Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(25): e2400566121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38870061

ABSTRACT

Intrinsic and acquired resistance to mitogen-activated protein kinase inhibitors (MAPKi) in melanoma remains a major therapeutic challenge. Here, we show that the clinical development of resistance to MAPKi is associated with reduced tumor expression of the melanoma suppressor Autophagy and Beclin 1 Regulator 1 (AMBRA1) and that lower expression levels of AMBRA1 predict a poor response to MAPKi treatment. Functional analyses show that loss of AMBRA1 induces phenotype switching and orchestrates an extracellular signal-regulated kinase (ERK)-independent resistance mechanism by activating focal adhesion kinase 1 (FAK1). In both in vitro and in vivo settings, melanomas with low AMBRA1 expression exhibit intrinsic resistance to MAPKi therapy but higher sensitivity to FAK1 inhibition. Finally, we show that the rapid development of resistance in initially MAPKi-sensitive melanomas can be attributed to preexisting subclones characterized by low AMBRA1 expression and that cotreatment with MAPKi and FAK1 inhibitors (FAKi) effectively prevents the development of resistance in these tumors. In summary, our findings underscore the value of AMBRA1 expression for predicting melanoma response to MAPKi and supporting the therapeutic efficacy of FAKi to overcome MAPKi-induced resistance.


Subject(s)
Adaptor Proteins, Signal Transducing , Drug Resistance, Neoplasm , Melanoma , Protein Kinase Inhibitors , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Humans , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Animals , Mice , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Xenograft Model Antitumor Assays , Mitogen-Activated Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Female
3.
Front Immunol ; 14: 1240678, 2023.
Article in English | MEDLINE | ID: mdl-37662956

ABSTRACT

Background: Therapeutic cancer vaccination against mutant calreticulin (CALR) in patients with CALR-mutant (CALRmut) myeloproliferative neoplasms (MPN) induces strong T-cell responses against mutant CALR yet fails to demonstrate clinical activity. Infiltration of tumor specific T cells into the tumor microenvironment is needed to attain a clinical response to therapeutic cancer vaccination. Aim: Determine if CALRmut specific T cells isolated from vaccinated patients enrich in the bone marrow upon completion of vaccination and explore possible explanations for the lack of enrichment. Methods: CALRmut specific T cells from four of ten vaccinated patients were expanded, enriched, and analyzed by T-cell receptor sequencing (TCRSeq). The TCRs identified were used as fingerprints of CALRmut specific T cells. Bone marrow aspirations from the four patients were acquired at baseline and at the end of trial. T cells were enriched from the bone marrow aspirations and analyzed by TCRSeq to identify the presence and fraction of CALRmut specific T cells at the two different time points. In silico calculations were performed to calculate the ratio between transformed cells and effector cells in patients with CALRmut MPN. Results: The fraction of CALRmut specific T cells in the bone marrow did not increase upon completion of the vaccination trial. In general, the T cell repertoire in the bone marrow remains relatively constant through the vaccination trial. The enriched and expanded CALRmut specific T cells recognize peripheral blood autologous CALRmut cells. In silico analyses demonstrate a high imbalance in the fraction of CALRmut cells and CALRmut specific effector T-cells in peripheral blood. Conclusion: CALRmut specific T cells do not enrich in the bone marrow after therapeutic cancer peptide vaccination against mutant CALR. The specific T cells recognize autologous peripheral blood derived CALRmut cells. In silico analyses demonstrate a high imbalance between the number of transformed cells and CALRmut specific effector T-cells in the periphery. We suggest that the high burden of transformed cells in the periphery compared to the number of effector cells could impact the ability of specific T cells to enrich in the bone marrow.


Subject(s)
Cancer Vaccines , Myeloproliferative Disorders , Neoplasms , Humans , Bone Marrow , T-Lymphocytes , Calreticulin/genetics , Vaccines, Subunit , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy
5.
Cancer Lett ; 552: 215982, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36309209

