Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
Semin Cancer Biol ; 80: 58-72, 2022 05.
Article in English | MEDLINE | ID: mdl-32070764

ABSTRACT

The recent advances in cancer immunotherapy confirm the crucial role of the immune system in cancer progression and treatment. Chronic inflammation and reduced immune surveillance are both features of the tumor microenvironment. Strategies aimed at reverting pro-tumor inflammation and stimulating the antitumor immune components are being actively searched, and the anticancer effects of many candidate drugs have been linked to their ability to modulate the immune system. Marine organisms constitute a rich reservoir of new bioactive molecules; some of them have already been exploited for pharmaceutical use, whereas many others are undergoing clinical or preclinical investigations for the treatment of different diseases, including cancer. In this review, we will discuss the immune-modulatory properties of marine compounds for their potential use in cancer prevention and treatment and as possible tools in the context of cancer immunotherapy.


Subject(s)
Neoplasms , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aquatic Organisms , Humans , Immunotherapy , Inflammation/drug therapy , Inflammation/prevention & control , Neoplasms/drug therapy , Tumor Microenvironment
2.
Pharmacol Res ; 196: 106914, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37714393

ABSTRACT

Immune checkpoint inhibitors (ICIs) are an expanding class of immunotherapeutic agents with the potential to cure cancer. Despite the outstanding clinical response in patient subsets, most individuals become refractory or develop resistance. Patient stratification and personalized immunotherapies are limited by the absence of predictive response markers. Recent findings show that dominant patterns of immune cell composition, T-cell status and heterogeneity, and spatiotemporal distribution of immune cells within the tumor microenvironment (TME) are becoming essential determinants of prognosis and therapeutic response. In this context, ICIs also function as investigational tools and proof of concept, allowing the validation of the identified mechanisms. After reviewing the current state of ICIs, this article will explore new comprehensive predictive markers for ICIs based on recent discoveries. We will discuss the recent establishment of a classification of TMEs into immune archetypes as a tool for personalized immune profiling, allowing patient stratification before ICI treatment. We will discuss the developing comprehension of T-cell diversity and its role in shaping the immune profile of patients. We describe the potential of strategies that score the mutual spatiotemporal modulation between T-cells and other cellular components of the TME. Additionally, we will provide an overview of a range of synthetic and naturally occurring or derived small molecules. We will compare compounds that were recently identified by in silico prediction to wet lab-validated drug candidates with the potential to function as ICIs and/or modulators of the cellular components of the TME.

3.
Mar Drugs ; 21(4)2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37103372

ABSTRACT

Palytoxin is considered one of the most potent biotoxins. As palytoxin-induced cancer cell death mechanisms remain to be elucidated, we investigated this effect on various leukemia and solid tumor cell lines at low picomolar concentrations. As palytoxin did not affect the viability of peripheral blood mononuclear cells (PBMC) from healthy donors and did not create systemic toxicity in zebrafish, we confirmed excellent differential toxicity. Cell death was characterized by a multi-parametric approach involving the detection of nuclear condensation and caspase activation assays. zVAD-sensitive apoptotic cell death was concomitant with a dose-dependent downregulation of antiapoptotic Bcl-2 family proteins Mcl-1 and Bcl-xL. Proteasome inhibitor MG-132 prevented the proteolysis of Mcl-1, whereas the three major proteasomal enzymatic activities were upregulated by palytoxin. Palytoxin-induced dephosphorylation of Bcl-2 further exacerbated the proapoptotic effect of Mcl-1 and Bcl-xL degradation in a range of leukemia cell lines. As okadaic acid rescued cell death triggered by palytoxin, protein phosphatase (PP)2A was involved in Bcl-2 dephosphorylation and induction of apoptosis by palytoxin. At a translational level, palytoxin abrogated the colony formation capacity of leukemia cell types. Moreover, palytoxin abrogated tumor formation in a zebrafish xenograft assay at concentrations between 10 and 30 pM. Altogether, we provide evidence of the role of palytoxin as a very potent and promising anti-leukemic agent, acting at low picomolar concentrations in cellulo and in vivo.


