Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 578
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38612419

RESUMO

Somatostatin receptor ligands (SRLs) with high affinity for somatostatin receptors 2 and 5 (SSTR2 and SSTR5) are poorly efficacious in NF-PitNETs, expressing high levels of SSTR3. ITF2984 is a pan-SSTR ligand with high affinity for SSTR3, able to induce SSTR3 activation and to exert antitumoral activity in the MENX rat model. The aim of this study was to test ITF2984's antiproliferative and proapoptotic effects in NF-PitNET primary cultured cells derived from surgically removed human tumors and to characterize their SSTR expression profile. We treated cells derived from 23 NF-PitNETs with ITF2984, and a subset of them with octreotide, pasireotide (SRLs with high affinity for SSTR2 or 5, respectively), or cabergoline (DRD2 agonist) and we measured cell proliferation and apoptosis. SSTR3, SSTR2, and SSTR5 expression in tumor tissues was analyzed by qRT-PCR and Western blot. We demonstrated that ITF2984 reduced cell proliferation (-40.8 (17.08)%, p < 0.001 vs. basal, n = 19 NF-PitNETs) and increased cell apoptosis (+41.4 (22.1)%, p < 0.001 vs. basal, n = 17 NF-PitNETs) in all tumors tested, whereas the other drugs were only effective in some tumors. In our model, SSTR3 expression levels did not correlate with ITF2984 antiproliferative nor proapoptotic effects. In conclusion, our data support a possible use of ITF2984 in the pharmacological treatment of NF-PitNET.


Assuntos
Antimitóticos , Tumores Neuroendócrinos , Neoplasias Hipofisárias , Humanos , Tumores Neuroendócrinos/tratamento farmacológico , Octreotida/farmacologia , Octreotida/uso terapêutico , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/genética , Receptores de Somatostatina/genética
2.
Bioorg Med Chem Lett ; 105: 129745, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38614151

RESUMO

A series of 8 novel pyridinyl 4-(2-oxoimidazolidin-1-yl)benzenesulfonates (PYRIB-SOs) were designed, prepared and evaluated for their mechanism of action. PYRIB-SOs were found to have antiproliferative activity in the nanomolar to submicromolar range on several breast cancer cell lines. Moreover, subsequent biofunctional assays indicated that the most potent PYRIB-SOs 1-3 act as antimitotics binding to the colchicine-binding site (C-BS) of α, ß-tubulin and that they arrest the cell cycle progression in the G2/M phase. Microtubule immunofluorescence and tubulin polymerisation assay confirm that they disrupt the cytoskeleton through inhibition of tubulin polymerisation as observed with microtubule-destabilising agents. They also show good overall theoretical physicochemical, pharmacokinetic and druglike properties. Overall, these results show that PYRIB-SOs is a new family of promising antimitotics to be further studied in vivo for biopharmaceutical and pharmacodynamic evaluations.


Assuntos
Antimitóticos , Proliferação de Células , Colchicina , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Colchicina/química , Colchicina/metabolismo , Colchicina/farmacologia , Sítios de Ligação , Antimitóticos/farmacologia , Antimitóticos/química , Antimitóticos/síntese química , Relação Estrutura-Atividade , Proliferação de Células/efeitos dos fármacos , Linhagem Celular Tumoral , Benzenossulfonatos/química , Benzenossulfonatos/farmacologia , Benzenossulfonatos/síntese química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Tubulina (Proteína)/metabolismo , Estrutura Molecular , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/síntese química , Piridinas/química , Piridinas/farmacologia , Piridinas/síntese química , Relação Dose-Resposta a Droga
3.
Science ; 383(6690): 1441-1448, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38547292

RESUMO

Mitotic duration is tightly constrained, and extended mitosis is characteristic of problematic cells prone to chromosome missegregation and genomic instability. We show here that mitotic extension leads to the formation of p53-binding protein 1 (53BP1)-ubiquitin-specific protease 28 (USP28)-p53 protein complexes that are transmitted to, and stably retained by, daughter cells. Complexes assembled through a Polo-like kinase 1-dependent mechanism during extended mitosis and elicited a p53 response in G1 that prevented the proliferation of the progeny of cells that experienced an approximately threefold extended mitosis or successive less extended mitoses. The ability to monitor mitotic extension was lost in p53-mutant cancers and some p53-wild-type (p53-WT) cancers, consistent with classification of TP53BP1 and USP28 as tumor suppressors. Cancers retaining the ability to monitor mitotic extension exhibited sensitivity to antimitotic agents.


