ABSTRACT
Background: Infiltration is a life-threatening growth pattern in malignant astrocytomas and a significant cause of therapy resistance. It results in the tumor cell spreading deeply into the surrounding brain tissue, fostering tumor recurrence and making complete surgical resection impossible. We need to thoroughly understand the mechanisms underlying diffuse infiltration to develop effective therapies. Methods: We integrated in vitro and in vivo functional assays, RNA sequencing, clinical, and expression information from public data sets to investigate the role of ADAM23 expression coupling astrocytoma's growth and motility. Results: ADAM23 downregulation resulted in increased infiltration, reduced tumor growth, and improved overall survival in astrocytomas. Additionally, we show that ADAM23 deficiency induces γ-secretase (GS) complex activity, contributing to the production and deposition of the Amyloid-ß and release of NICD. Finally, GS ablation in ADAM23-low astrocytomas induced a significant inhibitory effect on the invasive programs. Conclusions: Our findings reveal a role for ADAM23 in regulating the balance between cell proliferation and invasiveness in astrocytoma cells, proposing GS inhibition as a therapeutic option in ADAM23 low-expressing astrocytomas.
ABSTRACT
There are no effective strategies for the successful treatment of glioblastomas (GBM). Current therapeutic modalities effectively target bulk tumor cells but leave behind marginal GBM cells that escape from the surgical margins and radiotherapy field, exhibiting high migratory phenotype and resistance to all available anti-glioma therapies. Drug resistance is mostly driven by tumor cell plasticity: a concept associated with reactivating transcriptional programs in response to adverse and dynamic conditions from the tumor microenvironment. Autophagy, or "self-eating", pathway is an emerging target for cancer therapy and has been regarded as one of the key drivers of cell plasticity in response to energy demanding stress conditions. Many studies shed light on the importance of autophagy as an adaptive mechanism, protecting GBM cells from unfavorable conditions, while others recognize that autophagy can kill those cells by triggering a non-apoptotic cell death program, called 'autophagy cell death' (ACD). In this review, we carefully analyzed literature data and conclude that there is no clear evidence indicating the presence of ACD under pathophysiological settings in GBM disease. It seems to be exclusively induced by excessive (supra-physiological) stress signals, mostly from in vitro cell culture studies. Instead, pre-clinical and clinical data indicate that autophagy is an emblematic example of the 'dark-side' of a rescue pathway that contributes profoundly to a pro-tumoral adaptive response. From a standpoint of treating the real human disease, only combinatorial therapy targeting autophagy with cytotoxic drugs in the adjuvant setting for GBM patients, associated with the development of less toxic and more specific autophagy inhibitors, may inhibit adaptive response and enhance the sensibility of glioma cells to conventional therapies.
ABSTRACT
Aim: To characterize a method to isolate glioma-derived extracellular vesicles (GEVs) and understand their role in immune system modulation and glioma progression. Materials & methods: GEVs were isolated by differential centrifugation from C6 cell supernatant and characterized by size and expression of CD9, HSP70, CD39 and CD73. The glioma model was performed by injecting C6 glioma cells into the right striatum of Wistar rats in the following groups: controls (C6 cells alone), coinjection (C6 cells + GEVs) and GEVs by intranasal administration followed by immune cells, tumor size and cells proliferation analyses. Results: GEVs presented uniform size (175 nm), expressed CD9, HSP70, CD39, CD73 and produced adenosine. In vivo, we observed a reduction in tumor size, in cell proliferation (Ki-67) and in a regulatory cell marker (FoxP3). Conclusion: GEVs, administered before or at tumor challenge, have antiproliferative properties and reduce regulatory cells in the glioma microenvironment.
Subject(s)
Brain Neoplasms , Cell Proliferation/drug effects , Extracellular Vesicles , Glioma , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Glioma/drug therapy , Rats , Rats, Wistar , Tumor MicroenvironmentABSTRACT
BACKGROUND: Neoadjuvant chemoradiation for locally advanced rectal cancer combining 5-fluorouracil with radiation increases tumor regression compared with radiation alone. However, it occurs at the cost of significant treatment-related toxicity. Patients with rectal cancer using metformin have been associated with improved response to radiotherapy. OBJECTIVE: The purpose of this study was to evaluate the radiosensitizing effects of metformin in vitro and in vivo and compare it with a standard combination of radiation/5-fluorouracil. DESIGN: Colorectal cancer cell lines SW480, HT29, and HCT116 were used as models. Cell viability was compared under treatments with radiation, radiation/5-fluorouracil, metformin, radiation/metformin, and radiation/5-fluorouracil/metformin. Nude mice were injected subcutaneously with SW480 cells and treated for 1 week with radiation/5-fluorouracil, metformin, radiation/metformin, or radiation/5-fluorouracil/metformin. Tumor volume was evaluated for 4 weeks after treatment completion. The phosphorylation status of key proteins of the PI3K/Akt/mTOR pathway was determined by immunoblots. SETTINGS: This was an experimental study conducted in vitro and in vivo. PATIENTS: Animal models/cell lines were used. MAIN OUTCOME MEASURES: The end point was to investigate how metformin compares with 5-fluorouracil as a radiosensitizer. RESULTS: All cell lines significantly decreased cell viability after treatment with radiation/metformin when compared with radiation alone. Radiation/metformin was superior to radiation/5-fluorouracil in SW480 (37% vs 