Subject(s)
Leukemia , Leukocytes, Mononuclear , Animals , Humans , Leukocytes, Mononuclear/metabolism , Zebrafish/metabolism , Down-Regulation , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , bcl-X Protein/metabolism , bcl-X Protein/pharmacology
4.
Mar Drugs ; 19(6)2021 May 21.
Article in English | MEDLINE | ID: mdl-34063867

ABSTRACT

Aplysinopsins are a class of marine indole alkaloids that exhibit a wide range of biological activities. Although both the indole and N-benzyl moieties of aplysinopsins are known to possess antiproliferative activity against cancer cells, their mechanism of action remains unclear. Through in vitro and in vivo proliferation and viability screening of newly synthesized aplysinopsin analogs on myelogenous leukemia cell lines and zebrafish toxicity tests, as well as analysis of differential toxicity in noncancerous RPMI 1788 cells and PBMCs, we identified EE-84 as a promising novel drug candidate against chronic myeloid leukemia. This indole derivative demonstrated drug-likeness in agreement with Lipinski's rule of five. Furthermore, EE-84 induced a senescent-like phenotype in K562 cells in line with its cytostatic effect. EE-84-treated K562 cells underwent morphological changes in line with mitochondrial dysfunction concomitant with autophagy and ER stress induction. Finally, we demonstrated the synergistic cytotoxic effect of EE-84 with a BH3 mimetic, the Mcl-1 inhibitor A-1210477, against imatinib-sensitive and resistant K562 cells, highlighting the inhibition of antiapoptotic Bcl-2 proteins as a promising novel senolytic approach against chronic myeloid leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Indoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Sulfonamides/pharmacology , Tryptophan/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line , Cytotoxins/chemistry , Cytotoxins/toxicity , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Humans , Indoles/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Sulfonamides/chemistry , Tryptophan/chemistry , Tryptophan/pharmacology , Tryptophan/toxicity , Zebrafish
5.
Pharmacol Res ; 160: 105058, 2020 10.
Article in English | MEDLINE | ID: mdl-32619722

ABSTRACT

Despite the discovery of tyrosine kinase inhibitors (TKIs) for the treatment of breakpoint cluster region-Abelson (BCR-ABL)+ cancer types, patients with chronic myeloid leukemia (CML) treated with TKIs develop resistance and severe adverse effects. Combination treatment, especially with a histone deacetylase (HDAC) 6 inhibitor (HDAC6i), appears to be an attractive option to prevent TKI resistance, considering the potential capacity of an HDAC6i to diminish BCR-ABL expression. We first validated the in vivo anti-cancer potential of the compound 7b by significantly reducing the tumor burden of BALB/c mice xenografted with K-562 cells, without notable organ toxicity. Here, we hypothesize that the HDAC6i compound 7b can lead to BCR-ABL downregulation in CML cells and sensitize them to TKI treatment. The results showed that combination treatment with imatinib and 7b resulted in strong synergistic caspase-dependent apoptotic cell death and drastically reduced the proportion of leukemia stem cells, whereas this treatment only moderately affected healthy cells. Ultimately, the combination significantly decreased colony formation in a semisolid methylcellulose medium and tumor mass in xenografted zebrafish compared to each compound alone. Mechanistically, the combination induced BCR-ABL ubiquitination and downregulation followed by disturbance of key proteins in downstream pathways involved in CML proliferation and survival. Taken together, our results suggest that an HDAC6i potentiates the effect of imatinib and could overcome TKI resistance in CML cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fusion Proteins, bcr-abl/metabolism , Histone Deacetylase 6/antagonists & inhibitors , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Ubiquitination/drug effects , Animals , Caspases/drug effects , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , K562 Cells , Mice , Mice, Inbred BALB C , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
6.
Cancer ; 125(10): 1612-1628, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30840315

ABSTRACT

The immune system represents the major primary defense line against carcinogenesis and acts by identifying and eradicating nascent transformed cells. A growing body of evidence is indicating that aberrant epigenetic reprogramming plays a key role in tumor immune escape through: 1) impaired efficient recognition of neoplastic cells by the immune system, resulting from a downregulation or loss of the expression of tumor-associated antigens, human leukocyte antigens, antigen processing and presenting machinery, and costimulatory molecule genes; 2) aberrant expression of immune checkpoint proteins and their ligands; and 3) modification of cytokine profiles and tumor-associated immune cell populations toward an immunosuppressive state in the tumor microenvironment. Consistent with the inherent reversibility of epigenetic alterations, epigenetic drugs, including DNA methyltransferase and histone deacetylase inhibitors, have the unique potential to favorably modify the tumor microenvironment, restore tumor recognition and stimulate an antitumor immune response. The objective of this review is to highlight selected, naturally occurring epigenetic modulators, namely, butyrate, curcumin, (-)-epigallocatechin-3-gallate, resveratrol, romidepsin, and trichostatin A, with a special focus on their antitumor immune properties.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Neoplasms/pathology , Tumor Escape/genetics , DNA Modification Methylases/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Humans , Neoplasms/genetics , Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
7.
Pharmacol Res ; 129: 337-356, 2018 03.
Article in English | MEDLINE | ID: mdl-29133216