Assuntos
Proliferação de Células , Mitose , Neoplasias , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina Tiolesterase , Humanos , Proliferação de Células/genética , Instabilidade Genômica , Mitose/efeitos dos fármacos , Mitose/genética , Neoplasias/genética , Neoplasias/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Quinase 1 Polo-Like/metabolismo , Antimitóticos/farmacologia , Resistencia a Medicamentos Antineoplásicos
4.
Int J Mol Sci ; 25(3)2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38338967

RESUMO

Recently, the diarylpentanoid BP-M345 (5) has been identified as a potent in vitro growth inhibitor of cancer cells, with a GI50 value between 0.17 and 0.45 µM, showing low toxicity in non-tumor cells. BP-M345 (5) promotes mitotic arrest by interfering with mitotic spindle assembly, leading to apoptotic cell death. Following on from our previous work, we designed and synthesized a library of BP-M345 (5) analogs and evaluated the cell growth inhibitory activity of three human cancer cell lines within this library in order to perform structure-activity relationship (SAR) studies and to obtain compounds with improved antimitotic effects. Four compounds (7, 9, 13, and 16) were active, and the growth inhibition effects of compounds 7, 13, and 16 were associated with a pronounced arrest in mitosis. These compounds exhibited a similar or even higher mitotic index than BP-M345 (5), with compound 13 displaying the highest antimitotic activity, associated with the interference with mitotic spindle dynamics, inducing spindle collapse and, consequently, prolonged mitotic arrest, culminating in massive cancer cell death by apoptosis.


Assuntos
Antimitóticos , Antineoplásicos , Neoplasias , Humanos , Antimitóticos/farmacologia , Mitose , Proliferação de Células , Ciclo Celular , Fuso Acromático/metabolismo , Neoplasias/metabolismo , Apoptose , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/metabolismo
5.
Cancer Chemother Pharmacol ; 93(5): 427-437, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38226983

RESUMO

PURPOSE: Drug efflux transporter associated multi-drug resistance (MDR) is a potential limitation in the use of taxane chemotherapies for the treatment of metastatic melanoma. ABT-751 is an orally bioavailable microtubule-binding agent capable of overcoming MDR and proposed as an alternative to taxane-based therapies. METHODS: This study compares ABT-751 to taxanes in vitro, utilizing seven melanoma cell line models, publicly available gene expression and drug sensitivity databases, a lung cancer cell line model of MDR drug efflux transporter overexpression (DLKP-A), and drug efflux transporter ATPase assays. RESULTS: Melanoma cell lines exhibit a low but variable protein and RNA expression of drug efflux transporters P-gp, BCRP, and MDR3. Expression of P-gp and MDR3 correlates with sensitivity to taxanes, but not to ABT-751. The anti-proliferative IC50 profile of ABT-751 was higher than the taxanes docetaxel and paclitaxel in the melanoma cell line panel, but fell within clinically achievable parameters. ABT-751 IC50 was not impacted by P-gp-overexpression in DKLP-A cells, which display strong resistance to the P-gp substrate taxanes compared to DLKP parental controls. The addition of ABT-751 to paclitaxel treatment significantly decreased cell proliferation, suggesting some reversal of MDR. ATPase activity assays suggest that ABT-751 is a potential BCRP substrate, with the ability to inhibit P-gp ATPase activity. CONCLUSION: Our study confirms that ABT-751 is active against melanoma cell lines and models of MDR at physiologically relevant concentrations, it inhibits P-gp ATPase activity, and it may be a BCRP and/or MDR3 substrate. ABT-751 warrants further investigation alone or in tandem with other drug efflux transporter inhibitors for hard-to-treat MDR melanoma.


Assuntos
Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Melanoma , Sulfonamidas , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Melanoma/genética , Melanoma/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sulfonamidas/farmacologia , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Taxoides/farmacologia , Proliferação de Células/efeitos dos fármacos , Antimitóticos/farmacologia , Antineoplásicos/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores
6.
Microsc Res Tech ; 87(5): 1031-1043, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38205658

RESUMO

The genus Achyranthes belong to the family Amaranthaceae which constitutes an important group of herbs and shrubs with immense medicinal value. The present research work was conducted to investigate the anticancer potential of Achyranthes aspera L. leaves by focusing on the antioxidant, aniproliferative and antimitotic activities of leaf extracts. Plant extraction was carried out by soxhelt method with different solvents. Phytochemical characterization of the plants extracts using chemical methods identified the presence of cardiac glycosides, saponins, coumarins, proteins, tannins, flavonoids and triterpenes. Alkaloid was present in methanolic and ethanolic extract. High performance liquid chromatography showed presence of different concentration of myricetin, quercetin and kaempferol in different extracts with the highest concentration of myricetin (84.53 µg/mL) in n-butanolic extract. The extracts were then tested for antioxidant activity using 2,2-diphenylpicrylhydrazyl (DPPH) radical scavenging assay by spectrophotometric method. In DPPH radical scavenging assay, antioxidant activity of A. aspera ranged between 79.78 ± 0.034% and 58.63 ± 0.069%. Highest antioxidant activity was observed for methanolic extract and lowest for acetone. Antimitotic activity was determined by using Allium cepa assay in which microscopic investigation was carried out to observe normal and abnormal phases of mitosis. In this assay, n-butanolic extract had highest antimitotic activity with minimum mitotic index at 2 mg/mL (57 ± 0.0351%). The plant extracts also caused chromosomal and mitotic aberrations which were clearly observed under 40× and 100× magnification of compound microscope. Antiproliferative activity was determined by using yeast cell model in which light microscope with hemocytometer was used for cell counting. In case of Antiproliferative activity, the ethyl acetate extract of A. aspera had highest antiproliferative activity with lowest cell viability (22.14 ± 0.076%) at highest extract concentration (2 mg/mL) while methanol extract of A. aspera had highest antiproliferative activity with lower cell viability (24.24 ± 0.057%) at lowest extract concentration (0.25 mg/mL). The results of the study indicated that the leaves extract of A. aspera have strong potential to be used as a source of anti-cancer agent. RESEARCH HIGHLIGHTS: Achyranthes aspera L. leaves have various phytochemicals which contribute to its medicinal properties Various extracts of the leaves of A. aspera L. possess antioxidant, antimitotic and antiproliferative potential The results of the study indicated that the leaves extract of A. aspera have strong potential to be used as a source of anti-cancer agent.


Assuntos
Achyranthes , Antimitóticos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Antioxidantes/farmacologia , Achyranthes/química , Microscopia , Plantas , Metanol , Análise Espectral , Folhas de Planta
7.
J Evid Based Med ; 17(1): 37-53, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38243639

RESUMO

BACKGROUND: Plantar warts are common infectious cutaneous growths causing severe physiological and psychological discomforts in patients and heaving global financial burdens. However, paucity of clear-cut guidelines for plantar warts, selecting appropriate treatments for plantar warts remains challenging. The objective of the study is to evaluate the efficacy and safety of common treatments for plantar warts. METHODS: PubMed, EMbase, and The Cochrane Library were searched from inception to March 1, 2023 for randomized controlled trials (RCTs) of plantar warts. The primary outcome (complete response) and secondary outcome (recurrence and pain) were extracted and combined using Bayesian network meta-analysis (NMA) with random-effect and fixed-effect models. RESULTS: Totally, 33 RCTs were included in the systematic review and quantitative NMA. In NMA of complete response, topical application of 1% cantharidin, 20% podophylotoxin, 30% salicylic acid (CPS), microneedles plus bleomycin (MNB), and intralesional bleomycin injection (INB) were the only three treatments significantly superior to no treatment (NT) and CPS was of the highest possibility to be the top-ranked treatment (SUCRA = 0.9363). However, traditional warts treatments, salicylic acid (SA) and cryotherapy were not superior to NT. CONCLUSIONS: The NMA has produced evidence for using CPS, MNB, and INB, which are all topical antimitotic treatments, to improve the management of plantar warts. The classic treatment modalities for plantar warts, including SA and cryotherapy, may play a less important role in the clinical practice of plantar warts.


Assuntos
Antimitóticos , Verrugas , Humanos , Antimitóticos/uso terapêutico , Metanálise em Rede , Ensaios Clínicos Controlados Aleatórios como Assunto , Verrugas/tratamento farmacológico , Ácido Salicílico , Bleomicina/uso terapêutico , Resultado do Tratamento
8.
Sci Rep ; 14(1): 912, 2024 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-38195628

