Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Int J Biol Macromol ; 256(Pt 1): 128401, 2024 Jan.
Article En | MEDLINE | ID: mdl-38007027

Leishmaniasis, caused by a protozoan parasite, is among humanity's costliest banes, owing to the high mortality and morbidity ratio in poverty-stricken areas. To date, no vaccine is available for the complete cure of the disease. Current chemotherapy is expensive, has undesirable side effects, and faces drug resistance limitations and toxicity concerns. The substantial differences in homology between leishmanial DNA topoisomerase IB compared with the human counterparts provided a new lead in the study of the structural determinants that can be targeted. Several research groups explored this molecular target, trying to fill the therapeutic gap, and came forward with various anti-leishmanial scaffolds. This article is a comprehensive review of knowledge about topoisomerases as an anti-leishmanial drug target and their inhibitors collected over the years. In addition to information on molecular targets and reported scaffolds, the review details the structure-activity relationship of described compounds with leishmanial Topoisomerase IB. Moreover, the work also includes information about the structure of the inhibitors, showing common interacting residues with leishmanial topoisomerases that drive their mode of action towards them. Finally, in search of topoisomerase inhibitors at the stage of clinical trials, we have listed all the drugs that have been in clinical trials against leishmaniasis.


Leishmania , Leishmaniasis , Humans , DNA Topoisomerases/pharmacology , Leishmaniasis/drug therapy , DNA Topoisomerases, Type I/metabolism , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use
2.
Eur J Pharm Sci ; 192: 106632, 2024 Jan 01.
Article En | MEDLINE | ID: mdl-37949194

Antimicrobial resistance caused by the excessive and inappropriate use of antibacterial drugs is a global health concern. Currently, we are walking a fine line between the fact that most bacterial infections can still be cured with the antibiotics known so far, and the emergence of infections with bacteria resistant to several drugs at the same time, against which we no longer have an effective drug. Therefore, new antibacterial drugs are urgently needed to curb the hard-to-treat infections. Our group has developed new antibacterials from the class of novel bacterial topoisomerase inhibitors (NBTIs) that exhibit broad-spectrum antibacterial activity. This article reviews our efforts in developing highly potent NBTIs over the past decade. Following the discovery of an initial hit with potent enzyme inhibitory and broad-spectrum antibacterial activity, an extensive hit-to-lead campaign was conducted with the goal of optimizing physicochemical properties, reducing hERG inhibition, and maintaining antibacterial activity against both Gram-positive and Gram-negative bacteria, with a focus on methicillin-resistant Staphylococcus aureus (MRSA). This optimization strategy resulted in an amide-containing, focused NBTI library with compounds exhibiting potent antibacterial activity against Gram-positive bacteria, reduced hERG inhibition, no cardiotoxicity in in vivo zebrafish model, and favorable in vivo efficacy in a neutropenic murine thigh infection model for MRSA infections.


Methicillin-Resistant Staphylococcus aureus , Topoisomerase Inhibitors , Mice , Animals , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use , Topoisomerase Inhibitors/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , DNA Gyrase/chemistry , DNA Gyrase/pharmacology , Zebrafish , Gram-Positive Bacteria , Gram-Negative Bacteria , Microbial Sensitivity Tests , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use
3.
Cell Mol Biol (Noisy-le-grand) ; 69(15): 217-222, 2023 Dec 31.
Article En | MEDLINE | ID: mdl-38279438

IgG4-related sialadenitis is a systemic autoimmune disease that can lead to fibro-inflammatory conditions. This study aims to investigate the immune microenvironment and potential signaling pathways associated with IgG4-related sialadenitis. Datasets related to IgG4-related sialadenitis were retrieved from the GEO database. Immune cell infiltration analysis was conducted using the Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) method. Differentially immune-related expressed genes (DIEG) and immune-related functional enrichment were identified. Moreover, potential treatment targets for IgG4-related sialadenitis were predicted using The Connectivity Map. Only two datasets from GEO were included for further analysis. The CIBERSORT results indicated dominant immune cell populations in IgG4-related sialadenitis, including CD8+ T cells, resting NK cells, monocytes, and naïve B cells in peripheral blood mononuclear cells. Additionally, high abundance of plasma cells was observed in labial salivary gland tissues. Furthermore, a total of 42 DIEGs were identified, with tumor necrosis factor (TNF) signaling via the NF-Kappa B signaling pathway and the p53 signaling pathway being highly enriched. Autophagy inhibitors and DNA topoisomerase inhibitors were strongly associated with potential targets for the treatment of IgG4-related sialadenitis (P<0.05). This study reveals altered immune microenvironment in peripheral blood mononuclear cells and labial salivary gland tissues in IgG4-related sialadenitis. Autophagy inhibitors and DNA topoisomerase inhibitors show promise as potential targets for treating IgG4-related sialadenitis, providing a novel therapeutic strategy for this disease.