ABSTRACT

Alterations of the epigenetic machinery, affecting multiple biological functions, represent a major hallmark enabling the development of tumors. Among epigenetic regulatory proteins, histone deacetylase (HDAC)6 has emerged as an interesting potential therapeutic target towards a variety of diseases including cancer. Accordingly, this isoenzyme regulates many vital cellular regulatory processes and pathways essential to physiological homeostasis, as well as tumor multistep transformation involving initiation, promotion, progression and metastasis. In this review, we will consequently discuss the critical implications of HDAC6 in distinct mechanisms relevant to physiological and cancerous conditions, as well as the anticancer properties of synthetic, natural and natural-derived compounds through the modulation of HDAC6-related pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase 6/metabolism , Neoplasms/drug therapy , Humans , Neoplasms/metabolism , Signal Transduction
8.
Mar Drugs ; 16(12)2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30572618

ABSTRACT

Treatment of acute myeloid leukemia (AML) patients is still hindered by resistance and relapse, resulting in an overall poor survival rate. Recently, combining specific B-cell lymphoma (Bcl)-2 inhibitors with compounds downregulating myeloid cell leukemia (Mcl)-1 has been proposed as a new effective strategy to eradicate resistant AML cells. We show here that 1(R), 6(S), 1'(R), 6'(S), 11(R), 17(S)-fistularin-3, a bromotyrosine compound of the fistularin family, isolated from the marine sponge Suberea clavata, synergizes with Bcl-2 inhibitor ABT-199 to efficiently kill Mcl-1/Bcl-2-positive AML cell lines, associated with Mcl-1 downregulation and endoplasmic reticulum stress induction. The absolute configuration of carbons 11 and 17 of the fistularin-3 stereoisomer was fully resolved in this study for the first time, showing that the fistularin we isolated from the marine sponge Subarea clavata is in fact the (+)-11(R), 17(S)-fistularin-3 stereoisomer keeping the known configuration 1(R), 6(S), 1'(R), and 6'(S) for the verongidoic acid part. Docking studies and in vitro assays confirm the potential of this family of molecules to inhibit DNA methyltransferase 1 activity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Isoxazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Tyrosine/analogs & derivatives , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , HL-60 Cells , Humans , Isoxazoles/administration & dosage , Isoxazoles/chemistry , Isoxazoles/isolation & purification , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Docking Simulation , Porifera/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/administration & dosage , Tyrosine/administration & dosage , Tyrosine/chemistry , Tyrosine/isolation & purification , Tyrosine/pharmacology , U937 Cells
9.
Molecules ; 23(2)2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29401749

ABSTRACT

NAD⁺-dependent histone deacetylases (sirtuins) are implicated in cellular processes such as proliferation, DNA repair, and apoptosis by regulating gene expression and the functions of numerous proteins. Due to their key role in cells, the discovery of small molecule sirtuin modulators has been of significant interest for diverse therapeutic applications. In particular, it has been shown that inhibition of sirtuin 1 and 2 activities is beneficial for cancer treatment. Here, we demonstrate that the fungal metabolite eurochevalierine from the fungus Neosartorya pseudofischeri inhibits sirtuin 1 and 2 activities (IC50 about 10 µM) without affecting sirtuin 3 activity. The binding modes of the eurochevalierine for sirtuin 1 and 2 have been identified through computational docking analyses. Accordingly, this sequiterpene alkaloid induces histone H4 and α-tubulin acetylation in various cancer cell models in which it induces strong cytostatic effects without affecting significantly the viability of healthy PBMCs. Importantly, eurochevalierine targets preferentially cancer cell proliferation (selectivity factor ≫ 7), as normal human primary CD34⁺ stem/progenitor cells were less affected by the treatment. Finally, eurochevalierine displays suitable drug-likeness parameters and therefore represent a promising scaffold for lead molecule optimization to study the mechanism and biological roles of sirtuins and potentially a basis for development into therapeutics.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Protein Processing, Post-Translational , Sesquiterpenes/pharmacology , Sirtuin 1/antagonists & inhibitors , Sirtuin 2/antagonists & inhibitors , Acetylation , Alkaloids/chemistry , Alkaloids/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Binding Sites , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/isolation & purification , Histones/genetics , Histones/metabolism , Humans , Molecular Docking Simulation , Neosartorya/chemistry , Neosartorya/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism , Tubulin/genetics , Tubulin/metabolism
10.
Lancet Oncol ; 17(11): e502-e509, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27819248

