Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharm Sci ; 113(1): 214-227, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38498417

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is often chemotherapy-resistant, and novel drug combinations would fill an unmet clinical need. Previously we reported synergistic cytotoxic effects of gemcitabine and trabectedin on pancreatic cancer cells, but underlying protein-level interaction mechanisms remained unclear. We employed a reliable, sensitive, comprehensive, quantitative, high-throughput IonStar proteomic workflow to investigate the time course of gemcitabine and trabectedin effects, alone and combined, upon pancreatic cancer cells. MiaPaCa-2 cells were incubated with vehicle (controls), gemcitabine, trabectedin, and their combinations over 72 hours. Samples were collected at intervals and analyzed using the label-free IonStar liquid chromatography-mass spectrometry (LC-MS/MS) workflow to provide temporal quantification of protein expression for 4,829 proteins in four experimental groups. To characterize diverse signal transduction pathways, a comprehensive systems pharmacodynamic (SPD) model was developed. The analysis is presented in two parts. Here, Part I describes drug responses in cancer cell growth and migration pathways included in the full model: receptor tyrosine kinase- (RTK), integrin-, G-protein coupled receptor- (GPCR), and calcium-signaling pathways. The developed model revealed multiple underlying mechanisms of drug actions, provides insight into the basis of drug interaction synergism, and offers a scientific rationale for potential drug combination strategies.


Asunto(s)
Gemcitabina , Neoplasias Pancreáticas , Humanos , Trabectedina/farmacología , Desoxicitidina/farmacología , Proteómica , Cromatografía Liquida , Línea Celular Tumoral , Espectrometría de Masas en Tándem , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal
2.
Biomed Pharmacother ; 172: 116296, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38382330

RESUMEN

Ovarian cancer (OC) is the fifth most frequent cause of cancer-related death in women. Chemotherapy agent trabectedin, affecting cancer cells and tumor microenvironment, has been approved for the treatment of relapsed platinum-sensitive OC patients. CCR5-antagonist maraviroc inhibits tumor growth, metastasis, and enhances the antitumoral activity of DNA-damaging drugs. Here, we found that OC cells expressed CCR5 receptor but did not secret CCR5-ligands. Maraviroc treatment did not affect OC cell viability, but strongly potentiated the antiproliferative activity, apoptosis induction, cell cycle blockage, DNA damage, and ROS formation by trabectedin. In A2780cis cisplatin-resistant cells, the cross-resistance to trabectedin was overcame by the combination with maraviroc. Maraviroc enhanced trabectedin cytotoxicity in OC 3Dimensional spheroids and THP-1-monocytes. Both maraviroc and trabectedin interact with drug efflux pump MDR1/P-gp, overexpressed in recurrent OC patients. Maraviroc increased trabectedin intracellular accumulation and the MDR1-inhibitor verapamil, like maraviroc, increased trabectedin cytotoxicity. In OC tumor xenografts the combination with maraviroc further reduced tumor growth, angiogenesis, and monocyte infiltration by trabectedin. In conclusion, this study offers a preclinical rationale for the use of maraviroc as new option to improve trabectedin activity in relapsed chemoresistant OC patients.


Asunto(s)
Neoplasias Ováricas , Microambiente Tumoral , Humanos , Femenino , Maraviroc/farmacología , Trabectedina/farmacología , Recurrencia Local de Neoplasia , Neoplasias Ováricas/tratamiento farmacológico , Carcinoma Epitelial de Ovario
3.
Int J Mol Sci ; 25(4)2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38396735

RESUMEN

The in-silico strategy of identifying novel uses for already existing drugs, known as drug repositioning, has enhanced drug discovery. Previous studies have shown a positive correlation between expression changes induced by the anticancer agent trabectedin and those caused by irinotecan, a topoisomerase I inhibitor. Leveraging the availability of transcriptional datasets, we developed a general in-silico drug-repositioning approach that we applied to investigate novel trabectedin synergisms. We set a workflow allowing the identification of genes selectively modulated by a drug and possible novel drug interactions. To show its effectiveness, we selected trabectedin as a case-study drug. We retrieved eight transcriptional cancer datasets including controls and samples treated with trabectedin or its analog lurbinectedin. We compared gene signature associated with each dataset to the 476,251 signatures from the Connectivity Map database. The most significant connections referred to mitomycin-c, topoisomerase II inhibitors, a PKC inhibitor, a Chk1 inhibitor, an antifungal agent, and an antagonist of the glutamate receptor. Genes coherently modulated by the drugs were involved in cell cycle, PPARalpha, and Rho GTPases pathways. Our in-silico approach for drug synergism identification showed that trabectedin modulates specific pathways that are shared with other drugs, suggesting possible synergisms.


