ijms-logo

Journal Browser

Journal Browser

Pleiotropic Action of Selenium in the Prevention and Treatment of Cancer, and Related Diseases

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Bioactives and Nutraceuticals".

Deadline for manuscript submissions: closed (30 November 2018) | Viewed by 43800

Printed Edition Available!
A printed edition of this Special Issue is available here.

Special Issue Editor

Department of Cancer Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
Interests: HIFs; TGF-b; oncogenic microRNA-210 druggable targets; mechanism-based biologic and cytotoxic therapies; COVID-19 and selenium
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Gene cloning and sequence has provided the opportunity to identify and characterize the functional role of biomarkers expressed in and on tumor cells and the surrounding microenvironment. Molecular and immunologic heterogeneity of cells in the tumor microenvironment contribute to instability, enhanced angiogenesis and drug resistance of the tumor cell. Since tumor cells are the ultimate therapeutic targets for drugs and therapy development, the tumor microenvironment that regulates the growth and the delivery of effective drug concentrations to tumor cells is the gatekeeper. Thus, to have a significant impact on overall survival and cure of patients with advanced cancer, the stabilization of the tumor microenvironment should be the initial treatment, followed by treatment that targets and kills tumor cells.

Antiangeogenic therapies hold considerable promise in the treatment of a subset of cancer patients, and is reported to have significant impacts on the stabilization of the tumor microenvironment. More recently, selenium-containing molecules, such as Se-metylselenocysteine, seleno-L-methionine, and selenized yeast, among others, have been shown to target and modulate biomarkers associated with tumor cells and the tumor microenvironment. The effects are selenium type, dose, and schedule-dependent. The pleiotropic actions of selenium are necessary for tumor cells sensitization, and synergy with mechanism-based combinations. This Special Issue is devoted to highlighting evidence for the potential role of specific types, doses, and schedules of selenium alone and in combination with mechanism-based biologic and cytotoxic therapies for the prevention and treatment of cancer and related diseases. The collection of contributions should provide a comprehensive overview of the pharmacology, metabolism, and delineation of the pleiotropic action of different types of selenium molecules, relevant to the use of selenium as a potential modulator of the therapeutic efficacy and toxicity of biologic and cytotoxic therapies for cancer and related diseases. The pleiotropic action of specific types of selenium, doses and schedule, as selective and efficacious modulator of genetic, immunologic, and epigenetic biomarkers, should stimulate expanded preclinical research that could ultimately impact on the development of new and novel approaches for the treatment of cancer.

Prof. Dr. Youcef M. Rustum
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Selenium-containing molecules
  • Immunologic and epigenetic biomarkers
  • Tumor microenvironment
  • Mechanism-based biologic and cytotoxic therapies
  • Molecular, epigeneticand immunological targets (replacement)
  • Prevention and therapeutic potential
  • Pharmacology and metabolism
  • HIFs (Hypoxia-inducible factor), VEGF (Vascular endothelial growth factor)
  • VEGFR, PD-L1 and miRNAs targets