Immunoglobulin G , Sialadenitis , Humans , Leukocytes, Mononuclear/pathology , Sialadenitis/drug therapy , Sialadenitis/pathology , Plasma Cells , Topoisomerase Inhibitors/therapeutic use
4.
Biochem Pharmacol ; 205: 115294, 2022 11.
Article En | MEDLINE | ID: mdl-36241099

Combination therapy can enhance therapeutic effect by activation of multiple downstream pathways. The present study was aimed to investigate a novel strategy to successfully inhibit the EGFR pathway in EGFR wild and mutated types lung cancer by combination method. Topotecan (TPT) and crizotinib (CRI) were used to evaluate the effect on EGFR-wild, primary and secondary mutant non-small cell lung cancer (NSCLC) cell lines (H1299, HCC827 and H1975 cells). The combination group significantly inhibited the lung cancer growth with combination index (CI) < 1, and they synergistically induced the cell apoptosis by disrupting the balance of Bax and Bcl-xL, loss of mitochondrial membrane potential (MMP), and accumulation of reactive oxygen species (ROS). In addition, EGFR downstream signaling pathways including AKT, ERK, JNK, and p38 MAPK were regulated when treated with the combination regimen. Meanwhile, a nano-liposomes co-loaded CRI and TPT was prepared and exhibited strong cytotoxicity to the lung cancer cells especially H1299 and H1975 cells. The animal study confirmed the synergy between TPT and CRI from the results that they remarkable repressed the tumor growth with the inhibition rate of 81.32 %. The nano-liposomes of TPT and CRI achieved an optimal curative effect (71.52 % of inhibition rate) at 2 mg/kg. Moreover, the synergistic mechanism of the combination was consistent with the in vitro cell experiment by regulating EGFR signaling pathways. Collectively, we proposed a preclinical rationale and potential formulation for the use of a combination therapy consisting of the topoisomerase inhibitor TPT and the ALK-TKI CRI for treatment of lung cancers.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Lung Neoplasms/metabolism , Crizotinib/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Reactive Oxygen Species , ErbB Receptors/metabolism , Topoisomerase Inhibitors/therapeutic use , Quinazolines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , bcl-2-Associated X Protein , Topotecan/pharmacology , Topotecan/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cell Line, Tumor , p38 Mitogen-Activated Protein Kinases , Drug Resistance, Neoplasm
5.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article En | MEDLINE | ID: mdl-36232843

Topoisomerases are essential enzymes that recognize and modify the topology of DNA to allow DNA replication and transcription to take place. Topoisomerases are divided into type I topoisomerases, that cleave one DNA strand to modify DNA topology, and type II, that cleave both DNA strands. Topoisomerases normally rapidly religate cleaved-DNA once the topology has been modified. Topoisomerases do not recognize specific DNA sequences, but actively cleave positively supercoiled DNA ahead of transcription bubbles or replication forks, and negative supercoils (or precatenanes) behind, thus allowing the unwinding of the DNA-helix to proceed (during both transcription and replication). Drugs that stabilize DNA-cleavage complexes with topoisomerases produce cytotoxic DNA damage and kill fast-dividing cells; they are widely used in cancer chemotherapy. Oligonucleotide-recognizing topoisomerase inhibitors (OTIs) have given drugs that stabilize DNA-cleavage complexes specificity by linking them to either: (i) DNA duplex recognizing triplex forming oligonucleotide (TFO-OTIs) or DNA duplex recognizing pyrrole-imidazole-polyamides (PIP-OTIs) (ii) or by conventional Watson-Crick base pairing (WC-OTIs). This converts compounds from indiscriminate DNA-damaging drugs to highly specific targeted DNA-cleaving OTIs. Herein we propose simple strategies to enable DNA-duplex strand invasion of WC-OTIs giving strand-invading SI-OTIs. This will make SI-OTIs similar to the guide RNAs of CRISPR/Cas9 nuclease bacterial immune systems. However, an important difference between OTIs and CRISPR/Cas9, is that OTIs do not require the introduction of foreign proteins into cells. Recent successful oligonucleotide therapeutics for neurodegenerative diseases suggest that OTIs can be developed to be highly specific gene editing agents for DNA lesions that cause neurodegenerative diseases.