ABSTRACT

DNA methyltransferase (DNMT) inhibitors are used for treatment of certain hematological malignancies and exert anti-cancer activity through diverse mechanisms, including reexpression of tumor suppressor genes and anti-viral responses triggered by expression of endogenous retroviruses. Despite advances in the pharmacokinetic properties of DNMT inhibitors, the efficacy of these drugs in solid cancers remains low. Here, we show in cell lines and clinical and experimental tumors across multiple cancer types that DNMT inhibition induces the expression of interleukin-1 (IL-1), a cytokine with proinflammatory and protumorigenic properties. Specifically, this tumor-intrinsic IL-1 expression modulates the chemokine landscape of tumors and leads to the recruitment of monocytic myeloid-derived suppressor cells to the tumor microenvironment, processes that can be blocked by IL-1 antagonists. Molecular analysis demonstrates complex patterns of IL-1 and interferon activation and crosstalk in response to DNMT inhibition, which depend on the integrity of IRF- and NF-κB-mediated antiviral pathways and may determine the outcome of DNMT-inhibitor treatment. Together, our results show that DNMT inhibitors may negatively affect the microenvironment of a large subset of tumors and suggest that co-treatment with IL-1 antagonists may be a favorable combination for these patients.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Humans , Tumor Microenvironment , Interleukin-1 , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Modification Methylases , DNA , Cell Line, Tumor
6.
Front Oncol ; 12: 998000, 2022.
Article in English | MEDLINE | ID: mdl-36276095

ABSTRACT

Cancer/testis antigens are receiving attention as targets for cancer therapy due to their germ- and cancer cell-restricted expression. However, many of these antigens are inconsistently expressed among cancer types and individual tumors. Here, we show that members of the SSX cancer/testis antigen family comprise attractive targets in the majority of melanoma patients, as SSX is expressed in more than 90% of primary melanomas and metastases and plays a critical role in metastatic progression. Accordingly, SSX silencing in melanoma mouse xenograft models reduced tumor growth and completely abolished the formation of metastatic lesions in lungs and livers. Mechanistically, we demonstrate that silencing SSX in melanoma cells induces cell cycle S-phase stalling, leading to proliferative arrest and enhanced apoptosis, which elucidates the inhibitory effect of SSX loss on tumor growth and colonization capacity. Silencing SSX further compromised the capacity of melanoma cells to migrate and invade, influencing these cells' capability to spread and colonize. Taken together, these studies highlight SSX proteins as pivotal targets in melanoma with implications for blocking metastatic progression.

7.
Eur J Pharm Sci ; 178: 106282, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35995349

ABSTRACT

Rat epidermal keratinocyte (REK) Organotypic culture (ROC) is an epidermis model that is robust and inexpensive to develop and maintain, and it has in previous studies been shown to have permeability characteristics close to those of human skin. Here, we characterize the model further by structural comparison to native human and rat skin and by investigating functional characteristics of lipid packing, polarity, and permeability coefficients. We show that the ROC model has structural similarities to native human skin and observe human skin-like permeability coefficients for testosterone and mannitol. We develop a transwell device that allows live cell microscopy of the tissue at the air-liquid interface and establish transgenic cell lines expressing different fluorescently tagged proteins. This enables showing the migration of keratinocytes during the first days after seeding, finding that keratinocytes have a higher mean migration rate in the earlier days of development. Collectively, our results show that the ROC model is an inexpensive and robust epidermis model that works reproducibly across laboratories.


Subject(s)
Keratinocytes , Skin , Animals , Humans , Keratinocytes/metabolism , Lipids/chemistry , Mannitol , Rats , Skin/metabolism , Testosterone/metabolism
8.
Genes (Basel) ; 13(1)2022 01 12.
Article in English | MEDLINE | ID: mdl-35052473

ABSTRACT

The transcription factor ZBED1 is highly expressed in trophoblast cells, but its functions in the processes of trophoblast and placental biology remain elusive. Here, we characterized the role of ZBED1 in trophoblast cell differentiation using an in vitro BeWo cell model. We demonstrate that ZBED1 is enhanced in its expression early after forskolin-induced differentiation of BeWo cells and regulates many of the genes that are differentially expressed as an effect of forskolin treatment. Specifically, genes encoding markers for the differentiation of cytotrophoblast into syncytiotrophoblast and factors essential for trophoblast cell fusion and invasion were negatively regulated by ZBED1, indicating that ZBED1 might be important for maintaining a steady pool of cytotrophoblast cells. In addition, ZBED1 affected genes involved in the regulation of trophoblast cell survival and apoptosis, in agreement with the observed increase in apoptosis upon knockdown of ZBED1 in forskolin-treated BeWo cells. In addition, genes implicated in the differentiation, recruitment, and function of innate immune cells by the placenta were affected by ZBED1, further suggesting a role for this protein in the regulation of maternal immune tolerance. In conclusion, our study implicates ZBED1 in major biological processes of placental biology.


