Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
Add more filters

Country/Region as subject
Publication year range
1.
CA Cancer J Clin ; 73(4): 376-424, 2023.
Article in English | MEDLINE | ID: mdl-36512337

ABSTRACT

Cancer development is driven by the accumulation of alterations affecting the structure and function of the genome. Whereas genetic changes disrupt the DNA sequence, epigenetic alterations contribute to the acquisition of hallmark tumor capabilities by regulating gene expression programs that promote tumorigenesis. Shifts in DNA methylation and histone mark patterns, the two main epigenetic modifications, orchestrate tumor progression and metastasis. These cancer-specific events have been exploited as useful tools for diagnosis, monitoring, and treatment choice to aid clinical decision making. Moreover, the reversibility of epigenetic modifications, in contrast to the irreversibility of genetic changes, has made the epigenetic machinery an attractive target for drug development. This review summarizes the most advanced applications of epigenetic biomarkers and epigenetic drugs in the clinical setting, highlighting commercially available DNA methylation-based assays and epigenetic drugs already approved by the US Food and Drug Administration.


Subject(s)
Epigenesis, Genetic , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/drug therapy , DNA Methylation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology
2.
Semin Cancer Biol ; 92: 84-101, 2023 07.
Article in English | MEDLINE | ID: mdl-37003397

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous disease with a genetic, epigenetic, and transcriptional etiology mainly presenting somatic and germline abnormalities. AML incidence rises with age but can also occur during childhood. Pediatric AML (pAML) accounts for 15-20% of all pediatric leukemias and differs considerably from adult AML. Next-generation sequencing technologies have enabled the research community to "paint" the genomic and epigenomic landscape in order to identify pathology-associated mutations and other prognostic biomarkers in pAML. Although current treatments have improved the prognosis for pAML, chemoresistance, recurrence, and refractory disease remain major challenges. In particular, pAML relapse is commonly caused by leukemia stem cells that resist therapy. Marked patient-to-patient heterogeneity is likely the primary reason why the same treatment is successful for some patients but, at best, only partially effective for others. Accumulating evidence indicates that patient-specific clonal composition impinges significantly on cellular processes, such as gene regulation and metabolism. Although our understanding of metabolism in pAML is still in its infancy, greater insights into these processes and their (epigenetic) modulation may pave the way toward novel treatment options. In this review, we summarize current knowledge on the function of genetic and epigenetic (mis)regulation in pAML, including metabolic features observed in the disease. Specifically, we describe how (epi)genetic machinery can affect chromatin status during hematopoiesis, leading to an altered metabolic profile, and focus on the potential value of targeting epigenetic abnormalities in precision and combination therapy for pAML. We also discuss the possibility of using alternative epidrug-based therapeutic approaches that are already in clinical practice, either alone as adjuvant treatments and/or in combination with other drugs.


Subject(s)
Epigenomics , Leukemia, Myeloid, Acute , Humans , Child , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Prognosis , Mutation
3.
Int J Cancer ; 154(6): 1029-1042, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-37947765

ABSTRACT

Non-small cell lung cancer (NSCLC) patients are often elderly or unfit and thus cannot tolerate standard aggressive therapy regimes. In our study, we test the efficacy of the DNA-hypomethylating agent decitabine (DAC) in combination with all-trans retinoic acid (ATRA), which has been shown to possess little systemic adverse effects. Screening a broad panel of 56 NSCLC cell lines uncovered a decrease in cell viability after the combination treatment in 77% of the cell lines. Transcriptomics, proteomics, proliferation and migration profiling revealed that fast proliferating and slowly migrating cell lines were more sensitive to the drug combination. The comparison of mutational profiles found oncogenic KRAS mutations only in sensitive cells. Additionally, different cell lines showed a heterogeneous gene expression response to the treatment pointing to diverse mechanisms of action. Silencing KRAS, RIG-I or RARB partially reversed the sensitivity of KRAS-mutant NCI-H460 cells. To study resistance, we generated two NCI-H460 cell populations resistant to ATRA and DAC, which migrated faster and proliferated slower than the parental sensitive cells and showed signs of senescence. In summary, this comprehensive dataset uncovers a broad sensitivity of NSCLC cells to the combinatorial treatment with DAC and ATRA and indicates that migration and proliferation capacities correlate with and could thus serve as determinants for drug sensitivity in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Tretinoin/pharmacology , Tretinoin/therapeutic use , Decitabine/pharmacology , Decitabine/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Cell Line, Tumor , Cell Proliferation
4.
Eur J Immunol ; 53(4): e2048980, 2023 04.
Article in English | MEDLINE | ID: mdl-36647268