Neurodegenerative Diseases , Oligonucleotides , DNA/metabolism , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , DNA, Superhelical , Humans , Imidazoles , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Nylons , Oligonucleotides/chemistry , Pyrroles , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use
6.
Breast ; 66: 169-177, 2022 Dec.
Article En | MEDLINE | ID: mdl-36302269

Antibody drug conjugates (ADCs) combine the potent cytotoxicity of chemotherapy with the antigen -specific targeted approach of antibodies into one single molecule. Trophoblast cell surface antigen 2 (TROP-2) is a transmembrane glycoprotein involved in calcium signal transduction and is expressed in multiple tumor types. TROP-2 expression is higher in HER2-negative breast tumors (HR+/HR-) and is associated with worse survival. Sacituzumab govitecan (SG) is a first-in-class TROP-2-directed ADC with an anti-TROP-2 antibody conjugated to SN-38, a topoisomerase inhibitor via a hydrolysable linker. This hydrolysable linker permits intracellular and extracellular release of the membrane permeable payload enabling the "bystander effect" contributing to the efficacy of this agent. There was significant improvement in progression free survival (PFS) and overall survival (OS) with SG versus chemotherapy in pretreated metastatic triple negative breast cancer (TNBC), resulting in regulatory approval. Common adverse events (AE) reported were neutropenia and diarrhea. SG also demonstrated clinical activity versus chemotherapy in a phase III trial of HR+/HER2-metastatic breast cancer (MBC) and is under evaluation in first-line metastatic and early stage TNBC as well. Datopotamab deruxtecan (Dato-DXd) is a TROP-2 ADC that differs from SG in that it has a cleavable tetrapeptide linker and a more potent topoisomerase inhibitor payload. This construct is highly stable in circulation with a longer half-life than SG, and undergoes cleavage in presence of intracellular lysosomal proteases. Dato-DXd demonstrated preliminary efficacy in unselected metastatic TNBC, with common AEs of low-grade nausea and stomatitis. Dato-DXd is being investigated in phase III studies in metastatic TNBC and HR+/HER2- MBC. These novel TROP-2 ADCs have the potential to deliver enhanced efficacy with reduced toxicity in MBC and possibly in early stage breast cancer (EBC).


Antineoplastic Agents , Breast Neoplasms , Immunoconjugates , Triple Negative Breast Neoplasms , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/chemically induced , Immunoconjugates/therapeutic use , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Irinotecan/therapeutic use , Topoisomerase Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/drug therapy
7.
Acta Biomater ; 144: 121-131, 2022 05.
Article En | MEDLINE | ID: mdl-35304322

Overexpressed DNA topoisomerase II alpha (TOP-2A) is closely related to the invasion and metastasis of malignant breast tumors. Mitoxantrone (MTX) has been identified as a TOP-2A inhibitor with significant inhibitory activity against breast tumors. The tumor-homing ability of MTX has been further enhanced by using nanodrug delivery systems (nano-DDSs), reducing off-target side effects. However, conventional MTX nano-DDSs are still limited by low drug-loading capacity and material carrier-related toxicity. In this study, we developed metal iron-coordinated carrier-free supramolecular co-nanoassemblies of dual DNA topoisomerase-targeting inhibitors with high drug loading for superimposed DNA damage-augmented tumor regression. By introducing iron ions (Ⅲ) and another TOP-2A inhibitor quercetin (QU) onto the building blocks, Fe3+-mediated QU-MTX co-nanoassemblies are fabricated (QU-MTX-Fe) via intermolecular coordination interactions. The PEGylated co-nanoassemblies (P-QU-MTX-Fe) exhibit distinct advantages over QU/MTX solution (Sol) alone or MTX-QU mixture Sol in terms of therapeutic efficacy and systemic toxicity. Meanwhile, P-QU-MTX-Fe could efficiently suppress primary and distal breast tumor relapse by activating the CD 8+-mediated antitumor immune response. Overall, such iron-coordinated nanomedicines provide insights into the rational design of drug-likeness compounds with undesirable therapeutic performance for cancer therapy. STATEMENT OF SIGNIFICANCE: Aimed at the key target TOP-2A in the malignant breast tumor, the metal coordination-mediated supramolecular co-assemble strategy of one-target dual inhibitors was firstly proposed for superimposed DNA damage for cancer therapy. Multiple interactions involving π-π stacking interactions, hydrogen bonds and coordination forces maintained the stability of co-nanoassemblies. Meanwhile, this co-nanoassemblies not only had potentials to increase therapeutic efficacy and decrease systemic toxicity, but also activated the CD 8+-mediated antitumor immune response against distal breast tumor relapse. Such a facile and safe nanoplatform is expected to provide an important prospective for promoting the clinical transformation of drug-likeness compounds in the suppression of difficult-to-treat breast tumor.


Breast Neoplasms , Nanoparticles , Breast Neoplasms/drug therapy , Cell Line, Tumor , DNA Topoisomerases , Female , Humans , Ions , Iron/therapeutic use , Nanoparticles/chemistry , Neoplasm Recurrence, Local/drug therapy , Prospective Studies , Quercetin , Topoisomerase Inhibitors/therapeutic use
8.
Front Immunol ; 12: 691713, 2021.
Article En | MEDLINE | ID: mdl-34925310