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

17 pages, 3804 KiB  
Article
Identification of a Novel Quinoxaline-Isoselenourea Targeting the STAT3 Pathway as a Potential Melanoma Therapeutic
by Verónica Alcolea, Deepkamal N. Karelia, Manoj K. Pandey, Daniel Plano, Parvesh Singh, Juan Antonio Palop, Shantu Amin, Carmen Sanmartín and Arun K. Sharma
Int. J. Mol. Sci. 2019, 20(3), 521; https://doi.org/10.3390/ijms20030521 - 26 Jan 2019
Cited by 11 | Viewed by 3646
Abstract
The prognosis for patients with metastatic melanoma remains very poor. Constitutive signal transducer and activator of transcription 3 (STAT3) activation has been correlated to metastasis, poor patient survival, larger tumor size, and acquired resistance against vemurafenib (PLX-4032), suggesting its potential as a molecular [...] Read more.
The prognosis for patients with metastatic melanoma remains very poor. Constitutive signal transducer and activator of transcription 3 (STAT3) activation has been correlated to metastasis, poor patient survival, larger tumor size, and acquired resistance against vemurafenib (PLX-4032), suggesting its potential as a molecular target. We recently designed a series of isoseleno- and isothio-urea derivatives of several biologically active heterocyclic scaffolds. The cytotoxic effects of lead isoseleno- and isothio-urea derivatives (compounds 1 and 3) were studied in a panel of five melanoma cell lines, including B-RAFV600E-mutant and wild-type (WT) cells. Compound 1 (IC50 range 0.8–3.8 µM) showed lower IC50 values than compound 3 (IC50 range 8.1–38.7 µM) and the mutant B-RAF specific inhibitor PLX-4032 (IC50 ranging from 0.4 to >50 µM), especially at a short treatment time (24 h). These effects were long-lasting, since melanoma cells did not recover their proliferative potential after 14 days of treatment. In addition, we confirmed that compound 1 induced cell death by apoptosis using Live-and-Dead, Annexin V, and Caspase3/7 apoptosis assays. Furthermore, compound 1 reduced the protein levels of STAT3 and its phosphorylation, as well as decreased the expression of STAT3-regulated genes involved in metastasis and survival, such as survivin and c-myc. Compound 1 also upregulated the cell cycle inhibitor p21. Docking studies further revealed the favorable binding of compound 1 with the SH2 domain of STAT3, suggesting it acts through STAT3 inhibition. Taken together, our results suggest that compound 1 induces apoptosis by means of the inhibition of the STAT3 pathway, non-specifically targeting both B-RAF-mutant and WT melanoma cells, with much higher cytotoxicity than the current therapeutic drug PLX-4032. Full article
Show Figures

Graphical abstract

15 pages, 1396 KiB  
Article
Methylseleninic Acid Induces Lipid Peroxidation and Radiation Sensitivity in Head and Neck Cancer Cells
by John T. Lafin, Ehab H. Sarsour, Amanda L. Kalen, Brett A. Wagner, Garry R. Buettner and Prabhat C. Goswami
Int. J. Mol. Sci. 2019, 20(1), 225; https://doi.org/10.3390/ijms20010225 - 08 Jan 2019
Cited by 14 | Viewed by 4974
Abstract
Combination radiation and chemotherapy are commonly used to treat locoregionally advanced head and neck squamous cell carcinoma (HNSCC). Aggressive dosing of these therapies is significantly hampered by side effects due to normal tissue toxicity. Selenium represents an adjuvant that selectively sensitizes cancer cells [...] Read more.
Combination radiation and chemotherapy are commonly used to treat locoregionally advanced head and neck squamous cell carcinoma (HNSCC). Aggressive dosing of these therapies is significantly hampered by side effects due to normal tissue toxicity. Selenium represents an adjuvant that selectively sensitizes cancer cells to these treatments modalities, potentially by inducing lipid peroxidation (LPO). This study investigated whether one such selenium compound, methylseleninic acid (MSA), induces LPO and radiation sensitivity in HNSCC cells. Results from 4,4-difluoro-4-bora-3a,4a-diaza-S-indacene (BODIPY) C11 oxidation and ferric thiocyanate assays revealed that MSA induced LPO in cells rapidly and persistently. Propidium iodide (PI) exclusion assay found that MSA was more toxic to cancer cells than other related selenium compounds; this toxicity was abrogated by treatment with α-tocopherol, an LPO inhibitor. MSA exhibited no toxicity to normal fibroblasts at similar doses. MSA also sensitized HNSCC cells to radiation as determined by clonogenic assay. Intracellular glutathione in cancer cells was depleted following MSA treatment, and supplementation of the intracellular glutathione pool with N-acetylcysteine sensitized cells to MSA. The addition of MSA to a cell-free solution of glutathione resulted in an increase in oxygen consumption, which was abrogated by catalase, suggesting the formation of H2O2. Results from this study identify MSA as an inducer of LPO, and reveal its capability to sensitize HNSCC to radiation. MSA may represent a potent adjuvant to radiation therapy in HNSCC. Full article
Show Figures

