Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cannabis Cannabinoid Res ; 9(2): 437-448, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38377580

RESUMEN

Background: Δ9-tetrahydrocannabinol (THC), the primary intoxicating compound in cannabis, has been tested extensively in controlled administration human studies. Some studies require a high THC dose that may induce adverse events (AEs), such as those testing novel treatments for cannabinoid overdose. Although there are ethical concerns related to administering high THC doses, there is no systematic analysis on studies utilizing these doses. In this review, we examine studies that administered oral THC doses ≥30 mg ("high-dose THC"), focusing on reported tolerability, subjective effects, and pharmacokinetics (PK), with the objective to inform the design of future studies. Methods: A comprehensive PubMed search was performed to identify studies meeting pre-specified criteria. Results: Our search identified 27 publications from 17 high-dose oral THC laboratory studies, with single doses up to 90 mg and multiple doses up to 210 mg per day. The maximum plasma THC concentration (Cmax) appeared to increase in a dose-proportional manner over this dose range. All high-dose THC studies enrolled participants with previous cannabis experience, although current use ranged from nonusers to regular cannabis users. High-dose THC was generally well tolerated with transient mild to moderate AE, including nausea and vomiting, anxiety, paranoia, and sedation. There were occasional participant withdrawals due to AEs, but there were no serious AE. Participants with frequent cannabis use tolerated high-dose THC best. Conclusion: Although based on limited data, THC was generally adequately tolerated with single oral doses of at least 50 mg in a controlled laboratory setting in healthy participants with past cannabis experience.


Asunto(s)
Cannabinoides , Cannabis , Humanos , Dronabinol/efectos adversos , Cannabinoides/efectos adversos , Proyectos de Investigación , Ansiedad
2.
Int J Biol Macromol ; 253(Pt 2): 126694, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37673150

RESUMEN

The following article provides an insight into the production of chitosan aerogels as potential materials for tissue engineering. Chitosan aerogels were prepared following two different protocols: formation in ethanol and formation in sodium hydroxide in an ethanol solution. The main objective was to apply a new route to obtain chitosan aerogels with no external cross-linkers and compare the mentioned preparation approaches. Forming chitosan aerogels in ethanol implies a simple, environmentally friendly, and efficient method. The prepared materials showed specific surface areas of up to 450 m2/g, highly porous networks and great mechanical properties. In vitro degradation studies revealed high stability for up to 10 weeks. The differences between the samples were significant. While the chitosan aerogels prepared in ethanol showed superior textural, morphological and mechanical properties, the chitosan aerogels prepared in the sodium hydroxide solution proved that a considerable influence on end properties could be made simply by adjusting the ageing medium. In vitro cell analysis with primary human osteoblasts showed good biocompatibility and pointed towards the potential use of these aerogels for orthopedic applications. This testing showed further that adjustments in structural properties by sodium hydroxide also come with a cost regarding their suitability to host bone cells.


Asunto(s)
Quitosano , Humanos , Quitosano/farmacología , Quitosano/química , Geles/química , Etanol , Hidróxido de Sodio , Osteoblastos
3.
Cells ; 11(15)2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35954159

RESUMEN

Determining the viability of cells is fraught with many uncertainties. It is often difficult to determine whether a cell is still alive, approaching the point of no return, or dead. Today, there are many methods for determining cell viability. Most rely on an indirect determination of cell death (metabolism, molecular transport, and leakage, to name a few). In contrast, we have developed a promising novel method for a "direct" determination of cell viability. The potential method assesses cell membrane integrity (which is essential for all viable cells) by measuring the electrical potential of the cell membrane. To test the assay, we chose two different cell types, blood macrophages (TLT) and breast cancer epithelial cells (MCF 7). We exposed them to seven different toxic scenarios (arsenic (V), UV light, hydrogen peroxide, nutrient starvation, Tetrabromobisphenol A, fatty acids, and 5-fluorouracil) to induce different cell death pathways. Under controlled test conditions, the assay showed good accuracy when comparing the toxicity assessment with well-established methods. Moreover, the method showed compatibility with live cell imaging. Although we know that further studies are needed to confirm the performance of the assay in other situations, the results obtained are promising for their wider application in the future.