Objective: To investigate the characteristics of the tumor immune microenvironment in patients with gastrointestinal stromal tumor (GIST) and identify cancer stem-like properties of GIST to screen potential druggable molecular targets. Methods: The gene expression data of 60 patients with GIST was retrieved from the Array Express database. CIBERSORT was applied to calculate the level of immune infiltration. ssGSEA and ESTIMATE were used to calculate the cancer stemness index and tissue purity. The Connectivity Map (CMAP) database was implemented to screen targeted drugs based on cancer stem-like properties of GIST. Result: There was a difference in the level of immune infiltration between the metastasis and non-metastasis GIST groups. The low level of T-cell infiltration was correlated with high tumor purity and tumor stemness index, and the correlation coefficients were -0.87 and -0.61 (p < 0.001), respectively. Furthermore, there was a positive correlation between cancer stemness index and cell purity (p < 0.001). The cancer stemness index in the metastasis group was higher than that in the non-metastasis group (p = 0.0017). After adjusting for tumor purity, there was no significant correlation between T-cell infiltration and cancer stemness index (p = 0.086). Through the pharmacological mechanism of topoisomerase inhibitors, six molecular complexes may be the targets of GIST treatment. Conclusion: Immune infiltration in GIST patients is related to cancer stem-like properties, and the correlation relies on tumor purity. Cancer stemness index can be used as a new predictive biomarker of tumor metastasis and targets of drug therapy for GIST patients.


Cell Self Renewal , Gastrointestinal Neoplasms/therapy , Gastrointestinal Stromal Tumors/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Molecular Targeted Therapy , Neoplastic Stem Cells/pathology , Cell Self Renewal/drug effects , Epithelial-Mesenchymal Transition , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gastrointestinal Stromal Tumors/secondary , Gene Expression Profiling , Gene Ontology , Humans , Neoplasm Metastasis , Protein Interaction Maps , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Support Vector Machine , T-Lymphocyte Subsets/immunology , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use , Tumor Microenvironment/immunology
9.
Phytomedicine ; 84: 153504, 2021 Apr.
Article En | MEDLINE | ID: mdl-33611211

BACKGROUND: DNA topoisomerase (Topo) inhibition plays key role in breast cancer treatment. Stephania hainanensis H. S. Lo et Y. Tsoong (S. hainanensis), a Li nationality plant that has abundant aporphine alkaloids, can inhibit Topo. PURPOSE: To identify a dual Topo inhibitor, a deep and systematic study of active aporphine alkaloids in S. hainanensis and their mechanisms of inhibiting breast cancer proliferation and Topo activity are essential. STUDY DESIGN: This study aimed to assess the anti-breast cancer and Topo inhibitory activities of oxocrebanine and explore the underlying mechanisms. METHODS: The growth inhibitory activities of 12 compounds in S. hainanensis were screened by MTT assay in MCF-7, SGC-7901, HepG-2 cells, and compared with the effects on human normal mammary epithelial MCF-10A cells as non cancer control cells. The Topo inhibitory activity was assessed by DNA relaxation and unwinding assays, kDNA decatenation assay and western blot. Cell cycle and autophagy analyses were carried out with flow cytometry and staining. Acridine orange staining and α-tubulin morphology were observed by fluorescence microscopy. Western blot was used to examine microtubule assembly dynamics and the expression levels of key proteins associated with DNA damage, autophagy and mitotic arrest. RESULTS: Oxocrebanine was the anti-breast cancer active alkaloid in S. hainanensis. It exhibited the best inhibitory effect on MCF-7 cells with an IC50 of 16.66 µmol/l, and had only weak effect on the proliferation of MCF-10A cells. Oxocrebanine inhibited Topo I and II α in a cell-free system and in MCF-7 cells. The DNA unwinding assay suggested that oxocrebanine intercalated with DNA as a catalytic inhibitor. Oxocrebanine regulated the levels of Topo I and IIα and DNA damage-related proteins. Oxocrebanine led to the mitotic arrest, and these effects occurred through both p53-dependent and p53-independent pathways. Oxocrebanine induced autophagy, abnormal α-tubulin morphology and stimulated enhanced microtubule dynamics. CONCLUSION: Oxocrebanine was the anti-breast cancer active aporphine alkaloid in S. hainanensis. Oxocrebanine was a Topo I/IIα dual inhibitor, catalytic inhibitor and DNA intercalator. Oxocrebanine caused DNA damage, autophagy, and mitotic arrest in MCF-7 cells. Oxocrebanine also disrupted tubulin polymerization. Accordingly, oxocrebanine held a great potential for development as a novel dual Topo inhibitor for effective breast cancer treatment.