Figure 1

12 pages, 1983 KiB  
Article
The Interaction of Selenium with Chemotherapy and Radiation on Normal and Malignant Human Mononuclear Blood Cells
by Richard J. Lobb, Gregory M. Jacobson, Ray T. Cursons and Michael B. Jameson
Int. J. Mol. Sci. 2018, 19(10), 3167; https://doi.org/10.3390/ijms19103167 - 15 Oct 2018
Cited by 23 | Viewed by 3580
Abstract
Selenium, a trace element with anticancer properties, can reduce harmful toxicities of chemotherapy and radiotherapy without compromising efficacy. However, the dose-response relationship in normal versus malignant human cells is unclear. We evaluated how methylseleninic acid (MSA) modulates the toxicity and efficacy of chemotherapy [...] Read more.
Selenium, a trace element with anticancer properties, can reduce harmful toxicities of chemotherapy and radiotherapy without compromising efficacy. However, the dose-response relationship in normal versus malignant human cells is unclear. We evaluated how methylseleninic acid (MSA) modulates the toxicity and efficacy of chemotherapy and radiation on malignant and non-malignant human mononuclear blood cells in vitro. We specifically investigated its effects on endoplasmic reticulum stress induction, intracellular glutathione concentration, DNA damage and viability of peripheral blood mononuclear cells and THP1 monocytic leukaemia cells in response to radiation, cytosine arabinoside or doxorubicin chemotherapy. MSA, at lower concentrations, induced protective responses in normal cells but cytotoxic effects in malignant cells, alone and in conjunction with chemotherapy or radiation. However, in normal cells higher concentrations of MSA were directly toxic and increased the cytotoxicity of radiation but not chemotherapy. In malignant cells higher MSA concentrations were generally more effective in combination with cancer treatments. Thus, optimal MSA concentrations differed between normal and malignant cells and treatments. This work supports clinical reports that selenium can significantly reduce dose-limiting toxicities of anticancer therapies and potentially improve efficacy of anticancer treatments. The optimal selenium compound and dose is not yet determined. Full article
Show Figures

Figure 1

17 pages, 2729 KiB  
Article
Novel Methylselenoesters Induce Programed Cell Death via Entosis in Pancreatic Cancer Cells
by Prajakta Khalkar, Nuria Díaz-Argelich, Juan Antonio Palop, Carmen Sanmartín and Aristi P. Fernandes
Int. J. Mol. Sci. 2018, 19(10), 2849; https://doi.org/10.3390/ijms19102849 - 20 Sep 2018
Cited by 23 | Viewed by 3477
Abstract
Redox active selenium (Se) compounds have gained substantial attention in the last decade as potential cancer therapeutic agents. Several Se compounds have shown high selectivity and sensitivity against malignant cells. The cytotoxic effects are exerted by their biologically active metabolites, with methylselenol (CH [...] Read more.
Redox active selenium (Se) compounds have gained substantial attention in the last decade as potential cancer therapeutic agents. Several Se compounds have shown high selectivity and sensitivity against malignant cells. The cytotoxic effects are exerted by their biologically active metabolites, with methylselenol (CH3SeH) being one of the key executors. In search of novel CH3SeH precursors, we previously synthesized a series of methylselenoesters that were active (GI50 < 10 µM at 72 h) against a panel of cancer cell lines. Herein, we refined the mechanism of action of the two lead compounds with the additional synthesis of new analogs (ethyl, pentyl, and benzyl derivatives). A novel mechanism for the programmed cell death mechanism for Se-compounds was identified. Both methylseleninic acid and the novel CH3SeH precursors induced entosis by cell detachment through downregulation of cell division control protein 42 homolog (CDC42) and its downstream effector β1-integrin (CD29). To our knowledge, this is the first time that Se compounds have been reported to induce this type of cell death and is of importance in the characterization of the anticancerogenic properties of these compounds. Full article
Show Figures