ABSTRACT

Epigenetics, as a discipline that aims to explain the differential expression of phenotypes arising from the same gene sequence and the heritability of epigenetic expression, has received much attention in medicine. Epigenetic mechanisms are constantly being discovered, including DNA methylation, histone modifications, noncoding RNAs and m6A. The immune system mainly achieves an immune response through the differentiation and functional expression of immune cells, in which epigenetic modification will have an important impact. Because of immune infiltration in the tumor microenvironment, immunotherapy has become a research hotspot in tumor therapy. Epigenetics plays an important role in autoimmune diseases and cancers through immunology. An increasing number of drugs targeting epigenetic mechanisms, such as DNA methyltransferase inhibitors, histone deacetylase inhibitors, and drug combinations, are being evaluated in clinical trials for the treatment of various cancers (including leukemia and osteosarcoma) and autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, systemic sclerosis). This review summarizes the progress of epigenetic regulation for cancers and autoimmune diseases to date, shedding light on potential therapeutic strategies.


Subject(s)
Autoimmune Diseases , Neoplasms , Humans , Autoimmunity/genetics , Epigenesis, Genetic , Translational Science, Biomedical , DNA Methylation , Neoplasms/genetics , Neoplasms/therapy , Tumor Microenvironment
5.
Ann Hematol ; 103(5): 1435-1454, 2024 May.
Article in English | MEDLINE | ID: mdl-37581713

ABSTRACT

Lymphomas, complex and heterogeneous malignant tumors, originate from the lymphopoietic system. These tumors are notorious for their high recurrence rates and resistance to treatment, which leads to poor prognoses. As ongoing research has shown, epigenetic modifications like DNA methylation, histone modifications, non-coding RNA regulation, and RNA modifications play crucial roles in lymphoma pathogenesis. Epigenetic modification-targeting drugs have exhibited therapeutic efficacy and tolerability in both monotherapy and combination lymphoma therapy. This review discusses pathogenic mechanisms and potential epigenetic therapeutic targets in common lymphomas, offering new avenues for lymphoma diagnosis and treatment. We also discuss the shortcomings of current lymphoma treatments, while suggesting potential areas for future research, in order to improve the prediction and prognosis of lymphoma.


Subject(s)
Lymphoma , Neoplasms , Humans , DNA Methylation , Epigenesis, Genetic , Neoplasms/drug therapy , Lymphoma/drug therapy , Lymphoma/genetics , Prognosis
6.
J Allergy Clin Immunol ; 151(6): 1622-1633.e10, 2023 06.
Article in English | MEDLINE | ID: mdl-37086924

ABSTRACT

BACKGROUND: X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus infection and N-linked glycosylation defect (XMEN) disease is a rare combined immunodeficiency caused by loss-of-function mutations in the magnesium transporter 1 (MAGT1) gene. MAGT1 deficiency impairs magnesium transport and the N-linked glycosylation of a panel of proteins, which subsequently abolishes the expression of key immune receptors such as natural killer group 2, member D (aka NKG2D). These effects induce immune system abnormalities, chronic Epstein-Barr virus infection, and neoplasia. Recent research shows that MAGT1 and tumor candidate suppressor 3 (TUSC3) share high sequence and functional similarity. OBJECTIVE: We sought to investigate the feasibility of activating TUSC3 expression to provide a potential therapeutic strategy for XMEN disease. METHODS: The expression profiles of MAGT1 and TUSC3 were analyzed using multiple databases, real-time quantitative PCR, and Western blot. The effects of decitabine and panobinostat on the regulation of TUSC3 expression were explored in both MAGT1 knockout (KO)/patient-derived lymphocytes and MAGT1 KO hepatocytes. RESULTS: Although TUSC3 is widely expressed, it is undetectable specifically in the immune system and liver, consistent with the main diseased tissues in patients with XMEN disease. CRISPR/Cas9-mediated KO of MAGT1 in the NKL cell line successfully mimicked the phenotypes of XMEN patient-derived lymphocytes, and exogenous expression of TUSC3 rescued the deficiencies in KO NKL cells. Using this in vitro model, we identified 2 epigenetic drugs, decitabine and panobinostat, by screening. Combination treatment using these 2 drugs significantly upregulated TUSC3 expression and rescued the immune and liver abnormalities. CONCLUSIONS: Epigenetic activation of TUSC3 expression constitutes an effective therapeutic strategy for XMEN disease.