Subject(s)
Cell Fusion , Choriocarcinoma/pathology , Gene Expression Regulation , Placenta/pathology , Transcription Factors/metabolism , Trophoblasts/pathology , Uterine Neoplasms/pathology , Cell Differentiation , Choriocarcinoma/genetics , Choriocarcinoma/metabolism , Female , Humans , Placenta/metabolism , Pregnancy , Transcription Factors/genetics , Trophoblasts/metabolism , Tumor Cells, Cultured , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
9.
Cancers (Basel) ; 13(20)2021 Oct 16.
Article in English | MEDLINE | ID: mdl-34680349

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that exhibits a high proliferation rate and early metastasis leading to a poor prognosis. HMGA2 is a DNA binding transcriptional regulator implicated in tumorigenesis. Here, we demonstrate that the HMGA2 promoter is demethylated in TNBC tumors, leading to increased expression of HMGA2 at both mRNA and protein levels. Importantly, high HMGA2 levels in TNBC tumors are correlated with poor prognosis. To detail the role of HMGA2 in TNBC development and progression, we studied its effect on core cancer phenotypes. Stable knockdown of HMGA2 in TNBC cells revealed that HMGA2 may support cell proliferation, cell migration and invasion. In addition, HMGA2 knockdown decreased cancer stem cell (CSC) features. Importantly, we found that silencing HMGA2 inhibited NF-kB signaling and lead to decreased expression of the downstream molecules IL-6 and IL-8 and reduced STAT3 pathway activation. Our results demonstrate that HMGA2 supports cancer hallmarks in TNBC and may represent a promising target for TNBC treatment.

10.
Front Oncol ; 10: 594163, 2020.
Article in English | MEDLINE | ID: mdl-33251148

ABSTRACT

Pericentromeric heterochromatin is maintained in a condensed structure by repressive epigenetic control mechanisms and perturbation of these may cause diseases. The chromosome 1q12 region harbors the largest pericentromeric heterochromatin domain in the genome and is among the most common breakpoints in both solid and hematopoietic cancers. Furthermore, the 1q arm is frequently amplified in cancer and this may support tumorigenesis by increasing the dosage of the many oncogenes of this genomic region. Recent studies have provided insight into the mechanisms leading to loss of 1q12 stability and 1q amplification and DNA hypomethylation seems to play a prominent role. This may be the result of decreased activity of DNA methyltransferases and instrumental for 1q12 destabilization or arise secondary to perturbation of other important epigenetic mechanisms that control repression of pericentromeric heterochromatin. Polycomb proteins were recently demonstrated to epigenetically reprogram demethylated 1q12 pericentromeric heterochromatin in premalignant and malignant cells to form large subnuclear structures known as polycomb bodies. This may influence the regulation and stability of 1q12 pericentromeric heterochromatin and/or the distribution of polycomb factors to support tumorigenesis. This review will discuss recent insight into the epigenetic perturbations causing the destabilization of 1q12 pericentromeric heterochromatin and its possible implications for tumor biology.

11.
Cells ; 9(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31963307

ABSTRACT

The stability of pericentromeric heterochromatin is maintained by repressive epigenetic control mechanisms, and failure to maintain this stability may cause severe diseases such as immune deficiency and cancer. Thus, deeper insight into the epigenetic regulation and deregulation of pericentromeric heterochromatin is of high priority. We and others have recently demonstrated that pericentromeric heterochromatin domains are often epigenetically reprogrammed by Polycomb proteins in premalignant and malignant cells to form large subnuclear structures known as Polycomb bodies. This may affect the regulation and stability of pericentromeric heterochromatin domains and/or the distribution of Polycomb factors to support tumorigeneses. Importantly, Polycomb bodies in cancer cells may be targeted by the cancer/testis-related SSX proteins to cause derepression and genomic instability of pericentromeric heterochromatin. This review will discuss the interplay between SSX and Polycomb factors in the repression and stability of pericentromeric heterochromatin and its possible implications for tumor biology.


Subject(s)
Centromere/metabolism , Heterochromatin/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Polycomb-Group Proteins/metabolism , Repressor Proteins/metabolism , Animals , Humans , Neoplasms/genetics , Protein Binding
12.
PLoS One ; 13(10): e0205461, 2018.
Article in English | MEDLINE | ID: mdl-30304065