Graphical abstract

Review

Jump to: Research

26 pages, 607 KiB  
Review
Selenium Species: Current Status and Potentials in Cancer Prevention and Therapy
by Heng Wee Tan, Hai-Ying Mo, Andy T. Y. Lau and Yan-Ming Xu
Int. J. Mol. Sci. 2019, 20(1), 75; https://doi.org/10.3390/ijms20010075 - 25 Dec 2018
Cited by 140 | Viewed by 8071
Abstract
Selenium (Se) acts as an essential trace element in the human body due to its unique biological functions, particularly in the oxidation-reduction system. Although several clinical trials indicated no significant benefit of Se in preventing cancer, researchers reported that some Se species exhibit [...] Read more.
Selenium (Se) acts as an essential trace element in the human body due to its unique biological functions, particularly in the oxidation-reduction system. Although several clinical trials indicated no significant benefit of Se in preventing cancer, researchers reported that some Se species exhibit superior anticancer properties. Therefore, a reassessment of the status of Se and Se compounds is necessary in order to provide clearer insights into the potentiality of Se in cancer prevention and therapy. In this review, we organize relevant forms of Se species based on the three main categories of Se—inorganic, organic, and Se-containing nanoparticles (SeNPs)—and overview their potential functions and applications in oncology. Here, we specifically focus on the SeNPs as they have tremendous potential in oncology and other fields. In general, to make better use of Se compounds in cancer prevention and therapy, extensive further study is still required to understand the underlying mechanisms of the Se compounds. Full article
Show Figures

Graphical abstract

14 pages, 780 KiB  
Review
Current Landscape and the Potential Role of Hypoxia-Inducible Factors and Selenium in Clear Cell Renal Cell Carcinoma Treatment
by Rohan Garje, Josiah J. An, Kevin Sanchez, Austin Greco, Jeffrey Stolwijk, Eric Devor, Youcef Rustum and Yousef Zakharia
Int. J. Mol. Sci. 2018, 19(12), 3834; https://doi.org/10.3390/ijms19123834 - 01 Dec 2018
Cited by 24 | Viewed by 5370
Abstract
In the last two decades, the discovery of various pathways involved in renal cell carcinoma (RCC) has led to the development of biologically-driven targeted therapies. Hypoxia-inducible factors (HIFs), angiogenic growth factors, von Hippel–Lindau (VHL) gene mutations, and oncogenic microRNAs (miRNAs) play [...] Read more.
In the last two decades, the discovery of various pathways involved in renal cell carcinoma (RCC) has led to the development of biologically-driven targeted therapies. Hypoxia-inducible factors (HIFs), angiogenic growth factors, von Hippel–Lindau (VHL) gene mutations, and oncogenic microRNAs (miRNAs) play essential roles in the pathogenesis and drug resistance of clear cell renal cell carcinoma. These insights have led to the development of vascular endothelial growth factor (VEGF) inhibitors, Mechanistic target of rapamycin (mTOR) inhibitors, and immunotherapeutic agents, which have significantly improved the outcomes of patients with advanced RCC. HIF inhibitors will be a valuable asset in the growing therapeutic armamentarium of RCC. Various histone deacetylase (HDAC) inhibitors, selenium, and agents like PT2385 and PT2977 are being explored in various clinical trials as potential HIF inhibitors, to ameliorate the outcomes of RCC patients. In this article, we will review the current treatment options and highlight the potential role of selenium in the modulation of drug resistance biomarkers expressed in clear cell RCC (ccRCC) tumors. Full article
Show Figures

Figure 1

14 pages, 870 KiB  
Review
Selenium-Binding Protein 1 in Human Health and Disease
by Mostafa Elhodaky and Alan M. Diamond
Int. J. Mol. Sci. 2018, 19(11), 3437; https://doi.org/10.3390/ijms19113437 - 02 Nov 2018
Cited by 61 | Viewed by 4729
Abstract
Selenium-binding protein 1 (SBP1) is a highly conserved protein that covalently binds selenium. SBP1 may play important roles in several fundamental physiological functions, including protein degradation, intra-Golgi transport, cell differentiation, cellular motility, redox modulation, and the metabolism of sulfur-containing molecules. SBP1 expression is [...] Read more.
Selenium-binding protein 1 (SBP1) is a highly conserved protein that covalently binds selenium. SBP1 may play important roles in several fundamental physiological functions, including protein degradation, intra-Golgi transport, cell differentiation, cellular motility, redox modulation, and the metabolism of sulfur-containing molecules. SBP1 expression is often reduced in many cancer types compared to the corresponding normal tissues and low levels of SBP1 are frequently associated with poor clinical outcome. In this review, the transcriptional regulation of SBP1, the different physiological roles reported for SBP1, as well as the implications of SBP1 function in cancer and other diseases are presented. Full article
Show Figures