ABSTRACT

Biosimilars are considered to be one of the solutions to combat the substantially increasing costs of cancer treatment, and its imminent introduction is expected to expand affordability worldwide. However, biosimilar monoclonal antibodies provide many challenges compared with first-generation biosimilars, growth factors, and hormones, because they have shown only a modest clinical effect, and are often used in combination with other more toxic therapies, making it difficult to design studies that allow appropriate efficacy and safety assessments compared with the original products. The value of comparative clinical trials for showing clinical equivalence of biosimilars that demonstrate a high degree of similarity in physical, chemical, structural, and biological characteristics with the original product is increasingly being questioned, and advances in analytical methods that provide robust non-clinical data might reduce the need for extensive clinical comparisons. In this Series paper, the third of three papers on drug safety in oncology, we review the safety and efficacy of biosimilars in oncology, assessing biosimilar monoclonal antibodies in relation to first-generation biosimilars, the issues surrounding interchangeability and extrapolation of biosimilars to other disease and patient indications, and reassessing the safety approval pathway in light of 10 years worth of biosimilar experience.


Subject(s)
Biosimilar Pharmaceuticals/adverse effects , Neoplasms/drug therapy , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Biosimilar Pharmaceuticals/therapeutic use , Humans
11.
Molecules ; 21(11)2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27886118

ABSTRACT

The remarkable complexity of cancer involving multiple mechanisms of action and specific organs led researchers Hanahan and Weinberg to distinguish biological capabilities acquired by cancer cells during the multistep development of human tumors to simplify its understanding. These characteristic hallmarks include the abilities to sustain proliferative signaling, evade growth suppressors, resist cell death, enable replicative immortality, induce angiogenesis, activate invasion and metastasis, avoid immune destruction, and deregulate cellular energetics. Furthermore, two important characteristics of tumor cells that facilitate the acquisition of emerging hallmarks are tumor-promoting inflammation and genome instability. To treat a multifactorial disease such as cancer, a combination treatment strategy seems to be the best approach. Here we focus on natural histone deacetylase inhibitors (HDACi), their clinical uses as well as synergies with modulators of the pro-inflammatory transcription factor signaling pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Drug Synergism , Genomic Instability/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction/drug effects , Transcription Factors/metabolism
12.
J Pharmacol Exp Ther ; 355(2): 308-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26330537

ABSTRACT

Cyclooxygenase-2 (COX-2) is an essential regulator of cancer promotion and progression. Extensive efforts to target this enzyme have been developed to reduce growth of cancer cells for chemopreventive and therapeutic reasons. In this context, cyclooxygenase-2 inhibitors present interesting antitumor effects. However, inhibition of COX-2 by anti-COX-2 compounds such as celecoxib was recently associated with detrimental cardiovascular side effects limiting their clinical use. As many anticancer effects of celecoxib are COX-2 independent, analogs such as 2,5-dimethyl-celecoxib (DMC), which lacks COX-2-inhibitory activity, represent a promising alternative strategy. In this study, we investigated the effect of this molecule on growth of hematologic cancer cell lines (U937, Jurkat, Hel, Raji, and K562). We found that this molecule is able to reduce the growth and induces apoptosis more efficiently than celecoxib in all the leukemic cell lines tested. Cell death was associated with downregulation of Mcl-1 protein expression. We also found that DMC induces endoplasmic reticulum stress, which is associated with a decreased of GRP78 protein expression and an alteration of cell cycle progression at the G1/S transition in U937 cells. Accordingly, typical downregulation of c-Myc and cyclin D1 and an upregulation of p27 were observed. Interestingly, for shorter time points, an alteration of mitotic progression, associated with the downregulation of survivin protein expression was observed. Altogether, our data provide new evidence about the mode of action of this compound on hematologic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/toxicity , Apoptosis , Calcium/metabolism , Celecoxib/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2 Inhibitors/toxicity , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Humans , Leukemia , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Pyrazoles/toxicity , Sulfonamides/toxicity , Zebrafish
13.
Mol Carcinog ; 54(10): 999-1013, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24798089