ABSTRACT

Drosophila DNA replication-related element binding factor (DREF) is a transcription regulatory factor that binds the promoters of many genes involved in replication and cell proliferation and is required for normal cell cycle progression. Human DREF/zinc finger BED domain-containing protein 1 (ZBED1), an orthologue of Drosophila DREF, also has DNA binding activity, but its cellular functions remain largely uncharacterized. Herein, we show that ZBED1 is a chromatin-associated nuclear protein with a wide expression profile in human tissues from all three primary germ layers. For instance, ZBED1 was expressed in mesodermal-derived epithelial cells of the reproductive system and urinary tract, in endodermal-derived epithelial cells throughout the gastrointestinal tract, and in epidermal epithelium from the ectoderm. ZBED1 was also expressed in connective tissue and smooth muscle cells of multiple organs. To investigate whether ZBED1 is implicated in cell proliferation, similar to Drosophila DREF, we compared the tissue distribution of ZBED1 to that of the proliferation marker Ki-67. ZBED1 and Ki-67 were co-expressed in many epithelial tissues, but ZBED1 expression extended widely beyond that of Ki-67-positive cells. In other tissues, ZBED1 expression was more restricted than Ki-67 expression. These results suggest that ZBED1 is not a cell proliferation-associated factor such as Drosophila DREF, and our study adds to the cumulative understanding of the functions of ZBED1 in human cells and tissues.


Subject(s)
Ectoderm/metabolism , Endoderm/metabolism , Mesoderm/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics
13.
Mol Cancer Res ; 16(3): 417-427, 2018 03.
Article in English | MEDLINE | ID: mdl-29330295

ABSTRACT

Repression of repetitive DNA is important for maintaining genomic stability, but is often perturbed in cancer. For instance, the megabase satellite domain at chromosome 1q12 is a common site of genetic rearrangements, such as translocations and deletions. Polycomb-group proteins can be observed as large subnuclear domains called polycomb bodies, the composition and cellular function of which has remained elusive. This study demonstrates that polycomb bodies are canonical subunits of the multiprotein polycomb repressive complex 1 deposited on 1q12 pericentromeric satellite DNA, which are normally maintained as constitutive heterochromatin by other mechanisms. Furthermore, the data reveal that polycomb bodies are exclusive to premalignant and malignant cells, being absent in normal cells. For instance, polycomb bodies are present in melanocytic cells of nevi and conserved in primary and metastatic melanomas. Deposition of polycomb on the 1q12 satellite DNA in melanoma development correlated with reduced DNA methylation levels. In agreement with this, inhibition of DNA methyltransferases, with the hypomethylating agent guadecitabine (SGI-110), was sufficient for polycomb body formation on pericentromeric satellites in primary melanocytes. This suggests that polycomb bodies form in cancer cells with global DNA demethylation to control the stability of pericentromeric satellite DNA. These results reveal a novel epigenetic perturbation specific to premalignant and malignant cells that may be used as an early diagnostic marker for detection of precancerous changes and a new therapeutic entry point.Implications: Pericentromeric satellite DNA is epigenetically reprogrammed into polycomb bodies as a premalignant event with implications for transcriptional activity and genomic stability. Mol Cancer Res; 16(3); 417-27. ©2018 AACR.


Subject(s)
DNA, Satellite/genetics , Neoplasms/genetics , Precancerous Conditions/genetics , Epigenesis, Genetic , Humans , Neoplasms/pathology , Precancerous Conditions/pathology , Transfection
14.
Int J Mol Sci ; 17(6)2016 Jun 06.
Article in English | MEDLINE | ID: mdl-27275820

ABSTRACT

Genomic instability is a hallmark of human cancer and an enabling factor for the genetic alterations that drive cancer development. The processes involved in genomic instability resemble those of meiosis, where genetic material is interchanged between homologous chromosomes. In most types of human cancer, epigenetic changes, including hypomethylation of gene promoters, lead to the ectopic expression of a large number of proteins normally restricted to the germ cells of the testis. Due to the similarities between meiosis and genomic instability, it has been proposed that activation of meiotic programs may drive genomic instability in cancer cells. Some germ cell proteins with ectopic expression in cancer cells indeed seem to promote genomic instability, while others reduce polyploidy and maintain mitotic fidelity. Furthermore, oncogenic germ cell proteins may indirectly contribute to genomic instability through induction of replication stress, similar to classic oncogenes. Thus, current evidence suggests that testis germ cell proteins are implicated in cancer development by regulating genomic instability during tumorigenesis, and these proteins therefore represent promising targets for novel therapeutic strategies.