Subject(s)
Epstein-Barr Virus Infections , Magnesium , Humans , Magnesium/metabolism , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human , Decitabine , Panobinostat , Epigenesis, Genetic
7.
Semin Cancer Biol ; 83: 152-165, 2022 08.
Article in English | MEDLINE | ID: mdl-32858230

ABSTRACT

The global burden of breast cancer (BC) is increasing significantly. This trend is caused by several factors such as late diagnosis, limited treatment options for certain BC subtypes, drug resistance which all lead to poor clinical outcomes. Recent research has reported the role of epigenetic alterations in the mechanism of BC pathogenesis and its hallmarks include drug resistance and stemness features. The understanding of these modifications and their significance in the management of BC carcinogenesis is challenging and requires further attention. Nevertheless, it promises to provide novel insight needed for utilizing these alterations as potential diagnostic, prognostic markers, predict treatment efficacy, as well as therapeutic agents. This highlights the importance of continuing research development to further advance the existing knowledge on epigenetics and BC carcinogenesis to overcome the current challenges. Hence, this review aims to shed light and discuss the current state of epigenetics research in the diagnosis and management of BC.


Subject(s)
Breast Neoplasms , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinogenesis , DNA Methylation , Epigenesis, Genetic , Epigenomics , Female , Humans
8.
Semin Cancer Biol ; 83: 503-522, 2022 08.
Article in English | MEDLINE | ID: mdl-33309850

ABSTRACT

Epigenetic modifications are heritable yet reversible, essential for normal physiological functions and biological development. Aberrant epigenetic modifications, including DNA methylation, histone modification, and non-coding RNA (ncRNA)-mediated gene regulation play a crucial role in cancer progression. In cellular reprogramming, irregular epigenomic modulations alter cell signaling pathways and the expression of tumor suppressor genes and oncogenes, resulting in cancer growth and metastasis. Therefore, alteration of epigenetic-status in cancer cells can be used as a potential target for cancer therapy. Several synthetic epigenetic inhibitors (epi-drugs) and natural epigenetic modulatory bioactives (epi-diets) have been shown to have the potential to alter the aberrant epigenetic status and inhibit cancer progression. Further, the use of combinatorial approaches with epigenetic drugs and diets has brought promising outcomes in cancer prevention and therapy. In this article, we have summarized the epigenetic modulatory activities of epi-drugs, epi-diets, and their combination against various cancers. We have also compiled the preclinical and clinical status of these epigenetic modulators in different cancers.


Subject(s)
Epigenomics , Neoplasms , DNA Methylation , Diet , Epigenesis, Genetic , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/prevention & control
9.
Subcell Biochem ; 100: 503-521, 2022.
Article in English | MEDLINE | ID: mdl-36301504

ABSTRACT

Origin of cancer is strongly related to the unusual epigenetic regulation of gene function as indicated by recent reports. The covalent modifications to DNA or histones without affecting genomes that finally lead to phenotypical changes in cells or organisms are referred as "Epigenetics." The possibility to reprogram the epigenetics in the cancer epigenome is the most important target for cancer treatment and drug resistance. The development of epigenetic drugs holds a great potential for the current cancer therapeutic approaches. Nevertheless, targeting cancer epigenetic pathways is still exciting due to the lack of selective and effective small molecule compounds or drug molecules. Therefore, the current book chapter highlights epigenetic pathways for cancer and potential small molecule inhibitors and epidrugs targeting DNA methyltransferase, histone modification, and more new therapies with nanomaterials and imaging to improve the effectiveness of cancer treatment. The structural aspects on discovery of novel small molecules or drugs targeting epigenetic pathways in cancer exploration as promising strategies will be also discussed.