ABSTRACT

Molecules targeting pro-inflammatory pathways have demonstrated beneficial effects in cancer treatment. More recently, combination of natural and synthetic anti-inflammatory drugs was suggested as an appealing strategy to inhibit tumor growth. Herein, we show that curcumin, a polyphenol from Curcuma longa and celecoxib induce apoptosis in hematopoietic cancer cell lines (Hel, Jurkat, K562, Raji, and U937). Further investigations on the most sensitive cell line, U937, indicated that these effects were tightly associated with an accumulation of the cells in S and G2/M for curcumin and in G0/G1 phase of cell cycle for celecoxib, respectively. The effect of celecoxib on cell cycle is associated with an induction of p27 and the down-regulation of cyclin D1. However, in the case of combination experiments, the pretreatment of U937 cells with celecoxib at non-apoptogenic concentrations counteracted curcumin-induced apoptosis. We found that this effect correlated with the prevention of the accumulation in S and G2/M phase of cell cycle induced by curcumin. Similar results have been obtained when celecoxib and curcumin were co-administrated at the same time. Overall our data suggest that this natural and synthetic drug combination is detrimental for cell death induction.


Subject(s)
Apoptosis/drug effects , Celecoxib/pharmacology , Curcumin/pharmacology , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Humans , Jurkat Cells , K562 Cells , U937 Cells
14.
Mol Carcinog ; 54(3): 229-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24155226

ABSTRACT

Cell division cycle (CDC) 25 proteins are key phosphatases regulating cell cycle transition and proliferation by regulating CDK/cyclin complexes. Overexpression of these enzymes is frequently observed in cancer and is related to aggressiveness, high-grade tumors and poor prognosis. Thus, targeting CDC25 by compounds, able to inhibit their activity, appears a good therapeutic approach. Here, we describe the synthesis of a new inhibitor (SV37) whose structure is based on both coumarin and quinone moieties. An analytical in vitro approach shows that this compound efficiently inhibits all three purified human CDC25 isoforms (IC50 1-9 µM) in a mixed-type mode. Moreover, SV37 inhibits growth of breast cancer cell lines. In MDA-MB-231 cells, reactive oxygen species generation is followed by pCDK accumulation, a mark of CDC25 dysfunction. Eventually, SV37 treatment leads to activation of apoptosis and DNA cleavage, underlining the potential of this new type of coumarin-quinone structure.


Subject(s)
Apoptosis/drug effects , Benzoquinones/chemistry , Breast Neoplasms/enzymology , Coumarins/chemistry , Coumarins/pharmacology , Naphthoquinones/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Cyclin-Dependent Kinases/metabolism , DNA Cleavage/drug effects , Female , Humans , MCF-7 Cells , Naphthoquinones/chemical synthesis , Naphthoquinones/chemistry , Protein Isoforms/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Viral Proteins
16.
Molecules ; 20(4): 7097-142, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25903364

ABSTRACT

Although considerable effort and progress has been made in the search for new anticancer drugs and treatments in the last several decades, cancer remains a major public health problem and one of the major causes of death worldwide. Many sources, including plants, animals, and minerals, are of interest in cancer research because of the possibility of identifying novel molecular therapeutics. Moreover, structure-activity-relationship (SAR) investigations have become a common way to develop naturally derived or semi-synthetic molecular analogues with improved efficacy and decreased toxicity. In 2012, approximately 138 molecules from marine sources, including isolated compounds and their associated analogues, were shown to be promising anticancer drugs. Among these, 62% are novel compounds. In this report, we review the marine compounds identified in 2012 that may serve as novel anticancer drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Animals , Antineoplastic Agents/chemistry , Biological Products/chemistry , Drug Discovery , Humans , Marine Biology , Molecular Structure , Neoplasms/drug therapy , Structure-Activity Relationship
17.
Cancer Treat Res ; 159: 123-43, 2014.
Article in English | MEDLINE | ID: mdl-24114478