Subject(s)
Ectopic Gene Expression , Gene Expression Regulation, Neoplastic , Genomic Instability , Germ Cells/metabolism , Neoplasms/genetics , Testis/metabolism , Animals , Aurora Kinase C/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomes/genetics , Chromosomes/metabolism , DNA Replication , Humans , Male , Meiosis/genetics , Neoplasms/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polyploidy , Protein Binding , Recombination, Genetic , Stress, Physiological , Synaptonemal Complex/metabolism
15.
BMC Cancer ; 16: 7, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26747105

ABSTRACT

BACKGROUND: GAGE cancer/testis antigens are frequently expressed in various types of malignancies and represent attractive targets for immunotherapy, however their role in cancer initiation and progression has remained elusive. GAGE proteins are expressed in normal cells during early development with migratory and invasive properties and were found to be upregulated in cancer cells with metastasizing potential in a gastric cancer model. METHODS: We have addressed the direct role of GAGE proteins in supporting metastasis using an isogenic metastasis model of human cancer, consisting of 4 isogenic cell lines, which are equally tumorigenic in immunodeficient mice, but differ with their ability to generate metastases in the lungs and lymph nodes. RESULTS: Although GAGE proteins were strongly upregulated in the highly metastatic clone (CL16) compared to non-metastatic (NM2C5), weakly metastatic (M4A4) and moderately metastatic clones (LM3), stable downregulation of GAGE expression did not affect the ability of CL16 cells to establish primary tumors and form metastasis in the lungs of immunodeficient mice. CONCLUSIONS: These results suggest that GAGE proteins per se do not support metastasis and that further studies are needed to clarify the contribution of GAGE proteins to the metastatic potential of different types of cancer cells.


Subject(s)
Antigens, Neoplasm/biosynthesis , Carcinogenesis/genetics , Lymphatic Metastasis/genetics , Neoplasms/genetics , Animals , Antigens, Neoplasm/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis/pathology , Mice , Multigene Family/genetics , Neoplasm Invasiveness/genetics , Neoplasms/pathology , Prognosis , Xenograft Model Antitumor Assays
16.
Nucleic Acids Res ; 42(18): 11433-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25249625

ABSTRACT

Polycomb group (PcG) complexes regulate cellular identity through epigenetic programming of chromatin. Here, we show that SSX2, a germline-specific protein ectopically expressed in melanoma and other types of human cancers, is a chromatin-associated protein that antagonizes BMI1 and EZH2 PcG body formation and derepresses PcG target genes. SSX2 further negatively regulates the level of the PcG-associated histone mark H3K27me3 in melanoma cells, and there is a clear inverse correlation between SSX2/3 expression and H3K27me3 in spermatogenesis. However, SSX2 does not affect the overall composition and stability of PcG complexes, and there is no direct concordance between SSX2 and BMI1/H3K27me3 presence at regulated genes. This suggests that SSX2 antagonizes PcG function through an indirect mechanism, such as modulation of chromatin structure. SSX2 binds double-stranded DNA in a sequence non-specific manner in agreement with the observed widespread association with chromatin. Our results implicate SSX2 in regulation of chromatin structure and function.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Polycomb-Group Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/chemistry , DNA/metabolism , Enhancer of Zeste Homolog 2 Protein , Histones/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Neoplasm Proteins/physiology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Repressor Proteins/physiology , Spermatogenesis
17.
Expert Opin Biol Ther ; 10(7): 1061-75, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20420535

ABSTRACT

IMPORTANCE OF THE FIELD: Immunotherapy holds great potential for disseminated cancer, and cancer-germline (CG) antigens are among the most promising tumor targets. They are widely expressed in different cancer types and are essentially tumor-specific, since their expression in normal tissues is largely restricted to immune-privileged sites. Although the therapeutic potential of these antigens may be compromised by their highly heterogeneous expression in many tumors and low frequency in some cancers, recent developments suggest that tumor-cell-selective enhancement of CG antigen gene expression can be achieved using epigenetic modifiers. AREAS COVERED IN THIS REVIEW: We provide an overview of the potential of CG antigens as targets for cancer immunotherapy, including advantages and disadvantages. We also discuss the current state of development of CG antigen vaccines, and the potential synergistic effect of combining CG antigen immunotherapeutic strategies with epigenetic modifiers. WHAT THE READER WILL GAIN: The reader will gain an overview of the past, present and future role of CG antigens in cancer immunotherapy. TAKE HOME MESSAGE: Chemoimmunotherapy using epigenetic drugs and CG antigen vaccines may be a useful approach for treating cancer.


Subject(s)
Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Epigenesis, Genetic/drug effects , Genetic Therapy/methods , Immunotherapy/methods , Neoplasms/therapy , Animals , Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Cancer Vaccines/immunology , DNA Modification Methylases/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...