Antineoplastic Agents, Phytogenic/therapeutic use , Aporphines/therapeutic use , Breast Neoplasms/drug therapy , Topoisomerase Inhibitors/therapeutic use , Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Aporphines/chemistry , Aporphines/pharmacology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Mitosis/drug effects , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/pharmacology
10.
J Nucl Med ; 61(11): 1544-1552, 2020 11.
Article En | MEDLINE | ID: mdl-33037092

In recent years, targeted radionuclide therapy (TRT) has emerged as a promising strategy for cancer treatment. In contrast to conventional radiotherapy, TRT delivers ionizing radiation to tumors in a targeted manner, reducing the dose that healthy tissues are exposed to. Existing TRT strategies include the use of 177Lu-DOTATATE, 131I-metaiodobenzylguanidine, Bexxar, and Zevalin, clinically approved agents for the treatment of neuroendocrine tumors, neuroblastoma, and non-Hodgkin lymphoma, respectively. Although promising results have been obtained with these agents, clinical evidence acquired to date suggests that only a small percentage of patients achieves complete response. Consequently, there have been attempts to improve TRT outcomes through combinations with other therapeutic agents; such strategies include administering concurrent TRT and chemotherapy, and the use of TRT with known or putative radiosensitizers such as poly(adenosine diphosphate ribose) polymerase and mammalian-target-of-rapamycin inhibitors. In addition to potentially achieving greater therapeutic effects than the respective monotherapies, these strategies may lead to lower dosages or numbers of cycles required and, in turn, reduce unwanted toxicities. As of now, several clinical trials have been conducted to assess the benefits of TRT-based combination therapies, sometimes despite limited preclinical evidence being available in the public domain to support their use. Although some clinical trials have yielded promising results, others have shown no clear survival benefit from particular combination treatments. Here, we present a comprehensive review of combination strategies with TRT reported in the literature to date and evaluate their therapeutic potential.


Neoplasms/radiotherapy , Cell Cycle/drug effects , Combined Modality Therapy , DNA Damage , DNA Repair/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hedgehog Proteins/antagonists & inhibitors , Humans , Immune Checkpoint Inhibitors/therapeutic use , NAD/metabolism , Octreotide/analogs & derivatives , Octreotide/therapeutic use , Organometallic Compounds/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use
11.
Curr Protein Pept Sci ; 21(11): 1103-1142, 2020.
Article En | MEDLINE | ID: mdl-32951576

Topoisomerases are reported to resolve the topological problems of DNA during several cellular processes, such as DNA replication, transcription, recombination, and chromatin remodeling. Two types of topoisomerases (Topo I and II) accomplish their designated tasks by introducing single- or double-strand breaks within the duplex DNA molecules, and thus maintain the proper structural conditions of DNA to release the topological torsions, which is generated by unwinding of DNA to access coded information, in the course of replication, transcription, and other processes. Both the topoisomerases have been looked at as crucial targets against various types of cancers such as lung, melanoma, breast, and prostate cancers. Conceptually, targeting topoisomerases will disrupt both DNA replication and transcription, thereby leading to inhibition of cell division and consequently stopping the growth of actively dividing cancerous cells. Since the discovery of camptothecin (an alkaloid) as an inhibitor of Topo I in 1958, a number of derivatives of camptothecin were developed as potent inhibitors of Topo I. Two such derivatives of camptothecin, namely, topotecan and irinotecan, have been commonly used as US Food and Drug Administration (FDA) approved drugs against Topo I. Similarly, the first Topo II inhibitor, namely, etoposide, an analogue of podophyllotoxin, was developed in 1966 and got FDA approval as an anti-cancer drug in 1983. Subsequently, several other inhibitors of Topo II, such as doxorubicin, mitoxantrone, and teniposide, were developed. These drugs have been reported to cause accumulation of cytotoxic non-reversible DNA double-strand breaks (cleavable complex). Thus, the present review describes the anticancer potential of plant-derived secondary metabolites belonging to alkaloids, flavonoids and terpenoids directed against topoisomerases. Furthermore, in view of the recent advances made in the field of computer-aided drug design, the present review also discusses the use of computational approaches such as ADMET, molecular docking, molecular dynamics simulation and QSAR to assess and predict the safety, efficacy, potency and identification of these potent anti-cancerous therapeutic molecules.


Antineoplastic Agents, Phytogenic/therapeutic use , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type I/chemistry , DNA, Neoplasm/genetics , Drug Design , Neoplasms/drug therapy , Topoisomerase Inhibitors/therapeutic use , Alkaloids/chemical synthesis , Alkaloids/isolation & purification , Alkaloids/therapeutic use , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/isolation & purification , Biological Products/chemistry , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/chemistry , DNA, Neoplasm/metabolism , Flavonoids/chemical synthesis , Flavonoids/isolation & purification , Flavonoids/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Nucleic Acid Conformation , Quantitative Structure-Activity Relationship , Terpenes/chemical synthesis , Terpenes/isolation & purification , Terpenes/therapeutic use , Topoisomerase Inhibitors/chemical synthesis , Topoisomerase Inhibitors/isolation & purification
12.
Biochem Pharmacol ; 177: 113986, 2020 07.
Article En | MEDLINE | ID: mdl-32330494