74%; p < 0.001). In HT29 and in HCT116, radiation/metformin was inferior to radiation/5-fluorouracil (40.0% vs 13.8%, p < 0.001 and 40.0% vs 7.0%, p < 0.001), mainly because of increased 5-fluorouracil toxicity (≤20% of cell viability). In vivo assays indicated that radiation/metformin treatment was comparable with radiation/5-fluorouracil (557 vs 398 mm; p > 0.05) and that the addition of metformin to the standard radiation/5-fluorouracil did not improve tumor response (349 mm; p > 0.05). Metformin exerted strong PI3K/Akt/mTOR pathway inactivation effects after 24-hour exposure (increasing pAMPK, p < 0.01; decreasing pAkt, p < 0.01; and pS6, p <0.05). LIMITATIONS: In vitro and in vivo chemoradiation regimens cannot be directly translated to human delivery methods. CONCLUSIONS: Metformin enhances tumor response to radiation in vitro and in vivo. Metformin is an attractive alternative radiosensitizing agent to be considered in future studies/trials. See Video Abstract at http://links.lww.com/DCR/B219. LA METFORMINA COMO AGENTE RADIOSENSIBILIZADOR ALTERNATIVO A 5FU DURANTE EL TRATAMIENTO NEOADYUVANTE PARA CÁNCER DE RECTO: La quimiorradiación neoadyuvante para el cáncer de recto localmente avanzado que combina 5FU con radiación aumenta la regresión tumoral en comparación con la radiación sola. Sin embargo, se produce a costa de una toxicidad significativa relacionada con el tratamiento. Los pacientes con cáncer de recto que usan metformina se han asociado con una mejor respuesta a la radioterapia.Evaluar los efectos radiosensibilizantes de metformina in vitro e in vivo y compararlo con la combinación estándar de radiación / 5FU.Se usaron como modelos las líneas celulares de cáncer colorrectal SW480, HT29 y HCT116. La viabilidad celular se comparó en tratamientos con radiación, radiación / 5FU, metformina, radiación / metformina y radiación / 5FU / metformina. A los ratones desnudos se les inyectó por vía subcutánea células SW480 y fueron tratados durante una semana con radiación / 5FU, metformina, radiación / metformina o radiación / 5FU / metformina. El volumen tumoral se evaluó durante 4 semanas después de la finalización del tratamiento. El estado de fosforilación de las proteínas clave de la vía PI3K / Akt / mTOR se determinó mediante inmunotransferencias.Estudio experimental in vitro e in vivo.Modelo animal / líneas celulares.El punto final fue investigar cómo la metformina se compara con 5FU como un radiosensibilizador.Todas las líneas celulares disminuyeron significativamente la viabilidad celular después del tratamiento con radiación / metformina en comparación con la radiación sola. La radiación / metformina fue superior a la radiación / 5FU en SW480 (37% frente a 74%; p <0,001). En el HT29 y el HCT116 la radiación / metformina fue inferior a la radiación / 5FU (40% vs 13.8%, p <0.001 y 40% vs 7%, p <0.001; respectivamente), debido principalmente al aumento de la toxicidad de 5FU (≤20% de la célula viabilidad). Los ensayos in vivo indicaron que el tratamiento con radiación / metformina era comparable a la radiación / 5FU (557 vs 398 mm, p > 0.05), y que la adición de metformina a la radiación estándar / 5FU no mejoró la respuesta tumoral (349 mm, p > 0.05). La metformina ejerció fuertes efectos de inactivación de la vía PI3K / Akt / mTOR después de 24 horas de exposición (aumentando pAMPK p < 0.01, disminuyendo pAkt, p < 0.01; y pS6, p < 0.05).Los regímenes de CRT in vitro e in vivo no se pueden traducir directamente a los métodos de entrega en humanos.La metformina mejora la respuesta tumoral a la radiación in vitro e in vivo. La metformina es un agente alternativo de radiosensibilización atractivo para ser considerado en futuros estudios / ensayos. Consulte Video Resumen en http://links.lww.com/DCR/B219. (Traducción-Dr Gonzalo Hagerman).
Subject(s)
Hypoglycemic Agents/pharmacology , Metformin/administration & dosage , Metformin/pharmacology , Neoadjuvant Therapy/methods , Rectal Neoplasms/therapy , Animals , Case-Control Studies , Chemoradiotherapy/standards , Combined Modality Therapy , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Hypoglycemic Agents/administration & dosage , Male , Mice , Mice, Nude , Models, Animal , Neoadjuvant Therapy/trends , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/pharmacology , Rectal Neoplasms/pathology , Signal Transduction/drug effectsABSTRACT
Glioblastoma multiforme is the most devastating tumor in the brain. Ursolic acid (UA) is found in a variety of plants, and exhibits several pharmacological activities. In this study, we investigated the effects of UA in vitro, clarifying the mechanisms that mediate its toxicity and the long-lasting actions of UA in C6 glioma cells. We also evaluated the antitumor activity of UA in an in vivo orthotopic glioma model. Cell numbers were assessed using the Trypan blue exclusion test, and the cell cycle was characterized by flow cytometry using propidium iodide staining. Apoptosis was analyzed using an Annexin V kit and by examining caspase-3. Akt immunocontent was verified by Western blot and the long-lasting actions of UA were measured by cumulative population doubling (CPD). In vivo experiments were performed in rats to measure the effects on tumor size, malignant features and toxicological parameters. In vitro results showed that UA decreased glioma cell numbers, increased the sub-G1 fraction and induced apoptotic death, accompanied by increased active caspase-3 protein levels. Akt phosphorylation/activation in cells was also diminished by UA. With regard to CPD, cell proliferation was almost completely restored upon single UA treatments, but when the UA was added again, the majority of cells died, demonstrating the importance of re-treatment cycles with chemotherapeutic agents for abolishing tumor growth. In vivo, ursolic acid slightly reduced glioma tumor size but did not decrease malignant features. Ursolic acid may be a potential candidate as an adjuvant for glioblastoma therapy.