Asunto(s)
Antineoplásicos , Tetrahidroisoquinolinas , Trabectedina/farmacología , Trabectedina/uso terapéutico , Tetrahidroisoquinolinas/farmacología , Dioxoles/farmacología , Sinergismo Farmacológico
4.
Bone ; 179: 116983, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38013019

RESUMEN

Stress fractures occur as a result of repeated mechanical stress on bone and are commonly found in the load-bearing lower extremities. Macrophages are key players in the immune system and play an important role in bone remodeling and fracture healing. However, the role of macrophages in stress fractures has not been adequately addressed. We hypothesize that macrophage infiltration into a stress fracture callus site promotes bone healing. To test this, a unilateral stress fracture induction model was employed in which the murine ulna of four-month-old, C57BL/6 J male mice was repeatedly loaded with a pre-determined force until the bone was displaced a distance below the threshold for complete fracture. Mice were treated daily with parathyroid hormone (PTH, 50 µg/kg/day) starting two days before injury and continued until 24 h before euthanasia either four or six days after injury, or treated with trabectedin (0.15 mg/kg) on the day of stress fracture and euthanized three or seven days after injury. These treatments were used due to their established effects on macrophages. While macrophages have been implicated in the anabolic effects of PTH, trabectedin, an FDA approved chemotherapeutic, compromises macrophage function and reduces bone mass. At three- and four-days post injury, callus macrophage numbers were analyzed histologically. There was a significant increase in macrophages with PTH treatment compared to vehicle in the callus site. By one week of healing, treatments differentially affected the bony callus as analyzed by microcomputed tomography. PTH enhanced callus bone volume. Conversely, callus bone volume was decreased with trabectedin treatment. Interestingly, concurrent treatment with PTH and trabectedin rescued the reduction observed in the callus with trabectedin treatment alone. This study reports on the key involvement of macrophages during stress fracture healing. Given these observed outcomes on macrophage physiology and bone healing, these findings may be important for patients actively receiving either of these FDA-approved therapeutics.


Asunto(s)
Fracturas por Estrés , Hormona Paratiroidea , Humanos , Masculino , Ratones , Animales , Lactante , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/uso terapéutico , Trabectedina/farmacología , Fracturas por Estrés/tratamiento farmacológico , Fracturas por Estrés/patología , Microtomografía por Rayos X/métodos , Ratones Endogámicos C57BL , Callo Óseo/patología , Curación de Fractura , Macrófagos
5.
J Pharm Sci ; 113(1): 235-245, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37918792

RESUMEN

Despite decades of research efforts, pancreatic adenocarcinoma (PDAC) continues to present a formidable clinical challenge, demanding innovative therapeutic approaches. In a prior study, we reported the synergistic cytotoxic effects of gemcitabine and trabectedin on pancreatic cancer cells. To investigate potential mechanisms underlying this synergistic pharmacodynamic interaction, liquid chromatography-mass spectrometry-based proteomic analysis was performed, and a systems pharmacodynamics model (SPD) was developed to capture pancreatic cancer cell responses to gemcitabine and trabectedin, alone and combined, at the proteome level. Companion report Part I describes the proteomic workflow and drug effects on the upstream portion of the SPD model related to cell growth and migration, specifically the RTK-, integrin-, GPCR-, and calcium-signaling pathways. This report presents Part II of the SPD model. Here we describe drug effects on pathways associated with cell cycle, DNA damage response (DDR), and apoptosis, and provide insights into underlying mechanisms. Drug combination effects on protein changes in the cell cycle- and apoptosis pathways contribute to the synergistic effects observed between gemcitabine and trabectedin. The SPD model was subsequently incorporated into our previously-established cell cycle model, forming a comprehensive, multi-scale quantification platform for evaluating drug effects across multiple scales, spanning the proteomic-, cellular-, and subcellular levels. This approach provides a quantitative mechanistic framework for evaluating drug-drug interactions in combination chemotherapy, and could potentially serve as a tool to predict combinatorial efficacy and assist in target selection.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Gemcitabina , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Trabectedina/farmacología , Trabectedina/uso terapéutico , Desoxicitidina/farmacología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Proteómica , Línea Celular Tumoral , Ciclo Celular , Proliferación Celular , Apoptosis , Reparación del ADN
6.
Front Immunol ; 14: 1211068, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37675104

RESUMEN

In recent years, the central role of cell bioenergetics in regulating immune cell function and fate has been recognized, giving rise to the interest in immunometabolism, an area of research focused on the interaction between metabolic regulation and immune function. Thus, early metabolic changes associated with the polarization of macrophages into pro-inflammatory or pro-resolving cells under different stimuli have been characterized. Tumor-associated macrophages are among the most abundant cells in the tumor microenvironment; however, it exists an unmet need to study the effect of chemotherapeutics on macrophage immunometabolism. Here, we use a systems biology approach that integrates transcriptomics and metabolomics to unveil the immunometabolic effects of trabectedin (TRB) and lurbinectedin (LUR), two DNA-binding agents with proven antitumor activity. Our results show that TRB and LUR activate human macrophages toward a pro-inflammatory phenotype by inducing a specific metabolic rewiring program that includes ROS production, changes in the mitochondrial inner membrane potential, increased pentose phosphate pathway, lactate release, tricarboxylic acids (TCA) cycle, serine and methylglyoxal pathways in human macrophages. Glutamine, aspartate, histidine, and proline intracellular levels are also decreased, whereas oxygen consumption is reduced. The observed immunometabolic changes explain additional antitumor activities of these compounds and open new avenues to design therapeutic interventions that specifically target the immunometabolic landscape in the treatment of cancer.


Asunto(s)
Neoplasias , Humanos , Trabectedina/farmacología , Macrófagos , Ácido Láctico , Microambiente Tumoral
7.
Dis Model Mech ; 16(6)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37158111

RESUMEN

This study exploited a novel patient-derived xenograft (PDX) of desmoplastic small round cell tumor (DSRCT), which reproduces histomorphological and molecular characteristics of the clinical tumor, to assess the activity of cytotoxic and targeted anticancer agents. Antitumor effect was moderate for doxorubicin, pazopanib and larotrectenib [maximum tumor volume inhibition (max TVI), 55-66%], while trabectedin had higher activity (max TVI, 82%). Vinorelbine, irinotecan and eribulin achieved nearly complete tumor growth inhibition (max TVI, 96-98%), although tumors regrew after the end of treatment. The combination of irinotecan with either eribulin or trabectedin resulted in complete responses, which were maintained until the end of the experiment for irinotecan plus trabectedin. Irinotecan-based combinations nearly abrogated the expression of proteins of the G2/M checkpoint, preventing cell entrance in mitosis, and induced apoptotic and necroptotic cell death. Consistently, irinotecan plus trabectedin resulted in reprogramming of DSCRT transcriptome, with downregulation of E2F targets, G2/M checkpoint and mitotic spindle gene sets. This study emphasizes the importance of patient-derived preclinical models to explore new treatments for DSRCT and fosters clinical investigation into the activity of irinotecan plus trabectedin.


Asunto(s)
Antineoplásicos , Tumor Desmoplásico de Células Pequeñas Redondas , Humanos , Trabectedina/uso terapéutico , Trabectedina/farmacología , Irinotecán/farmacología , Irinotecán/uso terapéutico , Tumor Desmoplásico de Células Pequeñas Redondas/tratamiento farmacológico , Tumor Desmoplásico de Células Pequeñas Redondas/patología , Xenoinjertos , Antineoplásicos/uso terapéutico
8.
Biomed Pharmacother ; 161: 114548, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36940615

RESUMEN

Immune cells have an important role in the tumor-microenvironment. Macrophages may tune the immune response toward inflammatory or tolerance pathways. Tumor-associated macrophages (TAM) have a string of immunosuppressive functions and they are considered a therapeutic target in cancer. This study aimed to analyze the effects of trabectedin, an antitumor agent, on the tumor-microenvironment through the characterization of the electrophysiological and molecular phenotype of macrophages. Experiments were performed using the whole-cell configuration of the patch-clamp technique in resident peritoneal mouse macrophages. Trabectedin does not directly interact with KV1.5 and KV1.3 channels, but their treatment (16 h) with sub-cytotoxic concentrations of trabectedin increased their KV current due to an upregulation of KV1.3 channels. In vitro generated TAM (TAMiv) exhibited an M2-like phenotype. TAMiv generated a small KV current and express high levels of M2 markers. K+ current from TAMs isolated from tumors generated in mice is a mixture of KV and KCa, and in TAM isolated from tumors generated in trabectedin-treated mice, the current is mostly driven by KCa. We conclude that the antitumor capacity of trabectedin is not only due to its effects on tumor cells, but also to the modulation of the tumor microenvironment, due, at least in part, to the modulation of the expression of different macrophage ion channels.


Asunto(s)
Macrófagos , Microambiente Tumoral , Ratones , Animales , Trabectedina/farmacología , Macrófagos/metabolismo , Activación de Macrófagos , Fenómenos Electrofisiológicos
9.
Chem Biol Interact ; 369: 110262, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36396105

RESUMEN

BACKGROUND AND OBJECTIVES: Non-small cell lung cancer (NSCLC) is a global health concern. NSCLC treatment outcomes are generally poor due to treatment resistance or toxicity. Ferroptosis is a novel cell death triggered by iron accumulation, reactive oxygen species (ROS), and lipid peroxidation. Ferroptosis may kill cancer cells, particularly those resistant to apoptosis. MATERIALS AND METHODS: The Cell Counting Kit-8 assay assessed NSCLC cell viability after trabectedin treatment. Flow cytometry with Annexin V-FITC staining evaluated cell death. ROS, iron, lipid peroxidation, and GSH levels were measured using commercial kits. qRT-PCR and western blots evaluated messenger RNA and protein levels. Proteins were inhibited using short interfering RNA transfection and specific inhibitors. RESULTS: Trabectedin was cytotoxic to NSCLC cells regardless of p53 status. Trabectedin upregulated iron, ROS, and lipid peroxidation in NSCLC cells, causing ferroptosis. Trabectedin increases iron and ROS levels by upregulating transferrin receptor 1 and the HIF-1/IRP1 axis. In NSCLC cells, trabectedin suppresses glutathione peroxidase 4, followed by the Keap1/Nrf2 axis. CONCLUSIONS: Our findings imply that trabectedin may treat NSCLC effectively.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Ferroptosis , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Trabectedina/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Hierro/metabolismo
10.
Int J Mol Sci ; 23(22)2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36430780

RESUMEN

Trabectedin is used for the treatment of advanced soft tissue sarcomas (STSs). In this study, we evaluated if trabectedin could enhance the efficacy of irradiation (IR) by increasing the intrinsic cell radiosensitivity and modulating tumor micro-environment in fibrosarcoma (HS 93.T), leiomyosarcoma (HS5.T), liposarcoma (SW872), and rhabdomyosarcoma (RD) cell lines. A significant reduction in cell surviving fraction (SF) following trabectedin + IR compared to IR alone was observed in liposarcoma and leiomyosarcoma (enhancement ratio at 50%, ER50: 1.45 and 2.35, respectively), whereas an additive effect was shown in rhabdomyosarcoma and fibrosarcoma. Invasive cells' fraction significantly decreased following trabectedin ± IR compared to IR alone. Differences in cell cycle distribution were observed in leiomyosarcoma and rhabdomyosarcoma treated with trabectedin + IR. In all STS lines, trabectedin + IR resulted in a significantly higher number of γ-H2AX (histone H2AX) foci 30 min compared to the control, trabectedin, or IR alone. Expression of ATM, RAD50, Ang-2, VEGF, and PD-L1 was not significantly altered following trabectedin + IR. In conclusion, trabectedin radiosensitizes STS cells by affecting SF (particularly in leiomyosarcoma and liposarcoma), invasiveness, cell cycle distribution, and γ-H2AX foci formation. Conversely, no synergistic effect was observed on DNA damage repair, neoangiogenesis, and immune system.


Asunto(s)
Fibrosarcoma , Leiomiosarcoma , Liposarcoma , Fármacos Sensibilizantes a Radiaciones , Rabdomiosarcoma , Sarcoma , Neoplasias de los Tejidos Blandos , Humanos , Trabectedina/farmacología , Trabectedina/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Leiomiosarcoma/tratamiento farmacológico , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Liposarcoma/tratamiento farmacológico , Microambiente Tumoral
11.
Org Biomol Chem ; 20(43): 8438-8442, 2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36254754

RESUMEN

Ecteinascidin 743 is a famous marine drug used in anticancer treatments. In this work, a series of simplified hybrids/analogues have been synthesized by employing a newly developed chemistry that integrates the partial structural features of two anticancer bis-tetrahydroisoquinoline alkaloids ecteinascidin 743 and cribrostatin 4. The described Suzuki-coupling protocol enabled us to easily introduce variable functionalities at the C3 position of the basic skeleton of bis-tetrahydroisoquinoline alkaloids for the first time. Cytotoxic examination showed that analogue 21f exhibited inhibitory activities with IC50 values in the low 10-6 M range against the proliferation of the cancer cell lines A549, HepG2, and MDA-MB-231. This work reveals that diversifying the C3/C4 olefin in the skeleton of tetrahydroisoquinoline alkaloids is a useful means to generate potential pharmaceuticals.


Asunto(s)
Alcaloides , Antineoplásicos , Neoplasias , Tetrahidroisoquinolinas , Trabectedina/farmacología , Tetrahidroisoquinolinas/farmacología , Tetrahidroisoquinolinas/química , Alcaloides/farmacología , Alcaloides/química , Antineoplásicos/farmacología , Antineoplásicos/química , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad , Línea Celular Tumoral , Estructura Molecular , Relación Dosis-Respuesta a Droga
12.
Andrologia ; 54(11): e14599, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36168116

RESUMEN

Trabectedin is a chemotherapy agent originating from a tunicate, Ecteinascidia turbinata. In this study, DNA-independent action mechanisms of trabectedin are investigated in prostate cancer (PCa) cells. Cell viability was assessed via XTT assay. Apoptosis was evaluated via flow cytometry. Tetramethylrodamine ethyl ester (TMRE) dye was utilized to determine mitochondrial membrane potential (MMP). Cell cycle distribution was investigated via flow cytometric analysis. Reactive oxygen species (ROS) were monitored using fluorescence CM-H2DCFDA dye. Changes in CHOP, p-eIF2α, GRP78 and p-PERK which are endoplasmic reticulum (ER) stress-involved proteins were investigated via western blot. Trabectedin induced cytotoxicity and cell cycle arrest at the G2/M phase. Trabectedin decreased MMP via ROS generation in PCa cells. ER stress-related proteins CHOP, p-eIF2α, GRP78 and p-PERK were also elevated by trabectedin treatment indicating the induction of ER stress-induced apoptosis. The results of this study show that trabectedin may be an effective chemotherapeutic for PCa.


Asunto(s)
Estrés del Retículo Endoplásmico , Neoplasias de la Próstata , Masculino , Humanos , Trabectedina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Apoptosis , Neoplasias de la Próstata/tratamiento farmacológico
13.
Eur J Immunol ; 51(11): 2677-2686, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34570376

RESUMEN

A considerable proportion of cancer patients are resistant or only partially responsive to immune checkpoint blockade immunotherapy. Tumor-Associated Macrophages (TAMs) infiltrating the tumor stroma suppress the adaptive immune responses and, hence, promote tumor immune evasion. Depletion of TAMs or modulation of their protumoral functions is actively pursued, with the purpose of relieving this state of immunesuppression. We previously reported that trabectedin, a registered antitumor compound, selectively reduces monocytes and TAMs in treated tumors. However, its putative effects on the adaptive immunity are still unclear. In this study, we investigated whether treatment of tumor-bearing mice with trabectedin modulates the presence and functional activity of T-lymphocytes. In treated tumors, there was a significant upregulation of T cell-associated genes, including CD3, CD8, perforin, granzyme B, and IFN-responsive genes (MX1, CXCL10, and PD-1), indicating that T lymphocytes were activated after treatment. Notably, the mRNA levels of the Pdcd1 gene, coding for PD-1, were strongly increased. Using a fibrosarcoma model poorly responsive to PD-1-immunotherapy, treatment with trabectedin prior to anti-PD-1 resulted in improved antitumor efficacy. In conclusion, pretreatment with trabectedin enhances the therapeutic response to checkpoint inhibitor-based immunotherapy. These findings provide a good rational for the combination of trabectedin with immunotherapy regimens.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Antineoplásicos Alquilantes/farmacología , Neoplasias Experimentales/inmunología , Trabectedina/farmacología , Macrófagos Asociados a Tumores/efectos de los fármacos , Animales , Fibrosarcoma/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Macrófagos Asociados a Tumores/inmunología
14.
Hum Cell ; 34(5): 1569-1578, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34164773

RESUMEN

Spindle cell/sclerosing rhabdomyosarcoma (ssRMS) is a rare subtype of rhabdomyosarcoma (RMS) that has fascicular spindle cell and/or sclerosing morphology. SsRMS has a diverse molecular background and is categorized into three groups: congenital/infantile ssRMS with a gene fusion involving the NCOA2 and VGLL2, ssRMS with the MYOD1 mutation, and ssRMS with no recurrent identifiable genetic alterations. Because ssRMS is a newly defined disease concept of RMS, the optimal treatment methods have not been determined. This results in unfavorable prognosis and consequently signals the urgent need for continuous research. Patient-derived cell lines are essential tools in basic and translational research. However, only two ssRMS cell lines with the MYOD1 mutation have been reported to date. Thus, we established a novel ssRMS cell line named NCC-ssRMS2-C1 using a surgically resected tumor tissue from an adult ssRMS patient. NCC-ssRMS2-C1 cells retained the copy number alterations corresponding to the original tumor and are categorized into the group with no recurrent identifiable genetic alterations. NCC-ssRMS2-C1 cells demonstrated constant proliferation, spheroid formation, and capability for invasion in vitro, reflecting the malignant features of the original tumor tissue. In a drug screening test, ssRMS demonstrated remarkable sensitivity to romidepsin, trabectedin, actinomycin D, and bortezomib. Hence, we conclude that the NCC-ssRMS2-C1 cell line is the first ssRMS cell line which belongs to the group with no recurrent identifiable genetic alterations, and it will be a useful resource in both basic and translational studies for ssRMS.


Asunto(s)
Neoplasias de Cabeza y Cuello , Rabdomiosarcoma , Sarcoma , Adulto , Antineoplásicos/farmacología , Bortezomib/farmacología , Línea Celular Tumoral , Dactinomicina/farmacología , Depsipéptidos/farmacología , Fusión Génica , Neoplasias de Cabeza y Cuello/clasificación , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Proteínas Musculares/genética , Mutación , Proteína MioD/genética , Coactivador 2 del Receptor Nuclear/genética , Rabdomiosarcoma/clasificación , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Sarcoma/clasificación , Sarcoma/genética , Sarcoma/patología , Trabectedina/farmacología , Factores de Transcripción/genética
15.
J Exp Clin Cancer Res ; 40(1): 165, 2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-33975637

RESUMEN

BACKGROUND: Soft tissue sarcomas (STS) are a rare group of solid neoplasm including among others liposarcoma, leiomyosarcoma (L-sarcoma) and undifferentiated pleomorphic sarcoma (UPS) entities. The current first-line treatment is represented by anthracycline based- regimens, second-line may include trabectedin. Currently the activity of trabectedin and its mechanism of action is not completely elucidated. METHODS: Taking the advantages of our 3D patient-derived primary culture translational model we performed genomic-, chemobiogram, proteomic- and in vivo analysis in a UPS culture (S1). Furthermore pharmacological profiling of a UPS and L-sarcoma patient-derived case series and in silico analysis were carried out. RESULTS: Trabectedin exhibited an increased activity in 3D respect to 2D cultures suggesting an extracellular matrix (ECM) and timp1 involvement in its mechanism of action. Moreover 3D S1 xenotranspanted zebrafish model showed an increased sensitivity to trabectedin. Finally the results were further validated in a UPS and L-sarcoma case series. CONCLUSIONS: Taken together these results confirmed the activity of trabectedin in these STS histotypes. Moreover the data underline the ECM involvement in the cytotoxic effect mediated by trabectedin and could open the door for researches aimed to focus on the patient setting that could benefit from this agent.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Sarcoma/tratamiento farmacológico , Trabectedina/farmacología , Animales , Modelos Animales de Enfermedad , Matriz Extracelular , Humanos , Pez Cebra
16.
Hum Cell ; 34(3): 990-997, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33555519

RESUMEN

Dedifferentiated liposarcoma (DDLPS) is a highly aggressive subtype of liposarcoma that is histologically a transition form between an atypical lipomatous tumor/well-differentiated liposarcoma and a non-lipogenic sarcoma. DDLPS is genetically characterized by a complex karyotype with copy number variations and genomic complexity. DDLPS has a poor prognosis, a high local recurrence rate, and refractory behaviors for chemotherapy and radiation, which indicate a requirement for a novel therapeutic strategy for better clinical outcomes. We report here, a novel DDLPS cell line (NCC-DDLPS2-C1) developed from a tumor tissue. NCC-DDLPS2-C1 cells showed an amplified 12q13-15 region and exhibited constant growth, spheroid formation, and invasion. High-throughput drug screening revealed distinct sensitivity between monolayer- and three-dimensional cells. Romidepsin and trabectedin especially showed high anti-proliferative effects in both culture methods of NCC-DDLPS2-C1. Thus, the NCC-DDLPS2-C1 cell line may serve as a useful resource for DDLPS studies.


Asunto(s)
Liposarcoma/genética , Liposarcoma/patología , Anciano de 80 o más Años , Antineoplásicos/farmacología , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromosomas Humanos Par 12/genética , Depsipéptidos/farmacología , Femenino , Dosificación de Gen , Humanos , Cariotipo , Invasividad Neoplásica , Esferoides Celulares/patología , Trabectedina/farmacología
17.
Molecules ; 26(4)2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33578831

RESUMEN

Currently, SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) has infected people among all countries and is a pandemic as declared by the World Health Organization (WHO). SARS-CoVID-2 main protease is one of the therapeutic drug targets that has been shown to reduce virus replication, and its high-resolution 3D structures in complex with inhibitors have been solved. Previously, we had demonstrated the potential of natural compounds such as serine protease inhibitors eventually leading us to hypothesize that FDA-approved marine drugs have the potential to inhibit the biological activity of SARS-CoV-2 main protease. Initially, field-template and structure-activity atlas models were constructed to understand and explain the molecular features responsible for SARS-CoVID-2 main protease inhibitors, which revealed that Eribulin Mesylate, Plitidepsin, and Trabectedin possess similar characteristics related to SARS-CoVID-2 main protease inhibitors. Later, protein-ligand interactions are studied using ensemble molecular-docking simulations that revealed that marine drugs bind at the active site of the main protease. The three-dimensional reference interaction site model (3D-RISM) studies show that marine drugs displace water molecules at the active site, and interactions observed are favorable. These computational studies eventually paved an interest in further in vitro studies. Finally, these findings are new and indeed provide insights into the role of FDA-approved marine drugs, which are already in clinical use for cancer treatment as a potential alternative to prevent and treat infected people with SARS-CoV-2.