Virotherpay is emerging as a promising strategy against cancer, and three oncolytic viruses (OVs) have gained approval in different countries for the treatment of several cancer types. Beyond the capability to selectively infect, replicate and lyse cancer cells, OVs act through a multitude of events, including modification of the tumour micro/macro-environment as well as a complex modulation of the anti-tumour immune response by activation of danger signals and immunogenic cell death pathways. Most OVs show limited effects, depending on the viral platform and the interactions with the host. OVs used as monotherapy only in a minority of patients elicited a full response. Better outcomes were obtained using OVs in combination with other treatments, such as immune therapy or chemotherapy, suggesting that the full potential of OVs can be unleashed in combination with other treatment modalities. Here, we report the main described combination of OVs with conventional chemotherapeutic agents: platinum salts, mitotic inhibitors, anthracyclines and other antibiotics, anti-metabolites, alkylating agents and topoisomerase inhibitors. Additionally, our work provides an overview of OV combination with targeted therapies: histone deacetylase inhibitors, kinase inhibitors, monoclonal antibodies, inhibitors of DNA repair, inhibitors of the proteasome complex and statins that demonstrated enhanced OV anti-neoplastic activity. Although further studies are required to assess the best combinations to translate the results in the clinic, it is clear that combined therapies, acting with complementary mechanisms of action might be useful to target cancer lesions resistant to currently available treatments.


Antibodies, Monoclonal/therapeutic use , Combined Modality Therapy/methods , Immunotherapy/methods , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Alkylating Agents/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Antimitotic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Oncolytic Viruses/immunology , Platinum Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Topoisomerase Inhibitors/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
13.
J Med Chem ; 63(17): 8977-9002, 2020 09 10.
Article En | MEDLINE | ID: mdl-32320239

Histone deacetylases (HDACs) play an important role in regulating target gene expression. They have been highlighted as a novel category of anticancer targets, and their inhibition can induce apoptosis, differentiation, and growth arrest in cancer cells. In view of the fact that HDAC inhibitors and other antitumor agents, such as BET inhibitors, topoisomerase inhibitors, and RTK pathway inhibitors, exert a synergistic effect on cellular processes in cancer cells, the combined inhibition of two targets is regarded as a rational strategy to improve the effectiveness of these single-target drugs for cancer treatment. In this review, we discuss the theoretical basis for designing HDAC-involved dual-target drugs and provide insight into the structure-activity relationships of these dual-target agents.


Antineoplastic Agents/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , DNA Damage/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/chemistry , Humans , Neoplasms/drug therapy , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Kinases/pharmacology , Proteins/antagonists & inhibitors , Proteins/metabolism , Structure-Activity Relationship , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use
14.
EBioMedicine ; 54: 102711, 2020 Apr.
Article En | MEDLINE | ID: mdl-32279056

BACKGROUND: Cystic echinococcosis (CE), a condition caused by the larval stage of the dog tapeworm Echinococcus granulosus sensu stricto, is a globally distributed zoonotic disease. Current treatment options for CE are limited, and an effective and safe anti-echinococcal drug is urgently required. METHODS: Drug repurposing strategy was employed to identify new therapeutic agents against echinococcal cysts. An in vitro protoscolicidal assay along with in vivo murine models was applied in the drug screening. A microinjection procedure was employed to mimic the clinical PAIR (puncture, aspiration, injection and reaspiration) technique to evaluate the potential application of the candidate drug in clinical practice. FINDINGS: We repurposed pyronaridine, an approved antimalarial drug, for the treatment of CE. Following a three-dose intraperitoneal regimen (57 mg/kg, q.d. for 3 days), pyronaridine caused 100% cyst mortality. Oral administration of pyronaridine at 57 mg/kg, q.d. for 30 days significantly reduced the parasitic burden in the pre-infected mice compared with albendazole group (p < 0.001). Using a microinjection of drug into cysts, pyronaridine (200 µM) showed highly effective in term of inhibition of cyst growth (p < 0.05, compared with saline group). Pharmacokinetic analysis revealed that pyronaridine was highly distributed in the liver and lungs, the most affected organs of CE. Function analysis showed that pyronaridine inhibited the activity of topoisomerase I (IC50 = 209.7 ± 1.1 µM). In addition, classical apoptotic hallmarks, including DNA fragmentation and caspase activation, were triggered. INTERPRETATION: Given its approved clinical safety, the repurposing of pyronaridine offers a rapidly translational option for treating CE including PAIR. FUND: National Natural Science Foundation of China and International Cooperation Project of the Qinghai Science and Technology Department.