Subject(s)
Epigenesis, Genetic , Neoplasms , Humans , DNA Methylation , Neoplasms/drug therapy , Neoplasms/genetics , Histones/genetics , Histones/metabolism , DNA/metabolism
10.
Subcell Biochem ; 100: 427-472, 2022.
Article in English | MEDLINE | ID: mdl-36301502

ABSTRACT

Cancer begins due to uncontrolled cell division. Cancer cells are insensitive to the signals that control normal cell proliferation. This uncontrolled cell division is due to the accumulation of abnormalities in different factors associated with the cell division, including different cyclins, cell cycle checkpoint inhibitors, and cellular signaling. Cellular signaling pathways are aberrantly activated in cancer mainly due to epigenetic regulation and post-translational regulation. In this chapter, the role of epigenetic regulation in aberrant activation of PI3K/AKT, Ras, Wnt, Hedgehog, Notch, JAK/STAT, and mTOR signaling pathways in cancer progression is discussed. The role of epigenetic regulators in controlling the upstream regulatory proteins and downstream effector proteins responsible for abnormal cellular signaling-mediated cancer progression is covered in this chapter. Similarly, the role of signaling pathways in controlling epigenetic gene regulation-mediated cancer progression is also discussed. We have tried to ascertain the current status of potential epigenetic drugs targeting several epigenetic regulators to prevent different cancers.


Subject(s)
Epigenesis, Genetic , Neoplasms , Signal Transduction , Humans , Neoplasms/metabolism , Gene Expression Regulation, Neoplastic
11.
J Enzyme Inhib Med Chem ; 38(1): 2163242, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36629431

ABSTRACT

Androgen deprivation therapy (ADT) is a common treatment for recurrent prostate cancer (PC). However, after a certain period of responsiveness, ADT resistance occurs virtually in all patients and the disease progresses to lethal metastatic castration-resistant prostate cancer (mCRPC). Aberrant expression and function of the epigenetic modifiers EZH2 and BET over activates c-myc, an oncogenic transcription factor critically contributing to mCRPC. In the present work, we tested, for the first time, the combination of an EZH2 inhibitor with a BET inhibitor in metastatic PC cells. The combination outperformed single drugs in inhibiting cell viability, cell proliferation and clonogenic ability, and concomitantly reduced both c-myc and NF-kB expression. Although these promising results will warrant further in vivo validation, they represent the first step to establishing the rationale that the proposed combination might be suitable for mCRPC treatment, by exploiting molecular targets different from androgen receptor.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Transcription Factors , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/metabolism
12.
Int J Mol Sci ; 24(14)2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37511510

ABSTRACT

Amino acid metabolism has been implicated in tumorigenesis and tumor progression. Alterations in intracellular and extracellular metabolites associated with metabolic reprogramming in cancer have profound effects on gene expression, cell differentiation, and tumor immune microenvironment. However, the prognostic significance of amino acid metabolism in head and neck cancer remains to be further investigated. In this study, we identified 98 differentially expressed genes related to amino acid metabolism in head and neck cancer in The Cancer Genome Atlas. Using batch univariate Cox regression and Lasso regression, we extracted nine amino acid metabolism-related genes. Based on that, we developed the amino acid metabolism index. The prognostic value of this index was validated in two Gene Expression Omnibus cohorts. The results show that this model can help predict tumor recurrence and prognosis. The infiltration of immune cells in the tumor microenvironment was analyzed, and it was discovered that the high index is associated with an immunosuppressive microenvironment. In addition, this study demonstrated the impact of the amino acid metabolism index on clinical indicators, survival of patients with head and neck cancer, and the prediction of treatment response to immune checkpoint inhibitors. We conducted several cell experiments and demonstrated that epigenetic drugs could affect the index and enhance tumor immunity. In conclusion, our study demonstrates that the index not only has important prognostic value in head and neck cancer patients but also facilitates patient stratification for immunotherapy.