Asunto(s)
Peróxido de Hidrógeno , Rayos Ultravioleta , Recuento de Células , Supervivencia Celular , Peróxido de Hidrógeno/farmacología , Potenciales de la Membrana
4.
Biomedicines ; 10(2)2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35203587

RESUMEN

Mesenchymal stem cells (MSCs) represent the basis of novel clinical concepts in cellular therapy and tissue regeneration. Therefore, the isolation of MSCs from various tissues has become an important endeavour for stem cell biobanking and the development of regenerative therapies. Paravertebral adipose tissue is readily exposed during spinal procedures in children and could be a viable source of stem cells for therapeutic applications. Here, we describe the first case of MSCs isolated from paravertebral adipose tissue (PV-ADMSCs), obtained during a routine spinal surgery on a child. Using quantitative real-time PCR and flow cytometry, we show that PV-ADMSCs have different levels of stem marker expression compared to the MSCs from other sources while having the highest proliferation rate. Furthermore, we evaluate the multipotency of PV-ADMSCs by the three-lineage (adipogenic, osteogenic and chondrogenic) differentiation and compare it to the multipotency of MSCs from other sources. It was found that the PV-ADMSCs have a strong osteogenic potential in particular. Taken together, our data indicate that PV-ADMSCs meet the criteria for successful cell therapy, defined by the International Society for Cellular Therapy (ISCT), and thus, could provide a source of MSCs that is relatively easy to isolate and expand in culture. Due to their strong osteogenic potential, these cells provide a promising basis, especially for orthopaedic applications.

5.
Cell Death Differ ; 29(1): 230-245, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34453119

RESUMEN

Mounting evidence indicates that immunogenic therapies engaging the unfolded protein response (UPR) following endoplasmic reticulum (ER) stress favor proficient cancer cell-immune interactions, by stimulating the release of immunomodulatory/proinflammatory factors by stressed or dying cancer cells. UPR-driven transcription of proinflammatory cytokines/chemokines exert beneficial or detrimental effects on tumor growth and antitumor immunity, but the cell-autonomous machinery governing the cancer cell inflammatory output in response to immunogenic therapies remains poorly defined. Here, we profiled the transcriptome of cancer cells responding to immunogenic or weakly immunogenic treatments. Bioinformatics-driven pathway analysis indicated that immunogenic treatments instigated a NF-κB/AP-1-inflammatory stress response, which dissociated from both cell death and UPR. This stress-induced inflammation was specifically abolished by the IRE1α-kinase inhibitor KIRA6. Supernatants from immunogenic chemotherapy and KIRA6 co-treated cancer cells were deprived of proinflammatory/chemoattractant factors and failed to mobilize neutrophils and induce dendritic cell maturation. Furthermore, KIRA6 significantly reduced the in vivo vaccination potential of dying cancer cells responding to immunogenic chemotherapy. Mechanistically, we found that the anti-inflammatory effect of KIRA6 was still effective in IRE1α-deficient cells, indicating a hitherto unknown off-target effector of this IRE1α-kinase inhibitor. Generation of a KIRA6-clickable photoaffinity probe, mass spectrometry, and co-immunoprecipitation analysis identified cytosolic HSP60 as a KIRA6 off-target in the IKK-driven NF-κB pathway. In sum, our study unravels that HSP60 is a KIRA6-inhibitable upstream regulator of the NF-κB/AP-1-inflammatory stress responses evoked by immunogenic treatments. It also urges caution when interpreting the anti-inflammatory action of IRE1α chemical inhibitors.


Asunto(s)
Endorribonucleasas , Proteínas Serina-Treonina Quinasas , Retículo Endoplásmico/metabolismo , Endorribonucleasas/metabolismo , Humanos , Imidazoles , Muerte Celular Inmunogénica , Inflamación/metabolismo , Naftalenos , Pirazinas
6.
Biomedicines ; 9(10)2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34680532

RESUMEN

The ethical constraints and shortcomings of animal models, combined with the demand to study disease pathogenesis under controlled conditions, are giving rise to a new field at the interface of tissue engineering and pathophysiology, which focuses on the development of in vitro models of disease. In vitro models are defined as synthetic experimental systems that contain living human cells and mimic tissue- and organ-level physiology in vitro by taking advantage of recent advances in tissue engineering and microfabrication. This review provides an overview of in vitro models and focuses specifically on in vitro disease models of the endocrine pancreas and diabetes. First, we briefly review the anatomy, physiology, and pathophysiology of the human pancreas, with an emphasis on islets of Langerhans and beta cell dysfunction. We then discuss different types of in vitro models and fundamental elements that should be considered when developing an in vitro disease model. Finally, we review the current state and breakthroughs in the field of pancreatic in vitro models and conclude with some challenges that need to be addressed in the future development of in vitro models.

7.
Analyst ; 146(16): 4985-5007, 2021 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-34337638

RESUMEN

Multi-spectral imaging flow cytometry (MIFC) has become one of the most powerful technologies for investigating general analytics, molecular and cell biology, biotechnology, medicine, and related fields. It combines the capabilities of the morphometric and photometric analysis of single cells and micrometer-sized particles in flux with regard to thousands of events. It has become the tool of choice for a wide range of research and clinical applications. By combining the features of flow cytometry and fluorescence microscopy, it offers researchers the ability to couple the spatial resolution of multicolour images of cells and organelles with the simultaneous analysis of a large number of events in a single system. This provides the opportunity to visually confirm findings and collect novel data that would otherwise be more difficult to obtain. This has led many researchers to design innovative assays to gain new insight into important research questions. To date, it has been successfully used to study cell morphology, surface and nuclear protein co-localization, protein-protein interactions, cell signaling, cell cycle, cell death, and cytotoxicity, intracellular calcium, drug uptake, pathogen internalization, and other applications. Herein we describe some of the recent advances in the field of multiparametric imaging flow cytometry methods in various research areas.


Asunto(s)
Núcleo Celular , Ciclo Celular , Muerte Celular , Citometría de Flujo , Humanos , Microscopía Fluorescente
8.
Polymers (Basel) ; 13(14)2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34301068

RESUMEN

The development of novel polymer-based materials opens up possibilities for several novel applications, such as advanced wound dressings, bioinks for 3D biofabrication, drug delivery systems, etc. The aim of this study was to evaluate the viability of vascular and intestinal epithelial cells on different polymers as a selection procedure for more advanced cell-polymer applications. In addition, possible correlations between increased cell viability and material properties were investigated. Twelve polymers were selected, and thin films were prepared by dissolution and spin coating on silicon wafers. The prepared thin films were structurally characterized by Fourier transform infrared spectroscopy, atomic force microscopy, and goniometry. Their biocompatibility was determined using two epithelial cell lines (human umbilical vein endothelial cells and human intestinal epithelial cells), assessing the metabolic activity, cell density, and morphology. The tested cell lines showed different preferences regarding the culture substrate. No clear correlation was found between viability and individual substrate characteristics, suggesting that complex synergistic effects may play an important role in substrate design. These results show that a systematic approach is required to compare the biocompatibility of simple cell culture substrates as well as more complex applications (e.g., bioinks).

9.
Plants (Basel) ; 10(3)2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33802757

RESUMEN

Cannabis sativa is one of the oldest medicinal plants used by humans, containing hundreds of bioactive compounds. The biological effects and interplay of these compounds are far from fully understood, although the plant's therapeutic effects are beyond doubt. Extraction methods for these compounds are becoming an integral part of modern Cannabis-based medicine. Still, little is known about how different methods affect the final composition of Cannabis extracts and thus, their therapeutic effects. In this study, different extraction methods were tested, namely maceration, Soxhlet, ultrasound-assisted extraction (UAE), and supercritical CO2 extraction methods. The obtained extracts were evaluated for their cannabinoid content, antioxidant properties, and in vitro bioactivity on human colon cancer and healthy colon cells. Our data suggest that Cannabis extracts, when properly prepared, can significantly decrease cancer cell viability while protecting healthy cells from cytotoxic effects. However, post-processing of extracts poses a significant limitation in predicting therapeutic response based on the composition of the crude extract, as it affects not only the actual amounts of the respective cannabinoids but also their relative ratio to the primary extracts. These effects must be carefully considered in the future preparations of new therapeutic extracts.

10.
Pharmaceutics ; 13(4)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923814

RESUMEN

In this study, a multilayer bioactive coating based on carboxymethyl cellulose (CMC) and dexamethasone (DEX) was prepared on medical-grade stainless steel (AISI 316LVM). Its aim was the controlled drug delivery of the incorporated anti­inflammatory drug, which at the same time promotes osteogenic differentiation of mesenchymal stem cells. Due to DEX's limited solubility in physiological fluids, which limits the loading capacity of coatings, it was further combined with ß-cyclodextrin to increase its concentration in the bioactive coating. Controlled release of DEX from the multilayer coating was achieved in four steps: a "burst", i.e., very fast, release step (in an immersion interval of 0-10 min), a fast release step (10-30 min), a slow-release step (60-360 min), and a plateau step (360-4320 min), following a zero-order release or Higuchi model release mechanism. Successful layer-by-layer coating formation was confirmed using attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR). It was shown that the application of the coating significantly increases the hydrophilic character of AISI 316LVM, and also significantly increases the surface roughness, which is known to promote cell growth. In addition, electrochemical measurements demonstrated that the coating application does not increase the susceptibility of medical-grade stainless steel to corrosion. In vitro cell testing using all cell types with which such coatings come into contact in the body (osteoblasts, chondrocytes, and mesenchymal stem cells (MSCs)) showed very good biocompatibility towards all of the mentioned cells. It further confirmed that the coatings promoted MSCs osteogenic differentiation, which is the desired mode of action for orthopedic implants.

11.
Transl Oncol ; 12(7): 932-950, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31096110

RESUMEN

Development of novel bioactive compounds against KRAS and/or BRAF mutant colorectal cancer (CRC) is currently an urgent need in oncology. In addition, single or multitarget kinase inhibitors against MEK/ERK and PI3K/AKT pathways are of potential therapeutic advantage. A new compound based on the benzothiophene nucleus was synthesized, based on previous important outcomes on other pharmaceutical preparations, to be tested as potential anticancer agent. Treatments by 2-5 µM DPS-2 of several CRC and melanoma cell lines bearing either BRAF or KRAS mutations have shown a remarkable effect on cell viability in 2D and 3D cultures. More detailed analysis has shown that DPS-2 can kill cancer cells by apoptosis, reducing at the same time their autophagy properties. After testing activities of several signaling pathways, the compound was found to have a dual inhibition of two major proliferative/survival pathways, MEK/ERK and PI3K/AKT, in both CRC and melanoma, thus providing a mechanistic evidence for its potent anticancer activity. Antitumor activity of DPS-2 was further validated in vivo, as DPS-2 treatment of mouse xenografts of Colo-205 colorectal cancer cells remarkably reduced their tumor formation properties. Our findings suggest that DPS-2 has significant anti-KRAS/ anti-BRAF mutant CRC activity in preclinical models, potentially providing a novel treatment strategy for these difficult-to-treat tumors, which needs to be further exploited.

12.
Cell Death Differ ; 26(12): 2520-2534, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30850732

RESUMEN

Melanoma cells are highly resistant to conventional genotoxic agents, and BRAFV600/MEK-targeted therapies as well as immunotherapies frequently remain inefficient. Alternative means to treat melanoma, in particular through the induction of programmed cell death modalities such as apoptosis or necroptosis, therefore still need to be explored. Here, we report that melanoma cell lines expressing notable amounts of RIPK1, RIPK3 and MLKL, the key players of necroptosis signal transduction, fail to execute necroptotic cell death. Interestingly, the activity of transforming growth factor ß-activated kinase 1 (TAK1) appears to prevent RIPK1 from contributing to cell death induction, since TAK1 inhibition by (5Z)-7-Oxozeaenol, deletion of MAP3K7 or the expression of inactive TAK1 were sufficient to sensitize melanoma cells to RIPK1-dependent cell death in response to TNFα or TRAIL based combination treatments. However, cell death was executed exclusively by apoptosis, even when RIPK3 expression was high. In addition, TAK1 inhibitor (5Z)-7-Oxozeaenol suppressed intrinsic or treatment-induced pro-survival signaling as well as the secretion of cytokines and soluble factors associated with melanoma disease progression. Correspondingly, elevated expression of TAK1 correlates with reduced disease free survival in patients diagnosed with primary melanoma. Overall, our results therefore demonstrate that TAK1 suppresses the susceptibility to RIPK1-dependent cell death and that high expression of TAK1 indicates an increased risk for disease progression in melanoma.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Melanoma/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Decitabina/farmacología , Progresión de la Enfermedad , Humanos , Quinasas Quinasa Quinasa PAM/biosíntesis , Quinasas Quinasa Quinasa PAM/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Necroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/biosíntesis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal , Transfección , Zearalenona/análogos & derivados , Zearalenona/farmacología
13.
Cell Death Differ ; 26(8): 1365-1378, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30323272

RESUMEN

Malignant melanoma is a highly aggressive form of skin cancer responsible for the majority of skin cancer-related deaths. Recent insight into the heterogeneous nature of melanoma suggests more personalised treatments may be necessary to overcome drug resistance and improve patient care. To this end, reliable molecular signatures that can accurately predict treatment responsiveness need to be identified. In this study, we applied multiplex phosphoproteomic profiling across a panel of 24 melanoma cell lines with different disease-relevant mutations, to predict responsiveness to MEK inhibitor trametinib. Supported by multivariate statistical analysis and multidimensional pattern recognition algorithms, the responsiveness of individual cell lines to trametinib could be predicted with high accuracy (83% correct predictions), independent of mutation status. We also successfully employed this approach to case specifically predict whether individual melanoma cell lines could be sensitised to trametinib. Our predictions identified that combining MEK inhibition with selective targeting of c-JUN and/or FAK, using siRNA-based depletion or pharmacological inhibitors, sensitised resistant cell lines and significantly enhanced treatment efficacy. Our study indicates that multiplex proteomic analyses coupled with pattern recognition approaches could assist in personalising trametinib-based treatment decisions in the future.


Asunto(s)
Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Piridonas/farmacología , Pirimidinonas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Melanoma/metabolismo , Melanoma/patología
14.
BMC Cell Biol ; 19(1): 28, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30587121

RESUMEN

BACKGROUND: Dasatinib (Sprycel) was developed as a tyrosine kinase inhibitor targeting Bcr-Abl and the family of Src kinases. Dasatinib is commonly used for the treatment of acute lymphoblastic and chronic myelogenous leukemia. Previous clinical studies in melanoma returned inconclusive results and suggested that patients respond highly heterogeneously to dasatinib as single agent or in combination with standard-of-care chemotherapeutic dacarbazine. Reliable biomarkers to predict dasatinib responsiveness in melanoma have not yet been developed. RESULTS: Here, we collected comprehensive in vitro data from experimentally well-controlled conditions to study the effect of dasatinib, alone and in combination with dacarbazine, on cell proliferation and cell survival. Sixteen treatment conditions, covering therapeutically relevant concentrations ranges of both drugs, were tested in 12 melanoma cell lines with diverse mutational backgrounds. Melanoma cell lines responded heterogeneously and, importantly, dasatinib and dacarbazine did not synergize in suppressing proliferation or inducing cell death. Since dasatinib is a promiscuous kinase inhibitor, possibly affecting multiple disease-relevant pathways, we also determined if basal phospho-protein amounts and treatment-induced changes in phospho-protein levels are indicative of dasatinib responsiveness. We found that treatment-induced de-phosphorylation of p53 correlates with dasatinib responsiveness in malignant melanoma. CONCLUSIONS: Loss of p53 phosphorylation might be an interesting candidate for a kinetic marker of dasatinib responsiveness in melanoma, pending more comprehensive validation in future studies.


Asunto(s)
Dasatinib/farmacología , Melanoma/metabolismo , Melanoma/patología , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dacarbazina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos
15.
Cell Syst ; 7(2): 161-179.e14, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30007540

RESUMEN

Clinically used RAF inhibitors are ineffective in RAS mutant tumors because they enhance homo- and heterodimerization of RAF kinases, leading to paradoxical activation of ERK signaling. Overcoming enhanced RAF dimerization and the resulting resistance is a challenge for drug design. Combining multiple inhibitors could be more effective, but it is unclear how the best combinations can be chosen. We built a next-generation mechanistic dynamic model to analyze combinations of structurally different RAF inhibitors, which can efficiently suppress MEK/ERK signaling. This rule-based model of the RAS/ERK pathway integrates thermodynamics and kinetics of drug-protein interactions, structural elements, posttranslational modifications, and cell mutational status as model rules to predict RAF inhibitor combinations for inhibiting ERK activity in oncogenic RAS and/or BRAFV600E backgrounds. Predicted synergistic inhibition of ERK signaling was corroborated by experiments in mutant NRAS, HRAS, and BRAFV600E cells, and inhibition of oncogenic RAS signaling was associated with reduced cell proliferation and colony formation.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Quinasas raf/antagonistas & inhibidores , Proteínas ras/metabolismo , Línea Celular Tumoral , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Simulación del Acoplamiento Molecular , Mutación/efectos de los fármacos , Neoplasias/genética , Neoplasias/metabolismo , Multimerización de Proteína/efectos de los fármacos , Termodinámica , Quinasas raf/química , Quinasas raf/metabolismo , Proteínas ras/genética
16.
Oncol Res ; 25(9): 1489-1494, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28337955

RESUMEN

Antagonists of inhibitors of apoptosis proteins (IAPs), alone or in combination with genotoxic therapeutics, have been shown to efficiently induce cell death in various solid tumors. The IAP antagonist birinapant is currently being tested in phase II clinical trials. We herein aimed to investigate the antitumor efficacy of dacarbazine in vitro, both as a single agent and in combination with birinapant, in melanoma cell lines. Covering clinically relevant drug concentration ranges, we conducted a total of 5,400 measurements in a panel of 12 human melanoma cell lines representing different stages of disease progression. Surprisingly, functionally relevant synergies or response potentiation in combination treatments was not observed, and only one cell line modestly responded to birinapant single treatment (approximately 16% cell death). Although we did not study the underlying resistance mechanisms or more complex in vivo scenarios in which dacarbazine/birinapant response synergies may still possibly manifest, our findings are nevertheless noteworthy because IAP antagonists were demonstrated to strongly enhance responses to DNA-damaging agents in cell lines of other cancer types under comparable experimental conditions in vitro.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Dipéptidos/farmacología , Indoles/farmacología , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Dacarbazina/administración & dosificación , Dipéptidos/administración & dosificación , Humanos , Indoles/administración & dosificación , Melanoma/patología , Mutación , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...