Antimalarials/therapeutic use , Echinococcosis/drug therapy , Naphthyridines/therapeutic use , Topoisomerase Inhibitors/therapeutic use , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacokinetics , Antimalarials/toxicity , DNA Fragmentation , DNA Topoisomerases, Type I/metabolism , Drug Repositioning , Echinococcus granulosus/drug effects , Echinococcus granulosus/pathogenicity , Female , Liver/metabolism , Liver/parasitology , Lung/metabolism , Lung/parasitology , Mice , Mice, Inbred BALB C , Naphthyridines/administration & dosage , Naphthyridines/pharmacokinetics , Naphthyridines/toxicity , Tissue Distribution , Topoisomerase Inhibitors/administration & dosage , Topoisomerase Inhibitors/pharmacokinetics , Topoisomerase Inhibitors/toxicity
15.
Pharmacol Res ; 151: 104551, 2020 01.
Article En | MEDLINE | ID: mdl-31743776

Topoisomerase enzymes have shown unique roles in replication and transcription. These enzymes which were initially found in Escherichia coli have attracted considerable attention as target molecules for cancer therapy. Nowadays, there are several topoisomerase inhibitors in the market to treat or at least control the progression of cancer. However, significant toxicity, low solubility and poor pharmacokinetic properties have limited their wide application and these characteristics need to be improved. Nano-delivery systems have provided an opportunity to modify the intrinsic properties of molecules and also to transfer the toxic agent to the target tissues. These delivery systems leads to the re-introduction of existing molecules present in the market as novel therapeutic agents with different physicochemical and pharmacokinetic properties. This review focusses on a variety of nano-delivery vehicles used for the improvement of pharmacological properties of topoisomerase inhibitors and thus enabling their potential application as novel drugs in the market.


Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Neoplasms/drug therapy , Topoisomerase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , DNA Topoisomerases/metabolism , Drug Delivery Systems/methods , Humans , Neoplasms/metabolism , Topoisomerase Inhibitors/administration & dosage , Topoisomerase Inhibitors/pharmacology
16.
Future Med Chem ; 11(16): 2193-2203, 2019 08.
Article En | MEDLINE | ID: mdl-31538522

The significant increase in the detection of drug-resistant strains of Mycobacterium tuberculosis caused an urgent need for the discovery new antituberculosis drugs. Development of bioinformatics and computational sciences enabled the progress of new strategies leading to design, discovery and identification of a series of interesting drug candidates. In this short review, we would like to present recently discovered compounds targeting important mycobacterial proteins: DNA topoisomerases and the transcriptional repressor of EthA monooxygenase - EthR.


Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Repressor Proteins/antagonists & inhibitors , Topoisomerase Inhibitors/pharmacology , Tuberculosis/drug therapy , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Bacterial Proteins/metabolism , DNA Topoisomerases/metabolism , Drug Development , Humans , Mycobacterium tuberculosis/metabolism , Repressor Proteins/metabolism , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/therapeutic use , Tuberculosis/microbiology
17.
J Virol ; 93(23)2019 12 01.
Article En | MEDLINE | ID: mdl-31511390

Breast cancer is the second leading cause of cancer-related deaths in women in the United States. Triple-negative breast cancer constitutes a subset of breast cancer that is associated with higher rates of relapse, decreased survival, and limited therapeutic options for patients afflicted with this type of breast cancer. Mammalian orthoreovirus (reovirus) selectively infects and kills transformed cells, and a serotype 3 reovirus is in clinical trials to assess its efficacy as an oncolytic agent against several cancers. It is unclear if reovirus serotypes differentially infect and kill triple-negative breast cancer cells and if reovirus-induced cytotoxicity of breast cancer cells can be enhanced by modulating the activity of host molecules and pathways. Here, we generated reassortant reoviruses by forward genetics with enhanced infective and cytotoxic properties in triple-negative breast cancer cells. From a high-throughput screen of small-molecule inhibitors, we identified topoisomerase inhibitors as a class of drugs that enhance reovirus infectivity and cytotoxicity of triple-negative breast cancer cells. Treatment of triple-negative breast cancer cells with topoisomerase inhibitors activates DNA damage response pathways, and reovirus infection induces robust production of type III, but not type I, interferon (IFN). Although type I and type III IFNs can activate STAT1 and STAT2, triple-negative breast cancer cellular proliferation is only negatively affected by type I IFN. Together, these data show that reassortant viruses with a novel genetic composition generated by forward genetics in combination with topoisomerase inhibitors more efficiently infect and kill triple-negative breast cancer cells.IMPORTANCE Patients afflicted by triple-negative breast cancer have decreased survival and limited therapeutic options. Reovirus infection results in cell death of a variety of cancers, but it is unknown if different reovirus types lead to triple-negative breast cancer cell death. In this study, we generated two novel reoviruses that more efficiently infect and kill triple-negative breast cancer cells. We show that infection in the presence of DNA-damaging agents enhances infection and triple-negative breast cancer cell killing by reovirus. These data suggest that a combination of a genetically engineered oncolytic reovirus and topoisomerase inhibitors may provide a potent therapeutic option for patients afflicted with triple-negative breast cancer.