ABSTRACT

Over the centuries, plant extracts have been used to treat various diseases. Until now, natural products have played an important role in anticancer therapy as there are more than 500 compounds from terrestrial and marine plants or microorganisms, which have antioxidant, antiproliferative, or antiangiogenic properties and are therefore able to reduce tumor growth. The recent discovery of new natural products has been accelerated by novel technologies (high throughput screening of natural products in plants, animals, marine organisms, and microorganisms). Vincristine, irinotecan, etoposide, and paclitaxel are examples of compounds derived from plants that are used in cancer treatment. Similarly, actinomycin D, mitomycin C, bleomycin, doxorubicin, and L-asparaginase are drugs derived from microorganisms. In this review, we describe the molecular mechanisms of natural compounds with anti-inflammatory and anticancer activities.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Neoplasms/drug therapy , Animals , Humans
18.
Bioorg Med Chem Lett ; 24(16): 3797-801, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25042254

ABSTRACT

Histone deacetylases (HDACs) are well-established, promising targets for anticancer therapy due to their critical role in cancer development. Accordingly, an increasing number of HDAC inhibitors displaying cytotoxic effects against cancer cells have been reported. Among them, a large panel of chemical structures was described including coumarin-containing molecules. In this study, we described synthesis and biological activity of new coumarin-based derivatives as HDAC inhibitors. Among eight derivatives, three compounds showed HDAC inhibitory activities and antitumor activities against leukemia cell lines without affecting the viability of peripheral blood mononuclear cells from healthy donors.


Subject(s)
Antineoplastic Agents/pharmacology , Coumarins/pharmacology , Drug Design , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , K562 Cells , Molecular Structure , Structure-Activity Relationship , U937 Cells
19.
Molecules ; 19(12): 20839-63, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25514225

ABSTRACT

Cancer is a multifactorial disease that requires treatments able to target multiple intracellular components and signaling pathways. The natural compound, curcumin, was already described as a promising anticancer agent due to its multipotent properties and huge amount of molecular targets in vitro. Its translation to the clinic is, however, limited by its reduced solubility and bioavailability in patients. In order to overcome these pharmacokinetic deficits of curcumin, several strategies, such as the design of synthetic analogs, the combination with specific adjuvants or nano-formulations, have been developed. By taking into account the risk-benefit profile of drug combinations, as well as the knowledge about curcumin's structure-activity relationship, a new concept for the combination of curcumin with scaffolds from different natural products or components has emerged. The concept of a hybrid curcumin molecule is based on the incorporation or combination of curcumin with specific antibodies, adjuvants or other natural products already used or not in conventional chemotherapy, in one single molecule. The high diversity of such conjugations enhances the selectivity and inherent biological activities and properties, as well as the efficacy of the parental compound, with particular emphasis on improving the efficacy of curcumin for future clinical treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/analogs & derivatives , Neoplasms/drug therapy , Biological Availability , Chemistry, Pharmaceutical , Curcumin/pharmacokinetics , Drug Synergism , Humans
20.
Molecules ; 19(7): 10011-32, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25014531

ABSTRACT

Plumbagin is a plant naphtoquinone exerting anti-cancer properties including apoptotic cell death induction and generation of reactive oxygen species (ROS). The aim of this study was to elucidate parameters explaining the differential leukemia cell sensitivity towards this compound. Among several leukemia cell lines, U937 monocytic leukemia cells appeared more sensitive to plumbagin treatment in terms of cytotoxicity and level of apoptotic cell death compared to more resistant Raji Burkitt lymphoma cells. Moreover, U937 cells exhibited a ten-fold higher ROS production compared to Raji. Neither differential incorporation, nor efflux of plumbagin was detected. Pre-treatment with thiol-containing antioxidants prevented ROS production and subsequent induction of cell death by apoptosis whereas non-thiol-containing antioxidants remained ineffective in both cellular models. We conclude that the anticancer potential of plumbagin is driven by pro-oxidant activities related to the cellular thiolstat.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Leukemia/metabolism , Naphthoquinones/pharmacology , Oxidation-Reduction/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glutathione/metabolism , Humans , Reactive Oxygen Species/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL