation of T cells and selection on HERV-K(+) AaPC, more than 95% of genetically modified T cells expressed the CAR with an effector memory phenotype and lysed HERV-K env(+) tumor targets in an antigen-specific manner. Even though there is apparent shedding of this TAA from tumor cells that can be recognized by HERV-K env-specific CAR(+) T cells, we observed a significant antitumor effect. CONCLUSIONS: Adoptive cellular immunotherapy with HERV-K env-specific CAR(+) T cells represents a clinically appealing treatment strategy for advanced-stage melanoma and provides an approach for targeting this TAA on other solid tumors.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Melanoma/virology , T-Lymphocytes/transplantation , Viral Proteins/immunology , Animals , Genetic Engineering/methods , Humans , Immunohistochemistry , Melanoma/immunology , Mice , Mice, Inbred NOD , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 21(2): 471-83, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25370465

ABSTRACT

PURPOSE: To determine whether HERV-K envelope (ENV) protein could function as a tumor-associated antigen and elicit specific T-cell responses against autologous ovarian cancer cells. EXPERIMENTAL DESIGN: The expression of HERV-K transcripts and ENV protein, the presence of serum antibodies against HERV-K, reverse transcriptase (RT) activities, and cellular immune responses in primary ovarian cancer tissues and patient blood samples were analyzed and compared with samples from patients with benign ovarian diseases and normal female donors. RESULTS: Ovarian cancer cells in primary tumors and ascites expressed markers of cancer stem cells and markers of both mesenchymal and epithelial cells. Expression of HERV transcripts and HERV-K ENV protein and reverse transcriptase activities were higher in ovarian cancer compared with adjacent normal and benign tissues. The ovarian cancer patient plasma also had high reverse transcriptase activities and the ovarian cancer patient sera contained HERV-K immunoreactive antibodies. HERV-K-specific T cells generated from autologous dendritic cells pulsed with HERV-K ENV antigens exhibited phenotypes and functions consistent with a cellular immune response including T-cell proliferation, IFNγ production, and HERV-K-specific cytotoxic T lymphocyte (CTL) activity. Significantly higher CTL lysis of autologous tumor cells than of uninvolved normal cells was demonstrated in patients with ovarian cancer than patients with benign diseases and further enhanced lysis was observed if T regulatory cells were depleted. CONCLUSION: Endogenous retroviral gene products in ovarian cancer may represent a potentially valuable new pool of tumor-associated antigens for targeting of therapeutic vaccines to ovarian cancer. Clin Cancer Res; 21(2); 471-83. ©2014 AACR.


Subject(s)
Endogenous Retroviruses/genetics , Gene Products, env/genetics , Ovarian Neoplasms/blood , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/blood , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic , Endogenous Retroviruses/metabolism , Female , Gene Products, env/blood , Humans , Lymphocyte Activation , Ovarian Neoplasms/virology , RNA-Directed DNA Polymerase/blood , RNA-Directed DNA Polymerase/genetics , T-Lymphocytes, Cytotoxic/virology
12.
Int J Cancer ; 137(6): 1249-57, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-24890612

ABSTRACT

Harbored as relics of ancient germline infections, human endogenous retroviruses (HERVs) now constitute up to 8% of our genome. A proportion of this sequence has been co-opted for molecular and cellular processes, beneficial to human physiology, such as the fusogenic activity of the envelope protein, a vital component of placentogenesis. However, the discovery of high levels of HERV-K mRNA and protein and even virions in a wide array of cancers has revealed that HERV-K may be playing a more sinister role-a role as an etiological agent in cancer itself. Whether the presence of this retroviral material is simply an epiphenomenon, or an actual causative factor, is a hotly debated topic. This review will summarize the current state of knowledge regarding HERV-K and cancer and attempt to outline the potential mechanisms by which HERV-K could be involved in the onset and promotion of carcinogenesis.


Subject(s)
Carcinogenesis/genetics , Neoplasms/genetics , Neoplasms/virology , Retroviridae/genetics , Animals , Humans , Neoplasms/etiology , RNA, Messenger/genetics , RNA, Viral/genetics
13.
Carcinogenesis ; 35(9): 2074-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24858205