Subject(s)
Head and Neck Neoplasms , Neoplasm Recurrence, Local , Humans , Prognosis , Head and Neck Neoplasms/genetics , Carcinogenesis , Immunosuppressive Agents , Amino Acids , Tumor Microenvironment/genetics
13.
Int J Mol Sci ; 24(13)2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37446319

ABSTRACT

The SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin-remodeling complex is one of the most remarkably altered epigenetic regulators in cancer. Pathogenic mutations in genes encoding SWI/SNF-related proteins have been recently described in many solid tumors, including rare and aggressive malignancies with rhabdoid features with no standard therapies in advanced or metastatic settings. In recent years, clinical trials with targeted drugs aimed at restoring its function have shown discouraging results. However, preclinical data have found an association between these epigenetic alterations and response to immune therapy. Thus, the rationale for immunotherapy strategies in SWI/SNF complex alteration-related tumors is strong. Here, we review the SWI/SNF complex and how its dysfunction drives the oncogenesis of rhabdoid tumors and the proposed strategies to revert this alteration and promising novel therapeutic approaches, including immune checkpoint inhibition and adoptive cell therapy.


Subject(s)
DNA-Binding Proteins , Rhabdoid Tumor , Humans , DNA-Binding Proteins/genetics , Immunotherapy, Adoptive , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Rhabdoid Tumor/genetics , Rhabdoid Tumor/therapy , Rhabdoid Tumor/pathology
14.
Molecules ; 28(4)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36838987

ABSTRACT

A comprehensive understanding of the mechanisms involved in epigenetic changes in gene expression is essential to the clinical management of diseases linked to the SMYD family of lysine methyltransferases. The five known SMYD enzymes catalyze the transfer of donor methyl groups from S-adenosylmethionine (SAM) to specific lysines on histones and non-histone substrates. SMYDs family members have distinct tissue distributions and tissue-specific functions, including regulation of development, cell differentiation, and embryogenesis. Diseases associated with SMYDs include the repressed transcription of SMYD1 genes needed for the formation of ion channels in the heart leading to heart failure, SMYD2 overexpression in esophageal squamous cell carcinoma (ESCC) or p53-related cancers, and poor prognosis associated with SMYD3 overexpression in more than 14 types of cancer including breast cancer, colon cancer, prostate cancer, lung cancer, and pancreatic cancer. Given the importance of epigenetics in various pathologies, the development of epigenetic inhibitors has attracted considerable attention from the pharmaceutical industry. The pharmacologic development of the inhibitors involves the identification of molecules regulating both functional SMYD SET (Suppressor of variegation, Enhancer of Zeste, Trithorax) and MYND (Myeloid-Nervy-DEAF1) domains, a process facilitated by available X-ray structures for SMYD1, SMYD2, and SMYD3. Important leads for potential pharmaceutical agents have been reported for SMYD2 and SMYD3 enzymes, and six epigenetic inhibitors have been developed for drugs used to treat myelodysplastic syndrome (Vidaza, Dacogen), cutaneous T-cell lymphoma (Zoinza, Isrodax), and peripheral T-cell lymphoma (Beleodag, Epidaza). The recently demonstrated reversal of SMYD histone methylation suggests that reversing the epigenetic effects of SMYDs in cancerous tissues may be a desirable target for pharmacological development.


Subject(s)
Epigenesis, Genetic , Histone-Lysine N-Methyltransferase , Humans , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Transcription Factors/metabolism , Neoplasms/genetics
15.
Neuroendocrinology ; 112(6): 580-594, 2022.
Article in English | MEDLINE | ID: mdl-34348348

ABSTRACT

INTRODUCTION: Neuroendocrine transdifferentiation (NED) of prostate cancer (PC) cells is associated with the development of resistance to antiandrogen therapy and poor prognosis in patients with castration-resistant PC (CRPC). Many of the molecular events, involved in NED, appear to be mediated by epigenetic mechanisms. In this study, we evaluated the antitumor activity and epigenetic modulation of 2 epigenetic drugs, such as the demethylating agent 5-aza-2'-deoxycytidine (AZA) and the methyl donor S-adenosylmethionine (SAM), in 2 human CRPC cell lines with NED (DU-145 and PC-3). METHODS: The effects of AZA and SAM on cell viability, cell cycle, apoptosis, migration, and genome-wide DNA methylation profiling have been evaluated. RESULTS: Both drugs showed a prominent antitumor activity in DU-145 and PC-3 cells, through perturbation of cell cycle progression, induction of apoptosis, and inhibition of cell migration. AZA and SAM reversed NED in DU-145 and PC-3, respectively. Moreover, AZA treatment modified DNA methylation pattern in DU-145 cells, sustaining a pervasive hypomethylation of the genome, with a relevant effect on several pathways involved in the regulation of cell proliferation, apoptosis, and cell migration, in particular Wnt/ß-catenin. CONCLUSIONS: A relevant antitumor activity of these epigenetic drugs on CRPC cell lines with NED opens a new scenario in the therapy of this lethal variant of PC.


Subject(s)
Epigenesis, Genetic , Prostatic Neoplasms, Castration-Resistant , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
16.
Int J Cancer ; 149(10): 1732-1743, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34213777

ABSTRACT

Ovarian cancer therapy has remained fundamentally unchanged for 50 years, with surgery and chemotherapy still the frontline treatments. Typically asymptomatic until advanced stages, ovarian cancer is known as "the silent killer." Consequently, it has one of the worst 5-year survival rates, as low as 30%. The most frequent driver mutations are found in well-defined tumor suppressors, such as p53 and BRCA1/2. In recent years, it has become clear that, like the majority of other cancers, many epigenetic regulators are altered in ovarian cancer, including EZH2, SMARCA2/4 and ARID1A. Disruption of epigenetic regulators often leads to loss of transcriptional control, aberrant cell fate trajectories and disruption of senescence, apoptotic and proliferation pathways. These mitotically inherited epigenetic alterations are particularly promising targets for therapy as they are largely reversible. Consequently, many drugs targeting chromatin modifiers and other epigenetic regulators are at various stages of clinical trials for other cancers. Understanding the mechanisms by which ovarian cancer-specific epigenetic processes are disrupted in patients can allow for informed targeting of epigenetic pathways tailored for each patient. In recent years, there have been groundbreaking new advances in disease modeling through ovarian cancer organoids; these models, alongside single-cell transcriptomic and epigenomic technologies, allow the elucidation of the epigenetic pathways deregulated in ovarian cancer. As a result, ovarian cancer therapy may finally be ready to advance to next-generation treatments. Here, we review the major developments in ovarian cancer, including genetics, model systems and technologies available for their study and the implications of applying epigenetic therapies to ovarian cancer.


Subject(s)
Epigenesis, Genetic , Epigenomics/methods , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Humans , Nuclear Proteins/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Protein-Arginine N-Methyltransferases/genetics , Transcription Factors/genetics
17.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799504

ABSTRACT

Garcinol extracted from Garcinia indica fruit peel and leaves is a polyisoprenylated benzophenone. In traditional medicine it was used for its antioxidant and anti-inflammatory properties. Several studies have shown anti-cancer properties of garcinol in cancer cell lines and experimental animal models. Garcinol action in cancer cells is based on its antioxidant and anti-inflammatory properties, but also on its potency to inhibit histone acetyltransferases (HATs). Recent studies indicate that garcinol may also deregulate expression of miRNAs involved in tumour development and progression. This paper focuses on the latest research concerning garcinol as a HAT inhibitor and miRNA deregulator in the development and progression of various cancers. Garcinol may be considered as a candidate for next generation epigenetic drugs, but further studies are needed to establish the precise toxicity, dosages, routes of administration, and safety for patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Epigenesis, Genetic/drug effects , Histone Acetyltransferases/genetics , MicroRNAs/genetics , Neoplasms/drug therapy , Terpenes/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Garcinia/chemistry , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Plant Extracts/chemistry
18.
Rinsho Ketsueki ; 62(9): 1367-1381, 2021.
Article in Japanese | MEDLINE | ID: mdl-34615796