Apoptosis , Breast Neoplasms/therapy , Oncolytic Virotherapy/methods , Reoviridae/physiology , Topoisomerase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/immunology , Cell Death , Cell Line, Tumor , Cell Survival , DNA Damage , Female , High-Throughput Screening Assays , Humans , Immunity, Innate , Interferons/metabolism , Kinetics , Oncolytic Viruses/physiology , Reoviridae/genetics , Reoviridae Infections/virology , Topoisomerase Inhibitors/therapeutic use , Virus Replication , Interferon Lambda
18.
Nutrients ; 11(6)2019 Jun 25.
Article En | MEDLINE | ID: mdl-31242602

Cancers are one of the leading causes of deaths affecting millions of people around the world, therefore they are currently a major public health problem. The treatment of cancer is based on surgical resection, radiotherapy, chemotherapy or immunotherapy, much of which is often insufficient and cause serious, burdensome and undesirable side effects. For many years, assorted secondary metabolites derived from plants have been used as antitumor agents. Recently, researchers have discovered a large number of new natural substances which can effectively interfere with cancer cells' metabolism. The most famous groups of these compounds are topoisomerase and mitotic inhibitors. The aim of the latest research is to characterize natural compounds found in many common foods, especially by means of their abilities to regulate cell cycle, growth and differentiation, as well as epigenetic modulation. In this paper, we focus on a review of recent discoveries regarding nature-derived anticancer agents.


Antimitotic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Diet , Neoplasms/drug therapy , Topoisomerase Inhibitors/therapeutic use , Animals , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Energy Metabolism/drug effects , Epigenesis, Genetic/drug effects , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
19.
Curr Drug Targets ; 20(1): 70-80, 2019.
Article En | MEDLINE | ID: mdl-29697027

BACKGROUND: The discovery of new chemotherapeutic agents still remains a continuous goal to achieve. DNA polymerases and topoisomerases act in nucleic acids metabolism modulating different processes like replication, mitosis, damage repair, DNA topology and transcription. It has been widely documented that Polymerases serve as molecular targets for antiviral and antitumoral chemotherapy. Furthermore, telomerase is a ribonucleoprotein with exacerbated activity in most of the tumor cell lines, becoming as an emergent target in Cancer treatment. METHODS: We undertook an exhaustive search of bibliographic databases for peer-reviewed research literature related to the last decade. The characteristics of screened bibliography describe structure activity relationships and show the principal moieties involved. This work tries to summarize the investigation about natural and semi-synthetic products with natural origin with the faculty to inhibit key enzymes that play a crucial role in DNA metabolism. RESULTS: Eighty-five data references were included in this review, showing natural products widely distributed throughout the plant kingdom and their bioactive properties such as tumor growing inhibitory effects, and anti-AIDS activity. CONCLUSION: The findings of this review confirm the importance to find new drugs and biologically active natural products, and their potential medicinally useful benefits.


Antiviral Agents/pharmacology , Biological Products/pharmacology , Neoplasms/drug therapy , Nucleic Acid Synthesis Inhibitors/pharmacology , Topoisomerase Inhibitors/pharmacology , Virus Diseases/drug therapy , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Biological Products/chemistry , Biological Products/therapeutic use , DNA/metabolism , DNA Topoisomerases/chemistry , DNA Topoisomerases/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Humans , Molecular Targeted Therapy/methods , Neoplasms/genetics , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/therapeutic use , Structure-Activity Relationship , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/therapeutic use , Virus Diseases/genetics , Virus Diseases/virology
20.
Curr Med Chem ; 26(33): 6074-6106, 2019.
Article En | MEDLINE | ID: mdl-29874992

BACKGROUND: Dual-targeting in cancer treatment by a single drug is an unconventional approach in relation to drug combinations. The rationale for the development of dualtargeting agents is to overcome incomplete efficacy and drug resistance frequently present when applying individual targeting agents. Consequently, -a more favorable outcome of cancer treatment is expected with dual-targeting strategies. METHODS: We reviewed the literature, concentrating on the association between clinically relevant and/or novel dual inhibitors with the potential to modulate multidrug resistant phenotype of cancer cells, particularly the activity of P-glycoprotein. A balanced analysis of content was performed to emphasize the most important findings and optimize the structure of this review. RESULTS: Two-hundred and forty-five papers were included in the review. The introductory part was interpreted by 9 papers. Tyrosine kinase inhibitors' role in the inhibition of Pglycoprotein and chemosensitization was illustrated by 87 papers. The contribution of naturalbased compounds in overcoming multidrug resistance was reviewed using 92 papers, while specific dual inhibitors acting against microtubule assembling and/or topoisomerases were described with 55 papers. Eleven papers gave an insight into a novel and less explored approach with hybrid drugs. Their influence on P-glycoprotein and multidrug resistance was also evaluated. CONCLUSION: These findings bring into focus rational anticancer strategies with dual-targeting agents. Most evaluated synthetic and natural drugs showed a great potential in chemosensitization. Further steps in this direction are needed for the optimization of anticancer treatment.


Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Humans , Polyphenols/chemistry , Polyphenols/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Topoisomerase Inhibitors/therapeutic use , Tubulin Modulators/therapeutic use
...