ABSTRACT

Aberrant expression of subgroup k human endogenous retroviruses (HERV-K) has been observed in prostate cancer. This subgroup is unique because it encodes sequences in the human genome containing open reading frames for near intact retroviruses. We hypothesized that HERV-K reactivation could serve as a non-invasive early disease detection marker for prostate cancer. We evaluated HERV-K gag messenger RNA (mRNA) expression in blood samples of African-American and European-American men using a case-control design via quantitative real-time PCR. Additionally, we examined HERV-K envelope protein expression in prostate tumors by immunohistochemistry. HERV-K envelope protein was commonly upregulated in prostate tumors, but more so in tumors of African-American than European-American patients (61% versus 40%, P < 0.01). Examining HERV-K gag expression in peripheral blood mononuclear cells (PBMC) from 294 cases and 135 healthy men, we found that the abundance of HERV-K gag message was significantly higher in cases than controls and was associated with increased plasma interferon-γ. Men with gag expression in the highest quartile had >12-fold increased odds {odds ratio = 12.87 [95% confidence interval 6.3-26.25]} of being diagnosed with prostate cancer than those in the lowest quartile. Moreover, our results showed that HERV-K expression may perform better as a disease biomarker in older than younger men (whereas the sensitivity of prostate-specific antigen (PSA) testing decreases with age) and in men with a smoking history compared with never smokers. Combining non-invasive HERV-K testing with PSA testing may improve the efficacy of prostate cancer detection specifically among older men and smokers who tend to develop a more aggressive disease.


Subject(s)
Adenocarcinoma/blood , Gene Products, gag/blood , Leukocytes, Mononuclear/metabolism , Prostatic Neoplasms/blood , Smoking/blood , Adenocarcinoma/diagnosis , Adenocarcinoma/virology , Chemokine CXCL10/blood , Endogenous Retroviruses/enzymology , Gene Expression , Humans , Interferon-gamma/blood , Leukocytes, Mononuclear/virology , Male , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/virology , RNA, Messenger/blood , RNA, Messenger/genetics , Risk Factors
14.
Int J Cancer ; 134(3): 587-95, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23873154

ABSTRACT

A simple and accurate test to detect early-stage breast cancer has not been developed. Previous studies indicate that the level of human endogenous retrovirus type K (group HERV-K(HML-2)) transcription may be increased in human breast tumors. We hypothesized that HERV-K(HML-2) reactivation can serve as a biomarker for early detection of breast cancer. Serum samples were collected from women without cancer (controls) and patients with ductal carcinoma in situ (DCIS) and invasive breast cancer. ELISA assays were used to detect serum anti-HERV-K(HML-2) antibody titers. RNA was extracted from sera and analyzed by real-time RT-PCR to quantitate the level of HERV-K(HML-2) mRNA. We measured significantly higher serum mRNA and serum antibody titers against HERV-K(HML-2) proteins in women with DCIS and stage I disease than in women without cancer. At optimized cutoffs for the antibody titers, the assay produced an area under the receiver operating characteristic curve (AUC) of 0.89 (95% confidence interval 0.77-1.00) for DCIS and of 0.95 (95% confidence interval 0.89-1.00) for invasive breast cancer. These AUCs are comparable to those observed for mammograms. We also found that serum HERV-K(HML-2) mRNA tended to be higher in breast cancer patients with a primary tumor who later on developed the metastatic disease than in patients who did not develop cancer metastasis. Our results show that HERV-K(HML-2) antibodies and mRNA are already elevated in the blood at an early stage of breast cancer, and further increase in patients who are at risk of developing a metastatic disease.


Subject(s)
Antibodies, Viral/blood , Biomarkers, Tumor/blood , Breast Neoplasms/blood , Carcinoma, Intraductal, Noninfiltrating/blood , Endogenous Retroviruses/immunology , RNA, Messenger/blood , RNA, Viral/blood , Breast Neoplasms/pathology , Breast Neoplasms/virology , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/virology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Neoplasm Metastasis , Real-Time Polymerase Chain Reaction
15.
J Natl Cancer Inst ; 104(3): 189-210, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22247020

ABSTRACT

BACKGROUND: The envelope (env) protein of the human endogenous retrovirus type K (HERV-K) family is commonly expressed on the surface of breast cancer cells. We assessed whether HERV-K env is a potential target for antibody-based immunotherapy of breast cancer. METHODS: We examined the expression of HERV-K env protein in various malignant (MDA-MB-231, MCF-7, SKBR3, MDA-MB-453, T47D, and ZR-75-1) and nonmalignant (MCF-10A and MCF-10AT) human breast cell lines by immunoblot, enzyme-linked immunosorbent assay, immunofluorescence staining, and flow cytometry. Anti-HERV-K env monoclonal antibodies (mAbs; 6H5, 4D1, 4E11, 6E11, and 4E6) were used to target expression of HERV-K, and antitumor effects were assessed by quantifying growth and apoptosis of breast cancer cells in vitro, and tumor growth in vivo in mice (n = 5 per group) bearing xenograft tumors. The mechanisms responsible for 6H5 mAb-mediated effects were investigated by microarray assays, flow cytometry, immunoblot, and immunofluorescence staining. The expression of HERV-K env protein was assessed in primary breast tumors (n = 223) by immunohistochemistry. All statistical tests were two-sided. RESULTS: The expression of HERV-K env protein in malignant breast cancer cell lines was substantially higher than nonmalignant breast cells. Anti-HERV-K-specific mAbs inhibited growth and induced apoptosis of breast cancer cells in vitro. Mice treated with 6H5 mAb showed statistically significantly reduced growth of xenograft tumors compared with mice treated with control immunoglobulin (control [mIgG] vs 6H5 mAb, for tumors originating from MDA-MB-231 cells, mean size = 1448.33 vs 475.44 mm(3); difference = 972.89 mm(3), 95% CI = 470.17 to 1475.61 mm(3); P < .001). Several proteins involved in the apoptotic signaling pathways were overexpressed in vitro in 6H5 mAb-treated malignant breast cells compared with mIgG-treated control. HERV-K expression was detected in 148 (66%) of 223 primary breast tumors, and a higher rate of lymph node metastasis was associated with HERV-K-positive compared with HERV-K-negative tumors (43% vs 23%, P = .003). CONCLUSION: Monoclonal antibodies against HERV-K env protein show potential as novel immunotherapeutic agents for breast cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/virology , Endogenous Retroviruses , Gene Products, env/antagonists & inhibitors , Immunotherapy/methods , Retroviridae Proteins/antagonists & inhibitors , Adult , Aged , Animals , Antibodies, Anti-Idiotypic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Bromodeoxyuridine/metabolism , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/virology , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Feasibility Studies , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Gene Products, env/metabolism , Humans , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/analysis , Mice , Mice, Nude , Middle Aged , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Staging , Pilot Projects , Protein Array Analysis , Random Allocation , Retroviridae Proteins/metabolism , Signal Transduction , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism , Up-Regulation
16.
Genes Cancer ; 2(9): 914-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22593804

ABSTRACT

We previously observed that the HERV type K (HERV-K) envelope (env) protein was expressed in the majority of human breast tumors from a U.S. cohort of women from Texas. We also made the preliminary observation that the expression of HERV-K env transcripts was associated with markers of disease progression. In this follow-up study, env protein expression was evaluated immunohistochemically in an additional 195 paraffin-embedded breast tumors from a second U.S. patient cohort (Baltimore, Maryland) and in 110 tumors from Chinese patients. Moreover, we compared env transcript expression between fresh-frozen normal and cancerous breast tissues. We observed that while env mRNA and protein expression was undetectable in normal breast tissue and in a subset of uninvolved normal-appearing tissue adjacent to the tumor epithelium, it was overexpressed in most tumors. Furthermore, env expression was associated with breast cancer progression. In Baltimore cohort women, HERV-K tumor positivity was significantly associated with disease stage and lymph node metastasis. In Chinese women, HERV-K env positivity was significantly associated with tumor size, TNM stage, and lymph node metastases, which is consistent with the observations in the U.S. cohort. We also found that Chinese breast cancer patients with a high expression of HERV-K had a decreased overall survival compared with patients who had either a moderate or low HERV-K expression in their tumors (P = 0.049, χ(2) log rank test). In conclusion, the HERV-K env gene is expressed in the majority of breast cancers from U.S. or Chinese women but not in normal breast tissue. High expression of HERV-K env protein in breast cancer patients is associated with markers of disease progression and poor disease outcome, indicating that HERV-K env protein is a novel candidate prognostic marker for breast cancer.

17.
Cancer Res ; 68(14): 5869-77, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632641

ABSTRACT

Recent evidence indicates that human cancer cells reactivate the expression of latent human endogenous retroviral (HERV) proteins. However, the extent to which cancer patients mount de novo immune responses against expressed HERV elements is unclear. In this study, we determined the extent of HERV-K env expression in human breast cancer (BC) and whether both humoral and cell-mediated immunity against HERV-K can be found in BC patients. We found HERV-K env protein expression in 88% of BC (n = 119) but not in normal breast (n = 76) tissues. ELISA screening assays detected significant titers of anti-HERV-K env IgG in a large proportion of BC patients. T-cell responses against HERV-K were also detected in peripheral blood mononuclear cells (PBMC) from BC patients stimulated with autologous dendritic cells pulsed with HERV-K env SU antigens. These responses included induction of T-cell proliferation (P = 0.0043), IFN-gamma production measured by enzyme-linked immunospot (P < 0.0001), and multiplex cytokine secretion (P = 0.0033). Multiplex cytokine analysis found a T-helper 1 cytokine response, including interleukin (IL)-2 (P = 0.0109), IL-6 (P = 0.0396), IL-8 (P = 0.0169), and IP-10 (P = 0.0045) secretion during in vitro stimulation of BC PBMC with HERV-K antigen. We also found HERV-K-specific CTLs that were capable of lysing target cells expressing HERV-K env protein in BC patients but not in normal female controls without cancer. These findings suggest that retroviral gene products are capable of acting as tumor-associated antigens activating both T-cell and B-cell responses in BC patients.


Subject(s)
Antigens, Neoplasm/chemistry , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Endogenous Retroviruses/metabolism , Gene Expression Regulation, Neoplastic , Breast/pathology , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/cytology , Humans , Immune System/pathology , Immunoglobulin G/chemistry , Interferon-gamma/metabolism , Leukocytes, Mononuclear/cytology , Models, Biological , T-Lymphocytes/cytology
18.
Int J Cancer ; 121(10): 2346-54, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17657741

ABSTRACT

Previous studies suggest that underlying ovarian stromal cues may regulate the ovarian surface epithelium. However, little is known about the interaction between ovarian stromal cells (OSC) and ovarian surface epithelial cells (OSE) under normal physiologic and pathologic conditions, largely because of the lack of a suitable model. In the current study, the OSC obtained from a sheep were immortalized with SV-40 T/t antigen (designated IOSC) and telomerase reverse transcriptase (designated IOSCH), followed by transfection with the oncogenic allele of the human H-Ras oncogene (designated IOSChR). IOSC cells transfected with H-Ras before immortalization with telomerase were designated IOSCRH. These sheep OSCs were used in both in vitro and in vivo model systems to evaluate mechanisms by which OSCs influence ovarian tumor progression. Normal sheep OSCs were found to inhibit the growth of SKOV3 and OVCAR3 human ovarian cancer cells, but not normal sheep OSE and human OSE cells (hOSE137 cells). In contrast, IOSChR and IOSCRH cells stimulated the growth of normal sheep and human OSE cells, as well as cancer cells. These findings were confirmed by in vivo studies. Our data provide compelling support for the importance of stromal-epithelial cell interactions during tumor progression, and show for the first time that immortalized and transformed OSCs promote growth of ovarian epithelial tumors.


Subject(s)
Epithelial Cells/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Stromal Cells/metabolism , Animals , Antigens, Polyomavirus Transforming/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Disease Progression , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Nude , Sheep , Telomerase/metabolism , Xenograft Model Antitumor Assays , ras Proteins/metabolism
20.
Int J Cancer ; 120(1): 81-90, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17013901

ABSTRACT

Individual classes of human endogenous retrovirus (HERV) genes and proteins are expressed in cancer, but expression of more than one type of HERV is rare. We report here the expression of multiple HERV genes and proteins in ovarian cell lines and tissues. Expression of HERV-K env mRNA was greater in ovarian epithelial tumors than in normal ovarian tissues (N = 254). The expression of this protein on the surface and in the cytoplasm of ovarian cancer cells was confirmed using anti-HERV-K specific antibody by flow cytometric analysis. The frequency of expression of HERV-K env protein in multitissue microarrays (N = 641) was determined by immunohistochemistry and a significant correlation with tumor histotype was found. A significantly increased expression of HERV-K was observed in tumors with low malignant potential and low grade, relative to expression in normal ovarian tissues. The increase in expression of HERV-K env protein took place in a stepwise fashion in serous papillary adenocarcinoma. Interestingly, we found that other classes of HERV env mRNAs, including ERV3 and HERV-E, are expressed in the same ovarian cancer tissues that expressed HERV-K. Furthermore, anti-HERV antibodies including anti-ERV3 (30%), anti-HERV-E (40%) and anti-HERV-K (55%) were detected in patients with ovarian cancer, but not in normal female controls. HERV env proteins are frequently transcribed and translated in ovarian epithelial tumors, and multiple HERV families are detectable in ovarian cancer. HERV env proteins, and especially those expressed on the cell surface, may serve as novel tumor targets for detection, diagnosis and immunotherapy of ovarian cancer.


Subject(s)
Endogenous Retroviruses/metabolism , Gene Products, env/physiology , Membrane Proteins/metabolism , Ovarian Neoplasms/metabolism , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/virology , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/virology , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Base Sequence , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/virology , Case-Control Studies , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/virology , Endogenous Retroviruses/genetics , Endogenous Retroviruses/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Products, env/genetics , Gene Products, env/metabolism , Humans , Immunoenzyme Techniques , Membrane Proteins/genetics , Middle Aged , Molecular Sequence Data , Ovarian Neoplasms/virology , Ovary/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...