Figure 1

20 pages, 7275 KiB  
Review
Non-Coding Micro RNAs and Hypoxia-Inducible Factors Are Selenium Targets for Development of a Mechanism-Based Combination Strategy in Clear-Cell Renal Cell Carcinoma—Bench-to-Bedside Therapy
by Youcef M. Rustum, Sreenivasulu Chintala, Farukh A. Durrani and Arup Bhattacharya
Int. J. Mol. Sci. 2018, 19(11), 3378; https://doi.org/10.3390/ijms19113378 - 29 Oct 2018
Cited by 18 | Viewed by 3387
Abstract
Durable response, inherent or acquired resistance, and dose-limiting toxicities continue to represent major barriers in the treatment of patients with advanced clear-cell renal cell carcinoma (ccRCC). The majority of ccRCC tumors are characterized by the loss of Von Hippel–Lindau tumor suppressor gene function, [...] Read more.
Durable response, inherent or acquired resistance, and dose-limiting toxicities continue to represent major barriers in the treatment of patients with advanced clear-cell renal cell carcinoma (ccRCC). The majority of ccRCC tumors are characterized by the loss of Von Hippel–Lindau tumor suppressor gene function, a stable expression of hypoxia-inducible factors 1α and 2α (HIFs), an altered expression of tumor-specific oncogenic microRNAs (miRNAs), a clear cytoplasm with dense lipid content, and overexpression of thymidine phosphorylase. The aim of this manuscript was to confirm that the downregulation of specific drug-resistant biomarkers deregulated in tumor cells by a defined dose and schedule of methylselenocysteine (MSC) or seleno-l-methionine (SLM) sensitizes tumor cells to mechanism-based drug combination. The inhibition of HIFs by selenium was necessary for optimal therapeutic benefit. Durable responses were achieved only when MSC was combined with sunitinib (a vascular endothelial growth factor receptor (VEGFR)-targeted biologic), topotecan (a topoisomerase 1 poison and HIF synthesis inhibitor), and S-1 (a 5-fluorouracil prodrug). The documented synergy was selenium dose- and schedule-dependent and associated with enhanced prolyl hydroxylase-dependent HIF degradation, stabilization of tumor vasculature, downregulation of 28 oncogenic miRNAs, as well as the upregulation of 12 tumor suppressor miRNAs. The preclinical results generated provided the rationale for the development of phase 1/2 clinical trials of SLM in sequential combination with axitinib in ccRCC patients refractory to standard therapies. Full article
Show Figures

Figure 1

19 pages, 871 KiB  
Review
Role of Hypoxic Stress in Regulating Tumor Immunogenicity, Resistance and Plasticity
by Stéphane Terry, Rania Faouzi Zaarour, Goutham Hassan Venkatesh, Amirtharaj Francis, Walid El-Sayed, Stéphanie Buart, Pamela Bravo, Jérome Thiery and Salem Chouaib
Int. J. Mol. Sci. 2018, 19(10), 3044; https://doi.org/10.3390/ijms19103044 - 06 Oct 2018
Cited by 63 | Viewed by 5766
Abstract
Hypoxia, or gradients of hypoxia, occurs in most growing solid tumors and may result in pleotropic effects contributing significantly to tumor aggressiveness and therapy resistance. Indeed, the generated hypoxic stress has a strong impact on tumor cell biology. For example, it may contribute [...] Read more.
Hypoxia, or gradients of hypoxia, occurs in most growing solid tumors and may result in pleotropic effects contributing significantly to tumor aggressiveness and therapy resistance. Indeed, the generated hypoxic stress has a strong impact on tumor cell biology. For example, it may contribute to increasing tumor heterogeneity, help cells gain new functional properties and/or select certain cell subpopulations, facilitating the emergence of therapeutic resistant cancer clones, including cancer stem cells coincident with tumor relapse and progression. It controls tumor immunogenicity, immune plasticity, and promotes the differentiation and expansion of immune-suppressive stromal cells. In this context, manipulation of the hypoxic microenvironment may be considered for preventing or reverting the malignant transformation. Here, we review the current knowledge on how hypoxic stress in tumor microenvironments impacts on tumor heterogeneity, plasticity and resistance, with a special interest in the impact on immune resistance and tumor immunogenicity. Full article
Show Figures

Figure 1

Back to TopTop