Asunto(s)
Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , SARS-CoV-2/fisiología , Inhibidores de Serina Proteinasa/farmacología , Dominio Catalítico , Depsipéptidos/química , Depsipéptidos/farmacología , Reposicionamiento de Medicamentos , Furanos/química , Furanos/farmacología , Humanos , Cetonas/química , Cetonas/farmacología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Péptidos Cíclicos , Relación Estructura-Actividad Cuantitativa , SARS-CoV-2/efectos de los fármacos , Inhibidores de Serina Proteinasa/química , Trabectedina/química , Trabectedina/farmacología , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
18.
Cancer Lett ; 500: 182-193, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33326840

RESUMEN

Classical Hodgkin lymphoma (cHL) tumor cells are surrounded by a protective tumor microenvironment (TME). Trabectedin, an anticancer drug targeting both tumor cells and TME, demonstrated a potent antitumor activity against Hodgkin Reed Sternberg (HRS) cells. It was cytotoxic against cHL cell lines, including the doxorubicin-resistant clones, with subnanomolar IC50 values, and inhibited clonogenic growth and heterospheroid cell viability. It induced necroptosis, caused DNA damage, G2/M cell cycle arrest, and increased reactive oxygen species production. It reduced HRS cell secretion of CCL5, M-CSF, IL-6, IL-13 and TARC, and inhibited migration. Conditioned medium from trabectedin-treated HRS cells was less chemoattractive toward monocytes, mesenchymal stromal cells and lymphocytes, and less effective in educating monocytes to become immunosuppressive macrophages. These monocytes expressed lower levels of indoleamine 2,3-dioxygenase-1, CD206 and PD-L1, secreted lower amounts of IL-10, TARC, and TGF-ß, and were less able to inhibit the growth of activated lymphocytes. In vivo, trabectedin inhibited by >75% the growth of cHL murine xenografts with minimal weight loss; tumors of trabectedin-treated mice had fewer TAMs and less angiogenesis. Altogether, this study offers a preclinical rationale for trabectedin use as a new drug candidate in relapsed/refractory cHL patients.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Enfermedad de Hodgkin/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Humanos , Inmunosupresores/farmacología , Ratones , Monocitos/efectos de los fármacos , Proteínas de Neoplasias/genética , Trabectedina/farmacología , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Dev Comp Immunol ; 108: 103669, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32192994

RESUMEN

Among the diseases that afflict the human population, cancer is one for which many drug treatments are not yet known or effective. Moreover, the pharmacological treatments used often create serious side effects in sick patients and for this reason, it is essential to find effective and less harmful treatments. To date, marine biodiversity is a real source of metabolites with antitumoral activity and among invertebrates' ascidians have been the main source to obtain them. Mediterranean area is the richest in biodiversity and contains several ascidian species used in drugs development during the years. However, many more Mediterranean ascidian species have not been studied and could be a source of useful bioactive compounds. This review aims to summarize the scientific studies that analyzed the antitumor compounds obtained from different Mediterranean ascidians species, encouraging them to search further compounds in other new species to improve pharmacological treatments and human population life.


Asunto(s)
Antineoplásicos/uso terapéutico , Productos Biológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Urocordados/inmunología , Animales , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Productos Biológicos/aislamiento & purificación , Productos Biológicos/farmacología , Depsipéptidos/aislamiento & purificación , Depsipéptidos/farmacología , Depsipéptidos/uso terapéutico , Humanos , Mar Mediterráneo , Neoplasias/inmunología , Péptidos Cíclicos/aislamiento & purificación , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Trabectedina/aislamiento & purificación , Trabectedina/farmacología , Trabectedina/uso terapéutico
20.
Curr Drug Targets ; 21(10): 996-1007, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31994460

RESUMEN

The use of predictive biomarkers provides potential individualized cancer therapeutic options to prevent therapy failure as well as serious toxicities. Several recent studies showed that predictive and prognostic biomarkers are a notable personalized strategy to improve patients' care in several cancers. Trabectedin (Yondelis®) is a cytotoxic agent, derived from a marine organism, harbouring a significant antitumor activity against several cancers such as soft tissue sarcoma, ovarian, and breast cancers. Recently and with the advent of molecular genetic testing, BRCA mutational status was found as an important predictor of response to this anticancer drug, especially in gynecological cancers. The aim of this updated review is to discuss the mechanisms of action of trabectedin against the wellknown cancer hallmarks described until today. The current advances were also examined related to genomic biomarkers that can be used in the future to predict the efficacy of this potent anticancer natural molecule in various gynecological cancers.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Trabectedina/farmacología , Trabectedina/uso terapéutico , Proteína BRCA1/genética , Proteína BRCA2/genética , Biomarcadores Farmacológicos , Femenino , Genómica , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...