RESUMO

This study aimed at encapsulation of commonly administered, highly cytotoxic anticancer drug Docetaxel (DTX) in camel milk fat globule-derived liposomes for delivery in triple negative breast cancer cells. Prior to liposomal encapsulation of drug, in silico analysis of Docetaxel was done to predict off target binding associated toxicities in different organs. For this purpose, the ADMET Predictor (TM) Cloud version 10.4.0.5, 64-bit, was utilized to simulate Docetaxel's pharmacokinetic and physicochemical parameters. Freshly milked camel milk was bought from local market, from two breeds Brella and Marecha, in suburbs of Islamabad. After extraction of MFGM-derived liposomes from camel milk, docetaxel was loaded into liposomes by thin film hydration method. The physiochemical properties of liposomes were analyzed by SEM, FTIR and Zeta analysis. The results from SEM showed that empty liposomes (Lp-CM-ChT80) had spherical morphology while DTX loaded liposomes (Lp-CM-ChT80-DTX) exhibited rectangular shape, FTIR revealed the presence of characteristic functional groups which confirmed the successful encapsulation of DTX. Zeta analysis showed that Lp-CM-ChT80-DTX had size of 836.6 nm with PDI of 0.088 and zeta potential of - 18.7 mV. The encapsulation efficiency of Lp-CM-ChT80 turned out to be 25% while in vitro release assay showed slow release of DTX from liposomes as compared to pure DTX using dialysis membrane. The in vitro anticancer activity was analyzed by cell morphology analysis and MTT cytotoxicity assay using different concentrations 80 µg/ml, 120 µg/ml and 180 µg/ml of Lp-CM-ChT80-DTX on MDA-MB-231 cells. The results showed cytotoxic effects increased in time and dose dependent manner, marked by rounding, shrinkage and aggregation of cells. MTT cytotoxicity assay showed that empty liposomes Lp-CM-ChT80 did not have cytotoxic effect while Lp-CM-ChT80-DTX showed highest cytotoxic potential of 60.2% at 180 µg/ml. Stability analysis showed that liposomes were stable till 24 h in solution form at 4 °C.


Assuntos
Antimitóticos , Neoplasias de Mama Triplo Negativas , Animais , Humanos , Docetaxel , Camelus , Lipossomos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Preparações de Ação Retardada , Diálise Renal
9.
Mol Cancer Ther ; 23(2): 235-247, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37816248

RESUMO

E7130 is a novel anticancer agent created from total synthetic study of the natural compound norhalichondrin B. In addition to inhibiting microtubule dynamics, E7130 also ameliorates tumor-promoting aspects of the tumor microenvironment (TME) by suppressing cancer-associated fibroblasts (CAF) and promoting remodeling of tumor vasculature. Here, we demonstrate TME amelioration by E7130 using multi-imaging modalities, including multiplexed mass cytometry [cytometry by time-of-flight (CyTOF)] analysis, multiplex IHC analysis, and MRI. Experimental solid tumors characterized by large numbers of CAFs in TME were treated with E7130. E7130 suppressed LAP-TGFß1 production, a precursor of TGFß1, in CAFs but not in cancer cells; an effect that was accompanied by a reduction of circulating TGFß1 in plasma. To our best knowledge, this is the first report to show a reduction of TGFß1 production in TME. Furthermore, multiplex IHC analysis revealed reduced cellularity and increased TUNEL-positive apoptotic cells in E7130-treated xenografts. Increased microvessel density (MVD) and collagen IV (Col IV), an extracellular matrix (ECM) component associated with endothelial cells, were also observed in the TME, and plasma Col IV levels were also increased by E7130 treatment. MRI revealed increased accumulation of a contrast agent in xenografts. Moreover, diffusion-weighted MRI after E7130 treatment indicated reduction of tumor cellularity and interstitial fluid pressure. Overall, our findings strongly support the mechanism of action that E7130 alters the TME in therapeutically beneficial ways. Importantly, from a translational perspective, our data demonstrated MRI as a noninvasive biomarker to detect TME amelioration by E7130, supported by consistent changes in plasma biomarkers.


Assuntos
Antimitóticos , Fibroblastos Associados a Câncer , Neoplasias Experimentais , Neoplasias , Animais , Humanos , Fibroblastos Associados a Câncer/patologia , Remodelação Vascular , Microambiente Tumoral , Células Endoteliais/patologia , Neoplasias/tratamento farmacológico , Antimitóticos/farmacologia
10.
Int J Mol Sci ; 24(24)2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38139302

RESUMO

Antimitotic agents are one of the more successful types of anticancer drugs, but they suffer from toxicity and resistance. The application of approved drugs to new indications (i.e., drug repurposing) is a promising strategy for the development of new drugs. It relies on finding pattern similarities: drug effects to other drugs or conditions, similar toxicities, or structural similarity. Here, we recursively searched a database of approved drugs for structural similarity to several antimitotic agents binding to a specific site of tubulin, with the expectation of finding structures that could fit in it. These searches repeatedly retrieved frentizole, an approved nontoxic anti-inflammatory drug, thus indicating that it might behave as an antimitotic drug devoid of the undesired toxic effects. We also show that the usual repurposing approach to searching for targets of frentizole failed in most cases to find such a relationship. We synthesized frentizole and a series of analogs to assay them as antimitotic agents and found antiproliferative activity against HeLa tumor cells, inhibition of microtubule formation within cells, and arrest at the G2/M phases of the cell cycle, phenotypes that agree with binding to tubulin as the mechanism of action. The docking studies suggest binding at the colchicine site in different modes. These results support the repurposing of frentizole for cancer treatment, especially for glioblastoma.


Assuntos
Antimitóticos , Antineoplásicos , Antimitóticos/farmacologia , Tubulina (Proteína)/metabolismo , Linhagem Celular Tumoral , Relação Estrutura-Atividade , Colchicina/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Moduladores de Tubulina/química , Ensaios de Seleção de Medicamentos Antitumorais , Proliferação de Células , Sítios de Ligação
11.
Int J Mol Sci ; 24(21)2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37958555

RESUMO

MYCN amplification occurs in approximately 20-30% of neuroblastoma patients and correlates with poor prognosis. The TH-MYCN transgenic mouse model mimics the development of human high-risk neuroblastoma and provides strong evidence for the oncogenic function of MYCN. In this study, we identified mitotic dysregulation as a hallmark of tumor initiation in the pre-cancerous ganglia from TH-MYCN mice that persists through tumor progression. Single-cell quantitative-PCR of coeliac ganglia from 10-day-old TH-MYCN mice revealed overexpression of mitotic genes in a subpopulation of premalignant neuroblasts at a level similar to single cells derived from established tumors. Prophylactic treatment using antimitotic agents barasertib and vincristine significantly delayed the onset of tumor formation, reduced pre-malignant neuroblast hyperplasia, and prolonged survival in TH-MYCN mice. Analysis of human neuroblastoma tumor cohorts showed a strong correlation between dysregulated mitosis and features of MYCN amplification, such as MYC(N) transcriptional activity, poor overall survival, and other clinical predictors of aggressive disease. To explore the therapeutic potential of targeting mitotic dysregulation, we showed that genetic and chemical inhibition of mitosis led to selective cell death in neuroblastoma cell lines with MYCN over-expression. Moreover, combination therapy with antimitotic compounds and BCL2 inhibitors exploited mitotic stress induced by antimitotics and was synergistically toxic to neuroblastoma cell lines. These results collectively suggest that mitotic dysregulation is a key component of tumorigenesis in early neuroblasts, which can be inhibited by the combination of antimitotic compounds and pro-apoptotic compounds in MYCN-driven neuroblastoma.


Assuntos
Antimitóticos , Neuroblastoma , Humanos , Camundongos , Animais , Proteína Proto-Oncogênica N-Myc/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , Linhagem Celular Tumoral , Camundongos Transgênicos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Neuroblastoma/patologia , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica
12.
Bioorg Chem ; 140: 106820, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37672952

RESUMO

4-(3-Alkyl-2-oxoimidazolidin-1-yl)-N-phenylbenzenesulfonamides (PAIB-SAs) are members of a new family of prodrugs bioactivated by cytochrome P450 1A1 (CYP1A1) in breast cancer cells into their potent 4-(2-oxoimidazolidin-1-yl)-N-phenylbenzenesulfonamide metabolites (PIB-SAs). One of the predominant problems for the galenic formulation and administration of PAIB-SAs in animal studies is their poor hydrosolubility. To circumvent that difficulty, we report the design, the synthesis, the chemical characterization, the evaluation of the aqueous solubility, the antiproliferative activity and the mechanism of action of 18 new Na+, K+ and Li+ salts of PAIB-SAs. Our results evidenced that the latter exhibited highly selective antiproliferative activity toward MCF7 and MDA-MB-468 breast cancer cells expressing endogenously CYP1A1 compared to insensitive MDA-MB-231 and HaCaT cells. Moreover, PAIB-SA salts 1-18 are significantly more hydrosoluble (3.9-9.4 mg/mL) than their neutral counterparts (< 0.0001 mg/mL). In addition, the most potent PAIB-SA salts 1-3 and 10-12 arrested the cell cycle progression in the G2/M phase and disrupted the cytoskeleton's dynamic assembly. Finally, PAIB-SA salts are N-dealkylated by CYP1A1 into their corresponding PIB-SA metabolites, which are potent antimitotics. In summary, our results show that our water-soluble PAIB-SA salts, notably the sodium salts, still exhibit potent antiproliferative efficacy and remain prone to CYP1A1 bioactivation. In addition, these PAIB-SA salts will allow the development of galenic formulations suitable for further biopharmaceutical and pharmacodynamic studies.


Assuntos
Antimitóticos , Neoplasias da Mama , Citocromo P-450 CYP1A1 , Pró-Fármacos , Animais , Antimitóticos/química , Antimitóticos/farmacocinética , Antimitóticos/farmacologia , Citocromo P-450 CYP1A1/metabolismo , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Sais , Humanos
13.
Life Sci ; 330: 122033, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37598976

RESUMO

Aim Overcoming resistance to apoptosis and antimitotic chemotherapy is crucial for effective treatment of lung cancer. Diosgenin (DG), a promising phytochemical, can regulate various molecular pathways implicated in tumor formation and progression. However, the precise biological activity of DG in lung cancer remains unclear. This study aimed to investigate the antiproliferative activity of DG in NCI-H460 lung carcinoma cells to explore the underlying antimitotic mechanisms and alternative cell death pathways. MATERIALS AND METHODS: In a 2D culture system, we analyzed cell viability, multinucleated cell frequency, cell concentration, cell cycle changes, cell death induction, intracellular reactive oxygen species (ROS) production, and nuclear DNA damage, particularly in relation to target gene expression. We also evaluated the antiproliferative activity of DG in a 3D culture system of spheroids, assessing volume changes, cell death induction, and inhibition of proliferation recovery and clonogenic growth. KEY FINDINGS: DG reduced cell viability and concentration while increasing the frequency of cells with multiple nuclei, particularly binucleated cells resulting from daughter cell fusion. This effect was associated with genes involved in cytokinesis regulation (RAB35, OCRL, BIRC5, and AURKB). Additionally, DG-induced cell death was linked to necroptosis, as evidenced by increased intracellular ROS production and RIPK3, MLKL, TRAF2, and HSPA5 gene expression. In tumor spheroids, DG increased spheroid volume, induced cell death, and inhibited proliferation recovery and clonogenic growth. SIGNIFICANCE: Our study provides new insights into the biological activities of DG in lung cancer cells, contributing to the development of novel oncological therapies.


Assuntos
Antimitóticos , Diosgenina , Neoplasias Pulmonares , Humanos , Citocinese , Necroptose , Espécies Reativas de Oxigênio , Neoplasias Pulmonares/tratamento farmacológico , Divisão Celular , Diosgenina/farmacologia , Pulmão
14.
Dalton Trans ; 52(34): 11859-11874, 2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37464882

RESUMO

Antimitotic agents are among the most important drugs used in anticancer therapy. Kinesin spindle protein (KSP) was proposed as a promising target for new antimitotic drugs. Herein, we report the synthesis of Ru, Os, Rh, and Ir half-sandwich complexes with the KSP inhibitor ispinesib and its (S)-enantiomer. Conjugation of the organometallic moiety with ispinesib and its (S)-enantiomer resulted in a significantly increased cytotoxicity of up to 5.6-fold compared to the parent compounds, with IC50 values in the nanomolar range. The most active derivatives were the ispinesib Ru and Rh conjugates which were able to generate reactive oxygen species (ROS), which may at least partially explain their high cytotoxicity. At the same time, the Os and Ir derivatives acted as KSP inhibitors with no effects on ROS generation.


Assuntos
Antimitóticos , Antineoplásicos , Compostos Organometálicos , Antimitóticos/farmacologia , Espécies Reativas de Oxigênio , Quinazolinas , Benzamidas/metabolismo , Benzamidas/farmacologia , Compostos Organometálicos/farmacologia
15.
Curr Drug Targets ; 24(9): 698-717, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37424350

RESUMO

BACKGROUND: Tropolone and thailandepsin B are naturally occurring substances that are primarily isolated from fungi and plants, although they can also be found in certain bacteria. Tropolones belong to an important class of aromatic compounds with a seven-membered nonbenzenoid ring structure. Thailandepsins are a group of natural products that were initially discovered in the culture broth of the Gram-negative bacterium Burkholderia thailandensis. Tropolonebased structures have been identified in over 200 natural compounds, ranging from simple tropolone derivatives to complex multicyclic systems like pycnidione and pyrerubrine A. These natural compounds exhibit a diverse range of pharmacological effects, including antibacterial, antifungal, insecticidal, phytotoxic, anti-inflammatory, antimitotic, anti-diabetic, enzyme inhibitory, anticancer, cytoprotective, and ROS scavenging properties. It is worth noting that thujaplicane, a compound similar to tropolone, displays all of the listed biological activities except for antimitotic action, which has only been observed in one natural tropolone compound, colchicine. Tropolone can be synthesized from commercially available seven-membered rings or derived through various cyclization and cycloaddition reactions. Thailandepsin B, on the other hand, can be synthesized by macro-lactonization of the corresponding secoacid, followed by the formation of internal disulfide bonds. It is important to mention that thailandepsin B exhibits different selective inhibition profiles compared to FK228. OBJECTIVE: We investigated the HDAC inhibitory activity of the Tropolones and Thailandepsin B and discussed the biosynthesis of the naturally occurring compounds and their synthetic scheme. RESULTS AND CONCLUSION: It has been observed that Tropolone derivatives act as isoenzyme-selective inhibitors of proven anticancer drug targets, histone deacetylases (HDACs). Some monosubstituted tropolones show remarkable levels of selectivity for HDAC2 and strongly inhibit the growth of T-lymphocyte cell lines. And Thailandepsins have different selective inhibition profiles than FK228. They exhibit comparable inhibitory activities to FK228 against human HDAC1, HDAC2, HDAC3, HDAC6, HDAC7, and HDAC9, but less potent inhibitory activities than FK228 toward HDAC4 and HDAC8, the latter of which may be useful. Thailandepsins possess potent cytotoxic activities toward some types of cell lines.


Assuntos
Antimitóticos , Antineoplásicos , Humanos , Inibidores de Histona Desacetilases/farmacologia , Tropolona/farmacologia , Tropolona/química , Antineoplásicos/química , Histona Desacetilases , Proteínas Repressoras
16.
Cell Biochem Biophys ; 81(2): 269-283, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37233844

RESUMO

The tubulin-microtubule system is a major target for a variety of small molecules which can interfere in cell cycle progression. Therefore, it serves as a prospective to control the incessant division of cancer cells. To identify novel inhibitors of the tubulin-microtubule system, a group of estrogen derivatives has been tested with tubulin as a target since literature surveys portray coveted behaviour from the same. Out of them, ß-Estradiol-6-one 6- (O-carboxy methyl Oxime) abbreviated as Oxime, disrupts the cytoskeleton network and induces apoptosis with nuclei fragmentation. It has been revealed from the work that Oxime targets the colchicine binding site and binds tubulin in an entropy-driven manner. This suggests that structural variation might play a key role in modulating the anti-mitotic role of estrogen derivatives. Our work reveals that Oxime might serve as a lead molecule to nurture anti-cancer research, having the potential for recovery of the vast cancer population.


Assuntos
Antimitóticos , Antineoplásicos , Tubulina (Proteína)/química , Antimitóticos/metabolismo , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo , Mitose , Estudos Prospectivos , Microtúbulos/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Sítios de Ligação , Estrogênios/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/metabolismo
17.
J Med Chem ; 66(4): 2477-2497, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36780426

RESUMO

Phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) are a new family of antimitotic prodrugs bioactivated in breast cancer cells expressing CYP1A1. In this study, we report that the 14C-labeled prototypical PAIB-SO [14C]CEU-818 and its antimitotic counterpart [14C]CEU-602 are distributed in whole mouse body and they show a short half-life in mice. To circumvent this limitation, we evaluated the effect of the homologation of the alkyl side chain of the imidazolidin-2-one moiety of PAIB-SOs. Our studies evidence that PAIB-SOs bearing an n-pentyl side chain exhibit antiproliferative activity in the nanomolar-to-low-micromolar range and a high selectivity toward CYP1A1-positive breast cancer cells. Moreover, the most potent n-pentyl PAIB-SOs were significantly more stable toward rodent liver microsomes. In addition, PAIB-SOs 10 and 14 show significant antitumor activity and low toxicity in chorioallantoic membrane (CAM) assay. Our study confirms that homologation is a suitable approach to improve the rodent hepatic stability of PAIB-SOs.


Assuntos
Antimitóticos , Neoplasias , Pró-Fármacos , Camundongos , Animais , Antimitóticos/química , Pró-Fármacos/química , Citocromo P-450 CYP1A1 , Roedores , Microssomos Hepáticos , Benzenossulfonatos/química
18.
Anticancer Drugs ; 34(2): 281-289, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36730487

RESUMO

BACKGROUND: Prolonging the time which plasma concentrations of antimitotic drugs, such as the taxanes, exceed cytotoxic threshold levels may be beneficial for their efficacy. Orally administered docetaxel offers an undemanding approach to optimize such time above threshold plasma concentrations (t C>threshold ). METHODS: A nonsystematic literature screen was performed to identify studies reporting in-vitro half-maximal inhibitory concentration (IC 50 ) values for docetaxel. Pharmacokinetics of intravenously (i.v.) docetaxel (75 mg/m 2 ) and orally administered docetaxel (ModraDoc006) co-administered with ritonavir (r) given twice daily (30 + 20 mg concomitant with 100 mg ritonavir bis in die) were simulated using previously developed population models. T C>threshold was calculated for a range of relevant thresholds in terms of in-vitro cytotoxicity and plasma concentrations achieved after i.v. and oral administration of docetaxel. A published tumor growth inhibition model for i.v. docetaxel was adapted to predict the effect of attainment of time above threshold levels on tumor dynamics. RESULTS: Identified studies reported a wide range of in vitro IC 50 values [median 0.04 µmol/L, interquartile range (IQR): 0.0046-0.62]. At cytotoxic thresholds <0.078 µmol/L oral docetaxel shows up to ~7.5-fold longer t C>threshold within each 3-week cycle for a median patient compared to i.v.. Simulations of tumor dynamics showed the increased relative potential of oral docetaxel for inhibition of tumor growth at thresholds of 0.075, 0.05 and 0.005 µmol/L. CONCLUSION: ModraDoc006/r is superior to i.v. docetaxel 75 mg/m 2 in terms of median time above cytotoxic threshold levels <0.078 µmol/L. This may indicate superior cytotoxicity and inhibition of tumor growth compared to i.v. administration for relatively docetaxel-sensitive tumors.


Assuntos
Antimitóticos , Antineoplásicos , Neoplasias da Mama , Humanos , Feminino , Docetaxel , Neoplasias da Mama/patologia , Ritonavir , Antineoplásicos/farmacologia , Taxoides , Administração Oral
19.
Mol Cancer Ther ; 22(1): 12-24, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36279567

RESUMO

Innate and adaptive resistance to cancer therapies, such as chemotherapies, molecularly targeted therapies, and immune-modulating therapies, is a major issue in clinical practice. Subpopulations of tumor cells expressing the receptor tyrosine kinase AXL become enriched after treatment with antimitotic drugs, causing tumor relapse. Elevated AXL expression is closely associated with drug resistance in clinical samples, suggesting that AXL plays a pivotal role in drug resistance. Although several molecules with AXL inhibitory activity have been developed, none have sufficient activity and selectivity to be clinically effective when administered in combination with a cancer therapy. Here, we report a novel small molecule, ER-851, which is a potent and highly selective AXL inhibitor. To investigate resistance mechanisms and identify driving molecules, we conducted a comprehensive gene expression analysis of chemoresistant tumor cells in mouse xenograft models of genetically engineered human lung cancer and human triple-negative breast cancer. Consistent with the effect of AXL knockdown, cotreatment of ER-851 and antimitotic drugs produced an antitumor effect and prolonged relapse-free survival in the mouse xenograft model of human triple-negative breast cancer. Importantly, when orally administered to BALB/c mice, this compound did not induce retinal toxicity, a known side effect of chronic MER inhibition. Together, these data strongly suggest that AXL is a therapeutic target for overcoming drug resistance and that ER-851 is a promising candidate therapeutic agent for use against AXL-expressing antimitotic-resistant tumors.


Assuntos
Antimitóticos , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Receptor Tirosina Quinase Axl , Antimitóticos/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral , Inibidores de Proteínas Quinases/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Pharm Pharmacol ; 75(4): 445-465, 2023 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-36334086

RESUMO

OBJECTIVES: Haploid germ cell-specific nuclear protein kinase (Haspin) is a serine/threonine kinase as an atypical kinase, which is structurally distinct from conventional protein kinases. KEY FINDINGS: Functionally, Haspin is involved in important cell cycle progression, particularly in critical mitosis regulating centromeric sister chromatid cohesion during prophase and prometaphase, and subsequently ensuring proper chromosome alignment during metaphase and the normal chromosome segregation during anaphase. However, increasing evidence has demonstrated that Haspin is significantly upregulated in a variety of cancer cells in addition to normal proliferating somatic cells. Its knockdown or small molecule inhibition could prevent cancer cell growth and induce apoptosis by disrupting the regular mitotic progression. Given the specificity of its expressed tissues or cells and the uniqueness of its current known substrate, Haspin can be a promising target against cancer. Consequently, selective synthetic and natural inhibitors of Haspin have been widely developed to determine their inhibitory power for various cancer cells in vivo and in vitro. SUMMARY: Here our perspective includes a comprehensive review of the roles and structure of Haspin, its relatively potent and selective inhibitors and Haspin's preliminary studies in a variety of cancers.


Assuntos
Antimitóticos , Neoplasias , Humanos , Fosforilação , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Serina-Treonina Quinases/metabolismo , Mitose , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...