ABSTRACT

Epigenetics is the study that involves understanding of the DNA sequence-independent mechanism of transcriptional regulation. The epigenetic regulation of gene expression is exerted via the alteration of chromatin structures through covalent modifications of core histone tails and methylation of CpG dinucleotides. In general, histone acetylation and DNA methylation are associated with transcriptional activation and repression, respectively. Histone methylation offers an additional layer for transcriptional regulation. Epigenetic abnormalities underlie the development of various hematological malignancies; for example, recurrent mutations of the DNA methyltransferase DNMT3A or DNA demethylase TET2 transform hematopoietic stem cells into preleukemic stem cells. Consequently, preleukemic stem cells give rise to T-cell lymphomas, such as angioimmunoblastic T-cell lymphoma and T-cell lymphoblastic lymphoma. Epigenetic alterations could be ideal therapeutic targets; indeed, HDAC inhibitors and DNA demethylating agents have already been used for the treatment of peripheral T-cell lymphomas. It is anticipated that more number of epigenetic drugs would be developed for clinical application in the near future.


Subject(s)
Epigenesis, Genetic , Lymphoma, Non-Hodgkin , Humans , Lymphoma, Non-Hodgkin/genetics
19.
Cancer Immunol Immunother ; 69(11): 2169-2178, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32648166

ABSTRACT

T-cell receptor (TCR)- and chimeric antigen receptor (CAR)-based adoptive cell transfer (ACT) has shown promising results in hematological malignancies, but remains immature in solid cancers. The challenges associated with identification of tumor-specific targets, the heterogenic antigen expression, limited T-cell trafficking to tumor sites and the hostile tumor microenvironment (TME), are all factors contributing to the limited efficacy of ACT therapies against solid tumors. Epigenetic priming of tumor cells and the microenvironment may be a way of overcoming these obstacles and improving the clinical efficacy of adoptive T-cell therapies in the future. Here, we review the current literature and suggest combining epigenetic modulators and ACT strategies as a way of augmenting the efficacy of TCR- and CAR-engineered T cells against solid tumors.


Subject(s)
Combined Modality Therapy/methods , Epigenesis, Genetic , Immunotherapy, Adoptive/methods , Neoplasms , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/immunology , DNA Modification Methylases/antagonists & inhibitors , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
20.
Rev Endocr Metab Disord ; 21(4): 509-519, 2020 12.
Article in English | MEDLINE | ID: mdl-32514818

ABSTRACT

Epigenetic alterations in cancer play a variety of roles. Aberrant DNA methylation, as one of the epigenetic mechanisms, has been widely studied in both tumor and liquid biopsies and provide a useful bench mark for treatment response in cancer. Recently, several studies have reported an association between the type of diet and epigenetic modifications. Whereby there is a growing interest in finding the "anti-cancer diet formula", if such a thing exists. In this sense, ketogenic diets (KD) have reported potentially beneficial effects, which were able to prevent malignancies and decrease tumor growth. Some studies have even shown increased survival in cancer patients, reduced side effects of cytotoxic treatments, and intensified efficacy of cancer therapies. Although the biological mechanisms of KD are not well understood, it has been reported that KD may affect DNA methylation by modulating the expression of crucial genes involved in tumor survival and proliferation. However, there are many considerations to take into account to use ketotherapy in cancer, such as epigenetic mark, type of cancer, immunological and metabolic state or microbiota profile. In this review, we argue about ketotherapy as a potential strategy to consider as coadjuvant of cancer therapy. We will focus on mainly epigenetic mechanisms and dietary approach that could be included in the current clinical practice guidelines.


Subject(s)
Circadian Rhythm , DNA Methylation , Diet, Ketogenic , Epigenesis, Genetic , Neoplasms/diet therapy , Circadian Rhythm/physiology , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Humans , Neoplasms/immunology , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL